21.11.2014 Views

Supplements - Haematologica

Supplements - Haematologica

Supplements - Haematologica

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

haematologica<br />

journal ofh<br />

hematology<br />

volume 85<br />

supplement<br />

to no. 12<br />

december 2000<br />

Mensile – Sped. Abb. Post. – 45% art. 2, comma 20B, Legge 662/96 - Filiale di Pavia. Il mittente chiede la restituzione dei fascicoli non consegnati impegnandosi a pagare le tasse dovute<br />

ISSN 0390-6078


haematologica<br />

journal ofh<br />

hematology<br />

volume 85<br />

supplement to no. 12<br />

december 2000<br />

ISSN 0390-6078<br />

editor-in-chief<br />

edoardo ascari<br />

published by the<br />

Ferrata Storti Foundation<br />

established in 1920


<strong>Haematologica</strong> – Vol. 85, supplement to n. 12, December 2000


haematologica<br />

journal of<br />

hematology<br />

hOfficial Organ of<br />

the Italian Society of Hematology,<br />

the Italian Society of Experimental Hematology,<br />

the Spanish Association of Hematology and Hemotherapy,<br />

the Italian Society of Hemostasis and Thrombosis,<br />

and<br />

the Italian Society of Pediatric Hematology/Oncology<br />

Editor-in-Chief<br />

Edoardo Ascari (Pavia)<br />

Executive Editor<br />

Mario Cazzola (Pavia)<br />

Editorial Committee<br />

Tiziano Barbui (President of the Italian Society of Hematology, Bergamo); Ciril<br />

Rozman (Representative of the Spanish Association of Hematology and<br />

Hemotherapy, Barcelona); Pier Mannuccio Mannucci (Italian Society of<br />

Hemostasis and Thrombosis, Milan)<br />

Associate Editors<br />

Carlo Brugnara (Boston), Red Cells & Iron. Federico Caligaris Cappio (Torino),<br />

Lymphocytes & Immunology. Carmelo Carlo-Stella (Milano), Hematopoiesis &<br />

Growth Factors. Paolo G. Gobbi (Pavia), Lymphoid Neoplasia. Franco Locatelli<br />

(Pavia), Pediatric Hematology. Francesco Lo Coco (Roma), Translational Research.<br />

Alberto Mantovani (Milano), Leukocytes & Inflammation. Giuseppe Masera<br />

(Monza), Geographic Hematology. Cristina Mecucci (Perugia), Cytogenetics.<br />

Pier Giuseppe Pelicci (Milano, Italian Society of Experimental Hematology),<br />

Molecular Hematology. Paolo Rebulla (Milano), Transfusion Medicine. Miguel<br />

Angel Sanz (Valencia), Myeloid Neoplasia. Salvatore Siena (Milano), Medical<br />

Oncology. Jorge Sierra (Barcelona), Transplantation & Cell Therapy. Vicente<br />

Vicente (Murcia), Hemostasis & Thrombosis<br />

Editorial Board<br />

Adriano Aguzzi (Zürich), Adrian Alegre Amor (Madrid), Claudio Anasetti (Seattle),<br />

Jeanne E. Anderson (San Antonio), Nancy C. Andrews (Boston), William Arcese<br />

(Roma), Andrea Bacigalupo (Genova), Carlo Balduini (Pavia), Luz Barbolla (Madrid),<br />

Giovanni Barosi (Pavia), Javier Batlle Fourodona (La Coruña), Yves Beguin (Liège),<br />

Marie Christine Béné (Nancy), Yves Beuzard (Paris), Andrea Biondi (Monza), Mario<br />

Boccadoro (Torino), Niels Borregaard (Copenhagen), David T. Bowen (Dundee),<br />

Ronald Brand (Leiden), Salut Brunet (Barcelona), Ercole Brusamolino (Pavia), Clara<br />

Camaschella (Torino), Dario Campana (Memphis), Maria Domenica Cappellini<br />

(Milano), Angelo Michele Carella (Genova), Gian Carlo Castaman (Vicenza), Marco<br />

Cattaneo (Milano), Zhu Chen (Shanghai), Alan Cohen (Philadelphia), Eulogio Conde<br />

Garcia (Santander), Antonio Cuneo (Ferrara), Björn Dahlbäck (Malmö), Riccardo<br />

Dalla Favera (New York), Armando D’Angelo (Milano), Elisabetta Dejana (Milano),<br />

Jean Delaunay (Le Kremlin-Bicêtre), Consuelo Del Cañizo (Salamanca), Valerio De<br />

Stefano (Roma), Joaquin Diaz Mediavilla (Madrid), Francesco Di Raimondo<br />

(Catania), Charles Esmon (Oklahoma City), Elihu H. Estey (Houston), Renato Fanin<br />

(Udine), José-María Fernández Rañada (Madrid), Evarist Feliu Frasnedo (Barcelona),<br />

Jordi Fontcuberta Boj (Barcelona), Francesco Frassoni (Genova), Renzo Galanello<br />

(Cagliari), Arnold Ganser (Hannover), Alessandro M. Gianni (Milano), Norbert<br />

C. Gorin (Paris), Alberto Grañena (Barcelona), Eva Hellström-Lindberg (Huddinge),<br />

Martino Introna (Milano), Rosangela Invernizzi (Pavia), Achille Iolascon (Bari),<br />

Sakari Knuutila (Helsinki), Myriam Labopin (Paris), Catherine Lacombe (Paris),<br />

Francesco Lauria (Siena), Mario Lazzarino (Pavia), Roberto M. Lemoli (Bologna),<br />

Giuseppe Leone (Roma), A. Patrick MacPhail (Johannesburg), Ignazio Majolino<br />

(Palermo), Patrice Mannoni (Marseille), Guglielmo Mariani (Palermo), Estella<br />

Matutes (London), Alison May (Cardiff), Roberto Mazzara (Barcelona), Giampaolo<br />

Merlini (Pavia), José Maria Moraleda (Murcia), Enrica Morra (Milano), Kazuma<br />

Ohyashiki (Tokyo), Alberto Orfao (Salamanca), Anders Österborg (Stockholm),<br />

Pier Paolo Pandolfi (New York), Ricardo Pasquini (Curitiba), Andrea Pession<br />

(Bologna), Franco Piovella (Pavia), Giovanni Pizzolo (Verona), Domenico Prisco<br />

(Firenze), Susana Raimondi (Memphis), Alessandro Rambaldi (Bergamo), Fernando<br />

Ramos Ortega (León), José Maria Ribera (Barcelona), Damiano Rondelli (Bologna),<br />

Giovanni Rosti (Ravenna), Bruno Rotoli (Napoli), Domenico Russo (Udine), Stefano<br />

Sacchi (Modena), Giuseppe Saglio (Torino), Jesus F. San Miguel (Salamanca),<br />

Guillermo F. Sanz (Valencia), Hubert Schrezenmeier (Berlin), Mario Sessarego<br />

(Genova), Pieter Sonneveld (Rotterdam), Yoshiaki Sonoda (Kyoto), Yoichi Takaue<br />

(Tokyo), José Francisco Tomás (Madrid), Giuseppe Torelli (Modena), Antonio Torres<br />

(Cordoba), Pinuccia Valagussa (Milano), Andrea Velardi (Perugia), Ana Villegas<br />

(Madrid), Françoise Wendling (Villejuif), Pier Luigi Zinzani (Bologna)<br />

Publication Policy Committee<br />

Edoardo Storti (Chair, Pavia), John W. Adamson (Milwaukee), Carlo Bernasconi<br />

(Pavia), Gianni Bonadonna (Milano), Gianluigi Castoldi (Ferrara), Albert de la<br />

Chapelle (Columbus), Peter L. Greenberg (Stanford), Fausto Grignani (Perugia),<br />

Lucio Luzzatto (New York), Franco Mandelli (Roma), Massimo F. Martelli (Perugia),<br />

Emilio Montserrat (Barcelona), David G. Nathan (Boston), Alessandro Pileri<br />

(Torino), Vittorio Rizzoli (Parma), Eduardo Rocha (Pamplona), Pierluigi Rossi Ferrini<br />

(Firenze), George Stamatoyannopoulos (Seattle), Sante Tura (Bologna), Herman<br />

van den Berghe (Leuven), Zhen-Yi Wang (Shanghai), David Weatherall (Oxford),<br />

Soledad Woessner (Barcelona)<br />

Editorial Office<br />

Igor Ebuli Poletti, Paolo Marchetto, Michele Moscato, Lorella Ripari,<br />

Rachel Stenner


h<br />

journal of<br />

hematology<br />

haematologica<br />

Associated with USPI, Unione Stampa Periodica<br />

Italiana. Premiato per l’alto valore culturale dal<br />

Ministero dei Beni Culturali ed Ambientali<br />

Editorial policy<br />

<strong>Haematologica</strong> – Journal of Hematology (ISSN 0390-6078) is owned by<br />

the Ferrata Storti Foundation, a non-profit organization created through<br />

the efforts of the heirs of Professor Adolfo Ferrata and of Professor<br />

Edoardo Storti. The aim of the Ferrata Storti Foundation is to stimulate<br />

and promote the study of and research on blood disorders and their<br />

treatment in several ways, in particular by supporting and expanding<br />

<strong>Haematologica</strong>.<br />

The journal is published monthly in one volume per year and has both a<br />

paper version and an online version (<strong>Haematologica</strong> on Internet, web<br />

site: http://www.haematologica.it). There are two editions of the print<br />

journal: 1) the international edition (fully in English) is published by the<br />

Ferrata Storti Foundation, Pavia, Italy; 2) the Spanish edition (the international<br />

edition plus selected abstracts in Spanish) is published by Ediciones<br />

Doyma, Barcelona, Spain.<br />

The contents of <strong>Haematologica</strong> are protected by copyright. Papers are<br />

accepted for publication with the understanding that their contents, all<br />

or in part, have not been published elsewhere, except in abstract form or<br />

by express consent of the Editor-in-Chief or the Executive Editor. Further<br />

details on transfer of copyright and permission to reproduce parts of<br />

published papers are given in Instructions to Authors. <strong>Haematologica</strong><br />

accepts no responsibility for statements made by contributors or claims<br />

made by advertisers.<br />

Editorial correspondence should be addressed to: <strong>Haematologica</strong> Journal<br />

Office, Strada Nuova 134, 27100 Pavia, Italy (Phone: +39-0382-531182<br />

– Fax: +39-0382-27721 – E-mail: office@haematologica.it).<br />

Year 2001 subscription information<br />

International edition<br />

All subscriptions are entered on a calendar-year basis, beginning in January<br />

and expiring the following December. Send subscription inquiries<br />

to: <strong>Haematologica</strong> Journal Office, Strada Nuova 134, 27100 Pavia, Italy<br />

(Phone: +39-0382-531182 Fax: +39-0382-27721 - E-mail:<br />

office@haematologica.it). Payment accepted: major credit cards (American<br />

Express, VISA and MasterCard), bank transfers and cheques.<br />

Subscription rates, including postage and handling, are reported below.<br />

Individual subscriptions are intended for personal use. Subscribers to the<br />

print edition are entitled to free access to <strong>Haematologica</strong> on Internet,<br />

the full-text online version of the journal. Librarians interested in getting<br />

a site licence that allows concurrent user access should contact the<br />

<strong>Haematologica</strong> Journal Office.<br />

Rates Institutional Personal<br />

Print edition<br />

Europe Euro 300 Euro 150<br />

Rest of World (surface) Euro 300 Euro 150<br />

Rest of World (airmail) Euro 350 Euro 200<br />

Countries with limited resources Euro 35 Euro 25<br />

<strong>Haematologica</strong> on Internet<br />

Worldwide Free Free<br />

Spanish print edition<br />

The Spanish print edition circulates in Spain, Portugal, South and Central<br />

America. To subscribe to it, please contact: Ediciones Doyma S.A.,<br />

Travesera de Gracia, 17-21, 08021 Barcelona, Spain (Phone: +34-93-<br />

414-5706 – Fax +34-93-414-4911 – E-mail: info@doyma.es). Subscribers<br />

to the Spanish print edition are also entitled to free access to<br />

the online version of the journal.<br />

Change of address<br />

Communications concerning changes of address should be addressed to<br />

the Publisher. They should include both old and new addresses and<br />

should be accompanied by a mailing label from a recent issue. Allow six<br />

weeks for all changes to become effective.<br />

Back issues<br />

Inquiries about single or replacement copies of the journal should be<br />

addressed to the Publisher.<br />

Advertisements<br />

Contact the Advertising Manager, <strong>Haematologica</strong> Journal Office, Strada<br />

Nuova 134, 27100 Pavia, Italy (Phone: +39-0382-531182 – Fax: +39-<br />

0382-27721 – E-mail: office@haematologica.it).<br />

16 mm microfilm, 35 mm microfilm, 105 mm microfiche and article<br />

copies are available through University Microfilms International, 300<br />

North Zeeb Road, Ann Arbor, Michigan 48106, USA.<br />

apple


haematologica<br />

hInstructions to Authors<br />

For additional information, the scientific<br />

staff of <strong>Haematologica</strong> can be reached<br />

through:<br />

mailing address: <strong>Haematologica</strong>, Strada<br />

Nuova 134, I-27100 Pavia, Italy.<br />

Tel. +39.0382.531182.<br />

Fax +39.0382.27721.<br />

e-mail: office@haematologica.it<br />

web: http://www.haematologica.it<br />

<strong>Haematologica</strong> publishes monthly Editorials, Original Papers, Reviews and Scientific Correspondence<br />

on subjects regarding experimental, laboratory and clinical hematology.<br />

Editorials and Reviews are normally solicited by the Editor, but suitable papers of this type<br />

may be submitted for consideration. Appropriate papers are published under the headings<br />

Decision Making and Problem Solving and Molecular Basis of Disease.<br />

Review and Action. Submission of a paper implies that neither the article nor any essential<br />

part of it has been or will be published or submitted for publication elsewhere before<br />

appearing in <strong>Haematologica</strong>. Each paper submitted for publication is first assigned by the<br />

Editor to an appropriate Associate Editor who has knowledge of the field discussed in the<br />

manuscript. The first step of manuscript selection takes place entirely inhouse and has<br />

two major objectives: a) to establish the article’s appropriateness for <strong>Haematologica</strong>’s<br />

readership; b) to define the manuscript’s priority ranking relative to other manuscripts<br />

under consideration, since the number of papers that the journal receives is greater than<br />

which it can publish. Manuscripts that are considered to be either unsuitable for the journal’s<br />

readership or low-priority in comparison with other papers under evaluation will not<br />

undergo external in-depth review. Authors of these papers are notified promptly; within<br />

about 2 weeks, that their manuscript cannot be accepted for publication. The remaining<br />

articles are reviewed by at least two different external referees (second step or classical<br />

peer-review). After this peer evaluation, the final decision on a paper’s acceptability<br />

for publication is made in conjunction by the Associate Editor and one of the Editors,<br />

and this decision is then transmitted to the authors.<br />

Conflict of Interest Policies. Before final acceptance, authors of research papers or<br />

reviews will be asked to sign the following conflict of interest statement: Please provide<br />

any pertinent information about the authors’ personal or professional situation that might<br />

affect or appear to affect your views on the subject. In particular, disclose any financial support<br />

by companies interested in products or processes involved in the work described. A note<br />

in the printed paper will indicate that the authors have disclosed a potential conflict of<br />

interest. Reviewers are regularly asked to sign the following conflict of interest statement:<br />

Please indicate whether you have any relationship (personal or professional situation, in particular<br />

any financial interest) that might affect or appear to affect your judgment. Research<br />

articles or reviews written by Editorial Board Members are regularly processed by the Editor-in-Chief<br />

and/or the Executive Editor.<br />

Time to publication. <strong>Haematologica</strong> strives to be a forum for rapid exchange of new<br />

observations and ideas in hematology. As such, our objective is to review a paper in 4<br />

weeks and communicate the editorial decision by fax within one month of submission.<br />

However, it must be noted that <strong>Haematologica</strong> strongly encourages authors to send their<br />

papers via Internet. <strong>Haematologica</strong> believes that this is a more reliable way of speeding<br />

up publication. Papers sent using our Internet Submission page will be processed in 2-3<br />

weeks and then, if accepted, published immediately on our web site. Papers sent via regular<br />

mail or otherwise are expected to require more time to be processed. Detailed instructions<br />

for electronic submission are available at<br />

http://www.haematologica.it.<br />

Submit papers to:<br />

http://www.haematologica.it/submission<br />

or<br />

the Editorial Office, <strong>Haematologica</strong>, Strada Nuova 134, 27100 Pavia, Italy<br />

Manuscript preparation. Manuscripts must be written in English. Manuscripts with<br />

inconsistent spelling will be unified by the English Editor. Manuscripts should be prepared<br />

according to the Uniform Requirements for Manuscripts Submitted to Biomedical Journals,<br />

N Engl J Med 1997; 336:309-15; the most recent version of the Uniform Requirements can<br />

be found on the following web site:<br />

http://www.ama-assn.org/public/peer/wame/uniform.htm<br />

With respect to traditional mail submission, manuscripts, including tables and figures,<br />

should be sent in triplicate to facilitate rapid reference. In order to accelerate processing,<br />

author(s) should also enclose a 3.5” diskette (MS-DOS or Macintosh) containing the manuscript<br />

text; if the paper includes computerized graphs, the diskette should contain these<br />

documents as well. Computer programs employed to prepare the above documents should<br />

be listed.<br />

Title Page. The first page of the manuscript must contain: (a) title, name and surname of<br />

the authors; (b) names of the institution(s) where the research was carried out; (c) a running<br />

title of no more than 50 letters; (d) acknowledgments; (e) the name and full postal<br />

address of the author to whom correspondence regarding the manuscript as well as<br />

requests for abstracts should be sent. To accelerate communication, phone, fax number<br />

and e-mail address of the corresponding author should also be included.<br />

Abstract. The second page should carry an informative abstract of no more than 250 words<br />

which should be intelligible without reference to the text. Original paper abstracts must<br />

be structured as follows: background and objectives, design and methods, results, interpretation<br />

and conclusions. After the abstract, add three to five key words.<br />

Editorials should be concise. No particular format is required for these articles, which<br />

should not include a summary.<br />

Original Papers should normally be divided into abstract, introduction, design and methods,<br />

results, discussion and references.<br />

The section Decision Making and Problem Solving presents papers on health decision science<br />

specifically regarding hematologic problems. Suitable papers will include those<br />

dealing with public health, computer science and cognitive science. This section may also<br />

include guidelines for diagnosis and treatment of hematologic disorders and position<br />

papers by scientific societies.<br />

Reviews provide a comprehensive overview of issues of current interest. No particular format<br />

is required but the text should be preceded by an abstract which should be structured<br />

as follows: background and objective, evidence and information sources, state of<br />

art, perspectives. Within review articles, <strong>Haematologica</strong> gives top priority to: a) papers<br />

on molecular hematology to be published in the section Molecular basis of disease; b)<br />

papers on clinical problems analyzed according to the methodology typical of Evidence-<br />

Based Medicine.<br />

Scientific Correspondence should be no longer than 500 words (a word count should be<br />

included by the authors), can include one or two figures or tables, and should not contain<br />

more than ten strictly relevant references. Letters should have a short abstract (≤ 50<br />

words) as an introductory paragraph, and should be signed by no more than six authors.<br />

Correspondence, i.e. comments on articles published in the Journal will only appear in


haematologica<br />

hInstructions to Authors<br />

For additional information, the scientific<br />

staff of <strong>Haematologica</strong> can be reached<br />

through:<br />

mailing address: <strong>Haematologica</strong>, Strada<br />

Nuova 134, I-27100 Pavia, Italy.<br />

Tel. +39.0382.531182.<br />

Fax +39.0382.27721.<br />

e-mail: office@haematologica.it<br />

web: http://www.haematologica.it<br />

our Internet edition. Pictures of particular interest will be published in appropriate spaces<br />

within the journal.<br />

Tables and Illustrations. Tables and illustrations must be constructed in consideration of<br />

the size of the Journal and without repetitions. They should be sent in triplicate with each<br />

table typed on a separate page, progressively numbered with Arabic numerals and accompanied<br />

by a caption in English. All illustrations (graphs, drawings, schemes and photographs)<br />

must be progressively numbered with Arabic numerals. In place of original drawings,<br />

roentgenograms, or other materials, send sharp glossy black-and-white photographic<br />

prints, ideally 13 by 18 cm but no larger than 20 by 25 cm. In preparing illustrations, the<br />

final base should be considered the width of a single column, i.e. 8 cm (larger illustrations<br />

will be accepted only in special cases). Letters and numbers should be large enough to<br />

remain legible (> 1 mm) after the figure has been reduced to fit the width of a single column.<br />

In preparing composite illustrations, each section should be marked with a small letter<br />

in the bottom left corner. Legends for illustrations should be typewritten on a separate<br />

page. Authors are also encouraged to submit illustrations as electronic files together<br />

with the manuscript text (please, provide what kind of computer and software<br />

employed).<br />

Units of measurement. All hematologic and clinical chemistry measurements should be<br />

reported in the metric system according to the International System of Units (SI) (Ann<br />

Intern Med 1987; 106:114-29). Alternative non-SI units may be given in addition. Authors<br />

are required to use the standardized format for abbreviations and units of the International<br />

Committee for Standardization in Hematology when expressing blood count results<br />

(<strong>Haematologica</strong> 1991; 76:166).<br />

References should be prepared according to the Vancouver style (for details see:<br />

http://www.ama-assn.org/ public/peer/wame/uniform.htm or also N Engl J Med 1997;<br />

336:309-15). References must be numbered consecutively in the order in which they are<br />

first cited in the text, and they must be identified in the text by Arabic numerals (in parentheses).<br />

Journal abbreviations are those of the List of the Journals Indexed, printed annually<br />

in the January issue of the Index Medicus [this list (about 1.3 Mb) can also be obtained<br />

on Internet through the US National Library of Medicine website, at the following worldwide-web<br />

address: http://www.nlm.nih.gov/tsd/serials/lji.html).<br />

List all authors when six or fewer; when seven or more, list only the first three and add et<br />

al. Examples of correct forms of references follow (please note that the last page must be<br />

indicated with the minimum number of digits):<br />

Journals (standard journal article, 1,2 corporate author, 3 no author given, 4 journal supplement<br />

5 ):<br />

1. Najfeld V, Zucker-Franklin D, Adamson J, Singer J, Troy K, Fialkow PJ. Evidence for clonal<br />

development and stem cell origin of M7 megakaryocytic leukemia. Leukemia 1988;<br />

2:351-7.<br />

2. Burgess AW, Begley CG, Johnson GR, et al. Purification and properties of bacterially<br />

synthesized human granulocyte-macrophage colony stimulating factor. Blood 1987;<br />

69:43-51.<br />

3. The Royal Marsden Hospital Bone-Marrow Transplantation Team. Failure of syngeneic<br />

bone-marrow graft without preconditioning in post-hepatitis marrow aplasia. Lancet<br />

1977; 2:242-4.<br />

4. Anonymous. Red cell aplasia [editorial]. Lancet 1982; 1:546-7.<br />

5. Karlsson S, Humphries RK, Gluzman Y, Nienhuis AW. Transfer of genes into hemopoietic<br />

cells using recombinant DNA viruses [abstract]. Blood 1984; 64(Suppl 1):58a.<br />

Books and other monographs (personal authors, 6,7 chapter in a book, 8 published proceeding<br />

paper, 9 abstract book, 10 monograph in a series, 11 agency publication 12 ):<br />

6. Ferrata A, Storti E. Le malattie del sangue. 2nd ed. Milano: Vallardi; 1958.<br />

7. Hillman RS, Finch CA. Red cell manual. 5th ed. Philadelphia: FA Davis; 1985.<br />

8. Bottomley SS. Sideroblastic anaemia. In: Jacobs A, Worwood M, eds. Iron in biochemistry<br />

and medicine, II. London: Academic Press; 1980. p. 363-92.<br />

9. DuPont B. Bone marrow transplantation in severe combined immunodeficiency with an<br />

unrelated MLC compatible donor. In: White HJ, Smith R, eds. Proceedings of the third<br />

annual meeting of the International Society for Experimental Hematology. Houston:<br />

International Society for Experimental Hematology; 1974. p. 44-6.<br />

10.Bieber MM, Kaplan HS. T-cell inhibitor in the sera of untreated patients with Hodgkin’s<br />

disease [abstract]. Paper presented at the International Conference on Malignant<br />

Lymphoma Current Status and Prospects, Lugano, 1981:15.<br />

11.Worwood M. Serum ferritin. In: Cook JD, ed. Iron. New York: Churchill Livingstone; 1980.<br />

p. 59-89. (Chanarin I, Beutler E, Brown EB, Jacobs A, eds. Methods in hematology; vol<br />

1).<br />

12.Ranofsky AL. Surgical operation in short-stay hospitals: United States-1975. Hyattsville,<br />

Maryland: National Center for Health Statistics; 1978. DHEW publication no. (PHS)<br />

78-1785, (Vital and health statistics; series 13; no. 34).<br />

References to Personal Communications and Unpublished Data should be incorporated in<br />

the text and not placed under the numbered References. Please type the references exactly<br />

as indicated above and avoid useless punctuation (e.g. periods after the initials of<br />

authors’ names or journal abbreviations).<br />

Galley Proofs and Reprints. Galley proofs should be corrected and returned by fax or<br />

express delivery within 72 hours. Minor corrections or reasonable additions are permitted;<br />

however, excessive alterations will be charged to the authors. Papers accepted for publication<br />

will be printed without cost. The cost of printing color figures will be communicated<br />

upon request. Reprints may be ordered at cost by returning the appropriate form sent<br />

by the publisher.<br />

Transfer of Copyright and Permission to Reproduce Parts of Published Papers. Authors<br />

will grant copyright of their articles to the Ferrata Storti Foundation. No formal permission<br />

will be required to reproduce parts (tables or illustrations) of published papers, provided<br />

the source is quoted appropriately and reproduction has no commercial intent. Reproductions<br />

with commercial intent will require written permission and payment of royalties.


haematologica<br />

hTable of Contents<br />

2000; vol. 85; supplement to no. 12<br />

(indexed by Current<br />

Contents/Life Sciences and in Faxon<br />

Finder and Faxon XPRESS, also available<br />

on diskette with abstracts)<br />

Papers<br />

CD34-positive cells: biology and clinical relevance<br />

Carmelo Carlo Stella, Mario Cazzola, Paolo De Fabritiis,<br />

Armando De Vincentiis, Alessandro Massimo Gianni,<br />

Francesco Lanza, Francesco Lauria, Roberto M. Lemoli,<br />

Corrado Tarella, Paola Zanon, Sante Tura .........................................1<br />

Peripheral blood stem cells in acute myeloid leukemia:<br />

biology and clinical applications<br />

Massimo Aglietta, Armando De Vincentiis, Luigi Lanata,<br />

Francesco Lanza, Roberto Massimo Lemoli, Giacomo<br />

Menichella, Agostino Tafuri, Paola Zanon, Sante Tura ....19<br />

Peripheral blood stem cell transplantation for<br />

the treatment of multiple myeloma: biological<br />

and clinical implications<br />

Federico Caligaris Cappio, Michele Cavo,<br />

Armando De Vincentiis, Luigi Lanata, Roberto Massimo<br />

Lemoli, Ignazio Majolino, Corrado Tarella, Paola Zanon,<br />

Sante Tura ......................................................................................................................32<br />

Allogeneic hematopoietic stem cells from sources other<br />

than bone marrow: biological and technical aspects<br />

Francesco Bertolini, Armando De Vincentiis, Luigi Lanata,<br />

Roberto Massimo Lemoli, Rita Maccario, Ignazio Majolino,<br />

Luisa Ponchio, Damiano Rondelli, Antonio Tabilio,<br />

Paola Zanon, Sante Tura..................................................................................49<br />

Clinical use of allogeneic hematopoietic stem cells from<br />

sources other than bone marrow<br />

William Arcese, Franco Aversa, Giuseppe Bandini,<br />

Armando De Vincentiis, Michele Falda, Luigi Lanata,<br />

Roberto Massimo Lemoli, Franco Locatelli,<br />

Ignazio Majolino, Paola Zanon, Sante Tura................................69<br />

Ex vivo expansion of hematopoietic cells<br />

and their clinical use<br />

Massimo Aglietta, Francesco Bertolini, Carmelo<br />

Carlo-Stella, Armando De Vincentiis, Luigi Lanata, Roberto<br />

Massimo Lemoli, Attilio Olivieri, Salvatore Siena, Paola<br />

Zanon, Sante Tura ..................................................................................................92<br />

Cell therapy: achievements and perspectives<br />

Claudio Bordignon, Carmelo Carlo-Stella, Mario Paolo<br />

Colombo, Armando De Vincentiis, Luigi Lanata, Roberto<br />

Massimo Lemoli, Franco Locatelli, Attilio Olivieri,<br />

Damiano Rondelli, Paola Zanon, Sante Tura ...........................117<br />

Antitumor vaccination: where we stand<br />

Monica Bocchia, Vincenzo Bronte, Mario Paolo Colombo,<br />

Armando De Vincentiis, Massimo Di Nicola, Guido Forni,<br />

Luigi Lanata, Roberto Massimo Lemoli, Massimo Massaia,<br />

Damiano Rondelli, Paola Zanon, Sante Tura ...........................156


eview<br />

CD34-positive cells:<br />

biology and clinical relevance<br />

haematologica 2000; 85(supplement to no. 12):1-18<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia L. e A. Seràgnoli,<br />

Policlinico S. Orsola, via Massarenti 9, 40138 Bologna, Italy.<br />

Acknowledgments: the preparation of this manuscript was supported<br />

by grants from Dompé Biotec SpA, Milano, and Amgen S.p.A., Milano,<br />

Italy. Received October 18, 1994; accepted May 2, 1995.<br />

CARMELO CARLO STELLA, MARIO CAZZOLA,*<br />

PAOLO DE FABRITIIS,° ARMANDO DE VINCENTIIS, #<br />

ALESSANDRO MASSIMO GIANNI, @ FRANCESCO LANZA,^<br />

FRANCESCO LAURIA,** ROBERTO M. LEMOLI,°°<br />

CORRADO TARELLA, ## PAOLA ZANON, @@ SANTE TURA°°<br />

Cattedra di Ematologia, Università di Parma, Parma; *Dipartimento<br />

di Medicina Interna e Terapia Medica, Sezione di Medicina<br />

Interna e Oncologia Medica, Università di Pavia e IRCCS<br />

Policlinico S. Matteo, Pavia; °Dipartimento di Biopatologia<br />

Umana, Sezione di Ematologia, Università “La Sapienza”,<br />

Roma; # Dompé Biotec SpA, Milano; @ Centro Trapianto Midollo<br />

Osseo “Cristina Gandini”, Divisione di Oncologia Medica C,<br />

Istituto Nazionale Tumori, Milano; ^Istituto di Ematologia e<br />

Fisiopatologia dell’Emostasi, Università degli Studi di Ferrara,<br />

Ferrara; **Istituto di Scienze Mediche, Università degli Studi<br />

di Milano, Milano; °°Istituto di Ematologia “Lorenzo e Ariosto<br />

Seragnoli”, Università degli Studi di Bologna, Bologna; ## Cattedra<br />

di Ematologia, Università di Torino, Torino; @@ Amgen<br />

S.p.A., Milano, Italy<br />

The CD34-positive cell: definition and<br />

morphology<br />

Cellular expression of the CD34 antigen identifies a<br />

morphologically and immunologically heterogeneous<br />

cell population that is functionally characterized by the<br />

in vitro capability to generate clonal aggregates derived<br />

from early and late progenitors and the in vivo capacity<br />

to reconstitute the myelo-lymphopoietic system in<br />

a supralethally irradiated host. 1-3<br />

Immunohistochemical studies have demonstrated<br />

that the CD34 antigen is stage but not lineage specific.<br />

In fact, independently of the differentiative lineage,<br />

it is expressed only by ontogenetically early cells. 4 For<br />

years a major obstacle to the morphological identification<br />

of putative hematopoietic stem cell has been the<br />

difficulty in separating them from their direct progeny.<br />

The use of CD34 and other suitable cell surface markers<br />

(i.e. CD33, CD38, HLA-DR antigens) in fluorescenceactivated<br />

cell-sorting techniques or other cell separation<br />

methods has allowed considerable progress in this<br />

field.<br />

Positively selected, lineage committed CD34 + cells<br />

and more immature, lineage negative CD34 + CD33 –<br />

HLA-DR – cells are shown in Figure 1 and Figure 2,<br />

respectively. On May-Grünwald-Giemsa stained preparations,<br />

CD34 + cells are medium sized cells having large<br />

nuclei, eccentrically surrounded by narrow rims of deep<br />

blue cytoplasm occasionally containing cytoplasmic<br />

granules. Some normal CD34 + cell nuclei show one or<br />

more pale blue nucleoli. Taken together, these findings<br />

reflect the heterogeneous proliferative status and protein<br />

synthesis of this cell population. Conversely, earlier<br />

hematopoietic progenitors, identified as CD34 +<br />

CD33– HLA-DR–, seem to be more homogeneous in size<br />

(small lymphocyte-like cells) and lack cytoplasmic granules<br />

and prominent nucleoli. Again, the morphology of<br />

this cellular population appears to reflect the functional<br />

characteristics of these cells (e.g. low protein synthesis,<br />

very low proliferative activity with predominantly<br />

G0 phase).<br />

Several monoclonal antibodies (MY10, 12.8, B1-3C5,<br />

115.2, ICH3, TUK3, etc.) raised against the leukemic cell<br />

lines KG1 or KG1a and an anti-endothelial cell antibody<br />

(QBEND10) assigned to the CD34 cluster have<br />

been shown to identify a transmembrane glycoproteic<br />

antigen of 105-120 kD expressed on 1-3% of normal<br />

bone marrow cells, 0.01-0.1% peripheral blood cells<br />

and 0.1-0.4% cord blood cells. 5 Different antibodies<br />

recognize distinct epitopes of the same antigen. CD34<br />

antigen expression is associated with concomitant<br />

expression of several other markers that can be classified<br />

as lineage non-specific markers (Thy1, CD38, HLA-<br />

DR, CD45RA, CD71) and lineage specific markers,<br />

including T-lymphoid (TdT, CD10, CD7, CD5, CD2), B-<br />

lymphoid (TdT, CD10, CD19), myeloid (CD33, CD13) and<br />

megakaryocytic (CD61, CD41, CD42b) markers. 5 The<br />

expression of lineage non-specific markers allows the<br />

heterogeneous CD34 + population to be divided into two<br />

distinct subpopulations characterized, respectively, by<br />

low or high expression of Thy1, CD38, HLA-DR, CD45RA,<br />

CD71. These two cell subpopulations contain early and<br />

late hematopoietic progenitor cells, respectively. 6-8<br />

In addition to conventional immunological markers<br />

classified on the basis of their assignation to specific<br />

clusters of differentiation, CD34 cells express receptors<br />

for a number of growth factors. Two distinct families of<br />

haematologica vol. 85(suppl. to n. 12):December 2000


2<br />

C. Carlo Stella et al.<br />

Figure 1. May-Grünwald-Giemsa (MGG) staining of cytospin<br />

preparation of enriched CD34+ cells, highly purified by<br />

avidin-biotin immunoaffinity.<br />

Figure 2. MGG staining of cytospin preparation of enriched<br />

CD34+ CD33– HLA-DR– cells. The CD34+ cell fraction was<br />

further depleted of CD33+ HLA-DR+ cells by immunomagnetic<br />

separation.<br />

related receptors have been identified: (i) tyrosine kinase<br />

receptors, including the stem cell factor receptor (SCF-<br />

R, CD117) and the macrophage colony-stimulating factor<br />

receptor (M-CSF-R, CD115); (ii) hematopoietic<br />

receptors not containing a tyrosine kinase domain, such<br />

as the granulocyte-macrophage colony-stimulating factor<br />

receptor (GM-CSF-R, CDw116). 5,9<br />

The identification of new markers selectively<br />

expressed on primitive lymphohematopoietic cells<br />

(CD34 + CD38 – ) represents a stimulating research field. In<br />

this context, stem cell tyrosine kinase receptors (STK),<br />

such as STK-1, a human homologue of the murine Flk-<br />

2/Flt-3, are of particular relevance. 10-12 The ligands for<br />

these receptors might represent new factors able to<br />

selectively control stem cell self-renewal, proliferation<br />

and differentiation. 13-15<br />

Clonogenic and biologic activity<br />

The structural and functional integrity of the<br />

hematopoietic system is maintained by a relatively small<br />

population of stem cells, located mainly in the bone<br />

marrow, that can (i) undergo self-renewal to produce<br />

more stem cells or (ii) differentiate to produce progeny<br />

which are progressively unable to self-renew, irreversibly<br />

committed to one or another of the various hematopoietic<br />

lineages, and able to generate clones of up to 10 5<br />

lineage-restricted cells that mature into specialized<br />

cells. 16-18<br />

The decision of a stem cell to either self-renew or differentiate<br />

and the selection of a specific differentiation<br />

lineage by a multipotent progenitor during commitment<br />

are intrinsic properties of stem cell progenitor cells and<br />

are regulated by stochastic mechanisms. 19 Survival and<br />

amplification of hematopoietic progenitors are controlled<br />

by a number of regulatory molecules (hematopoietic<br />

growth factors) interacting according to complex<br />

modalities (synergism, recruitment, antagonism). 19 A further<br />

level of hematopoietic control is exerted by nuclear<br />

transcription factors that activate lineage-specific genes<br />

regulating growth factor responsiveness and/or the proliferative<br />

capacity of hematopoietic cells. 20<br />

Detection of the most primitive hematopoietic cell<br />

types is now possible due to the technique of long-term<br />

bone marrow culture. In the case of human bone marrow,<br />

a 5- to 8-week time period between initiating cultures<br />

and assessing clonogenic progenitor numbers<br />

allows quantification of a very primitive cell in the starting<br />

population, the so-called long-term culture-initiating<br />

cell (LTC-IC). 21 Committed progenitors of the various<br />

hematopoietic cell classes can be quantitated by a number<br />

of short-term culture clonogenic assays. 19 CD34 antigen<br />

expression associated with low CD38 and CD45RA<br />

expression and variable HLA-DR expression is a typical<br />

feature of LTC-IC, CFU-Blast, CFU-T, CFU-B. In contrast,<br />

CD34 antigen expression associated with CD38 and HLA-<br />

DR expression is a typical feature of multipotent (CFU-<br />

GEMM) and lineage-restricted (CFU-GM, CFU-G, CFU-M,<br />

BFU-E, CFU-E, BFU-Meg, CFU-Meg) hematopoietic progenitor<br />

cells 5 (Figure 3). Recently reported data have<br />

shown that low or absent expression of the Thy1 or SCF<br />

receptor can be efficiently used to enrich primitive<br />

hematopoietic progenitors from the heterogeneous<br />

CD34 + cell population. 7,9 Although the CD34 antigen is<br />

virtually expressed by all progenitor cells, the percentage<br />

of CD34 + cells with assayable in vitro clonogenic activity<br />

ranges from 10 to 30%. The problem of non-clonogenic<br />

CD34 + cells is still open and not adequately<br />

explained by the presence of lymphoid progenitors which<br />

are not assayable with current in vitro systems. Nonproliferating<br />

CD34 + cells might represent a subpopulation<br />

that is not responsive to conventional myeloid<br />

hematopoietic growth factors. The non-proliferating<br />

CD34+ subset might require the presence of co-factors,<br />

such as the ligand of STK-1 or the hepatocyte growth<br />

factor, able to activate stem cell-specific genes whose<br />

expression is a prerequisite for acquiring responsiveness<br />

to conventional growth factors. 14,22,23<br />

In lethally irradiated non-human primates, both autologous<br />

and allogeneic CD34 + cells have been shown to<br />

have the capacity to reconstitute the myelo-lymphopoietic<br />

system, thus suggesting that the stem cell responsible<br />

for hematopoietic reconstitution is CD34 + . 24,25 It<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 3<br />

has also been shown that human CD34 + HLA-DR – cells<br />

transplanted in utero in the fetus of sheep initiate and<br />

sustain a chimeric hematopoiesis producing human<br />

progenitor cells of all differentiative lineages. 26 Autologous<br />

CD34 + cells enriched by avidin-biotin columns have<br />

been shown to be able to reconstitute myelo-lymphopoiesis<br />

in patients receiving high-dose chemoradiotherapy.<br />

27 The results of studies using CD34 + bone marrow<br />

cells in the allogeneic setting in patients receiving<br />

both related as well as unrelated allogeneic marrow<br />

transplants will soon be available. 27 In addition, trials are<br />

planned that will use allogeneic peripheral blood CD34 +<br />

cells either alone or with marrow. 27<br />

Figure 3. Cellular organization of the hematopoietic system.<br />

Characterization and function of the CD34<br />

cell surface molecule<br />

The CD34 cell surface molecule has been biochemically<br />

characterized and both the human cDNA and gene<br />

have been cloned and sequenced in the last few<br />

years. 28,29<br />

CD34 is a one-pass type I transmembrane glycoprotein<br />

with a molecular weight of 105-120 kDs in either<br />

the reduced or unreduced form 28 (Figure 4). CD34 protein<br />

is not homologous to any other known protein. The<br />

minimum size of the CD34 protein is 354-amino acids<br />

and contains nine sites for N-glycosylation and a several<br />

for O-glycosylation that are essential constituents<br />

of the three epitopes of the molecule; this molecule is<br />

also rich in sialic acid. Its biochemical composition suggests<br />

a mucin-like structure and in some respects<br />

resembles leucosialin (CD43). Sequence comparisons<br />

between human and mouse CD34 show a very low level<br />

of identity in the glycosylated region, 70% identity in<br />

the globular domain, and 92% in the transmembrane<br />

and cytoplasmic regions.<br />

Using a KG1 cell line library, it has been shown that<br />

the human CD34 gene is located on chromosome 1, and<br />

recent studies with in situ hybridization have assigned<br />

its localization to band 1q32, in close proximity to other<br />

genes that encode growth factors or function molecules<br />

such as CD1, CD45, TGF2, laminin, LAM/GMP,<br />

etc. 28<br />

Seven CD34 monoclonal antibodies (MoAbs) were<br />

clustered at the 4th Workshop on Leukocyte Differentiation<br />

Antigens (Vienna, 1988), 30,31 and another 15<br />

MoAbs were verified as recognizing the CD34 molecule<br />

during the 5th International Workshop on Leukocyte<br />

Differentiation Antigens (Boston, 1983), the most direct<br />

evidence being reactivity with cells transfected with<br />

CD34 cDNA and binding to CD34 protein. 32,33 The epitope<br />

specificity of the CD34 antibodies was classified<br />

into three distinct groups according to the sensitivity of<br />

the epitopes to enzymatic cleavage (which was performed<br />

using neuraminidase, chymopapain and glycoprotease<br />

from Pasteurella haemolytica), reactivity with<br />

fibroblasts and high endothelial venules, and cross<br />

blocking experiments (Table 1). 34,35 We know, in fact,<br />

that glycoprotease from Pasteurella haemolytica specifically<br />

cleaves only proteins containing sialylated O-<br />

linked glycans. 34 Based on these data, it can be further<br />

postulated that class III epitopes are more proximal to<br />

the extracellular side of the cell membrane than class I<br />

and class III epitopes.<br />

Furthermore, for most CD34 MoAbs (with few exceptions)<br />

cross blocking experiments are in agreement with<br />

the classification based on enzymatic cleavage of the<br />

CD34 protein. In other words, using a cocktail of CD34<br />

MoAbs, CD34 reactivity is blocked only in the case that<br />

MoAbs belonging to the same CD34 epitope are simultaneously<br />

employed. On the contrary, the combined use<br />

of MoAbs recognizing class II and III or class I and II or<br />

class III epitopes does not affect cell reactivity. Moreover,<br />

CD34 MoAbs defining class III epitopes are unable<br />

to react with CD34 glycoprotein in Western blots<br />

because this epitope is sensitive to denaturation.<br />

The pattern of expression of CD34 antibodies exhibited<br />

by CD34 + acute leukemias is partially in accordance<br />

with that derived from epitope mapping based on the<br />

differential sensitivity of CD34 to enzymatic treatment.<br />

However, about one third of CD34 MoAbs do not seem<br />

to belong to any of these subgroups and for this peculiar<br />

pattern of expression are referred to as atypical<br />

CD34 reagents. The widest variation in CD34 MoAb<br />

reactivity has been demonstrated in acute myeloid<br />

leukemia (AML) samples, allowing us to postulate the<br />

occurrence of aberrant antigens or of distinct epitopes<br />

in subgroups of leukemias. Alternatively, it can be<br />

hypothesized that the expression of different antibod-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


4<br />

C. Carlo Stella et al.<br />

Lipid bilayer<br />

GPI anchor<br />

N-glycosylation site<br />

O-glycosylation site<br />

Figure 4. Schematic representation of the CD34 cell surface<br />

molecule.<br />

Fig. 4<br />

ies could reflect the degree of maturation of leukemic<br />

cells. The presence of new, distinct non overlapping epitopes<br />

could be proposed on the basis of the data published<br />

so far in the literature. As far as the expression of<br />

CD34 in normal and leukemic cells is concerned, it has<br />

been calculated by flow cytometry that the number of<br />

molecular equivalents of soluble fluorochrome (MESF)<br />

expressed by leukemic and normal progenitors ranges<br />

from 18,200 to 322,000 and from 8,000 to 124,000,<br />

respectively.<br />

Recent data collected by Lanza et al. 36 seem to indicate<br />

that class I-type MoAbs are more sensitive to freezing<br />

procedures than class II and III MoAbs, since the<br />

epitope is not identifiable following a frozen/thawed<br />

methodology.<br />

The function of CD34 surface glycoprotein in hematopoietic<br />

stem and progenitor cells is still the object of<br />

debate. In light of recent findings, it would seem to play<br />

a relevant role in modulating cell adhesion. 36 Furthermore,<br />

it has been demonstrated that CD34 probably acts<br />

as an adhesive ligand for L-selectin. It has been further<br />

postulated that the CD34 molecule could play a protective<br />

role against proteolytic enzyme-mediated damage<br />

due to its high number of O-glycosylation sites. The cytoplasmic<br />

domain contains two sites for protein kinase C<br />

phosphorylation and one for tyrosine phosphorylation. 28<br />

Given the discordant reactivity of these molecules,<br />

the choice of the CD34 MoAb to employ may be important<br />

when analyzing cell positivity for the CD34 molecule<br />

in both leukemic and normal samples, and may be<br />

responsible for the differences reported by various<br />

authors in the literature concerning the prognostic role<br />

played by this antigen in acute myeloid leukemias. 37 The<br />

type of CD34 MoAb used to enumerate progenitor cells<br />

is probably relevant in the peripheral blood stem cell<br />

autograft setting as well, since both early and late<br />

engraftment following transplantation are, to some<br />

extent, related to the number of hemopoietic stem cells<br />

collected at the time of blood or bone marrow harvests,<br />

and to the degree of progenitor cell maturation related<br />

to the expression of lineage markers such as HLA-DR,<br />

CD71, CD38, CD33, and myeloperoxidase.<br />

Techniques for CD34 + cell separation<br />

A number of different techniques have been proposed<br />

for separating CD34 + cells. The common aim is to produce<br />

a cell population with optimal purity and viability<br />

by means of a low cost, rapid and simple separation<br />

technique. The first separation techniques exploited<br />

parameters such as size and cell density and were represented<br />

by Ficoll-Hypaque and Percoll density gradi-<br />

Epitope class Clones CD34 reactivity<br />

paraffin frozen western<br />

section section blotting<br />

I. Sensitive to neuraminidase (from Vibrio<br />

cholerae), chymopapain, glycoprotease (from<br />

Pasteurella haemolytica)<br />

II. Resistant to neuraminidase. Glycoprotease,<br />

and chymopapain sensitive<br />

14G3, BI3C5, My10,* 12.8,*<br />

ICH3,*<br />

Immu-133, Immu-409<br />

43A1, MD34.3,<br />

MD34.1, MD34.2, QBend10,<br />

4A1, 9044, 9049<br />

positive negative positive<br />

positive positive positive<br />

III. Resistant to neuraminidase, chymopapain<br />

and glycoprotease<br />

CD34 9F2,HPCA2,<br />

581, 553.563, Tuk 3, 115.2<br />

negative positive negative<br />

Table 1. Epitope specificity of<br />

CD34 MoAbs as assessed by<br />

their differential sensitivity.<br />

*Incomplete digestion by neuroaminidase.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 5<br />

ents. In the last two decades, the development of monoclonal<br />

antibodies has allowed a more specific and careful<br />

cell selection by identifying surface antigens used as<br />

targets for cell separation (Table 2).<br />

FACS (Fluorescence Activated Cell Sorter)<br />

Flow cytometry is able to physically separate different<br />

populations after incubation of cells with fluorochrome-conjugated<br />

monoclonal antibodies. This cell<br />

sorting technique can yield a highly purified (> 98%)<br />

cell population. In addition, the use of electronic gates<br />

allows selection and recovery of several subpopulations<br />

according to antigenic expression and different characteristics<br />

such as size and cytoplasmic granularity. This<br />

technique has been very useful for studying CD34 + subpopulations<br />

but cannot be employed to select large<br />

numbers of cells due to its complexity and low recovery.<br />

The recent development of high-speed cell sorting,<br />

however, might allow clinical utilization of this technique.<br />

Panning<br />

Anti-CD34 monoclonal antibodies bound to one of<br />

the surfaces of cell culture flasks were recently used to<br />

select CD34 + cells. When a cell suspension is introduced<br />

into the flask, the positive population is blocked on the<br />

plastic surface, while CD34 negative cells remain in suspension<br />

and can be easily eliminated. Adherent cells<br />

should contain the CD34 + population with a viability<br />

> 90%. 38<br />

Immunomagnetic systems<br />

Immunomagnetic beads are uniform, super-paramagnetic,<br />

polystyrene beads with affinity purified antimouse<br />

Ig covalently bound to the surface. They are<br />

equally suited for negative and positive cell separation;<br />

the rosetted target cell can easily be isolated by applying<br />

a magnet on the outer wall of the test tube for 1-2<br />

minutes. Immunomagnetic beads coupled with CD34<br />

monoclonal antibodies can be utilized for positive selection<br />

of CD34 + cells to obtain a population with > 80%<br />

viable CD34 + cells. 39 Similarly, immunomagnetic beads<br />

can be employed for negative depletion with monoclonal<br />

antibodies binding lineage-specific antigens.<br />

High-affinity chromatography based on<br />

biotin-avidin interaction<br />

This method is based on an immunoadsorption technique<br />

that relies on the high affinity interaction between<br />

the protein avidin and the vitamin biotin. Avidin-biotin<br />

immunoadsorption has been employed for both positive<br />

selection and depletion of specific cell populations. 40<br />

The instrument includes a set of non-reusable products<br />

including biotinylated anti CD34 antibody, plastic bags,<br />

filters and a column of avidin-biotin beads. An automated<br />

version controlled by a computer which guarantees<br />

reproducibility of operation and reduces risks of<br />

operator errors has been developed for clinical use. Its<br />

capacity has recently been increased so that a single<br />

column can process more than 50×10 10 bone marrow or<br />

peripheral blood cells in 1-2 hours and sustain bone<br />

marrow engraftment in patients submitted to autograft.<br />

41<br />

CD34-positive subpopulations:<br />

phenotypic and functional analysis<br />

The normal CD34 + cell population likely contains<br />

progenitors of all human lympho-hematopoietic lineages,<br />

including stem cells capable of hematopoietic<br />

reconstitution after bone marrow transplantation. 1 Levels<br />

of CD34 expression decline with differentiation; consequently,<br />

the earliest clonogenic cells (CFU-blast, LTC-<br />

IC, etc.) express the highest levels of CD34, while the<br />

most differentiated (CFU-G, CFU-M, CFU-E, CFU-Meg)<br />

express only low levels of CD34 (Figure 5). The CD34<br />

antigen has been used to identify, enumerate and isolate<br />

cells from different lympho-hematopoietic lineages,<br />

as well as develop in vitro tests for indirect evaluation<br />

of cells with different functional and clonogenic capacity.<br />

42<br />

Pre-clinical studies<br />

Several animal-human systems have been created to<br />

utilize chimeras for the study of lympho-hematopoiesis<br />

in vivo. The first experiments demonstrated the feasibility<br />

of transplanting human fetal stem cells to sheep<br />

fetuses; the postnatal presence of human cells in the<br />

sheep was documented at several points in time. 43 Furthermore,<br />

some early CD34 + subpopulations were able<br />

to repopulate sheep bone marrow; animals were transplanted<br />

in utero with CD34 + /DR – cells and the presence<br />

of a chimeric population with the functional characteristics<br />

of hemopoietic progenitors was demonstrated in<br />

the marrow and peripheral blood in a percentage of cases.<br />

44 Berenson et al. 45 also showed how hematopoietic<br />

progenitors (positive for the Ia antigen and subsequently<br />

for the CD34 antigen) could reconstitute the marrow of<br />

lethally irradiated dogs. Of the seven animals treated, all<br />

showed complete marrow engraftment after reinfusion<br />

of Ia-positive cells; only one dog died from infection. 45<br />

Similarly, marrow cells from 5 primates (baboons) were<br />

treated in vitro with a biotinylated anti-CD34 antibody<br />

and then passed through a column of avidin; after autograft,<br />

all the animals showed marrow engraftment followed<br />

by hematologic reconstitution comparable to that<br />

of control animals. 46 Furthermore, the demonstration<br />

that allogeneic CD34 + cells can reconstitute the hematopoietic<br />

system in lethally irradiated baboons confirmed<br />

that this cell population includes pluripotent<br />

stem cells. 25<br />

Lymphoid precursor cells<br />

The CD34 + cell compartment contains all the cells<br />

expressing terminal deoxynucleotidyltransferase (TdT),<br />

which is an intranuclear enzyme expressed in early lymphoid<br />

cells undergoing immunoglobulin or T-cell receptor<br />

gene rearrangement. Flow cytometry has shown that<br />

the great majority of TdT + cells in the marrow coexpress<br />

CD34, CD19 and CD10 (B-cell precursors), as well as T-<br />

cell differentiation antigens such as CD7, CD5 and CD2.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


6<br />

C. Carlo Stella et al.<br />

A small proportion of CD34 + /TdT + cells coexpress CD10,<br />

which might represent a common lymphoid progenitor<br />

for both B and T lineages. 47 Recently, Miller et al. reported<br />

that CD34 + cells may also generate NK cells in vitro. 48<br />

Granulocyte-macrophage precursors<br />

Marrow erythroid progenitors lack specific markers<br />

and therefore are difficult to identify. Glycophorin A-<br />

directed monoclonal antibodies recognize all hemoglobinized<br />

cells, but this molecule is expressed in only a<br />

small proportion of CD34 + cells and is absent in clonogenic<br />

cells. 49<br />

High levels of CD45 are present on BFU-E, but this<br />

antigen is lost by the CFU-E stage; 50 however, the<br />

CD45RO isoform is well represented on earlier erythroid<br />

progenitors, while the CD45RA isoform is present on<br />

committed myeloid progenitors. The expression of CD71<br />

(transferrin receptor) is currently considered to be the<br />

specific antigen for the CD34 + erythroid population.<br />

CD71 is present at high levels on erythroid progenitor<br />

cells and at very low levels in all the other progenitor<br />

cells. 51 Expression of CD71 increases from stem cells to<br />

BFU-E, then declines during erythroid maturation. In<br />

addition, marrow CD34 + erythroid cells might express IL-<br />

3, GM-CSF (CD116) and erythropoietin receptors, based<br />

on the action of these growth factors on CFU-erythroid<br />

cells. 52<br />

Myeloid precursor clonogenic cells (CFU-GM, CFU-G,<br />

CFU-M, CFU-MK) coexpress CD34, HLA-DR, CD117 (ckit),<br />

CD45RA, CD33 and CD13; CD15 is present at low<br />

levels on CFU-G, while CFU-M specifically express<br />

CD115 (M-CSF receptor). CFU-MK are the only CD34 +<br />

cells which express the platelet glycoproteins identified<br />

by the CD61, CD42 and CD41 monoclonal antibodies. 5<br />

Dendritic cells also originate from bone marrow, but the<br />

conditions that direct their growth and differentiation<br />

are still poorly characterized. GM-CSF stimulates the<br />

growth of dendritic cells from mouse peripheral blood;<br />

however, it was recently reported that CD34 + cells may<br />

give rise to dendritic/Langherans cells after stimulation<br />

with GM-CSF and tumor necrosis factor-α. 53<br />

Multilineage progenitors and stem cells<br />

CFU-GEMM contain precursor clonogenic cells of both<br />

myeloid and erythroid lineages and express CD34, HLA-<br />

DR, CD38, CD117 and CD45RA. They also express low<br />

amounts of CD33, but not CD13. In a hypothetical differentiation<br />

scheme involving pluripotent stem cells, the<br />

lympho-hemopoietic compartment is the next cell type<br />

and can be identified in vitro with CFU-Blast and LTC-IC.<br />

The lack of expression of CD38 is the most important<br />

characteristic of these early progeny, which represent<br />

1% of CD34 and less than 0.01% of mononuclear cells.<br />

The lack of CD38 allows separation of committed progenitors<br />

(CD34 + /CD38 + ) from earlier compartments<br />

(CD34 + /CD38 – ) by a single marker combination. 54<br />

Furthermore, the earliest CD34 + cells coexpress low<br />

levels of CD45RO6 and are negative for staining with the<br />

fluorescent dye rhodamine 123. The role of HLA-DR in<br />

defining earlier cell types is still controversial; a series<br />

Table 2. Recovery of CD34-positive cells after different separation techniques.<br />

Enrichment Recovery Large-scale<br />

separation<br />

(% CD34+<br />

in the recovered<br />

(% CD34+<br />

of the original<br />

population) sample)<br />

Negative depletion<br />

by immunomagnetic<br />

beads<br />

20-60 30-60 no<br />

Positive selection by<br />

immunomagnetic<br />

beads<br />

60-80 30-60 yes<br />

Fluorescence activated<br />

cell sorter (FACS) > 95 30 - 50 time consuming<br />

Panning 50 - 80 30 - 60 yes<br />

Ceprate SC® 50 - 80 40 - 70 yes<br />

of evidence indicates that the stem cell compartment<br />

has the CD34 + /CD38 – /HLA-DR + phenotype. 8,54 Recent<br />

works, however, have not found HLA-DR in the earliest<br />

cells. 55 These data were confirmed by the possibility that<br />

HLA-DR expression may discriminate the Ph-positive<br />

leukemic compartment (HLA-DR + ) from normal residual<br />

cells (HLA-DR – ) in chronic myeloid leukemia. 56<br />

Adhesion molecules and cytokine<br />

receptors<br />

A number of molecules within the integrin family have<br />

been shown to mediate interactions between early<br />

CD34-positive cells and bone marrow stromal cells.<br />

These include VLA-4/VCAM-1, VLA-5/fibronectin, LFA-1<br />

or ICAM, and several others. Each adhesion molecule<br />

appears to mediate a specific cell interaction.<br />

Hematopoietic growth factors may be active in a soluble<br />

form or in a membrane-anchored form; adhesion<br />

molecules may be crucial for allowing anchored growth<br />

factors to bind the target cell. Table 3 lists the main<br />

adhesion molecule and growth factor receptors<br />

expressed on CD34 + cells.<br />

CD34 expression on normal and neoplastic<br />

cells<br />

It has been known that CD34 monoclonal antibodies<br />

bind specifically to vascular endothelium ever since a<br />

110 kd protein extracted from freshly isolated umbilical<br />

vessel endothelium was identified with CD34 antibodies<br />

in Western blots and in Northern blot analysis. 57,58<br />

CD34 molecules have a striking ultrastructural localization<br />

on endothelial cells: they are concentrated primarily<br />

on the luminal side, in particular on membrane<br />

processes, many of which interdigitate between adjacent<br />

endothelial cells. 57,58 Since this region is an important<br />

site for leukocyte adhesion and transendothelial<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 7<br />

traffic, in contrast to previous experiences, 33 it has been<br />

hypothesized that CD34 may be antagonistic or<br />

inhibitory to the adhesive functions of vascular endothelium.<br />

This was supported by the demonstration that<br />

CD34 gene expression at the mRNA level is reciprocally<br />

down-regulated when adhesion molecules ICAM-1<br />

and ELAM-1 are up-regulated by IL-1 during inflammatory<br />

skin lesions associated with leukocyte infiltration.<br />

33,59 Furthermore, additional studies conducted on<br />

both paraffin embedded and cryopreserved sections<br />

demonstrated that fibroblasts also react with anti-CD34<br />

MoAbs. 60 However, it should be noted that while CD34<br />

MoAbs reacted with all classes of epitopes on cryopreserved<br />

sections, class III epitopes were not recognized by<br />

specific anti-CD34 MoAbs on paraffin embedded sections.<br />

Levels of CD34 expression, highest in immature<br />

hematopoietic precursor cells, decrease progressively<br />

with cell maturation.<br />

Regarding hematologic malignancies, CD34 is<br />

expressed in a large percentage of acute leukemias. 61<br />

The fluorescence intensity of CD34 expression is variable<br />

and higher in acute lymphoblastic (ALL) than in acute<br />

myeloblastic leukemia (AML). In these latter patients,<br />

the CD34 antigen is found on 40-60% of leukemic blasts<br />

and is most frequently associated with M0, M1 and M4<br />

FAB cytotype, secondary leukemias, karyotypic abnormalities<br />

involving chromosome 5 or 7, P170 expression<br />

and poor prognosis. 61-64 Thus, CD34 expression may be<br />

considered the most predictive negative factor in AML<br />

patients strictly correlated with intensity of expression.<br />

65,66 In ALL, CD34 is expressed in 70% of patients,<br />

particularly in those with a B-lineage phenotype. In<br />

these patients, unlike AML cases, the clinical relevance<br />

of CD34 expression is controversial; however, according<br />

to the findings of a Pediatric Oncology Group, 67 its<br />

expression in B-lineage cases was associated with<br />

hyperdiploidy, lower frequency of initial central nervous<br />

system (CNS) leukemia and a favorable prognosis. In T-<br />

cell ALL cases, on the other hand, CD34 expression<br />

showed a positive correlation with initial CNS leukemia<br />

and CD10 negativity, but not with any presenting favorable-risk<br />

characteristics. 67,68<br />

Lastly, CD34 and HLA-DR expression may be very useful<br />

in discriminating between the very few benign primitive<br />

hematopoietic progenitors and their malignant<br />

counterparts in patients with chronic myeloid leukemia<br />

(CML). In fact, it has been demonstrated that normal<br />

progenitor cells are CD34 + and HLA-DR – , while malignant<br />

progenitor cells, which exhibit Ph and bcr/abl<br />

rearrangement, express HLA-DR antigens. 56<br />

Positive and negative regulators of<br />

hematopoietic progenitor cells<br />

The hematopoietic stem cell is defined by its extensive<br />

self-renewal capacity, multilineage differentiation<br />

potential and capacity for of long-term reconstitution<br />

of normal marrow function in lethally irradiated animals.<br />

68 Transplantation of retrovirally marked murine<br />

Figure 5. Functional differentiation and antigenic expression of hematopoietic cells.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


8<br />

C. Carlo Stella et al.<br />

Table 3. Adhesion molecule and growth factor receptors<br />

expressed on CD34-positive cells.<br />

Antigen name CD Ligand<br />

GlyCAM-L/selectin none adhesion molecule<br />

ICAM1,2,3 CD11a/CD18 adhesion molecule<br />

H-CAM CD44 adhesion molecule<br />

VLA-4 CD49d/CD29 adhesion molecule<br />

VLA-5 CD49d/CD29 adhesion molecule<br />

FGF-R none growth factor<br />

M-CSF CD115 growth factor<br />

GM-CSF-R CD116 growth factor<br />

SCF (c-kit) CD117 growth factor<br />

Interferon γ-R CD119 growth factor<br />

IL 7-R CD127 growth factor<br />

stem cells has shown that only a few multilineage progenitor<br />

cells induce the repopulation of engrafted<br />

hematopoietic tissue, 69 suggesting that hematopoiesis<br />

may be supported by a succession of short-lived clones.<br />

Moreover, experimental evidence indicates that the<br />

processes of self-renewal, differentiation and selection<br />

of lineage potentials are intrinsic properties of the stem<br />

cells and occur according to a stochastic (random) model.<br />

70 In the steady state, most of the stem cells are quiescent<br />

(G0 phase) and begin active cycling randomly. 71<br />

Conversely, survival and proliferation of hematopoietic<br />

progenitors are regulated by cytokines, which also act<br />

in preventing apoptosis. 72 According to this model, the<br />

induction of differentiation by a cytokine may be considered<br />

as the consequence of the proliferation of a specific<br />

population stimulated by that factor. The broad<br />

term cytokines includes growth factors such as fibroblast<br />

growth factor, colony stimulating factors (CSFs) (e.g.<br />

granulocyte-CSF, granulocyte-macrophage-CSF), and<br />

interleukins (ILs). Depending on their target cells and<br />

different proliferative potential, cytokines can be divided<br />

into three categories: 71 1) late-acting, lineage-specific<br />

factors; 2) intermediate-acting, lineage-nonspecific<br />

factors; 3) early-acting growth factors inducing the<br />

recruitment of dormant early progenitors in the cell<br />

cycle (Figure 6). 71<br />

The majority of late-acting factors support the proliferation<br />

and maturation of lineage-committed progenitors<br />

and the functional properties of terminally differentiated<br />

cells. Erythropoietin (Epo) is the physiologic<br />

regulator of erythropoiesis 73 and thrombopoietin has the<br />

same role in thrombopoiesis, 74 while M-CSF and IL-5<br />

are considered specific for macrophages and eosinophils,<br />

respectively. G-CSF exerts its activity on committed<br />

neutrophil precursors, although it has been shown to<br />

be a synergistic factor for primitive hematopoietic<br />

cells. 75<br />

Intermediate-acting, lineage-nonspecific factors<br />

include IL-3, IL-4 and GM-CSF. Their activity is mainly<br />

directed toward progenitor cells in the intermediate<br />

stages of hematopoietic development. However, they<br />

interact with later-acting growth factors for the production<br />

of more mature cells, 76 as well as with the<br />

cytokines capable of triggering stem cell cycling. On<br />

their own, they act as survival factors for stem cells and<br />

appear to stimulate early hematopoietic precursors only<br />

after their exit from G0. 77<br />

Several cytokines have recently been identified for<br />

their capacity to stimulate the proliferation of the earliest<br />

hematopoietic cells. Mapping studies of normal<br />

blast cell colony formation from single progenitors have<br />

shown that IL-6, G-CSF, IL-11, stem cell factor (SCF), IL-<br />

12 and leukemia inhibitory factor (LIF) recruit dormant<br />

stem cells into the cell cycle that are then able to<br />

respond to additional growth factors. 77 Whereas the permissive<br />

factors retain limited proliferative potential by<br />

themselves, they strongly enhance the stimulatory activity<br />

of IL-3, GM-CSF, G-CSF and EPO on CD34 + and more<br />

immature CD34 + lineage-progenitors. 78 In addition to<br />

positive interaction with intermediate-and late-acting<br />

growth factors, SCF synergistically or additively augments<br />

the colony-forming ability of other early-acting<br />

growth factors, such as IL-11, IL-12 and IL-6, on myeloid<br />

and bilineage (i.e. lymphomyeloid) primitive cells. 79<br />

Recently, the ligands for the STK-1 or FLT3 receptor, and<br />

the hepatocyte growth factor were shown to be able to<br />

stimulate very primitive hematopoietic progenitor cells.<br />

However, their biological activity is still under investigation.<br />

The main sources of both positive and negative regulatory<br />

proteins are accessory myeloid cells and the<br />

stromal component of the bone marrow. In general,<br />

microenvironment cells do not constitutively produce<br />

cytokines, rather transcription and/or translation<br />

processes are rapidly induced by cytokines such as IL-1,<br />

TNF. The extracellular matrix also participates in the<br />

regulation of hematopoiesis by binding growth factors<br />

and presenting them in a biologically active form to<br />

bone marrow progenitor cells. 80<br />

Most data suggest that stem cells express low levels<br />

of growth factor receptors and require multiple proliferative<br />

stimuli to enter into the cell cycle, while committed<br />

progenitor cells can be effectively stimulated by<br />

individual cytokines. 23 Combinations of two or more<br />

growth factors 81 can stimulate hematopoietic cells<br />

either by amplifying the progeny cell production of single<br />

precursors (synergy) or by inducing additional clonogenic<br />

cells to proliferate (recruitment). Examples of<br />

these two types of enhancement are given in Figures 7<br />

and 8, where CD34 + CD33 – DR – cells are simultaneously<br />

stimulated by two or three growth factors. The molecular<br />

basis regulating the complex interplay between<br />

cytokines is still largely unknown. However, one proposed<br />

mechanism for growth factor synergism is the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 9<br />

induction of CSF receptors on early hematopoietic progenitor<br />

cells. 82 This appears to be a coordinate cascade<br />

transactivation via up-modulation of growth factor<br />

receptors that leads to proliferation and differentiation<br />

of human marrow cells. 83 Conversely, the structural<br />

homology between some growth factors, 84 the presence<br />

of shared receptor subunits on the cell membrane 85 and<br />

common signal-transduction proteins 86 provide a potential<br />

explanation for their functional similarities and the<br />

apparent redundancy of their activity.<br />

The growth of hematopoietic progenitor cells is also<br />

regulated by soluble negative regulators such as<br />

macrophage inflammatory protein-1a (MIP-1a), tumor<br />

necrosis factor-α (TNF-α), interferons (IFNs), prostaglandins<br />

and transforming growth factor-β (TGF-β). The<br />

TGF-β family of proteins includes at least five isoforms<br />

(TGF-β1-5) which are encoded by different genes 87 and<br />

produced by stromal cells, platelets and bone cells.<br />

Moreover, a subset of very primitive murine hematopoietic<br />

cells has been shown to secrete active TGF-β1 by an<br />

autocrine mechanism. 88 TGF-β1 and 2 isoforms are<br />

bimodal regulators of murine and human hematopoietic<br />

progenitor cells, and their activity is based upon the<br />

differentiation state of the target cells and the presence<br />

of growth factors. 89 In the human system, for<br />

instance, CFU-GEMM derived from purified CD34 +<br />

CD33 – cells are inhibited by TGF-β1, whereas more committed<br />

progenitors such as CFU-G or CFU-GM are not<br />

affected. In addition, high proliferative potential-CFC<br />

(HPP-CFC) responsive to a combination of CSFs are<br />

markedly inhibited by TGF-β1, while more mature CFU-<br />

GM are actually enhanced when GM-CSF is used as<br />

colony forming factor. 78 TGF-β1 and 2-induced myelosuppression<br />

is partially counteracted by early-acting<br />

growth factors such as G-CSF, IL-6 and fibroblast<br />

growth factor. 90 On the other hand, TGF-β3 has been<br />

shown to be a more potent suppressor of human BM<br />

precursors and its activity on hematopoiesis is only<br />

inhibitory, 89 although the synergistic growth factors IL-<br />

11 and IL-9 seem to be able to oppose the negative regulation<br />

of TGF-β3 on human CD34 + cells. 91 Several<br />

potential modes of action of the TGF-β family have been<br />

suggested, including down-modulation of cytokine<br />

receptors, 92 interaction with the underphosphorylated<br />

form of the retinoblastoma gene product in late G1<br />

phase, 93 and alteration of gene expression. 94 Early studies<br />

have shown that TGF-β1 and 3 exert their activity on<br />

normal and leukemic cells by lengthening or arresting<br />

the G1 phase of the cell cycle, 95 and this effect is functional<br />

to protect normal CD34 positive cells from the<br />

toxicity of alkylating agents in vitro. 96 More recent<br />

investigations have demonstrated that TGF-β regulates<br />

the responsiveness of mice hematopoietic cells to SCF,<br />

which is known to be the main synergistic factor for<br />

both murine and human stem cells, through a decrease<br />

in c-kit mRNA stability that leads to decreased cell-surface<br />

expression. 97<br />

Similarly to TGF-β, TNF-α has been reported to have<br />

both inhibitory and stimulatory effects on hematopoietic<br />

progenitor cells. TNF-α potentiated the IL-3 and<br />

GM-CSF-mediated growth of human CD34 + cells in<br />

short-term liquid culture assay. However, it inhibited<br />

STEM CELLS<br />

SCF<br />

<br />

IL-6<br />

<br />

<br />

IL-1<br />

<br />

IL-3<br />

IL-9<br />

IL-11<br />

IL-12<br />

G-CSF<br />

GM-CSF<br />

PROGENITOR<br />

CELLS<br />

IL-4<br />

G-CSF<br />

<br />

IL-3<br />

<br />

SCF<br />

M-CSF<br />

IL-5<br />

<br />

IL-3<br />

EPO<br />

IL-9<br />

SCF<br />

<br />

SCF<br />

IL-3<br />

<br />

IL-6<br />

IL-11<br />

<br />

GM-CSF<br />

GM-CSF<br />

GM-CSF<br />

MATURING<br />

CELLS<br />

<br />

<br />

<br />

IL-5<br />

<br />

<br />

EPO<br />

<br />

<br />

<br />

IL-6<br />

<br />

EPO<br />

<br />

G-CSF<br />

GM-CSF<br />

M-CSF<br />

MEG-CSF<br />

<br />

Neutrophils<br />

<br />

<br />

<br />

<br />

Red cells<br />

Platelets<br />

Figure 6. Cytokines exert their activity at different levels of the hematopoietic differentiation pathway. Each progenitor cell is<br />

concurrently affected by multiple regulators.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


10<br />

C. Carlo Stella et al.<br />

the growth promoting activity of G-CSF. 98 Two TNF<br />

receptors with molecular weights of 55 and 75 kd,<br />

respectively, have recently been identified. 99 Whereas<br />

the p55 receptor mediates TNF-α effects on committed<br />

progenitor cells, the p75 receptor is involved in signaling<br />

the inhibition of murine primitive cells.<br />

Furthermore, it was recently shown that TNF-α is<br />

capable of inhibiting the multi-growth factor (GM-CSF,<br />

IL-3, G-CSF, SCF, IL-1)-dependent growth of human<br />

HPP-CFC derived from CD34 + cells through interaction<br />

with both p55 and p75 receptors, while the p55 receptor<br />

exclusively mediates the bifunctional activity of TNFα<br />

on more mature BM precursors responsive to single<br />

cytokines. 100<br />

MIP-1a is a peptide of 69-amino acids with a molecular<br />

weight of 7.8 kd produced by activated macrophages,<br />

T-cells and fibroblasts. 101 It belongs to a large<br />

family of putative cytokines that includes MIP-1β, MIP-<br />

2 and IL-8 (chemokines). Biologic characterization has<br />

shown that MIP-1α enhances the M-CSF- and GM-CSFdependent<br />

growth of CFU-GM, while it inhibits the<br />

colony-forming ability of hematopoietic precursors present<br />

in a cell population enriched for CD34 ++ DR + cells<br />

stimulated with erythropoietin, IL-3 and GM-CSF. 102<br />

Figure 7. Human CD34 + CD33- DR- cells were stimulated by<br />

IL-9 (A) or IL-9 and SCF (B) in the presence of EPO. The addition<br />

of SCF induced the growth of large multicentered BFU-<br />

E colonies containing > 10,000 cells. The different size of<br />

the colonies indicates amplification of stem cell progeny<br />

(synergy).<br />

A<br />

B<br />

Taken together, these results indicate that a complex<br />

interplay between positive and negative regulatory proteins<br />

determines the proliferation or inhibition of early<br />

hematopoietic progenitor cells (Figure 9). In general, the<br />

activity of inhibitors of hemopoiesis appears to be<br />

reversible, lineage-nonspecific and directed at the early<br />

stages of differentiation. In addition, TGF-β has shown<br />

some degree of differential activity between normal and<br />

neoplastic lymphoid cells. 96 Thus, negative regulators<br />

may be clinically relevant to the protection of the hematopoietic<br />

stem cell compartment from the dose-limiting<br />

toxicity of neoplastic disease therapy. 96,103,104<br />

Collection of CD34 + cells<br />

Bone marrow and peripheral blood are the only<br />

sources of immature hemopoietic precursors identified<br />

as CD34 + cells. A diagnostic marrow sample contains<br />

only a few CD34 + cells, while even fewer of them are<br />

present in peripheral blood samples taken under steady<br />

state conditions. Large quantities of CD34 + cells can be<br />

collected with massive marrow harvests, such as for<br />

transplantation purposes. Nevertheless, marrow CD34 +<br />

cells are somewhat elusive due to their scattering<br />

among the predominant CD34 + hematopoietic population.<br />

105 Recently developed cell separation procedures<br />

allow collection of highly enriched CD34 + cell populations.<br />

However, this is generally accomplished through<br />

aspecific and often unacceptable cell loss. 106 So far the<br />

limited number of immature precursors commonly<br />

obtained from both bone marrow and peripheral blood<br />

has been the major obstacle to a simple identification<br />

and analysis of marrow CD34 + cells. Indeed, the growing<br />

interest in CD34 + cells is primarily the result of the<br />

development of new therapeutic modalities that allow<br />

easy access to large quantities of hemopoietic precursors<br />

through the peripheral blood. The key role in these<br />

innovative approaches is represented by the introduction<br />

of hemopoietic growth factors for clinical use. 107<br />

At present GM-CSF and G-CSF are the most extensively<br />

employed and studied hemopoietic growth factors<br />

in the clinical setting. From the very beginning, it was<br />

observed that GM-CSF or G-CSF administration was<br />

associated with an increase in circulating hemopoietic<br />

progenitors. 108,109<br />

Later on, it was demonstrated that this phenomenon<br />

could be extensively and reproducibly amplified by combining<br />

growth factor administration with high-dose<br />

chemotherapy. 110-112<br />

Indeed hemopoietic progenitors are massively, though<br />

transiently, mobilized into peripheral blood during<br />

hemopoietic recovery following high-dose chemotherapy<br />

given with growth factor support. Such an abundance<br />

of immature hemopoietic cells makes them easily<br />

recognizable by cell sorting techniques that employ<br />

anti-CD34 MoAbs. 113 Under optimal conditions, the proportion<br />

of CD34 + cells may reach values as high as 20-<br />

30% of the total leukocyte count. In steady state conditions<br />

CD34 + cells do not exceed 4% of the total marrow<br />

population, while they are undetectable by cell sort-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 11<br />

Figure 8. The addition of SCF to IL-11, in the presence of<br />

EPO, results in a much higher number of colony-forming<br />

cells derived from CD34+ CD33– DR– progenitors (right), as<br />

compared to the IL-11 and EPO combination (left) (recruitment).<br />

ing techniques in the peripheral blood. Chemotherapy<br />

and growth factors do not merely induce a relative<br />

increase of immature hemopoietic cells; their absolute<br />

number is amplified several times over basal conditions.<br />

This allows collection of sufficient amounts of hemopoietic<br />

progenitors for autografting purposes by means<br />

of a few leukapheresis procedures. 110,113,114<br />

Values of 10-20×10 4 CFU-GM/kg represent the minimal<br />

required dose for marrow engraftment with peripheral<br />

blood progenitors. 115-117 In fact, much higher quantities<br />

of circulating progenitors can be collected using<br />

appropriate mobilization procedures. Under optimal<br />

conditions, circulating CD34 + cell peak values may range<br />

between 150 and 700/µL on days of maximal mobilization.<br />

As a rule, at least 8×10 6 CD34 + cells/kg or more can<br />

thus be collected with 1 to 3 leukapheresis procedures.<br />

114 These huge quantities, approximately corresponding<br />

to 50×10 4 CFU-GM/kg, must be considered<br />

the ideal threshold dose of peripheral blood progenitors<br />

for autografting purposes. Indeed values of 8×10 6 CD34 +<br />

cells/kg or more guarantee a rapid and durable hemopoietic<br />

recovery when circulating progenitors are used<br />

as the sole source for marrow reconstitution following<br />

submyeloablative treatment. 118-121<br />

Thus far, massive CD34 + cell mobilization has been<br />

most commonly observed when growth factor is administered<br />

following high-dose cyclophosphamide, given at<br />

7 g/m 2 . Indeed chemotherapy that induces profound<br />

leukocytopenia seems to be crucial for optimal mobilization;<br />

for instance, cyclophosphamide at doses lower<br />

than 7 g/m 2 produces a reduced mobilizing stimulus.<br />

111,122 Several other chemotherapy schedules have<br />

also been successfully employed for mobilization purposes.<br />

The principal chemotherapy protocols reported<br />

to be highly effective in inducing CD34 + cell mobilization<br />

are summarized in Table 4.<br />

110, 111, 115, 117, 122-127<br />

As stressed earlier, hemopoietic growth factors play a<br />

key role in mobilization. This has been clearly documented<br />

with cyclophosphamide. A median peak value of<br />

75 CD34 + cells/µL has been recorded following highdose<br />

cyclophosphamide alone, whereas 420 and 500/µL<br />

are the median values recorded when GM-CSF or G-<br />

CSF, respectively, are added to cyclophosphamide.<br />

112,113,128,129 Extensive growth factor-induced mobilization<br />

is further substantiated by the possibility of collecting<br />

sufficient CD34 + cells using growth factor<br />

alone. 130,131 In this setting the most promising experiences<br />

have been produced with G-CSF. The results<br />

reported indicate new opportunities for the utilization<br />

of mobilized progenitors in allogeneic transplantation<br />

procedures. 132,133 Combined chemotherapy and growth<br />

Self renewal<br />

Differentiation<br />

Negative Positive Survival<br />

IFN , , TGF- 1-2<br />

TGF- 3 TNF-<br />

MIP-1<br />

SCF<br />

IL-11<br />

IL-12<br />

IL-6<br />

G-CSF<br />

IL-1<br />

IL-9(?)<br />

IL-11<br />

GM-CSF<br />

Figure 9. Interplay between positive<br />

and negative regulatory proteins<br />

acting on hematopoietic<br />

stem cells.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


12<br />

C. Carlo Stella et al.<br />

factors remain the most efficient mobilization procedure<br />

at this time. However, ongoing studies are directed<br />

at improving mobilization efficiency with growth factors<br />

alone. For example, G-CSF at doses higher than 5<br />

µg/kg/day up to 20 µg/kg/day may have higher mobilization<br />

capacity. 134 Further improvement might derive<br />

from the clinical availability of new cytokines, such as<br />

IL-3, SCF and others, to be employed alone or in combination<br />

with G- or GM-CSF. 135-137 However, the potentially<br />

high mobilizing activity of such new cytokine combinations<br />

must be accompanied by no or very few side<br />

effects in order to be considered for a wide clinical<br />

applicability.<br />

Mobilizing protocols generally include daily delivery<br />

of growth factors, starting 1 to 3 days after chemotherapy<br />

administration and continuing until harvesting procedures<br />

are completed. Growth factor is usually administered<br />

for a total of 7-12 consecutive days. The most<br />

convenient route of delivery is subcutaneous, with 1 to<br />

2 doses per day. Progenitor cell harvests are performed<br />

during hemopoietic recovery, provided that progenitor<br />

cell mobilization is documented. Indeed various parameters<br />

have been considered as an indirect indication of<br />

progenitor mobilization, including an increase in WBC or,<br />

alternatively, in monocytes, basophils or platelets. 138-140<br />

However, CD34 + cell evaluation remains mandatory for<br />

an accurate definition of the extent of progenitor mobilization.<br />

141,142<br />

CD34 + cells should be monitored daily from the early<br />

Table 4. Main chemotherapy protocols employed in hematopoietic progenitor<br />

mobilization.<br />

Authors Protocol Drug(s) Dosage<br />

characteristics employed<br />

Gianni et al.110 single agent cyclophosphamide high<br />

Tarella et al.123 single agent etoposide high<br />

Gianni et al.124<br />

Kotasek et al.121 single agent cyclophosphamide intermediate<br />

Tarella et al.122<br />

Dreyfus et al.125 single agent cytarabine high<br />

Schimazaki et al.126 multiple drugs etoposide high<br />

cytarabine<br />

Kawano et al.115 multiple drugs daunorubicin intermediate<br />

cyclophosphamide<br />

etoposide<br />

Pettengell et al.127 multiple drugs doxorubicin intermediate<br />

cyclophosphamide<br />

etoposide<br />

Hass et al.117 multiple drugs cytarabine high<br />

mitoxantrone<br />

stages of hemopoietic recovery. Detection of circulating<br />

CD34 + cells is not sufficient reason for starting leukapheresis<br />

procedures; an adequate number of CD34 + cells<br />

(>20-50/µL) and WBC > 1.0×10 9 /L and platelet count<br />

>30×10 9 /L are required for safe and effective progenitor<br />

cell harvesting. 114,142 Leukaphereses are performed<br />

using continuous-flow blood cell separators. A complete<br />

leukapheresis procedure generally takes 2-3 hours and<br />

the total blood volume processed ranges from 6 to 10<br />

liters. 114,140,143,144 The harvested cells are resuspended in<br />

freezing medium and then cryopreserved for subsequent<br />

transplantation. One to 3 leukaphereses repeated on<br />

consecutive days usually provide large amounts of progenitor<br />

cells capable of rapid engraftment after submyeloablative<br />

treatments.<br />

Factors and procedures favoring CD34 + cell mobilization<br />

have been well established. However, other conditions<br />

adversely influencing the mobilization phenomenon<br />

should be considered. A major limitation is represented<br />

by impaired marrow function, as can occur in<br />

previously treated patients. 111 In fact, patients at first<br />

relapse following a single treatment protocol maintain<br />

an adequate mobilization capacity; 145 however, few if<br />

any mobilized CD34 + cells can be obtained from heavily<br />

treated patients previously exposed to multiple<br />

chemotherapy courses. Mobilization is also profoundly<br />

reduced and often totally abolished in patients previously<br />

exposed to radiotherapy, especially if it was delivered<br />

to the pelvis or to extended vertebral areas. 117,123<br />

Lastly, marrow invasion by tumor cells may negatively<br />

affect mobilization capacity. This is typically reported in<br />

myeloma patients in whom the extent of CD34 + cells<br />

often correlates with the degree of marrow involvement<br />

by tumoral plasma cells. 122 In conclusion, several new<br />

findings have dramatically improved the procedures for<br />

collection of large amounts of CD34 + cells. However,<br />

optimal marrow function is a prerequisite for exploiting<br />

fully the activity of all those factors known for their<br />

mobilization-inducing capacity.<br />

CD34 + positive cells in umbilical cord blood<br />

Significant numbers of human hematopoietic stem<br />

cells can be found in umbilical cord blood and can be<br />

used for allogeneic bone marrow transplantation.<br />

Mayani et al. 146 found that 1-2% of the total number of<br />

cord blood-derived low-density cells express high levels<br />

of the CD34 antigen and low or undetectable levels of<br />

the antigens CD45RA and CD71. These populations were<br />

highly enriched in clonogenic cells (34%), in particular<br />

in multipotent progenitors (12%). By culturing these<br />

cells at low concentration for 8-10 days in highly<br />

defined serum-free liquid cultures supplemented with<br />

various hematopoietic cytokines, it was possible to<br />

achieve a significant expansion (about 100-fold) of the<br />

CD34 + cell population.<br />

These last results were recently confirmed by Traycott<br />

et al. 147 in an elegant study showing that umbilical cord<br />

blood CD34 + cells rapidly exit G0-G1 phases and start to<br />

cycle in response to stem cell factor. This feature would<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 13<br />

make cord blood CD34 + cells more suitable candidates<br />

than bone marrow cells for ex vivo expansion.<br />

It is clear that in vitro expansion and maturation of<br />

hematopoietic progenitor cells might be of particular<br />

relevance for transplantation of cord blood hematopoietic<br />

cells; however, information about the effects of<br />

such an expansion on the cells required for long-term<br />

hematopoietic reconstitution is highly desirable.<br />

The dual role of peripheral blood<br />

hematopoietic progenitor cells in oncohematology<br />

CD34 + progenitor cells circulating in the peripheral<br />

blood represent an enriched and easily accessible source<br />

of two distinct cell populations: committed progenitor<br />

cells and hematopoietic stem cells.<br />

Although circulating progenitors are commonly called<br />

stem cells, the presence of circulating stem cells has<br />

been formally proved only in mice. 148 In humans the<br />

presence of hematopoietic stem cells in the peripheral<br />

blood is almost certain (see below), but to call reinfusion<br />

of circulating progenitors transplantation of<br />

peripheral blood stem cells (PBSC or similar acronyms)<br />

is hardly appropriate. This terminology overlooks the fact<br />

that the tremendous interest in peripheral blood autografting<br />

is not (at least so far) a consequence of its being<br />

a simple surrogate for autologous bone marrow transplantation<br />

(as the term PBSC would suggest), but rather<br />

derives from the unique property of this procedure to<br />

reduce the duration of the severe pancytopenia that follows<br />

submyeloablative treatments from two-three<br />

weeks (when bone marrow cells are used) to a few days<br />

only. 110,149 The reason for the rapid recovery which occurs<br />

after circulating progenitor autotransfusion (CPAT) has<br />

not been formally proved, but it is most likely a consequence<br />

of the much larger (10- to 100-fold higher)<br />

amount of committed progenitors reinfused when a<br />

patient is autografted with blood-derived (as opposed to<br />

marrow-derived) cells. The most likely hypothesis is that<br />

these late progenitors (post-progenitors) of granulocytes<br />

and platelets are capable of giving rise to mature<br />

progeny within a few days, thus allowing submyeloablated<br />

patients to survive the initial aplastic phase.<br />

The fundamental role of committed progenitor cells<br />

was elegantly proved in mice by Jones et al. 150 None of<br />

the lethally irradiated animals transplanted with a pure<br />

population of stem cells free of more mature progenitors<br />

(CFU-GM, CFU-S) survived the initial aplasia. A<br />

more recent paper 151 challenged this ‘conventional wisdom’<br />

model, and maintained that peripheral blood stem<br />

cells per se are capable of rapidly maturing in vivo.<br />

Other authors, using a very similar approach, reached<br />

an opposing conclusion. 152 In humans the most convincing,<br />

albeit indirect, evidence in favor of the role of<br />

committed progenitors in accelerating post-transplant<br />

recovery was provided by Robertson et al., 153 who documented<br />

a significantly prolonged hematopoietic recovery<br />

for patients undergoing autologous bone marrow<br />

transplantation purged ex vivo with anti-CD33 monoclonal<br />

antibodies. This result, which occurred after a<br />

treatment that selectively kills the most mature progenitor<br />

cells (expressing the CD33 surface antigen), represents<br />

convincing indirect proof of the role of committed<br />

progenitors in early engraftment.<br />

The clinical role of committed progenitors in reducing<br />

the morbidity and mortality of high-dose therapy<br />

has expanded since hemopoietic growth factors have<br />

become clinically available. In fact, infusion of rhGM-<br />

CSF or rhG-CSF, in particular after administration of<br />

myelotoxic doses of certain stem cell-sparing agents,<br />

allows easy collection of an amount of CFU-GM/kg body<br />

weight 10 to 100 times higher than that contained in a<br />

bone marrow harvest. 110<br />

As already documented by a large number of papers,<br />

the use of circulating progenitors has brought about a<br />

dramatic change in the perspectives of high-dose submyeloablative<br />

regimens. In fact, these once specialized,<br />

expensive and highly toxic treatments are now well tolerated,<br />

easy to administer, and clinically useful (cost<br />

effective). As an example, it is worth mentioning the<br />

initial Milan Cancer Institute experience in a group of<br />

over 50 poor-risk breast cancer patients who received<br />

high-dose melphalan plus an optimal amount of circulating<br />

progenitors (≥ 5×10 4 CFU-GM/kg body weight).<br />

These patients required a median of 10.5, 11 and 12.5<br />

days to score > 0.5, 1.0 and 2.0×10 9 /L neutrophils/µL,<br />

respectively. Moreover, more than half of them did not<br />

require platelet transfusions, while the remaining ones<br />

needed only one or two transfusions during the first<br />

week after autografting. Such mild to moderate toxicity<br />

was never described before the clinical availability of<br />

committed progenitor cells.<br />

In conclusion, born as a compassionate surrogate of<br />

bone marrow autografting, today CPAT is rapidly replacing<br />

ABMT. In fact, today it is the latter that should be<br />

considered a compassionate need procedure, useful in<br />

those few patients unable to mobilize a sufficient number<br />

of circulating progenitor cells.<br />

The second, distinct role of circulating progenitors is<br />

related to the presence of stem cells, i.e. totipotent precursors<br />

responsible for durable reconstitution of all lympho-hematopoietic<br />

lineages following marrow ablative<br />

therapy. Since virtually no high-dose treatment that can<br />

be safely administered to humans is genuinely myeloablative,<br />

formal proof of the existence of circulating<br />

stem cells must await either stable transduction of DNA<br />

markers into autografted cells or the use of this cell<br />

population for allografting.<br />

These experiments are presently underway in several<br />

laboratories, 154,155 and preliminary data do confirm the<br />

presence of stem cells in the peripheral blood of humans.<br />

Their utilization is expected to revolutionize fields like<br />

allogeneic bone marrow transplantation and somatic<br />

gene therapy, whenever the target of genetic manipulations<br />

is the hematopoietic cell. 156<br />

haematologica vol. 85(suppl. to n. 12):December 2000


14<br />

C. Carlo Stella et al.<br />

References<br />

1. Civin CI, Strauss LC, Brovall C, Fackler MJ, Schwartz<br />

JF, Shaper JH. Antigenic analysis of hematopoiesis III.<br />

A hematopoietic progenitor cell surface antigen<br />

defined by a monoclonal antibody raised against KG-<br />

1a cells. J Immunol 1984; 133:157-65.<br />

2. Sutherland RD, Keating A. The CD34 antigen: structure,<br />

biology and potential clinical applications. J<br />

Hematother 1992; 1:115-29.<br />

3. Andrews RG, Singer JW, Bernstein ID. Precursors of<br />

colony-forming cells in humans can be distinguished<br />

from colony-forming cells by expression of the CD33<br />

and CD34 antigens and light scatter properties. J Exp<br />

Med 1989; 169:1721-31.<br />

4. Andrews RG, Singer JW, Bernstein ID. Monoclonal<br />

antibody 12.8 recognizes a 115-kd molecule present<br />

on both unipotent and multipotent hematopoietic<br />

colony-forming cells and their precursors. Blood<br />

1986; 67:842-52.<br />

5. Civin CI, Gore SD. Antigenic analysis of hematopoiesis:<br />

a review. J Hematother 1993; 2:137-44.<br />

6. Lansdorp PM, Sutherland HJ, Eaves CJ. Selective<br />

expression of CD45 isoforms on functional subpopulations<br />

of CD34+ hematopoietic cells from human<br />

bone marrow. J Exp Med 1990; 172:363-6.<br />

7. Baum CM, Weissman IL, Tsukamoto AS, Buckle AM.<br />

Isolation of a candidate human hematopoietic stemcell<br />

population. Proc Natl Acad Sci USA 1992; 89:<br />

2804-8.<br />

8. Huang S, Terstappen LWMM. Lymphoid and myeloid<br />

differentiation of single human CD34+, HLA-DR–,<br />

CD38– hematopoietic stem cells. Blood 1994; 83:<br />

1515-26.<br />

9. Gunji Y, Nakamura M, Osawa H, et al. Human primitive<br />

hematopoietic progenitor cells are more enriched<br />

in KIT low cells than in KIT high cells. Blood 1993;<br />

82:3283-9.<br />

10. Rosnet O, Marchetto S, de Lapeyriere O, Birnbaum D.<br />

Murine Flt3, a gene encoding a novel tyrosine kinase<br />

receptor of the PDGFR/CSF1R family. Oncogene<br />

1991; 6:1641-50.<br />

11. Matthews W, Jordan CT, Wiegand GW, Pardoll D,<br />

Lemischka IR. A receptor tyrosine kinase specific to<br />

hematopoietic stem and progenitor cell-enriched populations.<br />

Cell 1991; 65: 1143-52.<br />

12. Small D, Levenstein M, Kim E, et al. STK-1, the human<br />

homologue of Flk-2/Flt-3, is selectively expressed in<br />

CD34+ human bone marrow cells and is involved in<br />

the proliferation of early progenitor/stem cells. Proc<br />

Natl Acad Sci USA 1994; 91:459-63.<br />

13. Lyman SD, James L, van den Bos T, et al. Molecular<br />

cloning of a ligand for the flt3/flk-2 tyrosine kinase<br />

receptor: a proliferative factor for primitive hematopoietic<br />

cells. Cell 1993; 75: 1157-67.<br />

14. Lyman SD, James L, Johnson L, et al. Cloning of the<br />

human homologue of the murine flt3 ligand: a growth<br />

factor for early hematopoietic progenitor cells. Blood<br />

1994; 83:2795-801.<br />

15. Hannum C, Culpepper J, Campbell D, et al. Ligand for<br />

FLT3/FLK2 receptor tyrosine kinase regulates growth<br />

of haematopoietic stem cells and is encoded by variant<br />

RNAs. Nature 1994; 368:643-8.<br />

16. McCulloch EA. Stem cells in normal and leukemic<br />

hemopoiesis (Henry Stratton lecture, 1982). Blood<br />

1983; 62:1-13.<br />

17. Metcalf D. The molecular control of cell division, differentiation<br />

commitment and maturation in haemopoietic<br />

cells. Nature 1989; 339:27-30.<br />

18. Orlic D, Bodine DM. What defines a pluripotent<br />

hematopoietic stem cell (PHSC): will the real PHSC<br />

stand up! Blood 1994; 84:3919-94.<br />

19. Ogawa M. Differentiation and proliferation of hematopoietic<br />

stem cells. Blood 1993; 81:2844-53.<br />

20. Tsai FY, Keller G, Kuo FC, et al. An early haematopoietic<br />

defect in mice lacking the transcription factor<br />

GATA-2. Nature 1994; 371:221-6.<br />

21. Sutherland HJ, Eaves CJ, Eaves AJ, Dragowskas W,<br />

Lansdorp PM. Characterization and partial purification<br />

of human marrow cells capable of initiating longterm<br />

hematopoiesis in vitro. Blood 1989; 74:1563-<br />

70.<br />

22. Kmiecik TE, Keller JE, Rosen E, van de Woude GF.<br />

Hepatocyte growth factor is a synergistic factor for<br />

the growth of hematopoietic progenitor cells. Blood<br />

1992; 80:2454-7.<br />

23. Metcalf D. Hematopoietic regulators: redundancy or<br />

subtlety? Blood 1993; 82:3515-23.<br />

24. Berenson RJ, Andrews RG, Bensinger WI, et al. CD34+<br />

marrow cells engraft lethally irradiated baboons. J Clin<br />

Invest 1988; 81:951-9.<br />

25. Andrews RG, Bryant EM, Bartelmez SH, et al. CD34<br />

marrow cells, devoid of T and B lymphocytes, reconstitute<br />

stable lymphopoiesis and myelopoiesis in<br />

lethally irradiated allogeneic baboons. Blood 1992;<br />

80:1693-9.<br />

26. Srour EF, Zanjani ED, Cornetta K, et al. Persistence of<br />

human multilineage, self-renewing lymphohematopoietic<br />

stem cells in chimeric sheep. Blood 1993;<br />

82:3333-42.<br />

27. Berenson R. Transplantation of hematopoietic stem<br />

cells. J Hematother 1993; 2:347-9.<br />

28. Greaves MF, Brown J, Molgaard HV, et al. Molecular<br />

features of CD34: a hemopoietic progenitor cell-associated<br />

molecule. Leukemia 1992; 6:31-6.<br />

29. Silvestri F, Banavali S, Baccarani M, Preisler HD. Progenitor<br />

cell associated antigen CD34: biology and clinical<br />

applications. <strong>Haematologica</strong> 1992; 77:265-72.<br />

30. Peschle CH, Köller U. Cluster report: CD34. In: Knapp<br />

W, Dörken B, Gilks WR, Rieber EP, Schmidt RE, Stein<br />

H, von dem Borne AEGKr, eds. Leukocyte typing IV:<br />

white cell differentiation antigens. Oxford:Oxford University<br />

Press, 1989: 817-8.<br />

31. Civin CI, Trishmann TM, Fackler MJ, et al. Report on<br />

the CD34 cluster workshop. In: Knapp W, Dörken B,<br />

Gilks WR, Rieber EP, Schmidt RE, Stein H, von dem<br />

Borne AEGKr, eds. Leukocyte typing IV: white cell differentiation<br />

antigens. Oxford:Oxford University Press,<br />

1989: 818-25.<br />

32. Lanza F, Moretti S, Papa S, Malavasi F, Castoldi G.<br />

Report on the Fifth international Workshop on<br />

Human Leukocyte Differentiation Antigens, Boston,<br />

November 3-7, 1993. <strong>Haematologica</strong> 1994; 79:374-<br />

86.<br />

33. Greaves MF, Colman SM, Bühring HJ, et al. Report on<br />

the CD34 cluster workshop. In: Schlossman SF,<br />

Boumsell L, Gilks W, et al, eds. Leukocyte typing V:<br />

White cell differentiation antigens. Oxford:Oxford<br />

University Press, 1995, in press.<br />

34. Sutherland DR, Marsh JCW, Davidson J, Baker MA,<br />

Keating A, Mellors A. Differential sensitivity of CD34<br />

epitopes to cleavage by Pasteurella haemolytica glycoprotease:<br />

implications for purification of CD34-<br />

positive progenitor cells. Exp Hematol 1992; 20:590-<br />

9.<br />

35. Lanza F, Castoldi GL. Large scale enrichment of<br />

CD34+ cells by Percoll density gradients: a CML-based<br />

study design. In: Wunder E, Serke S, Solovat H, Henon<br />

P, eds. Hematopoietic stem cells. Dayton:AlphaMed<br />

Press, 1993:255-70.<br />

36. Lanza F, Bi S, Castoldi GL, Goldman JM. Abnormal<br />

expression of N-CAM (CD56) adhesion molecule on<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 15<br />

myeloid and progenitor cells from chronic myeloid<br />

leukemia. Leukemia 1993; 7:1570-5.<br />

37. Lanza F, Rigolin GM, Moretti S, Latorraca A, Castoldi<br />

GL. Prognostic value of immunophenotypic characteristic<br />

of blasts cells in acute myeloid leukemia.<br />

Leuk Lymphoma 1994; 13(Suppl 1):81-5.<br />

38. Morecki S, Topalian SL, Myers WW, Okrongly D,<br />

Okarma TB, Rosenberg SA. Separation and growth<br />

of human CD4+ and CD8+ tumor infiltrating lymphocytes<br />

and peripheral blood mononuclear cells by<br />

direct positive panning on covalently attached monoclonal<br />

antibody coated-flasks. J Biol Resp Modifiers<br />

1990; 9:463-74.<br />

39. Civin CI, Strauss LC, Fackler MJ, Trismann TM, Wiley<br />

JM, Loken RL. Positive stem cell selection. Basic science.<br />

In: S Gross, A Gee, DA Worthington White, eds.<br />

Bone Marrow Purging and Processing. New York:<br />

Wiley-Liss, 1990:387-402.<br />

40. Berenson RJ, Bensinger WI, Kalamasz D. Elimination<br />

of Daudi lymphoblasts from human bone marrow<br />

using avidin-biotin immunoadsorption. Blood 1986;<br />

67:509-15.<br />

41. Berenson RJ, Bensinger WI, Hill RS, et al. Engraftment<br />

after infusion of CD34+ marrow cells in patients with<br />

breast cancer or neuroblastoma. Blood 1991; 77:<br />

1717-22.<br />

42. Carlo-Stella C, Mangoni L, Piovani G, Garau D, Almici<br />

C, Rizzoli V. Identification of Philadelphia-negative<br />

granulocyte-macrophage colony-forming units generated<br />

by stroma-adherent cells from chronic myelogenous<br />

leukemia patients. Blood 1994; 83:1373-80.<br />

43. Zanjani ED, Pallavicini MG, Ascensao JL, et al.<br />

Engraftment and long-term expression of human fetal<br />

hemopoietic stem cells in sheep following transplantation<br />

in utero. J Clin Invest 1992; 89:1178-88.<br />

44. Srour EF, Zanjani ED, Brandt JE, et al. Sustained<br />

human hematopoiesis in sheep transplanted in utero<br />

during early gestation with fractionated adult human<br />

bone marrow cells. Blood 1992; 79:1404-12.<br />

45. Berenson RJ, Bensinger WI, Kalamasz D, et al. Engraftment<br />

of dogs with Ia-positive marrow cells isolated by<br />

Avidin-Biotin immunoadsorption. Blood 1987; 69:<br />

1363-7.<br />

46. Berenson RJ, Andrews RG, Bensinger WI, et al. Antigen<br />

CD34+ marrow cells engraft lethally irradiated<br />

baboons. J Clin Invest 1988; 81:951-5.<br />

47. Gore SD, Kastan MB, Civin CI. Normal human bone<br />

marrow precursors that express terminal deoxynucleotidyl<br />

transferase include T-cell precursors and possible<br />

lymphoid stem cells. Blood 1991; 77:1681-90.<br />

48. Miller JS, Verfaillie C, McGlave P. The generation of<br />

human natural killer cells from CD34+/DR– primitive<br />

progenitors in long term bone marrow culture. Blood<br />

1992; 80:2182-7.<br />

49. Loken MR, Civin CI, Bighee WL, Langlois RG, Jensen<br />

RH. Coordinate glycosylation and cell surface expression<br />

of glycophorin A during normal human erythropoiesis.<br />

Blood 1987; 70:1959-61.<br />

50. Shaw VO, Civin CI, Loken MR. Flow cytometric analysis<br />

of human bone marrow. IV. Differential quantitative<br />

expression of T-200 common leukocyte antigen<br />

during normal hematopoiesis. J Immunol 1988;<br />

140:1861-7.<br />

51. Loken MR, Shaw VO, Dattilio K, Civin CI. Flow cytometric<br />

analysis of human bone marrow. I. Normal erythroid<br />

development. Blood 1987; 69:255-63.<br />

52. Sawada K, Krantz SB, Kans JS, et al. Purification of<br />

human colony-forming units-erythroid and demonstration<br />

of specific binding of erythropoietin. J Clin<br />

Invest 1987; 80:357-66.<br />

53. Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau<br />

J. GM-CSF and TNF-α cooperate in the generation<br />

of dendritic Langherans cells. Nature 1992;<br />

360:258-61.<br />

54. Terstappen LWMM, Huang S, Safford M, Lansdorp P,<br />

Loken MR. Sequential generation of hematopoietic<br />

colonies derived from single nonlineage-committed<br />

CD34+/CD38– progenitor cells. Blood 1991; 77:<br />

1218-27.<br />

55. De Fabritiis P, Dowding C, Bungey J, et al. Phenotypic<br />

characterization of normal and CML CD34-positive<br />

cells: only the most primitive CML progenitors<br />

include Ph-neg cells. Leuk Lymphoma 1993; 11:51-<br />

61.<br />

56. Verfaillie C, Miller WJ, Boylan K, McGlave PB. Selection<br />

of benign primitive hematopoietic progenitors in<br />

chronic myelogenous leukemia on the basis of HLA-<br />

DR antigen expression. Blood 1992; 79:1003-10.<br />

57. Beschoner WE, Civin CI, Strauss LC. Localization of<br />

hemopoietic progenitor cells in tissue with the anti-<br />

My-10 monoclonal antibody. Am J Pathol 1985;<br />

119:1-6.<br />

58. Fina L, Molgaard HV, Robertson D, et al. Expression<br />

of the CD34 gene in vascular endothelial cells. Blood<br />

1990; 75: 2417-26.<br />

59. Delia D, Lampugnani MG, Resnati M, et al. CD34<br />

expression is regulated reciprocally with adhesion molecules<br />

in vascular endothelial cells in vitro. Blood<br />

1993; 81:1001-8.<br />

60. Majdic O, Johannes Stockl, Pickl WF, et al. Signaling<br />

and induction of enhanced cytoadhesiveness via the<br />

hematopoietic progenitor cell surface molecule CD34.<br />

Blood 1994; 83: 1226-34.<br />

61. Borowitz MJ, Gockerman JP, Moore JO, et al. Clinicopathologic<br />

and cytogenetic features of CD34 (MY10)-<br />

positive acute nonlymphocytic leukemia. Am J Clin<br />

Pathol 1989; 91:265-70.<br />

62. Campos L, Guyotat D, Archimbaud E, et al. Surface<br />

marker expression in adults with acute myelocytic<br />

leukemia: correlations with initial characteristics, morphology<br />

and response to therapy. Br J Haematol<br />

1989; 72:161-6.<br />

63. Geller RB, Zahurak M, Hurwitz CA, et al. Prognostic<br />

importance of immunophenotyping in adults with<br />

acute myelocytic leukaemia: the significance of the<br />

stem-cell glycoprotein CD34 (My10). Br J Haematol<br />

1990; 76:340-7.<br />

64. Myint H, Lucie NP. The prognostic significance of the<br />

CD34 antigen in acute myeloid leukaemia. Leuk Lymphoma<br />

1992; 7:425-9.<br />

65. Lauria F, Raspadori D, Ventura MA, et al. CD7 expression<br />

does not affect the prognosis in acute myeloid<br />

leukemia. Blood 1994; 83:3097-8.<br />

66. Borowitz Mj, Shuster JJ, Civin CI, et al. Prognostic significance<br />

of CD34 expression in childhood B-precursor<br />

acute lymphocytic leukemia: a Pediatric Oncology<br />

Group Study. J Clin Oncol 1990; 8:1389-96.<br />

67. Pui CH, Hancock ML, Head DR, et al. Clinical significance<br />

of CD34 expression in childhood acute lymphoblastic<br />

leukemia. Blood 1993; 82:889-98.<br />

68. Till JE, McCulloch EA. Hemopoietic stem cell differentiation.<br />

Biochim Biophys Acta 1980; 605:431-59.<br />

69. Van Zant G, Chen JJ, Scott-Micus K. Developmental<br />

potential of hematopoietic stem cells determined<br />

using retrovirally marked allogenic marrow. Blood<br />

1991; 77:756-63.<br />

70. Till JE, McCulloch EA, Siminovitc L. A stochastic model<br />

of stem cell proliferation, based on the growth of<br />

spleen colony-forming cells. Proc Natl Acad Sci USA<br />

1964; 51:29-34.<br />

71. Ogawa M. Differentiation and proliferation of<br />

hematopoietic stem cells. Blood 1993; 81:2844-53.<br />

72. Bergamaschi G, Rosti V, Danova M, Lucotti C, Cazzola<br />

M. Apoptosis: biological and clinical aspects.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


16<br />

C. Carlo Stella et al.<br />

<strong>Haematologica</strong> 1994; 79:86-93.<br />

73. Barosi G. Control of erythropoietin production in<br />

man. <strong>Haematologica</strong> 1993; 78:77-9.<br />

74. Cazzola M. The end of a long search: at last thrombopoietin.<br />

<strong>Haematologica</strong> 1994; 79:397-9.<br />

75. Ikebuchi K, Clark SC, Ihle JN, Souza LM, Ogawa M.<br />

Granulocyte colony-stimulating factor enhances interleukin-3-dependent<br />

proliferation of multipotential<br />

hemopoietic progenitors. Proc Natl Acad Sci USA<br />

1988; 85:3445-9.<br />

76. Sonoda Y, Yang YC, Wong GG, Clark SC, Ogawa M.<br />

Analysis in serum-free culture of the targets of recombinant<br />

human hemopoietic factors: interleukin-3 and<br />

granulocyte/macrophage colony-stimulating factor<br />

are specific for early developmental stages. Proc Natl<br />

Acad Sci USA 1988; 85: 4360-6.<br />

77. Leary AG, Zeng HQ, Clark SC, Ogawa M. Growth factor<br />

requirements for survival in G0 and entry into the<br />

cell cycle of primitive human hematopoietic progenitors.<br />

Proc Natl Acad Sci USA 1992; 89:4013-7.<br />

78. Moore MAS. Clinical implications of positive and negative<br />

hematopoietic stem cell regulators. Blood 1991;<br />

78:1-19.<br />

79. Hyrayama F, Shih JP, Awgulewitsch A, Warr GW,<br />

Clark SC, Ogawa M. Clonal proliferation of murine<br />

lymphohematopoietic progenitors in culture. Proc<br />

Natl Acad Sci USA 1992; 89: 5907-11.<br />

80. Gordon MY, Riley GP, Watt SM, Greaves MF.<br />

Compartimentalization of a haematopoietic growth<br />

factor (GM-CSF) by glycosaminoglycans in the bone<br />

marrow microenvironment. Nature 1987; 326:403-5.<br />

81. Montagna C, Massaro P, Morali F, Foa P, Maiolo AT,<br />

Eridani S. In vitro sensitivity of human erythroid progenitors<br />

to hemopoietic growth factors: studies on primary<br />

and secondary polycythemia. <strong>Haematologica</strong><br />

1994; 79:311-8.<br />

82. Jacobsen SEW, Ruscetti FW, Dubois CM, Wine J,<br />

Keller JR. Induction of colony-stimulating factor receptor<br />

expression on hematopoietic progenitor cells: Proposed<br />

mechanism for growth factor synergism. Blood<br />

1992; 80:678-87.<br />

83. Testa U, Pelosi E, Gabbianelli M, et al. Cascade of<br />

transactivation of growth factor receptors in early<br />

human hematopoiesis. Blood 1993; 81:1442-56.<br />

84. Hirano T, Yasukawa K, Harada H, et al. Complementary<br />

DNA for a novel human interleukin (BSF-2) that<br />

induces B lymphocytes to produce immunoglobulin.<br />

Nature 1986; 324: 73-6.<br />

85. Kitamura T, Sato N, Arai K, Miyajima A. Expression<br />

cloning of the human IL-3 receptor cDNA reveals a<br />

shared b subunit for the human IL-3 and GM-CSF<br />

receptors. Cell 1991; 66: 1165-9.<br />

86. Yin T, Taga T, Lik-Shing Tsang M, Yasukawa K, Kishimoto<br />

T, Yang YC. Involvement of IL-6 signal transducer<br />

gp130 in IL-11-mediated signal transduction. J<br />

Immunol 1993; 151: 2555-61.<br />

87. Massague J. The transforming growth factor-family.<br />

Ann Rev Cell Biol 1990; 6:597-641.<br />

88. Ploemacher RE, van Soest PL, Boudewijn A. Autocrine<br />

transforming growth factor 1 blocks colony formation<br />

and progenitor cell generation by hemopoietic<br />

stem cells stimulated with steel factor. Stem Cells<br />

1993; 11:336-47.<br />

89. Jacobsen SEW, Keller JR, Ruscetti FW, Kondaiah P,<br />

Roberts AB, Falk LA. Bidirectional effects of transforming<br />

growth factor β (TGF-β) on colony-stimulating<br />

factor-induced human myelopoiesis in vitro: differential<br />

effects of distinct TGF- isoforms. Blood 1991;<br />

78:2239-47.<br />

90. Keller JR, Jacobsen SEW, Dubois CM, Hestdal K,<br />

Ruscetti FW. Transforming growth factor-α: a bidirectional<br />

regulator of hematopoietic cell growth. Int J<br />

Cell Cloning 1992; 10:2-11.<br />

91. Lemoli RM, Fogli M, Fortuna A, Tura S. Interleukin-11<br />

(IL-11) and IL-9 counteract the inhibitory activity of<br />

transforming growth factor β3 (TGF β3) on human<br />

primitive hemopoietic progenitor cells. <strong>Haematologica</strong><br />

1995; 80:5-12.<br />

92. Jacobsen SEW, Ruscetti FW, Dubois CM, Lee J, Boone<br />

TC, Keller JR. Transforming growth factor-β transmodulates<br />

the expression of colony stimulating factor<br />

receptors on murine hematopoietic progenitor cell<br />

lines. Blood 1991; 77:1706-16.<br />

93. Laiho M, Di Caprio JA, Ludlow JW, Livingstone DM,<br />

Massague J. Growth inhibition by TGF-β linked to suppression<br />

of retinoblastoma protein phosphorylation.<br />

Cell 1990; 62: 175-85.<br />

94. Takehara K, Le Roy EC, Grotendorst GR. TGF-β inhibition<br />

of endothelial cell proliferation: alteration of<br />

EGF binding and EGF-induced growth-regulatory<br />

(competence) gene expression. Cell 1987; 49:415-22.<br />

95. Strife A, Lambek C, Perez A, et al. The effects of TGFβ3<br />

on the growth of highly enriched hematopoietic<br />

progenitor cells derived from normal human bone<br />

marrow and peripheral blood. Cancer Res 1991; 15:<br />

4828-36.<br />

96. Lemoli RM, Strife A, Clarkson BD, Haley JD, Gulati<br />

SC. TGF-β3 protects normal human hematopoietic<br />

progenitor cells treated with 4-hydroperoxycyclophosphamide<br />

in vitro. Exp Hematol 1992; 20: 1252-<br />

6.<br />

97. Dubois CM, Ruscetti FW, Stankowa J, Keller JR. Transforming<br />

growth factor-β regulates c-kit message stability<br />

and cell-surface protein expression in hematopoietic<br />

progenitors. Blood 1994; 83:3138-45.<br />

98. Caux C, Saeland S, Favre C, Duvert V, Mannoni P,<br />

Bancherau J. Tumor necrosis factor-a strongly potentiates<br />

interleukin-3 and granulocyte-macrophage<br />

colony-stimulating factor-induced proliferation of<br />

human CD34+ hematopoietic progenitor cells. Blood<br />

1990; 75:2292-8.<br />

99. Schall TJ, Lewis M, Koller KJ, et al. Molecular cloning<br />

and expression of a receptor for human tumor necrosis<br />

factor. Cell 1990; 61:361-4.<br />

100. Rusten LS, Jacobsen FW, Lesslauer W, Loetscher H,<br />

Smeland EB, Jacobsen SEW. Bifunctional effects of<br />

tumor necrosis factor-α (TNF-α) on the growth of<br />

mature and primitive human hematopoietic progenitor<br />

cells: involvement of p55 and p75 TNF receptors.<br />

Blood 1994; 83:3152-9.<br />

101. Graham GJ, Wright EG, Hewick R, et al. Identification<br />

and characterization of an inhibitor of hematopoietic<br />

stem cell proliferation. Nature 1990; 344:442-<br />

4.<br />

102. Broxmeyer HE, Sherry B, Lu L, et al. Enhancing and<br />

suppressing effects of recombinant murine macrophage<br />

inflammatory proteins on colony formation in<br />

vitro by bone marrow myeloid progenitor cells. Blood<br />

1992; 76:1110-6.<br />

103. Dunlop DJ, Wright EG, Lorimore S, et al. Demonstration<br />

of stem cell inhibition and myeloprotective<br />

effects of SCI/MIP1α in vivo. Blood 1992; 79:2221-5.<br />

104. Moreb J, Zucali JR, Rueth S. The effects of tumor<br />

necrosis factor-α on early human hematopoietic progenitor<br />

cells treated with 4-hydroperoxycyclophosphamide.<br />

Blood 1990; 76:681-9.<br />

105. Andrews RG, Singer JW, Bernstein ID. Precursors of<br />

colony forming cells in human can be distinguished<br />

from colony forming cells by expression of the CD33<br />

and CD34 antigens and light scatter properties. J Exp<br />

Med 1989; 169:1721-31.<br />

106. Gabbianelli M, Sargiacomo M, Pelosi E, Testa U, Isacchi<br />

G, Peschle C. Pure human hematopoietic progenitors:<br />

permissive action of basic fibroblast growth fac-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


CD34 + cells: biology and clinical relevance 17<br />

tor. Science 1990; 249:1561-4.<br />

107. Peters W, Rosner G, Ross M, et al. Comparative<br />

effects of granulocyte-macrophage colony-stimulating<br />

factor (GM-CSF) and granulocyte colony-stimulating<br />

factor (G-CSF) on priming peripheral blood<br />

progenitor cells for use with autologous bone marrow<br />

transplantation after high-dose chemotherapy.<br />

Blood 1993; 81:1709-19.<br />

108. Duhrsen U, Villeval JL, Boyd J, Kannourakis G,<br />

Morstyn G, Metcalf D. Effects of recombinant human<br />

granulocyte colony-stimulating factor on hematopoietic<br />

progenitor cells in cancer patients. Blood 1988;<br />

72:2074-81.<br />

109. Socinski MA, Elias A, Schnipper L, Cannistra SA,<br />

Antman KH, Griffin JD. Granulocyte-macrophage<br />

colony-stimulating factor expands the circulating<br />

haemopoietic progenitor cell compartment in man.<br />

Lancet 1988; i:1194-8.<br />

110. Gianni AM, Siena S, Bregni M, et al. Granulocytemacrophage<br />

colony-stimulating factor to harvest circulating<br />

haematopoietic stem cells for autotransplantation.<br />

Lancet 1989; ii:580-5.<br />

111. Kotasek D, Shepherd KM, Sage RE, et al. Factors<br />

affecting blood stem cell collections following highdose<br />

cyclophosphamide mobilization in lymphoma,<br />

myeloma and solid tumors. Bone Marrow Transplant<br />

1992; 9:11-7.<br />

112. Teshima T, Harada M, Takamatsu Y, et al. Cytotoxic<br />

drug and cytotoxic drug/G-CSF mobilization of<br />

peripheral blood stem cells and their use for autografting.<br />

Bone Marrow Transplant 1992; 10:215-20.<br />

113. Siena S, Bregni M, Brando B, Ravagnani F, Bonadonna<br />

G, Gianni AM. Circulation of CD34-positive<br />

hematopoietic stem cells in the peripheral blood of<br />

high-dose cyclophosphamide treated patients:<br />

enhancement by intravenous recombinant human<br />

GM-CSF. Blood 1989; 74:1905-14.<br />

114. Ravagnani F, Siena S, Bregni M, Sciorelli G, Gianni<br />

AM, Pellegris G. Large scale collection of circulating<br />

haematopoietic progenitors in cancer patients treated<br />

with high-dose cyclophosphamide and recombinant<br />

human GM-CSF. Eur J Cancer 1990; 26:562-4.<br />

115. Kawano Y, Takaue Y, Watanabe T, et al. Effects of<br />

progenitor cell dose and preleukapheresis use of<br />

human recombinant granulocyte colony-stimulating<br />

factor on the recovery of hematopoiesis after blood<br />

stem cell autografting in children. Exp Hematol 1993;<br />

21:103-8.<br />

116. Bensinger W, Singer J, Appelbaum F, et al. Autologous<br />

transplantation with peripheral blood mononuclear<br />

cells collected after administration of recombinant<br />

granulocyte stimulating factor. Blood 1993; 81:<br />

3158-63.<br />

117. Haas R, Mohle R, Fruhauf S, et al. Patient characteristics<br />

associated with successful mobilizing and autografting<br />

of peripheral blood progenitor cells in malignant<br />

lymphoma. Blood 1994; 83:3787-94.<br />

118. To LB, Dyson PG, Juttner CA. Cell-dose effect in circulating<br />

stem-cell autografting. Lancet 1986; ii:404-5.<br />

119. Gianni AM, Tarella C, Siena S, et al. Durable and complete<br />

hematopoietic reconstitution after autografting<br />

of rhGM-CSF exposed peripheral blood progenitor<br />

cells. Bone Marrow Transplant 1991; 6:143-5.<br />

120. Hohaus S, Goldschmidt H, Ehrhardt R, Haas R. Successful<br />

autografting following myeloablative conditioning<br />

therapy with blood stem cells mobilized by<br />

chemotherapy plus rhG-CSF. Exp Hematol 1993; 21:<br />

508-14.<br />

121. Siena S, Bregni M, Di Nicola M, et al. Durability of<br />

hematopoiesis following myeloablative cancer tharapy<br />

and autografting with peripheral blood hematopoietic<br />

progenitors. Ann Oncol 1994; 5:935-41.<br />

122. Tarella C, Boccadoro M, Omedè P, et al. Role of<br />

chemotherapy and GM-CSF on hemopoietic progenitor<br />

cell mobilization in multiple myeloma. Bone Marrow<br />

Transplant 1993; 11: 271-7.<br />

123. Tarella C, Ferrero D, Siena S, et al. Conditions influencing<br />

the expansion of the circulating hemopoietic<br />

progenitor cell compartment. <strong>Haematologica</strong> 1990;<br />

75:11-4.<br />

124. Gianni AM, Bregni M, Siena S, et al. Granulocytemacrophage<br />

colony stimulating factor or granulocytecolony<br />

stimulating factor infusion makes high-dose<br />

etoposide a safe outpatient regimen that is effective in<br />

lymphoma and myeloma patients. J Clin Oncol 1992;<br />

10:1955-62.<br />

125. Dreyfus F, Leblond V, Belanger C, et al. Peripheral<br />

blood stem cell collection and autografting in high<br />

risk lymphoma. Bone Marrow Transplant 1992; 10:<br />

409-13.<br />

126. Shimazaki C, Oku N, Ashihara E, et al. Collection of<br />

peripheral blood stem cells mobilized by high-dose<br />

Ara-C plus VP-16 or aclarubicin followed by recombinant<br />

human granulocyte-colony stimulating factor.<br />

Bone Marrow Transplant 1992; 10:341-6.<br />

127. Pettengell R, Testa NG, Swindell R, Crowther D, Dexter<br />

TM. Transplantation potential of hematopoietic<br />

cell released into the circulation during routine<br />

chemotherapy for non-Hodgkin’s lymphoma. Blood<br />

1993; 82:2239-48.<br />

128. Tarella C, Ferrero D, Bregni M, et al. Peripheral blood<br />

expansion of early progenitor cells after high-dose<br />

cyclophosphamide and rhGM-CSF. Eur J Cancer<br />

1991; 27:22-7.<br />

129. Bender JG, Lum L, Unverzagt KL, et al. Correlation of<br />

colony-forming cells, long-term culture initiating cells<br />

and CD34+ cells in apheresis products from patients<br />

mobilized for peripharal blood progenitors with different<br />

regimens. Bone Marrow Transplant 1994;<br />

13:479-85.<br />

130. Haas R, Ho AD, Bredthauer U, et al. Successful autologous<br />

transplantation of blood stem cells mobilized<br />

with recombinant human granulocyte-macrophage<br />

colony-stimulating factor. Exp Hematol 1990; 18:94-<br />

8.<br />

131. Sheridan WP, Begley CG, Juttner C, et al. Effect of<br />

peripheral blood progenitor cells mobilized by filgrastim<br />

(G-CSF) on platelet recovery after high-dose<br />

chemotherapy. Lancet 1992; i:640-4.<br />

132. Weaver CH, Buckner CD, Longin K, et al. Syngeneic<br />

transplantation with peripheral blood mononuclear<br />

cells collected after the administration of recombinant<br />

human granulocyte colony-stimulating factor.<br />

Blood 1993; 82:1981-4.<br />

133. Matsunaga T, Sakamaki S, Ohi S, Hirayama Y, Niitsu<br />

Y. Recombinant human granulocyte colony-stimulating<br />

factor can mobilize sufficient amounts of peripheral<br />

blood stem cells in healthy volunteers for allogeneic<br />

transplantation. Bone Marrow Transplant<br />

1993; 11:103-8.<br />

134. Schwinger W, Mache Ch, Urban Ch, Beaufort F, Toglhofer<br />

W. Single dose of filgrastim (rhG-CSF) increases<br />

the number of hematopoietic progenitors in the<br />

peripheral blood of adult volunteers. Bone Marrow<br />

Transplant 1993; 11:489-92.<br />

135. Brugger W, Bross K, Frisch J, et al. Mobilization of<br />

peripheral blood progenitor cells by sequential administration<br />

of interleukin-3 and granulocyte-macrophage<br />

colony-stimulating factor following polychemotherapy<br />

with etoposide, ifosfamide and cisplatin. Blood<br />

1992; 79:1193-200.<br />

136. D’Hondt V, Guillaume T, Humblet Y, et al. Tolerance<br />

of sequential or simultaneous administration of IL-3<br />

and G-CSF in improving peripheral blood stem cell<br />

haematologica vol. 85(suppl. to n. 12):December 2000


18<br />

C. Carlo Stella et al.<br />

harvesting following multi-agent chemotherapy: a<br />

pilot study. Bone Marrow Transplant 1994; 13:261-<br />

4.<br />

137. de Revel T, Appelbaum FR, Storb R, et al. Effects of<br />

granulocyte colony-stimulating factor and stem cell<br />

factor, alone and in combination, on the mobilization<br />

of peripheral blood cells that engraft lethally irradiated<br />

dogs. Blood 1994; 83:3795-9.<br />

138. Liu KY, Akashi K, Harada M, Takamatsu Y, Niho Y.<br />

Kinetics of circulating haematopoietic progenitors<br />

during chemotherapy-induced mobilization with or<br />

without granulocyte colony-stimulating factor. Br J<br />

Haematol 1993; 84:31-8.<br />

139. Bishop MR, Anderson JR, Jackson JD, et al. High-dose<br />

therapy and peripheral blood progenitor cell transplantation:<br />

effects of recombinant human granulocyte-macrophage<br />

colony-stimulating factor on the<br />

autograft. Blood 1994; 83: 610-6.<br />

140. Jansen WE. Peripheral blood and bone marrow<br />

hematopoietic stem cells: are they the same? Semin<br />

Oncol 1993; 20:19-27.<br />

141. Kessinger A, Armitage JO. The evolving role of autologous<br />

peripheral stem cell transplantation following<br />

high-dose therapy for malignancies. Blood 1991;<br />

77:211-3.<br />

142. Siena S, Bregni M, Brando B, et al. Flow cytometry for<br />

clinical estimation of circulating hematopoietic progenitors<br />

for autologous transplantation in cancer<br />

patients. Blood 1992; 77:400-9.<br />

143. Menichella G, Pierelli L, Foddai ML, et al. Autologous<br />

blood stem cell harvesting and transplantation in<br />

patients with advanced ovarian cancer. Br J Haematol<br />

1991; 79:444-50.<br />

144. Pettengell R, Morgenstern GR, Woll PJ, et al. Peripheral<br />

blood progenitor cell transplantation in lymphoma<br />

and leukemia using a single apheresis. Blood<br />

1993; 82:3770-7.<br />

145. Caracciolo D, Gavarotti P, Aglietta M, et al. Highdose<br />

sequential (HDS) chemotherapy with blood and<br />

marrow cell autograft as salvage treatment in very<br />

poor prognosis, relapsed non-Hodgkin lymphoma.<br />

Bone Marrow Transplant 1993; 12:621-5.<br />

146. Mayani H, Dragowska W, Lansdorp PM. Cytokineinduced<br />

selective expansion and maturation of erythroid<br />

versus myeloid progenitors from purified cord<br />

blood precursor cells. Blood 1993; 81:3252-8.<br />

147. Traycott CM, Abboud MR, Laver J, Clapp DW, Srour<br />

EF. Rapid exit from G0-G1 phases of cell cycle in<br />

response to stem cell factor confers on umbilical cord<br />

blood CD34+ cells an enhanced ex vivo expansion<br />

potential. Exp Hematol 1994; 22:1264-72.<br />

148. Molineux G, Pojda Z, Hampson IN, Lord BI, Dexter<br />

TM. Transplantation potential of peripheral blood<br />

stem cells induced by granulocyte colony-stimulating<br />

factor. Blood 1990; 76:2153-8.<br />

149. Gianni AM, Bregni M, Siena S, et al. Rapid and complete<br />

hemopoietic reconstitution following combined<br />

transplantation of autologous blood and bone marrow<br />

cells. A changing role for high dose chemo-radiotherapy?<br />

Hematol Oncol 1989; 7:139-48.<br />

150. Jones RJ, Wagner JE, Celano P, Zicha MS, Sharkis SJ.<br />

Separation of pluripotent hematopoietic stem cells<br />

from spleen colony forming cells. Nature 1990;<br />

347:188-9.<br />

151. Uchida N, Aguila HL, Fleming WH, Jerabek L, Weissman<br />

IL. Rapid and sustained hematopoietic recovery<br />

in lethally irradiated mice transplanted with purified<br />

Thy-1.1loLin-Sca-1+ hematopoietic stem cells. Blood<br />

1994; 83:3758-79.<br />

152. Bradford G, Williams N, Barber L, Bertoncello I. Temporal<br />

thrombocytopenia after engraftment with<br />

defined stem cells with long-term marrow reconstitution<br />

activity. Exp Hematol 1993; 21:1615-20.<br />

153. Robertson MJ, Soiffer RJ, Freedman AS, et al. Human<br />

bone marrow depleted of CD33-positive cells mediates<br />

delayed but durable reconstitution of hematopoiesis:<br />

clinical trial of MY9 monoclonal antibodypurged<br />

autografts for the treatment of acute myeloid<br />

leukemia. Blood 1992; 79:2229-36.<br />

154. Dreger P, Suttorp M, Haferlach T, et al. Allogeneic<br />

granulocyte colony-stimulating factor-mobilized peripheral<br />

blood progenitor cells for treatment of engraftment<br />

failure after bone marrow transplantation.<br />

Blood 1993; 81:401-9.<br />

155. Brenner MK, Rill DR, Moen RC, et al. Gene marking<br />

to trace origin of relapse after autologous bone-marrow<br />

transplantation. Lancet 1993; i: 85-6.<br />

156. Bregni M, Magni M, Siena S, Di Nicola M, Bonadonna<br />

G, Gianni AM. Human peripheral blood<br />

hematopoietic progenitors are optimal targets of<br />

retroviral mediated gene transfer. Blood 1992; 80:<br />

1418-22.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


eview<br />

Peripheral blood stem cells in acute<br />

myeloid leukemia: biology and clinical<br />

applications<br />

haematologica 2000; 85(supplement to no. 12):19-31<br />

MASSIMO AGLIETTA, ARMANDO DE VINCENTIIS, LUIGI LANATA,<br />

FRANCESCO LANZA, ROBERTO M. LEMOLI, GIACOMO MENICHELLA,<br />

AGOSTINO TAFURI, PAOLA ZANON, SANTE TURA<br />

Clinica Medica, Università di Torino, Novara; Dompé Biotec<br />

SpA, Milano; Amgen Italia SpA; Cattedra di Ematologia, Università<br />

di Ferrara, Ferrara; Istituto di Ematologia Seràgnoli,<br />

Università di Bologna, Bologna; Servizio di Ematologia ed<br />

Emotrasfusione, Università Cattolica del Sacro Cuore, Roma;<br />

and Cattedra di Ematologia, Università La Sapienza, Roma<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia L. e A.<br />

Seràgnoli, Policlinico S. Orsola, via Massarenti 9, 40138 Bologna,<br />

Italy.<br />

Received September 22, 1995; accepted November 27, 1995.<br />

Ackowledgments: preparation of this manuscript was supported by a<br />

grant from Dompé Biotec SpA and Amgen Italia SpA, Milan, Italy.<br />

Clinical application of circulating stem cells for<br />

autologous transplantation is steadily expanding. 1<br />

It has become increasingly clear that mobilized<br />

peripheral blood progenitor cells (PBSC) induce faster<br />

hematopoietic recovery, fewer febrile days, lower transfusion<br />

requirement and shorter hospitalization than<br />

bone marrow (BM)-derived cells. 2,3 More recently, rapid<br />

and sustained engraftment has also been reported using<br />

granulocyte colony-stimulating factor (G-CSF)-mobilized<br />

allogeneic PBSC following myeloablative therapy. 4<br />

In contrast to solid tumors and many hematological<br />

malignancies, PBSC transplantation is not widely used<br />

for acute myeloblastic leukemia (AML) patients. In this<br />

setting there are still unanswered questions such as the<br />

role of autologous stem cell transplantation in postremission<br />

therapy, as well as major issues concerning<br />

PBSC mobilization and collection: the expression of<br />

CD34 antigen on leukemic stem cells as compared to<br />

their normal counterparts, the biologic significance of<br />

CD34 + AML, the response of leukemic cells to CSFs used<br />

to optimize PBSC harvest, the potential contamination<br />

of PBSC grafts by residual AML cells and the role of exvivo<br />

purging of leukemic cells.<br />

This review analyzes the most recent advances in this<br />

field, addressing clinical and biological issues relevant<br />

to the use of autologous PBSC for AML patients.<br />

Growth factor receptor expression and<br />

response of leukemic cells to human CSFs<br />

The CD34 antigen is a 105-120 KD glycoprotein<br />

expressed on the cell surface of hematopoietic progenitors<br />

and stem cells, but it is not expressed on late<br />

hematopoietic cells or on many tumor cells. 1 CD34 + cells<br />

are responsible for the self-renewal and the expansion<br />

of the large majority of AML. It has recently been shown<br />

that most of the clonogenic cells in AML derive from the<br />

CD34 + cell fraction as opposed to CD34 – cells. 5,6 Moreover,<br />

CD34 + cells co-expressing differentiation markers<br />

(CD33, CD38) have a reduced proliferative potential<br />

since in vitro they give rise to small colonies unable to<br />

originate secondary clones. This phenomenon is likely<br />

the expression of a limited self-renewal potential. 6 Lapidot<br />

et al. provided the most convincing evidence of the<br />

stem cell role of CD34 + cells in AML by showing that<br />

only the CD34 + /CD38 – cell fraction was capable of generating<br />

acute leukemia when transplanted into SCID<br />

mice. 7<br />

These observations indicate the relevance of defining<br />

the growth and receptor expression pattern of leukemic<br />

CD34 + cells, their response to CSFs as well as their<br />

kinetic status compared to their normal counterparts.<br />

Among the different cytokines involved in the regulation<br />

of hemopoiesis, a key role in the pathogenesis of<br />

the leukemic growth is probably played by stem cell<br />

factor (SCF), interleukin 3 (IL-3), granulocyte-macrophage<br />

CSF (GM-CSF) and G-CSF. 8-12<br />

SCF receptor (c-kit) is expressed by the vast majority<br />

of AML. 8,9 Both high and low affinity receptors have<br />

been demonstrated (Table 1). c-kit shares structural<br />

similarities with the receptors for M-CSF and PDGF. A<br />

linear correlation between the percentage of CD34 +<br />

cells and c-kit expression has been documented, thus<br />

indicating that CD34 + AML express high levels of c-kit.<br />

In adult patients, the presence of a high number of<br />

CD34 + cells has been shown to correlate with a bad<br />

prognosis.<br />

c-kit activation plays a foundamental role in the regulation<br />

of the early phases of CD34 + cell stimulation.<br />

The interaction of SCF with its ligand exerts a modest<br />

proliferative stimulus on immature quiescent cells and<br />

up-regulates the expression of receptors for other<br />

growth factors. While in normal hematopoiesis this triggers<br />

myeloid differentiation, in AML it may activate<br />

self-renewal and expansion of the leukemic population.<br />

11-14<br />

High affinity receptors for GM-CSF and IL-3 (Table 1)<br />

are expressed by nearly all AML, irrespective of the FAB<br />

subtype. 15,16 IL-3, GM-CSF (and IL-5) receptors consist of<br />

an α subunit (ligand specific) and a shared β subunit.<br />

While the α subunit has a low affinity for the ligand and<br />

alone is incapable of transducing the signal, the association<br />

of the two subunits gives rise to a functioning<br />

haematologica vol. 85(suppl. to n. 12):December 2000


20<br />

M. Aglietta et al.<br />

high affinity receptor which is a type I receptor devoid<br />

of endogenous tyrosine-kinase activity. β chain activation<br />

induces several tyrosine-kinases like Fyn, Lyn, Fps,<br />

Jaks, which transduce a signal common to IL-3 and GM-<br />

CSF, 17 whereas a subunit activation induces ligand specific<br />

pathways. In the majority of AML cases IL-3 and<br />

GM-CSF induce the proliferation of CD34 + , although to<br />

variable extents. Correlations have been observed<br />

between responses to different factors but no significant<br />

additive effects have been noted.<br />

Exposure of leukemic cells to GM-CSF or IL-3 in vitro<br />

can give rise to a generation of mature cells. However,<br />

the persistence of blast cells capable of secondary<br />

leukemic colony formation indicates that the differentiation<br />

potential of IL-3 and GM-CSF is negligible and<br />

that they are unable to abolish the self-renewal of the<br />

leukemic population. 18-20 SCF synergizes with IL-3 and<br />

GM-CSF in inducing large clones primarily composed of<br />

undifferentiated cells.<br />

G-CSF receptor is expressed by nearly all AML (Table<br />

1). 16,21 However, M2 and M3 AML appear to express the<br />

highest number of receptors. In vitro growth stimulation<br />

is not consistent except for M2 and M3 AML; G-CSF<br />

action is additive or synergic with that of IL-3, SCF and,<br />

to a lesser extent, with that of GM-CSF. G-CSF also<br />

induces some degree of differentiation of leukemic<br />

CD34 + cells, and the presence of the growth factor<br />

affects the formation of secondary colonies. In addition,<br />

CSF treatment seems to prevent cell death in AML. 22<br />

The in vivo use of growth factors in AML patients<br />

derives from contrasting hypotheses:<br />

a) use of growth factors before and during cytostatic<br />

treatment to induce the proliferation of quiescent<br />

leukemic progenitors. The increased proliferative rate<br />

and, possibly, the intracellular accumulation of some<br />

cytotoxic drugs (i.e. Ara-CTP) should increase the fraction<br />

of cells killed. 23,24 This approach has never been tested<br />

in randomized trials specifically addressing this issue.<br />

It seems, however, to be of modest value with G-CSF or<br />

GM-CSF. 25,26 It remains to be seen if this approach would<br />

be more useful with molecules such as SCF that are particularly<br />

active on leukemic CD34 cells;<br />

Table 1. High affinity receptors for hematopoietic growth factors in AML.<br />

Affinity, kd<br />

(range)<br />

No. of receptors<br />

per cell<br />

Reference<br />

SCF 16-158 pmol/L 200-8000 14<br />

G-CSF 36-130 pmol/L 55-1200 16<br />

GM-CSF 64-404 pmol/L 40-1263 15,16<br />

IL-3 26-467 pmol/L 21-145 15,16<br />

b) use of growth factors as differentiating agents with<br />

the aim of exhausting the self-renewal potential of the<br />

leukemic progenitors. On the basis of in vitro and preliminary<br />

(although still to be confirmed) in vivo data, G-<br />

CSF seems the most promising molecule; 27<br />

c) use of growth factors for accelerating the recovery<br />

of residual normal progenitors after induction chemotherapy.<br />

This approach has been pursued with G-CSF<br />

and GM-CSF in AML patients > 60 years of age, for<br />

whom the pancytopenia following cytotoxic treatment<br />

is particularly profound and long-lasting and carries a<br />

relevant risk of life-threatening infections. In this setting,<br />

both G-CSF and GM-CSF given after induction<br />

chemotherapy reduce the duration of neutropenia without<br />

affecting the rate of severe infections. 28,29 Moreover,<br />

G-CSF, but not GM-CSF, appears to increase the complete<br />

remission rate. Both cytokines, however, have no<br />

impact on the survival rate. Of interest, no evidence of<br />

accelerated growth of residual leukemic cells has been<br />

observed.<br />

All these data demonstrate how controversial the use<br />

of hemopoietic growth factors in the treatment of AML<br />

is, although the most recent results suggest the safety<br />

of G-CSF administration following induction-consolidation<br />

treatment. 29<br />

Stem cell kinetics in AML<br />

The hematopoietic cell renewal process is supported<br />

by a small population of bone marrow cells termed<br />

hematopoietic stem cells. They are defined as cells capable<br />

of long-term hematopoietic reconstitution and differentiation<br />

into multiple hematopoietic lineages. It is<br />

generally held that, in the steady state, the majority of<br />

normal stem cells are dormant in the cell cycle and only<br />

a few of them supply all the hematopoietic cells at a given<br />

time. More than thirty years ago, stem cell kinetic<br />

studies 30 proposed the concept of a true resting state<br />

and coined the term G0 as the state from which stem<br />

cells randomly move to the active cell cycle.<br />

Subsequent studies 31 confirmed Lajtha’s observations<br />

by showing that brief in vitro exposure of bone marrow<br />

cells to highly specific radioactive thymidine does not<br />

reduce the number of multipotential progenitors. As<br />

shown in Figure 1, most normal bone marrow CD34 +<br />

progenitor cells are indeed quiescent in G0. Culture of<br />

enriched human progenitors documented that they<br />

remain as single cells for as long as 2 weeks in culture<br />

and begin proliferation upon stimulation with combinations<br />

of cytokines. 32 Based on mathematical studies,<br />

stem cell function was seen as a model in which the<br />

decision to self-renew and differentiate followed a stochastic<br />

process. 33 By replating individual blast cell<br />

colonies, Till and coworkers showed that the production<br />

of secondary blast cell colonies is a self-renewal process<br />

and that the generation of secondary multilineage<br />

colonies is differentiation. Thus the self-renewal process<br />

is associated with renewed dormancy in the cell cycle<br />

while the differentiation process is characterized by<br />

continuous cell doubling.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Peripheral blood stem cells in acute myeloid leukemia<br />

21<br />

Similar work was performed on leukemic stem cells by<br />

several authors and two fundamental antithetical models<br />

were proposed, based on the presence of quiescent<br />

progenitor cells in human leukemia.<br />

It was postulated that leukemic progenitors were predominantly<br />

involved in rapid cell cycling as judged by<br />

measuring the proportion of S-phase cells using H3-<br />

TdR or hydroxyurea. 34 However, this was in contrast with<br />

previous observations obtained in vivo by continuous<br />

infusions of 3H-thymidine for 8-10 days. 35 In those<br />

experiments, 88 to 93% of the leukemic cells were<br />

labeled at the end of infusion, whereas almost all the<br />

smallest leukemic cells were not, suggesting that they<br />

were in an extended G0. Using in vivo pulse labeling with<br />

tritiated thymidine in AML patients, it was further<br />

shown that blasts with a high proliferative rate do not<br />

behave as a pool of normal self-maintaining cells, but<br />

rather as a normal multiplication-maturation compartment.<br />

36 Data obtained with different techniques (labeling<br />

and cell culture methods) and the heterogeneity of<br />

study cell populations may represent the reason for<br />

these discordant results. The hypothesis that AML progenitors<br />

are characterized by a substantial number of<br />

nonproliferating or very slowly proliferating blast cells<br />

(lower RNA content) 37,38 was the rationale for different<br />

approaches to AML treatment. For instance, the combined<br />

use of cytokines and chemotherapy to recruit quiescent<br />

cells into the cell cycle, enhancing the cytotoxicity<br />

of cycle-specific agents. 39,40<br />

Raymakers et al. 41 have studied the proliferative<br />

capacity of the bone marrow fraction double stained<br />

for CD34 and CD33 in AML patients. The cloning efficacy<br />

was highly variable in different AML samples, with<br />

predominant cluster growth. Cluster and colony growth<br />

was similar between CD34 – /CD33 + and CD34 + /CD33 + ,<br />

in contrast to what is observed in normal bone marrow.<br />

The most primitive CD34 + /CD33 – fraction was found in<br />

highly proliferative colony growth. When this analysis<br />

was extended to AML with a more mature phenotype<br />

(small fraction of CD34 + /33 – ), the highly proliferative<br />

colonies deriving from the CD34 + /33 – fraction were<br />

found to be disomic by in situ hybridization in all<br />

patients who were characterized by chromosomal<br />

abnormalities. Nevertheless, the authors could not<br />

exclude the presence of leukemic stem cells kinetically<br />

characterized by low or no proliferation under their<br />

experimental culture conditions.<br />

A further study 42 aimed at evaluating the specific<br />

activity of SCF on enriched CD34 + in suspension culture<br />

by measuring Ki67 expression and flow cytometric DNA<br />

content showed no difference in cell cycle distribution<br />

among progenitors obtained from normal bone marrow,<br />

umbilical cord blood and chronic myeloid leukemia<br />

CD34 + peripheral blood stem cells.<br />

Further investigations on the role of a family of proteins<br />

recently identified as cell cycle regulators, such as<br />

cyclin A, B, D, E and of their catalytic subunits, the cyclindependent<br />

kinases cdk2, cdk4, cdk6 and cdc2, may help<br />

to identify kinetic features and fine differences between<br />

normal and leukemic hemopoietic stem cells, as well as<br />

events involved in neoplastic transformation. 43<br />

In conclusion, the kinetic characteristics of leukemic<br />

stem cells have still not been defined, mainly because<br />

different experimental conditions allow evaluation of<br />

progenitors with different degrees of maturation and<br />

therefore with different proliferative characteristics. The<br />

heterogeneity among different leukemia subtypes<br />

should also be taken into account.<br />

Expression of CD34 antigen in AML and<br />

CD34 + leukemias: clinical and biological<br />

significance<br />

Based on current information, there is no doubt that<br />

a substantial number of acute leukemias express the<br />

CD34 antigen on the cell membrane of blast cells. However,<br />

the incidence of such expression in AML has been<br />

found to be highly variable (25-64% of the patients<br />

examined), depending on a number of factors, as shown<br />

in Table 2.<br />

The variability in the reported incidence of CD34 + AML<br />

has also influenced the prognostic relevance of CD34<br />

expression in AML. Most authors found a clear association<br />

between CD34 + AML and a lower incidence of<br />

complete remission following induction therapy. In<br />

addition, the relapse rate was higher in AML showing<br />

positivity for the CD34 antigen compared to that of the<br />

CD34 – group. 44-53 However, other authors did not confirm<br />

these results and found no significant difference in<br />

the complete remission rate or overall survival of CD34 +<br />

and CD34 – AML patients. 54-60<br />

Expression of the CD34 antigen in AML and its association<br />

with different survival rates could be due to a<br />

Figure 1. DNA/RNA cellular content (acridine orange) of<br />

normal enriched CD34+ cells. Cell cycle measurements confirms<br />

that the majority of progenitors are quiescent (G0)<br />

with only few going into cycle (G1).<br />

haematologica vol. 85(suppl. to n. 12):December 2000


22<br />

M. Aglietta et al.<br />

number of factors. First of all, the cut-off point for CD34<br />

positivity that should be used to decided whether an<br />

AML sample is carrying this antigen. Since the proportion<br />

of CD34 + cells is around 1% of normal bone marrow<br />

mononuclear cells and 0.01-0.1% of peripheral<br />

blood leukocytes, many authors have considered 5% as<br />

the optimal cutoff level for classifying CD34 + AML.<br />

Nowadays, most authors agree that the cutoff point for<br />

CD34 should be 20% in order to avoid misinterpretation<br />

of the data coming from surface marker analysis. However,<br />

there is no scientific basis for considering a sample<br />

with 20% positive cells as positive, while another<br />

specimen with 19% positivity as negative, since the level<br />

of CD34 expression in a substantial number of<br />

patients is characterized by a continuous spectrum.53<br />

Furthermore, it must be kept in mind that the choice of<br />

a cutoff level of 5% could give rise to erroneous results,<br />

since it can be influenced by the methods used to detect<br />

antigen expression, which are characterized by different<br />

levels of sensitivity and specificity. Indirect immunofluorescence<br />

staining is more sensitive, although less specific,<br />

while the opposite is true for the direct technique.<br />

As far as the instrumentation is concerned, it must be<br />

underlined that modern flow cytometers are highly sensitive<br />

in detecting surface marker positivity with respect<br />

to microscope analysis and immunoenzymatic techniques<br />

such as APAAP, PAP, etc. In addition, whenever<br />

possible immunophenotype analysis should be preferentially<br />

performed on fresh, not cryopreserved bone<br />

marrow samples, and if this is not possible the number<br />

Table 2. Possible explanations for the differences reported in the literature<br />

concerning the incidence of CD34 + AML.<br />

1. Cut-off levels for the discrimination of positive and negative cases<br />

2. Detection systems employed (flow cytometry, type of flow cytometer,<br />

immunoenzymatic technique-APAAP, immunogold, PAP-immunofluorescence<br />

microscope)<br />

3. Specimen analyzed (bone marrow, peripheral blood)<br />

4. Percentage of leukemic cells present in the sample examined<br />

5. Use of cryopreserved rather than fresh cells<br />

6. Use of different CD34 antibodies recognizing distinct<br />

CD34 epitopes<br />

7. Percentage value and level of intensity for CD34<br />

8. Light scattering properties of CD34+ cells<br />

9. Patients analyzed (de novo AML or secondary AML)<br />

10. Biologic characteristics of AML cells (chromosome aberrations,<br />

gene abnormalities, immunophenotypic profile of CD34+ AML blasts)<br />

11. Type of chemotherapy regimen employed<br />

of blasts present in the specimen analyzed should be<br />

carefully evaluated. 61-65<br />

A recent report showed that CD34 antigen expression<br />

in AML samples having a marked heterogeneity in cell<br />

size was found preferentially on small leukemic cells<br />

with little or no side scatter. This feature was also associated<br />

with shorter remission duration and survival, suggesting<br />

that this morphological heterogeneity could<br />

reflect a peculiar biological behavior of AML. 66,67<br />

Moreover, discrimination of blast cells from residual<br />

normal nucleated cells is less likely to be obtained in<br />

AML cells by looking at light scattering properties (forward<br />

and side scatter) and expression of the CD45 antigen.<br />

For this reason, a multiparametric approach using<br />

two-three-colour analysis is strongly recommended in<br />

order to define the predominant leukemic population as<br />

well as minor pathological clones or subclones. In addition,<br />

CD34 positivity has to be evaluated solely on the<br />

blast population in order to avoid misinterpretation of<br />

the data. In fact, the percentage of blasts could vary<br />

from 30% to 99% in the bone marrow, and from 1 to<br />

99% in the peripheral blood.<br />

Another point which deserves careful discussion is<br />

represented by the level of expression for CD34 in AML.<br />

In normal hemopoiesis, the CD34 antigen is expressed<br />

on virtually all colony forming cells (CFU) and lymphocyte<br />

progenitors of either T or B lineage. However, within<br />

the progenitor cell compartment the degree of positivity<br />

for CD34 decreases with cell differentiation (maximum<br />

for multipotent cells and minimum for unipotent<br />

cells), and disappears in morphologically identifiable<br />

bone marrow precursors. Studies performed at the V<br />

International Workshop on Leukocyte Differentiation<br />

Antigens (Boston, 1993) recognized three main subsets<br />

of CD34 + normal bone marrow cells with CD34 antigen<br />

densities: low (2,000-5,000 binding sites per cell-ABC),<br />

medium (10,000-20,000 ABC), high intensities (25,000-<br />

40,000 ABC). This heterogeneity in CD34 antigen expression<br />

in normal progenitors makes it difficult to use this<br />

molecule for the monitoring of minimal residual disease<br />

(MRD) in AML patients treated with chemotherapy<br />

and/or bone marrow transplantation. 68 Flow cytometry<br />

allows the recognition of a subset of CD34 + AML characterized<br />

by bright expression for CD34 (> 50,000 ABC),<br />

which could therefore be easily recognized even when<br />

present in a very low percentage (< 0.1% of nucleated<br />

cells). This subset represents about 20-30% of CD34 +<br />

AML, so the remaining AML patients should be checked<br />

for MRD by using alternative ways (strategical double or<br />

triple staining: CD34/CD56; CD34/CD65/TdT; cytogenetics,<br />

molecular biology, etc.). 68<br />

Another source of variability in detecting CD34 + AML<br />

is represented by the type of CD34 monoclonal antibody<br />

used for immunophenotypic analysis. It has been<br />

demonstrated that at least three distinct CD34 epitopes<br />

exist, based on their differential sensitivity to enzymatic<br />

cleavage (using neuroaminidase, chymopapain and<br />

glycoprotease), Western blotting analysis, cell reactivity<br />

studies, and cross blocking experiments. 1,69-75 So far<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Peripheral blood stem cells in acute myeloid leukemia<br />

23<br />

at least twenty-two CD34 monoclonal antibodies<br />

(McAbs) have been shown to recognize the CD34 molecule,<br />

the most direct evidence being reactivity with<br />

cells transfected with CD34 cDNA and binding to CD34<br />

glycoprotein. 72 Recently, it has been reported that a<br />

number of AML cases are positive for some CD34 McAbs<br />

and negative for others (especially if they belong to a<br />

different epitope class), confirming the necessity of<br />

using the same CD34 McAbs in order to achieve comparable<br />

results between different centers. 71,72,74<br />

Another point which needs to be considered when<br />

evaluating the incidence of CD34 + AML is represented by<br />

patient characteristics at diagnosis. The number of CD34 +<br />

cases is higher in secondary than in newly diagnosed<br />

AML. If we consider that both the biology and the clinical<br />

pattern of secondary AML are quite different from<br />

those of de novo AML, one may argue that including of<br />

both types of leukemias in a clinical setting could interfere<br />

significantly with the prognostic relevance of CD34<br />

expression on AML blast cells. In this context, the type of<br />

treatment utilized by various authors (chemotherapy regimen,<br />

allogeneic and autologous bone marrow transplantation),<br />

which can influence the outcome of the disease,<br />

is also of some relevance.<br />

CD34 + AML are characterized by a higher incidence of<br />

chromosomes abnormalities involving chromosomes 5<br />

(–5, 5q–), 7 (–7.7q–), and to a lesser extent chromosomes<br />

16 (16q), 17 (17p), 11 (trisomy 11), or multiple<br />

chromosomes at the same time, generating the socalled<br />

major karyotypic abnormalities. 44,46,48,52,57,76 Recent<br />

studies have found a close relationship between CD34<br />

expression in AML and previous exposure to chemotherapy,<br />

radiotherapy and/or pesticides. 47 CD34 + AML are<br />

also associated with trilineage myelodysplasia, dysgranulopoiesis,<br />

and/or abnormalities of the p53 tumor<br />

suppressor gene. 77<br />

The correlation between CD34 + AML and FAB subtypes<br />

is illustrated in Table 3. On the other hand, most<br />

biphenotypic acute leukemias (BAL) show positivity for<br />

the CD34 antigen.<br />

Finally, the antigenic profile of CD34 + AML is rather<br />

heterogeneous, depending essentially on the morphological<br />

subtype and to a lesser extent on the differentiation<br />

stage of the leukemic clone. The large majority of<br />

CD34 + AML coexpress a number of antigens which are<br />

not associated with cell commitment, such as HLA-DR,<br />

CD38, CD45RO, CD45RA, CD71, and IL3 receptor. In<br />

addition, some surface and cytoplasmic glycoproteins<br />

expressed by committed myeloid cells were found to be<br />

positive in CD34 + AML, i.e. CD33, CD13, CD117 (stem<br />

cell factor receptor), CDw116 (GM-CSF receptor), G-CSF<br />

receptor, myeloperoxidase, lysozyme 78-82 (Figures 2 and<br />

3). The stem cell- associated antigen Thy-1 (CD90) is<br />

negative in this AML subtype, while nuclear TdT is sometimes<br />

positive. CD34 + cells also express high levels of P-<br />

glycoprotein, which is the product of the multiple drug<br />

resistance (MDR) gene.<br />

Post-remission therapy of acute myeloid<br />

leukemia and potential role of autologous<br />

stem cell transplantation<br />

About two thirds of previously untreated adults with<br />

primary acute myeloid leukemia enter complete remission<br />

(CR) after induction therapy based on cytarabine<br />

and an anthracycline. 83 However, long-term disease-free<br />

survival occurs in a minority of cases since most subjects<br />

relapse from proliferation of occult residual leukemic<br />

cells. Following conventional consolidation treatment<br />

less than 25% of patients remain in complete remission<br />

at four years. 84,85<br />

In order to eradicate residual AML cells and improve<br />

disease-free survival, three approaches have been<br />

employed in the last ten years: (a) intensive postremission<br />

chemotherapy; (b) allogeneic bone marrow transplantation<br />

(BMT), and (c) myeloablative conditioning<br />

regimens followed by autologous BMT as supportive<br />

therapy.<br />

Intensive postremission chemotherapy is essentially<br />

based on the use of high-dose cytarabine (1 to 3 g/m 2<br />

3 6 to 12 doses), either alone or in combination with<br />

other agents. Results of uncontrolled studies performed<br />

in the late ’80s and early ’90s (reviewed by Cassileth et<br />

al. 85 ) indicated that intensive post-remission therapy<br />

achieves long-term disease-free survival in 25-30% of<br />

patients in first CR. Mayer et al. 83 recently reported a<br />

prospective study aimed at evaluating the effect of the<br />

intensity of postremission chemotherapy on survival of<br />

leukemic patients. Acute leukemia individuals in first CR<br />

were randomly treated with four courses of cytarabine<br />

at one of three doses: 100 mg/m 2 per day for 5 days by<br />

continuous infusion; 400 mg/m 2 per day for 5 days by<br />

continuous infusion, or 3 g/m 2 in a 3-hour infusion every<br />

Table 3. Clinical and biological characteristics of CD34 + AML.<br />

Incidence: 30-50% (de novoAML); 50-70% (secondary AML)<br />

History: previous exposure to chemo-radiotherapy or pesticides<br />

Correlation with FAB subtypes: M0,M1,M5 (70-90%); M2,M4 (20-60%); M3:<br />

1-5%; M6, M7: 20-50%<br />

Prognosis: poor (mean survival rates less than 12-18 months)<br />

Cellular density for CD34: variable from case to case (range: 3,000- 100,000<br />

per blast cell)<br />

Immunophenotypic profile: in most cases CD45+, HLA-DR+, CD38+, CD33+,<br />

CD117+, Thy1–<br />

Chromosome abnormalities: -5, -7, 5q-, 7q-, 16q, 17p, major karyotype<br />

aberrations<br />

Therapy: to be defined (more aggressive chemotherapy regimens?)<br />

haematologica vol. 85(suppl. to n. 12):December 2000


24<br />

M. Aglietta et al.<br />

Figure 2. Dual color fluorescence analysis with a flow cytometer<br />

(CD34/FITC; c-kit-CD117-PE) in a patient with AML FAB M4. Bone marrow<br />

sample shows a blast percentage of 84%.<br />

A) Light scattering properties of blast cells (forward scatter= cell volume;<br />

side scatter= cell granularity).<br />

B)Contour plot diagram showing 83% of CD34 + cells, 71% of c-kit +<br />

cells, and 68.5% of cells co-expressing CD34 and c-kit.<br />

C)Histogram distribution of cells stained for CD34 monoclonal antibody.<br />

D)Histogram distribution of cells labelled with CD117 monoclonal antibody.<br />

Figure 3. Discordant reactivity between different epitope class antibodies<br />

is shown in a patient with AML M1 (% blasts = 90%); 36.5% of<br />

blasts are positive for class I, 0.1% for class II, and 47% for class III.<br />

The intensity of fluorescence varied significantly from antibody to antibody<br />

even within the same epitope class.<br />

12 hours on days 1, 3 and 5. In patients 60 years of age<br />

or younger the probability of remaining disease-free<br />

after four years correlated with the postremission<br />

cytarabine dose: 24% for the 100-mg group, 29% for<br />

the 400-mg group, and 44% for the 3-g group (p =<br />

0.002). In patients older than 60 the probability of<br />

remaining disease-free after four years was 16% or less<br />

in each of the three postremission cytarabine groups,<br />

with no significant difference between groups. It should<br />

be noted that a significant proportion of AML patients<br />

achieving CR cannot proceed to intensive chemotherapy<br />

due to persistent bone marrow aplasia.<br />

Allogeneic bone marrow transplantation offers many<br />

advantages, including the graft-versus-leukemia effect;<br />

however, the availability of a histocompatible sibling<br />

donor is restricted to approximately 25% of potential<br />

candidates. Published studies report disease-free survival<br />

rates at four years ranging from 45 to 58%. 86-88<br />

These figures should be considered with caution since<br />

they are biased by the exclusion of patients who<br />

relapsed before allogeneic BMT. The new approach<br />

recently described by Aversa et al., 89 i.e. a strong<br />

immunosuppressive and myeloablative conditioning<br />

regimen followed by transplantation of a large number<br />

of haploidentical stem cells depleted of T lymphocytes,<br />

may open new perspectives for allogeneic bone marrow<br />

transplantation in AML patients. In this setting, the<br />

mobilization and collection of allogeneic PBSC is crucial<br />

to overcoming HLA-disparity. 89<br />

Pilot studies on the use of autologous BMT as postremission<br />

therapy 90,91 indicate that disease-free survival at<br />

four years is on the order of 50% (i.e. comparable to that<br />

of allogeneic BMT). Autologous BMT has the advantage<br />

of lower procedure-related mortality than allogeneic BMT<br />

(approximately 10-15%), but involves a high risk of<br />

leukemic relapse (about one half treatment failures). A<br />

recent trial by Zittoun et al. 92 showed that both autologous<br />

and allogeneic BMT performed in first CR resulted<br />

in a significantly better disease-free survival than intensive<br />

consolidation chemotherapy. The projected rate at<br />

four years was 55% for allogeneic BMT, 48% for autologous<br />

BMT and 30% for intensive chemotherapy with no<br />

differences between allogeneic and autologous BMT.<br />

Taken together, these results demonstrate the potential<br />

benefit of autologous stem cell transplantation for<br />

leukemic patients. However, the high incidence of<br />

leukemic relapse and delayed hematological recovery<br />

after ABMT have prompted several authors to investigate<br />

the use of mobilized PBSC.<br />

CD34 + mobilized hematopoietic cells for<br />

support of intensive postremission<br />

chemotherapy of AML<br />

As stated above, autologous transplantation of mobilized<br />

hematopoietic progenitor cells has been shown to<br />

reconstitute hematopoiesis more efficiently than BMderived<br />

grafts. 2 Moreover, early studies have failed to<br />

detect neoplastic cells in the peripheral blood of AML<br />

patients during the early recovery phase following<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Peripheral blood stem cells in acute myeloid leukemia<br />

25<br />

Table 4. Reported studies on autologous stem cell transplantation in AML<br />

patients. PBSC indicates peripheral stem cell transplantation, while ABMT<br />

refers to autologous bone marrow transplantation.<br />

PBSC/ABMT PBSC/ABMT PBSC/ABMT<br />

Reference # 99 102 100<br />

Pts 28/683 38/13° 20/23*<br />

CFU-GM reinfused<br />

(x104/Kg)<br />

NR 86.6/12.1** 2.3/0.1<br />

(0.2-4.1/0-1)<br />

Median time to:<br />

> 0.5 x 109PMN/L<br />

(9-60/9-389)<br />

15.5/27<br />

(9-17/12-35)<br />

11/22 14/42<br />

NR<br />

> 50 x 109PLT/L<br />

(11-713/10-700)<br />

58.5/50<br />

(9-NA/21-NA)<br />

13.5/32 30/46<br />

NR<br />

PLT transfusion NR 5.4/8.8** NR<br />

Days on antibiotics NR 9.3/14.3** NR<br />

Hospitalization<br />

(days)<br />

NR 27.5/35.1** 45/73<br />

Relapse rate 57%/48% NR NR<br />

DFS 39%/42% NR 35%/51%<br />

°AML pts = 19 in PBSC group and 1 in ABMT group.<br />

* Comparison was made with 23 pts receiving purged marrow. ** Results expressed as<br />

the mean. Abbreviations: NR, not reported; NA, not achieved; DFS, disease free survival;<br />

PMN, neutrophil; PLT, platelet.<br />

remission induction/consolidation chemotherapy. 93,94<br />

Based on these reports, several investigators have<br />

addressed the question of whether the use of PBSC<br />

might result in a more rapid engraftment and a lower<br />

risk of relapse in AML patients. Relevant issues include<br />

the level of malignant cell contamination, the threshold<br />

dose of hematopoietic precursors (i.e. CFU-GM and<br />

CD34 + cells), the optimal timing for stem cell collection<br />

and the potential benefit derived from the use of selected<br />

cytokines to improve PBSC harvest.<br />

To et al. 93 studied leukemia-associated cytogenetic<br />

abnormalities in myeloid colonies derived from early<br />

remission PBSC. At a sensitivity level of 2:100 cells, they<br />

were not able to detect the t(8;21) in 293 samples<br />

examined. Recently, the more sensitive nested reverse<br />

transcriptase polymerase chain reaction (RT-PCR) was<br />

used to monitor minimal residual disease in the BM and<br />

peripheral blood of leukemic patients considered in<br />

remission by morphologic analysis. 95 In that study, the<br />

authors found no differences between PBSC collections<br />

and simultaneous BM harvests. However, the degree of<br />

leukemic contamination may have been different<br />

among the PCR-positive groups because the two-step<br />

PCR is highly sensitive but not quantitative.<br />

The issue of leukemia-free autograft was recently<br />

underscored by gene-marking studies showing that<br />

residual contaminating AML cells contribute to relapse<br />

when reinfused into patients. 96 In this regard, leukemic<br />

recurrence remains the most frequent cause of treatment<br />

failure in AML patients 97,98 and preliminary nonrandomized<br />

clinical studies have not reported any<br />

advantage for ABMT over PBSC 99-101 in terms of diseasefree<br />

survival and overall survival rate (Table 4). Moreover,<br />

it could be argued that the interval between complete<br />

remission and myeloablative therapy may be<br />

shorter for PBSC patients, since the exclusion rate is<br />

higher for ABMT patients. 101 Thus, randomized studies<br />

are warranted to rule out selection bias. Reinfusion of<br />

PBSC markedly shortened the period of marrow aplasia<br />

compared to purged and unpurged ABMT 99-102 and<br />

reduced morbidity and resource utilization (Table 4). In<br />

particular, To et al. 102 demonstrated an advantage of 11<br />

and 19 days in the median time to achieve 0.5×10 9 neutrophils/L<br />

and 50×10 9 platelets/L, respectively, in favor<br />

of PBSC, whereas two other studies 99,100 showed a more<br />

rapid neutrophil engraftment (28 days and 12 days,<br />

respectively) but not a highly significant faster platelet<br />

recovery. Most likely the acceleration of hematopoietic<br />

reconstitution derives from the reinfusion of a higher<br />

number of early pluripotent precursors and large<br />

amounts of committed progenitor cells which require<br />

less time to reach maturation. 103<br />

Early studies in acute leukemia indicated that an optimal<br />

CFU-GM dose of 50×10 4 /kg body weight is required<br />

for complete and sustained reconstitution of BM function.<br />

104 Other reports 99-101 and our own preliminary experience<br />

(Tables 4 and 5) have demonstrated similar<br />

results with lower CFU-GM numbers. These differences<br />

are probably related to different assay methods, whereas<br />

the more reproducible evaluation of progenitor cells<br />

by flow cytometry has suggested a threshold dose of<br />

2×10 6 CD34 + cells/kg. 105 Interestingly, the number of<br />

progenitor cells infused is only indirectly related to longterm<br />

engraftment, suggesting that additional measurements<br />

of CD34 + cell fraction subsets may be helpful in<br />

predicting sustained recovery of hematopoiesis. Moreover,<br />

a transient secondary fall in neutrophil and<br />

platelet count has been described 104 during the 3 rd -8 th<br />

weeks after transplantation, indicating a time lag<br />

between exhaustion of the committed progenitor cell<br />

pool, which is responsible for early engraftment, and<br />

expansion of the more immature pluripotent stem cell<br />

compartment.<br />

Lastly, the timing of BM harvest or PBSC collection in<br />

the autologous setting plays an important role in the<br />

quality of the autograft. It has been clearly established<br />

that the amount and the length of previous therapy<br />

affects the number of circulating CD34 + cells; 106 however,<br />

reinfusion of PBSC collected in the recovery phase<br />

of induction therapy has resulted in a higher relapse<br />

rate 101,107 indicating that returning a larger quantity of<br />

cells to patients without careful analysis of minimal<br />

residual disease may increase the probability of transplantation<br />

of leukemic cells. Thus, at least one consolidation<br />

cycle of treatment should be performed in the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


26<br />

M. Aglietta et al.<br />

case of stem cell mobilization to take advantage of in<br />

vivo purging, coupled with a low number of apheresis<br />

procedures. To this end, it has been shown 108 that<br />

administration of G-CSF during the recovery phase of<br />

consolidation chemotherapy in acute leukemia patients<br />

increased the peak level of CFU-GM and CFU-MIX by 5.8<br />

and 4.3 times, respectively, compared to cycles were G-<br />

CSF was not used, and significantly prolonged the period<br />

of mobilization of stem cells. Although the role of<br />

cytokine treatment in AML patients is still under evaluation,<br />

preliminary data from a large cohort of leukemic<br />

patients suggest that G-CSF administration does not<br />

affect either the remission or relapse rate, 109,110 whereas<br />

a protective effect regarding relapse was shown in a<br />

randomized study. 29<br />

In practice, leukapheresis sessions should be started<br />

after a careful evaluation of CD34 + cells in the peripheral<br />

blood by flow cytometry, at time of hematopoietic<br />

recovery from transient myelosuppression.<br />

When adequate mobilization of progenitors (CD34 +<br />

cells > 10-15/µL) occurs, daily leukaphereses should be<br />

performed until the collection of a minimum number of<br />

2×10 6 /kg CD34 + cells. The leukapheresis products should<br />

be evaluated for the presence of residual contaminating<br />

leukemic cells by immunophenotyping, karyotypic<br />

analysis and RT-PCR-based molecular analysis in those<br />

samples deriving from patients who had shown a specific<br />

phenotypic and/or molecular marker at diagnosis.<br />

Moreover, because the content of circulating progenitors<br />

is generally low (< 1% of the mononuclear cell<br />

fraction), blood cell separator efficiency must be optimized.<br />

Collection efficiency (CE) is the percentage of<br />

cells entering the system that are eventually collected:<br />

CE (%) =<br />

No. harvested cells<br />

× processed blood volume<br />

No. of cells in preapheresis<br />

blood unit volume<br />

Acceptable CE should not be lower than 50%. CE is a<br />

useful parameter for evaluating blood cell separator<br />

effectiveness in harvesting PBSC, independently of the<br />

patient’s clinical condition.<br />

Purging in AML<br />

Considering the possibility of relapse from minimal<br />

residual disease (MRD) derived from autologous graft,<br />

several investigators addressed the issue of ex vivo purging<br />

of leukemic cells prior to stem cell reinfusion. Using<br />

the Brown Norway myelocytic leukemia rat model, it<br />

has been shown that injection of 25 leukemic cells<br />

induces leukemia in 50% of recipients. 111 By applying<br />

the same mathematical model to humans, it has been<br />

suggested that reinfusion of 10,000 residual leukemic<br />

cells may result in a relapse rate as high as 50%. 111 More<br />

recently, the role of residual tumor cells in clinical<br />

relapse after autograft was indicated by a clinical study<br />

Table 5. Experience of the Institute of Hematology “Seragnoli”, Bologna<br />

on autologous stem cell transplantation in AML patients. The results are<br />

expressed as median (range) and refer to AML (n=7) and RAEB-T (n=2)<br />

patients in I CR. PBSC collections were carried out following consolidation<br />

treatment. PMN and PLT recovery was recorded as such when the PB<br />

count was > 0.5 and 20 x 10 9 /L, respectively.<br />

Apheresis products<br />

Pts PBSC MNC CFU-GM CD34+<br />

collections (108/Kg) (104/Kg) (106/Kg)<br />

9 3 (2-3) 7 (2.8-11.6) 11.8 (2.8-78.2) 7 (3.1-17.5)<br />

Hematological reconstitution<br />

Pts day to PMN day to PLT PLT RBC<br />

Hospital<br />

recovery recovery transfusion transfusion<br />

discharge<br />

8 14 (11-34) 18 (10-NR) 2.5 (0->10) 4 (1-9) 18 (14-38)<br />

Abbreviations: MNC, mononuclear cells; RBC, red blood cells.<br />

involving 114 B-cell lymphoma patients with t(14;18)<br />

who received autologous marrow treated with a combination<br />

of monoclonal antibodies directed against B-<br />

cell associated antigens plus complement. 112 Following<br />

purging, no lymphoma cells could be detected by PCR<br />

amplification of the bcl-2 gene in the marrow of 57<br />

patients. Disease-free survival was increased in these<br />

individuals with respect to that of patients whose marrow<br />

contained detectable tumor cells. Moreover, the<br />

ability to remove lymphoma cells was the most important<br />

prognostic factor for predicting relapse (39% versus<br />

5% of purged patients after a median follow-up of<br />

23 months). 112 Lastly, genetic marking of marrow cells<br />

with the neomycin-resistance gene has provided the<br />

formal proof that reinfusion of residual leukemic cells in<br />

AML patients contributes to a recurrence of the disease.<br />

113 Taken together, these findings demonstrate the<br />

need for effective ex vivo treatments to improve the<br />

outcome of autologous transplantation.<br />

Among the many purging methods proposed for the<br />

elimination of MRD, 114 cyclophosphamide (Cy) derivatives<br />

are the most widely used agents in AML patients,<br />

since preclinical models demonstrated that these compounds<br />

were able to eliminate residual BM neoplastic<br />

cells in the Brown Norway rat system. 115 The main mechanism<br />

of action of Cy active metabolites is based upon<br />

marked inhibition of leukemic progenitor cell (CFU-L)<br />

growth 115 while sparing normal primitive hematopoietic<br />

cells. 116 Furthermore, these alkylating agents seem to<br />

induce apoptotic death of leukemic cells 117 as well as the<br />

activation of immune mechanisms capable of controlling<br />

leukemic cell proliferation. 118 Combinations of 4-<br />

hydroperoxycyclophosphamide (4-HC) or nitrogen mustard<br />

and VP-16 have also been proposed to increase the<br />

selective toxicity of pharmacologic purging towards<br />

neoplastic cells. 119 Several monoclonal antibodies that<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Peripheral blood stem cells in acute myeloid leukemia<br />

27<br />

recognize tumor-associated or cell-differentiation antigens<br />

not expressed by primitive cells responsible for<br />

hematopoietic engraftment have been selected for clinical<br />

trials after in vitro studies demonstrated a high<br />

purging efficacy with the use of complement, toxins and<br />

radioactive molecules. However, the heterogeneity of<br />

antigen expression on neoplastic cells and the lack of<br />

tumor-specific determinants may greatly affect the efficiency<br />

of antibody-based strategies for depletion of<br />

leukemic cells. In this context, the combination of two<br />

purging techniques has been explored with promising<br />

results. 120 Other approaches include the use of photoactive<br />

compounds which sensitize leukemic cells and<br />

specifically damage their cell membranes upon exposure<br />

to light, 121 and biological methods based on the different<br />

proliferative patterns of leukemic cells and their normal<br />

counterparts when cultured ex vivo for several days<br />

in the presence of stromal cells. 122<br />

Clinical trials<br />

Clinical retrospective data supporting the beneficial<br />

effect of purging have been progressively accumulating.<br />

The Baltimore team provided indirect evidence in favor<br />

of purging by correlating effective CFU-GM colony elimination<br />

with a significant decrease in relapse. 123 Furthermore,<br />

the same authors associated the sensitivity to<br />

4-HC of CFU-L grown in remission with the posttransplant<br />

outcome. 124 The 3 most recent surveys of the<br />

Leukemia Working Party of the EBMT group have consistently<br />

reported lower relapse rates following reinfusion<br />

of BM purged with mafosfamide, 91,125,126 especially<br />

in patients transplanted within 6 months of CR and in<br />

slow remitters (> 40 days to achievement of CR), two<br />

patient populations considered at high risk of disease<br />

recurrence. In fact, the proportion of patients relapsing<br />

in the purged and unpurged groups was 29±5% vs<br />

50±4%, respectively, following a conditioning regimen<br />

that included total body irradiation (p < 0.0001). More<br />

striking differences were found when considering only<br />

those patients autografted early after CR (16±6% vs<br />

60±6%) and patients with an interval from diagnosis to<br />

CR greater than 40 days (20±8% vs 61±6%). Gulati et<br />

al. and Laporte et al. 127, 128 reported disease free survival<br />

which approximated 60% in AML patients in I CR reinfused<br />

with autologous marrow treated with 4-HC and<br />

VP-16 and mafosfamide. In the same paper by the Paris<br />

group, 151 it was suggested that the higher the initial<br />

content of BM CFU-GM, the lower the risk of transplant<br />

related mortality and the higher the chance of curing<br />

the disease.<br />

Despite these results in favor of ex vivo elimination of<br />

contaminating leukemic cells, this procedure is not routinely<br />

performed in the majority of transplant centers.<br />

The main reasons might be: 1) the delay in hematological<br />

recovery after reinfusion of purged autografts; 2)<br />

the increase in the cost of ABMT; 3) the need for technical<br />

training and, most of all, 4) the lack of results<br />

derived from prospective clinical studies demonstrating<br />

the effects of purging. The feasibility of such trials is<br />

rather limited due to the high number of patients needed<br />

to obtain adequate statistical power.<br />

So far, no data are available on purging protocols for<br />

PBSC collections; however, there are several reasons for<br />

proposing purging strategies for autograft of circulating<br />

autologous stem cells. Unlike solid tumors and<br />

malignant lymphomas, acute leukemias easily involve<br />

PB. Moreover, the number of hematopoietic progenitor<br />

cells (and possibly leukemic precursors) in PB autotransplants<br />

is usually at least 10 times higher than that<br />

of ABMT. Finally, as discussed above, the interval<br />

between CR and autotransplant may be shorter for PBSC<br />

patients, who may be thus considered high risk patients<br />

for relapse. Critical issues for designing experimental<br />

studies in this setting would include the proper assessment<br />

of MRD before and after purging, the establishment<br />

of reproducible technical protocols (cell concentration,<br />

RBC contamination, etc.), and careful evaluation<br />

of the toxicity of purging agents on PB progenitors, since<br />

the kinetic status of circulating stem cells following<br />

mobilization protocols (especially if CSFs are used) may<br />

be different from BM stem cells. 129<br />

Conclusions<br />

Autologous BMT has been widely used as consolidation<br />

therapy in AML patients in first or second remission;<br />

however, delayed hematopoietic engraftment occurs in<br />

a substantial proportion of patients resulting in significant<br />

morbidity and mortality. This is mainly due to the<br />

adverse effects of prior intensive chemotherapy on BM<br />

harvest, a decrease in the normal stem cell pool in<br />

leukemic patients and, perhaps, toxic damage to the<br />

marrow microenvironment. Thus, several groups have<br />

investigated the use of circulating progenitor cells with<br />

the twofold aim of reducing transplant-related toxicity<br />

and widening the number of potential candidates for<br />

myeloablative therapy with the support of autologous<br />

stem cells.<br />

As for hematopoietic reconstitution, previous studies<br />

have provided evidence that PBSC transplantation may<br />

offer some advantages over BM autografting. However,<br />

crucial issues such as asynchronous mobilization of normal<br />

vs leukemic cells and potential contamination of<br />

PBSC collections, timing of PBSC harvest, detection of<br />

minimal residual disease, and the role of growth factors<br />

to accelerate hematological recovery and optimize stem<br />

cell collection have not been fully addressed.<br />

In the present paper, the latest advances in this field<br />

have been reviewed with special focus on the biology of<br />

putative leukemic stem cells; operative guidelines have<br />

also been provided for those investigators who wish to<br />

design proper clinical trials on PBSC autotransplantation<br />

in acute leukemia.<br />

Definitive answers regarding the role of PBSC will be<br />

coming from a large European randomized trial which<br />

is currently comparing peripheral blood stem cell and<br />

BM-derived graft.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


28<br />

M. Aglietta et al.<br />

References<br />

1. Carlo Stella C, Cazzola M, De Fabritiis P, et al. CD34-<br />

positive cells: biology and clinical applications.<br />

<strong>Haematologica</strong> 1995; 80:367-87.<br />

2. Siena S, Bregni M, Di Nicola M, et al. Durability of<br />

hematopoiesis following autografting with peripheral<br />

blood hematopoietic progenitors. Ann Oncol 1994;<br />

5:935-41.<br />

3. To LB. Is our current strategy in manipulating hemopoiesis<br />

in autologous transplantation correct? Stem<br />

Cells 1993; 11: 283-9.<br />

4. Majolino I, Aversa F, Bacigalupo A, Bandini G, Arcese<br />

W, Reali G. Allogeneic transplants of the rhG-CSFmobilized<br />

peripheral blood stem cells (PBSC) from<br />

normal donors. <strong>Haematologica</strong> 1995; 80:40-3.<br />

5. Yin M, Silvestri FF, Banavali SD, et al. Clonogenic<br />

potential of myeloid leukemia cells in vitro is restricted<br />

to leukemia cells expressing the CD34 antigen. Eur<br />

J Cancer 1993; 29A: 2279-83.<br />

6. Carlo-Stella C, Mangoni L, Almici C, Frassoni F, Fiers<br />

W, Rizzoli V. Growth of CD34+ acute myeloblastic<br />

leukemia colony-forming cells in response to recombinant<br />

hemopoietic growth factors. Leukemia 1990;<br />

4:561-6.<br />

7. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating<br />

human acute myeloid leukaemia after transplantation<br />

into SCID mice. Nature 1994; 367:645-8.<br />

8. Ikeda H, Kanakura Y, Tamaki T, et al. Expression and<br />

functional role of the proto-oncogene c-kit in acute<br />

myeloblastic leukemia cells. Blood 1991; 78:2962-8.<br />

9. Broudy VC, Smith FO, Lin N, Zsebo K, Egrie J, Berstein<br />

ID. Blasts from patients with acute myelogenous<br />

leukemia express functional receptors for stem cell factor.<br />

Blood 1992; 80:60-7.<br />

10. Motoji T, Watanabe M, Uzumaki H, et al. Granulocyte<br />

colony-stimulating factor (G-CSF) receptors on<br />

acute myeloblastic leukemia cells and their relationship<br />

with the proliferative response to G-CSF in clonogenic<br />

assay. Br J Haematol 1991; 77:54-9.<br />

11. Budel LM, Touw IP, Delwel R, Clark SC, Lowenberg B.<br />

Interleukin-3 and granulocyte-monocyte colony stimulating<br />

factor receptors on acute myelocytic leukemia<br />

cells and relationship to the proliferative response.<br />

Blood 1989; 74:565-71.<br />

12. Pietsch T, Kyas U, Steffens U, et al. Effects of human<br />

stem factor (c-kit ligand) on proliferation of myeloid<br />

leukemia cells: heterogeneity in response and synergy<br />

with other hematopoietic growth factors. Blood 1992;<br />

80:1199-206.<br />

13. Piacibello W, Sanavio F, Bresso P, et al. Stem cell factor<br />

improvement of proliferation and maintenance of<br />

hemopoietic progenitors in myelodysplastic syndrome.<br />

Leukemia 1994; 8:250-7.<br />

14. Carlo Stella C, Rizzoli V. Stem cells and stem cell factor(s).<br />

<strong>Haematologica</strong> 1995; 80:1-4.<br />

15. Miyajima A, Mui ALF, Ogorochi T, Sakamaki K.<br />

Receptors for granulocyte-macrophage colony stimulating<br />

factor, interleukin-3 and interleukin-5. Blood<br />

1993; 82:1960-74.<br />

16. Lowenberg B, Touw IP. Hemopoietic growth factors<br />

and their receptors in acute leukemia. Blood 1993;<br />

81:281-92.<br />

17. Mui ALF, Miyajima A. Interleukin-3 and granulocytemacrophage<br />

colony-stimulating factor receptor signal<br />

transduction. Proc Soc Exp Biol Med 1994; 206:<br />

284-8.<br />

18. Begley CG, Metcalf D, Nicola NA. Primary human<br />

myeloid leukemia cells: comparative responsiveness<br />

to proliferative stimulation by GM-CSF or G-CSF and<br />

membrane expression of CSF receptors. Leukemia<br />

1987; 1:1-8.<br />

19. Aglietta M, DeFelice L, Stacchini A, et al. Effect of<br />

hemopoietic growth factors on the proliferation of<br />

acute myeloid and lymphoid leukemias. Leuk Lymphoma<br />

1990; 2:207-14.<br />

20. Damiani D, Michieli M, Revignas MG, et al. Proliferation<br />

and maturation effects of in vivo granulocytemacrophage<br />

colony stimulating factor in acute nonlymphocyte<br />

leukemia. <strong>Haematologica</strong> 1992; 77: 25-9.<br />

21. Kondo S, Okamma S, Asano Y, Harada M, Niko Y.<br />

Human granulocyte colony stimulating factor receptors<br />

in acute myelogenous leukemia. Eur J Haematol<br />

1991; 46:223-30.<br />

22. Sachs L, Lotem J. Control of programmed cell death<br />

in normal and leukemic cells: new implications for<br />

therapy. Blood 1993; 82:15-21.<br />

23. Aglietta M, De Felice L, Stacchini A, et al. In vivo effect<br />

of granulocyte-macrophage colony-stimulating factor<br />

on the kinetics of human acute myeloid leukemia cells.<br />

Leukemia 1991; 5:979-84.<br />

24. Bettelheim P, Valent P, Andreeff M, et al. Recombinat<br />

human granulocyte-macrophage colony stimulating<br />

factor in combination with standard induction<br />

chemotherapy in de novo acute myeloid leukemia.<br />

Blood 1991; 77:700-11.<br />

25. Estey E, Thall P, Andreeff M, et al. Use of granulocyte<br />

colony-stimulating factor before, during and after fludarabine<br />

plus cytarabine induction therapy of newly<br />

diagnosed acute myelogenous leukemia or myelodysplastic<br />

syndromes: comparison with fludarabine plus<br />

cytarabine without granulocyte colony-stimulating<br />

factor. J Clin Oncol 1994; 12:671-8.<br />

26. Ohno R, Naoe T, Kanamaru A, et al. A double-blind<br />

controlled study of granulocyte colony-stimulating<br />

factor started two days before induction chemotherapy<br />

in refractory acute myeloid leukemia. Blood 1994;<br />

83:2066-92.<br />

27. Giralt S, Escudier S, Kantarjian H, et al. Preliminary<br />

results of treatment with filgrastim for relapse of<br />

leukemia and myelodysplasia after allogenic bone<br />

marrow transplantation. N Engl J Med 1993; 329:<br />

757-61.<br />

28. Stone RM, Berg DT, George SL, et al. Granulocytemacrophage<br />

colony stimulating factor after initial<br />

chemotherapy for elderly patients with primary acute<br />

myelogenous leukmia. N Engl J Med 1995; 332:1671-<br />

7.<br />

29. Dombret H, Chastang C, Fenaux P, et al. A controlled<br />

study of recombinant human granulocyte colony stimulating<br />

factor in elderly patients after treatment for<br />

acute myelogenous leukemia. N Engl J Med 1995;<br />

332:1678-83.<br />

30. Lajtha LG. On the concept of the cell cycle. J Cell<br />

Comp Physiol 1963; 62:143-5.<br />

31. Hodgson GS, Bradley TR. Properties of haematopoietic<br />

stem cells surviving 5-fluorouracil treatment: evidence<br />

for a pre-CFU-S cell? Nature 1979; 281:381-2.<br />

32. Leary AG, Hirai Y, Kishimoto T, Clark SG, Ogawa M.<br />

Survival of hemopoietic progenitors in G0 does not<br />

require early hemopoietic regulators. Proc Natl Acad<br />

Sci USA 1989; 86:4535-9.<br />

33. Till JE, McCulloch EA, Siminovitch L. A stochastic<br />

model of stem cell proliferation, based on the growth<br />

of spleen colony-forming cells. Proc Natl Acad Sci USA<br />

1964; 51:29-34.<br />

34. Minden MD, Till JE, McCulloch EA. Proliferative state<br />

of blast cell progenitors in acute myeloblastic<br />

leukemia (AML). Blood 1978; 52:592-6.<br />

35. Clarkson B, Fried J, Striff A, Saki Y, Ota K, Ohkita T.<br />

Studies of cellular proliferation in human leukemia.<br />

III. Behavior of leukemic cells in three adults with acute<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Peripheral blood stem cells in acute myeloid leukemia<br />

29<br />

leukemia given continuous infusions of 3H-thymidine<br />

for 8-10 days. Cancer 1970; 25:1237-60.<br />

36. Gavosto F, Pileri A, Gabutti V, Masera P. Non selfmaintaining<br />

kinetics of proliferating blasts in human<br />

acute leukaemia. Nature 1967; 216:188-9.<br />

37. Andreeff M, Darzynkiewicz Z, Sharpless TK, Clarkson<br />

BD, Melamed MR. Discrimination of human<br />

leukemia subtypes by flow cytometric analysis of cellular<br />

DNA and RNA. Blood 1980; 55:282-93.<br />

38. Raza A, Maheshwari Y, Preisler HD. Differences in cell<br />

cycle characteristics among patients with acute nonlymphocytic<br />

leukemia. Blood 1987; 69:1647-53.<br />

39. Lista P, Brizzi MF, Rossi M, Resegotti L, Clark SC,<br />

Pegoraro L. Different sensitivity of normal and<br />

leukaemic progenitor cells to Ara-C and IL-3 combined<br />

treatment. Br J Haematol 1990; 76:21-6.<br />

40. Tafuri A, Andreeff M. Kinetic rationale for cytokineinduced<br />

recruitment of myloblastic leukemia followed<br />

by cycle-specific chemotherapy in vitro. Leukemia<br />

1990;12:826-34.<br />

41. Raymakers R, Wittebol S, Pennings A, Linders E, Poddighe<br />

P, De Witte T. Residual normal, highly proliferative<br />

progenitors can be isolated from the CD34+/33–<br />

fraction of AML with a more differentiated phenotype<br />

(CD33+). Leukemia 1995; 9:450-7.<br />

42. Gore SD, Amin S, Weng LJ, Civin C. Steel factor supports<br />

the cycling of isolated human CD34+ cells in<br />

the absence of other growth factors. Exp Hematol<br />

1995; 23:413-21.<br />

43. Hunter T, Pines J. Cyclins and Cancer II: cyclin D and<br />

CDK inhibitors come of age. Cell 1994; 79:573-82.<br />

44. Vaughan W, Civin C, Welsenburger D, et al. Acute<br />

leukemia expressing the normal human haematopoietic<br />

stem cell membrane glycoprotein CD34+ (MY10).<br />

Leukemia 1988; 2:661-6.<br />

45. Campos L, Guyotat D, Archimbaud E, et al. Surface<br />

marker expression in adult acute myeloid leukemia:<br />

correlations with initial characteristics, morphology<br />

and response to therapy. Br J Haematol 1989; 72:<br />

161-6.<br />

46. Borowitz M, Gockerman J, Moore J, et al. Clinicopathologic<br />

and cytogenetic features of CD34+<br />

(MY10) positive acute nonlymphocytic leukemia. Am<br />

J Clin Pathol 1989; 91:265-70.<br />

47. Geller RB, Zahurak M, Hurwitz C. Prognostic importance<br />

of immunophenotyping in adults with acute<br />

myelocytic leukemia: the significance of the stem cell<br />

glycoprotein CD34+ (MY10). Br J Haematol 1990;<br />

76:340-7.<br />

48. Guinot M, Sanz G F, Sempere A, et al. Prognostic value<br />

of CD34 expression in de novo acute myeloblastic<br />

leukemia. Br J Haematol 1991; 78:533-4.<br />

49. Lee E, Yang J, Leavitt R. The significance of CD34+<br />

and TdT determinations in patients with untreated<br />

de novo acute myeloid leukemia. Leukemia 1992; 6:<br />

1203-9.<br />

50. Solary E, Casasnovas R, Campos L and the Groupe<br />

d’Etude Immunologique des Leucemies. Surface<br />

markers in adult acute myeloblastic leukemia: correlation<br />

of CD19+, CD34+ and CD14+/DR+ phenotype<br />

with shorter survival. Leukemia 1992; 6:93-9.<br />

51. Myint H, Lucie N. The prognostic significance of the<br />

CD34+ antigen in acute myeloid leukemia. Leuk Lymphoma<br />

1992; 7:425-9.<br />

52. Fagioli F, Cuneo A, Carli M, et al. Chromosome aberrations<br />

in CD34+ positive acute myeloid leukemia:<br />

correlations with clinicopathologic features. Cancer<br />

Genet Cytogenet 1993; 71:119-24.<br />

53. Lanza F, Rigolin M, Moretti S, Latorraca A, Castoldi<br />

G. Prognostic value of immunophenotypic characteristics<br />

of blast cells in acute myeloid leukemia. Leuk<br />

Lymphoma 1994; 13:81-5.<br />

54. Smith FO, Lampkin B, Versteeg C, et al. Expression of<br />

lymphoid-associated cell surface antigens by childhood<br />

acute myeloid leukemia cells lacks prognostic<br />

significance. Blood 1992; 79:2415-22.<br />

55. Kuerbit SJ, Civin CI, Krisher JP, et al. Expression of<br />

myeloid-associated and lymphoid-associated cell surface<br />

antigens in acute myeloid leukemia of childhood:<br />

a pediatric oncology group study. J Clin Oncol 1992;<br />

10:1419-29.<br />

56. Selleri C, Notaro R, Catalano L, Fontana R, Del Vecchio<br />

L, Rotoli B. Prognostic irrelevance of CD34+ in<br />

acute myeloid leukemia. Br J Haematol 1992; 82:479-<br />

82.<br />

57. Sperling C, Buchner T, Sauerland C, Fonatsch C, Thiel<br />

E, Ludwig W. CD34 expression in de novo acute<br />

myeloid leukemia. Br J Haematol 1993; 85:635-7.<br />

58. Ciolli S, Leoni F, Caporale R, Pascarella A, Salti F,<br />

Rossi-Ferrini P. CD34+ expression fails to predict the<br />

outcome in adult acute myeloid leukemia. <strong>Haematologica</strong><br />

1993; 78:151-5.<br />

59. Del Poeta G, Stasi R, Venditti A, et al. Prognostic value<br />

of cell marker analysis in de novo acute myeloid<br />

leukemia. Leukemia 1994; 8:388-94.<br />

60. Sperling C, Buchner T, Creutzig U. Clinical, morphologic,<br />

cytogenetic and prognostic implications of<br />

CD34+ expression in childhood and adult de novo<br />

AML. Leuk Lymphoma 1995; 17:417-26.<br />

61. Terstappen L, Safford M, Konemann S, et al. Flow<br />

cytometric characterization of acute myeloid<br />

leukemia. Part II. Phenotypic heterogeneity at diagnosis.<br />

Leukemia 1991; 9:757-67.<br />

62. van’t Veer M, Kluin-Nelemans J, van Der Schoot C,<br />

van Putten W, Adriaansen H, van Wering E. Quality<br />

assessment of immunological marker analysis and the<br />

immunological diagnosis in leukemia and lymphoma:<br />

a multi-centre study. Br J Haematol 1992; 80:458-<br />

65.<br />

63. Brando B, Sommaruga E. Nationwide quality control<br />

trial on lymphocyte immunophenotyping and flow<br />

cytometer performance in Italy. Cytometry 1993; 14:<br />

294-306.<br />

64. Castoldi G, Liso V, Fenu S, Vegna L, Mandelli F.<br />

Reproducibility of the morphological diagnostic criteria<br />

for acute myeloid leukemia: the GIMEMA group<br />

experience. Ann Haematol 1993; 66:171-4.<br />

65. Drexler H, Eckhard T, Wolf-Dieter L. Acute myeloid<br />

leukemias expressing lymphoid-associated antigens:<br />

diagnostic incidence and prognostic significance.<br />

Leukemia 1993; 7:489-98.<br />

66. Kawada H, Ichikawa Y, Watanabe S, Nagao T, Arimori<br />

S. Flow cytometric analysis of cell surface antigen<br />

expressions on acute myeloid leukemia cell populations<br />

according to their cell size. Leuk Res 1994;<br />

18:29-35.<br />

67. Syrjala M, Ruutu T, Jansson SE. A flow cytometric<br />

assay of CD34-positive cell populations in the bone<br />

marrow. Br J Haematol 1994; 88:679-84.<br />

68. Campana D, Pui CH. Detection of minimal residual<br />

disease in acute leukemia: methodologic advances<br />

and clinical significance. Blood 1995; 85:1416-34.<br />

69. Greaves MF, Brown J, Molgaard HV, et al. Molecular<br />

features of CD34: a hemopoietic progenitor cell-associated<br />

molecule. Leukemia 1992; 6:31-6.<br />

70. Sutherland DR, Marsh JCW, Davidson J, Baker MA,<br />

Keating A, Mellors A. Differential sensitivity of CD34<br />

epitopes to cleavage by Pasteurella haemolytica glycoprotease:<br />

implications for purification of CD34-<br />

positive progenitor cells. Exp Hematol 1992; 20:590-<br />

9.<br />

71. Egeland T, Gaudernack G. Immunomagnetic isola-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


30<br />

M. Aglietta et al.<br />

tion of CD34+ cells: methodology and monoclonal<br />

antibodies. In: Wunder S, Serke S, Solovat H, Henon<br />

P, eds. Hematopoietic stem cell: the Mulhouse manual.<br />

Dayton: AlphaMed Press, 1994:141-8.<br />

72. Greaves MF, Colman SM, Böhring HJ, et al. Report on<br />

the CD34 cluster workshop. In: Schlossman SF,<br />

Boumsell L, Gilks W et al, eds. Leucocyte typing V:<br />

White cell differentiation antigens. Oxford: Oxford<br />

University Press, 1995:840-7.<br />

73. Silvestri F, Banavali S, Baccarani M, Preisler HD. Progenitor<br />

cell associated antigen CD34: biology and clinical<br />

applications. <strong>Haematologica</strong> 1992; 77:265-72.<br />

74. Lanza F, Castoldi G. Large scale enrichment of CD34+<br />

cells by percoll density gradients. A CML-based study<br />

design. In: Wunder E, Serke S, Sovalat H, Henon P,<br />

eds. Hematopoietic stem cells: the Mulhouse manual.<br />

Dayton: Alpha Med Press, 1994:255-70.<br />

76. Slovak ML, Traweek ST, Willman CL, et al. Trisomy<br />

11: an association with stem/progenitor cell immunophenotype.<br />

Br J Haematol 1995;90:266-73.<br />

77. Lai J, Preudhomme C, Zandecki M, et al. Myelodysplastic<br />

syndromes and acute myeloid leukemia with<br />

17p deletion. An entity characterized by specific dysgranulopoiesis<br />

and a high incidence of p53 mutations.<br />

Leukemia 1995; 9:370-81.<br />

78. Pierelli L, Teofili L, Menichella G, et al. Further investigations<br />

on the expression of HLA-DR+, CD33+ and<br />

CD13+ surface antigens in purified bone marrow and<br />

peripheral blood CD34+ haematopoietic progenitor<br />

cells. Br J Haematol 1993; 84:24-30.<br />

79 . Craig W, Kay R, Cutler R, Lansdorp P. Expression of<br />

THY-1 on human haematopoietic progenitor cells. J<br />

Exp Med 1993; 177:1331-42.<br />

80. Holyoake T, Alcorn M. CD34+ positive hemopoietic<br />

cells: biology and clinical applications. Blood Rev<br />

1994; 8:113-24.<br />

81. Lanza F, Moretti S, Castagnari B, et al. CD34+<br />

leukemic cells assessed by different CD34 monoclonal<br />

antibodies. Leuk Lymphoma 1995 (in press).<br />

82. Meckenstock G, Heyll A, Schneider E, et al. Acute<br />

leukemia coexpressing myeloid, B- and T-lineage associated<br />

markers: multiparameter analysis of criteria<br />

defining lineage commitment and maturational stage<br />

in a case of undifferentiated leukemia. Leukemia<br />

1995; 9:260-4.<br />

83. Mayer RJ, Davis RB, Schiffer CA, et al. Intensive<br />

postremission chemotherapy in adults with acute<br />

myeloid leukemia. N Engl J Med 1994; 331:896-903.<br />

84. Cassileth PA, Linch E, Hines JD, et al. Varying intensity<br />

of postremission therapy in acute myeloid leukemia.<br />

Blood 1992; 79:1924-30.<br />

85. Cassileth PA, Nowell PC, Larson RA. Acute leukemia.<br />

In: McArthur JR, Schrier S, eds. Hematology - 1993.<br />

St. Louis: The American Society of Hematology,<br />

1993:38-48.<br />

86. Clift RA, Buckner CD, Appelbaum FR, et al. Allogeneic<br />

bone marrow transplantation in patients with acute<br />

myeloid leukemia in first remission: a randomized trial<br />

of two irradiation regimens. Blood 1990; 76:1867-<br />

71.<br />

87. Young JW, Papadopoulos EB, Cunningham I, et al. T-<br />

cell-depleted allogeneic bone marrow transplantation<br />

in adults with acute nonlymphocytic leukemia in first<br />

remission. Blood 1992; 79:3380-7.<br />

88. Bortin MM, Horowitz MM, Rowlings PA, et al. 1993<br />

Progress report from the International Bone Marrow<br />

Transplant Registry. Bone Marrow Transplant 1993;<br />

12:97-104.<br />

89. Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment<br />

of T-cell-depleted haploidentical three-loci<br />

incompatible transplants in leukemia patients by<br />

addition of recombinant human granulocyte colonystimulating<br />

factor-mobilized peripheral blood progenitor<br />

cell to bone marrow inoculum. Blood 1994;<br />

84:3948-55.<br />

90. Cassileth PA, Andersen J, Lazarus HM, et al. Autologous<br />

bone marrow transplant in acute myeloid<br />

leukemia in first remission. J Clin Oncol 1993; 11:314-<br />

9.<br />

91. Gorin NC, Labopin M, Meloni G, et al. Autologous<br />

bone marrow transplantation for acute myeloblastic<br />

leukemia in Europe: further evidence of the role of<br />

marrow purging by mafosfamide. Leukemia 1991;<br />

5:896-904.<br />

92. Zittoun RA, Mandelli F, Willemze R, et al. Autologous<br />

or allogeneic bone marrow transplantation compared<br />

with intensive chemotherapy in acute myelogenous<br />

leukemia. N Engl J Med 1995; 332:217-23.<br />

93. To LB, Russel J, Moore S, et al. Residual leukemia cannot<br />

be detected in very early remission peripheral<br />

blood stem cell collection in acute non-lymphoblastic<br />

leukemia. Leuk Res 1987; 11:327-9.<br />

94. Juttner CA, To LB, Haylock DN, et al. Circulating<br />

autologous stem cells collected in very early remission<br />

from acute non-lymphoblastic leukemia produce<br />

prompt but incomplete hemopoietic reconstitution<br />

after high dose melphalan or supralethal chemoradiotherapy.<br />

Br J Haematol 1985; 61:739-45.<br />

95. Nagafuji K, Harada M, Takamatsu Y, et al. Evaluation<br />

of leukemic contamination in peripheral blood stem<br />

cell harvests by reverse transcriptase polymerase chain<br />

reaction. Br J Haematol 1993; 85:578-83.<br />

96. Brenner MK, Rill DR, Moen RC, et al. Gene-marking<br />

to trace the origin of relapse after autologous bonemarrow<br />

transplantation. Lancet 1993; 341:85-6.<br />

97. Gorin NC, Dicke K, Lowenberg B. High dose therapy<br />

for acute myelocytic leukemia treatment strategy:<br />

what is the choice? Ann Oncol 1993; 4 (Suppl. 1):59-<br />

80.<br />

98. Bassan R, Barbui T. Remission induction therapy for<br />

adults with acute myelogenous leukemia: towards the<br />

ICE age? <strong>Haematologica</strong> 1995; 80:82-90.<br />

99. Reiffers J, Korbling M, Labopin M, et al. Autologous<br />

blood stem cell transplantation versus autologous<br />

bone marrow transplantation for acute myeloid<br />

leukemia in first complete remission. Int J Cell Cloning<br />

1992; 7(Suppl. 1):111.<br />

100. Korbling M, Fliedner TM, Holle R, et al. Autologous<br />

blood stem cell (ABSCT) versus purged bone marrow<br />

transplantation (pABMT) in standard risk AML: influence<br />

of source and cell composition of the autograft<br />

on hemopoietic reconstitution and disease free survival.<br />

Bone Marrow Transplant 1991; 7:343-9.<br />

101. Sanz MA, de la Rubia J, Sanz GF, et al. Busulfan plus<br />

cyclophosphamide followed by autologous blood<br />

stem cell transplantation for patients with acute<br />

myeloblastic leukemia in first complete remission: a<br />

report from a single institution. J Clin Oncol 1993;<br />

11:1661-7.<br />

102. To LB, Roberts MM, Haylock DN, et al. Comparison<br />

of hematological recovery times and supportive care<br />

requirements of autologous recovery phase peripheral<br />

blood stem cell transplants, autologous bone marrow<br />

transplants and allogeneic bone marrow transplants.<br />

Bone Marrow Transplant 1992; 9:277-84.<br />

103. Siena S, Bregni M, Brando B, et al. Flow cytometry for<br />

clinical estimation of circulating hematopoietic progenitors<br />

for autologous transplantation in cancer<br />

patients. Blood 1991; 77:400-9.<br />

104. To LB, Haylock DN, Dyson PG, Thorp D, Roberts<br />

MM, Juttner CA. An unusual pattern of hemopoietic<br />

reconstitution in patients with acute myeloid leukemia<br />

transplanted with autologous recovery phase peripheral<br />

blood. Bone Marrow Transplant 1990; 6:109-14.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Peripheral blood stem cells in acute myeloid leukemia<br />

31<br />

105. Bender JG, To LB, Williams S, Schwartzberg LS. Defining<br />

a therapeutic dose of peripheral blood stem cells.<br />

J Hematother 1992; 1:329-42.<br />

106. Kotasek D, Sheperd KM, Gage RE, et al. Factors<br />

affecting blood stem cell collections following highdose<br />

cyclophosphamide mobilization in lymphoma,<br />

myeloma and solid tumors. Bone Marrow Transplant<br />

1992; 9:4-17.<br />

107. Laporte JP, Gorin NC, Feuchtenbaum J, et al. Relapse<br />

after autografting with peripheral blood stem cells<br />

[letter]. Lancet 1987; 2:1393.<br />

108. Yan Liu K, Akashi K, Harada M, Takamatsu Y, Niho Y.<br />

Kinetics of circulating haematopoietic progenitors<br />

during chemotherapy-induced mobilization with or<br />

without granulocyte colony-stimulating factor. Br J<br />

Haematol 1993; 84:31-8.<br />

109. Onho R, Tomonaga M, Kobayashi T, et al. Effect of<br />

granulocyte colony-stimulating factor after intensive<br />

induction in relapsed or refractory acute leukemia. N<br />

Engl J Med 1990; 323:871-7.<br />

110. Onho R, Hiraoka A, Tanimoto M, et al. No increase<br />

of leukemia relapse in newly diagnosed patients with<br />

acute myeloid leukemia who received granulocyte<br />

colony-stimulating factor for life-threatening infection<br />

during remission induction and consolidation therapy.<br />

Blood 1993; 81:561-2.<br />

111. Hagenbeek A, Schultz FW, Martens ACM. In vitro or<br />

in vivo treatment of leukemia to prevent relapse after<br />

autologous bone marrow transplantation. In: Dicke<br />

KA, Spitzer G, Jagannath S, eds. Autologous bone<br />

marrow transplantation: Proceedings of the Fourth<br />

International Symposium. MD Anderson Hospital<br />

Publ., 1989:107-12.<br />

112. Gribben JH, Freedman AS, Neuberg D, et al. Immunologic<br />

purging of marrow assessed by PCR before autologous<br />

bone marrow transplantation for B-cell lymphoma.<br />

N Engl J Med 1991; 325:1525-33.<br />

113. Brenner MK, Rill DR, Moen RC, et al. Gene-marking<br />

to trace origin of relapse after autologous bone marrow<br />

transplantation. Lancet 1993; 341:85-6.<br />

114. Gulati SC, Lemoli RM, Acaba L, Igarashi T, Wasserheit<br />

C, Fraig M. Purging in autologous and allogeneic bone<br />

marrow transplantation. Curr Opin Oncol 1992; 4:<br />

543-50.<br />

115. Sharkis SJ, Santos GW, Colvin MO. Elimination of<br />

acute myelogenous leukemia cells from marrow and<br />

tumor suspensions in the rat with 4-hydroperoxycyclophosphamide.<br />

Blood 1980; 55:521-3.<br />

116. Siena S, Castro-Malaspina H, Gulati SC, et al. Effects<br />

of in vitro purging with 4-hydroperoxycyclophosphamide<br />

on the hematopoietic and microenvironment<br />

elements of human bone marrow. Blood 1985;<br />

65:655-62.<br />

117. Bullock G, Tang C, Tourkina E, et al. Effect of combined<br />

treatment with interleukin-3 and interleukin-6<br />

on 4-hydroperoxycyclophosphamide induced programmed<br />

cell death or apoptosis in human myeloid<br />

leukemia cells. Exp Hematol 1993; 21:1640-4.<br />

118. Skorski T, Kawalec M, Hoser G, Ratajczac M, Gnatowski<br />

B, Kawiak J. The kinetic of immunologic and<br />

hematologic recovery in mice after lethal total body<br />

irradiation and reconstitution with syngeneic bone<br />

marrow cells treated or untreated with mafosfamide<br />

(Asta Z 7654). Bone Marrow Transplant 1988; 3:543-<br />

51.<br />

119. Lemoli RM, Gulati SC. In vitro cytotoxicity of VP-16-<br />

213 and nitrogen mustard: agonistic on tumor cells<br />

but not on normal human bone marrow progenitors.<br />

Exp Hematol 1990; 18:1008-12.<br />

120. Lemoli RM, Gasparetto C, Scheinberg DA, et al.<br />

Autologous bone marrow transplantation in acute<br />

myelogenous leukemia: in vitro treatment with<br />

myeloid-specific monoclonal antibodies and drugs in<br />

combination. Blood 1991; 77:1829-36.<br />

121. Lemoli RM, Igarashi T, Knizewski M, et al. Dye-mediated<br />

photolysis is capable of eliminating drug-resistant<br />

tumor cells. Blood 1993; 31:793-800.<br />

122. Chang J, Morgenstern GR, Coutinho LH, et al. The<br />

use of bone marrow cells grown in long-term culture<br />

for autologous bone marrow transplantation in acute<br />

myeloid leukemia: an update. Bone Marrow Transplant<br />

1989; 4:5-10.<br />

123. Rowley SD, Jones RJ, Piantadosi S, et al. Efficacy of ex<br />

vivo purging for autologous bone marrow transplantation<br />

in the treatment of acute non lymphoblastic<br />

leukemia. Blood 1989; 74:501-6.<br />

124. Miller CB, Zenhbauer BA, Piantadosi S, Rowley SD,<br />

Jones RJ. Correlation of occult clonogenic leukemia<br />

drug sensitivity with relapse after autologous bone<br />

marrow transplantation. Blood 1991; 78:1125-31.<br />

125. Gorin NC, Aegerter P, Auvert P, et al. Autologous<br />

bone marrow transplantation for acute myelocytic<br />

leukemia in first remission. A European survey of the<br />

role of marrow purging. Blood 1990; 75:1606-14.<br />

126. Labopin M, Gorin NC. Autologous bone marrow<br />

transplantation in 2502 patients with acute leukemia<br />

in Europe: a retrospective study. Leukemia 1992; 6<br />

(Suppl. 4):95-9.<br />

127. Gulati SC, Acaba L, Yahalom J, et al. Autologous bone<br />

marrow transplantation for acute myelogenous<br />

leukemia using 4-hydroperoxycyclophosphamide and<br />

VP-16 purged bone marrow. Bone Marrow Transplant<br />

1992; 10:129-34.<br />

128. Laporte JP, Douay L, Lopez M, et al. One hundred<br />

twenty-five adult patients with primary acute leukemia<br />

autografted with marrow purged with mafosfamide:<br />

a 10-year single institution experience. Blood 1994;<br />

84:3810-8.<br />

129. Roberts AW, Metcalf D. Noncycling state of peripheral<br />

blood progenitor cells mobilized by granulocyte<br />

colony-stimulating factor and other cytokines. Blood<br />

1995; 86:1600-5.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


eview<br />

Peripheral blood stem cell<br />

transplantation for the treatment<br />

of multiple myeloma:<br />

biological and clinical implications<br />

haematologica 2000; 85(supplement to no. 12):32-48<br />

FEDERICO CALIGARIS CAPPIO, MICHELE CAVO,<br />

ARMANDO DE VINCENTIIS, LUIGI LANATA, ROBERTO MASSIMO<br />

LEMOLI, IGNAZIO MAJOLINO, CORRADO TARELLA, PAOLA ZANON,<br />

SANTE TURA<br />

Cattedra di Immunologia Clinica, Dipartimento di Scienze<br />

Biomediche e Oncologia Umana, Università di Torino, Turin;<br />

Istituto di Ematologia ed Oncologia Medica“Lorenzo e Ariosto<br />

Seràgnoli”, Università di Bologna, Bologna; Dompé Biotec<br />

SpA, Milan; Dipartimento di Ematologia, Unità Trapianti di<br />

Midollo Osseo, Ospedale Cervello, Palermo; Divisione Universitaria<br />

di Ematologia, Università di Torino, Azienda<br />

Ospedaliera S. Giovanni, Turin; Amgen Italia SpA, Milan; Italy<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia ed Oncologia<br />

Medica “Seràgnoli”, Policlinico S. Orsola, via Massarenti 9,<br />

40138 Bologna, Italy.<br />

Received January 11, 1996; accepted June 4, 1996.<br />

Acknowledgments: preparation of this manuscript was supported by<br />

grants from Dompé Biotec SpA and Amgen Italia SpA, Milan, Italy.<br />

The aim of this review is to define the role of<br />

peripheral blood stem cell transplantation for the<br />

treatment of multiple myeloma. Therefore, we<br />

first review our present knowledge of this disease and<br />

then analyze the clinical trials based on the use of<br />

autologous bone marrow or peripheral stem cell<br />

transplantation. Optimal methods for peripheral<br />

blood stem cell transplantation will also be discussed.<br />

Myelomagenesis<br />

Multiple myeloma (MM), the prototype plasma cell<br />

malignancy, is characterized by the uncontrolled<br />

accumulation of plasma cells that replace normal<br />

bone marrow (BM) and by the overproduction of<br />

monoclonal immunoglobulins (Ig) and cytokines. A<br />

number of observations provided both by basic sciences<br />

and by clinical investigation allow us to place<br />

the disease and its unusual features in a more coherent<br />

perspective and to discuss new therapeutic<br />

options properly.<br />

Epidemiology<br />

The reported incidence of MM is available for the<br />

years up to 1982 and varies substantially in different<br />

countries. 1 A striking increase in the incidence of MM<br />

has been noticed in the last thirty years and is only<br />

partially 2 explained by amelioration of diagnostic<br />

capabilities. 3 Between 1973 and 1990 an increase of<br />

40% among people over 65 and of almost 15%<br />

among people under 65 has been recorded in US Cancer<br />

Death rates. 4 Ethnic differences are apparent: the<br />

incidence is twice as high and the mortality rate has<br />

quadrupled in blacks, while doubling in whites. 4 By<br />

contrast, rates among Asians are lower than those of<br />

whites living in the same geographic area. 5<br />

Both genetic and environmental factors can be<br />

invoked to explain these ethnic differences. A significant<br />

increase has been detected in first-degree relatives<br />

of patients. 5 Moreover, an increased risk has<br />

been observed to be associated with occupational<br />

and environmental elements that include farming<br />

exposure to pesticides, exposure to ionizing radiations,<br />

petroleum and rubber processing, as well as<br />

persistent (viral) infections. 3 The main conclusion that<br />

can be drawn from a large body of observations is<br />

the necessity of discriminating the genetic roots from<br />

the environmental links of the disease. As a corollary,<br />

it may be asked which elements (genetic vs. environmental)<br />

are associated with the development of monoclonal<br />

gammopathy of undetermined significance<br />

(MGUS) and how they relate to the progression of<br />

MGUS to overt MM.<br />

Cytogenetics and molecular biology<br />

Two major pieces of information have emerged<br />

from cytogenetic studies. The first is that no consistent<br />

(yet not random) chromosome abnormalities<br />

have been detected in MM. 6 The second is that<br />

numeric chromosome abnormalities are shared by<br />

MGUS and MM. 7,8 Both facts lead us to ask what the<br />

prerequisite is and what the additional events are in<br />

the development of plasma cell malignancies. We still<br />

do not know the prerequisite events that lead to<br />

MGUS, to MM or to the evolution of MGUS into MM,<br />

or how they differ from collateral elements that simply<br />

favor the malignant process. Along the same vein,<br />

it is interesting that no known specific oncogene has<br />

yet been related to the development of MM or to the<br />

transition from MGUS to overt MM. The genes most<br />

commonly implicated in MM, like N-RAS, P53 and<br />

retinoblastoma gene (RB), are all involved in the late<br />

stages of the disease. 9<br />

If the same cytogenetic abnormalities are shared by<br />

two clinical situations as different as MGUS and overt<br />

MM, a patrolling role for the immune system can be<br />

envisaged in the natural history of plasma cell disorders.<br />

It is not unreasonable to suspect that if the<br />

immune system is able to keep a malignant clone<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

33<br />

under control, a benign MGUS is the resulting disease;<br />

the breakdown of this control would lead to MM. Little<br />

direct, but much indirect evidence is available in murine<br />

models to suggest the immunomodulation of myeloma<br />

cell growth by host effector cells. 10<br />

Immunochemistry and B cell differentiation<br />

studies<br />

Three major findings have been obtained through<br />

immunochemistry and by a more proper understanding<br />

of the differentiation processes of B lineage cells. First,<br />

MM paraproteins may be directed against a wide variety<br />

of infectious agents, suggesting that MM development<br />

and antigen (Ag) stimulation may be causally related.<br />

11-13 Second, the Ig isotype of MM plasma cells is generally<br />

IgG or IgA, demonstrating that the predominant<br />

phenotype of MM tumor cells is post-switch. 9 Third,<br />

clonal proliferation involves a cell population that has<br />

already passed through the stage of Ig genes somatic<br />

hypermutation. 14,15 Since this process occurs in the germinal<br />

centers (GC) of secondary follicles, 16 its presence<br />

is a clear marker of the differentiative and functional<br />

level reached by the cell population being analyzed.<br />

By and large, the observation that MM is a neoplasm<br />

of plasma cells that have a post-switch phenotype,<br />

show somatic mutations and may produce monoclonal<br />

Ig with targeted antibody (Ab) activity leads to the conclusion<br />

that MM is an Ag-driven process, even if the<br />

specific causal Ag is generally unknown. This assumption<br />

has to be confronted with the simple, though basic,<br />

lesson from clinical medicine that MM is a BM disorder.<br />

In contrast with the distribution of normal plasma cells,<br />

MM plasma cells localize uniquely within the BM. 9<br />

Although the lamina propria of the intestine contains<br />

more Ig-producing cells than all other tissues in the<br />

body, it is never a site where MM develops, not even<br />

IgA1- and IgA2-producing MM. 17 Likewise, involvement<br />

of the spleen and/or lymph nodes, though typical of<br />

Waldenström’s macroglobulinemia, is very unusual in<br />

MM. 17 The exclusive BM localization of MM plasma cells<br />

appears to conflict with the extensive somatic hypermutations<br />

of the Ig they produce, which indicate a<br />

peripheral origin of malignant cells. However, while the<br />

steps of Ag processing and presentation that lead to<br />

the generation of somatically mutated IgG and IgA plasma<br />

cells occur only in secondary lymphoid follicles, the<br />

BM is a major site of IgG and IgA production in T-celldependent<br />

secondary immune responses. 18-20 Plasma cell<br />

precursors with specific traffic commitments originate<br />

from secondary lymphoid organs and migrate to the BM<br />

a few days after the Ag challenge (Figure 1). 20,21<br />

The issue whether MM plasma cell precursors are early<br />

BM stem cells or late peripheral B cells is misleading.<br />

The cell whose original transformation has ultimately<br />

generated the malignant plasma cell progeny that we<br />

see in MM cannot be equated with the B cell population<br />

that disseminates the disease throughout the axial<br />

skeleton. 21 The identity of the hypothetical MM stem<br />

cell is unknown, i.e. we do not know either the cellular<br />

target of the primary transforming event or where,<br />

when and how the unknown cellular target was hit by<br />

the transforming event. By contrast, the information<br />

available on the B cell population that feeds the downstream<br />

compartment of plasma cells and disseminates<br />

the disease indicates that this population has been generated<br />

in peripheral lymphoid organs during secondary<br />

T-cell-dependent Ab response, is programmed to home<br />

to the BM, and is committed to differentiate in close<br />

association with the BM microenvironment (Figure<br />

2). 21,22 On the basis of existing data, the most likely candidate<br />

for the physiological B lymphocyte equivalent of<br />

the MM plasma cell precursor is either a B memory cell<br />

or a plasma blast (Figure 1). 14,15,23,24<br />

Microenvironment and cytokines<br />

It is assumed that BM-seeking plasma cell precursors<br />

receive a differentiation signal after contact with the<br />

BM stromal microenvironment (Figure 2). 25,26 Microenvironmental<br />

stromal cells play an essential role in the<br />

growth of plasma cell tumors both in mice 27 and in<br />

humans. 28 MM BM stromal cells are well equipped with<br />

a large series of adhesion and extracellular matrix molecules<br />

that mediate homotypic and heterotypic interactions<br />

and provide anchorage sites to cells selectively<br />

exposed to locally released growth factors. 22,29,30 MM<br />

BM stromal cells produce cytokines like IL-6 known to<br />

play a crucial role in the evolution of the disease both<br />

in experimental systems, including IL-6 transgenic mice,<br />

extracellular matrix proteins<br />

B memory cell<br />

Stromal cell<br />

plasma blast<br />

centroblast<br />

stromal cell released cytokines (IL-6)<br />

Plasma cell<br />

BONE MARROW<br />

B memory cells<br />

centrocyte<br />

LYMPHOID FOLLICLES<br />

Figure 1. Plasma cell precursors generated in peripheral<br />

lymphoid organs differentiate in contact with bone marrow<br />

stromal cells.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


34<br />

F. Caligaris Cappio et al.<br />

Precursors<br />

Macrophages<br />

Fibroblasts<br />

T lymphocytes<br />

Bone Marrow<br />

IL-1, TNF-, M-CSF<br />

IL-1, IL-3, GM-CSF,<br />

TNF<br />

IL-6, IL-7,<br />

stem cell factor<br />

Microenvironment cells<br />

Osteoclasts<br />

Plasma cells<br />

Reciprocal<br />

Activation<br />

Figure 2. Model of multiple myeloma growth and progression<br />

based upon a series of Fig. mutual 2 interactions between the<br />

B-cell clone and the bone marrow microenvironment.<br />

and in vivo. 31-34 High levels of IL-6 are observed in the<br />

sera of patients with aggressive or progressive MM, 35<br />

and infusion of anti-IL-6 antibodies in patients with<br />

plasma cell leukemia or MM refractory to therapy has<br />

decreased the size of the plasma cell pool and hampered<br />

the proliferative activity of plasma cells. 36<br />

Malignant MM plasma cells are not inert vehicles of<br />

monoclonal Ig. They also produce a number of cytokines,<br />

including interleukin (IL)-1b, tumor necrosis factor<br />

(TNF)- and monocyte-macrophage colony stimulating<br />

factor (M-CSF), that activate stromal and accessory<br />

cells, aa well as having significant osteoclast activating<br />

factor (OAF) activity. 22,37 A minority of human MM cell<br />

lines autonomously produce small amounts of IL-6, but<br />

it is unclear whether fresh MM plasma cells can also<br />

produce IL-6. 34 IL-6, besides promoting B cell proliferation<br />

and differentiation, has recently been shown to<br />

have important OAF activity. 32,33<br />

These experimental findings linked to clinical observations<br />

lead to the attractive hypothesis (Figure 2) that<br />

a self-maintaining series of mutual interactions between<br />

the malignant B cell clone and the BM microenvironment<br />

may explain the progression of MM 22 through the<br />

production of ever-increasing amounts of cytokines<br />

capable of recruiting and activating several microenvironmental<br />

cells, including osteoclasts.<br />

The role of autologous transplantation<br />

in the treatment of multiple myeloma<br />

Investigations into the use of myeloablative therapy<br />

for the management of MM were pionereed in the mid-<br />

1980s and were stimulated by a persistent lack of<br />

progress in prognosis with conventional chemotherapy.<br />

38,39 As is the case with any experimental approach,<br />

initial trials were restricted to the treatment of patients<br />

with advanced refractory or relapsing disease and were<br />

focused mainly on defining the feasibility and toxicity of<br />

the procedure. These preliminary experiences were performed<br />

without the support of hemopoietic stem cells<br />

and demonstrated that high-dose melphalan (HDM), given<br />

intravenously (i.v.) at doses ranging between 100 and<br />

140 mg/m 2 , yielded an increase in the complete remission<br />

(CR) rate, albeit at the expense of prolonged marrow<br />

aplasia and an unacceptably high early mortality<br />

rate. 40-42 On the basis of these observations later studies<br />

with chemotherapeutic agents administered at myeloablative<br />

doses, and possibly added total body irradiation<br />

(TBI), were carried out with the support of autologous<br />

BM and/or peripheral blood hemopoietic stem cells<br />

(PBSC). 43 Demonstration of the safety and relative efficacy<br />

of autotransplants in refractory MM 41,44-46 encouraged<br />

subsequent application of this procedure in earlier<br />

phases of the disease 45,46 and, more recently, in newly<br />

diagnosed patients as well. 47,48 Over the past decade<br />

interest in this new treatment strategy has progressively<br />

grown, and the number of reported patients receiving<br />

autologous hemopoietic stem cell-supported myeloablative<br />

therapy is now approximately one thousand<br />

worldwide.<br />

What lessons have we learned from this collective<br />

experience? It is difficult to draw firm conclusions from<br />

published trials since none of them were controlled and<br />

patient populations were different, as were the preparative<br />

treatments and the criteria used for evaluating<br />

tumor response. In addition, the bias introduced by<br />

patient selection and, in most of the cases, the lack of<br />

an adequate follow-up also helped complicate correct<br />

interpretation of the data. As a consequence, the exact<br />

role of autotransplantation in the management of MM<br />

still remains poorly defined and could be properly<br />

addressed only in controlled clinical studies comparing<br />

autografting and conventional chemotherapy. There are<br />

at least several such trials in progress at the moment in<br />

Europe and the United States. Data reported at the last<br />

ASH meeting in Seattle (1995) by the Intergroupe<br />

Français du Myelome are promising and suggest an<br />

advantage for autografted patients in terms of increased<br />

CR rate and extended survival duration. 49<br />

Obviously, these results warrant confirmation in larger<br />

independent series. For this reason, similar investigations<br />

are currently being conducted in the United<br />

States under the auspices of the National Cancer Institute.<br />

While the conclusions of these studies are being<br />

awaited, analyses of available transplant data have provided<br />

the following important information.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

35<br />

Group No. % Source % % Median mos.<br />

pts. sens. % BM % PB ED CR PFS Surv.<br />

EBMT 130 68 63 25 6 48 17 27<br />

Univ. Arkansas (USA) 287 60 unknown


36<br />

F. Caligaris Cappio et al.<br />

Remission duration<br />

As previously emphasized, myeloablative therapy<br />

requiring autologous hemopoietic stem cell support provides<br />

substantial antitumor response, especially in<br />

patients with good prognosis (see below). However, even<br />

in this favorable condition, a considerable relapse rate,<br />

approaching 60% at 3 years, is reported after autotransplant<br />

and no plateau is yet apparent on relapse-free<br />

survival curves. 50-52 These results contrast with the 30%<br />

probability of long-term unmaintained remissions (and<br />

possible cures) reported by several groups for patients<br />

receiving allogeneic transplantation. 60 It has been suggested<br />

that the lack of an immunological effect by the<br />

donor’s marrow T lymphocytes on the residual myeloma<br />

cells (i.e. graft-versus myeloma) 61,62 and/or possible<br />

tumor reseeding may account for the apparently less<br />

durable duration of disease control following autologous<br />

as opposed to allogeneic transplantation. For this<br />

reason, important issues currently under clinical investigation<br />

in the autografting setting include further<br />

increases in the cytotoxic dose intensity level and depletion<br />

of tumor cells from the graft (see below).<br />

Prognostic variables<br />

Several important variables affecting the outcome of<br />

autologous transplantation have been identified (Table<br />

3), including 2 -microglobulin ( 2 -M) levels, 45,47,50-52,56<br />

pre-transplant disease status, 45,51,52 age, 45,51,52 performance<br />

status, 45 Ig isotype 45,51,52 and response to myeloablative<br />

therapy (e.g. attainment or non-attainment of<br />

CR). 47,48 In particular, at multivariate regression analysis<br />

early mortality was reported to be highest among resistant<br />

relapsing patients, who also had the poorest<br />

response to myeloablative therapy and the shortest<br />

relapse-free survival duration. 45 In contrast, low serum<br />

2 -M levels, both at diagnosis and before autografting,<br />

and prior responsiveness to conventional chemotherapy<br />

conferred the highest CR rate, as well as prolonged<br />

relapse-free and overall survival durations. 45,47,50-52,56 In<br />

addition, the timing of autotransplant also emerged as an<br />

important and independent prognostic parameter. 56,64<br />

This observation, on the one hand, was related to the<br />

generally reported improved outcome of patients transplanted<br />

earlier and, on the other hand, reflected the<br />

acquisition of multiple biological abnormalities in<br />

advanced phases of the disease 63 that ultimately led to<br />

refractoriness even to high-dose therapy. 64 Conversely,<br />

retaining sensitivity to high-dose therapy in earlier phases<br />

of MM assured better results, even in patients with<br />

primary refractory disease. 45,47,65<br />

New perspectives under clinical investigation<br />

Based on the assumption that the failure of the conditioning<br />

regimen to eradicate the myeloma clone contributes<br />

most to post-transplant relapse, attempts to<br />

increase the intensity, and possibly the efficacy, of<br />

treatment by means of repeated courses of myeloablative<br />

therapy have recently been undertaken. 46,53 The<br />

more rapid recovery of hemopoiesis assured by the<br />

combined use of PBSC and post-transplant administration<br />

of hemopoietic growth factors 59 made the double<br />

transplant strategy feasible for approximately 60%<br />

of patients within one year. 53 Results of pilot trials in<br />

primary refractory MM indicated that such an approach<br />

provided superior antitumor effect with improved<br />

event-free and overall survival durations with respect<br />

to a single transplant. 53<br />

A controlled clinical study comparing in a randomized<br />

fashion single vs. double autografting in newly<br />

diagnosed patients is currently underway in France. A<br />

similar trial is already in the early accrual stage in Italy.<br />

These studies will clarify in the next several years<br />

whether double transplant is associated with better<br />

prognosis. Alternatively, efforts to improve the clinical<br />

impact of autotransplant have been carried out by several<br />

groups and have included depletion of tumor cells<br />

from autografts by both negative selection of myeloma<br />

cells and positive selection of CD34 + hemopoietic<br />

stem cells, 66,67 as well as post-transplant immunomodulation<br />

with interferon- (IFN-). 47,49,68<br />

In summary, hemopoietic stem cell-supported myeloablative<br />

therapy holds the promise of being a safe and<br />

effective treatment modality for MM. It yields better<br />

overall response and CR rates than conventional<br />

chemotherapy and may prolong the duration of survival.<br />

49<br />

These conclusions, while encouraging, have been<br />

drawn mainly from uncontrolled studies carried out in<br />

select groups of patients and obviously warrant confirmation<br />

in controlled clinical trials which are currently<br />

under way. Therefore the next several years will clarify<br />

whether newly diagnosed patients with symptomatic<br />

MM can be routinely offered a single or double autotransplant<br />

as first-line or early salvage therapy for their<br />

disease.<br />

Table 3. Variables affecting the outcome of autotransplants<br />

for multiple myeloma. @<br />

Disease status<br />

Variable Refractory Refractory + Responsive<br />

CR RFS Surv. CR RFS Surv.<br />

Low 2 M – +* +* + +* +*<br />

Early transplant + + + + +* +*<br />

CR achievement +* +*<br />

Double transplant +* +*<br />

CT responsiveness + + +<br />

Younger age – + + + + +<br />

Non IgA isotype – + + – + +<br />

*In multivariate analyses. Abbreviations: CT, conventional chemotherapy;<br />

RFS, relapse-free survival. @ Ref.: 45,47,48,50,51,52,56,63,64,65.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

37<br />

While the results of these studies are being awaited,<br />

wider application of myeloablative therapy should probably<br />

be encouraged. Less heavily pretreated patients<br />

who did not respond to prior conventional chemotherapy<br />

are more likely to benefit primarily from autotransplant.<br />

In addition, data available from the literature do<br />

suggest that a superior outcome of this procedure can<br />

be anticipated in patients with chemosensitive disease<br />

and low tumor burden at diagnosis. Hence, ongoing<br />

clinical trials aimed at comparing conventional versus<br />

myeloablative therapy will also address the important<br />

issue of the role of autotransplant as early consolidation<br />

therapy in patients with intrinsically good prognosis.<br />

However, even in this favorable situation, recurrence<br />

of the underlying malignant disease remains a major<br />

problem and is the most common cause of treatment<br />

failure. For this reason, attempts to improve the clinical<br />

impact of autografting are under active clinical<br />

investigation.<br />

In addition, many other problems regarding autologous<br />

transplantation for MM are still unresolved and<br />

should be formally addressed in future clinical trials.<br />

The most important of these issues include the choice<br />

of the best conditioning regimen, the optimal source of<br />

hemopoietic stem cells, the nature of relapse after autografting,<br />

the benefit from purging techniques and, finally,<br />

the likelihood of long-term disease control, especially<br />

for patients with molecularly defined CR. 69<br />

Advantages offered by the use of<br />

PBSCs in the treatment of multiple<br />

myeloma<br />

The use of PBSC in support of high-dose chemoradiotherapy<br />

(peripheral blood stem cell transplantation)<br />

(PBSCT) is a valid alternative to autologous bone<br />

marrow transplantation (ABMT) in the treatment of<br />

both hematologic and non-hematologic neoplastic disorders.<br />

70-72 The growing interest in this procedure can be<br />

explained by: i) the possibility of mobilizing and collecting<br />

large amounts of hemopoietic progenitors, 73,74<br />

and ii) the rapid hemopoietic recovery observed following<br />

PBSCT. 70,71,74-78<br />

Progenitor collection represents the critical step in the<br />

procedure. Daily monitoring of circulating CD34 + cells is<br />

an essential assay in predicting the number and timing<br />

of leukaphereses. 79,80 Under proper conditions, only a few<br />

leukapheresis procedures are required to collect enough<br />

progenitor cells for marrow reconstitution after myeloablative<br />

treatments. Indeed, when circulating CD34 +<br />

cells rise to >50/µL, 1-2 leukaphereses may yield more<br />

than 5010 4 /CFU-GM/kg or 810 6 /CD34 + cells/kg,<br />

which are considered the ideal values for optimal<br />

engraftment. 80-82 In addition, it has been shown that<br />

large quantities of very immature elements, identified as<br />

long-term culture-initiating cells (LTC-IC), are mobilized<br />

as well. 83-85<br />

Inclusion in the harvested material of very immature<br />

elements is responsible for the stable and durable marrow<br />

reconstitution observed in patients autografted with<br />

circulating progenitors. 77,83 Thus the term PBSC, now<br />

commonly employed to identify mobilized hemopoietic<br />

progenitors, relies on both biological and clinical observations.<br />

As previously emphasized, the rapidity of<br />

engraftment is the major advantage offered by PBSC.<br />

Nevertheless, some authors argued that BM cells stimulated<br />

by growth factor administration might be at least<br />

as efficient as mobilized progenitors in ensuring rapid<br />

engraftment following myeloablative treatment. 87-89<br />

However, it has recently been shown that both committed<br />

and early progenitors are by far more frequent<br />

in PB than in BM during maximal mobilization. 90 This<br />

conclusive observation points toward the preferential<br />

use of PBSC as the hemopoietic cell source for grafting<br />

purposes.<br />

Since its introduction into clinical practice, PBSCT<br />

has been considered a promising approach for MM<br />

patients. 91,92 Several studies have been designed in the<br />

last few years.<br />

Reported results have shown a significant decrease in<br />

hemopoietic toxicity following this procedure as compared<br />

to ABMT, with recovery of granulocytes >0.510 9 /L<br />

and plateles >25-3010 9 /L within approximately 2 weeks<br />

after autograft (Table 4) 42,44-48,52,54,56,93-98 This was paralleled<br />

by rather good tolerability with rare early fatal<br />

events. 52,56,97,98<br />

In addition, hemopoietic reconstitution by PBSC<br />

seems to be long lasting. MM patients may require<br />

repeated exposure to high-dose cytotoxic therapy.<br />

Reducing hemopoietic toxicity might be critical for the<br />

ultimate treatment outcome. Therefore, also for its<br />

long-term effect, PBSCT may have a positive impact on<br />

the life expectancy of those patients who are suitable<br />

for intensified chemo-radiotherapy treatments. 99<br />

PBSC mobilization and collection in<br />

multiple myeloma<br />

PBSC mobilization in myeloma patients<br />

PBSC collection presents specific problems in patients<br />

with MM, where a decrease of progenitors in the bone<br />

marrow is due in part to a defect of the monocyte/macrophage<br />

activation pathway. In fact, CD34 + cells<br />

from MM patients grow normal numbers of colonies<br />

when stimulated by normal monocytes, while normal<br />

CD34 + cells have a reduced growth rate with MM monocytes.<br />

100 Another aspect is prior treatment. Repeated<br />

courses of chemo-radiotherapy are able to exhaust the<br />

pool of pluripotent stem cells, 101 resulting in insufficient<br />

progenitor cell harvests. 59,102-105 Studies specifically<br />

addressed at MM patients show that melphalan 106 and<br />

treatment-free interval prior to PBSC mobilization 107 also<br />

have an influence on the release of progenitors into the<br />

peripheral blood, while the value of BM plasmacytosis as<br />

an independent factor is more questionable. 108,109 As a<br />

consequence of these and other unknown factors, progenitor<br />

yields in MM are often unpredictable and lower<br />

than those observed in other malignant disorders. 110<br />

haematologica vol. 85(suppl. to n. 12):December 2000


38<br />

F. Caligaris Cappio et al.<br />

Time to recovery from°<br />

Intensified treatments* leukopenia thrombocytopenia Treatment-related References<br />

(days) (days) deaths (%)<br />

Without autograft 28 # 27 17 42,46,54,93-95<br />

With BMT 20 26 7 44,45,47,48,96<br />

With PBSCT 14 18 3.7 52,56,97,98<br />

Table 4. Toxicity of intensified<br />

treatments with or without<br />

autologous stem cell support in<br />

multiple myeloma patients.<br />

*intensified treatments consisted of HDM (60-200 mg/sqm) in most studies; the association of TBI/HDM was also used in some programs with autograft;<br />

°time to recovery from leukopenia and thrombocytopenia was reported as days to reach >0.5x10 9 ANC/µL and > 25x10 9 platelets/L, respectively,<br />

in nearly all studies; # table data have been calculated as medians from median values of hemopoietic recovery and from percentages of treatmentrelated<br />

deaths reported in each quoted study.<br />

Nonetheless, cell harvests sufficient for one or two subsequent<br />

autografts are usually obtained, 59,97,108,111-113 even<br />

in patients with markedly infiltrated marrow or primary<br />

resistant disease. 109 To avoid the adverse influence of premobilization<br />

treatment, PBSC collection in MM patients<br />

should be planned as early as possible in the course of<br />

disease, and alkylating drugs should be omitted in the<br />

primary treatment. It should also be kept in mind that<br />

heavily pre-treated patients require more leukaphereses<br />

and show slower platelet recovery after autograft. 109 The<br />

key issues in the apheretic harvest of PBSCs in MM are<br />

presented in Table 5.<br />

PBSC also may be collected from patients with malignancies<br />

in steady state conditions; 114 however, multiple<br />

aphereses are required with this method. Mobilization<br />

of progenitors with cytotoxic chemotherapy, hemopoietic<br />

growth factors, or a combination of the two is<br />

therefore generally preferred. The hematopoietic recovery<br />

that occurs after cytotoxic chemotherapy is accompanied<br />

by a PBSC rise that is proportional to the intensity<br />

of myelosuppression.<br />

102, 108<br />

In MM, chemotherapy alone with either HDM, 115 or<br />

CHOP-like regimens 112,113,116 or intermediate- to highdose<br />

cyclophosphamide (Cy) 97,117,118 has been used to<br />

mobilize PBSC. However, the failure rate, defined as the<br />

percentage of patients with a low progenitor cell peak<br />

in the blood or poor collections at the end of the<br />

apheresis program, was relatively high, ranging from 20<br />

to 30%. Moreover, when using high-dose therapy protocols<br />

without growth factor support, one should consider<br />

that this implies an undue risk of severe toxicity. 118<br />

G-CSF 73,119-121 and GM-CSF, 75,112 as well as other<br />

cytokines are able to promote a dramatic rise of progenitors<br />

in the circulation. In a study of MM patients,<br />

administration of G-CSF at 10 µg/kg alone for six days<br />

induced a considerable increase in CFU-GM and CD34 +<br />

cells, 111 with rapid recovery of counts after autograft.<br />

However, the use of growth factors alone in patients<br />

with neoplastic disorders produces little enthusiasm<br />

among hematologists. In fact, the spike of progenitor<br />

cells can be further amplified by combining growth factors<br />

with chemotherapy. 71 Together with the demonstration<br />

that tumor cells are also mobilized by growth<br />

factors, 123 this fact makes the combination of<br />

chemotherapy with G-CSF or GM-CSF the most reliable<br />

approach. 86,109,112,113<br />

In MM as in other diseases, 74,77 the use of growth factors<br />

following cytotoxic treatment proves to be superior<br />

to chemotherapy alone in terms of progenitor cell<br />

yield, 108,112 and significantly contributes to minimizing<br />

treatment toxicity. 112,124 High progenitor peak levels are<br />

reported 108 with high-dose chemotherapy, namely Cy at<br />

7 g/m 2 or etoposide (VP16) at 2 g/m 2 followed by G-CSF<br />

or GM-CSF, and results seem to compare favorably with<br />

intermediate-dose Cy with or without G-CSF or GM-<br />

CSF. In conclusion, the optimal schedule for PBSC mobilization<br />

in MM has not yet been defined, though the<br />

most experience is with Cy at 7 g/m 2 followed by G-CSF<br />

or GM-CSF. A review of the mobilization schedules<br />

reported so far in MM patients is presented in Table 6.<br />

Target of collections and cell monitoring<br />

CD34 + cell number and CFU-GM dose are both reliable<br />

predictors of engraftment time. 125-129 The amount of<br />

PBSC necessary for engraftment is not clearly defined,<br />

but values of 10 to 2010 4 /kg CFU-GM represent a reasonable<br />

minimal dose. 110,120 Irrespective of disease, rapid<br />

neutrophil engraftment has been reported with<br />

2010 4 /kg CFU-GM or 210 6 /kg CD34 + cells. 125,130,131<br />

However, a higher dose may be necessary for rapid and<br />

full platelet engraftment. 105,132 In a recent study of MM<br />

autografts, Tricot et al. 59 found that platelet engraftment<br />

is influenced by previous history and cell dose. In<br />

patients with more than 24 months of chemotherapy<br />

before the autograft, they found a dose ≥510 6 /kg to<br />

be required for rapid and full platelet recovery post<br />

graft. This number of CD34 + cells may be obtained with<br />

1 or 2 apheretic runs, and only a minority of patients,<br />

namely those with prolonged pre-mobilization treatment,<br />

need a higher number of apheretic procedures.<br />

The number of cells needed is obviously greater when a<br />

double autograft is planned. When this is the case, since<br />

recovery after a second autograft is influenced by the<br />

same factors as the first, 59 the number of CD34 + cells to<br />

be collected simply has to be doubled.<br />

CD34 + cell monitoring in blood and collection products<br />

is undoubtely the most reliable and rapid method<br />

for apheresis planning, 131,133-135 though the assay<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

39<br />

Issue<br />

Dysregulated or suppressed hematopoiesis<br />

Methods for mobilization<br />

Toxicity of mobilization therapy<br />

Kinetics of recovery after mobilization<br />

Prediction of harvest<br />

Progenitor cell assays<br />

Target of collections<br />

Apheresis method<br />

Tumor contamination of harvest<br />

Related aspects<br />

Decreased rate of progenitors,* defective monocyte activation,* prior<br />

chemotherapy<br />

Type (and doses) of chemotherapy, use of growth factors<br />

Fever, allergy, infections, thrombosis<br />

Timed and asynchronous use of WBC, monocytes and platelets<br />

Prior chemotherapy, G-CSF test<br />

CD34 + cells, CFU-C<br />

Need for >510 6 /kg CD34 + cells in heavily pre-treated patients*<br />

Cell separator, volume processed, schedule of aphereses<br />

Purging technique<br />

Table 5. Key issues in mobilization<br />

and collection of<br />

PBSCs.<br />

Note. Most aspects are<br />

shared with other malignant<br />

disorders, and only a few<br />

may specifically affect multiple<br />

myeloma. These latter<br />

are marked with an *.<br />

requires skillful personnel and carries a substantial cost.<br />

The issue has been reviewed extensively by Rowley. 136<br />

Siena et al. 133 initially suggested starting the collection<br />

program as soon as CD34 + cells were detectable in the<br />

peripheral blood. However, in terms of efficiency, the<br />

best collections are performed when CD34 + cells are at<br />

their peak. In practice, aphereses should be started as<br />

soon as the CD34 + cells in the blood exceed a given level.<br />

We suggest a value of 20 CD34 + cells/µL combined<br />

with a WBC level >1.010 9 /L and a platelet count<br />

>3010 9 /L before starting collections. 82,133 Mononuclear<br />

cells (MNC) in DNA synthesis also predict a good<br />

yield when their level in the blood is >5% (or<br />

>250/µL). 137<br />

Few studies report detailed data on apheretic PBSC<br />

collection in MM. Dimopoulos et al. 109 began the<br />

aphereses when the MNC count went above 0.310 9 /L,<br />

having as target the collection of > 210 6 /kg CD34 +<br />

cells. They were able to collect >3.010 6 /kg CD34 + cells<br />

daily in patients with ≤ 4 months of prior chemotherapy,<br />

but the mean daily yield was uniformly lower<br />

(< 110 6 CD34 + cells/kg) in patients with more than 12<br />

months of chemo-radiotherapy. Tricot et al. 59 initiated<br />

collections upon recovery of a WBC count > 0.510 9 /L,<br />

and assumed a target of > 63,108/kg MNC to support<br />

two autografts. In a recent study 113 aphereses were<br />

started as soon as the WBC count exceeded 510 9 /L<br />

after a CHOP-like regimen followed by G-CSF, and<br />

>610 6 /kg CD34 + cells were collected from all patients<br />

in 1 to 3 aphereses.<br />

A predictive test with G-CSF, a single dose of 10<br />

mcg/kg, followed by CD34 + cell monitoring on days 4<br />

and 5 has been proposed. 138 The study included patients<br />

with MM, but the sample was too small to draw any<br />

conclusions. Steady-state CD34 + cell counts seem to<br />

predict the yield of PBSCs after mobilization with<br />

chemotherapy and G-CSF, 139 but not after G-CSF<br />

alone. 140 Table 7 shows the first apheresis day reported<br />

with different mobilization methods. 98,108,111,112,117,141 It<br />

is clear that the CD34 + cell peak occurs very early<br />

(approximately day 5 or 6) during mobilization with<br />

growth factors alone. When chemotherapy is included<br />

in the mobilization schedule, the CD34 + cell peak day<br />

occurs later (approximately day 20), but the subsequent<br />

use of growth factors will shorten it by a week or so.<br />

To conclude, we suggest (Table 8) mobilizing PBSC<br />

with the combination of chemotherapy and growth factors<br />

(G-CSF or GM-CSF), and performing serial determinations<br />

of CD34 + cells in the blood. Aphereses should<br />

be started as soon as the level of CD34 + cells exceeds<br />

20/µL, and collections should be performed daily with<br />

twice the blood volume processed each time. Continuous-flow<br />

separators are to be preferred. As target for<br />

collections, the figure of 210 6 /kg CD34 + cells per single<br />

autograft should be adopted for patients with < 24<br />

months of prior chemotherapy, while a greater number<br />

(> 510 6 /kg) should be collected in patients with a<br />

longer treatment history.<br />

Assessment of myeloma cells in the<br />

peripheral blood and role of ex vivo<br />

purging<br />

PBSC collections are generally believed to have lower<br />

tumor cell contamination than BM harvests in cancer<br />

patients eligible for autografting. Moreover, the use<br />

of circulating progenitor cells has shown more rapid<br />

hematopoietic reconstitution than reinfusion of BMderived<br />

cells, thus reducing the incidence of serious<br />

infections and virtually eliminating mortality. 142 Consequently,<br />

PBSCT is widely used after myeloablative therapy<br />

for the treatment of myeloma patients. 53,56,59 However,<br />

myeloma-related B-cells bearing the same idiotypic<br />

determinant as the neoplastic plasma cells have<br />

been identified in the blood of MM patients under<br />

steady-state conditions, 143-149 and they may play a crucial<br />

role in the pathogenesis of the disease. 144,147 Therefore<br />

in this chapter we will review the published data<br />

concerning: i) the presence of MM elements in PB and<br />

their kinetics in response to mobilization protocols; ii)<br />

methods for myeloma cell assessment; iii) methods for<br />

ex vivo removal of contaminating tumor cells and the<br />

role of purging with respect to disease relapse.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


40<br />

F. Caligaris Cappio et al.<br />

Table 6. PBSC mobilization schedules in multiple myeloma.<br />

Authors No. pts Treatment Growth factor Day of Peaked Peaked Notes<br />

progenitor peak CD34 + /µL CFU-GM/mL<br />

Reiffers 117 15 Cy 7 g/m 2 no nr nr nr 5/13 failures<br />

Jagannath 97 36 Cy 6 g/m 2 no nr nr nr better with GM-CSF<br />

39 Cy 6 g/m 2 GM-CSF 17 nr nr<br />

Tarella 108 11 Cy 7 g/m 2 or VP16 2 g/m 2 GM-CSF 15 (13-16) 126 6432<br />

4 Cy 21.2 g/m 2 no 16 (16-18) 31 462<br />

4 Cy 21.2 g/m 2 GM-CSF 14 (14-15) 77 2588<br />

Ossenkoppele 111 6 no G-CSF6 gg 6 845<br />

Majolino 112 7 VCAD no 20 (17-30) 622<br />

7 VCAD G-CSF 13 (9-17) 22 893<br />

Vasta 113 6 VCED G-CSF 13 (12-15) 70 2391<br />

Legend. Cy: cyclophosphamide; VCAD: vincristine 1 mg, cyclophosphamide 4x500 mg/m 2 , adriamycin 2x50 mg/m 2 , dexamethasone 4x40 mg. VCED was identical to VCAD except that epirubicin 2x60 to 80<br />

mg/m 2 was substituted for adriamycin. nr: not reported.<br />

Identification of circulating myeloma cells<br />

Circulating B-cells belonging to the malignant clone<br />

were originally thought to be pre-B-cells on the basis of<br />

the surface expression of the CD10 (CALLA) Ag, 150 an<br />

endopeptidase present on all fetal pre-B and B-cells, on<br />

adult pre-B-cells and their neoplastic counterparts. 151<br />

However, the CD10 Ag has also been found on activated<br />

B-cells 151 and does not seem to be restricted to the early<br />

stages of B-lineage differentiation. Moreover, PB<br />

abnormal B-lymphocytes express plasma cell markers<br />

such as PCA-1 and PC-1 and the CD45RO Ag isoform,<br />

which is typical of late B-cells. 145 Thus phenotypic analysis<br />

of circulating CD19 + cells indicates a heterogeneous,<br />

continuously differentiating B-lineage. 145 By physical<br />

parameters, CD19 + cells include a small and a large subset<br />

that are mainly late B-cells (pre-plasma cells) coexpressing<br />

CD20, CD10, PCA-1, CD45RO and CD24 Ag. 148<br />

The majority of large B-cells also express the CD56 Ag<br />

and high density CD38, whereas small lymphocytes show<br />

only minor expression of these 2 antigenic determinants.<br />

This phenotypic profile (i.e. CD19 + CD20 + CD38 ++ CD56 + )<br />

is not found in normal resting B-cells. Interestingly,<br />

malignant cells were detected at diagnosis, irrespective<br />

of tumor burden and stage of disease, 148 and treatment<br />

had no detectable effect on the large B-cell subset. Conversely,<br />

a significant decrease in the number of small B-<br />

lymphocytes followed chemotherapy, although these<br />

cells returned to baseline value once the therapy was<br />

discontinued. In this regard, it was previously shown that<br />

circulating CD19 + cells in MM express the functional<br />

multidrug transporter p-glycoprotein, 147,169 thus suggesting<br />

that blood B-cells include a highly drug-resistant<br />

subset capable of inducing disease recurrence in myeloma<br />

patients. However, it should be noted that mature<br />

plasma cells do not always express the CD19 Ag, whereas<br />

the presence of the CD56 Ag discriminates clonal plasma<br />

cells from normal ones. 152 In addition, the recently<br />

described monoclonal antibody B-B4 152 seems to be highly<br />

specific for BM and circulating terminal plasma cells.<br />

More recently, the issue of myeloma cell contamination<br />

in leukapheresis products and the kinetics of circulating<br />

tumor cells in response to mobilization protocols have<br />

been addressed. 67,69,153-155 These studies have consistently<br />

shown that the majority of PBSC collections, if not all,<br />

are contaminated by myeloma cells, which represent up<br />

to 10% of PB mononuclear cells by immunophenotyping<br />

and molecular analysis using polymerase chain reaction<br />

(PCR) with consensus oligonucleotides to the Ig<br />

heavy chain complementary determining region III (CDR<br />

III) (see below). 155 The same pattern of contamination<br />

has been shown following high-dose Cy and either G- or<br />

GM-CSF, 67,69,155 as well as after G-CSF alone, 154 suggesting<br />

that growth factors for stem cell mobilization,<br />

regardless of the use of chemotherapy, may influence<br />

the expression of adhesion molecules associated with<br />

the myeloma cell membrane. Notably, kinetic analysis<br />

has demonstrated that following high-dose Cy and G-<br />

CSF, the concomitant mobilization of plasma cells and<br />

hematopoietic progenitor cells in the PB takes place with<br />

the maximum peak of neoplastic elements occurring<br />

within the optimal time period for collection of circulating<br />

CD34 + cells. 67 Conversely, GM-CSF seems to<br />

reduce asynchronous mobilization of neoplastic elements<br />

and hematopoietic stem cells into PB, so that the<br />

contamination of actively proliferating myeloma cells is<br />

minimal in the first two days of apheresis. 156<br />

Methods for assessment of minimal residual<br />

disease<br />

A number of methods have been proposed to detect<br />

malignant cells in the blood of myeloma patients, including<br />

immunologic assessment by monoclonal antibodies,<br />

flow cytometry analysis of DNA and cytoplasmic Ig,<br />

studies on gene rearrangement. Each of these techniques<br />

has limitations in sensitivity and, in some cases, specificity.<br />

For instance, analysis of the hypervariable region<br />

of the Ig heavy chain (IgH) gene using a set of familyspecific<br />

primers (IgH fingerprinting) requires 0.1% mon-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma 41<br />

Regimen Day of Day of No. apheresis References<br />

progenitor peak first apheresis<br />

G-CSF 10 mcg/kg/day 6 d 6 6 phlebotomy 2 112<br />

Cy 7 g/m 2 nr 20 6 117<br />

Cy 7 g/m 2 + GM-CSF 15 nr 4 98-108<br />

Cy 7 g/m 2 + G-CSF 15 14 2-3 141<br />

VCAD 20 14 6 112<br />

VCAD + G-CSF 13 12 2-3 112<br />

Table 7. Day of cell peak and<br />

of first apheresis after PBSC<br />

mobilization in patients with<br />

MM. The addition of G-CSF<br />

or GM-CSF shortens the<br />

time to progenitor peak and<br />

consequently the time to<br />

apheresis. Mean number of<br />

apheretic procedures was<br />

lower when growth factors<br />

were employed.<br />

Legend: nr: not reported.<br />

oclonal cells 157 and may produce false positive results.<br />

Conversely, dual-parameter flow cytometric analysis<br />

(e.g. CD19/monoclonal light chain) and evaluation of<br />

intracytoplasmic monoclonal heavy or light chain are<br />

highly specific and allow detection as low as 0.1% 67 (Figure<br />

3); however, they only assess mature Ig + B-cells.<br />

Recently, several laboratories have described applications<br />

of PCR techniques to increase significantly the sensitivity<br />

and specificity of detection of minimal residual<br />

disease (MRD). Both consensus oligonucleotides (ODN) 146<br />

and family-specific primers 69 have been used to amplify<br />

the CDRIII of rearranged heavy chain alleles (Figure 4)<br />

from myeloma samples. From the sequence of the amplified<br />

products, allele-specific (tumor-specific) oligonucleotides<br />

(ASO) were synthesized and used directly in<br />

PCR amplification reactions (ASO-PCR) for each patient<br />

sample to detect the malignant clone. The sensitivity of<br />

this method is 1:10 5 normal cells and a quantitative<br />

analysis can be performed by generating titrations<br />

curves of tumor cells. Alternatively, direct fingerprinting<br />

of CDRIII IgH gene rearrangement may be used, although<br />

the sensitivity is 1:10 4 normal cells. 67<br />

The biological and prognostic significance of cancer<br />

cells present in autologous grafts is still unknown and<br />

circulating myeloma cells may only reflect advanced<br />

stages of the disease; therefore relapse may be caused<br />

by regrowth of residual clonogenic cells in vivo. However,<br />

considering that MM is a disease intrinsic to BM<br />

and recent studies clearly show that reseeding of reinfused<br />

malignant cells contributes to relapse, 158 several<br />

attempts have been made to remove myeloma cells<br />

from BM or PBSC autografts using different strategies.<br />

Ex vivo purging of myeloma cells<br />

Of the purging methods proposed for the elimination<br />

of MRD, the cyclophosphamide derivative 4-hydroperoxycyclophosphamide<br />

(4-HC) was the first used, 159 on the<br />

basis of in vitro models demonstrating that this compound<br />

was able to eliminate BM-infiltrating MM cell<br />

lines. 160 The main mechanism of action of 4-HC is based<br />

on a marked inhibition of myeloma cell growth, whereas<br />

it spares normal primitive hematopoietic cells. 161 Moreover,<br />

this alkylating agent seems to induce the apoptotic<br />

death of tumor cells 162 as well as activate immune<br />

mechanisms capable of controlling malignant cell proliferation.<br />

163 Because 4-HC does not affect surface antigen<br />

expression of myeloma cells, it is also a potential candidate<br />

for combined treatment with monoclonal antibodies<br />

(MoAbs), and preliminary in vitro data confirm the<br />

additive effect of these two purging techniques. 160 Several<br />

MoAbs directed against tumor-associated or cell differentiation<br />

antigens not expressed by primitive cells<br />

responsible for hematopoietic engraftment have been<br />

selected for clinical trials after in vitro studies demonstrated<br />

high purging efficacy with the use of complement,<br />

164,165 toxins 166,167 or immunoaffinity columns. 168<br />

Gobbi et al. developed a series of MoAbs that recognize<br />

mature plasma cells as well as B-cell precursors. One of<br />

them (8A) was conjugated with the ribosome-inactivat-<br />

Table 8. Recommendations for PBSC mobilization and their<br />

apheretic harvest in patients with multiple myeloma.<br />

• Mobilization with chemotherapy + growth factors (G-CSF or GM-CSF)<br />

• Serial CD34 + determinations according to institutional protocol<br />

• Start apheresis when CD34 + cells in blood >2010 6 /L<br />

• Continuous flow separator, volume processed 2 blood volume per run<br />

• Collect at least 210 6 /kg CD34 + cells in patients with < 24 months prior<br />

chemothrapy, at least 510 6 /kg CD34 + cells in patients with > 24 months<br />

prior chemotherapy<br />

Figure 3. Circulating monoclonal B-lymphocytes and plasma<br />

cells assessed by double fluorescence immunostaining:<br />

intracytoplasmic Ig (green)/nuclear BRDU (red). Bromodeoxyuridine<br />

(BRDU) is incorporated in actively proliferating<br />

cells.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


42<br />

F. Caligaris Cappio et al.<br />

ing toxin momordin and clinically tested in 8 advanced<br />

stage MM patients to eliminate, ex vivo, contaminating<br />

myeloma cells prior to ABMT. 166 Although a marked tumor<br />

reduction was observed in all evaluable patients, none of<br />

them achieved CR and hematopoietic reconstitution following<br />

the myeloablative conditioning therapy was significantly<br />

delayed in 3 patients. These preliminary results<br />

showed the feasibility of this purging approach despite<br />

the poor selection of patients.<br />

The same MoAbs were also employed in vitro to remove<br />

myeloma cells through the avidin-biotin immunoabsorption<br />

technique, and the result was a greater than 3 log<br />

reduction in tumor cells with acceptable recovery of BM<br />

progenitors. 168 More recently, Goldmacher et al. 167 reported<br />

the development of an anti-CD38 immunotoxin capable<br />

of killing 4-6 logs of human myeloma and lymphoma<br />

cell lines. The immunotoxin was composed of an anti-<br />

CD38 antibody conjugated to a chemically modified ricin<br />

molecule (blocked ricin). However, the CD38 Ag may not<br />

be the proper target for purging because it is strongly<br />

expressed on myeloma plasma cells (see above) and on<br />

committed hematopoietic progenitor cells, 169 which are<br />

thought to be essential for rapid BM reconstitution. More<br />

specific antibodies directed either toward B-cells (anti-<br />

CD10 and CD20) or mature plasma cells (PCA-1) and complement<br />

were used to deplete tumor cells from the graft<br />

before ABMT by Anderson et al. 165 Following a TBI-containing<br />

conditioning regimen, a neutrophil count greater<br />

than 0.510 9 /L and an unsupported platelet count<br />

greater than 2010 9 /L were reached at a median of 21<br />

days (range 12-46) and 23 days (range 12-53), respectively.<br />

Similarly, immunologic reconstitution was not different<br />

from that commonly observed in cancer patients<br />

receiving unmanipulated autograft. This study documented<br />

that high-dose chemo-radiotherapy can produce<br />

a high response rate in pretreated patients with sensitive<br />

disease, and MoAb-based purging methods do not prevent<br />

rapid and sustained engraftment. However, the occurrence<br />

of relapses post-ABMT and partial responses will<br />

not define the need, if any, for marrow purging until more<br />

effective ablative strategies are developed. Taken together,<br />

these data demonstrate that the heterogeneity of Ag<br />

expression on neoplastic cells and the lack of true tumorspecific<br />

determinants may greatly influence the efficacy<br />

of antibody-based strategies for the depletion of myeloma<br />

cells. Alternatively, long-term Dexter-type marrow<br />

cultures have been used to select normal myeloid progenitors<br />

from heavily infiltrated myeloma BM, on the basis<br />

of the selective growth advantage of benign cells over<br />

malignant cells in this system. 170<br />

Enrichment of hematopoietic CD34 + cells has lately<br />

been shown to be an alternative approach to myeloma<br />

cell removal with a limited loss of normal stem cells. The<br />

CD34 Ag is a 110-120 kD glycoprotein that is mainly<br />

CDRIII junctional region<br />

VH N DH N JH<br />

FRI CDRI FRII CDRII FRIII<br />

VH family<br />

consensus oligo<br />

VH consensus oligo<br />

CDRIII<br />

VH N DH N JH<br />

clonal CDR-III PCR product at diagnosis<br />

JH consensus oligo<br />

Sequencing<br />

ASO probe construction<br />

(allele specific oligonucleotide probe)<br />

ASO<br />

hybridization negative<br />

ASO<br />

hybridization positive<br />

VH N DH N JH<br />

non clonal CDRIII PCR product<br />

= molecular remission<br />

VH N DH N JH<br />

clonal CDRIII PCR product<br />

= molecular persistance of minimal residual disease<br />

Figure 4. Schematic representation of the genomic region of rearranged CDRIII of IgH gene and further utilization of the PCR<br />

product for detection of MRD. For further details see text.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

43<br />

expressed on the earliest identifiable precursor cells and<br />

committed myeloid progenitors. 169 In normal individuals,<br />

CD34 + cells represent 1% to 4% of the mononuclear cell<br />

fraction in the BM, whereas they are barely detectable in<br />

the PB. 169 In addition, the CD34 Ag is not expressed on<br />

the surface of mature plasma cells in MM, although the<br />

possibility that this glycoprotein may be present on clonally<br />

less differentiated B-lymphocytes is still a matter of<br />

debate. As reported above, recent data support the<br />

hypothesis that MM originates in the later stages of B-<br />

cell differentiation when B lymphocytes have lost the<br />

CD34 Ag, 171 whereas other studies have found CD34 +<br />

cells to be part of the neoplastic clone. 172,173 It should be<br />

underlined, however, that reverse transcription-PCR,<br />

which was used to detect MRD in those studies, is an<br />

extremely sensitive technique, and the potential contamination<br />

of the CD34 + cell fraction by unwanted cells<br />

should be carefully avoided.<br />

In this respect, Vescio et al. 171 did not find IgH gene<br />

clonal rearrangement in collections of 99.99% pure<br />

CD34 + cells obtained after using a combination of two<br />

methods of stem cell purification. Schiller et al. 66 and<br />

Lemoli et al. 67 reported the first studies on purified, CD34-<br />

selected PBSCT conducted in patients with advanced MM.<br />

A median of 4.65 and 410 6 CD34 + cells/kg were reinfused<br />

in the two trials with a median purity of 77% and<br />

88.5%, respectively. The median time to neutrophil and<br />

platelet recovery was 12 days and 10 and 11 days, respectively,<br />

with no difference with respect to a group of<br />

patients receiving unmanipulated PBSCs. 67<br />

Both reports utilized rigorously quantitative immunofluorescence<br />

and/or IgH gene rearrangement analysis,<br />

and tumor cell depletion ranging from 2.5 to 4.5 logs was<br />

achieved. However, the persistence of myeloma cells in<br />

the CD34 + cell fraction was documented by sensitive PCR<br />

assay in all cases heavily contaminated before positive<br />

selection of CD34 + cells. Thus an additional purging step<br />

may be necessary to achieve a virtually tumor-free autograft.<br />

In this regard, studies aimed at optimizing myeloma<br />

cell depletion by positive selection of primitive CD34 +<br />

Lin – Thy + cells have already been performed 155 and clinical<br />

trials are currently in progress.<br />

In summary, all these studies show the capacity of<br />

purging techniques to eliminate a substantial proportion<br />

of the myeloma cells from autologous grafts without<br />

affecting their engraftment potential. The clinical impact<br />

of purging on disease relapse remains to be determined<br />

in future randomized trials.<br />

Post-transplant (immuno)therapy<br />

In MM as well as in other hematologic malignancies,<br />

the primary objective of high-dose therapy with hemopoietic<br />

stem cell support is to prolong survival and possibly<br />

to cure an otherwise incurable disease. The aim of<br />

post-transplant therapy is to prevent recurrence of the<br />

disease while assuring good quality of life. From this<br />

latter point of view, there is no room for additional<br />

chemotherapy as a preventive means. In addition, highdose<br />

chemotherapy itself involves a risk of secondary<br />

myelodysplastic syndrome or acute myeloid leukemia.<br />

This risk is apparently related to prolonged alkylating<br />

agent therapy prior to transplantation and would<br />

undoubtedly increase with additional post-transplant<br />

chemotherapy.<br />

In the past few years interferon- (IFN-) has been<br />

extensively evaluated in the management of MM, either<br />

as part of the induction program or as maintenance therapy.<br />

173 Although controversial findings were frequently<br />

reported, several clinical trials showed a prolongation of<br />

the remission phase, and even of the survival duration,<br />

for patients receiving IFN- after a favorable response<br />

to conventional chemotherapy. 174,175 These results suggested<br />

that IFN- might be particularly useful in patients<br />

with low tumor burden or minimal residual disease, and<br />

led to clinical investigations of this agent in the autograft<br />

setting.<br />

The European Group for Blood and Marrow Transplantation<br />

(EBMT) has recently presented a retrospective study<br />

of a large series of MM patients treated with autologous<br />

stem cell transplantation. 50 Interestingly, post-transplant<br />

treatment with IFN- was independently associated with<br />

extended survival of responding patients, i.e. those achieving<br />

either CR or partial remission. Moreover, Powels et<br />

al. 176 designed a randomized clinical trial aimed at comparing<br />

maintenance IFN- therapy with no maintenance<br />

after HDM and ABMT. 175 The authors found that IFN-<br />

prolonged remission and improved the survival after autotransplant,<br />

and that this effect was particularly marked in<br />

the group of patients achieving CR.<br />

Maintenance IFN- is usually started three months<br />

after transplant and is given sc at a dosage of 310 6<br />

U/m 2 , 3 times weekly. This dose usually induces mild<br />

hematological and non-hematological toxicity, thus<br />

allowing good quality of life. Available data indicate that<br />

about 50% of the MM patients who achieve CR and are<br />

then treated with IFN- remain in remission four years<br />

after transplantation.<br />

Alternatively, maintenance treatments aimed at prolonging<br />

the duration of disease control after transplantation<br />

may also include the administration of interleukin<br />

2 (as nonspecific immunotherapy) 177 or humanized antiidiotype<br />

monoclonal antibodies, which could allow selective<br />

killing of myeloma cells and might be particularly<br />

useful for controlling minimal residual disease.<br />

References<br />

1. Herrinton LJ, Weiss NS, Olshan AF. The epidemiology<br />

of myeloma. In: Malpas JS, Bergsagel DE, Kyle RA, eds.<br />

Myeloma, biology and management, Oxford:Oxford<br />

Medical Publ, 1995:127-68.<br />

2. Turesson I, Zettervall O, Cuzick J, Waldenström JB,<br />

Velez R. Comparison of trends in the incidence of multiple<br />

myeloma in Malmo, Sweden and other countries.<br />

N Engl J Med 1984; 310:421-4.<br />

3. Obrams GI, Potter M. Epidemiology and biology of<br />

multiple myeloma. Berlin:Springer-Verlag, 1991.<br />

4. Evaluating the National Cancer Program: an ongoing<br />

haematologica vol. 85(suppl. to n. 12):December 2000


44<br />

F. Caligaris Cappio et al.<br />

process. National Cancer Institute, Bethesda, Md,<br />

USA, 1994.<br />

5. Riedel DA, Pottern LM. The epidemiology of multiple<br />

myeloma. Hematol Oncol Clin N Am 1992; 6:225-47.<br />

6. Sawyer JR, Waldron JA, Jagannath S, Barlogie B. Cytogenetic<br />

findings in 200 patients with multiple myeloma.<br />

Cancer Genet Cytogen 1995; 82:41-9.<br />

7. Zandecki M, Obein V, Bernardi F, et al. Monoclonal<br />

gammopathy of undetermined significance: chromosome<br />

changes are a common finding within bone marrow<br />

plasma cells. Br J Haematol 1995; 90:693-6.<br />

8. Drach J, Angerler J, Schuster J, et al. Interphase fluorescence<br />

in situ identifies chromosomal abnormalities<br />

in plasma cells from patients with monoclonal gammopathy<br />

of undetermined significance. Blood 1995;<br />

86:3915-21.<br />

9. Barlogie B. Plasma cell myeloma. In: Beutler E, Lichtman<br />

MA, Coller BS, Kipps TJ, eds. Williams’ Hematology,<br />

5 th ed. New York:1995: 1109-26.<br />

10. Hoover RG, Kornbluth J. Immunoregulation of murine<br />

and human myeloma. Hematol Oncol Clin N Am<br />

1992; 6:407-24.<br />

11. Potter M. Myeloma proteins (M-components) with<br />

antibody-like activity. N Engl J Med 1971; 284:831-8.<br />

12. Seligmann M, Brouet JC. Antibody activity of human<br />

myeloma globulins. Semin Hematol 1973; 10:163-77.<br />

13. Konrad RJ, Kricka LJ, Goodman DBP, Goldman J, Silberstein<br />

LE. Myeloma-associated paraprotein directed<br />

against the HIV-1 p24 antigen in an HIV-1-seropositive<br />

patient. N Engl J Med 1993; 328:1817-9.<br />

14. Bakkus MHC, Heirman C, Van Riet I, Van Camp B,<br />

Thielemans K. Evidence that multiple myeloma Ig heavy<br />

chain VDJ genes contain somatic mutations but show<br />

no intraclonal variation. Blood 1992; 80:2326-35.<br />

15. Vescio RA, Cao J, Hong CH, et al. Myeloma Ig heavy<br />

chain V region sequences reveal prior antigenic selection<br />

and marked somatic mutation but no intraclonal<br />

diversity. J Immunol 1995; 155:2487-97.<br />

16. Kelsoe G. B cell diversification and differentiation in<br />

the periphery. J Exp Med 1994; 180:5-6.<br />

17. Caligaris-Cappio F, Gregoretti MG. Basic concepts:<br />

plasma cells in multiple myeloma. In: BGM Durie, G.<br />

Gahrton, eds. Multiple Myeloma. London:E. Arnold<br />

Publisher, in press.<br />

18. Benner R, Hijmans W, Haajman JJ. The bone marrow:<br />

the major source of serum immunoglobulins, but still<br />

a neglected site of antibody formation. Clin Exp<br />

Immunol 1981; 46:1-8.<br />

19. DiLosa RM, Maeda K, Masuda A, Szakal AK, Tew JG.<br />

Germinal center B cells and antibody production in the<br />

bone marrow. J Immunol 1991; 146:4071-7.<br />

20. Liu YJ, Johnson GD, Gordon J, MacLennan ICM. Germinal<br />

centres in T-cell dependent antibody responses.<br />

Immunol Today 1992; 13:17-21.<br />

21. MacLennan ICM. In which cells does neoplastic transformation<br />

occur in myelomatosis? Curr Top Microbiol<br />

Immunol 1992; 182:209-13.<br />

22. Caligaris-Cappio F, Gregoretti MG, Ghia P, Bergui L. In<br />

vitro growth of human multiple myeloma: implications<br />

for biology and therapy. Hematol Oncol Clin N 1992;<br />

6:257-71.<br />

23. Corradini P, Boccadoro M, Voena C, Pileri A. Evidence<br />

for a bone marrow B cell transcribing malignant plasma<br />

cell VDJ joined to Cm sequence in IgG and IgA<br />

secreting multiple myelomas. J Exp Med 1993; 178:<br />

1091-6.<br />

24. Billadeau D, Ahmann G, Greipp P, Van Ness B. The<br />

bone marrow of multiple myeloma patients contains B<br />

cell populations at different stages of differentiation<br />

that are clonally related to the malignant plasma cell.<br />

J Exp Med 1993; 178:1023-31.<br />

25. Rolink A, Melchers F. Generation and regeneration of<br />

cells of the B-lymphocyte lineage. Curr Opin Immunol<br />

1993; 5:207-17.<br />

26. Kinkade P, Lee G, Pietrangeli CE, Hayashi SH, Gimble<br />

J. Cells and molecules that regulate B lymphopoiesis in<br />

bone marrow. Annu Rev Immunol 1989; 7:111-43.<br />

27. Degrassi A, Hilbert DM, Rudikoff S, Anderson AO, Potter<br />

M, Coon HG. In vitro culture of primary plasmacytomas<br />

requires stromal cell feeder layers. Proc Natl<br />

Acad Sci USA 1993; 90:2060-4.<br />

28. Caligaris-Cappio F, Bergui L, Gregoretti MG, et al. Role<br />

of bone marrow stromal cells in the growth of human<br />

multiple myeloma. Blood 1991; 77:2688-93.<br />

29. Thiery JP, Boyer B. The junction between cytokines and<br />

cell adhesion. Current Opin Cell Biol 1992; 4:782-92.<br />

30. Hynes RO. Integrins: versatility, modulation and signalling<br />

in cell adhesion. Cell 1992; 69:11-25.<br />

31. Klein B, Zhang XG, Jourdan M, et al. Paracrine rather<br />

than autocrine regulation of myeloma-cell growth and<br />

differentiation by interleukin-6. Blood 1989; 73:517-<br />

26.<br />

32. Klein B, Zhang XG, Lu ZY, Bataille R. Interleukin-6 in<br />

human multiple myeloma. Blood 1995; 85:863-72.<br />

33. Kishimoto T, Akira S, Narazaki M, Taga T. Interleukin-<br />

6 family of cytokines and gp130. Blood 1995; 86:<br />

1243-54.<br />

34. Jernberg H, Pettersson M, Kishimoto T, Nilsson K.<br />

Heterogeneity in response to interleukin 6 (IL-6),<br />

expression of IL-6 and IL-6 receptor mRNA in a panel<br />

of established human multiple myeloma cell lines.<br />

Leukemia 1991; 5:255-65.<br />

35. Bataille R, Jourdan M, Zhang XG, Klein B. Serum levels<br />

of interleukin-6, a potent myeloma cell growth factor,<br />

as a reflection of disease severity in plasma cell<br />

dyscrasias. J Clin Invest 1989; 84:2008-11.<br />

36. Klein B, Wijdenes J, Zhang XG, et al. Murine anti-interleukin-6<br />

monoclonal antibody therapy for a patient<br />

with plasma cell leukemia. Blood 1991; 78:1198-204.<br />

37. Bataille R, Chappard D, Klein B. Mechanisms of bone<br />

lesions in multiple myeloma. Hematol Oncol Clin N<br />

Am 1992; 6:285-95.<br />

38. Bersagel DE. The role of chemotherapy in the treatment<br />

of multiple myeloma. Baillière Clin Haematol<br />

1995; 8:783-94.<br />

39. Gregory WN, Richards MA, Malpas JS. Combination<br />

chemotherapy versus melphalan and prednisolone in<br />

the treatment of multiple myeloma: an overview of<br />

published trials. J Clin Oncol 1992; 10:334-42.<br />

40. McElwain TJ, Powles RL. High-dose intravenous melphalan<br />

for plasma-cell leukemia and myeloma. Lancet<br />

1983; 1:822-4.<br />

41. Barlogie B, Hall R, Znader A, et al. High-dose melphalan<br />

with autologous bone marrow transplantation for<br />

multiple myeloma. Blood 1986; 67:1298-301.<br />

42. Selby PJ, McElwain TJ, Nandi AC, et al. Multiple myeloma<br />

treated with high dose intravenous melphalan. Br<br />

J Haematol 1987; 66:55-62.<br />

43. Cavo M, Benni M, Gozzetti A, et al. The role of haematopoietic<br />

stem cell-supported myeloablative therapy<br />

for the management of multiple myeloma. Baillière Clin<br />

Haematol 1995; 8:795-813.<br />

44. Barlogie B, Alexanian R, Dicke KA, et al. High-dose<br />

chemoradiotherapy and autologous bone marrow<br />

transplantation for resistant multiple myeloma. Blood<br />

1987; 70:869-72.<br />

45. Jagannath S, Barlogie B, Dicke K, et al. Autologous<br />

bone marrow transplantation in multiple myeloma:<br />

identification of prognostic factors. Blood 1990; 76:<br />

1860-6.<br />

46. Harosseuau JL, Milpied N, LaPorte JP, et al. Doubleintensive<br />

therapy in high-risk multiple myeloma. Blood<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

45<br />

1992; 79: 2827-33.<br />

47. Attal M, Huguet F, Schlaifer D, et al. Intensive combined<br />

therapy for previously untreated aggressive<br />

myeloma. Blood 1992; 79: 1130-6.<br />

48. Cunningham D, Paz-Arez L, Milan S, et al. High-dose<br />

melphalan and autologous bone marrow transplantation<br />

as consolidation in previously untreated myeloma.<br />

J Clin Oncol 1994; 12:759-63.<br />

49. Attal M, Harousseau JL, Stoppa AM, et al. High dose<br />

therapy in multiple myeloma: final analysis of a<br />

prospective randomized study of the “Intergroupe<br />

Français du Myelome” (IFM 90). Blood 1995; 86 (suppl.<br />

1): Abstract n° 485.<br />

50. Björkstrand B, Goldstone AH, Ljungman P, et al. Prognostic<br />

factors in autologous stem cell transplantation<br />

for multiple myeloma: An EBMT registry study. Leuk<br />

Lymphoma 1994; 15:265-72.<br />

51. Barlogie B, Jagannath S, Vesole D, et al. Autologous<br />

and allogeneic transplants for multiple myeloma.<br />

Semin Hematol 1995; 32:31-44.<br />

52. Harousseau JL, Attal M, Divine M, et al. Autologous<br />

stem cell transplantation after first remission induction<br />

treatment in multiple myeloma: a report of the<br />

French registry on autologous transplantation in multiple<br />

myeloma. Blood 1995; 85:3077-85.<br />

53. Vesole DH, Barlogie B, Jagannath S et al. High-dose<br />

therapy for refractory multiple myeloma: improved<br />

prognosis with better supportive care and double<br />

transplants. Blood 1994; 84:950-6.<br />

54. Gore ME, Selby PJ, Viner C, et al. Intensive treatment<br />

of multiple myeloma and criteria for complete remission.<br />

Lancet 1989; 2:879-81.<br />

55. Dimopoulos MA, Alexanian R, Przepiorka D, et al.<br />

Thiotepa, busulfan, and cyclophosphamide: a new<br />

preparative regimen for autologous marrow or blood<br />

stem cell transplantation in high-risk multiple myeloma.<br />

Blood 1993; 82:2324-8.<br />

56. Fermand JP, Chevret S, Ravaud P, et al. High-dose<br />

chemoradiotherapy and autologous blood stem cell<br />

transplantation in multiple myeloma: results of a phase<br />

II trial involving 63 patients. Blood 1993; 82:2005-9.<br />

57. Alegre A, Lamana M, Arranz R, et al. Busulfan and melphalan<br />

as conditioning regimen for autologous peripheral<br />

blood stem cell transplantation in multiple myeloma.<br />

Br J Haematol 1995; 91:380-6.<br />

58. Barlogie B, Jagannath S, Dixon DO, et al. High-dose<br />

melphalan and granulocyte-macrophage colony-stimulating<br />

factor for refractory multiple myeloma. Blood<br />

1990; 76:677-80.<br />

59. Tricot G, Jagannath S, Vesole D, et al. Peripheral blood<br />

stem cell transplants for multiple myeloma: identification<br />

of favourable variables for rapid engraftment in<br />

225 patients. Blood 1995; 85:588-96.<br />

60. Cavo M, Benni M, Cirio TM, et al. Allogeneic bone<br />

marrow transplantation for the treatment of multiple<br />

myeloma. An overview of published trials. Stem Cells<br />

1995; 13 (Suppl. 2): 121-6.<br />

61. Tricot G, Vesole DH, Jagannath S, Hilton J, Munshi N,<br />

Barlogie B. Graft-versus-myeloma effect: proof of principle.<br />

Blood 1996; 87:1196-8.<br />

62 Verdonck LF, Lokhorst HM, Dekker AW, Nieuwenhuis<br />

HK, Petersen EJ. Graft-versus-myeloma effect in two<br />

cases. Lancet 1996; 347:800-1.<br />

63. Joshua DE, Gibson J, Brown RD. Mechanisms of the<br />

escape phase of myeloma. Blood Rev 1994; 8:13-20.<br />

64. Alexanian R, Dimopoulos M, Smith T, et al. Limited<br />

value of myeloablative therapy for late multiple myeloma.<br />

Blood 1994; 83:512-6.<br />

65. Alexanian R, Dimopoulos MA, Smith T, et al. Early<br />

myeloablative therapy for multiple myeloma. Blood<br />

1994; 84:4278-82.<br />

66. Schiller G, Vescio R, Freytes C, et al. Transplantation of<br />

CD34 + peripheral bood progenitor cells after high-dose<br />

chemotherapy for patients with advanced multiple<br />

myeloma. Blood 1995; 86:390-7.<br />

67. Lemoli RM, Fortuna A, Motta MR, et al. Concomitant<br />

mobilization of plasma cells and hemopoietic progenitors<br />

into peripheral blood of multiple myeloma<br />

patients: positive selection and transplantation of<br />

enriched CD34 + cells to remove circulating tumour<br />

cells. Blood 1996; 87:1625-34.<br />

68. Cunningham D, Powles R, Viner C, et al. High-dose<br />

chemotherapy and autologous bone marrow transplantation<br />

in multiple myeloma. Abstract Book IV<br />

International Workshop on Multiple Myeloma<br />

(Rochester) 1993; p. 102.<br />

69. Corradini P, Voena C, Astolfi M, et al. High-dose<br />

sequential chemoradiotherapy in multiple myeloma:<br />

residual tumor cells are detectable in bone marrow and<br />

peripheral blood cell harvests and after autografting.<br />

Blood 1995; 85:1596-602.<br />

70. Gianni AM, Bregni M, Siena S, et al. Rapid and complete<br />

hemopoietic reconstitution following combined<br />

transplantation of autologous blood and bone marrow<br />

cells. A changing role for high-dose chemotherapy?<br />

Hematol Oncol 1989; 7:139-48.<br />

71. Gianni AM, Bregni M, Siena S, et al. Granulocytemacrophage<br />

colony stimulating factor to harvest circulating<br />

hemopoietic stem cells for autotransplantation.<br />

Lancet 1989; 2:580-5.<br />

72. Kessinger A, Armitage JO. The evolving role of autologous<br />

peripheral stem cell transplantation following<br />

high dose therapy for malignancies. Blood 1991; 77:<br />

211-2.<br />

73. Sheridan WP, Begley CG, Juttner C, et al. Effect of<br />

peripheral blood progenitor cells mobilized by filgrastim<br />

(G-CSF) on platelet recovery after high dose<br />

chemotherapy. Lancet 1992; 1:640-4.<br />

74. Tarella C, Ferrero D, Bregni M, et al. Peripheral blood<br />

expansion of early progenitor cells after high dose<br />

cyclophosphamide and rhGM-CSF. Eur J Cancer 1991;<br />

27:22-7.<br />

75. Haas R, Ho AD, Bredthauer U, et al. Successful autologous<br />

transplantation of blood stem cells mobilized<br />

with recombinant human granulocyte macrophage<br />

colony-stimulating factor. Exp Hematol 1990; 18:94-<br />

8.<br />

76. Shimazaki C, Oku N, Ashihara E, et al. Collection of<br />

peripheral blood stem cells mobilized by high-dose<br />

Ara-C plus VP-16 or aclarubicin followed by recombinant<br />

human granulocyte colony-stimulating factor.<br />

Bone Marrow Transplant 1992; 10:341-6.<br />

77. Teshima T, Harada M, Takamatsu Y, et al. Cytotoxic<br />

drug and cytotoxic drug/G-CSF mobilization of peripheral<br />

blood stem cells and their use for autografting.<br />

Bone Marrow Transplant 1992; 10:215-20.<br />

78. Brugger W, Birken R, Bertz H, et al. Peripheral blood<br />

progenitor cells mobilized by chemotherapy plus granulocyte-colony<br />

stimulating factor accelerate both neutrophil<br />

and platelet recovery after high-dose VP16, ifosfamide<br />

and cisplatin. Br J Haematol 1993; 84:402-7.<br />

79. Siena S, Bregni M, Brando B, et al. Flow cytometry for<br />

clinical estimation of circulating hematopoietic progenitors<br />

for autologous transplantation in cancer<br />

patients. Blood 1992; 77:400-9.<br />

80. Siena S, Bregni M, Brando B, et al. Practical aspects of<br />

flow cytometry to guide large-scale collection of circulating<br />

hematopoietic progenitors for autologous transplantation<br />

in cancer patients. Int J Cell Cloning 1992;<br />

10:26-9.<br />

81. Siena S, Bregni M, Di Nicola M, et al. Durability of<br />

hematopoiesis following myeloablative cancer therapy<br />

haematologica vol. 85(suppl. to n. 12):December 2000


46<br />

F. Caligaris Cappio et al.<br />

and autografting with peripheral blood hematopoietic<br />

progenitors. Ann Oncol 1994; 5:935-41.<br />

82. Ravagnani F, Siena S, Bregni M, Sciorelli G, Gianni AM,<br />

Pellegris G. Large cell collection of circulating<br />

haematopoietic progenitors in cancer patients treated<br />

with high-dose cyclophosphamide and recombinant<br />

human GM-CSF. Eur J Cancer 1990; 26:562-4.<br />

83. Tong J, Hoffman R, Siena S, et al. Characterization and<br />

quantitation of primitive hemopoietic progenitor cells<br />

present in peripheral blood autografts. Exp Hematol<br />

1994; 21:1016-24.<br />

84. Bender JG, Lum L, Unverzagt KL, et al. Correlation of<br />

colony-forming cells, long-term culture-initiating cells<br />

and CD34 + cells in apheresis products from patients<br />

mobilized for peripheral blood progenitors with different<br />

regimens. Bone Marrow Transplant 1994; 13:479-<br />

85.<br />

85. Pettengell R, Luft T, Henschler R, et al. Direct comparison<br />

by limiting dilution analysis of long-term culture-initiating<br />

cells in human bone marrow, umbilical<br />

cord blood, and blood stem cells. Blood 1994; 84:<br />

3653-9.<br />

86. Gianni AM, Tarella C, Siena S, et al. Durable and complete<br />

hematopoietic reconstitution after autografting<br />

of rhGM-CSF exposed peripheral blood progenitor<br />

cells. Bone Marrow Transplant 1990; 6:143-5.<br />

87. Johnsen HE, Hansen PB, Plesner T, et al. Increased yield<br />

of myeloid progenitor cells in bone marrow harvested<br />

for autologous transplantation by pretreatment with<br />

recombinant human granulocyte-colony stimulating<br />

factor. Bone Marrow Transplant 1992; 10:229-34.<br />

88. Janssen WE. Peripheral blood and bone marrow<br />

hematopoietic stem cells: are they the same? Semin<br />

Oncol 1993; 20 (suppl. 6):19-27.<br />

89. Eaves CJ. Peripheral blood stem cells reach new<br />

heights. Blood 1993; 82:1957-9.<br />

90. Tarella C, Benedetti G, Caracciolo D, et al. Both early<br />

and committed haematopoietic progenitors are more<br />

frequent in peripheral blood than in bone marrow during<br />

mobilization induced by high-dose chemotherapy<br />

+ G-CSF. Br J Haematol 1995; 91:535-43.<br />

91. Reiffers J, Marit G, Boiron JM. Autologous blood stem<br />

cell transplantation in high-risk multiple myeloma. Br<br />

J Haematol 1989; 72:296-7.<br />

92. Pileri A, Tarella C, Bregni M, et al. GM-CSF exposed<br />

peripheral blood progenitors as sole source of stem<br />

cells for autologous transplantation in two patients<br />

with multiple myeloma. <strong>Haematologica</strong> 1990; 75:79-<br />

82.<br />

93. Barlogie B, Alexanian R, Smallwood L, et al. Prognostic<br />

factors with high-dose melphalan for refractory multiple<br />

myeloma. Blood 1988; 72:2015-9.<br />

94. Lokhorst HM, Meuwissen OJA, Verdonk LF, Dekker A.<br />

High-risk multiple myeloma treated with high-dose<br />

melphalan. J Clin Oncol 1992; 10:47-51.<br />

95. Cunningham D, Paz-Ares L, Gore ME, et al. High-dose<br />

melphalan for multiple myeloma: long-term follow-up<br />

data. J Clin Oncol 1994; 12:764-8.<br />

96. Mansi J, da Costa F, Viner C, Judson I, Gore ME,<br />

Cunningham D. High-dose busulfan in patients with<br />

myeloma. J Clin Oncol 1992; 10:1569-73.<br />

97. Jagannath S, Vesole DH, Glenn L, Crowley J, Barlogie<br />

B. Low-risk intensive therapy for multiple myeloma with<br />

combined autologous bone marrow and blood stem<br />

cell support. Blood 1992; 80:1666-72.<br />

98. Gianni AM, Tarella C, Bregni M, et al. High-dose<br />

sequential (HDS) chemo-radiotherapy, a widely applicable<br />

regimen, confers survival benefit to patients with<br />

high-risk multiple myeloma. J Clin Oncol 1994;<br />

12:503-9.<br />

99. Tricot G, Jagannath S, Vesole DH, Crowley J, Barlogie<br />

B. Relapse of multiple myeloma after autologous transplantation:<br />

survival after salvage therapy. Bone Marrow<br />

Transplant 1995; 16:7-11.<br />

100. Wunder E. Stimulation of granulocyte macrophage<br />

progenitors via monocyte/macrophage activation: a<br />

fundamental regulatory pathway of terminal differentiation.<br />

In: Wunder EW, Henon PR, eds. Peripheral<br />

blood stem cell autografts. Berlin:Springer-Verlag,<br />

1993:58-66.<br />

101. Mauch P, Ferrara J, Hellman S. Stem cell self-renewal<br />

considerations in bone marrow transplantation. Bone<br />

Marrow Transplant 1989; 4:601-7.<br />

102. Kotasek D, Sheperd KM, Sage RE, et al. Factors affecting<br />

blood stem cell collections following high-dose<br />

cyclophosphamide mobilization in lymphoma, myeloma<br />

and solid tumors. Bone Marrow Transplant 1992;<br />

9:11-7.<br />

103. Haas R, Möhle R, Fruehauf S, et al. Patient characteristics<br />

associated with successful mobilizing and autografting<br />

of peripheral blood progenitor cells in malignant<br />

lymphoma. Blood 1994; 83:3787-94.<br />

104. Schneider JG, Crown JP, Wasserheit C, et al. Factors<br />

affecting the mobilization of primitive and committed<br />

hematopoietic progenitors into the peripheral blood<br />

of cancer patients. Bone Marrow Transplant 1994;<br />

14:877-84.<br />

105. Bensinger WI, Longin K, Appelbaum F, et al. Peripheral<br />

blood stem cells (PBSCs) collected after recombinant<br />

granulocyte colony stimulating factor (rhG-CSF):<br />

an analysis of factors correlating with the tempo of<br />

engraftment after transplantation. Br J Haematol<br />

1994; 87:825-31.<br />

106. Laporte JP, Gorin NC, Dupuy-Montbrun MC, et al.<br />

Failure to collect sufficient amount of peripheral blood<br />

stem cells (PBSC) for autografting in patients with endstage<br />

multiple myeloma. Bone Marrow Transplant<br />

1988; 3(suppl 1):89.<br />

107. Tarella C, Caracciolo D, Gavarotti P, et al. Circulating<br />

progenitors following high-dose sequential (HDS)<br />

chemotherapy with G-CSF: short intervals between<br />

drug courses severely impair progenitor mobilization.<br />

Bone Marrow Transplant 1995; 16:223-8.<br />

108. Tarella C, Boccadoro M, Omedé P, et al. Role of<br />

chemotherapy and GM-CSF on hemopoietic progenitor<br />

cell mobilization in multiple myeloma. Bone Marrow<br />

Transplant 1993; 11: 271-7.<br />

109. Dimopoulos MA, Hester J, Huh Y, Champlin R, Alexanian<br />

R. Intensive therapy with blood progenitor transplantation<br />

for primary resistant multiple myeloma. Br<br />

J Haematol 1994; 87:730-4.<br />

110. Haas R, Möhle R, Murea S, et al. Characterization of<br />

peripheral blood progenitor cells mobilized by cytotoxic<br />

chemotherapy and recombinant human granulocyte<br />

colony-stimulating factor. J Hematother 1994;<br />

3:323-30.<br />

111. Ossenkoppele GJ, Jonkhoff AR, Huijgens PC, et al.<br />

Peripheral blood progenitors mobilised by G-CSF (filgrastim)<br />

and reinfused as unprocessed autologous<br />

whole blood shorten the pancytopenic period following<br />

high-dose melphalan in multiple myeloma. Bone<br />

Marrow Transplant 1994; 13:37-41.<br />

112. Majolino I, Marcenò R, Buscemi F, et al. Mobilization<br />

of circulating progenitor cells in multiple myeloma during<br />

VCAD therapy with or without rhG-CSF. <strong>Haematologica</strong><br />

1995; 80:108-14.<br />

113. Vasta S, Majolino I, Morabito F, et al. The association<br />

of vincristine, cyclophosphamide, epirubicin and dexamethasone<br />

(VCED) followed by rhG-CSF may provide<br />

a good PBSC mobilization in patients with multiple<br />

myeloma. Submitted for publication.<br />

114. Kessinger A, Armitage JO, Smith DM, et al. High-dose<br />

haematologica vol. 85(suppl. to n. 12):December 2000


PBSC transplantation in multiple myeloma<br />

47<br />

therapy and autologous peripheral blood stem cell<br />

transplantation for patients with lymphoma. Blood<br />

1989; 74:1260-5.<br />

115. Hénon P, Beck G, Debecker A, Eisenman JC, Lepers M,<br />

Kandel G. Autograft using peripheral blood stem cells<br />

collected after high-dose melphalan in high-risk multiple<br />

myeloma. Br J Haematol 1988; 70:254-5.<br />

116. Fermand JP, Levy Y, Gerota J, et al. Treatment of aggressive<br />

multiple myeloma by high-dose chemotherapy and<br />

total body irradiation followed by blood stem cells<br />

autologous graft. Blood 1989; 73:20-3.<br />

117. Reiffers J, Marit G, Boiron JM. Peripheral blood stemcell<br />

transplantation in intensive treatment of multiple<br />

myeloma. Lancet 1989; ii:1336.<br />

118. Indovina A, Majolino I, Scimé R, et al. High dose<br />

cyclophosphamide: stem cell mobilizing capacity in 21<br />

patients. Leuk Lymphoma 1994; 14:71-7.<br />

119. Duhrsen V, Villeval JL, Boyd J, Kannourakis G, Moerstyn<br />

G, Metcalf D. Effects of recombinant human granulocyte<br />

colony-stimulating factor on hematopoietic progenitor<br />

cells in cancer patients. Blood 1988; 72:2074-<br />

81.<br />

120. Bensinger W, Singer J, Appelbaum F, et al. Autologous<br />

transplantation with peripheral blood mononuclear<br />

cells collected after administration of recombinant<br />

granulocyte stimulating factor. Blood 1993; 81:3158-<br />

63.<br />

121. Majolino I, Buscemi F, Scimè R, et al. Treatment of<br />

normal donors with rhG-CSF 16 µg/kg for mobilization<br />

of peripheral blood stem cells and their apheretic<br />

collection for allogeneic transplantation. <strong>Haematologica</strong><br />

1995; 80:219-26.<br />

122. Socinski MA, Cannistra SA, Elias A, Antman KH,<br />

Schnipper L, Griffin JD. Granulocyte-macrophage<br />

colony stimulating factor expands the circulating<br />

haemopoietic progenitor cell compartment in man.<br />

Lancet 1988; i:1194-8.<br />

123. Brugger W, Bross KJ, Glatt M, Weber F, Mertelsmann<br />

R, Kanz L. Mobilization of tumor cells and hematopoietic<br />

progenitor cells into the peripheral blood of<br />

patients with solid tumors. Blood 1994; 83:636-40.<br />

124. Gianni AM, Bregni M, Siena S, et al. Granulocytemacrophage<br />

colony-stimulating factor or granulocyte<br />

colony-stimulating factor infusion makes high-dose<br />

etoposide a safe outpatient regimen that is effective in<br />

lymphoma and myeloma patients. J Clin Oncol 1992;<br />

10:1955-62.<br />

125. Bender JG, To LB, Williams S, Schwartzberg LS. Defining<br />

a therapeutic dose of peripheral blood stem cells.<br />

J Hematother 1992; 1:329-41.<br />

126. Smith R, Sweetenham JW. A mononuclear cell dose of<br />

3x10 8 /kg is sufficient to predict early multilineage<br />

engraftment in patients undergoing high dose therapy<br />

and transplantation with cryopreserved peripheral<br />

blood progenitor cells for malignant lymphoma. Blood<br />

1994; 84 (Suppl. 1):364.<br />

127. Schwartzberg L, Birch R, Blanco R, et al. Rapid and<br />

sustained hematopoietic reconstitution by peripheral<br />

blood stem cell infusion alone following high-dose<br />

chemotherapy. Bone Marrow Transplant 1993; 11:<br />

369-74.<br />

128. Pierelli L, Iacone A, Quaglietta AM, et al. Haemopoietic<br />

reconstitution after autologous blood stem cell<br />

transplantation in patients with malignancies: a multicentre<br />

retrospective study. Br J Haematol 1994; 86:<br />

70-5.<br />

129. Buscemi F, Indovina A, Scimè R, et al. CD34 + cell subsets<br />

and platelet recovery after PBSC autograft. Bone<br />

Marrow Transplant, in press.<br />

130. Schiller G, Rosen L, Vescio R, et al. Threshold dose of<br />

autologous CD34 positive peripheral blood progenitor<br />

cells required for engraftment after myeloablative treatment<br />

for multiple myeloma. Blood 1994;84 (suppl 1):<br />

207.<br />

131. Zimmerman TM, Lee WJ, Bender JG, Mick R, Williams<br />

SF. Quantitative CD34 analysis may be used to guide<br />

peripheral blood stem cell harvests. Bone Marrow<br />

Transplant 1995; 9:439-44.<br />

132. Indovina A, Majolino I, Buscemi F, et al. Engraftment<br />

kinetics and long-term stability of hematopoiesis following<br />

autografting of peripheral blood stem cells.<br />

<strong>Haematologica</strong> 1995; 80:115-22.<br />

133. Siena S, Bregni M, Brando B, et al. Flow cytometry for<br />

clinical estimation of circulating hematopoietic progenitors<br />

for autologous transplantation in cancer<br />

patients. Blood 1991; 77: 400-9.<br />

134. Hénon PR, Wunder E, Zingsem J, Lepers M, Siegert W,<br />

Eckstein R. Collection of peripheral blood stem cells.<br />

Apheresis monitoring and procedure. In: Wunder EW,<br />

Henon PR, eds. Peripheral blood stem cell autografts.<br />

Berlin:Springer-Verlag, 1993:185-93.<br />

135. Buscemi F, Fabbiano F, Felice R, et al. Use of large<br />

polygonal contiguous gates for flow cytometry analysis<br />

of circulating progenitor cells. Bone Marrow Transplant<br />

1993; 12:305.<br />

136. Rowley S. Analysis of the collected product. In:<br />

Kessinger A, McMannis JD, eds. Practical considerations<br />

of apheresis in peripheral blood stem cell transplantation.<br />

Lakewood, Co: Cobe BCT, Inc., 1994:35-<br />

51.<br />

137. Legros M, Fleury J, Curé H, et al. New method for stem<br />

cell quantification: applications to the management of<br />

peripheral blood stem cell transplantation. Bone Marrow<br />

Transplant 1995; 15:1-8.<br />

138. Mijovic A, Mufti GK. Single dose of filgrastim (rhG-<br />

CSF) to predict mobilization of hemopoietic progenitors<br />

in patients with hematologic malignancies. Bone<br />

Marrow Transplant 1995; 15:813-4.<br />

139. Fruehauf S, Haas R, Conradt C, et al. Peripheral blood<br />

progenitor cell (PBPC) counts during steady-state<br />

hematopoiesis allow to estimate the yield of mobilized<br />

PBPC after filgrastim (R-metHuG-CSF)-supported<br />

cytotoxic chemotherapy. Blood 1995; 85:2619-26.<br />

140. Roberts AW, Begley CG, Grigg AP, Basser RL. Do<br />

steady-state peripheral blood progenitor cell (PBPC)<br />

counts predict the yield of PBPC mobilized by filgrastim<br />

alone? Blood 1995; 86:2451.<br />

141. Cavo M, Gozzetti A, Lemoli RM, et al. High-dose<br />

cyclophosphamide (7 g/m 2 ) for the treatment of newly<br />

diagnosed and refractory multiple myeloma patients.<br />

Submitted for publication.<br />

142. To LB. Is our current strategy in manipulating hemopoiesis<br />

in autologous transplantation correct? Stem<br />

Cells 1993; 11: 283-9.<br />

143. Berenson J, Wong R, Kim K, Brown N, Lichstentein A.<br />

Evidence of peripheral blood B lymphocyte but not T<br />

lymphocyte involvement in multiple myeloma. Blood<br />

1987; 70: 1550-3.<br />

144. Bergui L, Schena M, Gaidano GL, Riva M, Caligaris-<br />

Cappio F. Interleukin-3 and interleukin-6 synergistically<br />

promote the proliferation and diffentiation of malignant<br />

plasma cell precursors in multiple myeloma. J Exp<br />

Med 1989; 170:613-8.<br />

145. Pilarski LM, Jensen GS. Monoclonal circulating B cells<br />

in multiple myeloma: a continuously differentiating<br />

possibly invasive population as defined by expression<br />

of CD45 isoforms and adhesion molecules. Hematol<br />

Oncol Clin N Am 1992; 6:297-332.<br />

146. Billadeau D, Quam L, Thomas W, et al. Detection and<br />

quantitation of malignant cells in the peripheral blood<br />

of multiple myeloma patients. Blood 1992; 80:1818-<br />

24.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


48<br />

F. Caligaris Cappio et al.<br />

147. Pilarski LM, Belch AR. Circulating monoclonal B cells<br />

expressing P glycoprotein may be a reservoir of multidrug-resistant<br />

disease in multiple myeloma. Blood<br />

1994; 83:724-36.<br />

148. Bergsagel LP, Masellis Smith A, Szczepek A, Mant MJ,<br />

Belch AR, Pilarski LM. In multiple myeloma, clonotypic<br />

B lymphocytes are detectable among CD19 + peripheral<br />

blood cells expressing CD38, CD56, and monotypic<br />

Ig light chain. Blood 1995; 85: 436-47.<br />

149. Witzig T, Kyle R, O’Fallon W, Greipp P. Detection of<br />

peripheral blood plasma cells as a predictor of disease<br />

course in patients with smouldering multiple myeloma.<br />

Br J Haematol 1994; 87:266-72.<br />

150. Grogan TM, Durie BGM, Lomen C, et al. Delineation<br />

of a novel pre-B cell component in plasma cell myeloma:<br />

immunochemical, immunophenotypic, genotypic,<br />

cytologic, cell culture, and kinetic features. Blood<br />

1987; 70:932-42.<br />

151. LeBien TW, McCormack RT. The common acute<br />

lymphoblastic leukemia antigen (CD10). Emancipation<br />

from a functional enigma. Blood 1989; 73:625-<br />

35.<br />

152. Pellat-Deceunyck C, Bataille R, Robillard N, et al.<br />

Expression of CD28 and CD40 in human myeloma<br />

cells: a comparative study with normal plasma cells.<br />

Blood 1994; 84:2597-603.<br />

153. Witzig TE, Gertz MA, Pineda AA, Kyle RA, Greipp PR.<br />

Detection of monoclonal plasma cells in the peripheral<br />

blood stem cell harvests of patients with multiple<br />

myeloma. Br J Haematol 1995; 89:640-2.<br />

154. Vora A, Toh C, Peel J, Greaves M. Use of granulocyte<br />

colony-stimulating factor (G-CSF) for mobilizing<br />

peripheral blood stem cells: risk of mobilizing clonal<br />

myeloma cells in patients with bone marrow infiltration.<br />

Br J Haematol 1994; 86:180-2.<br />

155. Gazitt Y, Reading C, Hoffman R, et al. Purified CD34 +<br />

Lin – Thy + stem cells do not contain clonal myeloma<br />

cells. Blood 1995; 86:381-9.<br />

156 Gazitt Y, Tian E, Barlogie B, et al. Differential mobilization<br />

of myeloma cells and normal hematopoietic<br />

stem cells in multiple myeloma after treatment with<br />

cyclophosphamide and granulocyte-macrophagecolony-stimulating-factor.<br />

Blood, 1996; 87:805-11.<br />

157. Björkstrand B, Ljungman P, Bird JM, Samson D,<br />

Garthon G. Double high-dose chemoradiotherapy with<br />

autologous stem cell transplantation can induce molecular<br />

remissions in multiple myeloma. Bone Marrow<br />

Transplant 1995; 15:367-71.<br />

158. Brenner MK, Rill DR, Moen RC, et al. Gene-marking to<br />

trace origin of relapse after autologous bone marrow<br />

transplantation. Lancet 1993; 341:85-6.<br />

159. Reece DE, Barnett MJ, Connors JM. Treatment of multiple<br />

myeloma with intensive chemotherapy followed<br />

by autologous BMT using marrow purged with 4-<br />

hydroperoxycyclophosphamide. Bone Marrow Transplant<br />

1993; 11:139-46.<br />

160. Gulati SC, Shimazaki C, Lemoli RM, Atzpodien J, Clarkson<br />

BD. Ex vivo treatment of myeloma cells by 4-HC,<br />

VP-16, LAK cells and antibodies. Eur J Haematol 1989;<br />

43(Suppl. 51):164-72.<br />

161. Siena S, Castro-Malaspina H, Gulati SC, et al. Effects<br />

of in vitro purging with 4-hydroperoxycyclophosphamide<br />

on the hematopoietic and microenvironment<br />

elements of human bone marrow. Blood 1985; 65:<br />

655-62.<br />

162. Bullock G, Tang C, Tourkina E, et al. Effect of combined<br />

treatment with interleukin-3 and interleukin-6<br />

on 4-hydroperoxycyclophosphamide induced programmed<br />

cell death or apoptosis in human myeloid<br />

leukemia cells. Exp Hematol 1993; 21:1640-4.<br />

163. Skorski T, Kawalec M, Hoser G, Ratajczcac M, Gnatowski<br />

B, Kawiak J. The kinetic of immunologic and<br />

hematologic recovery in mice after lethal total body<br />

irradiation and reconstitution with syngeneic bone<br />

marrow cells treated or untreated with mafosphamide<br />

(Asta Z 7654). Bone Marrow Transplant 1988; 3:543-<br />

51.<br />

164. Tong AW, Lee JC, Fay JW, Stone MJ. Elimination of<br />

clonogenic stem cells from human multiple myeloma<br />

cell lines by a plasma cell-reactive monoclonal antibody<br />

and complement. Blood 1987; 70:1482-9.<br />

165. Anderson KC, Andersen J, Soiffer R, et al. Monoclonal<br />

antibody-purged bone marrow transplantation therapy<br />

for multiple myeloma. Blood 1993; 82:2568-76.<br />

166. Gobbi M, Cavo M, Tazzari PL, et al. Autologous bone<br />

marrow transplantation with immunotoxin-purged<br />

marrow for advanced multiple myeloma. Eur J Haematol<br />

1989; 43 (Suppl. 51):176-81.<br />

167. Goldmacher VS, Bourret LA, Levine BA, et al. Anti-<br />

CD38-blocked ricin: an immunotoxin for the treatment<br />

of multiple myeloma. Blood 1994; 84:3017-25.<br />

168. Lemoli RM, Gobbi M, Tazzari PL, et al. Bone marrow<br />

purging for multiple myeloma by avidin-biotin<br />

immunoadsorption. Transplantation 1989; 47:385-7.<br />

169. Carlo Stella C, Cazzola M, De Fabritiis P, et al. CD34-<br />

positive cells: biology and clinical relevance. <strong>Haematologica</strong><br />

1995; 80: 367-87.<br />

170. Visani G, Lemoli RM, Dinota A, et al. Evidence that<br />

long-term bone marrow culture of patients with multiple<br />

myeloma favors normal hemopoietic proliferation.<br />

Transplantation 1989; 48:1026-31.<br />

171. Vescio RA, Hong CH, Cao J, et al. The hematopoietic<br />

stem cell antigen, CD34, is not expressed on the malignant<br />

cells in multiple myeloma. Blood 1994; 84:3283-<br />

90.<br />

172. Takishita M, Kosaka M, Goto T, Saito S. Cellular origin<br />

and extent of clonal involvement in multiple myeloma:<br />

genetic and phenotypic studies. Br J Haematol<br />

1994; 87:735-42.<br />

173. Avvisati G, Petrucci MT, Mandelli F. The role of biotherapies<br />

(interleukins, interferons and erythropoietin)<br />

in multiple myeloma. Baillière Clinical Haematol 1995;<br />

8:815-29.<br />

174. Mandelli F, Avvisati G, Amadori S, et sl. Maintenance<br />

treatment with recombinant interferon alpha-2b in<br />

patients with multiple myeloma responding to conventional<br />

induction chemotherapy. N Engl J Med 1990;<br />

322:1430-4.<br />

175. Westin J, Rodjer S, Turesson I et Al. Interferon alpha-<br />

2b versus no maintenance therapy during the plateau<br />

phase in multiple myeloma: a randomized study. Br J<br />

Haematol 1995; 89:561-8.<br />

176. Powels R, Cunningham D, Malpas JS, et al. A randomized<br />

trial of maintenance therapy with Intron-A<br />

following high-dose melphalan and ABMT in myeloma.<br />

Blood 1994; 84 (Suppl. 1):535a.<br />

177. De Laurenzi A, Iudicone P, Zoli V, et al. Recombinant<br />

interleukin-2 treatment before and after autologous<br />

stem cell transplantation in hematologic malignancies:<br />

clinical and immunologic effects. J Hematother 1995;<br />

4:113-20.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


eview<br />

Allogeneic hematopoietic stem cells<br />

from sources other than bone marrow:<br />

biological and technical aspects<br />

haematologica 2000; 85(supplement to no. 12):49-68<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia ed Oncologia<br />

Medica “Seràgnoli”, Policlinico S. Orsola, via Massarenti 9, 40138<br />

Bologna, Italy. Received August 22, 1996; accepted January 20,<br />

1997.<br />

FRANCESCO BERTOLINI, ARMANDO DE VINCENTIIS, LUIGI LANATA,<br />

ROBERTO M. LEMOLI, RITA MACCARIO, IGNAZIO MAJOLINO,<br />

LUISA PONCHIO, DAMIANO RONDELLI, ANTONIO TABILIO,<br />

PAOLA ZANON, SANTE TURA<br />

Division of Oncology, IRCCS Fondazione Maugeri, Pavia;<br />

Dompé Biotec SpA, Milan; Amgen Italia SpA, Milan; Istituto<br />

di Ematologia ed Oncologia Medica “Seràgnoli”, University of<br />

Bologna, Bologna; Department of Pediatrics, IRCCS Policlinico<br />

S. Matteo di Pavia, Pavia; Department of Hematology and<br />

BMT Unit, Ospedale “V. Cervello”, Palermo; Department of<br />

Internal Medicine, University of Pavia and IRCCS Policlinico S.<br />

Matteo, Pavia; Institute of Hematology, University of Perugia,<br />

Italy<br />

Background and Objectives. Identification and characterization<br />

of hematopoietic stem cells in peripheral<br />

blood (PB) and cord blood (CB) have suggested feasible<br />

alternatives to conventional allogeneic bone marrow<br />

(BM) transplantation. The growing interest in this<br />

use of allogeneic stem cells has prompted the Working<br />

Group on CD34-positive Hematopoietic Cells to review<br />

this subject by analyzing its biological and technical<br />

aspects.<br />

Evidence and Information Sources. The method used<br />

for preparing this review was informal consensus development.<br />

Members of the Working Group met three<br />

times, and the participants at these meetings examined<br />

a list of problems previously prepared by the<br />

chairman. They discussed the individual points in order<br />

to reach an agreement on the various concepts, and<br />

eventually approved the final manuscript. Some of the<br />

authors of the present review have been working in the<br />

field of hematopoietic stem cell biology and processing,<br />

and have contributed original papers published in<br />

peer-reviewed journals. In addition, the material examined<br />

in the present review includes articles and<br />

abstracts published in journals covered by the Science<br />

Citation Index® and Medline®.<br />

State of Art. Several studies have now shown that<br />

hematopoietic stem cells collected from peripheral<br />

blood after the administration of G-CSF, or from cord<br />

blood upon delivery, are capable of supporting rapid<br />

and complete reconstitution of BM function in allogeneic<br />

recipients. Perhaps more importantly, reinfusion<br />

of large numbers of HLA-matched T-cells from PB collections<br />

or T-cells with various degrees of HLA disparity<br />

from CB did not result in a higher incidence or greater<br />

severity of acute graft-versus-host disease than expected<br />

with BM. Based on the data reviewed, operative<br />

guidelines for mobilization, collection and graft processing<br />

are provided.<br />

Perspectives. It should be remembered that despite<br />

the growing interest, these procedures must be still<br />

considered as advanced clinical research and should<br />

be included in formal clinical trials aimed at demonstrating<br />

their definitive role in stem cell transplantation.<br />

In this regard, a large European randomized study is<br />

currently comparing PB and BM allografts. However,<br />

the possibility of collecting large quantities of<br />

hematopoietic progenitor-stem cells, perhaps with<br />

reduced allo-reactivity, offers an exciting perspective<br />

for widening the number of potential stem cell donors<br />

and greater leeway for graft manipulation than is possible<br />

with BM.<br />

©1997, Ferrata Storti Foundation<br />

Key words: hematopoietic stem cells, bone marrow, cord blood,<br />

peripheral blood, allogeneic transplantation, graft-versus-host disease<br />

Allogeneic bone marrow transplantation has progressed<br />

from a highly experimental procedure to<br />

being accepted as the preferred form of treatment<br />

for a wide variety of diseases. 1 There have been<br />

impressive improvements in this therapeutic procedure<br />

in the last two decades, but the most important<br />

advances probably took place in the last few years and<br />

concern the source of hematopoietic stem cells itself.<br />

Whereas this had always been by definition the bone<br />

marrow since the very beginning, identification of stem<br />

cells in peripheral and cord blood has now provided<br />

useful alternatives.<br />

In 1994 the growing interest in the use of peripheral<br />

blood stem cells (PBSC) in the setting of allogeneic<br />

bone marrow transplantation induced the GITMO<br />

(Gruppo Italiano Trapianto di Midollo Osseo) to promote<br />

a Study Committee for evaluating the key aspects of<br />

allogeneic PBSC collection and transplantation. This<br />

Committee produced a list of recommendations that<br />

were published as a position paper in this Journal at the<br />

beginning of 1995. 2 In summary, the authors strongly<br />

recommended the use of allogeneic PBSC in experienced<br />

centers, in well-defined clinical settings, and possibly<br />

– for the time being – in patients with advanced<br />

disease.<br />

As the use of PBSC expanded both in the autologous<br />

and the allogeneic setting, expression of the CD34 antigen<br />

became increasingly important for their character-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


50<br />

F. Bertolini et al.<br />

ization. An ad hoc working group reviewed the biology<br />

and clinical relevance of CD34-positive cells in this journal<br />

in 1995. 3 In particular, techniques for CD34-positive<br />

cell separation and procedures for their collection from<br />

peripheral blood were analyzed. A brief chapter was also<br />

devoted to CD34-positive cells in cord blood. 3<br />

The working group on CD34-positive hematopoietic<br />

cells subsequently reviewed the use of PBSC in acute<br />

myeloid leukemia 4 and multiple myeloma. 5 The growing<br />

interest in the use of PBSC and cord blood stem cells in<br />

the setting of allogeneic transplantation has now<br />

prompted the working group to review this subject by<br />

analyzing its biological and technical aspects.<br />

PBSC mobilization and collection in<br />

normal donors<br />

Until recently, the collection of hematopoietic cells for<br />

allogeneic transplantation has required general or spinal<br />

anesthesia and multiple punctures of iliac bones. However,<br />

marrow harvesting is not completely devoid of<br />

complications, side effects or patient discomfort. In a<br />

report on 1270 harvest procedures in Seattle, 6 6 donors<br />

suffered life-threatening complications and 10 showed<br />

significant operative site morbidity. As many as 10 percent<br />

of donations were associated with fever, and<br />

increasing donor age was significantly linked to poor<br />

cell harvest. In a different survey, 10 percent of donors<br />

recovered completely from marrow donation only more<br />

than 30 days after the procedure. 7<br />

PBSC transplantation represents an alternative<br />

approach. In autologous transplantation peripheral<br />

blood is now replacing bone marrow as a source of progenitor<br />

cells. 8 The advantage is quicker hematopoietic<br />

recovery 9,10 with consequently fewer complications and<br />

shorter hospital stay.<br />

In the autologous setting, PBSC can be collected after<br />

mobilization with chemotherapy, 11,12 growth factors, 13<br />

or a combination of the two. 14 In a randomized study,<br />

leukaphereses created less anxiety and pain than bone<br />

marrow harvest. 15<br />

Table 1. Relationship between CD34 + cell yield and G-CSF<br />

dose in allogeneic PBSC donors. Only clinical experiences<br />

are reported.<br />

Authors (ref.#)<br />

Donors<br />

No.<br />

G-CSF,<br />

dose/kg and<br />

days of<br />

administration<br />

CD34+<br />

collected<br />

x 10 6<br />

Apheresis<br />

No.<br />

Weaver (31) 4 16 µg, 4 d 9.6/kg 2<br />

Korbling (16) 9 12 µg, 7 d 13.1/kg 3<br />

Bensinger (18) 8 16 µg, 6 d 13.1/kg 2<br />

Schmitz (17) 8 5-10 µg, 5-6 d 6.7/kg 1-3<br />

Russell (34) 9 6-8 µg, 2-4 d 4.7/kg 1-2<br />

Majolino (27) 5<br />

6<br />

10 µg, 5 d<br />

16 µg, 4 d<br />

754<br />

789<br />

Tabilio (1996) 39 12 µg, 4-7 d 132.6 2-4<br />

2<br />

2<br />

In allogeneic transplantation, the use of PBSC has<br />

been somewhat delayed by a possible increase in graftversus-host<br />

disease (GVHD) as a consequence of the<br />

much higher number of lymphocytes in the graft inoculum,<br />

and by the need for a mobilization treatment for<br />

healthy individuals in order to obtain a good cell yield.<br />

However, the clinical experience of the last two years<br />

suggests that the incidence of acute GVHD is not<br />

increased with PBSC as compared to marrow, and that<br />

in healthy donors a sufficient cell number can be<br />

obtained by using growth factors alone, in particular G-<br />

CSF. 16-20 As a consequence, the number of allogeneic<br />

PBSC transplants is increasing rapidly. The European<br />

Blood and Marrow Transplant Group (EBMT) registered<br />

only 12 PBSC allografts in 1993, but their number<br />

increased to 180 in 1994 and to 537 in 1995 (Gratwohl,<br />

personal communication).<br />

Collection of PBSC in normal donors<br />

On biological grounds, there are several means of<br />

mobilizing progenitor cells into the peripheral blood, but<br />

their ultimate modality of action is always detachment<br />

of the CD34 + progenitor cell from marrow stroma and<br />

endothelium, to which it is normally bound by interactions<br />

with different integrin-adhesion molecules. 3,21 We<br />

may induce detachment either by an inhibition of the<br />

link between CD34 + cells and stroma, or by inducing a<br />

stress to the hematopoietic system capable of favoring<br />

the egress of progenitor cells from marrow to circulation.<br />

The former is obtained by means of monoclonal<br />

antibodies directed against adhesion molecules, 22 while<br />

the latter is based on the use of a drug or a combination<br />

of drugs. Richman et al. 23 demonstrated for the first<br />

time in man that chemotherapy-induced cytopenia is<br />

followed by a substantial increase of CFU-C in blood.<br />

In normal donors, however, the use of chemotherapy<br />

is ethically unacceptable, and only growth factors must<br />

be employed. Though a number of other cytokines are<br />

able to induce an increase of PBSC, only G-CSF and GM-<br />

CSF have been utilized in clinical practice. G-CSF in particular<br />

has an excellent mobilizing effect when used<br />

alone. 13,24-29<br />

The pilot experience with stem cell mobilization in<br />

normal donors is the one reported by the Seattle group.<br />

They administered G-CSF 300 µg/day or 6 µg/kg/day to<br />

increase WBC levels in apheresis collections from granulocyte<br />

donors. 30 A number of different schedules were<br />

later applied to mobilize PBSC for allogeneic transplantation.<br />

The results in terms of CD34 + cell collection are<br />

reported in Table 1. In most of the studies the G-CSF<br />

dose ranged from 10 to 16 µg/kg/day. With 16 µg/kg/day<br />

for 5 days, Weaver et al. 31 collected 1.6 to 12.6 (median<br />

9.6)×10 6 /kg CD34 + cells with two aphereses. All transplants<br />

were syngeneic, and recovery of 0.5×10 9 /L granulocytes<br />

and 20×10 9 /L platelets occurred on day 13 and<br />

10, respectively. With the same dose administered for 4<br />

days, Majolino et al. 27 were able to mobilize (median)<br />

147×10 6 /L CD34 + cells on day 4, a 65-fold increase over<br />

the baseline level. The median collection was 754×10 6<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

51<br />

CD34 + cells and 270×10 8 CD3 + cells with 2 aphereses.<br />

With 12 µg/kg for 6 days, Körbling et al. 16 collected a<br />

mean of 13.1×10 6 /kg CD34 + cells with 3 aphereses, and<br />

their patients recovered >0.510 9 /L granulocytes on day<br />

10 and >20×10 9 /L platelets on day 14. However, their<br />

short recovery times were also influenced by the absence<br />

of methotrexate from GVHD prophylaxis.<br />

The relationship between G-CSF dose and CD34 + cell<br />

mobilization is supported in part by the study by Dreger<br />

et al., 26 who compared 5 µg/kg/day and 10 µg/kg/day G-<br />

CSF in normal volunteers. They found 10 µg/kg to be<br />

superior in terms of progenitor cell yield. With higher<br />

doses the advantage seems to decline, and no statistical<br />

difference was found between 10 µg/kg for 5 days<br />

and 16 µg/kg for 4 days in a retrospective non-randomized<br />

study (Figure 1). 32 Dührsen 33 has suggested that<br />

the maximal effect in terms of progenitor cell increase<br />

is that obtained at a dose level of 10 µg/kg/day. This is<br />

also the dose recommended by the GITMO in its recently<br />

published guidelines. 2<br />

The number of apheretic procedures necessary for a<br />

good collection is critical for the donor, and may vary<br />

with the dose and schedule of G-CSF as well as with the<br />

volume processed.<br />

Bensinger et al. 28 routinely employ a schedule of 16<br />

µg/kg/day for 5-6 days in an effort to minimize the<br />

number of apheretic procedures. With this dose, a median<br />

of approximately 7×10 6 /kg CD34 + cells are obtained<br />

with a single apheresis performed on day 5. At the M.D.<br />

Anderson Cancer Center in Houston 29 a schedule of 12<br />

µg/kg/day for 4-6 days is used. With a single large volume<br />

apheresis the target CD34 + cell dose of > 4×10 6 /kg<br />

is reached in nearly 80% of the donors. Russell et al. 34<br />

mobilized their donors with 6-8 µg/kg/day for 2-4 days.<br />

By daily monitoring of CD34 + levels, the target of<br />

2.5×10 6 /kg CD34 + cells was achieved with a single 2-4<br />

hour harvest in 12 out of 14 donors. With 24 µg/kg/day<br />

Figure 1. Schematic representation of timing in PBSC mobilization<br />

in healthy donors. G-CSF is administered at a daily<br />

dose of 10 µg/kg, apheretic harvest is performed on day 5<br />

(and 6). Day 5 collection cells are stored at 4°C till the following<br />

day, when they are infused together with day 6 cells.<br />

Parentheses indicate that G-CSF is given and aphereses<br />

performed only if the target number of CD34 + cells is not<br />

reached with the day 5 apheretic run.<br />

G-CSF for 4 days Waller et al. 35 were able to collect<br />

13×10 6 /kg CD34 + with a single apheresis; however, one<br />

donor suffered severe side effects and the G-CSF dose<br />

had to be halved.<br />

The mobilization kinetics of PBSC under low daily doses<br />

of G-CSF has also been investigated. With 2.5<br />

µg/kg/day G-CSF on days 1 to 6 followed by 5.0<br />

µg/kg/day on days 7 to 10, a CFU-GM peak was obtained<br />

on day 6, but continuing G-CSF administration at 5<br />

µg/kg/day did not increase the level of circulating CFU-<br />

GM. 36<br />

With a single G-CSF dose of 15 µg/kg a significant rise<br />

in CD34 + cells, CFU-GM and BFU-E was obtained, 37 but<br />

the reported counts of 250/mL, 3.2×10 3 /mL and<br />

1.75×10 3 /mL, respectively, are not comparable with<br />

those obtained with prolonged administration schedules.<br />

Bishop et al. 38 reported their experience with G-CSF<br />

at 5 µg/kg/day. Aphereses began on day 4 of G-CSF<br />

administration. However, the target cell CD34 + dose of<br />

>1×10 6 /kg required 3 to 4 aphereses. With this method,<br />

median time to ANC > 0.5×10 9 /L was 12 days but all<br />

patients received G-CSF after the allograft.<br />

With G-CSF doses ranging from 10 to 16 µg/kg/day,<br />

the progenitor cell peak occurs on day 4 or 5. 2,20,27,28,32,39<br />

Since the CD34 + cell level rapidly declines after growth<br />

factor withdrawal, it is highly recommended that its<br />

administration be continued till the end of apheretic<br />

harvests.<br />

In both the Seattle and the GITMO experiences, the<br />

WBC peak occurred approximately the same day as the<br />

CD34 + cell peak. In the GITMO study, 39 the level of CD34 +<br />

cells reached a peak of (mean) 135.5×10 6 /L CD34 + cells,<br />

a 19-fold increase over the mean baseline level.<br />

Lymphocytes also increased, doubling their counts on<br />

day 5. A number of CD34 + cells > 4×10 6 /kg was collected<br />

in 51% of donors with a single apheresis, in 85%<br />

with two. Optimal collections are obtained on days 4<br />

and 5 of G-CSF administration. 28,39 It is likely that starting<br />

PBSC collection on day 4 is best when using 16<br />

µg/kg/day, whereas day 5 is better when lower doses are<br />

employed (Figure 2).<br />

In normal volunteers GM-CSF has found application<br />

less frequently than G-CSF. Lane et al. 40 studied G- and<br />

GM-CSF alone and a combination of the two. The total<br />

number of CD34 + cells collected from the G-CSF group<br />

with a single apheresis was 119×10 6 , and was not significantly<br />

different from that collected from the group<br />

treated with G- and GM-CSF (101×10 6 CD34 + cells), but<br />

both were greater than that from the group treated with<br />

GM-CSF (12.6×10 6 ). However, a higher fraction of an<br />

early CD34 + /HLA-DR – /CD38 – cell population was found<br />

among the CD34 + cells after GM-CSF administration.<br />

Whether this early fraction is associated with more rapid<br />

engraftment is presently unknown.<br />

Predictive factors for progenitor cell yield have not<br />

been studied in normal volunteers. Though there is anecdotal<br />

experience of donors failing to respond, only age<br />

was reported to influence the quality of collections in a<br />

single study. 26<br />

haematologica vol. 85(suppl. to n. 12):December 2000


52<br />

F. Bertolini et al.<br />

The number of PBSC necessary for rapid and stable<br />

engraftment is unknown. In the autologous setting a<br />

dose of >2×10 6 /kg CD34 + cells has been suggested, 41<br />

but the requirement might be higher in allogeneic transplantations<br />

as a consequence of the immunological<br />

mechanisms involved. In Seattle 28 4 out of the 53 normal<br />

donors yielded only 0.6, 1.49, 1.55 and 1.74×10 6<br />

CD34 + cells/kg. Despite the low cell numbers, successful<br />

engraftment was achieved in all cases. We suggest<br />

that collection of >4×10 6 /kg CD34 + cells is the target for<br />

a safe allogeneic transplantation. Lower doses, however,<br />

may be sufficient. A lower limit of >210 6 /kg CD34 +<br />

cells would be reasonable for those patients whose<br />

donors respond poorly to cell mobilization.<br />

There are currently no contraindications to cryopreservation<br />

of cells for later use after thawing. Though<br />

most centers currently infuse freshly collected apheresis<br />

products, one may consider the advantage of separating<br />

the mobilization/collection phase from transplantation<br />

in terms of logistics and patient safety.<br />

Side effects and toxicity of the procedure<br />

Early toxic effects of G-CSF in healthy donors are now<br />

well known. The GITMO survey 39 on 76 healthy subjects<br />

aged 6 to 67 years receiving G-CSF for PBSC mobilization<br />

reveals that the side effects of G-CSF administration<br />

are acceptable, the only problem being moderate to<br />

severe bone pain in 13% of donors (Table 2).<br />

Twenty-three percent of donors also said the<br />

apheretic procedures were demanding. Comparable side<br />

effects are reported in other studies. 34,28 Additional problems<br />

could include pneumothorax due to jugular vein<br />

cannulation and paresthesia. 34 Nonetheless, donors who<br />

had previously given marrow mostly agreed that they<br />

preferred blood cell mobilization and collection to marrow<br />

harvest. 34-39 A good policy would be to avoid the use<br />

of venous catheters. In autologous PBSC harvesting<br />

where mobilization treatment often includes chemotherapy,<br />

central venous catheter (CVC) occlusion necessitating<br />

thrombolytic therapy was the most commonly<br />

Figure 2. Variations of blood cell counts in normal donors<br />

during G-CSF treatment and apheretic collection of mononuclear<br />

cells. The curves represent mean values. Data are<br />

those of 76 normal donors. 39<br />

Table 2. Incidence and grading of side effects reported during<br />

G-CSF administration in 76 healthy donors from the GIT-<br />

MO. 39<br />

% donors<br />

Absent Mild Moderate Severe<br />

Bone pain 27.4 59.6 9.6 3.2<br />

Arthralgias 54.8 32.2 11.2 1.6<br />

Headache 70.9 20.9 8 0<br />

Fatigue 74.5 22 3.3 0<br />

Fever 91.5 6.7 1.6 0<br />

observed complication, occurring in 15.9% of CVC-aided<br />

collections. 42<br />

Variations in blood counts mainly consist of a pronounced<br />

WBC increase, a moderate thrombocytopenia<br />

and a slight decrease of hematocrit values. In the Italian<br />

survey, thrombocytopenia from mild (< 70×10 9 /L) to<br />

moderate (< 50×10 9 /L) followed PBSC harvests in 40%<br />

and 10% of cases, respectively. WBC counts exceeded<br />

50×10 9 /L in 40% of cases, and 70×10 9 /L in 8%. Bensinger<br />

et al. 28 report their experience with 124 donors treated<br />

with G-CSF at various doses and scheduling. Forty-one<br />

were granulocyte donors, while 13 were PBSC syngeneic<br />

and 63 allogeneic donors. One donor had a myocardial<br />

infarction after the first apheresis, but he had a previous<br />

history of infarction. Thrombocytopenia was in part<br />

related to G-CSF dosage, in part to the volume of blood<br />

processed. A count


Non bone marrow allogeneic hemopoietic stem cells<br />

53<br />

The studies on leukemia development after G-CSF<br />

treatment must be considered with caution. A Japanese<br />

group 46 reported data on 170 children with aplastic anemia.<br />

Eleven out of the 108 receiving G-CSF had a transformation<br />

to MDS or leukemia, while this evolution<br />

occurred in none of the 62 patients not receiving G-<br />

CSF. Another study 47 reports the evolution to<br />

MDS/leukemia in 13 patients with congenital neutropenia<br />

treated with G-CSF, with the occurrence of<br />

monosomy 7 in 10 of them. However, as suggested by<br />

Smith et al. 48 in a study on the leukemic evolution of<br />

Kostmann’s disease, the fact that congenital neutropenia<br />

may evolve into MDS and AML under G-CSF treatment<br />

has no implication for normal donors, since it is<br />

the underlying hemopoietic defect that represents a preleukemic<br />

condition. In fact, in Kostmann’s disease not all<br />

the chromosomal aberrations involve chromosome 7,<br />

and when other abnormalities are detected leukemia<br />

does not develop.<br />

The use of G-CSF for mobilization of PBSC in children<br />

should be considered with more attention. Though there<br />

may be a specific advantage in collecting PBSC from<br />

children in the case of considerable disparity in body<br />

weight with the recipient, the GITMO stated that such<br />

a practice should be discouraged in standard allogeneic<br />

transplants. 2 This is also the opinion of the Italian<br />

Association of Pediatric Hemato-Oncology (AIEOP).<br />

PBSC have also been employed for allogeneic engraftment<br />

in MUD transplants. A small series was presented<br />

by Ringdén et al. 49 in Geneva. Six patients with high risk<br />

hematological malignancies received PBSC from unrelated<br />

donors, 4 of them as primary treatment and 2 for<br />

treatment of graft failure. For PBSC mobilization the<br />

donors received G-CSF 5 to 12 µg/kg and leukaphereses<br />

were performed using continuous flow devices. One<br />

donor complained of rib pain and one of nausea, dizziness<br />

and anxiety.<br />

One advantage of using PBSC for MUD transplants<br />

could derive from the higher number of progenitor cells,<br />

with better engraftment and reduction of failures. For<br />

the donor, the chance of obtaining stem cells for unrelated<br />

transplants without the need for general anesthesia<br />

is certainly appealing, and would probably encourage<br />

more volunteers to donate stem cells. It would also be<br />

easier to expand especially the number of donors belonging<br />

to ethnic minorities. Apheresis-derived mononuclear<br />

cells might be stored in liquid nitrogen and shipped when<br />

needed. Age limit for donors could be expanded. However,<br />

because of the limited experience with G-CSF<br />

mobilization in normal donors, National Marrow Donor<br />

Registries have not approved the use of PBSC as first<br />

choice. We expect this will remain the case in the foreseeable<br />

future.<br />

In general, the use of growth factors for any purpose<br />

in healthy subjects should be considered experimental.<br />

The donor should be informed of the potential short and<br />

long-term risks of growth factors and leukapheresis, as<br />

well as of anesthesia, and he should be given the possibility<br />

of choosing. Donor consent should be asked on<br />

the basis of a protocol previously approved by an official<br />

ethical committee. 2<br />

Characterization of CD34 + hematopoietic<br />

progenitor cells mobilized into peripheral<br />

blood of normal donors by rHG-CSF<br />

The preliminary results of clinical trials on allogeneic<br />

PBSC transplantation have demonstrated the capacity of<br />

G-CSF to mobilize true stem cells capable of long-term<br />

reconstitution of marrow function. Moreover, similarly<br />

to autografting, the most striking finding of PBSC transplantation<br />

has been the faster recovery of hematopoiesis<br />

after myeloablative conditioning regimens as compared<br />

to transplantation of BM-derived stem cells. Thus, clinical<br />

investigators asked the question of whether circulating<br />

progenitor cells may differ from their BM counterparts<br />

with respect to kinetic status, immunophenotype,<br />

frequency of both committed and primitive precursors,<br />

and their proliferative response to colony stimulating<br />

factors (CSFs).<br />

One early report 50 has shown a high expression of<br />

myeloid antigens on PB CD34 + cells (i.e. CD33, CD13) at<br />

the expense of B-lineage-associated antigens (i.e. CD10,<br />

CD19, CD20), coupled with a high colony-forming<br />

capacity of G-CSF-stimulated apheresis products. Moreover,<br />

Roberts and Metcalf 51 have clearly shown in an<br />

animal model and in humans that only a small minority<br />

of mobilized PBSC undergo active DNA synthesis,<br />

whereas BM cells contain more than 30% of S-phase<br />

clonogenic progenitors. This finding, coupled with the<br />

lack of expression of CD71 antigen (transferrin receptor)<br />

and the Rhodamine 123 dull status 52 observed in CD34 +<br />

cells from cancer patients mobilized with G-CSF, has<br />

suggested that PB progenitors may be functionally inactive<br />

since they are in deep G0-phase of the cell cycle.<br />

However, these results are somewhat in contrast with<br />

clinical data indicating rapid BM recovery after autologous<br />

and allogeneic transplantation and the experimental<br />

evidence that circulating CD34 + cells represent<br />

an optimal target for efficient retroviral infection requiring<br />

cell cycling for integration. 53 In addition, it is very<br />

important to assess the kinetic profile of the CD34 + cell<br />

fractions which are believed to ensure permanent<br />

engraftment after PBSC allografting, such as cells phenotypically<br />

identified as CD34 + /CD38 – , CD34 + /CD33 – /<br />

HLA-DR – or very primitive progenitor cells capable of<br />

generating clonogenic precursors in secondary semisolid<br />

assay after 5 or more weeks of liquid culture, longterm<br />

culture-initiating cells (LTC-IC). In this regard,<br />

defective long-term repopulating activity of early BM<br />

cells induced to S-phase by cytokines has recently been<br />

shown. 54<br />

To further elucidate the phenotypic profile and functional<br />

and kinetic characteristics of G-CSF-mobilized<br />

hematopoietic progenitor cells, highly purified CD34 +<br />

cells from the apheresis products of normal individuals<br />

undergoing PBSC collections for allogeneic transplantation<br />

were recently analyzed. The results were then com-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


54<br />

F. Bertolini et al.<br />

Cell-cycle distribution of CD34+ cells (%)<br />

Figure 3. Analysis of cell-cycle distribution of PB and BM<br />

CD34 + cells.<br />

pared with those obtained on CD34 + cells enriched from<br />

the BM of the same donors under steady-state conditions<br />

and after G-CSF administration on the same day as<br />

PBSC harvest. 55 The results confirmed the expression of<br />

CD33 and CD13 antigens on a higher percentage of circulating<br />

CD34 + cells compared to BM cells (91±31% SD<br />

and 85.3±10% SD versus 51.1±21% SD and 64.6±25%<br />

SD, respectively; p


Non bone marrow allogeneic hemopoietic stem cells<br />

55<br />

adhesion molecules critical for mobilization and related<br />

to cytokine-induced cell-cycle transit. 62 Moreover,<br />

pharmacological doses of steel factor determine a redistribution<br />

of stem cells in mice 63 and reduce the avidity<br />

of 4 1 and 5 1 integrins on the MO7e cell line, with<br />

a consequent inhibition of the specific cell adhesion of<br />

MO7e cells to VCAM-164. Other adhesion molecules,<br />

like L-selectin and VLA4, might play a role in the mobilization<br />

of hematopoietic progenitors in primates. 65<br />

Figure 4. Recovery and proliferative status of CFC and LTC-<br />

IC in steady-state normal blood and bone marrow and in the<br />

leukapheresis products of cancer patients obtained after<br />

chemotherapy + G-CSF. The cells were cultured for 16 hrs<br />

in a medium containing serum substitutes, SF (100<br />

ng/mL), IL-3 (20 ng/mL) and G-CSF (20 ng/mL) in the presence<br />

or absence of 3 H-Tdr (panel B and A, respectively).<br />

SEM of BM), whereas the proportion of LTC-IC cycling<br />

was superimposable on that of BM. The frequency was<br />

not different in the two compartments (48.2±35 SEM<br />

and 62.5±54 SEM for 10 4 CD34 + cells in PB and BM,<br />

respectively; p = ns). Thus these data, coupled with previous<br />

observations from nonhuman primates on cellcycle<br />

status and response to CSF of cytokine-mobilized<br />

CD34 + cells, 61 suggest that many circulating progenitors<br />

are not deeply quiescent in G0-phase. Rather, they<br />

are actively cycling and their high clonogenic efficiency<br />

and prompt proliferative response to CSFs may indicate<br />

a faster progression through cell-cycle mediated,<br />

perhaps, by G-CSF priming. A kinetic and functional pattern<br />

of CD34 + cells similar to that observed in normal<br />

PBSC donors has been found in acute leukemia and multiple<br />

myeloma patients mobilized with chemotherapy<br />

and G-CSF, and in lymphoma individuals receiving G-<br />

CSF alone (Lemoli, unpublished observations). Thus,<br />

regardless of the mobilization protocol, the administration<br />

of G-CSF and/or the change of compartment (i.e.<br />

egress into peripheral blood) induces a profound effect<br />

on the characteristics of hematopoietic progenitor cells.<br />

Further studies are presently directed toward investigating<br />

the modulation of the expression of integrin<br />

Stem cells from umbilical cord blood:<br />

biological aspects<br />

More than 20 years ago it was described in this Journal<br />

that hematopoietic progenitors circulate between<br />

the fetus and the placenta during gestation. 66 However,<br />

placental/umbilical cord blood (CB) from human<br />

newborns was not considered as a source of stem cells<br />

for clinical use until Broxmeyer et al. 67 enumerated the<br />

number of CFU-GM that could be collected from the CB<br />

remaining in the placenta after birth and suggested that<br />

the total number was sufficient for transplantation in<br />

pediatric patients. The Fanconi anemia patient who in<br />

1988 first received a CB transplant from his HLAmatched<br />

sibling 68 is still alive at the present time and<br />

cured from the hematological point of view, thus<br />

demonstrating the long-term engraftment capability of<br />

CB-derived stem cells.<br />

In the past five years interest in the biological aspects<br />

and clinical applications of CB has grown since large<br />

CB banks for unrelated stem cell transplantation have<br />

been implemented in the USA and Europe, and more<br />

than 300 CB transplants have been performed. However,<br />

many aspects of the properties of CB stem cells are<br />

still elusive. It is remarkable that a unit of CB used for<br />

transplantation contains 1-810 6 CD34 + cells and 10-<br />

12010 3 CAFC/LTC-IC, 56,69-70 i.e. 1-2 logs fewer than the<br />

total number of CD34 + cells and CAFC usually infused<br />

into recipients of allogeneic BM or PBSC. On average,<br />

recipients of CB transplants are given 0.05-0.5×10 6<br />

CD34 + cells/kg b.w., while it has been suggested that<br />

recipients of allogeneic PBSC must receive at least 2.5-<br />

5×10 6 CD34 + cells/kg b.w. to obtain safe hematopoietic<br />

engraftment. 71 On the other hand, despite the delay<br />

in the reconstitution of the megakaryocytic lineage, the<br />

rate of engraftment failure in CB transplant recipients<br />

is similar to that observed after BM or PBSC transplants.<br />

72 These observations have prompted a number of<br />

investigators to focus on the proliferation potential of<br />

CB stem cells. In an elegant study, Lansdorp et al. 73 sorted<br />

CB-derived, CD34 + CD45RA lo CD71 lo cells, defined as<br />

stem cell candidates. In liquid cultures supplemented<br />

with SCF, IL-3,-6 and Epo, these purified cells generated<br />

a number of CD34 + and mature cells significantly<br />

greater than that obtained in cultures of purified CD34 +<br />

CD45RA lo CD71 lo cells obtained from adult donors. This<br />

advantage was clearly ontogeny-related, since the proliferative<br />

potential of purified CD34 + CD45RA lo CD71 lo<br />

cells collected from fetal liver was superior to that of<br />

CB-derived cells. In this context, Hows et al. 74 demon-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


56<br />

F. Bertolini et al.<br />

strated in long-term stroma culture that both progenitor<br />

cell cultures and the lifespan of cultures were greater<br />

in CB than in adult BM, and Payne et al. 75 showed that<br />

the proportion of CD34 + cells that are CD38 – (lin – ) is<br />

significantly higher in CB than in other stem cell sources.<br />

In contrast to what has been documented in adult BM,<br />

Traycoff et al. 76 demonstrated that LTC-IC and cells presumably<br />

capable of in vivo engraftment reside in the<br />

CD34 + HLA-DR + Rh123 dull fraction. The cycling status of<br />

CB progenitors is still a matter of debate. In fact, some<br />

investigators using the tymidine suicide technique<br />

reported a higher frequency of ceIls in S phase, 77 whereas<br />

others did not find any difference between CB and<br />

adult BM in the frequency of actively cycling progenitors.<br />

78 More insight into this area is especially necessary<br />

since CB is a very attractive target for the transfer of<br />

genes able to correct inherited or non-inherited diseases<br />

such as thalassemia, Fanconi anemia, ADA-deficiency,<br />

etc, 79 and since entering S phase is required for gene<br />

transfer through safety modified retroviruses. Interestingly,<br />

a higher efficiency of retrovirus-mediated gene<br />

trasfer has been reported in CB than in BM progenitors.<br />

78,80,81 One possible explanation is the particularly<br />

rapid exit from the G0/G1 phases of the cell cycle in<br />

response to cytokines described by Traycoff et al. 82 in<br />

CB-derived CD34 + progenitors, which might also justify<br />

the ontogeny-related advantage in proliferative<br />

potential.<br />

The functional meaning of these differences in the in<br />

vitro behavior of phenotypically defined CB and BM populations<br />

is not yet fully understood, but these findings<br />

represent an interesting parameter to consider when<br />

assessing the suitability of a CB unit for transplantation<br />

in pediatric or adult patients. So far, in fact, most CB<br />

transplant recipients have been pediatric patients<br />

weighing less than 50 kg. Sporadic reports of CB transplants<br />

in adult recipients have indicated that the time<br />

to myeloid lineage engraftment is comparable to that of<br />

BM recipients, whereas the delay in platelet reconstitution<br />

seems to be more pronounced than in pediatric CB<br />

transplant recipients. 83 As described in the CB processing<br />

section, ex vivo expansion of CB progenitors prior to<br />

trasplantation might be useful to hasten hematopoietic<br />

engraftment; however, since the long-term engraftment<br />

capability of ex vivo cultured cells might be lost or<br />

impaired, 84 more work seems necessary to reach this<br />

important goal.<br />

CB collection<br />

Established advantages of CB banks over BM donor<br />

registries include the immediate availability of the frozen<br />

CB unit, minimal donor attrition, the presence of CB<br />

donors from minority groups that are poorly represented<br />

in BM donors registeries, and a much lower incidence<br />

of CMV infected donors. In fact, the time from the<br />

request for a CB unit to finding a matched donor is on<br />

average less than 2 months, and less than 1% of CB units<br />

are contaminated by CMV. 85<br />

Worldwide, the creation of large CB banks has prompted<br />

investigators to improve the methods for CB collection<br />

and fractionation. The first method described by<br />

Broxmeyer et al. 67 included CB collection in heparinized<br />

tubes by gravity. Further studies 69,86 indicated that this<br />

open system is much more prone to bacterial contamination<br />

than closed systems based upon CB collection in<br />

bags, as first proposed by Gluckman et al. 87 Another<br />

approach, proposed by Turner et al., 88 includes catheterization<br />

of the umbilical vein. However, in a recent report<br />

this procedure was found to cause significant contamination<br />

of the CB collection by maternal cells, including<br />

potentially harmful T cells. 89 It has been demonstrated,<br />

moreover, that CPD/CPD-A have an advantage over ACD<br />

and heparin because the former can anticoagulate blood<br />

over a wider volume range. Figure 5 describes results<br />

obtained using the method of CB collection in closed<br />

bags while the placenta is still in utero. Briefly, after the<br />

birth the umbilical cord is doubly clamped 1-2 cm from<br />

the newborn and transected before the newborn is<br />

removed from the operative field. The free end of the<br />

cord must be accurately disinfected before CB collection<br />

by venepuncture of the umbilical vein. As shown in Figure<br />

5, there is a strict correlation between the time of<br />

umbilical cord clamping, the volume and the total number<br />

of nucleated cells collected. If the clamping procedure<br />

is delayed to the second minute after birth, it seems difficult<br />

to collect systematically a number of nucleated<br />

cells sufficient for clinical use of the CB unit. In fact,<br />

after the birth the newborn is frequently positioned<br />

below the level of the uterus, and this determines the socalled<br />

transfusion effect from the placenta to the newborn.<br />

90,91 Interestingly, when newborns are delivered following<br />

the Leboyer method there is no transfusion effect<br />

since the newborn is kept above the level of uterus. 92<br />

Under these circumstances, early clamping of the cord is<br />

not required for collection of CB for clinical use. At the<br />

present time there is no consensus among neonatologists<br />

and pediatricians about the more appropriate timing<br />

of umbilical cord clamping. However, cord clamping<br />

in the first 30-60” after birth seems adequate to most<br />

reviewers, 93-97 and recent data on the immediate followup<br />

of newborns who underwent early clamping of the<br />

the umbilical cord and CB collection support the safety<br />

of this procedure. 98 In this retrospective study, none of<br />

the newborns who had CB collected were reported to<br />

suffer from weight loss, fatigue while feeding, tachypnea<br />

and tachycardia, hypoxia, or cardiac or pulmonary<br />

disease with reduced arterial oxygen saturation. The sole<br />

significant difference between the group of 59 CB donors<br />

and the control group was a slight reduction of Hb values,<br />

which corresponded to a loss of about 15 to 20 mg<br />

of iron.<br />

CB processing<br />

As mentioned above, a CB unit to be used for transplantation<br />

contains remarkably fewer CD34 + progenitor<br />

cells than BM or PBSC collections. For this reason, during<br />

CB manipulation the loss of progenitor cells must be<br />

carefully avoided. In pilot projects for large scale bank-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

57<br />

ing, CB was in fact stored as unmanipulated whole<br />

blood. The high cost of this procedure, which requires<br />

large liquid nitrogen space, has fueled intense research<br />

to concentrate CB nucleated cells in a reduced volume.<br />

Among the methods recently proposed for CB processing,<br />

however, some included density separation by<br />

Ficoll 99 or red cell sedimentation by means of animal<br />

gelatin, 69,100 i.e. reagents that are currently not (and<br />

probably will never be) licensed for use in humans by<br />

regulatory agencies like the the FDA. Consequently, procedures<br />

involving the use of licensed products like<br />

HES 101,102 should be recommended and a maximum loss<br />

of 10-15% of progenitors accepted. In this context, it<br />

must be noted that a number of patients have already<br />

been successfully transplanted with red cell-depleted<br />

CB. 101,103 Conversely, data on the engraftment potential<br />

of purified, CB-derived CD34 + cells are still poorly reproducible<br />

in the SCID-hu mouse model 104 and totally lacking<br />

in humans, so for the time being the storage of purified<br />

CD34 + CB progenitors for clincal use is not recommended.<br />

However, it has been shown in vitro that the<br />

proliferation potential of purified CD34 + cells is markedly<br />

superior to that of the low density or Ficoll fraction.<br />

70,104 This finding has major implications for the possible<br />

ex vivo expansion of an aliquot of the CB unit prior<br />

to transplantation. Two different strategies have been<br />

evaluated: the goal of some authors was to obtain multiple<br />

lineage expansion of progenitors by means of<br />

cytokine combinations including SCF, FLT-ligand IL-1,<br />

IL-3, IL-6, IL-11, G- and GM-CSF, 104-106 while others were<br />

interested in selected-lineage expansion of the<br />

myeloid 107 or megakaryocytic lineage. 108<br />

Rubinstein et al. 101 have recently proposed a new procedure<br />

for washing the CB unit prior to transplantation.<br />

Advantages of this approach include removal of free Hb<br />

from lysed red cells and a significant reduction in the<br />

DMSO infused, a molecule which is particularly toxic<br />

for pediatric transplant recipients. 109 In vitro data indicate<br />

that the washing procedure may improve the<br />

engraftment potential of the transplanted cells, but this<br />

finding should be futher confirmed in an in vivo model.<br />

Figure 5. Effect of the time of umbilical cord clamping on the<br />

mean (±1 SD) volume and nucleated cell count of cord blood<br />

collections (n=67). As in most European OB/GYN units,<br />

soon after birth the newborn is kept below the level of the<br />

uterus, and delayed clamping of the umbilical cord is associated<br />

with a reduction of cord blood volume and cellular<br />

content.<br />

Immunological features of cord blood<br />

lymphocytes<br />

The immune system which develops during fetal life<br />

is not fully competent at birth and continues the differentiatation<br />

process after birth in response to various<br />

antigenic challenges. At least three important elements<br />

control the development of the immune system during<br />

fetal life, thus determining the peculiar characteristics<br />

of the cord blood lymphocyte (CBL)-mediated immune<br />

response: i) limited or even absent antigenic experience,<br />

ii) immaturity of the majority of lymphocyte populations,<br />

and iii) feto-maternal immunological interaction.<br />

110,111 These elements are believed to influence the<br />

immunological features most strictly related to cord<br />

blood transplantation (CBT) and, in particular, the<br />

capacity to develop alloantigen-directed reactivity, antimicrobial<br />

immunity and anti-tumor immune surveillance.<br />

As a consequence of poor antigenic experience during<br />

pregnancy, the majority of CBL are naive cells expressing<br />

the RA isoform of the CD45 molecule. 112 The most<br />

prominent immunoregulatory function of CD45RA + T<br />

lymphocytes is suppressor activity. 113 These peculiar features<br />

of neonatal lymphocytes explain their incapacity to<br />

develop, both in vivo 114 and in vitro, 115 an immune<br />

response directed towards recall antigens (i.e. tetanus<br />

toxoid, influenza virus).<br />

The immaturity of the CBL population is also a direct<br />

consequence of its poor antigenic experience. Compared<br />

to adult blood, the distribution of the most relevant CBL<br />

subpopulations is characterized by a reduced percentage<br />

of CD3 + mature T lymphocytes and by the presence<br />

of immature T and NK lymphocyte subsets which are<br />

not detectable in adult peripheral blood. 116,117<br />

B lymphocytes are present in a normal or even augmented<br />

percentage in cord blood as compared with adult<br />

blood, even though immunoglobulin production is limited<br />

to the IgM class. 118 Other CBL peculiar features related<br />

to their immaturity are low expression of adhesion/costimulation<br />

molecules such as CD11a (LFA-1),<br />

CD18, CD54 (ICAM-1), CD58 (LFA-3) antigens, 113,117,119<br />

poor expression of CD40 ligand on activated T lymphocytes,<br />

120 and reduced ability to secrete some cytokines<br />

(i.e. -interferon, tumor necrosis factor and interleukin-<br />

4). 117<br />

Spontaneous NK activity is reduced in cord blood as<br />

compared to adult blood, possibly because of the low<br />

expression of adhesion molecules, known to be useful in<br />

promoting the capacity of NK lymphocytes to adhere to<br />

target cells. 121 On the other hand, antibody-dependent<br />

cell cytotoxicity (ADCC) and lymphokine activated killer<br />

(LAK) activity of cord blood reach values comparable to<br />

or even higher than those observed in adult blood. 121<br />

Moreover, recent data demonstrate that the innate<br />

haematologica vol. 85(suppl. to n. 12):December 2000


58<br />

F. Bertolini et al.<br />

immunity directed towards Epstein-Barr virus-infected<br />

cells is remarkably high in CBL collected from the majority<br />

of neonates. 122 The capacity of cord blood NK cells to<br />

be promptly activated in vitro suggests that innate<br />

immunity plays a key role in immune surveillance during<br />

fetal and perinatal life, as long as specific T-cell<br />

mediated adaptive immunity can be generated.<br />

From an immunological viewpoint, pregnancy can be<br />

considered as a successful HLA-mismatched transplantation.<br />

It is well known that the feto-maternal, anatomo-functional<br />

barrier allows the reciprocal transfer of<br />

lymphocyte subpopulations. It is thus conceivable to<br />

hypothesize that a very effective immunological network<br />

acts to prevent fetal rejection and graft-versushost<br />

reactions (GVHR). 111,123 Several lines of clinical and<br />

experimental evidence support this hypothesis, in particular:<br />

i) CBL preferentially display suppressor rather<br />

than helper immunological functions 111,113 and ii) both<br />

B and T lymphocytes maintain a state of hyporesponsivity<br />

towards noninherited maternal HLA molecules for<br />

a long time after birth. 124,125 A further confirmation of<br />

this peculiar state of tolerance derives from a recently<br />

reported observation 126 on the occurrence of acute<br />

GVHD in patients given CBT from donors who were disparate<br />

for the noninherited paternal allele, and on the<br />

absence of significant acute GVHD in recipients whose<br />

donors were disparate for the noninherited maternal<br />

allele.<br />

The fetal/neonatal period has been postulated to represent<br />

a crucial time in ontogeny, during which T and B<br />

lymphocytes learn to discriminate between self and nonself<br />

through the development of a state of tolerance<br />

toward antigens they encounter. 127 The concept of neonatal<br />

tolerance was recently re-examined in mice 128-130<br />

and it was demonstrated that induction of this phenomenon<br />

may depend on several elements, including<br />

the nature of the antigen-presenting cells, 128 the dose<br />

of antigen administered, 129 and the mode of immunization.<br />

130<br />

Poor antigenic experience, immaturity of lymphocyte<br />

subpopulations, feto-maternal immunological interactions,<br />

and neonatal tolerance may, altogether, contribute<br />

to the generation of a suppressive effect on CBL<br />

alloreactivity, thus permitting the use of HLA-partially<br />

matched donors for CBT. In agreement with this hypothesis,<br />

reduced proliferative and cytotoxic activity towards<br />

alloantigens was reported by several authors to be present<br />

in cord blood as compared with adult peripheral<br />

blood. 117,131-133 However, normal CBL alloreactivity has<br />

been documented in other studies. 134,135 The discrepancies<br />

observed between the above mentioned reports may<br />

depend on the high interindividual variability in the distribution<br />

of cord blood T and NK lymphocyte subpopulations.<br />

Interestingly, it has been recently reported that,<br />

even though proliferative response to alloantigen in a<br />

primary mixed lymphocyte culture (MLC) is comparable<br />

in adult and cord blood, restimulation in secondary MLC<br />

induces increased specificity and activity of adult alloreactive<br />

lymphocytes and a state of unresponsiveness in<br />

CBL. 136 These data strongly suggest that repeated in vitro<br />

stimulation with allogeneic cells amplifies the specific<br />

immune response of adult lymphocytes, while the same<br />

procedure induces a state of anergy in neonatal cells.<br />

Several clinical and experimental data obtained in the<br />

setting of allogeneic bone marrow transplantation<br />

(BMT) demonstrate that there is a strong correlation<br />

between GVHD and graft-versus-leukemia (GVL) effect.<br />

Thus, due to their low alloreactive capacity (responsible<br />

for the reduced GVHR), 126,135 CBL could be less efficient<br />

in mediating a GVL effect. As far as we know, no data<br />

concerning the identification of cord blood T lymphocytes<br />

capable of mediating specific anti-leukemic activity<br />

have been reported in the literature. This lack of<br />

information is not surprising since it is well known that<br />

the frequency of these cells is extremely low, even in the<br />

peripheral blood of healthy adult donors. However, some<br />

studies have recently demonstrated that innate antileukemic<br />

activity mediated by LAK cells and measured<br />

Table 3. Methods of T-Cell depletion in clinical trials.<br />

Method of T-Cell Depletion Cells Removed T-Cell Depletion<br />

(x log10)<br />

SBA lectin and E-rosette depletion<br />

T and B lymphocytes, monocytes,<br />

neutrophils 2.5 - 3.0<br />

Multiple E-rosette depletions T lymphocytes 2.0<br />

Mouse MoAb (anti-CD2, CD8) + rabbit C’ T lymphocytes 2.0<br />

Mouse MoAb (anti CD6) + human C’ T lymphocytes 1.5-2.0<br />

Rat MoAb(CAMPATH-1) + human C’ T and B lymphocytes, monocytes, 2.5<br />

Anti-CD5 immunotoxin-Ricin A Immunomagnetic T lymphocytes 2.0<br />

separation (anti-CD3, CD8)<br />

SBA lectin + immunomagnetic separation<br />

T and B lymphocytes, monocytes,<br />

3.1<br />

neutrophils<br />

AIS CD%/8T-CELLector T lymphocytes 2.5<br />

Autologous Immunorosettes<br />

(anti-CD2 and CD3 tetrametric complexes)<br />

T lymphocytes 2-3<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

59<br />

against long-term tumor cell lines, is comparable in<br />

adult and cord blood. 119,137 Even though the above mentioned<br />

studies are interesting, more experimental data<br />

and clinical observations are required to better define<br />

the potential GVL effect of CBL.<br />

PBSC processing<br />

T-cell depletion<br />

The role of T lymphocytes in bone marrow transplantation<br />

is very complex. They are in fact responsible for<br />

GvHD, a major contributing factor correlated with morbidity<br />

and mortality in allogeneic bone marrow transplantation.<br />

138,139 Indeed when T-cells are removed from<br />

the graft before transplantation, the incidence of GvHD<br />

decreases sharply. 71,140-143 On the other hand, the possible<br />

beneficial roles of T-lymphocytes include sustaining<br />

engraftment 144 and preventing relapses through the<br />

graft-versus-leukemia effect. 145,146 They are also crucial<br />

in immune-hematological reconstitution after transplantation<br />

because slow or deficient reconstitution may<br />

lead to a high incidence of viral infections or other<br />

infectious complications. Ex vivo manipulation of the T-<br />

lymphocyte content is easier and T-cell depleted allogeneic<br />

transplants may in the future be followed by<br />

infusion of non-alloreactive T-lymphocytes or of specifically<br />

engineered lymphocyte clones exerting an antiviral<br />

or anti-neoplastic effect.<br />

Standard T-cell depletion techniques<br />

Over the past 15 years, several techniques have been<br />

developed for depleting T-cells from human marrow<br />

allografts. 147 Table 3 summarizes the principles on which<br />

they are based and the degree of T-cell depletion each<br />

provides. Unfortunately, these methods may be timeconsuming,<br />

cumbersome and difficult to standardize in<br />

different transplantation centers. Results are therefore<br />

often variable and no general consensus has emerged on<br />

the use and benefit of bone marrow T-cell depletion.<br />

Very few data are available on the use of standard T-<br />

cell depletion methods in heterogeneous nucleated cell<br />

populations collected by leukaphereses from the peripheral<br />

blood of donors previously stimulated by hemopoietic<br />

growth factors.<br />

Kessinger et al. 148 first reported allogeneic transplantation<br />

utilizing T-cell depleted peripheral blood<br />

mononuclear cells and a sheep erythrocyte rosetting<br />

technique. Engraftment was rapid and grade II acute<br />

GvHD was observed.<br />

In a preliminary study after monocyte lysis with L-<br />

phenylalanine methyl ester, Suzue et al. 149 depleted T-<br />

lymphocytes from apheresis products harvested after<br />

stimulation of healthy donors with G-CSF using both E-<br />

rosettes with sheep red blood cells and panning with<br />

flasks coated with anti CD5/CD8 monoclonal antibodies.<br />

They reported an unsatisfactory depletion of T-cells<br />

(99.5%) and a stem cell recovery of only 7.5%.<br />

Aversa et al. 71 employed soybean lectin agglutination<br />

followed by 2 to 4 rounds of E-rosetting with sheep red<br />

blood cells on the leukapheresis product from donors<br />

stimulated with G-CSF. This approach achieves approximately<br />

3×log 10 T-lymphocyte depletion, as measured<br />

by cytofluorimetric assays. The main drawbacks are its<br />

complexity and lengthy laboratory times.<br />

Stem cell positive selection<br />

In principle, reducing T lymphocytes in the leukapheresis<br />

product by positively selecting CD34 + hemopoietic<br />

progenitors appears to be a valid technical alternative.<br />

Table 4 shows the basis of the main techniques<br />

for positive selection of hemopoietic progenitor cells.<br />

All use one monoclonal antibody which identifies an<br />

epitope on the human CD34 antigen. Separation is<br />

effected by collecting the antibody-sensitized cells onto<br />

a solid phase such as magnetic beads, plastic plates or<br />

columns of non magnetic particles, while non-target<br />

cells remain in suspension. Systems that utilize high<br />

speed flow cytometry to sort CD34 + cell populations<br />

have also been developed. 150<br />

The CD34 + stem cell selection systems adopted in<br />

clinical use are based on immunoadsorption and indirect<br />

immunomagnetic beads.<br />

Most clinical trials to date have been carried out with<br />

a ® Ceprate Stem Cell Concentrator (CellPro Inc., Bothell,<br />

WA, USA), which employs biotinylated 12.8 monoclonal<br />

antibody. The sensitized cells are applied to a column<br />

of avidin-coated polyacrylamide beads. Cells expressing<br />

Company Method Antibody Detachment<br />

CellPro Immunoadsorption 12.8 Mechanical<br />

Baxter Magnetic beads indirect 9C5 Chymopapain<br />

Baxter Magnetic beads indirect 9C5 PR34+ oligopeptide<br />

Dynal Magnetic beads direct BI3C5 Anti-Antibody<br />

(anti Fab of mouse MoAb)<br />

AIS Panning ICH3 Mechanical<br />

Immunotech Magnetic latex beads direct QBEND10 Not required<br />

Milteny Magnetic colloid indirect QBEND10 Not required<br />

Terry Fox Laboratory Magnetic Colloid Indirect 8G12 Not required<br />

System x FACS (high speed) Various Not required<br />

Table 4. Methods<br />

available for stem<br />

cell positive selection.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


60<br />

F. Bertolini et al.<br />

the CD34 antigen are retained and unlabelled cells<br />

washed through the column with gentle mechanical<br />

agitation. The CD34 + cells are then removed from the<br />

beads and collected.<br />

Using this system on the leukapheresis product, Link<br />

et al. 151 recovered a mean of 30% CD34 + cells, with a<br />

purity of 70%. Peripheral blood CD3 + cells were reduced<br />

by 3 logs. Other investigators have reported similar<br />

results. 152-155 This degree of T-cell depletion is known to<br />

prevent severe GvHD in severe combined immune deficiency<br />

(SCID) patients after matched or mismatched<br />

bone marrow transplantation. 140 In leukemia patients<br />

undergoing matched transplants it may not be enough<br />

to eliminate GvHD completely without the concomitant<br />

administration of immunosuppressive drugs. The threshold<br />

number of clonable T-lymphocytes in the inoculum<br />

should be below 1×10 5 /kg b.w., 156 which is difficult to<br />

achieve with one-step positive selection of hemopoietic<br />

stem cells. For a successful mismatched bone marrow<br />

transplant the T-lymphocyte threshold must be < 3-<br />

5×10 4 /kg b.w. 157 in the inoculum because of the greater<br />

likelihood and increased severity of GvHD in these<br />

patients. On the other hand, infusing a number well<br />

below the threshold value could expose the patient to<br />

a high risk of graft failure.<br />

Because T-cell depletion with the ® Ceprate system<br />

applied directly on the leukapheresis product does not<br />

reduce T-lymphocyte content from the graft by more<br />

than 3 logs, an additional T-cell depletion step is<br />

required.<br />

In 10 patients with different types of leukemias, Aversa<br />

et al. 157 employed an E-rosetting procedure before<br />

positively selecting hemopoietic progenitors with the<br />

®<br />

Ceprate system. This combined method yielded a T-cell<br />

depletion of 4.3 logs in the graft and a mean CD34 +<br />

recovery of 50-60%. 158<br />

In small-scale experiments, Fernandez et al. 159 applied<br />

the E-rosetting procedure after positive selection of<br />

CD34 + cells with this same ® Ceprate system to obtain a<br />

mean log 10 T-cell depletion of 4. Slaper-Cortenbach et<br />

al. 160 achieved a median recovery of 42.7% CD34 + cells<br />

and a T-lymphocyte reduction of 2-3 logs in 13 haploidentical<br />

transplants for SCID and in leukemia patients<br />

by employing autologous immunorosettes after positive<br />

selection of CD34 + cells.<br />

CD34 + progenitor cell immunomagnetic selection<br />

(Baxter, Irvine, CA, USA) achieved a 3 log T-cell depletion<br />

in preclinical experiments. 40,161 However, the main<br />

problem with this methodology was bead release from<br />

the target CD34 + cells. In fact release mediated by chymopapain<br />

may cause intractable cell clumping, particularly<br />

when a large number of cells are processed.<br />

Recently the PR34+TM stem cell releasing agent, an<br />

oligopeptide competing with the anti-CD34 monoclonal<br />

antibody for the release of the CD34 + cells from the<br />

magnetic beads, has also been proposed. 161 Preliminary<br />

results showed a reduction of non-target T cells by a<br />

factor of 2-3 logs with yields of CD34 + cells ranging<br />

from 31.1 to 85%. 162<br />

In conclusion, positive selection of CD34 + cells with<br />

the ® Ceprate system reduces the graft T-lymphocyte<br />

content under the threshold of risk for GvHD only when<br />

combined with standard T-depletion techniques such as<br />

E-rosetting with sheep red blood cells or autologous<br />

immunorosetting. Indirect immunomagnetic systems<br />

have to be evaluated more precisely for the use as a T-<br />

cell depletion system.<br />

Immunogenic activity of CD34 +<br />

hematopoietic cells<br />

Autologous transplantation of selected CD34 + cells<br />

induces rapid and complete hematologic reconstitution<br />

in myeloablated patients. In addition, isolation of CD34 +<br />

cells can be considered as an ex vivo means of purging<br />

neoplastic cells from the marrow or peripheral blood of<br />

patients with solid tumors or hematologic malignancies.<br />

163-165<br />

In the allogeneic setting, selection of CD34 + cells may<br />

be aimed at depleting donor T-cells and professional<br />

antigen-presenting cells (APC) such as monocytes, activated<br />

B-cells and dendritic cells, which are very potent<br />

stimulators of T-cell responses. Dendritic cells constitutively<br />

express the B7-2 (CD86) costimulatory molecule<br />

and upregulate B7-1 (CD80), B7-2 (CD86) and other<br />

molecules upon activation. 166-171 Furthermore, they were<br />

recently shown to derive from CD34 + marrow or peripheral<br />

blood cells, and can be rapidly generated in vitro in<br />

the presence of a specific combination of growth factors.<br />

172-177 Since it has been demonstrated that T-cell<br />

receptor (TCR): antigen interaction, in the absence of<br />

Authors<br />

TNC<br />

(x 108/kg)<br />

CD34+<br />

(x 106/kg)<br />

CD3+<br />

(x 106/kg)<br />

NK<br />

(x 106/kg)<br />

Dreger et al. (26) 13.52 8.16 404 N.R.<br />

Weaver et al. (191)* 20.53 9.6 450 N.R.<br />

Körbling et al. (16) 16.5 10.7 300 64.3<br />

Schmitz et al. (17) 8.6 13.1 340 94.0<br />

Bensinger et al. (18) 10.6 13.1 385 N.R.<br />

Majolino et al. (27) 9 6.84 250 27<br />

Rambaldi et al. (203) 8 6.9 279 N.R.<br />

Table 5. Median value of nucleated<br />

cells, CD34 + cells, CD3 + cells and<br />

natural killer cells infused in<br />

patients undergoing allogeneic<br />

peripheral blood stem cell transplantation.<br />

Legend. *Syngeneic transplants.<br />

N.R.=Not reported.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

61<br />

appropriate costimulation, may induce T-cell unresponsiveness<br />

or even apoptotic deletion, 171,178-181 the alloantigen<br />

presenting function of CD34 + marrow cells was<br />

recently investigated to evaluate whether transplantation<br />

of purified CD34 + cells could minimize the immune<br />

sensitization of an allogeneic receipient. 182 CD34 + marrow<br />

cells have been purified to >98% by a two-step<br />

procedure consisting of a first enrichment on an<br />

immunoaffinity chromatography column, followed by<br />

fluorescence activated cell sorting. Cytofluorimetric<br />

analysis of purified CD34 + marrow cells revealed the<br />

expression of HLA-DR and CD86 on >95% and 6% of<br />

the cells, respectively. Primary mixed leukocyte cultures<br />

demonstrated that irradiated CD34 + marrow cells<br />

induce brisk proliferation of allogeneic T-cells isolated<br />

from HLA-DR incompatible donors. On the basis of previous<br />

reports, 183,184 expression of CD18, the common<br />

chain of a family of leukointegrins, was investigated on<br />

CD34 + marrow cells and CD34 + /CD18 – cells were sorted<br />

to investigate whether this cell population was enriched<br />

in early hemopoietic precursors incapable of immunostimulating<br />

activity.<br />

On average, 25% of CD34 + marrow cells were CD18 –<br />

by direct immunofluorescent analysis. Purified CD34 + ,<br />

CD34 + /CD18 + and CD34 + /CD18 – marrow subsets were<br />

tested in bulk MLC with allogeneic T-cells, and it was<br />

observed that CD34 + , CD34 + /CD18 + and unseparated<br />

marrow mononuclear cells have a similar capacity to<br />

stimulate a T-cell response. Conversely, CD34 + /CD18 –<br />

cells do not elicit any T-lymphocyte proliferation. Moreover,<br />

limiting dilution assay (LDA) experiments showed,<br />

on a per cell basis, that CD34 + /CD18 – and CD34 + /CD86 –<br />

marrow cells have a very poor ability to induce a T-cell<br />

response, as opposed to CD34 + /CD18 + and CD34 + /CD86 +<br />

marrow cells. Since most marrow LTC-IC were included<br />

in the CD34 + /CD18 – cell fraction, it was concluded<br />

that CD34 + /CD18 – , or CD34 + /CD86 – marrow cells, may<br />

represent a useful source of progenitor cells for allogeneic<br />

transplantation because of their high stem cell<br />

activity combined with reduced immunogenicity. Data<br />

on normal human G-CSF mobilized CD34 + peripheral<br />

blood (PB) cells show that on average 30% of the cells<br />

express CD18 and only 3% express CD86, while functional<br />

in vitro results are consistent with what was previously<br />

observed in marrow. Thus, CD34 + PB cells can<br />

potently stimulate T cells, likely through the B7:CD28<br />

pathway, and CD34 + /CD18 – PB cells still have very weak<br />

immunostimulating activity.<br />

In a preliminary study sibling baboons were fully<br />

engrafted with allogeneic CD34 + marrow cells without<br />

GVHD, after receiving total body irradiation as conditioning<br />

regimen and standard GVHD prophylaxis. 184<br />

Development of mobilization regimens capable of<br />

increasing the number of peripheral blood hemopoietic<br />

stem cells in normal healthy donors allowed sufficient<br />

amounts of CD34 + PB cells to be harvested for allogeneic<br />

transplantation in humans. In fact, transplantation of<br />

enriched populations of G-CSF mobilized CD34 + cells<br />

resulted in rapid engraftment, similar to that observed<br />

in allogeneic PBSC transplants. 185-190 Purification of<br />

CD34 + cells on the Ceprate column obtains on average<br />

a 3 log depletion of CD3 + T cells in the graft; however,<br />

several studies reported contrasting rates of acute<br />

GVHD. In particular, > 80% of the patients transplanted<br />

with CD34 + PB cells in Seattle experienced aGVHD<br />

grade II-III after receiving a median number of 0.7×10 6<br />

T-cells/kg in the graft and GVHD prophylaxis with<br />

cyclosporin-A (CsA)± methotrexate (MTX). 187 Another<br />

study reported 2 cases out of 5 who died from aGVHD. 188<br />

By contrast, other groups reported a very low incidence<br />

of GVHD. 189,190 One of the reasons for these disparities<br />

may be that small numbers of patients, often with different<br />

malignancies and clinical characteristics, are<br />

included in these studies. Nevertheless, two hypotheses<br />

could be addressed: the first one suggests that infusion<br />

of as little as 0.5-1×10 6 CD3 + T cells/kg could be potentially<br />

capable of initiating GVHD, which would be prevented<br />

by further steps in T-cell depletion. 158 The second<br />

hypothesis, still to be tested, is whether APC in marrow<br />

or peripheral blood could play a role in the development<br />

of GVHD by presenting allogeneic peptides to donor T-<br />

cells.<br />

In this regard, a subset of CD34 + cells in the graft may<br />

induce the activation and proliferation of a limited number<br />

of T cells, such as those still present after CD34<br />

purification.<br />

Peripheral blood stem cells:<br />

immunological aspects<br />

Very few data are available on the effects of hemopoietic<br />

growth factors used to mobilize PBSC on peripheral<br />

blood lymphocytes.<br />

Weaver et al. 191 analyzed the influence of G-CSF on<br />

peripheral blood lymphocytes from 13 individuals (11<br />

autografts and 2 normal donors). In all cases they<br />

observed a slight increase in CD3, CD4, CD8, CD19 and<br />

CD20-positive lymphocytes, with a return to pretreatment<br />

values by days 4 and 5 of G-CSF administration.<br />

The change in the CD4/CD8 ratio was not statistically<br />

significant.<br />

The expression of CD2, CD3, CD4, CD7, CD8, CD20,<br />

CD25, CD57 and HLA-DR antigens was evaluated during<br />

administration of G-CSF (12 ug/kg/day for 5-7 days)<br />

to healthy donors. No significant variations were<br />

observed in the different lymphocyte subsets, in the<br />

CD4/CD8 ratio or in the expression of CD25 and HLA-DR<br />

antigens (unpublished data). G-CSF administration does<br />

not cause direct activation of T lymphocytes in vivo. This<br />

might be expected because lymphocytes do not possess<br />

the G-CSF receptor. 192 However, it is possible that activation<br />

could be caused by cytokine release from cells<br />

stimulated by G-CSF.<br />

Other important aspects of the PBSC allograft include<br />

the lymphocyte content, particularly T lymphocytes and<br />

natural killer cells, in the apheresis product. Table 5<br />

reports data on the total number of CD3 + lymphocytes<br />

derived from peripheral blood stem cells that were<br />

infused for allogeneic transplants. The number of<br />

haematologica vol. 85(suppl. to n. 12):December 2000


62<br />

F. Bertolini et al.<br />

infused T lymphocytes was always 1.5-2 logs greater<br />

than that derived from bone marrow. 193<br />

The exact relationship between the T-lymphocyte<br />

content in the graft and the development and severity<br />

of GvHD remains unclear. A linear relationship between<br />

the number of T lymphocytes infused and the development<br />

of GvHD has long been hypothesized, 139,194,195 but<br />

this correlation has not always been confirmed. 196,197<br />

Findings in allogeneic peripheral blood stem cell transplantation<br />

seem to suggest that the number of T lymphocytes<br />

is less important than donor-cell specificity in<br />

triggering GvHD. 16,17<br />

Another aspect of peripheral blood stem cell transplantation<br />

concerns the number of natural killer cells<br />

(NK) infused (Table 5) since they are important effector<br />

cells in graft-versus-leukemia activity. 198 The number of<br />

infused NK cells is about 20 times greater in an allogeneic<br />

peripheral blood stem cell transplant than in a<br />

bone marrow graft. 16,17 The question of whether this will<br />

translate into more potent GvL activity in patients allografted<br />

with peripheral blood stem cells compared to<br />

unmanipulated bone marrow cannot be answered at<br />

this time, but needs further study. However, preliminary<br />

data from a murine model demonstrated strong GvL<br />

activity for allogeneic NK cells without the induction of<br />

GvHD. 199<br />

An important technical point is the effect of freezing<br />

and thawing of the graft or of keeping the apheresis<br />

product at 4°C overnight on T-lymphocytes inactivation.<br />

Van Bekkum 200 described selective elimination of<br />

immunologically competent cells from bone marrow<br />

after storage at 4°C. Eckardt et al. 201 also noted that cryopreservation<br />

of allogeneic marrow may reduce the risk<br />

of acute GvHD. Selective depletion or induction of anergy<br />

in GvHD-inducing cells was hypothesized. 201 Storage<br />

of the apheresis product at 4°C overnight does not modify<br />

the surface expression of the CD3, CD4, CD8, or the<br />

CD57 antigens in a significant manner (Tabilio, 1996,<br />

unpublished results); however, how cryopreservation<br />

affects the alloreactivity of peripheral blood stem cells<br />

needs to be investigated further.<br />

Conclusions<br />

Several studies have now shown that hematopoietic<br />

stem cells collected from PB after the administration of<br />

G-CSF, or from CB upon delivery, are capable of supporting<br />

rapid and complete reconstitution of BM function<br />

in allogeneic recipients. 16-20,204,205 The faster recovery<br />

of hematopoiesis as compared to BM-derived allografts,<br />

together with a lower incidence of aGVHD than<br />

expected with BM transplantation, raises the question<br />

of whether PBSC collections may differ from conventional<br />

BM harvests with respect to the number of stem<br />

cells and their functional characteristics, lymphoid cell<br />

composition and T-cell reactivity. Moreover, recent clinical<br />

studies on transplantation of CB-derived cells from<br />

unrelated HLA-mismatch donors support the notion that<br />

sources of hematopoietic stem cells other than BM represent<br />

a feasible alternative to conventional transplantation.<br />

In this paper, the phenotypic, functional and<br />

kinetic features of circulating and CB hematopoietic<br />

cells were reviewed. We also emphasized the technical<br />

aspects of CB collection and processing, as well as the<br />

protocols for PBSC mobilization and collection from normal<br />

donors. Notably, novel data on the immunogenic<br />

and kinetic profile of BM and PB CD34 + cells may shed<br />

new light on stem cell biology and may help clinical<br />

investigators to design future trials on transplantation<br />

of purified hematopoietic progenitors.<br />

It should be remembered that despite growing interest<br />

these procedures must still be considered as<br />

advanced clinical research and should be included in<br />

formal clinical trials aimed at demonstrating their definitive<br />

role in stem cell transplantation. In this regard, a<br />

large European randomized study is currently comparing<br />

PBSC and BM allografts. However, the possibility of<br />

collecting a large quantity of hematopoietic progenitor<br />

stem cells from PB, perhaps with reduced allo-reactivity,<br />

offers an exciting perspective for widening the number<br />

of potential stem cell donors and greater leeway for<br />

graft manipulation than is possible with BM.<br />

Acknowledgments<br />

Preparation of this manuscript was supported by<br />

grants from Dompé Biotec SpA and Amgen Italia SpA,<br />

Milan, Italy.<br />

This review article was prepared by a group of experts<br />

designated by <strong>Haematologica</strong> and by representatives of<br />

two pharmaceutical companies, Amgen Italia SpA and<br />

Dompé Biotec SpA, both from Milan, Italy. This co-operation<br />

between a medical journal and pharmaceutical<br />

companies is based on the common aim of achieving an<br />

optimal use of new therapeutic procedures in medical<br />

practice. In agreement with the Journal’s Conflict of<br />

Interest Policy, the reader is given the following information.<br />

The preparation of this manuscript was supported<br />

by educational grants from the two companies.<br />

Dompé Biotec SpA sells G-CSF and rHuEpo in Italy, and<br />

Amgen Italia SpA has a stake in Dompé Biotec SpA. This<br />

paper has undergone a regular peer-review process and<br />

has been evaluated by two outside referees.<br />

References<br />

1. Thomas D. Bone marrow transplantation: past, present<br />

and future. <strong>Haematologica</strong> 1991; 76:353-6.<br />

2. Majolino I, Aversa F, Bacigalupo A, Bandini G, Arcese<br />

W, Reali G. Allogeneic transplants of rhG-CSF-mobilized<br />

peripheral blood stem cells (PBSC) from normal<br />

donors. <strong>Haematologica</strong> 1995; 80:40-3.<br />

3. Carlo-Stella C, Cazzola M, De Fabritiis P, et al. CD34-<br />

positive cells: biology and clinical applications.<br />

<strong>Haematologica</strong> 1995; 80:367-87.<br />

4. Aglietta M, De Vincentiis A, Lanata L, et al. Peripheral<br />

blood stem cells in acute myeloid leukemia: biology<br />

and clinical applications. <strong>Haematologica</strong> 1996;<br />

81:77-92.<br />

5. Caligaris-Cappio F, Cavo M, De Vincentiis A, et al.<br />

Peripheral blood stem cell transplantation for the<br />

treatment of multiple myeloma: biological and clini-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

63<br />

cal implications. <strong>Haematologica</strong> 1996; 81:356-75.<br />

6. Buckner CD, Clift RA, Sanders JE, et al. Marrow harvesting<br />

from normal donors. Blood 1984; 64:630-4.<br />

7. Stroncek DF, Holland PV, Bartch G, et al. Experiences<br />

of the first 493 unrelated marrow donors in the<br />

national marrow donor program. Blood 1993;<br />

81:1940-6.<br />

8. Gratwohl A, Schmitz N. Introduction to First International<br />

Symposium on allogeneic peripheral blood precursor<br />

cell transplants. Bone Marrow Transplant<br />

1996; 17(suppl 2):1-3.<br />

9. To LB, Roberts MM, Haylock DN, et al. Comparison<br />

of haematological recovery times and supportive care<br />

requirements of autologous recovery phase peripheral<br />

blood stem cell transplants, autologous bone marrow<br />

transplants and allogeneic transplants. Bone<br />

Marrow Transplant 1992; 9:277-84.<br />

10. Indovina A, Majolino I, Buscemi F, et al. Engraftment<br />

kinetics and long term stability of hematopoiesis following<br />

autografting of peripheral blood progenitor<br />

cells. <strong>Haematologica</strong> 1995; 80:115-22.<br />

11. To LB, Sheppard KM, Haylock DN, et al. Single high<br />

doses of cyclophosphamide enable the collection of<br />

high numbers of haematopoietic stem cells from the<br />

peripheral blood. Exp Hematol 1990; 18:442-7.<br />

12. Indovina A, Majolino I, Scimè R, et al. High dose<br />

cyclophosphamide: stem cell mobilizing capacity in<br />

21 patients. Leuk Lymphoma 1994; 14:71-7.<br />

13. Sheridan WP, Begley CG, Juttner CA, et al. Effect of<br />

peripheral blood progenitor cells mobilized by filgrastim<br />

(rhG-CSF) on platelet recovery after high-dose<br />

chemotherapy. Lancet 1992; 1:640-4.<br />

14. Gianni AM, Bregni M, Siena S, et al. Granulocytemacrophage<br />

colony-stimulating factor to harvest circulating<br />

hemopoietic stem cells for autotransplantation.<br />

Lancet 1989; 2:580-5.<br />

15. Auquier P, Macquart-Moulin G, Moatti JP, et al. Comparison<br />

of anxiety, pain and discomfort in two procedures<br />

of hematopoietic stem cell collection: leukacytapheresis<br />

and bone marrow harvest. Bone Marrow<br />

Transplant 1995; 16:541-7.<br />

16. Körbling M, Przepiorka D, Huh YO, et al. Allogeneic<br />

blood stem cell transplantation for refractory<br />

leukemia and lymphoma: potential advantage of<br />

blood over marrow allografts. Blood 1995; 85:1659-<br />

65.<br />

17. Schmitz N, Dreger P, Suttorp M, et al. Primary transplantation<br />

of allogeneic peripheral blood progenitor<br />

cells mobilized by filgrastim (granulocyte colony-stimulating<br />

factor). Blood 1995; 85:1666-72.<br />

18. Bensinger WI, Weaver CH, Appelbaum FR, et al.<br />

Transplantation of allogeneic peripheral blood stem<br />

cells mobilized by recombinant human granulocyte<br />

colony stimulating factor. Blood 1995; 85:1655-8.<br />

19. Bacigalupo A, Majolino I, Van Lint MT, et al. Transplantation<br />

of rhG-CSF mobilized allogeneic peripheral<br />

blood cells from HLA identical sibling donors<br />

[abstract]. Bone Marrow Transplant 1995; 15(Suppl.<br />

2):5.<br />

20. Majolino I, Saglio G, Scimé R, et al. High incidence of<br />

chronic GVHD after primary allogeneic peripheral<br />

blood stem cell transplantation in patients with hematologic<br />

malignancies. Bone Marrow Transplant 1996;<br />

17:555-60.<br />

21. Krause DS, Fackler MJ, Civin CI, May WS. CD34:<br />

structure, biology, and clinical utility. Blood 1996;<br />

87:1-13.<br />

22. Papayannopoulou T, Nakamoto B. Peripheralization<br />

of hemopoietic progenitors in primates treated with<br />

anti-VLA4 integrin. Proc Natl Acad Sci USA 1993;<br />

90:9374-8.<br />

23. Richman CM, Weiner RS, Yankee RA. Increase in circulating<br />

stem cells following chemotherapy in man.<br />

Blood 1976; 47:1031-4.<br />

24. Bungart B, Loeffler M, Goris H, Diehl V, Nijhof W.<br />

Differential effects of recombinant human colony<br />

stimulating factor (rh G-CSF) on stem cells in marrow,<br />

spleen and peripheral blood in mice. Br J Haematol<br />

1990; 76:174-9.<br />

25. Sheridan WP, Begley CG, To LB, et al. Comparison of<br />

autologous filgrastim (rhG-CSF)-mobilized peripheral<br />

blood progenitor cells to restore hemopoiesis after<br />

high-dose chemotherapy for lymphoid malignancies.<br />

Bone Marrow Transplant 1994; 14:105-11.<br />

26. Dreger P, Haferlach T, Eckstein V, et al. rhG-CSFmobilized<br />

peripheral blood progenitor cells for allogeneic<br />

transplantation: safety, kinetics of mobilization,<br />

and composition of the graft. Br J Haematol<br />

1994; 87:609-13.<br />

27. Majolino I, Buscemi F, Scimé R, et al. Treatment of<br />

normal donors with rhG-CSF 16 µg/kg for mobilization<br />

of peripheral blood stem cells and their apheretic<br />

collection for allogeneic transplantation. <strong>Haematologica</strong><br />

1995; 80:219-26.<br />

28. Bensinger WI, Buckner CD, Rowley S, Storb R, Appelbaum<br />

FR. Treatment of normal donors with recombinant<br />

growth factors for transplantation of allogeneic<br />

blood stem cells. Bone Marrow Transplant<br />

1996; 1(Suppl. 2):19-21.<br />

29. Anderlini P, Miller P, Sundberg J, et al. “High-dose” G-<br />

CSF (filgrastim) for stem cell mobilization in normal<br />

donors: a prospective study. ASCO Proc 1996;<br />

15:269.<br />

30. Bensinger WI, Prince TH, Dale DC, et al. The effects<br />

of daily recombinant human granulocyte colony stimulating<br />

factor administration on normal granulocyte<br />

donors undergoing leukapheresis. Blood 1993; 81:<br />

1883-8.<br />

31. Weaver CH, Buckner CD, Longin K, et al. Syngeneic<br />

transplantation with peripheral blood mononuclear<br />

cells collected after the administration of recombinant<br />

human granulocyte colony-stimulating factor.<br />

Blood 1993; 82:1981-4.<br />

32. Majolino I, Scimé R, Vasta S, et al. Mobilization and<br />

collection of PBSC in healthy donors. Comparison<br />

between two schemes or rhG-CSF administration. Eur<br />

J Haematol 1996; 57:214-21.<br />

33. Dührsen U, Villeval JL, Boyd J, Kannourakis G,<br />

Morstyn G, Metcalf D. Effects of recombinant human<br />

granulocyte colony-stimulating factor on hematopoietic<br />

progenitors cells in cancer patients. Blood 1988;<br />

72:2074-81.<br />

34. Russell JA, Luider J, Weaver M, et al. Collection of<br />

progenitor cells for allogeneic transplantation from<br />

peripheral blood of normal donors. Bone Marrow<br />

Transplant 1995; 15:111-5.<br />

35. Waller CF, Bertz H, Engelhardt M, et al. Mobilization<br />

of peripheral blood progenitor cells (PBPC) for allogeneic<br />

peripheral blood progenitor cell transplantation<br />

(allo-PBPCT): efficacy and toxicity of a high dose<br />

rhG-CSF regimen. Bone Marrow Transplant 1996; 17<br />

(Suppl. 2):71.<br />

36. Matsunaga T, Sakamaki S, Kohgo Y, Ohi S, Hirayama<br />

Y, Niitsu Y. Recombinant human granulocyte colonystimulating<br />

factor can mobilize sufficient amounts of<br />

peripheral blood stem cells in healthy volunteers for<br />

allogeneic transplantation. Bone Marrow Transplant<br />

1993; 11:103-8.<br />

37. Schwinger W, Mache C, Urban C, Beaufort F, Toglhofer<br />

W. Single dose of filgrastim (rhG-CSF) increases<br />

the number of hematopoietic progenitors in the<br />

peripheral blood of adult volunteers. Bone Marrow<br />

Transplant 1993; 11:489-92.<br />

38. Bishop MR, Tarantolo SR, Schmit-Pokorny K, et al.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


64<br />

F. Bertolini et al.<br />

Mobilization of blood stem cells from HLA-identical<br />

related donors with low-dose granulocyte colonystimulating<br />

factor for allogeneic transplantation.<br />

Blood 1995; 86(Suppl. 1):463.<br />

39. Majolino I, Cavallaro AM, Bacigalupo A, et al. Mobilization<br />

and collection of PBSC in healthy donors: a<br />

retrospective analysis of the Italian Bone Marrow<br />

Transplantation Group (GITMO). <strong>Haematologica</strong><br />

1997; 81:47-52.<br />

40. Lane TA, Law P, Maruyama M, et al. Harvesting and<br />

enrichment of hematopoietic progenitor cells mobilized<br />

into the peripheral blood of normal donors by<br />

granulocyte-macrophage colony-stimulating factor<br />

(GM-CSF) or G-CSF: potential role in allogeneic marrow<br />

transplantation. Blood 1995; 85:275-82.<br />

41. Bender JG, To LB, Williams S, Schwartzberg LS. Defining<br />

a therapeutic dose of peripheral blood stem cells.<br />

J Hematother 1992; 1:329-41.<br />

42. Goldberg SL, Mangan KF, Klumpp TR, et al. Complications<br />

of peripheral blood stem cell harvesting:<br />

review of 554 PBSC leukaphereses. J Hematother<br />

1995; 4:85-90.<br />

43. Fossa SD, Poulsen JP, Anders A. Alkaline phosphatase<br />

and lactate dehydrogenase changes during leukocytosis<br />

induced by G-CSF in testicular cancer. Lancet<br />

1992; 340:1544.<br />

44. Sakamaki S, Matsunaga T, Hirayama Y, Kuga T, Niitsu<br />

Y. <strong>Haematologica</strong>l study of healthy volunteers 5<br />

years after G-CSF. Lancet 1995; 346:1432-3.<br />

45. Hasenclever D, Sextro M. Safety of alloPBPCT donors:<br />

biometrical considerations on monitoring long term<br />

risks. Bone Marrow Transplant 1996; 17(suppl 2):28-<br />

30.<br />

46. Ohara A, Kojima S, Tsuchida M, et al. Evolution of<br />

acquired severe aplastic anemia to MDS/leukemia in<br />

childhood: a retrospective strudy on 170 severe aplastic<br />

anemia child patients. Blood 1995; 86(suppl 1):<br />

1333.<br />

47. Kalra R, Dale D, Freedman M, et al. Monosomy 7 and<br />

activating RAS mutations accompany malignant<br />

transformation in patients with congenital neutropenia.<br />

Blood 1995; 86:4579-86.<br />

48. Smith OP, Reeves BR, Kempski HM, Evans JP. Kostmann’s<br />

disease, recombinant HuG-CSF, monosomy 7<br />

and MDS/AML. Br J Haematol 1995; 91:150-3.<br />

49. Ringdén O, Potter MN, Oakhill A, et al. Transplantation<br />

of peripheral blood progenitor cells from unrelated<br />

donors. Bone Marrow Transplant 1996; 17(Suppl<br />

2):62-4.<br />

50. Tjonnfjord GE, Steen R, Evensen SA, et al. Characterization<br />

of CD 34+ peripheral blood cells from healthy<br />

adults mibilized by recombinant human granulocyte<br />

colony-stimulating factor. Blood 1994; 84:2795-801.<br />

51. Roberts AW, Metcalf D. Noncycling state of peripheral<br />

blood progenitor cells mobilized by granulocyte<br />

colony-stimulating factor and other cytokines. Blood<br />

1995; 86:1600-5.<br />

52. To LB, Haylock DN, Dowse T, et al. A comparative<br />

study of the phenotype and proliferative capacity of<br />

peripheral blood (PB) CD34+ cells mobilized by four<br />

different protocols and those of steady-state PB and<br />

bone marrow CD34+ cells. Blood 1994; 84:2930-9.<br />

53. Bregni M, Magni M, Siena S, et al. Human peripheral<br />

blood hematopoietic progenitors are optimal targets<br />

of retroviral-mediated gene transfer. Blood 1992;<br />

80:1418-22.<br />

54. Peters SO, Kittler ELW, Ramshaw HS, et al. Ex-vivo<br />

expansion of murine marrow cells with interleukin-3<br />

(IL-3), IL-6, IL-11 and stem cell factor leads to<br />

impaired engraftment in irradiated host. Blood 1996;<br />

87:30-7.<br />

55. Lemoli RM, Tafuri A, Fortuna A, et al. Cycling status<br />

of CD 34+ cells mobilized into peripheral blood of<br />

healthy donors by recombinant human granulocyte<br />

colony-stimulating factor. Blood 1997; in press.<br />

56. Pettengel R, Luft T, Henschler R, et al. Direct comparison<br />

by limiting dilution analysis of long-term culture-initiating<br />

cells in human bone marrow, umbelical<br />

cord blood, and blood stem cells. Blood 1994; 84:<br />

3653-9.<br />

57. Murray L, Chen B, Galy A, et al. Enrichment of human<br />

hematopoietic stem cell activity in the CD34+ Thy-1+<br />

Lin– subpopulation from mobilized peripheral blood.<br />

Blood 1995; 85:368-78.<br />

58. Sutherland HJ, Eaves CJ, Lansdorp PM, Phillips GL,<br />

Hogge DH. Kinetics of committed and primitive blood<br />

progenitor mobilization after chemotherapy and<br />

growth factor treatment and their use in autotransplants.<br />

Blood 1994; 83:3808-14.<br />

59. Dreger P, Klöss M, Petersen B, et al. Autologous progenitor<br />

cell transplantation: prior exposure to stem celltoxic<br />

drugs determines yield and engraftment of<br />

peripheral blood progenitor cell but not of bone marrow<br />

grafts. Blood 1995; 86:3970-8.<br />

60. Ponchio L, Conneally E, Eaves CJ. Quantitation of the<br />

quiescent fraction of longterm culture initianting cells<br />

(LTC-IC) in normal human blood and marrow and<br />

the kinetics of their growth factor-stimulated entry<br />

into S-phase in vitro. Blood 1995; 86:3314-21.<br />

61. Donahue RE, Kirby MR, Metzger ME, et al. Peripheral<br />

blood CD 34+ cells differ from bone marrow<br />

CD34+ cells in Thy-1 expression and cell cycle status<br />

in nonhuman primates mobilized or not mobilized<br />

with granulocyte colony-stimulating factor. Blood<br />

1996; 87:1644-53.<br />

62. Becker PS, Li Z, Quesenberry PJ. Cytokine regulation<br />

of cell adhesion receptor expression in hematopoietic<br />

cells. Blood 1994; 84:280a (Suppl. 1).<br />

63. Fleming WH, Alpern EJ, Uchida N, Ikuta K, Weissman<br />

IL. Steel factor influences the distribution and activity<br />

of murine hematopoietic stem cells in vivo. Proc<br />

Natl Acad Sci USA 1993; 90:3760-4.<br />

64. Kovach NL, Lin N, Yednock T, Harlan JM, Broudy VC.<br />

Stem cell factor modulates avidity of a4b1 and a5b1<br />

integrins expressed on hematopoietic cell lines. Blood<br />

1995; 85:159-67.<br />

65. Möhle R, Murea S, Kirsch M, Haas R. Differential<br />

expression of L-selectin, VLA-4, and LFA-1 on CD34+<br />

progenitor cells from bone marrow and peripheral<br />

blood durin G-CSF-enhanced recovery. Ex Hematol<br />

1995; 23:1535-42.<br />

66. Gabutti V, Foà R, Mussa F, Aglietta M. Behavior of<br />

human hematopoietic stem cells in cord and neonatal<br />

blood. <strong>Haematologica</strong> 1975; 60:492.<br />

67. Broxmeyer HE, Douglas GW, Hancgoc G, et al.<br />

Human umbilical cord as a potential source of transplantable<br />

hematopoietic stem/progenitor cells. Proc<br />

Natl Acad Sci USA, 1989; 86:3828.<br />

68. Gluckman E, Broxmeyer HE, Auerbach DA, et al.<br />

Hematopoietic reconstitution in a patient with Fanconi’s<br />

anemia by means of umbilical cord blood from<br />

an HLA-identical sibling. N Engl J Med 1989; 321:<br />

1174-8.<br />

69. Bertolini F, Lazzari L, Lauri L, et al. A comparative<br />

study of different procedures for the collection and<br />

banking of umbilical cord blood. J Hematother 1995;<br />

4:29-36.<br />

70. Bertolini F, Soligo D, Lazzari L, Corsini C, Servida F,<br />

Sirchia G. The effect of interkleukin 12 in ex-vivo<br />

expansion of human hematopoietic progenitors. Br J<br />

Haematol 1995; 90:935-8.<br />

71. Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment<br />

of T-cell-depleted haploidentical “three loci”<br />

incompatible transplants in leukemia patients by<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

65<br />

addition of recombinant human G-CSF-mobilized<br />

peripheral blood progenitor cells to bone marrow<br />

inocolum. Blood 1994; 84:3948-55.<br />

72. Wagner JE, Kernan NA, Steinbuch M, Broxmeyer HE,<br />

Gluckman E. Allogeneic sibling umbilical-cord-blood<br />

transplantation in children with malignant and nonmalignant<br />

disease. Lancet 1995; 346:214-9.<br />

73. Lansdorp PM, Dragowska W, Mayani H. Ontogenyrelated<br />

changes in proliferative potential of human<br />

hematopoietic cells. J Exp Med 1993; 178:787-91.<br />

74. Hows JM, Bradley BA, Marsh JCW, et al. Growth of<br />

human umbilical-cord blood in longterm haemopoietic<br />

cultures. Lancet 1992; 340:73-6.<br />

75. Payne TA, Traycoff CM, Laver J, Xu F, Srour EF,<br />

Abboud MR. Phenotypic analysis of early hematopoietic<br />

progenitors in cord blood and determination of<br />

their correlation with clonogenic progenitors: relevance<br />

to cord blood stem cell transplantation. Bone<br />

Marrow Transplant 1995; 15:187-92.<br />

76. Traycoff CM, Abboud MR, Laver J, et al. Evaluation of<br />

the in vitro behavior of phenotypically defined populations<br />

of umbilical cord blood hematopoietic progenitor<br />

cells. Exp Hematol 1994; 22:215-22.<br />

77. Gabutti V, Timeus F, Ramenghi U, et al. Expansion of<br />

cord blood progenitors and use for hemopoietic<br />

reconstitution. Stem Cells 1993; 11(Suppl. 2):105-<br />

12.<br />

78. Moritz T, Keller DC, Williams DA. Human cord blood<br />

cells as targets for gene transfer. Potential use in genetic<br />

therapies of SCID. J Exp Med 1993; 178:529-36.<br />

79. Kohn DB, Weinberg KI, Nolta JA, et al. Engraftment<br />

of gene-modified umbilical cord blood cells in<br />

neonates with adenosine deaminase deficiency.<br />

Nature Med 1995; 1:1017-23.<br />

80. Bertolini F, De Monte L, Corsini C, et al. Retrovirusmediated<br />

transfer of the multidrug resistance gene<br />

into human haematopoietic progenitors. Br J Haematol<br />

1994; 88:318-24.<br />

81. Bertolini F, Battaglia M, Corsini C, et al. Engineered<br />

stromal layers and continuous flow culture enhance<br />

multidrug resistance gene transfer in hematopoietic<br />

progenitors. Cancer Res 1996; 56:2566-72.<br />

82. Traycoff CM, Abboud MR, Laver J, Clapp DW, Srour<br />

EF. Rapid exit from G0/G1 phases of cell cycle in<br />

response to stem cell factor confers on umbilical cord<br />

blood CD34+ cells an enhanced ex vivo expansion<br />

potential. Exp Hematol 1994; 22:1264-72.<br />

83. Lambertenghi-Deliliers G, Bertolini F, Della Volpe A,<br />

et al. Unrelated mismatched cord blood transplantation<br />

in an adult patient with secondary AML. Bone<br />

Marrow Transplant 1997; in press .<br />

84. Traycoff CM, Cornetta K, Yoder MC, et al. Ex vivo<br />

expansion of murine hematopoietic progenitor cells<br />

generates classes of expanded cells possessing different<br />

levels of bone marrow repopulating potential. Exp<br />

Hematol 1996; 24:229-306.<br />

85. Rubinstein P, Rosenfield RE, Adamson JW, Stevens<br />

CE. Stored placental blood for unrelated bone marrow<br />

reconstitution. Blood 1993; 7:1679-90.<br />

86. Kögler G, Somville Th, Adams O, et al. Critical standards<br />

for stem cell preparations from unrelated cord<br />

blood within Eurocord. Exp Hematol 1995; 22:898.<br />

87. Gluckman E, Devergie A, Thierry D, et al. Clinical<br />

application of stem cells transfusion from cord blood<br />

and rationale for cord blood banking. Bone Marrow<br />

Transplant 1992; 9 Suppl 1:114-7.<br />

88. Turner CW, Luzins J, Hutcheson CA. A modified harvest<br />

technique for cord blood hematopoietic stem<br />

cells. Bone Marrow Transplant 1992; 10:89.<br />

89. Abecasis MM, Machado AM, Boavida G, et al. Haploidentical<br />

cord blood transplant contaminated with<br />

maternal T cells in a patient with advanced leukemia.<br />

Bone Marrow Transplant 1996; 17:891-5.<br />

90. Yao AC, Moinian M, Lind J. Distribution of blood<br />

between infant and placenta after birth. Lancet 1969;<br />

2:871.<br />

91. Yao AC, Lind J. Effect of gravity on placental transfusion.<br />

Lancet 1969; 2:508.<br />

92. Nelle M, Zilow EP, Kraus M, Bastert G, Linderkamp O.<br />

The effect of the Leboyer delivery on blood viscosity<br />

and other hemorheologic parameters in term<br />

neonates. Am J Ob Gyn 1993; 169:189-93.<br />

93. Sinclair JC, Bracken MB. eds. Effective care of newborn<br />

infant. Oxford: Oxford Medical Publ.; 1992.<br />

94. Roberton NRC, ed. Textbook of neonatology. London:<br />

Churchill Livingstone; 1992.<br />

95. Pritchard JA, McDonald PC, Grant NF, eds. William’s<br />

Obstetrics. Norwalk: Appelton-Century-Crofts; 1985.<br />

96. Taeush HW, Ballard RA, Avery ME. eds. Schaffer and<br />

Avery’s diseases of the newborn. Philadelphia :WB<br />

Saunders; 1991.<br />

97. Polin RA, Fox WW, eds. Fetal and neonatal physiology.<br />

Philadelphia: WB Saunders; 1992.<br />

98. Bertolini F, Battaglia M, De Iulio C, et al. Placental<br />

blood collection: Effects on newborns. Blood 1995;<br />

86:4699.<br />

99. Harris DT, Schumacher MJ, Rychlik S, et al. Collection,<br />

separation and cryopreservation of umbilical<br />

cord blood for use in transplantation. Bone Marrow<br />

Transplant 1994; 13:135-43.<br />

100. Almici C, Carlo-Stella C, Mangoni L, et al. Density<br />

separation of umbilical cord blood and recovery of<br />

hemopoietic progenitor cells: Implications for cord<br />

blood banking. Stem Cells 1995; 13:533-40.<br />

101. Rubinstein P, Dobrila L, Rosenfield RE, et al. Processing<br />

and cryopreservation of placental/umbilical<br />

cord blood for unrelated bone marrow reconstitution.<br />

Proc Natl Acad Sci USA 1995; 92:10119-22.<br />

102. Bertolini F, Battaglia M, Zibera C, et al. A new method<br />

for placental/cord processing in the collection bag.<br />

Analysis of factors involved in red blood cell removal.<br />

Bone Marrow Transplant 1996; in press.<br />

103. Pahwa RN, Fleischer A, Than S, Good RA. Successful<br />

hematopoietic reconstitution with transplantation of<br />

erythrocyte-depleted allogeneic human umbilical cord<br />

blood cells in a child with leukemia. Proc Natl Acad<br />

Sci USA 1994; 91:4485-8.<br />

104. Di Giusto DL, Lee R, Moon J, et al. Hematopoietic<br />

potential of cryopreserved and ex vivo manipulated<br />

umbilical cord blood progenitor cells evaluated in vitro<br />

and in vivo. Blood 1996; 87:1261-71.<br />

105. Petzer AL, Hogge DE, Lansdorp PM, Reid DS, Eaves<br />

CJ. Self-renewal of primitive hemetopoietic cells (longterm<br />

culture-initiating cells) in vitro and their expansion<br />

in defined medium. Proc Natl Acad Sci USA<br />

1996; 93:1470-4.<br />

106. Brugger W, Heimfeld S, Berenson RJ, Mertelsmann R,<br />

Kanz L. Reconstitution of hematopoiesis after highdose<br />

chemotherapy by autologous progenitor cells<br />

generated ex vivo. N Engl J Med 1995; 333:283-7.<br />

107. Williams SF, Lee WL, Bender JG, et al. Selection and<br />

expansion of peripheral blood CD34+ cells in autologous<br />

stem cell transplantation for breast cancer.<br />

Blood 1996; 87:1687-91.<br />

108. Choi ES, Nichol JL, Hokom MM, et al. Platelets generated<br />

in vitro from proplatelet-displaying human<br />

megakaryocytes are functional. Blood 1995; 85:402-<br />

13.<br />

109. Okamoto Y, Takaue Y, Saito S, et al. Toxicities associated<br />

with cryopreserved and thawed peripheral<br />

blood stem cell autografts in children with active cancer.<br />

Transfusion 1993; 33:578-81.<br />

110. Burgio GR, Hanson LA, Ugazio AG, eds. Immunology<br />

of the neonate. Berlin: Springer-Verlag; 1987. p.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


66<br />

F. Bertolini et al.<br />

188.<br />

111. Jacoby DR, Olding LB, Oldstone MBA. Immunologic<br />

regulation of fetal-maternal balance. Adv Immunol<br />

1984; 35:157-208.<br />

112. Picker LJ, Treer JR, Ferguson-Darnell B, Collins PA,<br />

Buck D, Terstappen LWMM. Control of lymphocyte<br />

recirculation in man. J Immunol 1993; 150:1105-21.<br />

113. Clement LT, Vink PE, Bradley GE. Novel immunoregulatory<br />

functions of phenotypically distinct subpopulations<br />

of CD4+ cells in the human neonate. J<br />

Immunol 1990; 145:102-8.<br />

114. Burgio GR, Curtoni E, Genova R, Magrini U. Skin reactivity<br />

in childhood: phytohemagglutinin skin test and<br />

streptokinase skin test. Ped Res 1971; 5:88-93.<br />

115. Clerici M, De Palma L, Roilides E, Baker R, Shearer<br />

GM. Analysis of T helper and antigen-presenting cell<br />

functions in cord blood and peripheral blood leukocytes<br />

from healthy children of different ages. J Clin<br />

Invest 1993; 91:2829-36.<br />

116. Maccario R, Nespoli L, Vitiello A, Ugazio AG, Burgio<br />

GR. Lymphocyte subpopulations in the neonate: identification<br />

of an immature subset of OKT8-positive,<br />

OKT3-negative cells. J Immunol 1983; 130:1129-31.<br />

117. Harris DT, Schumacher MJ, Locascio J, et al. Phenotypic<br />

and functional immaturity of human umbilical<br />

cord blood T lymphocytes. Proc Natl Acad Sci USA<br />

1992; 89:10006-10.<br />

118. Moller G. Ontogeny of human lymphocyte function.<br />

Immunol Rev 1981; 57:161.<br />

119. Keever CA, Abu-Hajir M, Graf W, et al. Characterization<br />

of alloreactivity and anti-leukemia reactivity of<br />

cord blood mononuclear cells. Bone Marrow Transplant<br />

1995; 15:407-19.<br />

120. Brugnoni D, Airo P, Graf D, et al. Ineffective expression<br />

of CD40 ligand on cord blood T cells may contribute<br />

to poor immunoglobulin production in the<br />

newborn. Eur J Immunol 1994; 24:1919-24.<br />

121. Montagna D, Maccario R, Ugazio AG, Mingrat G,<br />

Burgio GR. Natural cytotoxicity in the neonate: high<br />

levels of lymphokine activated killer (LAK) activity. Clin<br />

Exp Immunol 1988; 71:158-65.<br />

122. Moretta A, Comoli P, Montagna D, et al. High frequency<br />

of EBV-lymphoblastoid cell line-reactive lymphocytes<br />

in cord blood: evaluation of cytolytic activity<br />

and IL-2 production. Clin Exp Immunol 1996; in<br />

press.<br />

123. Moller G. Immunology of feto-maternal relationship.<br />

Immunol Rev 1983; 75:175.<br />

124. Claas FHJ, Gijbels Y, van der Velden-de Munck J, van<br />

Rood JJ. Induction of B cell unresponsiveness to noninherited<br />

maternal antigens during fetal life. Science<br />

1988; 241:1815-7.<br />

125. Zhang L, van Bree S, van Rood JJ, Claas FHJ. The influence<br />

of breast-feeding on the T cell allorepertoire.<br />

Transplantation 1991; 52:914-6.<br />

126. Wagner JE, Kernan NA, Steinbuch M, Broxmeyer HE,<br />

Gluckman E. Allogeneic sibling umbilical-cord-blood<br />

transplantation in children with malignant and nonmalignant<br />

disease. Lancet 1995; 346:214-9.<br />

127. Burnet FM. The clonal selection theory of acquired<br />

immunity. Nashville: Vanderbilt Univ. Press; 1959.<br />

128. Ridge JP, Fuchs EJ, Matzinger P. Neonatal tolerance<br />

revisited: turning on newborn T cells with dendritic<br />

cells. Science 1996; 271:1723-6.<br />

129. Sarzotti M, Robbins DS, Hoffman PM. Induction of<br />

protective CTL responses in newborn mice by a murine<br />

retrovirus. Science 1996; 271:1726-8.<br />

130. Forsthuber T, Yip HC, Lehmann PV. Induction of TH1<br />

and TH2 immunity in neonatal mice. Science 1996;<br />

271:1728.<br />

131. Risdon G, Gaddy J, Stehman FB, Broxmeyer HE. Proliferative<br />

and cytotoxic responses of human umbilical<br />

cord blood T lymphocytes following allogeneic stimulation.<br />

Cell Immunol 1994; 154:14-24.<br />

132. Harris DT, Schumacher MJ, LoCascio J, Booth A, Bard<br />

J, Boyse EA. Immunoreactivity of umbilical cord blood<br />

and post-partum maternal peripheral blood with<br />

regard to HLA-haploidentical transplantation. Bone<br />

Marrow Transplant 1994; 14:63-8.<br />

133. Harris DT, LoCascio J, Besencon FJ. Analysis of the<br />

alloreactive capacity of human umbilical cord blood:<br />

implications for graft-versus-host disease. Bone Marrow<br />

Transplant 1994; 14:545-53.<br />

134. Deacock SJ, Schwarer AP, Bridge J, Batchelor JR, Goldman<br />

JM, Lechler RI. Evidence that umbilical cord<br />

blood contains a higher frequency of HLA class II-specific<br />

alloreactive T cells than adult peripheral blood.<br />

Transplantation 1992; 53:1128-34.<br />

135. Apperley JF. Umbilical cord blood progenitor cell<br />

transplantation. Bone Marrow Transplant 1994; 14:<br />

187-96.<br />

136. Risdon G, Gaddy J, Horie M, Broxmeyer HE. Alloantigen<br />

priming induces a state of unresponsiveness in<br />

human umbilical cord blood T cells. Proc Natl Acad<br />

Sci USA 1995; 92:2413-7.<br />

137. Harris DT. In vitro and in vivo assessment of the graftversus-leukemia<br />

activity of cord blood. Bone Marrow<br />

Transplant 1995; 15:17-23.<br />

138. Butturini A, Franceschini F, Gale RP. Critical analysis<br />

of T-cell depletion in man. In: Martelli MF, Grignani<br />

F, Reisner Y, eds. T-cell depletion in allogeneic bone<br />

marrow transplantation. Rome: Serono Symposia<br />

Review; 1988. p. 1-13.<br />

139. Ferrara JLM, Deeg HJ. Graft-versus-host disease. N<br />

Engl J Med 1991; 324:667-74.<br />

140. Reisner Y, Kapoor N, Kirkpatrick D, et al. Transplantation<br />

for severe combined immunodeficiency with<br />

HLA-A, B, D, Dr incompatible parental marrow fractioned<br />

by soybean agglutinin and sheep red blood<br />

cells. Blood 1983; 61:341-8.<br />

141. O’ Reilly RJ, Kernan N, Cunningham I, et al. Soybean<br />

lectin agglutination and E-rosette depletion for<br />

removal of T-cells from HLA-identical and non-identical<br />

marrow grafts administered for the treatment of<br />

leukemia. In: Martelli MF, Grignani F, Reisner Y, eds.<br />

T cell depletion in allogeneic bone marrow transplantation.<br />

Rome: Serono Symposia Review; 1988. p. 123-<br />

9.<br />

142. Ash RC, Casper JT, Chitamba CR, et al. Successful<br />

allogeneic transplantation of T-cell depleted bone<br />

marrow from closely HLA-matched unrelated donors.<br />

N Engl J Med 1990; 322:485-94.<br />

143. Drobyski WR, Ash RC, Casper JT, et al. Effect of T-cell<br />

depletion as graft-versus-host disease prophylaxis on<br />

engraftment, relapse, and disease-free survival in unrelated<br />

marrow transplantation for chronic myelogenous<br />

leukemia. Blood 1994; 83:1980-7.<br />

144. Kernan NA, Flomenberg N, Dupont B, et al. Graft<br />

rejection in recipients of T-cell-depleted HLA-nonidentical<br />

marrow transplants for leukemia. Transplantation<br />

1987; 43:842-7.<br />

145. Goldman JM, Gale RP, Horowitz MM, et al. Bone<br />

marrow transplantation for chronic myelogenous<br />

leukemia in chronic phase: increased risk of relapse<br />

associated with T cell depletion. Ann Intern Med<br />

1988; 108:806-14.<br />

146. Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia<br />

reactions after bone marrow transplantation.<br />

Blood 1990; 75:555-62.<br />

147. O’Reilly R. T-cell depletion and allogeneic bone marrow<br />

transplantation. Semin Hematol 1992; 29(Suppl.<br />

1):20-6.<br />

148. Kessinger A, Smith DM, Strandjord SE, et al. Allogeneic<br />

transplantation of blood derived, T-cells<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Non bone marrow allogeneic hemopoietic stem cells<br />

67<br />

depleted hemopoietic stem cells after myeloablative<br />

treatment in a patient with acute lymphoblastic<br />

leukemia. Bone Marrow Transplant 1989; 4:643-6.<br />

149. Suzue T, Kawano Y, Takaue Y, et al. Cell processing<br />

protocol for allogeneic peripheral blood stem cells<br />

mobilized by granulocyte colony-stimulating factor.<br />

Exp Hematol 1994; 22:888-92.<br />

150. Keij JF, Groenewegen AC, Visser JWM. High-speed<br />

photodamage cell sorting: an evaluation of the ZAP-<br />

PER prototype. In: Darzynkiewicz Z, Robinson JP,<br />

Crissman HA, eds. Methods in cell biology. New York:<br />

Academic Press; 1994. p. 371-85.<br />

151. Link H, Arseniev L, Bahre O, et al. Combined transplantation<br />

of allogeneic bone marrow and CD34+<br />

blood cells. Blood 1995; 86:2500-8.<br />

152. Arseniev L, Tischler HJ, Battmer K, et al. Treatment of<br />

poor graft function with allogeneic CD34+ cells<br />

immunoselected from G-CSF-mobilized peripheral<br />

blood progenitor cells of the marrow donor. Bone<br />

Marrow Transplant 1994; 14:791-7.<br />

153. Di Persio J, Martin B, Abbond C, et al. Allogeneic BMT<br />

using bone marrow and CD34-selected mobilized<br />

PBSC; comparison to BM alone and mobilized PBSC<br />

alone. Exp Hematol 1994; 22:697a.<br />

154. Lemoli RM, Tazzari PL, Fortuna A, et al. Positive selection<br />

of hematopoietic CD34+ stem cells provides<br />

“indirect purging” of CD34– lymphoid cells and the<br />

purging efficiency is increased by anti CD2 and anti-<br />

CD30 immunotoxins. Bone Marrow Transplant 1994;<br />

13:465-71.<br />

155. Behringer D, Bertz H, Hardung-Backes M, et al. Allogeneic<br />

peripheral blood progenitor cell transplantation<br />

with or without CD34+ stem cell selection: follow<br />

up and immune reconstitution. Bone Marrow Transplant<br />

1996; 17 (Suppl. 1):298a.<br />

156. Kernan NA, Collins NH, Juliano L, et al. Clonable T<br />

lymphocytes in T cell-depleted bone marrow transplants<br />

correlate with development of graft-vs-host disease.<br />

Blood 1986; 68:770-3.<br />

157. Aversa F, Terenzi A, Tabilio A, et al. Addition of PBSCs<br />

to the bone marrow inoculum allows engraftment of<br />

T-cell-depleted HLA-incompatible transplants. Br J<br />

Haematol 1996; 93 (Suppl. 2):146.<br />

158. Tabilio A, Falzetti F, Aversa F, et al. T cell depletion of<br />

peripheral blood stem cells by CD34+ cell selection<br />

and E-rosettes. Submitted for publication.<br />

159. Fernandez JM, Yan Y, Bleans S, et al. T-cell depletion<br />

of peripheral blood progenitor cells (PBPC) by either<br />

CD34+ selection and E-rosette depletion or SBA<br />

agglutination and E-rosette depletion: comparison of<br />

T-cell and hematopoietic cell progenitor recovery.<br />

Blood 1995; 86 (Suppl. 1):626a.<br />

160. Slaper-Cortenbach ICM, Wijngaarden-du Bois MJGJ,<br />

de Vriesvan Rossen A, et al. New immunorosette technique<br />

for the depletion of T cells from allogeneic stem<br />

cell transplantation. Br J Haematol 1996; 93(Suppl.<br />

2):170a.<br />

161. Marolleau JP, Dal Cortivo L, Robert J, et al. The new<br />

clinical grade peptide PR34+TM stem cell releasing<br />

agent. In: Latest advancements in immunomagnetic<br />

Cell selection for grafts engineering in autologous and<br />

allogeneic stem cell transplantation. EBMT Baxter<br />

satellite symposium. Vienna, March 3rd, 1996.<br />

162. Kunkel L, Mills B, Burgess J, et al. Early experiences<br />

with selection of CD34+ cells and peptide release<br />

using a fully automated selection process. In: Latest<br />

advancements in immunomagnetic Cell selection for<br />

grafts engineering in autologous and allogeneic stem<br />

cell transplantation. EBMT Baxter satellite symposium.<br />

Vienna, March 3rd, 1996.<br />

163. Berenson RJ, Bensinger WI, Hill RS, et al. Engraftment<br />

after infusion of CD34+ marrow cells in patients with<br />

breast cancer or neuroblastoma. Blood 1991; 77:<br />

1717-22.<br />

164. Gorin NC, Lopez M, Laporte JP, et al. Preparation and<br />

succesful engraftment of purified CD34+ bone marrow<br />

progenitor cells in patients with non Hodgkin’s<br />

lymphoma. Blood 1995; 85:1647-54.<br />

165. Lemoli RM , Fortuna A, Motta MR, et al. Concomitant<br />

mobilization of plasma cells and hematopoietic<br />

progenitors into peripheral blood of multiple myeloma<br />

patients: positive selection and transplantation of<br />

enriched CD34+ cells to remove circulating tumor<br />

cells. Blood 1996; 87:1625-34.<br />

166. Steinman RM. The dendritic cell system and its role in<br />

immunogenicity. Annu Rev Immunol 1991; 9:271-96.<br />

167. Caux C, Vanbervliet B, Massacrier C, et al. B70/B7-2<br />

is identical to CD86 and is the major functional ligand<br />

for CD28 expressed on human dendritic cells. J Exp<br />

Med 1994; 180:1841-7.<br />

168. Sallusto F, Lanzavecchia A. Efficient presentation of<br />

soluble antigen by cultured human dendritic cells is<br />

maintained by granulocyte/macrophage colony-stimulating<br />

Factor plus Interleukin 4 and downregulated<br />

by tumor necrosis factor. J Exp Med 1994; 179:1109-<br />

18.<br />

169. Romani R, Gruner S, Brang D, et al. Proliferating dendritic<br />

cell progenitors in human blood. J Exp Med<br />

1994; 180:83-93.<br />

170. Hart DN, Starling GC, Calder VL, Fernando NS.<br />

B7/BB-1 is a leukocyte differentiation antigen on<br />

human dendritic cells induced by activation. Immunology<br />

1993; 79:616-20.<br />

171. Guinan EC, Gribben JG, Boussiotis VA, Freeman GJ,<br />

Nadler LM. Pivotal role of the B7:CD28 pathway in<br />

transplantation tolerance and tumor immunity. Blood<br />

1994; 84:3261-82.<br />

172. Reid CDL, Stackpoole A, Meager A, Tikerpae J. Interactions<br />

of tumor necrosis factor with granulocytemacrophage<br />

colony-stimulating factor and other<br />

cytokines in the regulation of dendritic cell growth in<br />

vitro from early bipotent CD34+ progenitors in<br />

human bone marrow. J Immunol 1992; 149:2681-8.<br />

173. Szalbocs P, Moore MAS, Young JW. Expansion of<br />

immunostimulatory dendritic cells among the myeloid<br />

progeny of human CD34+ bone marrow precursors<br />

cultured with c-kit ligand, GM-CSF and TNF-α. J<br />

Immunol 1995; 154:5851-61.<br />

174. Young JW, Szalbocs P, Moore MAS. Identification of<br />

dendritic cell colony-forming units among normal<br />

human CD34+ bone marrow progenitors that are<br />

expanded by c-kit ligand and yeld pure dendritic cell<br />

colonies in the presence of granulocyte/macrophage<br />

colony-stimulating factor and tumor necrosis factorα.<br />

J Exp Med 1995; 182:1111-20.<br />

175. Siena S, Di Nicola M, Bregni M, et al. Massive ex vivo<br />

generation of functional dendritic cells from mobilized<br />

CD34+ blood progenitors for anticancer therapy.<br />

Exp Hematol 1995; 23:1463-71.<br />

176. Rosenzwaig M, Canque B, Gluckman JC. Human dendritic<br />

cell differentiation pathway from CD34+ hematopoietic<br />

precursor cells. Blood 1996; 87:535-44.<br />

177. Strunk D, Rappesberger K, Egger C, et al. Generation<br />

of human dendritic cells/Langerhans cells from circulating<br />

CD34+ hematopoietic progenitor cells. Blood<br />

1996; 87:1292-302.<br />

178. Schwartz RH. A cell culture model for T lymphocyte<br />

clonal anergy. Science 1990; 48:1349-56.<br />

179. Tan P, Anasetti C, Hansen JA, et al. Induction of<br />

alloantigen-specific hyporesponsiveness in human T<br />

lymphocytes by blocking interaction of CD28 with its<br />

natural ligand B7/BB1. J Exp Med 1993; 177:165-73.<br />

180. DeSilva DS, Urdhal KB, Jenkins MK. Clonal anergy is<br />

induced in vitro by T cell receptor occupancy in the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


68<br />

F. Bertolini et al.<br />

absence of proliferation. J Immunol 1991; 147:2461-<br />

6.<br />

181. Jenkins MK. The ups and downs of T cell costimulation.<br />

Immunity 1994; 1:443-6.<br />

182. Rondelli D, Andrews RG, Hansen JA, Ryncarz R, Faerber<br />

MA, Anasetti C. Alloantigen presenting function of<br />

normal human CD34+ hematopoietic cells. Blood<br />

1996; 88:2619-25.<br />

183. Gunji Y, Nakamura M, Yanigisawa M, Miura Y, Suda<br />

T. Expression and function of adhesion molecules on<br />

human hematopoietic stem cells: CD34+LFA-1- cells<br />

are more primitive than CD34+LFA-1+ cells. Blood<br />

1992; 80:429-36.<br />

184. Krensky AM, Mentzer SJ, Clayberger C, et al. Heritable<br />

lymphocyte function-associated antigen-1 deficiency:<br />

abnormalities of cytotoxicity and proliferation<br />

associated with abnormal expression of LFA-1. J<br />

Immunol 1985; 135:3102-8.<br />

185. Andrews RG, Bryant EM, Bartelmez SH, et al. CD34+<br />

marrow cells, devoid of T and B lymphocytes, reconstitute<br />

stable lymphopoiesis and myelopoiesis in<br />

lethally irradiated allogeneic baboons. Blood 1992;<br />

80:1693-701.<br />

186. Bensinger WI, Rowley S, Appelbaum FR, et al. CD34<br />

selected allogeneic peripheral blood stem cell (PBSC)<br />

transplantation in older patients with advanced<br />

hematologic malignancies. Blood 1995; 86 (Suppl.1):<br />

376.<br />

187. Schiller G, Rowley S, Buckner CD, et al. Transplantation<br />

of allogeneic CD34+ peripheral blood stem cells<br />

(PBSC) in older patients with advanced hematologic<br />

malignancy. Blood 1995; 86 (Suppl.1):1545.<br />

188. Link H, Arseniev L, Bahre O, et al. Transplantation of<br />

allogeneic hematopoiesis by selected CD34+ blood<br />

cells. Blood 1995; 86 (Suppl.1):1150.<br />

189. Urbano-Ispizua A, Rozman C, Marìn P, et al. Selection<br />

of CD34+ peripheral blood progenitor cells (PBPC)<br />

for allogeneic transplantation. Blood 1995; 86(Suppl.1):901.<br />

190. Holland HK, Bray AM, Geller RB, et al. Transplantation<br />

of HLA-identical positively selected CD34+ cells<br />

of peripheral blood (PBSC) and marrow (BM) from<br />

related donors results in prompt engraftment and low<br />

incidence of GVHD in recipients undergoing allogeneic<br />

transplantation. Blood 1995; 86(Suppl.1):<br />

1543.<br />

191. Weaver CH, Longin K, Buckner CD, et al. Lymphocyte<br />

content in peripheral blood mononuclear cells collected<br />

after the administration of recombinant human<br />

granulocyte colony-stimulating factor. Bone Marrow<br />

Transplant 1994; 13:411-5.<br />

192. Nicola NA. Why do hemopoietic growth factor receptors<br />

interact with each other? Immunol Today 1987;<br />

8:134-40.<br />

193. Noga SJ, Davis JM, Vogelsgang GB, et al. The combined<br />

use of elutriation and CD8/magnetic bead to<br />

engineer the bone marrow allograft. In: Worthington-<br />

White DA, Gee AP, Gross S, eds. Advances in bone<br />

marrow purging and processing. New York: Wiley-Liss;<br />

1992. p. 411.<br />

194. Owens AH, Santos GW. The induction of graft versus<br />

host disease in mice treated with cyclophosphamide.<br />

J Exp Med 1968; 128:277-91.<br />

195. van Bekkum DW, de Vries MJ. Radiation chimaeras.<br />

New York: Academic Press; 1967.<br />

196. Atkinson K, Farrell C, Chapman G, et al. Female marrow<br />

donors increase the risk of acute graft-versushost-disease:<br />

effect of donor age and parity and analysis<br />

of cell subpopulations in the donor marrow inoculum.<br />

Br J Haematol 1986; 63:231-9.<br />

197. Jansen J, Goselink HM, Veenhof WFJ, et al. The impact<br />

of the composition of the bone marrow graft on<br />

engraftment and graft-versus-host-disease. Exp Hematol<br />

1983; 11:967-73.<br />

198. Murphy WJ, Reynolds CW, Tiberghien P, Longo DL.<br />

Natural killer cells and bone marrow transplantation.<br />

J Natl Cancer Inst 1993; 85:1475-82.<br />

199. Zeis M, Uharek L, Glass B, et al. Natural killer cells<br />

given after allogeneic BMT induce strong graft-vsleukemia<br />

(GVL) effects. Br J Haematol 1996; 93 (suppl<br />

2):76a.<br />

200. van Bekkum DW. The selective elimination of immunologically<br />

competent cells from bone marrow and<br />

lymphatic cell mixtures. Effect of storage at 4°C.<br />

Transplantation 1964; 2:393-8.<br />

201. Eckardt JR, Roodman GD, Boldt DH, et al. Comparison<br />

of engraftment and acute GvHD in patients<br />

undergoing cryopreserved or fresh allogeneic BMT.<br />

Bone Marrow Transplant 1993; 11:125-31.<br />

202. Dreger P, Suttorp M, Haferlach T, et al. Allogeneic<br />

granulocyte colony-stimulating factor mobilized peripheral<br />

blood progenitor cells for treatment of engraftment<br />

failure after bone marrow transplantation.<br />

Blood 1993; 81:1404-7.<br />

203. Rambaldi A, Viero P, Bassan R, et al. G-CSF-mobilized<br />

peripheral blood progenitor cells for allogeneic<br />

transplantation of resistant or relapsing acute<br />

leukemias. Leukemia 1996; 10:860-5.<br />

204. Kurtzberg J, Laughlin M, Graham ML, et al. Placental<br />

blood as a source of hematopoietic stem cells for<br />

transplantation into unrelated recipients. N Engl J<br />

Med 1996; 335:157-66.<br />

205. Laporte JP, Gorin NC, Rubinstein P, et al. Cord-blood<br />

transplantation from an unrelated donor in an adult<br />

with chronic myelogenous leukemia. N Engl J Med<br />

1996; 335:167-70.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


eview<br />

Clinical use of allogeneic<br />

hematopoietic stem cells from<br />

sources other than bone marrow<br />

haematologica 2000; 85(supplement to no. 12):69-91<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia ed Oncologia<br />

Medica “L. & A. Seràgnoli”, Policlinico S. Orsola, via Massarenti<br />

9, 40138 Bologna, Italy.<br />

WILLIAM ARCESE,* FRANCO AVERSA,° GIUSEPPE BANDINI, #<br />

ARMANDO DE VINCENTIIS, @ MICHELE FALDA,^ LUIGI LANATA, @<br />

ROBERTO M. LEMOLI, # FRANCO LOCATELLI, § IGNAZIO MAJOLINO, **<br />

PAOLA ZANON, °° SANTE TURA #<br />

*Department of Cellular Biotechnology and Hematology, University<br />

“La Sapienza”, Rome; °Department of Clinical Medicine,<br />

Pathology and Pharmacology, Section of Clinical Hematology<br />

and Immunology, University of Perugia, Perugia; # Institute of<br />

Hematology and Medical Oncology "L. & A. Seragnoli", University<br />

of Bologna, Bologna; @ Dompé Biotec SpA, Milan; ^Division of<br />

Hematology, Ospedale S. Giovanni Battista, Turin; § Department<br />

of Pediatrics, University of Pavia and IRCC Policlinico S. Matteo,<br />

Pavia; ** Department of Hematology and BMT Unit, Ospedale “V.<br />

Cervello”, Palermo; °°Amgen Italia SpA, Milan, Italy<br />

Background and Objectives. Peripheral blood stem cells<br />

(PBSC) are being increasingly used as an alternative to<br />

conventional allogeneic bone marrow (BM) transplantation.<br />

This has prompted the Working Group on CD34-<br />

Positive Hematopoietic Cells to evaluate current utilization<br />

of allogeneic PBSC in clinical hematology.<br />

Evidence and Information Sources. The method<br />

employed for preparing this review was that of informal<br />

consensus development. Members of the Working<br />

Group met three times, and the participants at these<br />

meetings examined a list of problems previously prepared<br />

by the chairman. They discussed the single<br />

points in order to reach an agreement on different<br />

opinions and eventually approved the final manuscript.<br />

Some of the authors of the present review have been<br />

working in the field of stem cell transplantation and<br />

have contributed original papers in peer-reviewed journals.<br />

In addition, the material examined in the present<br />

review includes articles and abstracts published in<br />

journals covered by the Science Citation Index® and<br />

Medline®.<br />

State of the Art. Review of the current literature shows<br />

that unmanipulated allogeneic PBSC give prompt and<br />

stable engraftment in HLA-identical sibling recipients.<br />

Despite the much higher number of T-cells infused, the<br />

incidence and severity of acute GVHD after PBSC transplant<br />

seems comparable to that observed with bone<br />

marrow (BM) cells. In comparison to the latter, PBSC<br />

probably ensure faster immunologic reconstitution in<br />

the early post-transplant period. Controversial results<br />

on the incidence and severity of acute-GVHD have been<br />

reported when CD34 + selection methods are used.<br />

Prospective randomized trials are underway to compare<br />

the results of PBSC and BM allogeneic transplantation.<br />

In mismatched family donor transplants, T-cell depleted<br />

PBSC successfully engraft immune-myeloablated<br />

recipients through a mega-cell-dose effect able to overcome<br />

the HLA barrier. Experience with PBSC in the context<br />

of unrelated donor transplants is currently anecdotal<br />

and prospective trials should be completed before<br />

that practice becomes routine. Finally, there is also<br />

limited evidence that, following induction chemotherapy,<br />

the addition of PBSC to donor lymphocyte<br />

infusion (DLI) for treatment of leukemia relapse after<br />

BMT may improve the safety and effectiveness of DLI<br />

itself. Concerning cord blood (CB) transplants, the<br />

most interesting aspects are the ease of CB collection<br />

and storage, the low risk of viral contamination and the<br />

low immune reactivity of CB cells. This last property has<br />

its clinical counterpart in an apparently reduced incidence<br />

and severity of acute GVHD both in sibling and<br />

unrelated CB transplants, probably making the level of<br />

donor/recipient HLA disparity acceptable a greater<br />

degree with respect to what is required for transplants<br />

from other sources.<br />

©2000, Ferrata Storti Foundation<br />

Key words: hematopoietic stem cells, bone marrow, cord blood, peripheral<br />

blood, allogeneic transplantation, graft-versus-host disease<br />

In the field of allogeneic transplantation the use of<br />

alternative sources of stem cells, namely peripheral<br />

blood stem cells (PBSC) 1 and placental cord blood (CB)<br />

stem cells, 2 is rapidly expanding. The European Group for<br />

Blood and Marrow Transplantation (EBMT) registered<br />

only 12 allogeneic PBSC transplants in 1993, but this<br />

number increased to 180 in 1994 and to 571 in 1995. 3<br />

Concerning cord blood (CB) transplants, following initial<br />

attempts 4,5 considerable experience has now been<br />

achieved in the USA and Europe so that this modality is<br />

entering a phase of extensive clinical application, with<br />

hundreds of procedures registered both from sibling and<br />

unrelated donors, 6,7 The ease of collection and storage of<br />

CB stem cells and the apparent tolerance-inducing property<br />

of CB CD8 + suppressor cells 8 are the most interesting<br />

aspects of this latter source.<br />

Stem cells in peripheral blood and CB both possess a<br />

migratory status and differ in part from those found in<br />

bone marrow with respect to their biological and functional<br />

properties. However, while PBSC are envisaged as<br />

a means of improving results by increasing the number<br />

of cells available, placental CB stem cells open the realistic<br />

perspective of increasing the number of transplants<br />

thanks to the availability of thousands of cord samples<br />

for patients who lack a compatible donor among family<br />

haematologica vol. 85(suppl. to n. 12):December 2000


70<br />

W. Arcese et al.<br />

members.<br />

This search for new stem cell sources also arises from<br />

the fact that allogeneic bone marrow transplantation still<br />

carries a high procedure-related mortality and disease<br />

recurrence rate. Cell dose has an influence on engraftment<br />

and chance of survival. In a retrospective study<br />

Bacigalupo et al. 9 showed that patients with hematologic<br />

malignancies who receive allogeneic bone marrow<br />

grafts with higher CFU-GM numbers have significantly<br />

higher platelet counts on day +80 and a lower mortality<br />

rate than those who receive fewer CFU-GM. The effect of<br />

CMV infection on platelet counts also appears to be less<br />

pronounced when the number of progenitor cells is higher.<br />

The use of more hematopoietic progenitors would then<br />

result in improved transplant outcome.<br />

The growing interest in allogeneic PBSC induced the<br />

Italian Bone Marrow Transplant Group (GITMO) to draw<br />

up a list of recommendations that were originally published<br />

in 1995 10 and recently revised in light of the<br />

increasing experience gained worldwide during the last<br />

two years. 11 Moreover, in a previous issue of this Journal 12<br />

a review article analyzed the biological and technical<br />

aspects of PB and CB stem cells. The key aspects dealt<br />

with were the mobilization and collection methods, the<br />

capacity for stable hemopoietic reconstitution, kinetic<br />

characteristics and immunological features.<br />

Historical background<br />

Interest in allogeneic transplantation of PBSC began<br />

thirty years ago. In the late sixties, based on an earlier<br />

demonstration that autologous PBSC were capable of<br />

restoring irradiation-myeloablated hematopoiesis, 13-15<br />

the Seattle group reported the first successful attempts<br />

at allogeneic PBSC transplantation in dogs 16,17 and nonhuman<br />

primates. 18 However, due to the high GVHD incidence,<br />

those experiments were unable to demonstrate<br />

long-term stability of the graft. Only a decade later did<br />

purification of PBSC and application of cytogenetic<br />

methods allow a group of German investigators 19,20 to<br />

document in dogs the stability of donor-derived hemopoietic<br />

function for more than ten years after PBSC allogeneic<br />

transplantation.<br />

A key issue at that time was the low number of progenitors<br />

in steady-phase peripheral blood, and clinical<br />

application of PBSC was limited to autologous transplantation<br />

in CML, 21 where hematopoietic progenitors,<br />

mostly of the leukemic counterpart, circulate in high<br />

numbers and can easily be collected by apheresis without<br />

any prior stimulation. A further step was the demonstration<br />

that the PBSC level increases dramatically during<br />

the post-chemotherapy recovery phase; 22 however, it<br />

was the advent of G-CSF and GM-CSF that provided the<br />

rapid expansion of PBSC technology and led to their use<br />

in allogeneic transplantation as well. The pioneer work<br />

of Socinski et al. 23 and Gianni et al. 24 established the ability<br />

of hematopoietic growth factors to expand the circulating<br />

progenitor cell pool either when used alone or<br />

in conjunction with chemotherapy. However, transferring<br />

growth-factor PBSC mobilization strategy from autograft<br />

patients to normal donors took some years. In fact,<br />

the safety of growth factors and the clinical applicability<br />

of allogeneic PBSC in terms of GVHD incidence and<br />

long-term engraftment represented serious reasons for<br />

caution. Clinical PBSC allogeneic transplantation began<br />

in 1989, when Kessinger et al. 25 reported the first attempt<br />

in an HLA-matched recipient with ALL. The patient was<br />

an 18-year-old man in third remission after CNS and<br />

testicular relapse. His sibling female donor preferred to<br />

donate PBSC rather than bone marrow, and she underwent<br />

10 apheretic procedures without any mobilization<br />

treatment. The apheresis product was T-depleted by<br />

sheep erythrocyte rosetting and infused after conditioning<br />

with high-dose Ara-C and TBI. The patient achieved<br />

full donor engraftment as demonstrated by cytogenetic<br />

studies but died on day +32, and sustained engraftment<br />

could not be demonstrated.<br />

Four years later, in 1993, Russell et al. 26 reported<br />

another transplant in a patient whose sibling donor presented<br />

an increased risk of complications from anesthesia.<br />

In this case, 10 mg/kg/day of G-CSF were given<br />

to mobilize PBSC. The cells were collected at two leukaphereses<br />

containing 36.8×10 4 /kg CFU-GM and infused<br />

without any prior manipulation. Engraftment occurred<br />

rapidly and GVHD did not develop despite the high T-cell<br />

content of the graft sample. The same year, a group of<br />

investigators from Kiel University 27 also successfully<br />

employed allogeneic PBSC. A 47-year old AML patient<br />

who failed to engraft after bone marrow transplantation<br />

from an HLA-identical sibling donor was infused with<br />

the unmanipulated product of 3 leukaphereses performed<br />

after treating the donor with 6 mg/kg/day G-<br />

CSF. Engraftment occurred on day +14, with moderate<br />

acute GVHD that responded to immunosuppression<br />

starting on day +18. Restriction fragment length polymorphism<br />

(RFLP) typing demonstrated full donor<br />

engraftment up to 60 days following transplantation.<br />

Another important step was the five PBSC transplants<br />

from syngeneic donors performed in Seattle and reported<br />

in 1993: 28 with a median of 9.6×10 6 /kg CD34 + cells<br />

infused, the patients engrafted 0.5×10 9 /L granulocytes<br />

on day 13 and 20×10 9 /L platelets on day 10. In 1995<br />

three separate reports appeared in the same issue of<br />

Blood, one from Seattle, 29 a second from Houston 30 and<br />

the other from Kiel; 31 a total of 25 patients were allografted<br />

with PBSC from their HLA-identical sibling<br />

donors. Acute GVHD was apparently not increased in<br />

those series. Molecular analysis of engraftment 30,31 furnished<br />

definitive proof of the experimental data 20 suggesting<br />

that allogeneic PBSC contain true long-term<br />

repopulating stem cells. The high engraftment potential<br />

of PBSC was exploited by the Perugia team 33 to successfully<br />

transplant leukemia patients from their haploidentical,<br />

three-loci-incompatible family donors<br />

through T-cell depletion. Finally, Ringdén et al. 34 recently<br />

reported the use of allogeneic PBSC in selected unrelated<br />

donor transplants.<br />

The kinetics of PBSC under cytokine mobilization was<br />

extensively analyzed in a previous review published in<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

71<br />

this Journal. 12 PBSC mobilization in healthy donors is<br />

best accomplished with G-CSF. The aspect of donor<br />

safety was analyzed in a cooperative GITMO study35 in<br />

which short-term side effects were shown to be minimal.<br />

Ten µg/kg/day of G-CSF for 5 days enabled the collection<br />

of >4×10 6 /kg CD34 + cells with two aphereses in<br />

85% of donors. Variations in blood counts included a<br />

sharp elevation of WBC and CD34 + cells and a moderate<br />

transitory thrombocytopenia. One problem, however,<br />

is the lack of data on the late effects of G-CSF. At the<br />

Geneva conference on allogeneic PBSC, Hasenclever and<br />

Sextro 36 presented a feasibility study of long-term risk<br />

analysis. In order to demonstrate a tenfold increase in<br />

leukemia risk, more than 2000 healthy PBSC donors<br />

would have to be followed for over 10 years. A control<br />

group of BMT donors of equal size would also be necessary.<br />

Such a study could only be carried out on a multi-national<br />

basis.<br />

Transplantation of allogeneic PBSC<br />

from HLA-identical siblings<br />

Conditioning regimens and GVHD prophylaxis<br />

Conditioning regimens employed in PBSC transplantation<br />

are the same as those used for bone marrow transplantation<br />

(BMT). As listed in Table 1, the majority of<br />

patients received CY-TBI or BU-CY with CY at 120 or 200<br />

mg/kg. Indeed, 33.8% and 24.5% of reported patients<br />

were conditioned with CY-TBI and BU-CY, respectively.<br />

Analogously to BM transplantation, patients with SAA<br />

received cyclophosphamide alone or in association with<br />

ATG.<br />

Recently, some innovative regimens have been developed<br />

in order to: i) increase antitumor activity; ii) reduce<br />

treatment-related toxicity. For instance, high dose Ara-<br />

C or VP16 has been employed for patients with more<br />

advanced disease. Others considered thiotepa, a potent<br />

myeloablative drug first introduced in conditioning by<br />

the Perugia group. 37 This latter compound was used along<br />

with classical BU-CY2 in a large series at the M.D. Anderson<br />

Cancer Center 38,39 or was associated with cyclophosphamide.<br />

40 In this study, thiotepa was introduced in the<br />

hope of reducing the liver toxicity of busulfan.<br />

It is interesting to note the recent introduction of fludarabine,<br />

a purine analogue initially proposed at conventional<br />

dosage by the M.D. Anderson group 41 and at<br />

a higher dose by the Perugia group in HLA-mismatched<br />

transplants. 42 Fludarabine has been associated with several<br />

other drugs or drug combinations including<br />

cyclophosphamide plus cis-platinum, high-dose (HD)<br />

Ara-C, idarubicine plus Ara-C or melphalan. 41,43 These<br />

fludarabine containing regimens were followed by full<br />

engraftment with complete donor chimerism in the<br />

absence of severe aplasia. Basically, these new regimens<br />

allow allograft even in older or medically infirm patients,<br />

since they reduce the toxicity but still maintain an effective<br />

graft versus leukemia reaction. For the same reason<br />

Adkins et al. 44 combined low-dose TBI (550 rad) at a<br />

high dose rate (30 cGy/min) with cyclophosphamide 120<br />

mg and methylprednisolone 2 g over two days.<br />

A non-myeloablative regimen with busulfan and<br />

methylprednisolone combined with immunosuppression<br />

with the CD3 monoclonal antibody was used by Tan et<br />

al., 45 while Slavin et al. 46 employed fludarabine and ATG<br />

as intensive immunosuppression associated with busulfan<br />

at 4 mg/kg/day over 2 days. Although these regimens<br />

are not specifically designed for PBSC transplantation,<br />

the high number of inoculated PB stem cells overcomes<br />

graft rejection, favoring rapid and stable chimerism.<br />

The large amount of stem cells in the inoculum might<br />

explain the full engraftment observed in a patient who<br />

could not complete the whole regimen and received<br />

busulfan alone as preparation. 47<br />

The large number of CD3 +ve cells present in the PBderived<br />

inoculum has raised some concerns about the<br />

severity of GVHD following PBSC allograft. However,<br />

GVHD prophylaxis has not been substantially modified<br />

from the standard regimens used for bone marrow transplants.<br />

Cyclosporin A (CsA) has been used alone (2.9%)<br />

or in association with either methotrexate (MTX) (49.8%)<br />

or methylprednisolone (MP) (21.2%) in 71% of patients<br />

(Table 1).<br />

New immunosuppressive regimens including tacrolimus<br />

(FK506) in association with methylprednisolone,<br />

methotrexate 39 or monoclonal antibodies 48 have been<br />

explored in 17% of cases.<br />

Finally, some centers have developed techniques for in<br />

vitro stem cell enrichment. However, using Ceprate-cell<br />

separation, a 2-3 log depletion of T-lymphocytes is not<br />

enough to avoid the risk of GVHD and further immunosuppression<br />

is generally required.<br />

In some institutions cryopreservation of collected PBSC<br />

is preferred to freshly collected material for several reasons.<br />

First, cryopreservation allows precise evaluation of<br />

the hemopoietic progenitor content in the harvested<br />

material. Second, the allograft may be scheduled at the<br />

proper time once adequate quantities of PBSC are collected.<br />

Finally, although still unproven, a reduced risk of<br />

acute GVHD in patients transplanted with cryopreserved<br />

BM cells has been suggested. 49<br />

Table 1. Conditioning regimens and GVHD prophylaxis: 301<br />

patients.<br />

Regimens No. % GVHD prophylaxis No. %<br />

CYTBI 102 33.8% CsA-MTX 150 49.8%<br />

BUCY 68 24.5% CsA-MP 82 21.2%<br />

TioBUCY 67 22.2% FK506-MTX 7 2.3%<br />

VP16TBI 10 3.3% FK506-MP 44 14.6%<br />

TioCY 8 2.6% CsA 15 2.9%<br />

CYATG 3 0.9% MTX 1 0.3%<br />

Others 43 16% Others 2 0.6%<br />

haematologica vol. 85(suppl. to n. 12):December 2000


72<br />

W. Arcese et al.<br />

Engraftment<br />

Engraftment kinetics following PBSC allograft has been<br />

extensively investigated. None of the studies include<br />

patients dying before day 21. The median time to reach<br />

an absolute neutrophil count(ANC) above 0.5×10 9 /L<br />

ranges between 10-16 days. The reported incidence of<br />

graft failure is definitely low: 1/59 in the EBMT survey, 50<br />

1/41 in the MD Anderson series, 51 1/26 in the Canadian<br />

experience. 52 The few rejections occurred in transplants<br />

with 1-2 antigen disparity. Platelet engraftment is also<br />

prompt, with median time to achieve an absolute platelet<br />

count (APC) of 20×10 9 /L ranging between 10 and 18 days<br />

in the reported series.<br />

Platelet more than neutrophil engraftment may be<br />

affected by acute GVHD or CsA toxicity as well as by early<br />

relapse or progressive disease. 50 In a recent report by<br />

the Genoa group a second infusion of PBSC without conditioning<br />

was required to achieve full engraftment of<br />

platelets in three out of thirty-one patients. 40<br />

The prompt engraftment offered by PBSC implies a<br />

reduction in transfusion need. The reported transfusion<br />

requirements range from 2 to 10 packed red cell units and<br />

from 3 to 12 platelet units.<br />

Furthermore, GVHD prophylaxis may adversely influence<br />

the time to engraftment. In particular, methotrexate<br />

given for GVHD prophylaxis delays neutrophil and<br />

platelet engraftment. 53,54<br />

Growth factors, mainly G-CSF, have been employed in<br />

several studies to speed-up engraftment. Urbano-<br />

Ispizua reported that G-CSF given to patients not receiving<br />

methotrexate accelerates neutrophil recovery<br />

(p=0.001); median time to >20×10 9 /L platelets was significantly<br />

delayed (p 0=0.01), although the time to reach<br />

5010 9 /L platelets was not affected. No difference in<br />

engraftment kinetics was seen between cryopreserved<br />

and fresh PBSC when G-CSF was administered following<br />

transplantation. 50,55<br />

The studies carried out so far are not sufficient to<br />

draw definitive conclusions about engraftment with<br />

PBSC as compared to engraftment with BM. Randomized<br />

studies are still in progress and results are not yet<br />

available. Most information comes from comparisons of<br />

PBSC results with historical data from BM transplants.<br />

A highly informative study was reported by the Seattle<br />

group, which compared 37 PBSC transplanted<br />

patients with a historical group of 37 bone marrow<br />

recipients. 56 Patients were well matched for diagnosis,<br />

disease stage, age and graft versus host prophylaxis.<br />

Faster neutrophil engraftment, 14 versus 16 days to<br />

reach more than 0.5×10 9 /L (p=0.0063), and earlier<br />

achievement of platelet transfusion independence, 11<br />

versus 15 days (p=0.0014), were observed in PBSC recipients<br />

compared to the BM control group. Consequently,<br />

the median number of platelet units transfused was<br />

24 versus 118 (p=0.0001) and the median number of red<br />

blood cell units transfused was 8 versus 17 (p=0.0005)<br />

in the PBSC group and in the BM group, respectively.<br />

Similar results have been reported by Russel et al.: 52<br />

duration of aplasia for both neutrophils (p=0.0002) and<br />

platelets (p=0.0003) was significantly reduced in<br />

patients receiving PBSC compared to BM recipients.<br />

Interestingly, the advantage of PBSC was also maintained<br />

if methotrexate was used as GVHD prophylaxis.<br />

A recent report by Rosenfeld et al. 57 evaluated 19<br />

patients transplanted with PBSC. No growth-factor was<br />

employed in the post-transplant phase. Significantly<br />

faster neutrophil recovery was observed in PBSC transplanted<br />

patients compared to historical control group<br />

transplanted with BM (p=0.01). However, the difference<br />

was not significant when the PBSC group was compared<br />

to BM recipients given G-CSF in the post-transplant<br />

phase.<br />

More recently, a prospective non-randomized study<br />

was carried out by the M.D. Anderson group. 39 The study<br />

included 74 adults transplanted with HLA-matched<br />

related donors. Thiotepa, busulfan and cyclophosphamide<br />

were employed as preparative regimen. The<br />

patients were divided into 3 cohorts: Group 1 received<br />

BMT using CsA and MTX as GVHD prophylaxis, Group 2<br />

received marrow using CsA and MP, and Group 3<br />

received PBSC with CsA and MP. All patients were given<br />

G-CSF post-transplant. Median time to neutrophils<br />

> 0.5×10 9 /L was 17, 9 and 10 days, and to platelets<br />

>20×10 9 /L was 32, 25 and 18 days in Groups 1, 2 and<br />

3, respectively. The use of CsA and MP for GVHD prophylaxis,<br />

rather than the source of engrafted cells was<br />

shown to be the most important factor for rapid neutrophil<br />

and platelet recovery. Provided that CsA/MP was<br />

used for GVHD prophylaxis, platelet transfusion requirement<br />

was found to be significantly lower in PBSC than<br />

in BMT recipients (p=0.04). Significant differences concerning<br />

regimen-related toxicity were seen for grade 2-<br />

4 stomatitis only between the BMT group using MTX in<br />

GVHD prophylaxis and the PBSC group using MP.<br />

Correlation between engraftment kinetics and quantity<br />

of PB cells infused is still an open question. The<br />

absolute number of nucleated PB cells or CD34 + cells did<br />

not correlate with time to neutrophils > 0.5×10 9 /L or<br />

with time to platelets > 20, > 50 or > 100×10 9 /L in a<br />

study by Rosenfeld et al. 57 Similarly, Urbano-Ispizua et<br />

al. did not find any correlation using several cut-off values<br />

of CD34 + cells at 2.5, 3, 4, 5.5 and 7×10 6 /kg. In contrast,<br />

Roy et al. 58 reported a correlation between CD34 +<br />

cells infused and engraftment using a mobilization regimen<br />

with G-CSF at a dose of 5 µg/kg. In a large series<br />

published by the M.D. Anderson group, 37 in univariate<br />

analysis of patients not given MTX prophylaxis the number<br />

of total nucleated cells infused positively affected<br />

ANC recovery. Moreover, platelet recovery was positively<br />

influenced by the number of CD34 + cells, as well as by<br />

young age and sex mis-matching.<br />

Immune reconstitution after transplantation<br />

of peripheral blood stem cells<br />

Patients undergoing allogeneic BMT experience a prolonged<br />

period of profound cellular and humoral immunodeficiency,<br />

mainly due to complete pre-transplant<br />

destruction of the host lymphohemopoietic system, the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

73<br />

use of immunosuppressive drugs for GVHD prophylaxis<br />

and the development of GVHD. 59-61 This immunodeficiency<br />

lasts until stem cells and mature lymphocytes<br />

contained in the transplanted marrow repopulate and<br />

reconstruct the hematopoietic and lymphopoietic systems<br />

which had been destroyed by the pre-transplant<br />

conditioning regimen. In particular, immunological<br />

reconstitution after BMT is considered to be dependent<br />

on two distinct phenomena. 59,60 In the early post-transplant<br />

period, there is an expansion of mature donorderived<br />

lymphocytes transferred with the graft, a<br />

process influenced by both the recipient’s environment<br />

and the cytokine storm 62 related to the transplant procedure.<br />

Thereafter, naive lymphocytes derived from the<br />

differentiation of donor hematopoietic stem cells colonize<br />

the lymphoid organs of the recipient and sustain<br />

the late immune response.<br />

The crucial role of the first step in immunological<br />

recovery is demonstrated by the observations that<br />

patients receiving a T-cell depleted transplant are at<br />

particular risk for infections and that patients transplanted<br />

using donors either recently vaccinated against<br />

or immune to a certain pathogen usually have a more<br />

rapid recovery of specific T-cell response than ones who<br />

received bone marrow from unprimed donors. 63-65 Formal<br />

proof of the contribution of transferred donorderived<br />

lymphocytes to recipient immune reconstitution<br />

has been recently reported. 62 In fact, using the combination<br />

of a cell culture method and a PCR amplified<br />

technique to study tetanus toxoid (TT)-specific T-cells<br />

clones, it was possible to demonstrate that patients after<br />

BMT display a small response that can be accounted for<br />

by a few donor-derived clones and that the T-cell clones<br />

transferred with the transplant were still detectable<br />

within the donor polyclonal T-cell lines for up to at least<br />

5 years after BMT. Moreover, the vaccination of donors<br />

with TT before BMT resulted in a more relevant transfer<br />

of antigen-experienced T-cells. 66<br />

The expansion of mature donor-derived lymphocytes<br />

transferred with the graft in recipients of peripheral<br />

blood stem cell (PBSC) transplantation could be expected<br />

to be more efficient than patients given BMT, in view<br />

of the higher number of donor lymphocytes transferred.<br />

However, at present, few reports specifically addressing<br />

the question of immune recovery after transplantation<br />

of PBSC are available.<br />

Ottinger et al. 67 demonstrated that, compared to BMT<br />

recipients, patients who were given a PBSC transplant<br />

had a more rapid recovery of both naive and memory<br />

CD4 + cells (expressing the RA and RO isoforms of the<br />

CD45 molecule, respectively) whose counts significantly<br />

exceeded those observed following marrow transplantation.<br />

This determined that in patients receiving<br />

PBSC transplantation the characteristic inversion of the<br />

CD4 + /CD8 + ratio observed after BMT was not encountered.<br />

Furthermore, the B-cell levels and, at least for the<br />

first 2 months after transplantation, the monocyte<br />

counts were augmented. Since monocytes of granulocyte<br />

colony-stimulating factor (G-CSF)-mobilized donors<br />

have been demonstrated to reduce the responsiveness of<br />

alloantigen specific T-cells, the increase in their count<br />

could contribute to the low incidence and reduced severity<br />

of acute GVHD reported after transplantation of<br />

PBSC. 68 Moreover, it must be noted that there is a prompt<br />

recovery of the lymphocyte counts after transplant of<br />

PBSC coupled with an enhanced in vitro response of lymphocytes<br />

to aspecific polyclonal activators (phytohemagglutinin<br />

and pokeweed mitogen) and to recall<br />

antigens (TT, Candida). The most likely hypothesis for<br />

explaining this accelerated recovery of helper T cells, B<br />

lymphocytes and monocytes is that the number of lymphocytes<br />

infused for each subset is more that one magnitude<br />

higher in recipients of PBSC transplant than in<br />

patients given BMT. However, alternative mechanisms<br />

cannot be excluded.<br />

Similar results in terms of more rapid recovery of CD4 +<br />

cells have also been reported by Bacigalupo et al. 40 in<br />

adults with advanced leukemia who received high-dose<br />

chemotherapy followed by G-CSF mobilized PBSC. More<br />

recently, two additional reports have further confirmed<br />

that recipients of PBSC transplants have a faster recovery<br />

of both naive and memory helper T cells. 69,70 Moreover,<br />

one of these studies documented that patients<br />

experiencing a more rapid recovery of the lymphocyte<br />

count had a significantly better probability of survival<br />

after transplantation. 69<br />

Whether the improved immune reconstitution<br />

observed after transplantation of PBSC is associated with<br />

a lower incidence of infectious complications still<br />

remains to be documented. In one of the previously mentioned<br />

studies, 69 the actuarial risk of reactivation of<br />

human cytomegalovirus (HCMV) infection in patients<br />

given a PBSC transplant was comparable to that<br />

observed in a historical control group of BMT recipients.<br />

This could be attributed to a greater viral load infused<br />

with the graft and correlated with the very large number<br />

of nucleated cells that can harbor HCMV transfused.<br />

Nonetheless, since the use of donor-derived adoptive<br />

immune therapy has been shown to be able to cure or<br />

prevent HCMV-related interstitial pneumonia and EBVinduced<br />

lymphoproliferative disorders, 71-73 it can be<br />

hypothesized that patients given PBSC transplants, with<br />

a more efficient transfer of antigen-experienced lymphocytes,<br />

may have a reduced incidence and/or reduced<br />

severity of infectious complications. Support for this theory<br />

is provided by the study reported by Bensinger et<br />

al. 74 in which a lower number of deaths from infectious<br />

complications was observed in patients given PBSC as<br />

compared to a historical group of BMT recipients.<br />

Acute and chronic GVHD<br />

PBSC collections contain a large number of T-cells –<br />

approximately 10 times more than unmanipulated marrow<br />

grafts. 75 Therefore concern for increased incidence<br />

and severity of GVHD after their infusion into an allogeneic<br />

host has been and still is a major issue after PBSC<br />

transplantation. Here we analyze the results reported<br />

so far in the most recent peer-reviewed studies pub-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


74<br />

W. Arcese et al.<br />

lished. Because of the relatively short follow-up of these<br />

studies, the assessment of chronic GVHD (cGVHD) is less<br />

complete and less accurate than that of the acute form.<br />

Some of the studies in fact do not address the problem<br />

of cGVHD. Acute GVHD, on the other hand, can now be<br />

evaluated in a rather significant number of patients. We<br />

shall look first at the characteristics of the studies, then<br />

analyze acute and cGVHD separately and finally make<br />

comparisons between marrow and PB blood transplants.<br />

A set of tentative comments will be made at the end of<br />

the chapter.<br />

Type of studies. Selected studies of PBSC transplantation<br />

for hematological malignancies are reported in<br />

Tables 2 and 3. Several of them have been analyzed in<br />

the section on hematological recovery. Table 2 focuses<br />

on the main demographic characteristics, while Table 3<br />

gives details of the transplant procedure and results of<br />

acute and chronic GVHD where applicable. Six studies<br />

are from a single institution, 39,40,55,75-77 while three are<br />

from several centers; 50,52,54 one study from the EBMT<br />

Group, 50 multicentric in nature, also reports several<br />

patients included in four of the other studies – a typical<br />

example of double reporting – so that its results<br />

reinforce what has already been observed. The figures<br />

from this last study were not calculated in any further<br />

statistical analysis in order to avoid the error of counting<br />

some of the patients twice. However, they are useful<br />

for comparisons and have been left in the tables. The<br />

total number of patients is 212. None of the studies is<br />

prospective or randomized, but four 52,55,75,77 compare the<br />

results of PBSC with those of marrow, although using<br />

different methods. We shall have to wait some time<br />

before seeing the results of the two prospective randomized<br />

studies comparing marrow and PBSC transplantation<br />

which are now in progress in Europe and the<br />

US; for the moment, the reports analyzed here represent<br />

the best we have. The 8 studies took place recently,<br />

between late 1993 and 1995, and mostly dealt with<br />

adults (median age 38 yrs, with a range from 1-57), but<br />

some included pediatric patients. Transplants were from<br />

fully HLA-identical siblings in 96% of the cases, but a<br />

minority received cells from family donors mismatched<br />

for one HLA antigen; a minority of patients (5 to 10%)<br />

also received a second allo transplant, usually from the<br />

original sibling who had donated the marrow. Patients<br />

showed a typical spectrum of hematological malignancies<br />

for which transplant is indicated. The majority<br />

(median 83%) were in advanced phases of their diseases,<br />

although definitions are quite variable with the<br />

term high-risk being used as a synonym for advanced<br />

phase, but 17% had early phase or low-risk disease at<br />

the time of transplant. These proportions differed widely<br />

within studies, some including 100% advanced diseases<br />

and others only 60%, with many more early phase<br />

patients. Pre-transplant regimens were obviously different,<br />

but despite their apparent disparities they can be<br />

grouped into those based on busulphan (54%) or TBI<br />

(31%). Only two studies differ considerably from the<br />

rest of the series: the Genoa group 40 purposely employed<br />

a low intensity regimen based on thiotepa and<br />

cyclophosphamide to reduce toxicity in a rather old<br />

patient population. The MD Anderson Hospital, 39,55 on<br />

the other hand, used a very intensive regimen combining<br />

busulphan, thiotepa and cyclophosphamide in a pop-<br />

Table 2. Main characteristics of the studies reviewed.<br />

Ref Type Of study Period of study Median N° pts Median 2nd transplants Hla family Phase of the disease<br />

Follow-up days (range) age yrs (range) mismatches<br />

74 SC 12/93-11/95 nr 37 38 (20-52) NO NO Advanced 100%<br />

52 MC 5/93-6/95 nr 26 40 (1-54) 3 (11%) 6 (23%) High risk 23 (88%)<br />

Standard risk 3 (12%)<br />

40 SC nr 136 (6-228) 31 44 (19-55) NO 3 (10%) Advanced 28 (90%)<br />

Early 3 (10%)<br />

55 SC nr 270 (180-600) 25 43 (17-57) 1 (4%) NO Relapse 21 (84%)<br />

Remission 4 (16%)<br />

76 MC 3/94-7/96 nr 24* 37 (16-57) 1 (4%) 1 (4%) Early 10 (42%)<br />

Advanced 14 (58%)<br />

54 SC 1/94-4/95 nr 33 36 (12-53) 8 (24%) NO Early 12 (36%)<br />

Advanced 21 (64%)<br />

39 SC 32 months nr 19 Not detailed 1 (5%) NO Early 18%<br />

Advanced 82%<br />

77 SC 3/94-4/95 111 (15-402) 17 33 (16-52) NO NO Early 6 (35%)<br />

Advanced 11 (65%)<br />

50 MC 1994 nr 51° 39 (2-54) NO NO Early 15 (25%)<br />

Advanced 44 (75%)<br />

LEGEND: SC = single center; MC = multicenter; n.r. = not reported; *includes 1 pt with SAA; °includes patients from studies #40, 76 and 54.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

75<br />

Table 3. Transplant modalities, aGVHD and cGVHD.<br />

Ref Fresh or Conditioning G-CSF GVHD Acute GVHD Chronic GVHD<br />

cryo- regimen* post TX prophylaxis n. grade grade GVHD related/ n. limited extensive<br />

preserved cells N° pts N° pts evaluable II-IV III-IV total deaths evaluable<br />

74 F TBI 32 no CsA/MTX 19 35 13 (37%) 5 (14%) 1/15 17 3 (18%) 4 (24%)<br />

Bus 5 CsA/PDN° 18<br />

52 C TBI 18 no CsA/MTX 26 nr 37% nr nr nr 53% overall<br />

40 F Thio-CTX 31 @ no CsA/MTX 31 31 17 (55%) 4 (13%) 4/12 28 15 (53%) 7 (25%)<br />

55 C Bus 25 yes CsA/PDN 25 25 11 (42%) 6 (22%) 3/7 nr nr nr<br />

76 F^ Bus 22 no Csa/MTX 23 22 10 (45%) 2 (9%) 0/7 16 1 (5%) 9 (50%)<br />

Other 3 CsA 1<br />

54 F 21 TBI 17 yes CsA 2<br />

C 1 Bus 16 (11 pts) CsA/MTX 22 32 11 (34%) 7 (22%) nr 11 4 (36% overall)<br />

CsA/PDN 9<br />

39 C Bus 19 yes FK-506/PDN 19 nr 22% 11% nr nr nr nr<br />

77 F Bus 17 no CsA/MTX 17 10 3 (33%) 4 (24%) 4/4 10 3 (33%) 0<br />

50 F 49 TBI 22 CsA/PDN 7<br />

Bus 22 yes CsA 6 57 30 (50%) 14 (23%) 7/29 49 17 (35%) 13 (26%)<br />

C 10 Thiotepa 11 (14 pts) Other 9<br />

Other 4<br />

*Conditioning regimen mainly based on; @ Regimen not including TBI or busulphan; CTX = cyclophosphamide; ^cells infused over 2 days: apheresis of day 1 stored at 4°C until infusion;<br />

°PDN=prednisone; n.r.=not reported<br />

ulation of similar age. Another difference is represented<br />

by the processing of the collected PBSC: in 130 cases<br />

(61%) they were infused fresh and in 82 cases (39%)<br />

they were cryopreserved instead until infusion. Finally,<br />

GVHD prophylaxis was not uniform: it was based on a<br />

combination of CsA and short-course methotrexate in<br />

130 (62%) of the cases, and on CsA plus prednisone in<br />

42 cases (20%); only one study 38 reports 19 patients<br />

(9%) who received a combination of tacrolimus and<br />

prednisone. CsA alone was used in three patients (1.4%).<br />

Acute GVHD. The incidence of aGVHD, grade II to IV,<br />

was about 40% on average. The Genoa study 40 reported<br />

a 55% incidence, but it also included the oldest patients<br />

in the series; the lowest incidence, 22%, was reported<br />

in the series from the MD Anderson Hospital where<br />

tacrolimus was used for GVHD prophylaxis. 39 The incidence<br />

of severe aGHVD, i.e. grade III and IV, was on average<br />

16% (range 11-24%). An interesting point is the<br />

fact that while most studies showed a direct correlation<br />

between the overall incidence of GVHD and severe<br />

GVHD – the latter being about half of the former – others<br />

did not. Two studies from Italy 40,76 which reported<br />

an overall incidence of over 50% also showed a low<br />

incidence of severe GVHD, which means that grade II<br />

accounted for the majority of the cases. No correlation<br />

could be made from the existing data between the variables<br />

known to influence aGVHD 78 and the results either<br />

within the single studies as discussed by their authors<br />

or by combining data as in this review. Of interest, on<br />

the specific issue of PBSC, no correlation was found with<br />

the number of T-lymphocytes infused or with the use of<br />

fresh or cryopreserved cells. However, it should be noted<br />

that the highest incidences of severe aGVHD (22-<br />

24%) were reported when prophylaxis was based on<br />

CsA/prednisone, 55 which is perhaps less effective than<br />

CsA/MTX or in the Spanish study which reported data<br />

from multiple institutions 54 with a good proportion of<br />

patients receiving CsA/prednisone for GVHD prophylaxis.<br />

Nevertheless, a high incidence was also reported in a<br />

study from Brazil where CsA/MTX was used in all cases.<br />

77 Mortality from aGVHD is not reported in all studies,<br />

as shown in Table 3; those giving the causes of death<br />

often do not mention, when infection was the main<br />

cause, whether GHVD was associated. However, considering<br />

the causes of death of 47 events analyzable in<br />

detail, GVHD was the main cause in 12 (25%). Incidentally,<br />

this figure is higher than the overall incidence of<br />

grade III-IV GVHD, but data on death are reported for<br />

fewer patients than data on GVHD.<br />

Chronic GVHD. The number of patients analyzable for<br />

cGHVD is smaller than for aGVHD; survival > 90, 100 or<br />

150 days is the requisite for evaluation. In addition,<br />

some studies give many details on cGVHD while others<br />

do not address the issue 55 or mention it very briefly. 39,52,54<br />

haematologica vol. 85(suppl. to n. 12):December 2000


76<br />

W. Arcese et al.<br />

We calculate that slightly more than 110 patients are<br />

evaluable. The overall incidence ranged from 36% to<br />

78%; considering the four studies which give detailed<br />

information, the extensive form of the disease occurred<br />

with nearly the same frequency or less than the limited<br />

form in three studies, 40,56,77 while one series reported a<br />

striking incidence of the extensive form, with the limited<br />

one being only minimally represented. 76 In that study<br />

cGVHD developed de novo in 5 out of 10 patients, at<br />

variance with the low incidence of aGVHD observed earlier.<br />

Another interesting observation is contained in a<br />

study from Seattle: 56 of 10 patients at risk, who had<br />

received CsA/prednisone for prophylaxis, 6 developed<br />

cGVHD, while only 1 of 7 given CsA/MTX did so.<br />

Comparisons between marrow and PBSC transplantation.<br />

Four studies 52,55,56,77 compared the incidence of GVHD<br />

after PBSC or marrow transplantation. These studies were<br />

carried out using matched-pair analysis with a historical<br />

control group of marrow recipients who were matched<br />

for diagnosis, disease and disease phase at transplant,<br />

age, GVHD prophylaxis 56 or age and disease status. 52 One<br />

study does not give the characteristics of the marrow<br />

recipients. 77 The Seattle study found a lower incidence of<br />

grade II-IV aGVHD for PBSC recipients – 37% vs 56% –<br />

severe GVHD (grade III-IV) was even more impressively<br />

lower in PBSC recipients, 14% vs 33% of marrow transplants.<br />

However, due to the small number of patients<br />

these data are not statistically significant. The overall<br />

incidence of cGVHD was similar in the two groups, with<br />

a tendency toward more severity in the PBSC than in the<br />

marrow group (42% vs 26% for any grade of clinical<br />

cGVHD), but again this was not statistically significant.<br />

The striking effect of MTX in the GVHD prophylaxis regimen<br />

observed in this study has already been mentioned.<br />

The multicenter Canadian study 52 reported a higher incidence<br />

of both aGVHD and cGVHD for the PBSC group<br />

than for the marrow recipients: 37% vs 21% grades II-<br />

IV aGVHD and 53% vs 48% for cGVHD, respectively. A<br />

different kind of comparison can be made in a study from<br />

the MD Anderson Hospital, 55 where patients with<br />

advanced hematological malignancies received, over a 3-<br />

year period, the same conditioning protocol but different<br />

forms of GVHD prophylaxis and different sources of allogeneic<br />

stem cells. The PBSC patients could be compared<br />

to the marrow group who received CsA/prednisone as<br />

GVHD prophylaxis: severe aGVHD was slightly less in the<br />

PBSC group, 22% vs 33%, but this difference was not statistically<br />

significant. No data on cGHVD were provided.<br />

The Brazilian study 77 reported more aGVHD in the PBSC<br />

than in the marrow group, grade III-IV 4/17 vs 3/21, but<br />

again this was not statistically significant. No comparative<br />

data on cGVHD were given.<br />

Comments. The fear of an unacceptably high rate of<br />

severe and perhaps uncontrollable acute GVHD after allo<br />

PBSC can now be allayed with a certain degree of confidence<br />

on the basis of the data analyzed here. In a population<br />

of adults with mainly advanced hematological<br />

malignancies who sometimes received second transplants<br />

or not fully HLA-identical grafts and, most important,<br />

were often not given the best GVHD prophylaxis<br />

available, acute GVHD was no more than what is expected<br />

with marrow transplants. Similar conclusions had<br />

been reached in an earlier review on the subject, 56<br />

although on a much smaller number of patients. It is<br />

more difficult to say whether GVHD is slightly more<br />

severe than after conventional marrow grafting, considering<br />

the wide variation in its occurrence, due to the<br />

multitude of factors which influence it. A broad comparison<br />

of the published data indicate that GVHD<br />

observed after PBSC is higher than in the best marrow<br />

series 79 but not worse than what is described in the large<br />

reports from registries. 80 Four studies have attempted a<br />

comparison with retrospective marrow transplants and<br />

none found significantly increased aGVHD incidence or<br />

severity. It is interesting to speculate why after infusion<br />

of 1 log more T-lymphocytes as compared to marrow<br />

aGVHD is not increased. One explanation is that any<br />

number of T-cells, once the 10 5 /kg threshold has been<br />

surpassed, 81 is already high enough to cause GVHD and<br />

even a 10-fold increase, with respect to the marrow,<br />

does not make any difference. Perhaps an extraordinarily<br />

large number of T-cells infused, that is usually not<br />

reached after G-CSF mobilization, for example 3 or 4<br />

logs, could be the next threshold above which more<br />

severe GVHD would regularly occur. Data from a study<br />

in Seattle, where 1500×10 6 /kg donor buffy-coat<br />

mononuclear cells were intentionally infused soon after<br />

BMT in patients with advanced disease to enhance a<br />

graft- versus-leukemia effect did show that acute, severe<br />

GVHD was indeed increased; in that study, unfortunately,<br />

GVHD prevention was based on MTX only so comparisons<br />

with today’s practices are not possible. 82 Other<br />

explanations for why aGVHD does not increase relate to<br />

the possible biological modifications of lymphokine production<br />

induced by G-CSF. For example, in mice G-CSF<br />

has been demonstrated to induce a polarization of T-<br />

lymphocytes towards the production of type-2 cytokines<br />

(namely IL-4 and IL-10), which display an anti-inflammatory<br />

effect. Such polarization was shown to be longlasting<br />

and was associated with a significant reduction<br />

in the severity of GVHD after transplantation of these<br />

cells into allogeneic mice recipients. 83 These results do<br />

not seem to be attributable to a direct effect of G-CSF<br />

on T-cells, since this subset of lymphocytes rarely<br />

expresses G-CSF receptors. Instead, these findings could<br />

be explained by the anti-inflammatory effects of G-CSF;<br />

in fact, administration of G-CSF decreases tumor necrosis<br />

factor (TNF) secretion. 84 Moreover, in normal subjects<br />

G-CSF is able to increase the production of two important<br />

cytokine antagonists such as soluble TNF-receptor<br />

and IL-1 receptor antagonists. 85<br />

Finally, leukapheresis products from G-CSF-mobilized<br />

donors contain a large number of monocytes. These cells<br />

have been demonstrated to significantly reduce the<br />

alloantigen specific proliferative response of T-lymphocytes.<br />

67 Also, monocytes from subjects treated with G-CSF<br />

or GM-CSF can induce the apoptosis of T-lymphocytes via<br />

the interaction of the FAS molecule with its ligand. 86 The<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

77<br />

use of G-CSF may inhibit the function of monocytes as<br />

antigen presenting cells and this, in turn, may explain<br />

the ability of this cytokine to polarize T-cells towards an<br />

anti-inflammatory cytokine profile. These findings could<br />

also contribute to explaining the unexpectedly low incidence<br />

and reduced severity of GVHD after transplantation<br />

of PBSC. However, more studies on the characterization<br />

of G-CSF mobilized lymphocytes are needed.<br />

With regard to cGHVD, it appears from this analysis<br />

that there is a trend toward a slightly increased incidence<br />

after PBSC, although not all individual studies had the<br />

same results. The clinical presentation of cGVHD was<br />

reported as peculiar in two studies, with many de novo<br />

cases 31,75 but also with a high response to treatment. 49 It<br />

should be noted that early data from the M.D. Anderson<br />

on PBSC transplants also reported an increase in cGVHD,<br />

with more liver and gastrointestinal manifestations compared<br />

to the marrow. 87 At the time of this writing, several<br />

patients were still on immunosuppressive treatment so<br />

the full magnitude of cGVHD will be appreciated only in<br />

the future after withdrawal of CsA, which is a critical<br />

time for the development of the syndrome. Furthermore,<br />

in a study on aplastic anemia 88 the infusion of donor<br />

buffy coat cells was associated with a significant increase<br />

of cGHVD. However, this increase of cGVHD was not<br />

observed in malignancies in the study. 82 Clearly a longer<br />

follow-up of a much larger number of patients is needed<br />

to answer the question of cGVHD.<br />

Transplantation of enriched allogeneic<br />

CD34 + cells<br />

As reported above, allogeneic PBSC transplantation<br />

results in the infusion of approximately 1 log more T-<br />

cells than conventional BM transplantation. Thus, in order<br />

to reduce the potential risk of severe aGVHD, several<br />

investigators have attempted to remove T-lymphocytes<br />

from allogeneic grafts. The only technique that has been<br />

utilized so far for T-cell depletion has been the positive<br />

selection of hematopoietic CD34 + cells.<br />

The CD34 antigen is present on the earliest identifiable<br />

progenitor cells and committed myeloid precursors,<br />

whereas it is not expressed on mature myeloid and B- and<br />

T-lymphoid cells. 89 However, CD34 + cells co-expressing<br />

both T-lymphoid (CD2, CD3, CD7) and B-lymphoid markers<br />

(CD19) are likely to be the early precursors of the T-<br />

and B-lymphoid lineages. 90 Preclinical studies have also<br />

shown the capacity of positive selection of CD34 + cells to<br />

eliminate 3 to 4 logs of T-cells, coupled with a substantial<br />

recovery of hematopoietic progenitors. 91,92 More<br />

recently, transplantation of autologous CD34 + cells has<br />

been proven to reconstitute normal hematopoiesis in cancer<br />

patients treated with myeloablative regimens. 93-96<br />

Based on these premises, Link et al. 97 transplanted 5<br />

patients with unmodified marrow and CD34 + selected<br />

PBSC and 5 patients with enriched marrow and PB CD34 +<br />

cells. They concluded that hematopoietic recovery was<br />

accelerated with respect to marrow allografts without<br />

an apparent increase in aGVHD following conventional<br />

CsA and MTX prophylaxis. In a subsequent study, 98 the<br />

same authors transplanted 10 individuals with positively<br />

selected circulating CD34 + cells alone. The patients<br />

were grouped according two different regimens of<br />

aGVHD prophylaxis: CsA alone or CsA and MTX. The median<br />

grades of aGVHD were 3 in group I (CsA) and 1 in<br />

group II (CsA plus MTX). Two patients in group I died from<br />

aGVHD and 2 leukemic relapses occurred in group II.<br />

Complete and stable donor hematopoiesis was shown in<br />

all patients with a median follow-up of 370 days (range<br />

45-481). It was concluded that despite a 3-log reduction<br />

of T-cells by CD34 + cell enrichment, CsA alone was not<br />

sufficient to avoid severe aGVHD.<br />

More recently, Bensinger et al. 99 transplanted 16<br />

patients with advanced hematologic malignancies with<br />

HLA-identical highly enriched PB CD34 + cells. Prophylaxis<br />

against aGVHD was CsA alone for 5 patients and CsA<br />

plus MTX for 11. A median of 8.96×10 6 CD34 + cells/kg of<br />

patient body weight were infused with a median purity<br />

of 62%. Positive selection of stem cells resulted in a<br />

median 2.8-log reduction of T-cells. Despite the prompt<br />

and sustained engraftment, 8 out of 16 patients died<br />

between 3 and 97 days post-transplant of transplantrelated<br />

causes and 1 of progressive disease. Grade 2-4<br />

aGVHD occurred in 86% of patients and 6 out of 8 evaluable<br />

patients developed clinical chronic GVHD.<br />

More promising results have been reported by Urbano-<br />

Ispizua et al. (1997), who recently transplanted 20 acute<br />

and chronic leukemia patients with allogeneic CD34 +<br />

cells. The median number of CD34 + cells and CD3 + cells<br />

infused was 2.9×10 6 /kg and 0.42×10 6 /kg, respectively.<br />

The patients were conditioned with fractionated TBI (total<br />

dose 13 Gy in 4 fractions) and cyclophosphamide 120<br />

mg/kg. Additional GVHD prophylaxis included CsA and<br />

methylprednisolone. No patients developed grade II- IV<br />

aGVHD. The overall procedure was associated with low<br />

morbidity and no transplant-related deaths occurred<br />

within the first 100 days. Although the median followup<br />

(7.5 months) is rather short for a full evaluation of<br />

cGVHD incidence and disease relapse, the absence of<br />

extensive cGVHD and the low rate of disease recurrence<br />

(only 3 out of 20 patients relapsed) encourage further<br />

studies in this direction. In comparison with previous<br />

studies, 99 it should be noted that the median age of the<br />

patient population was 40 years and only 35% of the<br />

individuals were older than 45 years. Moreover, the<br />

majority of the leukemic patients were transplanted in<br />

the early phase of their disease. Both these parameters<br />

are generally associated with lower transplant-related<br />

mortality and a lower incidence of severe GVHD.<br />

Although further studies involving larger numbers of<br />

patients are currently in progress, these results, taken<br />

together, demonstrate that infusion of CD34 + selected<br />

PBSC results in rapid and stable engraftment. However,<br />

transplantation of purified stem cells may induce a higher<br />

rate of acute and chronic GVHD than expected, thus<br />

requiring full GVHD prophylaxis. Therefore this approach<br />

for T-cell depletion should be carefully evaluated in the<br />

setting of HLA-identical PBSC transplantation and<br />

weighed against the potential increased risk of disease<br />

haematologica vol. 85(suppl. to n. 12):December 2000


78<br />

W. Arcese et al.<br />

relapse, and perhaps delayed immunological reconstitution,<br />

and the increased cost of the procedure.<br />

Allogeneic PBSC from haploidentical<br />

familial donors: the mega-stem-cell<br />

dose concept<br />

Allogeneic BMT has been largely confined to patients<br />

who are HLA-identical to their donors. At present, only<br />

about 30-35% of patients who might benefit from allogeneic<br />

BMT have an HLA-identical sibling. The establishment<br />

of large registries of HLA-typed individuals<br />

during recent years has led to a substantial increase in<br />

transplants from unrelated donors. 100-102 Although 40 to<br />

50% of patients are successful in locating HLA-A, B,<br />

DR-matched unrelated donors, many patients still fail to<br />

find an appropriate donor. 103,104 In contrast, nearly all<br />

patients have an HLA-haploidentical relative (parent,<br />

child, sibling,) who could serve as a donor.<br />

The feasibility and safety of transplants from partially<br />

matched family members have been investigated and<br />

the results of these studies have demonstrated that HLA<br />

matching is a critical and limiting factor in marrow<br />

transplantation. 105-107 In published works on mismatched<br />

transplants, there has been no large study involving<br />

patients mismatched with their donors by one full haplotype.<br />

These experiences have been limited because the<br />

problems of transplant increase with the number of<br />

antigenic disparities between donor and host. 106 In 2- or<br />

3-antigen mismatched transplants, studies by the Seattle<br />

program 105 and the International Bone Marrow Transplant<br />

Registry 108 reported graft failure in 20 to 30% of<br />

cases. The reported incidence of acute GvHD (grade II or<br />

greater) varied from 34% to 100% overall, but in 2- and<br />

3-antigen mismatched patients the incidence was at<br />

least 80%. 105,106 Severe GvHD was a greater problem<br />

than graft rejection, preventing more widespread use of<br />

mismatched related transplants during the latter 1970s<br />

and 1980s.<br />

By contrast, extensive experience in severe combined<br />

immunodeficiency (SCID) patients has shown that GVHD<br />

is largely preventable, even in 3-antigen mismatched<br />

transplants, when a 3-log T-cell depletion of the donor<br />

bone marrow is achieved. 109,110 In 1981 Reisner et al.<br />

reported the first case of leukemia treated with a T-cell<br />

depleted marrow transplant from a haploidentical, 3-<br />

loci incompatible, parental donor. 111 There was full<br />

engraftment and no GVHD. Subsequently, clinical trials<br />

in mismatched-sibling BMT for patients with leukemia<br />

were begun using the lectin or other T-cell depleting<br />

methods which included monoclonal antibodies with<br />

complement or conjugated to toxins, and counterflow<br />

centrifugal elutriation (reviewed in ref. #112). It was<br />

determined that the threshold dose below which GVHD<br />

was not seen in matched patients was 2.0×10 5 T<br />

cells/kg. 113 However, early enthusiasm for all methods<br />

was soon tempered by an increased incidence (> 50%)<br />

of graft rejection. 114<br />

In exploring the problem of failure in mismatched<br />

grafts, the inadequacy of immunosuppression was documented<br />

by the observation of residual host lymphocytes<br />

in patients who failed to engraft after being conditioned<br />

with conventional preparative regimens and<br />

given T-cell depleted mismatched transplants. 114 Work<br />

in esperimental models has shown that incompatible T-<br />

cell depleted transplants can be successfully performed<br />

by manipulating the conditioning regimen and/or the<br />

graft composition. 115 The immunologic response of the<br />

remaining host immune system against the graft can<br />

be overcome by increasing the total dose of TBI 116 or by<br />

adding selective anti-T measures with minimal toxicity,<br />

such as splenic irradiation 117 or in vivo treatment with<br />

anti-T monoclonal antibodies. 118 Engraftment is also<br />

improved by increasing the myeloablative effect of the<br />

conditioning regimen through the use of dimethylmyleran,<br />

busulfan or thiotepa, given with TBI. 119,120<br />

Different cytoreductive agents or radiation regimens<br />

were therefore added to the basic conditioning protocols<br />

used for conventional BMT. Although a marked beneficial<br />

effect was found in recipients of T-cell depleted<br />

HLA-identical bone marrow upon adding ATG and<br />

thiotepa to TBI and cyclophosphamide, 121 none of these<br />

agents were found to be useful in recipients of T-cell<br />

depleted haploidentical 3-antigen incompatible transplants.<br />

Others, using the monoclonal antibody Campath-<br />

1G instead of ATG, have observed similarly disappointing<br />

rejection rates. 122<br />

Concerning the composition of the graft, Lapidot et al.<br />

showed that megadoses of T-cell depleted incompatible<br />

bone marrow inoculum could obtain full donor-type<br />

engraftment in mice treated with sublethal irradiation,<br />

or presensitized with donor lymphocytes or partially<br />

reconstituted before the transplant by adding back a<br />

controlled number of host-type mature thymocytes. 123<br />

The means of overcoming graft failure elucidated in<br />

the experimental model can be applied in the clinical setting<br />

by combining approaches that increase both the<br />

conditioning of the host and the size of the stem cell<br />

inoculum. The major advance that finally made full haplotype-mismatched<br />

transplantation possible in leukemia<br />

patients was the availability of rhG-CSF 124 and the experience<br />

in autologous transplants in which G-CSF was used<br />

to mobilize high numbers of stem cells into the blood of<br />

patients without significant side effects. 125 Their employment<br />

made it feasible to increase the number of donor<br />

stem cells to a level which, in animal models, made transplantation<br />

across the histocompatibility barrier possible.<br />

117 On the basis of these concepts, the BMT team at<br />

the University of Perugia first introduced the megadose<br />

cell transplant in full haplotype-mismatched leukemia<br />

patients. 126 After a conditioning regimen which included<br />

8 Gy TBI in a single fraction at a fast dose rate (16 cGy/m),<br />

thiotepa (10 mg/kg), rabbit ATG (20 mg/kg in 4 days) and<br />

cyclophosphamide (100 mg/kg in 2 days), advanced<br />

leukemia patients, mostly adults, were given the combination<br />

of marrow and G-CSF-mobilized blood stem cells.<br />

Donors compatible with the patients for only one haplotype<br />

and 3-antigen disparate on the other haplotype<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

79<br />

underwent bone marrow harvest followed within a few<br />

days by treatment with G-CSF (12 µg/kg/d × 7 days). Four<br />

leukaphereses of progenitor cells were performed starting<br />

on the fourth day. The marrow as well as the leukapheresis<br />

product were each depleted of T-cells using soybean<br />

lectin agglutination and E-rosetting. 127 Both the<br />

CD34 + cells and CFU-GM were increased 7- to 10-fold<br />

over bone marrow alone, and the average number of CD3 +<br />

cells infused was 2.2×10 5 /kg recipient body weight. Following<br />

conditioning and stem cell infusion, patients<br />

received no additional GvHD prophylaxis. The results of<br />

the first 17 patients were reported in 1994 126,128 and subsequently<br />

27 additional leukemia patients, most of them<br />

in chemoresistant relapse at the time of transplant, were<br />

treated. For the first time a very rapid hematopoietic<br />

engraftment was observed in more than 80% of patients<br />

and, without any post-transplant prophylaxis, acute<br />

GVHD occurred in only 27%, and there was no significant<br />

chronic GvHD. As for survival, 7 patients are currently<br />

alive and disease free at a median follow-up of more than<br />

3 years. The major complications observed in this pilot<br />

study were interstitial pneumonitis, which occurred in<br />

43%, and infections in the setting of GvHD. Both were<br />

responsible for the 60% transplant-related mortality.<br />

This pilot experience showed that the megadose cell<br />

strategy, together with a highly immunosuppressive and<br />

myeloablative conditioning, resulted in a high incidence<br />

of durable engraftment with significantly reduced GvHD<br />

comparable to historical experience with unmanipulated<br />

transplants. It also confirmed that in humans, as in<br />

mice, the stem cell dose plays a crucial role in overcoming<br />

HLA-histocompatibility barriers. 129 This concept<br />

is also supported by the recent work by Rachamin et<br />

al. 130 demonstrating that purified CD34 + cells have a<br />

very powerful veto activity. They are able to specifically<br />

reduce, in a mixed lymphocyte culture, the frequency<br />

of CTL precursors against the stimulatory cells of the<br />

same subject and thereby help to overcome allogeneic<br />

rejection and enhance their own engraftment.<br />

The approach to haplotype-mismatched transplants<br />

has evolved since Aversa et al. 126,128 originally proposed<br />

the megadose cell concept. With their initial protocol<br />

GvHD was decreased but not eliminated, and it contributed<br />

to transplant-related mortality, which was significantly<br />

greater than in matched patients receiving<br />

similar conditioning (Aversa et al., unpublished data).<br />

These remaining problems were addressed in a subsequent<br />

trial, where it was possible to completely abrogate<br />

GVHD by improving the T-cell depletion method. 131 By<br />

processing the peripheral blood progenitor cells with an<br />

initial debulking of both mononuclear and T-cells with<br />

one-round E-rosetting followed by positive selection of<br />

CD34 + cells with the Ceprate stem cell concentrator<br />

(CellPro Inc. Bothell, WA, USA), it was possible to infuse<br />

a median of 3×10 4 CD3 + cells/kg and 13×10 6 CD34 +<br />

cells/kg in 24 high-risk leukemia patients. Conditioning-related<br />

toxicity was also reduced by modifying pretransplant<br />

chemotherapy. As a substitute for cyclophosphamide,<br />

which was considered a possible factor in<br />

the early mortality in the first pilot study, fludarabine<br />

was tested. In fact, it had been shown to have powerful<br />

immunosuppressive effect in patients treated for<br />

lymphoproliferative disorders, even at doses which were<br />

not associated with significant extra-hematologic toxicity.<br />

132 Furthermore, in a mouse model TBI+fludarabine<br />

(40 mg/m 2 /d × 5) was shown to provide an immunosuppressive<br />

effect comparable to TBI+cyclophosphamide.<br />

133<br />

At present, a regimen including TBI in a single fraction,<br />

thiotepa, fludarabine and ATG followed by the infusion<br />

of T-depleted bone marrow plus T-depleted CD34-<br />

selected blood cells is being evaluated for toxicity and<br />

efficacy. The preliminary results of this study were<br />

recently presented at the American Society of Hematology<br />

meeting in Orlando. 134,135 As hoped, with the<br />

decrease in the number of T-cells infused and the modifications<br />

in conditioning, the problem of GvHD was<br />

largely prevented (only 2 patients developed grade II<br />

acute GvHD and one progressed to chronic GvHD); the<br />

engraftment rate was 95% and there was a decrease in<br />

transplant-related mortality to 29% compared to the<br />

previous 60%.<br />

A more recent update on 48 patients was presented<br />

in Mannheim. 136 The abstract reports that 22/28 patients<br />

were in chemoresistant relapse at the time of transplant;<br />

age ranged from 4 to 53 years (median 27). Forty<br />

of 48 patients engrafted, grade II-IV acute GvHD<br />

occurred in only two patients and no one developed<br />

chronic GvHD. Twenty patients were alive and disease<br />

free at a median follow-up of 5 months (range 1-16).<br />

There were 11 relapses and 17 nonhematologic deaths.<br />

Transplant-related mortality was 35%.<br />

An unsolved problem remains the slow immunologic<br />

recovery of engrafted patients that is responsible for<br />

infections. Counting of peripheral blood lymphocytes<br />

which exhibited a phenotype of NK cells (CD56 + ), helper<br />

T cells (CD3 + /CD4 + ), cytotoxic T-cells (CD3 + /CD4 + ), cytotoxic<br />

T-cells (CD3 + /CD8 + ) and B-cells (IgM + ) revealed early<br />

recovery (within 2-4 weeks) of NK cells and extremely<br />

delayed recovery of T cells. In particular, CD4 + cells<br />

reached near normal values after 10 to 12 months. 137 In<br />

addition, the frequencies of T-cells responding to polyclonal<br />

activators in a sensitive limiting dilution assay<br />

were approximately 1 in 100 within the first post-grafting<br />

month and 1 in 10 at 10 months post-transplant<br />

(control responder cell frequencies are in the range of 1<br />

in 2). 137 The low number of T-cells, combined with their<br />

functional peculiarities (i.e. failure to respond to TcR<br />

stimulation) are certainly implicated in the high frequency<br />

of infectious complications and are strongly<br />

indicative of a markedly distorted T-cell maturational<br />

process.<br />

Interestingly, looking at post-transplant immune<br />

reconstitution, Albi et al. observed a large donor-type<br />

TcR- + CD8 + cell population that co-express NK-like<br />

receptors for specific MHC class I alleles. 137 NK cells<br />

expressed multiple, clonotypically distributed membrane<br />

receptors with different specificities for families of MHC<br />

haematologica vol. 85(suppl. to n. 12):December 2000


80<br />

W. Arcese et al.<br />

class I alleles (termed killer cell inhibitory receptors, KIR).<br />

The interaction between these receptors and the appropriate<br />

alleles produces a signal which inhibits killing of<br />

the target cells. Analysis of more than 900 clones<br />

revealed that 40% to 80% of these KIR + T-cells exhibit<br />

NK-like functions, i.e. they were able to lyse class I-negative<br />

targets and were functionally blocked by the<br />

expression of specific class I alleles on target cells. Furthermore,<br />

these cells do not lyse autologous hemopoietic<br />

cells, but are able to lyse fresh leukemic cells. 137<br />

This might suggest that they could provide a graft-versus-leukemia<br />

effect without causing GVHD.<br />

In a period of twenty years transplants across the histocompatibility<br />

barrier have advanced from being experimentally<br />

to clinically possible. The principles outlined<br />

at the beginning – adequate cell dose, adequate<br />

immunosuppression and myeloablation, avoidance of<br />

GvHD – have been successfully combined. Two other<br />

groups have recently reported on successful engraftment<br />

in haplotype mismatched transplants by combining bone<br />

marrow and G-CSF-mobilized blood stem cells after<br />

CD34-positive selection for patients with advanced<br />

leukemia. 138,139 Refinements of this protocol should make<br />

haplotype mismatched transplants an attractive therapeutic<br />

option for patients with high-risk leukemia without<br />

a matched related or unrelated donor.<br />

Furthermore, there are enormous potential applications<br />

of the concept of the stem cell dose for the future<br />

treatment of non-neoplatic diseases like aplastic anemia,<br />

Fanconi’s anemia, SCID, thalassemia, and for induction<br />

of tolerance in organ transplantation. This approach<br />

should be applicable not only in mismatched transplants<br />

but also for overcoming problems which remain in the<br />

matched transplant setting, such as rejection in aplastic<br />

anemia, regimen-related toxicity in Fanconi’s anemia<br />

and thalassemia.<br />

Transplantation of allogeneic PBSC<br />

from unrelated donors<br />

Two retrospective studies have recently suggested<br />

that the number of hematopoietic cells present in BM<br />

harvest correlates with the clinical outcome in the setting<br />

of stem cell transplantation from both HLA-identical<br />

siblings and from HLA-matched unrelated donors. 9,140<br />

In the latter case, the number of cells infused has proven<br />

to be the most potent prognostic factor for survival.<br />

Therefore, given the much higher number of progenitor<br />

cells collected in primed PB as compared to conventional<br />

BM harvest, the use of PBSC appears to be a<br />

promising alternative for improving the results of transplantation<br />

from unrelated donors. In this regard, Ringden<br />

et al. 34 and Stockschlader et al. 141 recently reported<br />

their preliminary experience with transplantation of<br />

allogeneic PBSC from full-matched or 1-antigen mismatched<br />

unrelated donors. In particular, Ringden et al.<br />

transplanted 6 patients with high-risk hematologic disease.<br />

Four of them received allogeneic PBSC as primary<br />

treatment while 2 others were treated after a BM graft<br />

failure. Five PBSC collections were infused without any<br />

manipulation; in 1 case Campath-1 monoclonal antibody<br />

was used for T-cell depletion. In the German study,<br />

1 AML patient in 2 nd CR received purified CD34 + cells<br />

from an HLA-matched unrelated donor. The total number<br />

of patients transplanted is too small and the followup<br />

too short to draw any conclusion; however, these<br />

preliminary data showing a rapid rate of engraftment<br />

are encouraging, whereas the role of T-cell depletion<br />

remains to be clarified.<br />

Transplantation of umbilical cord blood<br />

progenitor cells<br />

The existence of hematopoietic progenitors circulating<br />

between the fetus and the placenta during gestation<br />

was first described in this Journal more than 20 years<br />

ago, 142 but their clinical application began only when it<br />

became evident that the progenitor cell content of CB<br />

was sufficient for bone marrow repopulation in pediatric<br />

patients given myeloablative chemo-radiotherapy. 143 In<br />

1988, a patient affected with Fanconi’s anemia was first<br />

transplanted with CB progenitor cells from his HLAmatched<br />

healthy sibling. 4 Subsequently, successful CB<br />

transplants (CBT) were sporadically reported in patients<br />

affected by both malignant and nonmalignant disorders.<br />

5,144-147 The establishment of large CB banks in<br />

Europe and USA, and improvement of the methods of<br />

cell collection, manipulation and freezing have permitted<br />

a rapidly increasing use of CB progenitor cells, which<br />

are now extensively employed for allogeneic transplantation.<br />

148-150<br />

The biological and functional characteristics of CB<br />

hematopoietic stem cells have been already reviewed<br />

by the Working Group. 12<br />

Clinical results after cord blood<br />

transplantation<br />

As mentioned above, the use of human umbilical CB<br />

hematopoietic progenitors represents an alternative<br />

modality of transplantation. Advantages of CBT include<br />

ease of hematopoietic stem cell collection, absence of<br />

donor risks, low risk of viral contamination (cytomegalovirus,<br />

Epstein-Barr virus, etc.) and, for transplantation<br />

among unrelated individuals, prompt availability of<br />

hematopoietic stem cells. Over the past decade, placental<br />

blood has been used to transplant hundreds of<br />

patients (mainly children) and information on the rate<br />

and kinetics of engraftment and on the risk of severe<br />

acute or chronic GVHD is now available for CBT recipients<br />

from both related and unrelated donors.<br />

In the two largest cohorts of patients transplanted<br />

from an HLA-identical sibling reported to date, 7,148 the<br />

probability of engraftment of donor hematopoiesis was<br />

79% and 85%, respectively, even though it must be<br />

underlined that in the cohort analyzed by Wagner and<br />

colleagues rejections were mainly observed in patients<br />

affected by bone marrow failure syndromes or hemoglobinopathies,<br />

which are diseases with a high risk of<br />

graft failure. In the cohort reported by Wagner et al., the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

81<br />

median time to achieve granulocyte (PMN >0.5×10 9 /L)<br />

and platelet (PLT >50×10 9 /L) recovery was 22 and 49<br />

days, respectively; these values were greater than those<br />

observed with BMT. Comparable time for PMN and PLT<br />

recovery were observed in the European experience. 7 In<br />

particular, in this latter report, patients receiving a higher<br />

number of nucleated cells (i.e. more than 37×10 6 /kg)<br />

experienced faster engraftment than those given a lower<br />

number of cord blood progenitors, suggesting that<br />

the number of cells infused is the main factor influencing<br />

the rate of hematologic recovery. More prolonged<br />

periods of profound leukopenia and thrombocytopenia<br />

have also been described in children receiving CBT from<br />

unrelated donors. In fact, in the first two series of<br />

patients transplanted from an unrelated donor, 149,150<br />

PMN recovery occurred in a median of 22 and 24 days,<br />

respectively, whereas the median time for PLT recovery<br />

was 82 and 67 days, respectively. The importance of the<br />

number of cells infused on the kinetics of PMN and PLT<br />

engraftment in the Eurocord Transplant Group experience<br />

was also observed in the group of patients given<br />

an unrelated CBT. Moreover, unlike BMT, where the use<br />

of hematopoietic growth factors has been demonstrated<br />

to hasten myeloid recovery significantly, 151,152 administration<br />

of these cytokines has produced conflicting<br />

results in CBT recipients. In fact, in the cohort of patients<br />

receiving CBT from HLA-identical or disparate family<br />

donors studied by Wagner et al., the use of G-CSF or<br />

GM-CSF did not influence the kinetics of PMN reconstitution.<br />

142 In contrast, in a group of children transplanted<br />

using unrelated CB units reported by the same<br />

authors, patients receiving hematopoietic growth factors<br />

experienced faster myeloid recovery than those who<br />

were not given the cytokines. 150<br />

The delayed rate of neutrophil engraftment and the<br />

conflicting data mentioned above could be explained by<br />

the infusion of fewer progenitor cells with CBT with<br />

respect to BMT, as suggested by the European experience,<br />

or, alternatively, by the particular characteristics<br />

of the proliferative, self-renewing and differentiating<br />

capacity of CB cells. A practical consequence of the<br />

above observation is that specific attention should be<br />

paid to the risk of infectious complications in children<br />

receiving CBT.<br />

During the first few months after transplant CBT<br />

recipients show a steady, impressive increase in HbF<br />

whose values are significantly higher than those<br />

observed in patients receiving BMT. Moreover, the subsequent<br />

decline is usually less pronounced than that<br />

observed in normal children in the first year of life (Figure<br />

1). 153,154 This preferential production of chains in<br />

erythroid progenitors seems to reproduce the normal<br />

ontogeny of erythropoiesis, even though the persistence<br />

of HbF levels higher than those observed in the first year<br />

of age suggests a more delayed switch from fetal to<br />

adult hemoglobin synthesis.<br />

The dose of CB progenitor cells necessary to ensure<br />

early and sustained hematopoietic engraftment and<br />

favorable clinical outcome has still not been precisely<br />

defined. Wagner et al. 148 claimed that the lowest dose<br />

of CB nucleated cells reported to be capable of yielding<br />

complete and sustained engraftment is 1×10 7 /kg of<br />

recipient body weight. However, as previously mentioned,<br />

the Eurocord Transplant Group documented that<br />

a dose of nucleated cells available before thawing of<br />

fewer than 3.7×10 7 /kg recipient body weight was highly<br />

predictive of both graft failure and poor survival after<br />

CBT. 7 The importance of this value also emerges from<br />

Kurtzberg et al’s experience. 149 Ten out of 13 patients<br />

undergoing CBT from an unrelated donor and having<br />

Figure 1. HbF levels observed in the 5 CBT recipients in<br />

the first year after transplant. Normal percentiles of HbF<br />

values (dotted line, mean±2SD) observed in the first year<br />

of age (Galanello et al., 1981) are also reported (F.<br />

Locatelli, personal data).<br />

haematologica vol. 85(suppl. to n. 12):December 2000


82<br />

W. Arcese et al.<br />

received fewer than 3.710 7 /kg nucleated cells failed to<br />

benefit from the procedure. Although the importance<br />

of this cellular dose appears evident from these two<br />

reports, it should be noted that rarely is such a number<br />

of cells available in the case of adult patients. In fact,<br />

since the average leukocyte count in placental blood is<br />

about 10×10 6 /mL and the average volume of donated<br />

blood is about 80 mL, the average number of nucleated<br />

cells before thawing in one cord blood unit may reach<br />

800×10 6 . About 30% of nucleated cells are lost during<br />

the thawing and washing procedure, and even though<br />

the loss mostly involves mature cells which have no role<br />

in transplantation, it is reasonable to expect fewer than<br />

3.7×10 7 /kg viable cells for patients with body weight<br />

greater than 30-40 kg.<br />

The reduced immune reactivity of cord blood cells<br />

found a clinical counterpart in 38 children reported by<br />

Wagner et al. 148 who received CBT from an HLA-identical<br />

or 1-antigen mismatched sibling. In these patients,<br />

the incidence of grade II-IV acute GVHD and limited<br />

chronic GVHD was 3% and 6%, respectively, with no<br />

patient dying of GVHD. Confirmatory results were<br />

obtained by the Eurocord Transplant Group, which<br />

reported a 9% incidence of grade II-IV acute GVHD in<br />

CBT recipients from an HLA-identical relative. However,<br />

it is noteworthy that the same group documented a<br />

50% incidence of grade II-IV acute GVHD in patients<br />

transplanted from an HLA nonidentical family donor.<br />

In CBT performed between unrelated subjects with, in<br />

some cases, a disparity of 2-3 HLA antigens, the incidence<br />

of acute grade III-IV GVHD is reduced (approximately<br />

10-20%) 7,149,150 with respect to that observed<br />

after unmanipulated BMT between unrelated subjects<br />

for whom, notwithstanding complete HLA identity<br />

between recipient and donor, the observed risk of acute<br />

grade III-IV GVHD reaches at least 30-40%. In particular,<br />

in the cohort of patients given CBT from an unrelated<br />

donor reported by Kurtzberg et al., 149 no patient developed<br />

grade IV acute GVHD or experienced hepatic<br />

involvement or died of acute GVHD, and only 4 out of 65<br />

patients given an unrelated CBT reported by the Eurocord<br />

Transplant Group showed grade IV acute GVHD.<br />

From the data collected up to now, therefore, it clearly<br />

appears that CBT, from both familial and unrelated<br />

donors, is associated with a reduced risk of acute and<br />

chronic GVHD. 148-150 In view of this observation, different<br />

centers tend to adopt less intensive schemes of<br />

GVHD prophylaxis. Typically, children transplanted with<br />

a CB unit collected from an HLA-identical sibling receive<br />

GVHD prophylaxis consisting of CsA alone, whereas for<br />

patients undergoing unrelated CBT the most widely used<br />

regimens are those based on a combination of CsA with<br />

either low- or high-dose steroids. 149,150 The association<br />

of CsA with short-term methotrexate as proposed by<br />

the Seattle group in BMT recipients 155 is not generally<br />

employed due to concerns about the prolongation of<br />

time required for engraftment and possible damage to<br />

hematopoietic progenitors with a reduction in the<br />

potential for marrow repopulation. Procedures involving<br />

T-cell depletion of CB cells are also discouraged.<br />

The reported low incidence of GVHD 7,148-150 might, on<br />

the other hand, be a major drawback to the use of CB as<br />

a source of stem cells for allogeneic transplantation in<br />

leukemic patients. In fact, since the role of allogeneic<br />

lymphocytes in the control and/or eradication of malignancy<br />

is well established, the potential absence of GVL<br />

activity could represent a theoretical concern in leukemic<br />

subjects given CBT. Currently available data do not conclusively<br />

establish whether CBT really predisposes<br />

patients to an increased risk of leukemia relapse. However,<br />

considering the concern mentioned above, the<br />

choice of less intensive GVHD prophylaxis schemes could<br />

represent a possible means for partially sparing the<br />

immune-mediated GVL effect, which may significantly<br />

contribute to preventing regrowth of leukemia cells.<br />

Immunological reconstitution following<br />

cord blood transplantation<br />

Although immunological reconstitution after BMT has<br />

been extensively studied, 58,59 few data are available on<br />

the kinetics of immune recovery in CBT recipients.<br />

144,153,156 After CBT, recovery of T-cell immunity, as<br />

well as that of natural killer subpopulations, mimicks<br />

what is described in BMT recipients. 153 In particular, in<br />

the early post-transplant period recovery of CD8 + lymphocytes<br />

seems to be faster than that of CD4 + cells,<br />

determining a characteristic inversion of the ratio<br />

between the two subpopulations during the first 6<br />

months after CBT, similarly to what is described in BMT<br />

recipients. Considering the much lower number of lymphoid<br />

cells transferred with CBT as compared to BMT,<br />

the recovery of T-lymphocyte number and function<br />

towards normal must be considered rapid. The prompt<br />

recovery of T-cell immunity following CBT could be positively<br />

influenced by the reduced incidence and severity<br />

of both acute and chronic GVHD, which per se<br />

adversely affects the acquisition of lymphocyte function.<br />

However, it must be noted that the prompt recovery<br />

of lymphocyte function in vitro does not necessarily<br />

correlate with effective in vivo immunity. In fact, at<br />

present there are insufficient data to prove that this<br />

rapid T-cell recovery translates into a low incidence of<br />

viral and fungal infections after CBT.<br />

In contrast to what is observed in BMT recipients, 58,59<br />

an impressive increase in the percentage and absolute<br />

number of B-lymphocytes, apparently not related to<br />

viral infections, has been documented in children receiving<br />

CBT. 153,157 Possible hypotheses to account for this<br />

observation could involve the physiological characteristics<br />

of B-cell ontogeny in the first year of life and/or<br />

different distribution of mature memory lymphocytes in<br />

bone marrow and CB. 158,159<br />

Ethical problems of cord blood<br />

transplantation<br />

Like any other innovative treatment, CBT also poses<br />

some ethical questions that have still not been completely<br />

resolved. In particular, these ethical considera-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

83<br />

tions can be subdivided into those concerning transplants<br />

between HLA-compatible siblings and those<br />

regarding CBT from unrelated donors.<br />

The two main ethical problems regarding transplantation<br />

from a family donor are those of conceiving a sibling<br />

with the hope of producing a compatible donor for<br />

a previous child who requires transplantation of hematopoietic<br />

stem cells, and of his/her HLA typing in utero.<br />

Of course, any decision to conceive a child for the sole<br />

purpose of making it become a cord blood donor entails<br />

belittling the value of the individual to be born. However,<br />

it cannot be ignored that it is extremely difficult to<br />

separate the reasons that lead to conceiving a child solely<br />

for the joy of procreating from those linked to the<br />

possibility of saving a living, sick child. On the other<br />

hand, even this last reason does not lessen the importance<br />

of the future child who will bring happiness to the<br />

family in addition to being the person who, in the case<br />

of successful transplantation, allowed the family to save<br />

the life of a child who would have otherwise been lost. 160<br />

In the meantime, it is important to stress the inappropriateness<br />

of performing HLA typing in utero; because<br />

of the increased abortion rate due to the procedure<br />

(about 1-2%), it entails the risk of causing the death of<br />

a healthy human being and would in any case be deeply<br />

despicable if it were used to dispose of a conceived child<br />

found to be HLA-incompatible with the sick patient.<br />

From the point of view of the unborn child, HLA typing<br />

in utero quite obviously poses critical problems and offers<br />

no advantages, but only tangible risks for that unborn<br />

child’s survival. HLA typing in utero should be carried out<br />

only when other, far more important reasons (for example,<br />

advanced age of the mother with consequent higher<br />

risk of chromosome-21 trisomy for the fetus) suggest<br />

performing prenatal diagnostic procedures.<br />

Since the donor is a newborn infant, the use of cord<br />

blood for an unrelated sick patient has raised many questions<br />

of ethical interest. These ethical aspects go beyond<br />

the scope of this review, but we would like to comment<br />

briefly on some of them. Particular attention has been<br />

devoted to the problems raised by tests required to determine<br />

whether cord blood is suitable and usable without<br />

the risk of transmitting to the recipient any disease carried<br />

by the donor cells (namely infectious diseases and<br />

genetically transmissible disorders). In fact, for this specific<br />

aspect the ethical question is: what kind of behavior<br />

should be adopted by the medical operator who works<br />

with a woman (or with the parents of a child) if a disease<br />

for which there is no therapy is detected in the<br />

infant? 161 Such possibly dramatic news must cause as<br />

little damage as possible. We must by all means prevent<br />

our increasingly profound biological awareness of our<br />

selves from leading to a culture of anguish. One might<br />

recall in this regard that, for example, it has been stated<br />

that minors should not be tested for abnormal genes<br />

unless there is an effective curative or preventive treatment<br />

that must be instituted early in life. 162<br />

Another heavily debated problem regarding unrelated<br />

transplants is the case of a cord blood unit assigned<br />

to allotransplantation, making these cells unavailable<br />

in the case the donor needs them for an autologous<br />

transplant. However, to ensure that every CB donor has<br />

the right to use the donated blood for himself if necessary,<br />

there would be no way to provide cord blood units<br />

for allotransplants. Therefore the very nature of the<br />

technique originally conceived for allotransplants would<br />

be profoundly transformed, and this would punish all<br />

donation ethics at their very core.<br />

Strictly linked to these considerations is the problem<br />

of private banking of cord blood cells. 163 In any case, we<br />

firmly believe that involving money-making aspects in<br />

CB transplantation technology is unacceptable. In particular,<br />

as stated by other authors as well, 164 no part of<br />

the human body should be commercialized and CB<br />

should not be used for the benefit of financial speculators.<br />

Rational use of PBSC in the treatment<br />

of leukemic relapse after allogeneic<br />

transplant<br />

The cure rate of patients receiving an allogeneic transplant<br />

for hematological malignancies is negatively<br />

affected by relapse. The incidence and time of disease<br />

recurrence depend on several factors such as diagnosis,<br />

disease phase at the time of transplant, conditioning<br />

regimen, GVHD prophylaxis and T-cell content of the<br />

infused graft. 165<br />

The response rate and clinical outcome of relapsing<br />

patients with acute leukemia treated with chemotherapy<br />

alone are extremely poor. 166,167 Interferon therapy<br />

significantly prolongs the survival of CML patients<br />

relapsed after transplant, but its benefit is not durable<br />

over long-term follow-up. 168-170 Finally, a second transplant<br />

offers some possibilities of cure for relapsed<br />

patients but it carries high morbidity and regimen-related<br />

mortality. 171-173<br />

During the past few years the therapeutic approach to<br />

post-transplant relapse has been substantially modified.<br />

Following its first report by Kolb et al., 174 donor lymphocyte<br />

infusion (DLI) is currently being used as a form<br />

of adoptive immunotherapy for patients with hematological<br />

malignancies who relapse after transplant 175-188<br />

or develop EBV lymphoproliferative disorders. 72,189<br />

A number of observations in allogeneic transplants<br />

support the evidence that a GVL effect, whether associated<br />

or not with GvHD and mediated by donor<br />

immunocompetent cells, contributes to the eradication<br />

of the neoplastic clone. 82,190-195<br />

The rationale for the use of DLI in post-transplant<br />

relapse is based on two main factors: 1) the persistence<br />

of an immunotolerant status versus donor cells in the<br />

relapsing host; 2) the cytotoxic activity exerted by HLAunrestricted<br />

NK and LAK cells or by HLA-restricted T-<br />

cells of donor origin against host malignant cells. 188,196<br />

However, the effectiveness of DLI therapy is variable<br />

since it greatly depends on the type of disease and its<br />

stage at the time of relapse. Following DLI, a high pro-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


84<br />

W. Arcese et al.<br />

portion of CML patients with molecular, cytogenetic or<br />

chronic phase hematologic relapse will likely experience<br />

a long-term disease free survival, but the success rate<br />

is substantially lower in recurrent AML and virtually<br />

absent in patients relapsing with ALL or blast crisis<br />

CML. 186 Furthermore, the therapeutic success of DLI is<br />

counteracted by related severe complications such as<br />

GVHD and myelosuppression, which occur in up to 90%<br />

and 50% of cases, respectively. The mortality due to DLI<br />

may approach 20%.<br />

Therefore in order to optimize adoptive immunotherapy<br />

with DLI and to improve the general management of posttransplant<br />

relapse, several biological and clinical conditions<br />

should be considered.<br />

One of the most important factors is the time required<br />

for GVL to destroy the host neoplastic cells. In early stage<br />

CML this time seems to be enough to allow a GVL reaction<br />

to build up and eliminate residual CML cells. By contrast,<br />

neoplastic growth is so fast in acute leukemia that<br />

it may not be challenged by the GVL effect.<br />

A further variable influencing the response to DLI is the<br />

potential of the neoplastic clone to mature and differentiate<br />

into dendritic cells, which contribute to the GVL reaction<br />

by enhancing the antigen presentation capacity of<br />

tumor cells. This property is spontaneously attained by<br />

CML cells in chronic phase and, to some extent, by AML<br />

cells, particularly when cell differentiation follows the<br />

tumor reduction induced by chemotherapy. 197 Dendritic<br />

cells derived from bone marrow or produced in vitro by<br />

CD34 + cell cultures in the presence of cytokines 198,199 can<br />

exert their action through an HLA restricted mechanism. 200<br />

Therefore, in treating leukemia relapse weakly expressing<br />

HLA or tumor-specific antigens, donor hematopoietic<br />

progenitor cells may improve the immunologic effect<br />

mediated by DLI.<br />

Finally, bone marrow chimerism detected by PCR prior<br />

to DLI may predict either response to treatment or the<br />

occurrence of myelosuppression. Although long-term persistence<br />

of donor T-cells in the peripheral blood during<br />

relapse has been reported, 184 this observation does not<br />

provide any prognostic information on the post-DLI clinical<br />

outcome. Southern blot RFLP analysis, erythrocyte<br />

phenotype and cytogenetics have been employed to<br />

detect residual donor cells, but no correlation was found<br />

among pre-DLI BM chimerism, response to treatment and<br />

the risk of myelosuppression. However, pre-DLI BM<br />

chimerism assessed by quantitative PCR of VNTR<br />

sequences in relapsed CML patients is associated with<br />

cytogenetic and molecular remission and strongly predicts<br />

the development of aplasia, thereby providing an<br />

early indication for the reinfusion of PBSC from the<br />

donor. 201,202<br />

Donor PBSC reinfusion has frequently been adopted as<br />

from rescue of DLI-associated myelosuppression. As to the<br />

combined use of donor PBSC and DLI, the reported experiences<br />

are limited to a small number of patients who<br />

relapsed with acute leukemia. 203-205<br />

In these studies, donors were stimulated with G-CSF at<br />

doses ranging from 2.5 mg/kg for 10 days to 16 mg/kg for<br />

5 days. The apheresis products obtained over 1 to 3 consecutive<br />

days contained a median of 4×10 8 /kg CD34 + cells<br />

and a median of 3.5×10 8 /kg CD3 + cells. All patients<br />

received chemotherapy prior to PBSC infusion and most<br />

of them achieved CR with prompt hematopoietic reconstitution<br />

which in the cases analyzed originated from<br />

donor cells. The majority of patients developed acute or<br />

chronic GVHD and related complications. In one of the<br />

reported series, the median duration of CR after this combined<br />

treatment was longer than the median time from<br />

transplant to relapse. 206 These results compare favorably<br />

with those recently reported in patients receiving DLI<br />

alone for relapse of acute leukemia or myelodysplasia after<br />

BMT. 187 Of the eight patients receiving this treatment, only<br />

one achieved CR and 7 died of progressive disease.<br />

In conclusion, these preliminary experiences suggest<br />

that patients relapsing with acute leukemia or advanced<br />

phase CML after BMT should be treated with intensive<br />

chemotherapy regimens, not necessarily including<br />

immunosuppressive drugs, followed by donor mobilized<br />

PBSC.<br />

This approach might result in certain therapeutic<br />

advantages such as: 1) reduction of the tumor burden; 2)<br />

slowing down of the neoplastic growth; 3) acceleration of<br />

donor hematopoiesis recovery and promotion of dendritic<br />

cell differentiation; 4) the possibility for the immunocompetent<br />

donor cells to express their GVL activity to a<br />

greater extent. Whether the additional administration of<br />

cytokines (IFN, IL-2, G-CSF, GM-CSF) would improve the<br />

efficacy of chemotherapy and PBSC is unknown at present<br />

and awaits further investigation.<br />

Finally, the GVL reaction exerted by donor lymphocytes<br />

against CML cells which retain biological features of early<br />

stage disease is potent enough that patients might be<br />

spared a repetition of previous chemotherapy. However,<br />

donor PBSC infusion should be considered for CML<br />

patients with either cytogenetic or chronic phase relapse<br />

who show minimal (< 10%) or no BM chimerism. 206 In<br />

such cases the use of donor PBSC is mainly indicated to<br />

counteract the risk of severe BM aplasia following the<br />

infusion of DLI alone.<br />

Contributions and Acknowledgments<br />

All authors equally contributed to the conception and<br />

writing of this review article.<br />

Disclosures<br />

Conflict of interest: this review article was prepared<br />

by a group of experts designated by <strong>Haematologica</strong> and<br />

by representatives of two pharmaceutical companies,<br />

Amgen Italia SpA and Dompé Biotec SpA, both from<br />

Milan, Italy. This co-operation between a medical journal<br />

and pharmaceutical companies is based on the common<br />

aim of achieving an optimal use of new therapeutic<br />

procedures in medical practice. In agreement with the<br />

Journal’s Conflict of Interest Policy, the reader is given<br />

the following information. The preparation of this manuscript<br />

was supported by educational grants from the<br />

two companies. Dompé Biotec SpA sells G-CSF and rHuEpo<br />

in Italy, and Amgen Italia SpA has a stake in Dompé<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

85<br />

Biotec SpA.<br />

Redundant publications: no substantial overlapping<br />

with previous papers.<br />

Manuscript processing<br />

Received on July 29, 1997; accepted on November 28,<br />

1997.<br />

References<br />

1. Majolino I, Cavallaro AM, Scimè R. Peripheral blood<br />

stem cells for allogeneic transplantation. Bone Marrow<br />

Transplant 1996; 18(suppl 2):171-4.<br />

2. Rubinstein P, Rosenfield RE, Adamson JW, Stevens<br />

CE. Stored placental blood for unrelated bone marrow<br />

reconstitution. Blood 1993; 81:1679-90.<br />

3. Gratwohl A, Hermans J, Baldomero H, for the European<br />

Group for Blood and Marrow Transplantation<br />

(EBMT). Blood and marrow transplantation activity<br />

in Europe 1995. Bone Marrow Transplant 1997;<br />

19:407-19.<br />

4. Gluckman E, Broxmeyer HE, Auerbach AD, et al.<br />

Hematopoietic reconstitution in a patient with Fanconi’s<br />

anemia by means of umbilical-cord blood from<br />

an HLA-identical sibling. N Engl J Med 1989; 321:<br />

1174-8.<br />

5. Vilmer E, Sterkers G, Rahimy C, et al. HLA-mismatched<br />

cord blood transplantation in a patient with<br />

advanced leukemia. Transplantation 1992; 53:1155-<br />

7.<br />

6. Rubinstein P, Carrier C, Adamson J, et al. New York<br />

Blood Center’s program for unrelated placental/umbilical<br />

cord blood (PCB) transplantation: 243<br />

transplants in the first 3 years [abstract]. Blood 1996;<br />

88 (suppl 1):142a.<br />

7. Gluckman E, Rocha V, Boyer-Chammard A, et al. for<br />

the Eurocord Transplant Group and the European<br />

Blood and Marrow Transplantation Group. Outcome<br />

of Cord-Blood Transplantation from Related and<br />

Unrelated Donors. N Engl J Med 1997; 337:373-81.<br />

8. Risdon G, Gaddy J, Horrie M, Broxmeyer HE. Alloantigen<br />

priming induces a state of unresponsiveness in<br />

human umbilical cord blood T-cells. Proc Natl Acad<br />

Sci USA 1995; 92:2413-7.<br />

9. Bacigalupo A, Piaggio G, Podestà M, et al. Influence<br />

of marrow CFU-GM content on engraftment and survival<br />

after allogeneic bone marrow transplantation.<br />

Bone Marrow Transplant 1995; 15:221-6.<br />

10. Majolino I, Aversa F, Bacigalupo A, Bandini G, Arcese<br />

W, Reali G. Allogeneic transplants of rhG-CSF-mobilized<br />

peripheral blood stem cells (PBSC) from normal<br />

donors. <strong>Haematologica</strong> 1995; 80:40-3.<br />

11. Majolino I, Aversa F, Bacigalupo A, Bandini G, Arcese<br />

W. Peripheral blood stem cells for allogeneic transplantation.<br />

Recommendations from the GITMO<br />

1996. <strong>Haematologica</strong> 1996; 82:529-31.<br />

12. Bertolini F, De Vincentiis A, Lanata L, et al. Allogeneic<br />

hematopoietic stem cells from sources other than<br />

bone marrow: biological and technical aspects.<br />

<strong>Haematologica</strong> 1997; 82:220-38.<br />

13. Goodman JW, Hodgson GS. Evidence for stem cells in<br />

the peripheral blood of mice. Blood 1962; 19:702-<br />

14.<br />

14. Thomas ED, Collins JA, Herman Jr EC, Ferrebee JW.<br />

Marrow transplants in lethally irradiated dogs given<br />

methotrexate. Blood 1962; 19:217-28.<br />

15. Cavins JA, Scheer SC, Thomas ED, Ferrebee JW. The<br />

recovery of lethally irradiated dogs given infusions of<br />

autologous leukocytes preserved at –80°C. Blood<br />

1964; 21:38-43.<br />

16. Epstein RB, Graham TC, Buckner CD, Bryant J,<br />

Thomas ED. Allogeneic marrow engraftment by cross<br />

circulation in lethally irradiated dogs. Blood 1966;<br />

28:692-707.<br />

17. Storb R, Epstein RB, Radge H, Bryant J, Thomas ED.<br />

Marrow engraftment by allogeneic leukocytes in<br />

lethally irradiated dogs. Blood 1967; 30:805-11.<br />

18. Storb R, Graham TC, Epstein RB, Sale GE, Thomas<br />

ED. Demonstration of hemopoietic stem cells in the<br />

peripheral blood of baboons by cross circulation.<br />

Blood 1977; 50:537-42.<br />

19. Körbling M, Fliedner TM, Calvo W, Ross WM, Nothdurft<br />

W, Steinbach I. Albumin density gradient purification<br />

of canine hemopoietic blood stem cells<br />

(HBSC): long-term allogeneic engraftment without<br />

GVH-reaction. Exp Hematol 1979; 7:277-88.<br />

20. Carbonell F, Calvo W, Fliedner TM, et al. Cytogenetic<br />

studies in dogs after total body irradiation and allogeneic<br />

transfusion with cryopreserved blood mononuclear<br />

cells: observations in long-term chimeras. Int J<br />

Cell Cloning 1984; 2:81-8.<br />

21. Goldman JM, Catovsky D, Hows J, Spiers ASD, Galton<br />

DAG. Cryopreserved peripheral blood cells functioning<br />

as autografts in patients with chronic granulocytic<br />

leukemia in transformation. Br Med J 1979; 1:1310-<br />

3.<br />

22. Richman CM, Weiner RS, Yankee RA. Increase in circulating<br />

stem cells following chemotherapy in man.<br />

Blood 1976; 47:1031-4.<br />

23. Socinski MA, Cannistra SA, Elias A, Antman KH,<br />

Schnipper L, Griffin JD. Granulocyte-macrophage<br />

colony stimulating factor expands the circulating<br />

hemopoietic cell compartment in man. Lancet 1988;<br />

i:1194-8.<br />

24. Gianni AM, Siena S, Bregni M, et al. Granulocytemacrophage<br />

colony-stimulating factor to harvest circulating<br />

hematopietic stem cells for autotransplantation.<br />

Lancet 1989; 2:580-5.<br />

25. Kessinger A, Smith DM, Strandjord SE, et al Allogeneic<br />

transplantation of blood-derived, T-cell depleted<br />

hemopoietic stem cells after myeloablative treatment<br />

in a patient with acute lymphoblastic leukemia. Bone<br />

Marrow Transplant 1989; 4:643-6.<br />

26. Russell NH, Hunter A, Rogers S, et al. Peripheral<br />

blood stem cells as an alternative to marrow for allogeneic<br />

transplantation. Lancet 1993; 341:1482.<br />

27. Dreger P, Suttorp M, Haferlach T, Loffler M, Schmitz<br />

N, Schroyens W. Allogeneic granulocyte colony-stimulating<br />

factor-mobilized peripheral blood progenitor<br />

cells for treatment of engraftment failure after bone<br />

marrow transplantation. Blood 1993; 81:1404-7.<br />

28. Weaver CH, Buckner CD, Longin K, et al. Syngeneic<br />

transplantation with peripheral blood mononuclear<br />

cells collected after the administration of recombinant<br />

human granulocyte colony-stimulating factor.<br />

Blood 1993; 82:1981-4.<br />

29. Bensinger WI, Weaver CH, Appelbaum FR, et al.<br />

Transplantation of allogeneic peripheral blood stem<br />

cells mobilized by recombinant human granulocyte<br />

colony stimulating factor. Blood 1995; 85:1655-8.<br />

30. Körbling M, Przepiorka D, Huh YO, et al. Allogeneic<br />

blood stem cell transplantation for refractory<br />

leukemia and lymphoma. Potential advantages of<br />

blood over marrow allografts. Blood 1995; 85: 1659-<br />

65.<br />

31. Schmitz N, Dreger P, Suttorp M, et al. Primary transplantation<br />

of allogeneic peripheral blood cells mobilized<br />

by filgrastim (granulocyte colony-stimulating factor).<br />

Blood 1995; 85:1666-72.<br />

32. Majolino I, Saglio G, Scime’ R, et al. High incidence<br />

of chronic GVHD after primary allogeneic peripheral<br />

blood stem cell transplantation in patients with hema-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


86<br />

W. Arcese et al.<br />

tologic malignancies. Bone Marrow Transplant 1996;<br />

17:555-60.<br />

33. Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment<br />

of T-depleted haploidentical three loci incompatible<br />

transplants in leukemia patients by addition of<br />

recombinant human granulocyte-colony-stimulating<br />

factor mobilized peripheral blood progenitor cells to<br />

bone marrow inoculum. Blood 1994; 84:3948-55.<br />

34. Ringdén O, Potter MN, Oakhill A, et al. Transplantation<br />

of peripheral blood progenitor cells from<br />

unrelated donors. Bone Marrow Transplant 1996;<br />

17(suppl 2):62-4.<br />

35. Majolino I, Cavallaro AM, Bacigalupo A, et al. Mobilization<br />

and collection of PBSC in healthy donors. A<br />

retrospective analysis of the Italian Bone Marrow<br />

Transplantation Group (GITMO). <strong>Haematologica</strong><br />

1997; 82:47-52.<br />

36. Hasenclever D, Sextro M. Safety of alloPBPCT donors:<br />

biometrical considerations on monitoring long term<br />

risks. Bone Marrow Transplant 1996; 17(suppl 2):28-<br />

30.<br />

37. Aversa F, Pelicci PG, Terenzi A, et al. Results of T-<br />

depleted BMT in chronic myelogenous leukaemia<br />

after a conditioning regimen that included Thiotepa.<br />

Bone Marrow Transplant 1991; 7:24.<br />

38. Körbling M, Mirza N, Thall P, et al. Experience with<br />

100 HLA-identical allogeneic blood stem cell transplantations:<br />

CD34 cell dose and engraftment.<br />

[abstract]. Blood 1996; 88 (Suppl.I Part 1):2451.<br />

39. Przepiorka D, Ippoliti C, Khoury I, et al. Allogeneic<br />

transplantation for advanced leukemia: improved<br />

short-term outcome with blood stem cell grafts and<br />

tacrolimus. Transplantation 1996; 62:1806-10.<br />

40. Bacigalupo A, Van Lint MT, Valbonesi M, et al. Thiotepa<br />

cyclophosphamide followed by granulocyte<br />

colony-stimulating factor mobilized allogeneic peripheral<br />

blood cells in adults with advanced leukemia.<br />

Blood 1996; 88:353-7.<br />

41. Khouri I, Keating MJ, Przepiorka D, et al. Engraftment<br />

and induction of GVL with Fludarabine (FAMP) based<br />

non-ablative preparative regimen in patients with<br />

chronic lymphocytic leukemia (CLL) and lymphoma<br />

[abstract]. Blood 1996; 88 (Suppl. I Part 1):1194.<br />

42. Martelli MF, Aversa F, Velardi A, et al. New tools for<br />

crossing the HLA barrier: fludarabine and megadose<br />

stem cell transplants [abstract]. Blood 1996; 88 (Suppl<br />

1):1924.<br />

43. Giralt S, Estey E, van Besien K, et al. Induction of graftversus-leukemia<br />

without myeloablative therapy using<br />

allogeneic PBSC after purine analog containing regimens<br />

[abstract]. Blood 1996; 88 (Suppl.I Part<br />

1):2444.<br />

44. Adkins D, Spitzer G, Brown R, et al. High-dose rate<br />

(30 cGy/min) low-dose (550 cGy) single exposure<br />

total body irradiation (TBI) as conditioning for allogeneic<br />

peripheral blood stem cell (ALLOPBSC) transplantation<br />

results in complete donor chimerism and<br />

rapid engraftment without severe acute graft versus<br />

host disease (AGVHD) or toxicity [abstract]. Blood<br />

1996; 88 (Suppl.I Part 1):443.<br />

45. Tan P, Yeow Tee G. Stem cell allografting using a nonmyeloablative<br />

regimen (Abstract). Blood 1996; 88<br />

(Suppl.I Part 1):449.<br />

46. Slavin S, Nagler A, Naperstek E, et al. Immunotherapy<br />

of leukemia in conjunction with non-myeloablative<br />

conditioning: engraftment of blood stem cells and<br />

eradication of host leukemia with non-myeloablative<br />

conditioning based on fludarabine and anti-thymocyte<br />

globulin (ATG) [abstract]. Blood 1996; 88 (Suppl.I<br />

Part 1):2443.<br />

47. Majolino I, Cavallaro AM, Santoro A, et al. Successful<br />

engraftment of allogeneic PBSC after conditioning<br />

with busulfan alone. Bone Marrow Transplant 1997;<br />

19:621-3.<br />

48. Harada M, Kodera Y, Asano S. Clinical results of allogeneic<br />

peripheral blood stem cell transplantation:<br />

Japanese survey 1995. Bone Marrow Transplant 1996;<br />

17(Suppl 2):S47.<br />

49. Eckart JR, Roodman G, Boldt D, et al. Comparison of<br />

engraftment and acute GVHD in patients undergoing<br />

cryopreserved or fresh allogeneic BMT. Bone Marrow<br />

Transplant 1993; 11:125-31.<br />

50. Schmitz N, Bacigalupo A, Labopin M, et al. Transplantation<br />

of peripheral blood progenitor cells from<br />

HLA-identical sibling donors. Br J Haematol 1996;<br />

95:715-23.<br />

51. Körbling M, Huh YO, Durett A, et al. Allogeneic blood<br />

stem cell transplantation: peripheralization and yield<br />

of donor-derived primitive hematopoietic progenitor<br />

cells(CD34+ Thy-1 dim) and lymphoid subsets, and<br />

possible predictors of engraftment and graft-versushost<br />

disease. Blood 1996; 86:2842-8.<br />

52. Russel JA, Brown C, Bowen T, et al. Allogeneic blood<br />

cell transplants for hematological malignancy: preliminary<br />

comparison of outcomes with bone marrow<br />

transplantation. Bone Marrow Transplant 1996;<br />

17:703-8.<br />

53. Bensinger W, Buckner C, Demirez T, Storb R, Appelbaum<br />

F. Transplantation of allogeneic peripheral<br />

blood stem cells. Bone Marrow Transplant 1996; 17<br />

(Suppl 2):S56.<br />

54. Urbano-Ispizua A, Solano C, Brunet S, et al. Allogeneic<br />

peripheral blood progenitor cell transplantation:<br />

analysis of short term engraftment and acute<br />

GVHD incidence in 33 cases. Bone Marrow Transplant<br />

1996; 18:35-40.<br />

55. Przepiorka D, Anderlini P, Ippoliti C, et al. Allogeneic<br />

blood stem cell transplantation: reduction in early<br />

treatment-related morbidity and mortality for patients<br />

with advanced hematologic malignancies. Bone Marrow<br />

Transplant, in press.<br />

56. Bensinger W, Clift R, Anasetti C, et al. Transplantation<br />

of allogeneic peripheral blood stem cells mobilized by<br />

recombinant human granulocyte colony stimulating<br />

factor. Stem cells 1996; 14:90-105.<br />

57. Rosenfeld C, Collins R, Piñeiro L, Agura E, Nemunaitis<br />

J. Allogeneic blood cell transplantation without posttransplant<br />

colony-stimulating factors in patients with<br />

hematopoietic neoplasm: a phase II study. J Clin<br />

Oncol 1996; 14:1314-9.<br />

58. Roy J, Delage R, Demers C, et al. Stem cells collected<br />

with intermediate dose G-CSF lead to prompt engraftment<br />

in allogeneic blood transplant recipients<br />

[abstract]. Blood 1996; 88 (Suppl.I Part 1):1603.<br />

59. Atkinson K. Reconstitution of the haemopoietic and<br />

immune systems after marrow transplantation. Bone<br />

Marrow Transplant 1990; 5:209-26.<br />

60. Lum LG. The kinetics of immune reconstitution after<br />

human marrow transplantation. Blood 1987; 69:369-<br />

80.<br />

61. Voltarelli JC, Stites DP. Immunologic monitoring of<br />

bone marrow transplantation. Diagn Immunol 1986;<br />

4:171-93.<br />

62. Ferrara JLM, Deeg HJ. Graft-versus-host disease. N<br />

Engl J Med 1991; 310:667-74.<br />

63. Kato S, Yabe M, Yabe M, et al. Studies on transfer of<br />

varicella-zoster-virus specific T-cell immunity from<br />

bone marrow donor to recipient. Blood 1990; 75:<br />

806-9.<br />

64. Boland GJ, Vlieger AM, Ververs C, Gast GC. Evidence<br />

for transfer of cellular and humoral immunity to<br />

cytomegalovirus from donor to recipient in allogeneic<br />

bone marrow transplantation. Clin Exp Immunol<br />

1992; 88:506-11.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

87<br />

65. Rouleau M, Senik A, Leroy E, Vernant JP. Long-term<br />

persistence of transferred PPD-reactive T cells after<br />

allogeneic bone marrow transplantation. Transplantation<br />

1993; 55:72-6.<br />

66. Vavassori M, Maccario R, Moretta A, et al. Restricted<br />

TCR repertoire and long-term persistence of donorderived<br />

antigen-experienced CD4+ T-cells in allogeneic<br />

bone marrow transplantation recipients. J Immunol<br />

1996; 157:5739-47.<br />

67. Ottinger HD, Beelen DW, Scheulen B, Schaefer UW,<br />

Grosse-Wilde H. Improved immune reconstitution<br />

after allotransplantation of peripheral blood stem<br />

cells instead of bone marrow. Blood 1996; 88:2775-<br />

9.<br />

68. Mielcarek M, Martin P, Torok-Storb B. Suppression of<br />

alloantigen-induced T cell proliferation by large numbers<br />

of CD14+ cells in G-CSF mobilized peripheral<br />

blood mononuclear cells. Blood 1996; 88 (Suppl<br />

1):249a.<br />

69. Pavletic ZS, Bishop MR, Joshi SS, et al. Immunologic<br />

reconstitution after allogeneic blood stem cell transplantation:<br />

fast lymphocyte recovery correlates with<br />

better survival. Blood 1996; 88 (Suppl 1):422a.<br />

70. Donnenberg AD, Donnenberg VS, Meyer EM, Patton<br />

TJ, Griffin EL, Ball ED. Accelerated immune reconstitution<br />

after matched allogeneic PBSC transplantation.<br />

Blood 1996; 88 (Suppl 1):612a.<br />

71. Walter EA, Greenberg PD, Gilbert MJ, et al. Reconstitution<br />

of cellular immunity against cytomegalovirus<br />

in recipients of allogeneic bone marrow by transfer of<br />

T-cell clones from the donor. N Engl J Med 1995;<br />

333:1038-44.<br />

72. Papadopoulos EB, Ladanyi M, Emanuel D, et al. Infusions<br />

of donor leukocytes as treatment of Epstein-Barr<br />

virus associated lymphoproliferative disorders complicating<br />

allogeneic marrow transplantation. N Engl J<br />

Med 1994; 330:1185-91.<br />

73. Rooney CM, Smith CA, Ny CYC, et al. Use of genemodified<br />

virus-specific T lymphocytes to control<br />

Epstein-Barr-virus-related lymphoproliferation. Lancet<br />

1995; 245:9-13.<br />

74. Bensinger W, Clift R, Martin P, et al. Allogeneic<br />

peripheral blood stem cell transplantation in patients<br />

with advanced hematologic malignancies: a retrospective<br />

comparison with marrow transplantation.<br />

Blood 1996; 88:2794-800.<br />

75. Anderlini P, Przepiorka D, Champlin R, Korbling M.<br />

Biologic and clinical effects of granulocyte colony<br />

stimulating factor in normal individuals. Blood 1996;<br />

88:2819-25.<br />

76. Majolino I, Scimé R, Cavallaro AM, et al. Transplantation<br />

of unmanipulated allogeneic PBSC: preliminary<br />

report on 24 patients. Leuk Lymphoma 1998;<br />

in press.<br />

77. Azevedo VM, Aranha FJP, Gouvea JV, et al. Allogeneic<br />

transplantation with blood stem cells mobilized by<br />

rhG-CSF for hematological malignancies. Bone Marrow<br />

Transplant 1995; 16:647-53.<br />

78. Deeg HJ, Huss R. Acute graft-versus-host disease. In:<br />

Atkinson K, ed. Clinical bone marrow transplantation:<br />

a reference textbook. Cambridge: Cambridge University<br />

Press 1994. p. 297-311.<br />

79. Chao MJ, Schmidt GM, Niland JC, et al. Cyclophosphamide,<br />

methotrexate, and prednisone compared<br />

with cyclosporine and prednisone for prophylaxis<br />

of acute graft-versus-host disease. N Engl J Med<br />

1993; 329:1225-30.<br />

80. Gratwhol A, Hermans J, Apperley J, et al. Acute graftversus-host<br />

disease: grade and outcome in patients<br />

with chronic myelogenous leukemia. Blood 1995;<br />

85:813-8.<br />

81. Kernan NA, Collins NM, Juliano M, Cartagenia T,<br />

Dupont B, O’Reilly RJ. Clonable T lymphocytes in T-<br />

cell depleted bone marrow transplants correlate with<br />

development of graft-versus-host disease. Blood<br />

1986; 68:770-3.<br />

82. Sullivan KM, Storb R, Buckner CD, et al. Graft-versushost<br />

disease as adoptive immunotherapy in patients<br />

with advanced hematologic neoplasms. N Engl J Med<br />

1989; 320:828-34.<br />

83. Pan L, Delmonte J Jr, Jalonen CK, Ferrara JLM. Pretreatment<br />

of donor mice with granulocyte-colony<br />

stimulating factor polarizes donor T lymphocytes<br />

toward type-2 cytokine production and reduces severity<br />

of experimental graft versus host disease. Blood<br />

1995; 86:4422-8.<br />

84. Gorgen I, Hartung T, Leist M, et al. Granulocyte<br />

colony-stimulating factor treatment protects rodents<br />

against lipopolysaccharide-induced toxicity via suppression<br />

of systemic tumor necrosis factor-a. J<br />

Immunol 1992; 149:918-24.<br />

85. Hartung T, Docke WD, Grabner F, et al. Effect of granulocyte<br />

colony-stimulating factor treatment on ex vivo<br />

blood cytokine response in human volunteers. Blood<br />

1995; 85:2482-9.<br />

86. Talmadge JE, Singh RK, Agetois A, Ozerol I, Ino K. T<br />

cell apoptosis (mediated in part by FAS ligand) by<br />

monocytes in cytokine mobilized stem cell products:<br />

resultant immmunosuppression. Blood 1996; 88<br />

(suppl 1):610 a.<br />

87. Anderlini P, Przepiorka D, Khouri I, et al. Chronic<br />

graft-vs-host disease after allogeneic marrow or blood<br />

stem cell transplantation [abstract]. Blood 1995;<br />

86(suppl 1):109 .<br />

88. Storb R, Doney KC, Thomas ED, et al. Marrow transplantation<br />

with or without donor buffy-coat cells for<br />

65 transplanted aplastic anemia patients. Blood<br />

1982; 59:236-46.<br />

89. Carlo-Stella C, Tabilio A. Stem cells and stem cell<br />

transplatation. <strong>Haematologica</strong> 1996; 81:573-87.<br />

90. Strauss LC, Rowley SD, Larussa VF, et al. Antigenic<br />

analysis of hematopoiesis. V. Characterization of<br />

MY10 antigen expression by normal lymphohematopoietic<br />

progenitor cells. Exp Hematol 1986; 14:878-<br />

86.<br />

91. Lemoli RM, Tazzari PL, Fortuna A, et al. Positive selection<br />

of hematopoietic CD34+ stem cells provides<br />

“indirect purging” of CD34– lymphoid cells and the<br />

purging efficiency is increased by anti-CD2 and anti-<br />

CD30 immunotoxins. Bone Marrow Transplant 1994;<br />

13:465-71.<br />

92. Dreger P, Viehmann K, Steinmann J, et al. G-CSFmobilized<br />

peripheral blood progenitor cells for allogeneic<br />

transplantation: comparison of T cell depletion<br />

strategies using different CD34+ selection systems<br />

or CAMPATH 1. Exp Hematol 1995; 23:147-54.<br />

93. Shpall EJ, Jones RB, Bearman SI, et al. Transplantation<br />

of enriched CD34-positive autologous marrow into<br />

breast cancer patients following high-dose chemotherapy:<br />

Influence of CD34-positive peripheral blood<br />

progenitors and growth factors on engraftment. J Clin<br />

Oncol 1994; 12:28-36.<br />

94. Brugger W, Henschler R, Heimfeld S, et al. Positively<br />

selected autologous blood CD34+ cells and unseparated<br />

peripheral blood progenitor cells mediate identical<br />

hematopoietic engraftment after high dose VP<br />

16, ifosfamide, carboplatin, and epirubicin. Blood<br />

1994; 84:1421-6.<br />

95. Gorin NC, Lopez M, Laporte JP, et al. Preparation and<br />

successful engraftment of purified CD34+ bone marrow<br />

progenitor cells in patients with non-Hodgkin’s<br />

lymphoma. Blood 1995; 86:1647-54.<br />

96. Lemoli RM, Fortuna A, Motta MR, et al. Concomitant<br />

mobilization of plasma cells and hematopoietic prog-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


88<br />

W. Arcese et al.<br />

enitors into peripheral blood of multiple myeloma<br />

patients: positive selection and transplantation of<br />

enriched CD34+ cells to remove circulating tumor<br />

cells. Blood 1996; 1625-34.<br />

97. Link H, Arseniev L, Bahre O, et al. Combined transplantation<br />

of allogeneic bone marrow and CD34+<br />

blood cells. Blood 1995; 86:2500-8.<br />

98. Link H, Arseniev L, Bähre O, Kadar J, Diedrich H, Poliwoda<br />

H. Transplantation of allogeneic CD34+ blood<br />

cells. Blood 1996; 87:4903-9.<br />

99. Bensinger W, Buckner C, Shannon-Dorcy K, et al.<br />

Transplantation of allogeneic CD34+ peripheral<br />

blood stem cells in patients with advanced hematologic<br />

malignancy. Blood 1996; 88:4132-8.<br />

100. McCullough J, Hansen J, Perkins H, Stroncek D,<br />

Bartsch G. The National Marrow Donor Program:<br />

how it works, accomplishments to date. Oncology<br />

1989; 3:63-72.<br />

101. Kernan NA, Bartsch G, Ash RC, et al. Retrospective<br />

analysis of 462 unrelated marrow transplants facilitated<br />

by the National Marrow Donor Program<br />

(NMDP) for treatment of acquired and congenital disorders<br />

of the lymphohematopoietic system and congenital<br />

metabolic disorders. N Engl J Med 1993; 328:<br />

593-602.<br />

102. Stroncek DF, Clift RA, Sanders JE, et al. Experiences of<br />

the first 493 unrelated marrow donors in the national<br />

marrow donor program. Blood 1993; 81:1940-6.<br />

103. Beatty PG, Dahlberg S, Mickelson EM, et al. Probability<br />

of finding HLA-matched unrelated marrow<br />

donors. Transplantation 1988; 45:714-8.<br />

104. Anasetti C, Hansen J. Bone marrow transplantation<br />

from HLA-partially matched related donors and unrelated<br />

volunteer donors. In :Forman SJ, Blume KG,<br />

Thomas ED, eds. Bone Marrow Transplantation.<br />

Boston: Blackwell Scientific, 1994:665-79.<br />

105. Anasetti C, Amos D, Beatty PG, et al. Effect of HLA<br />

compatibility on engraftment of bone marrow transplants<br />

in patients with leukemia or lymphoma. N Engl<br />

J Med 1989; 320:197-204.<br />

106. Anasetti C, Beatty PG, Storb R, et al. Effect of HLAincompatibility<br />

on graft-versus-host disease, relapse,<br />

and survival after marrow transplantation for patients<br />

with leukemia or lymphoma. Hum Immunol 1990;<br />

29:79-91.<br />

107. Beatty PG, Clift RA, Mickelson EM, et al. Marrow<br />

transplantation from related donors other than HLAidentical<br />

siblings. N Engl J Med 1985; 313:765-71.<br />

108. Ash RC, Horowitz MM, Gale RP, et al. Bone marrow<br />

transplantation from related donors other than HLAidentical<br />

siblings: effect of T-cell depletion. Bone Marrow<br />

Transplant 1991; 7:443-52.<br />

109. Reisner Y, Kapoor N, Kirkpatrick D, et al. Transplantation<br />

for severe combined immunodeficiency<br />

with HLA-A, B, D, DR incompatible parental marrow<br />

fractionated by soybean agglutinin and sheep red<br />

blood cells. Blood 1983; 61:341-8.<br />

110. Reisner Y, Lapidot T, Singer TS, Schwartz E. The host<br />

barrier in animal models of T-cell depleted allogeneic<br />

bone marrow transplantation. In: Martelli MF, Grignani<br />

F, Reisner Y, eds. T-cell depletion in allogeneic<br />

bone marrow transplantation. Serono Symposia<br />

Review 1988; 13:37-47.<br />

111. Reisner Y, Kirkpatrick D, Dupont B, et al. Transplantation<br />

for acute leukaemia with HLA-A and B nonidentical<br />

parental marrow cells fractionated with soybean<br />

agglutinin and sheep red blood cells. Lancet 1981;<br />

2:327-31.<br />

112. O’Reilly RJ, Kernan NA, Cunningham I, et al. T cell<br />

depleted marrow transplants for the treatment of<br />

leukemia. In: Gale RP, Champlin RE, eds. Bone marrow<br />

transplantation: current controversies. New York,<br />

Alan R Liss, 1989. p. 477-93.<br />

113. Kernan NA, Collins NH, Juliano L, Cartagena BS,<br />

Dupont B, O’Reilly RJ. Clonable T lymphocytes in T-<br />

cell depleted bone marrow transplants correlate with<br />

development of graft versus host disease. Blood 1986;<br />

68:770-73.<br />

114. Kernan NA, Flomenberg N, Dupont B, O’Reilly RJ.<br />

Graft rejection in recipients of T-cell depleted HLA<br />

nonidentical transplants for leukemia: identification<br />

of host derived anti-donor allocytotoxic T-lymphocytes.<br />

Transplantation 1987; 43:482-7.<br />

115. Reisner Y, Martelli MF. Bone marrow transplantation<br />

across HLA barriers by increasing the number of transplanted<br />

cells. Immunol Today 1995; 16:437-40.<br />

116. Schwartz E, Lapidot T, Dalia G, Singer T, Reisner Y.<br />

Abrogation of bone marrow allograft resistance in<br />

mice by increased total body irradiation correlates<br />

with eradication of host clonable T cells and alloreactive<br />

cytotoxic precursors. J Immunol 1987; 138:<br />

460-5.<br />

117. Lapidot T, Singer TS, Salomon O, Terenzi A, Schwartz<br />

E, Reisner Y. Booster irradiation to the spleen following<br />

total body irradiation: A new immunosuppressive<br />

approach for allogeneic bone marrow transplantation.<br />

J Immunol 1988; 141: 2619-24.<br />

118. Cobbold SP, Martin G, Quin S, Waldman H. Monoclonal<br />

antobodies to promote marrow engraftment<br />

and tissue graft tolerance. Nature 1986; 323:164-6.<br />

119. Lapidot T, Terenzi A, Singer TS, Salomon O, Reisner<br />

Y. Enhancement by dimethyl myleran of donor type<br />

chimerism in murine recipients of bone marrow allografts.<br />

Blood 1989; 73:2025-32.<br />

120. Terenzi A, Lubin I, Lapidot T, et al. Enhancement of<br />

T-cell depleted bone marrow allografts in mice by<br />

thiotepa. Transplantation 1990; 50:717-20.<br />

121. Aversa F, Terenzi A, Carotti A, et al. Thiotepa improves<br />

results of T-cell depleted bone marrow transplants for<br />

acute leukemia. A seven year experience [abstract]. Br<br />

J Haematol 1996; 93(Suppl 2):616.<br />

122. Hale-G, Waldmann H. CAMPATH-1 monoclonal<br />

antibodies in bone marrow transplantation. J Hematother<br />

1994; 3:15-31.<br />

123. Lapidot T, Terenzi A, Singer TS, Salomon O, Reisner<br />

Y. Quantitative aspects of bone marrow allograft<br />

rejection in mice: new model with emphasis on relevance<br />

to leukemia patients. Bone Marrow Transplant<br />

1988; 3 (Suppl 1):18-22.<br />

124. Groopman JE, Molina JM, Scadden DT. Hematopoietic<br />

growth factors. Biology and clinical applications.<br />

N Engl J Med 1989; 321:1449-59.<br />

125. Gianni AM, Bregni M, Siena S, et al. Rapid and complete<br />

hematopoietic reconstitution following combined<br />

transplantation of autologous blood and bone<br />

marrow cells. A changing role for high dose<br />

chemotherapy? Hematol Oncol 1989; 7:139-48.<br />

126. Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment<br />

of T-cell depleted “three loci” mismatched transplants<br />

in leukemia patients by addition of rhG-CSFmobilized<br />

peripheral blood progenitor cells to bone<br />

marrow inoculum [abstract]. Exp Hematol 1994;<br />

22:146.<br />

127. Reisner Y, Friederich W, Fabian I. A shorter procedure<br />

for preparation of E-rosette depleted bone marrow<br />

for transplantation. Transplantation 1986; 42:312-6.<br />

128. Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment<br />

of T-cell depleted haploidentical “three loci”<br />

incompatible transplants in leukemia patients by<br />

addition of recombinant human granulocyte colonystimulating<br />

factor mobilized peripheral blood progenitor<br />

cells to bone marrow inoculum. Blood 1994;<br />

84:3948-55.<br />

129. Bachar-Lustig E, Rachamim N, Hong-Wei Li, Lan F,<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

89<br />

Reisner Y. Megadose of T cell depleted bone marrow<br />

overcomes MHC barriers in sublethally irradiated<br />

mice. Nature Med 1995; 12:1268-73.<br />

130. Rachamim N, Gan J, Segall H, et al. Human CD34<br />

hematopoietic stem cells exhibit potent veto activity.<br />

[Submitted for publication].<br />

131. Tabilio A, Falzetti F, Aversa F, et al. T-cell depletion for<br />

peripheral blood stem cells by CD34+ cell selection<br />

and E-rosettes. [Submitted for publication].<br />

132. Keating MJ, Kantarjian H, Talpaz M, et al. Fludarabine:<br />

a new agent with major activity against chronic<br />

lymphocytic leukemia. Blood 1989; 74:19-25.<br />

133. Terenzi A, Aversa F, Perruccio K, et al. Efficacy of fludarabine<br />

as immunosuppressor for bone marrow<br />

transplantation conditioning [abstract]. Blood 1996;<br />

88 (Suppl 1):2373.<br />

134. Martelli MF, Aversa F, Velardi A, et al. New tools for<br />

crossing the HLA barrier: fludarabine and megadose<br />

stem cell transplants [abstract]. Blood 1996; 88 (Suppl<br />

1):1924.<br />

135. O’Reilly RJ, Hansen JA, Kurtzberg J, et al. Allogeneic<br />

marrow transplants: approches for the patients lacking<br />

a donor. In: Schechter GP, McArthur JR, eds.<br />

Hematology 1996, Education Program of the American<br />

Society of Hematology. Orlando: American Society<br />

of Hematology; 1996. p. 132-46.<br />

136. Aversa F, Terenzi A, Velardi A, et al. Megadose stem<br />

cell transplants for crossing the HLA barrier<br />

[abstract]. J Mol Med 1997; 75:176.<br />

137. Albi N, Ruggeri L, Aversa F, et al. Natural killer (NK)-<br />

cell function and anti-leukemic activity of a large population<br />

of CD3+/CD8+ T-cells expressing NK receptors<br />

for major histocompatibility complex class I after<br />

“three loci” HLA-incompatible bone marrow transplantation.<br />

Blood 1996 ; 87:3993-4000.<br />

138. Bacigalupo A, Mordini N, Pitto A, et al. CD34+ selected<br />

stem cell transplants in patients with advanced<br />

leukemia from 3-loci mismatched family donors<br />

[abstract]. Blood 1995; 86 (Suppl 1):937.<br />

139. Kato K, Kojima S, Kondo M, Inaba J, Matsuyama.<br />

Allogeneic bone marrow and peripheral stem cell<br />

transplantation from a haplo-identical mother and<br />

CD34 positive selection for CML. Bone Marrow<br />

Transplant 1996; 18:449-52.<br />

140. Sierra J, Storer B, Hansen JA, et al. Transplantation of<br />

marrow cells from unrelated donors for treatment of<br />

high-risk acute leukemia: The effect of leukemic burden,<br />

donor HLA-matching and marrow cell dose.<br />

Blood 1997; 89: 4226-35.<br />

141. Stockschlader M, Loliger C, Kruger W, et al. Transplantation<br />

of allogeneic rh-G-CSF mobilized peripheral<br />

CD34+ cells from an HLA-identical unrelated<br />

donor. Bone Marrow Transplant 1995; 16:719-22.<br />

142. Gabutti V, Foà R, Mussa F, Aglietta M. Behavior of<br />

human haematopoietic stem cells in cord and neonatal<br />

blood [letter]. <strong>Haematologica</strong> 1975; 60: 492.<br />

143. Broxmeyer HE, Gordon GW, Hangoc G, et al. Human<br />

umbilical cord blood as a potential source of transplantable<br />

hematopoietic stem/progenitor cells. Proc<br />

Natl Acad Sci USA 1989; 86:3828-32.<br />

144. Wagner JE, Broxmeyer HE, Byrd RL, et al. Transplantation<br />

of umbilical cord blood after myeloablative<br />

therapy: analysis of engraftment. Blood 1992 79:<br />

1874-81.<br />

145. Vowels MR, Lam-PO-Tang R, Berdoukas V, et al. Brief<br />

report: correction of X-linked lymphoproliferative disease<br />

by transplantation of cord-blood stem cells. N<br />

Engl J Med 1993; 1623-5.<br />

146. Bogdanic V, Nemet D, Kastelal A, et al. Umbilical cord<br />

blood transplantation in a patient with Philadelphiachromosome<br />

positive chronic myelogenous leukemia.<br />

Transplantation 1993; 58:477-9.<br />

147. Laporte JP, Gorin NC, Rubinstein P, et al. Cord-blood<br />

transplantation from an unrelated donor in an adult<br />

with chronic myelogenous leukemia. N Engl J Med<br />

1996; 335:167-70.<br />

148. Wagner JE, Kernan NA, Steinbuch M, Broxmeyer HE,<br />

Gluckman E. Allogeneic sibling umbilical cord blood<br />

transplantation in forty-four children with malignant<br />

and non-malignant disease. Lancet 1995; 346:214-9.<br />

149. Kurtzberg J, Laughlin M, Graham M, et al. Placental<br />

blood as a source of hematopoietic stem cells for<br />

transplantation into unrelated recipients. N Engl J<br />

Med 1996; 335:157-66.<br />

150. Wagner JE, Rosenthal J, Sweetman R, et al. Successful<br />

transplantation of HLA-matched and HLA-mismatched<br />

umbilical cord blood from unrelated donors:<br />

analysis of engraftment and acute graft-versus-host<br />

disease. Blood 1996; 88:795-802.<br />

151. Gisselbrecht C, Prentice HG, Bacigalupo A, et al.<br />

Placebo-controlled phase I/II trial of lenograstim in<br />

bone-marrow transplantation. Lancet 1994; 343:<br />

696-700.<br />

152. Locatelli F, Pession A, Zecca M, et al. Use of recombinant<br />

human granulocyte colony-stimulating factor<br />

in children given allogeneic bone marrow transplantation<br />

for acute or chronic leukemia. Bone Marrow<br />

Transplant 1996; 17:31-7.<br />

153. Locatelli F, Maccario R, Comoli P, et al. Haematopoietic<br />

and immune recovery after transplantation of<br />

cord blood progenitor cells in children. Bone Marrow<br />

Transplant 1996; 18:1095-101.<br />

154. Galanello R, Melis MA, Ruggeri R, Cao A. Prospective<br />

study of red blood cell indices, hemoglobin A2 and<br />

hemoglobin F in infants heterozygous for b-thalassemia.<br />

J Pediat 1981; 99:105-7.<br />

155. Storb R, Deeg HJ, Whitehead J, et al. Methotrexate<br />

and cyclosporine compared with cyclosporine alone<br />

for prophylaxis of acute graft-versus-host disease after<br />

marrow transplantation for leukemia. N Engl J Med<br />

1986; 314:729-35.<br />

156. Sweetman R, Rosenthal J, Sender LS, et al. Successful<br />

engraftment, delayed immune reconstitution and minimal<br />

GVHD following unrelated cord blood transplantation<br />

in children and adults [abstract]. Blood<br />

1995; 86 (Suppl. 1):388a.<br />

157. Locatelli F, Maccario R, Zecca M. Placental blood<br />

transplantation [letter]. N Engl J Med 1997; 336:69.<br />

158. Vitetta ES, Berton MT, Burger C, Kepron M, Lee WT,<br />

Yin XM. Memory B and T cells. Ann Rev Immunol.<br />

1991; 9:193-217.<br />

159. Gathings WE, Kubagawa H, Cooper MD. A distinctive<br />

pattern of B cell immunity in perinatal humans.<br />

Immunol Rev 1981; 57:107-26.<br />

160. Burgio GR, Nespoli L, Locatelli F. Bone marrow transplantation<br />

in children: between primum non nocere<br />

(above all, do not harm) and primum adiuvare (above<br />

all, help). In: GR Burgio, JD Lantos, eds. Primum non<br />

nocere today. A symposium on pediatric bioethics.<br />

Amsterdam:Elsevier, 1994. p. 77-93.<br />

161. Silberstein LE, Jefferies LC. Placental-blood banking.<br />

A new frontier in transfusion medicine. N Engl J Med<br />

1996; 335:199-201.<br />

162. Marshall E. Clinical promise, ethical quandary. Science<br />

1996; 271:586-8.<br />

163. Lind SE. Ethical considerations related to the collection<br />

and distribution of cord blood stem cells for<br />

transplantation to reconstitute hematopoietic function.<br />

Transfusion 1994; 34:828-34.<br />

164. Gluckman E, O’Reilly RY, Wagner J, Rubinstein P.<br />

Patents versus transplants. Nature 1996; 382:108.<br />

165. Kumar L. Leukemia: management of relapse after allogeneic<br />

bone marrow transplantation. J Clin Oncol<br />

1994; 12:1710-7.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


90<br />

W. Arcese et al.<br />

166. Frassoni F, Barrett AJ, Granena A, et al. Relapse after<br />

allogeneic bone marrow transplantation for acute<br />

leukaemia: a survey by the E.B.M.T. of 117 cases. Br<br />

J Haematol 1988; 70:317-20.<br />

167. Mortimer J, Blinder MA, Schulman S, et al. Relapse of<br />

acute leukemia after marrow transplantation: natural<br />

history and results of subsequent therapy. J Clin<br />

Oncol 1989; 7:50-7.<br />

168. Arcese W, Mauro FR, Alimena G, et al. Interferon therapy<br />

for Ph-1 positive CML patients relapsing after T-<br />

cell depleted allogeneic bone marrow transplantation.<br />

Bone Marrow Transplant 1990; 5:309-15.<br />

169. Higano CS, Raskind W, Singer JW. Use of alpha-interferon<br />

for the treatment of relapse of chronic myelogenous<br />

leukemia after allogeneic bone marrow transplantation.<br />

Blood 1992; 80:1437-42.<br />

170. Arcese W, Goldman JM, D’Arcangelo E, et al. Outcome<br />

for patients who relapse after allogeneic bone<br />

marrow transplantation for chronic myeloid leukemia.<br />

Blood 1993; 82:3211-9.<br />

171. Sanders JE, Buckner CD, Clift RA, et al. Second marrow<br />

transplants in patients with leukemia who relapse<br />

after allogeneic bone marrow transplantation. Bone<br />

Marrow Transplant 1988; 3:11-9.<br />

172. Mrsic M, Horowitz MM, Atkinson K, et al. Second<br />

HLA-identical sibling transplants for leukemia recurrence.<br />

Bone Marrow Transplant 1992; 9:269-75.<br />

173. Radich JP, Sanders J, Buckner C, et al. Second allogeneic<br />

bone marrow transplantation for patients with<br />

recurrent leukemia after initial transplant with total<br />

body irradiation containing regimens. J Clin Oncol<br />

1993; 11:304-13.<br />

174. Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte<br />

infusion for treatment of recurrent chronic myelogenous<br />

leukemia in marrow transplant patients.<br />

Blood 1990; 76:2462-5.<br />

175. Drobyski WR, Roth MS, Thibodeau SN, et al. Molecular<br />

remission occurring after donor leukocyte infusions<br />

for the treatment of relapse chronic myelogenous<br />

leukemia after allogeneic bone marrow transplantation.<br />

Bone Marrow Transplant 1992; 10:301-4.<br />

176. Drobyski WR, Keever CA, Roth MS, et al. Salvage<br />

immunotherapy using donor leukocyte infusions as<br />

treatment for relapsed chronic myelogenous leukemia<br />

after allogeneic bone marrow transplantation: efficacy<br />

and toxicity of a defined T cell dose. Blood 1993;<br />

82:2310-8.<br />

177. Helg C, Roux E, Beris P, et al. Adoptive immunotherapy<br />

for recurrent CML after BMT. Bone Marrow<br />

Transplant 1993; 12:125-30.<br />

178. Hertenstein B, Wiesmeth M, Novotny J, et al. Interferon-a<br />

and donor buffy coat transfusions for treatment<br />

of relapsed chronic myeloid leukemia after allogeneic<br />

bone marrow transplantation. Transplantation<br />

1993; 56:1114-8.<br />

179. Szer J, Grigg AP, Phillips GL, et al. Donor leucocyte<br />

infusions after chemotherapy for patients relapsing<br />

with acute leukaemia following allogeneic BMT. Bone<br />

Marrow Transplant 1993; 11:109-11.<br />

180. Bar BMAM, Schattenberg A, Mensink EJBM, et al.<br />

Donor leucocyte infusion for chronic myeloid<br />

leukemia relapsed after allogeneic bone marrow transplantation.<br />

J Clin Oncol 1993; 11:513-9.<br />

181. Porter D, Roth M, McGarigle C, et al. Induction of<br />

graft-versus-host disease as immunotherapy for<br />

relapsed chronic myeloid leukemia. N Engl J Med<br />

1994; 330:100-6.<br />

182. van Rhee F, Lin F, Cullis JO, et al. Relapse of chronic<br />

myeloid leukemia after allogeneic bone marrow transplant:<br />

the case for giving donor leukocyte transfusions<br />

before the onset of hematologic relapse. Blood 1994;<br />

83:3377-83.<br />

183. Ferster A, Bujan W, Mouraux T, et al. Complete remission<br />

following donor leukocyte infusion in ALL relapsing<br />

after haploidentical bone marrow transplantation.<br />

Bone Marrow Transplant 1994; 14:331-2.<br />

184. Mackinnon S, Papadopoulos EB, Carabasi MM, et al.<br />

Adoptive immunotherapy evaluating escalating doses<br />

of donor leukocytes for relapse of chronic myeloid<br />

leukemia after bone marrow transplantation: separation<br />

of graft-versus-leukemia responses from graft-versus-host<br />

disease. Blood 1995; 86: 1261-8.<br />

185. Giralt S, Hester J, Huh Y, et al. CD-8 depleted donor<br />

lymphocyte infusion as treatment for relapsed chronic<br />

myelogenous leukemia after allogeneic bone marrow<br />

transplantation. Blood 1995; 86:4337-43.<br />

186. Kolb HJ, Schattenberg A, Goldman JM, et al. Graftversus-leukemia<br />

effect of donor lymphocyte transfusion<br />

in marrow grafted patients. Blood 1995;<br />

86:2041-50.<br />

187. Porter DL, Roth MS, Lee SJ, et al. Adoptive immunotherapy<br />

with donor mononuclear cell infusions to<br />

treat relapse of acute leukemia or myelodysplasia after<br />

allogeneic bone marrow transplantation. Bone Marrow<br />

Transplant 1996; 18:975-80.<br />

188. Slavin S, Naparstek E, Nagler A, et al. Allogeneic cell<br />

therapy with donor peripheral blood cells and recombinant<br />

human interleukin-2 to treat leukemia relapse<br />

after allogeneic bone marrow transplantation. Blood<br />

1996; 87:2195-204.<br />

189. Heslop HE, Brenner MK, Rooney CM. Donor T cells<br />

as therapy for EBV lymphoproliferation post bone<br />

marrow transplant. N Engl J Med 1994; 331:679.<br />

190. Weiden PL, Sullivan KM, Fluornoy N, et al. Antileukemic<br />

effect of chronic graft-versus-host disease.<br />

Contribution to improved survival after allogeneic<br />

marrow transplantation. N Engl J Med 1981; 304:<br />

1529-33.<br />

191. Apperley JF, Mauro FR, Goldman JM, et al. Bone marrow<br />

transplantation for chronic myeloid leukemia in<br />

first chronic phase: importance of a graft-versusleukemia<br />

effect. Br J Haematol 1988; 69:239-45.<br />

192. Frassoni F, Sessarego M, Bacigalupo A, et al. Competition<br />

between recipient and donor cells after bone<br />

marrow transplantation for chronic myeloid leukemia.<br />

Br J Haematol 1988; 69:471-3.<br />

193. Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia<br />

reactions after bone marrow transplantation.<br />

Blood 1990; 75:555-62.<br />

194. Higano CS, Brixey M, Bryant EM, et al. Durable complete<br />

remission of acute non-lymphocytic leukemia<br />

associated with discontinuation of immunosuppression<br />

following relapse after allogeneic bone marrow<br />

transplantation. A case report of a probable graft-versus-leukemia<br />

effect. Transfusion 1990; 50:175-7.<br />

195. Collins RH, Rogers ZR, Bennett M, et al. Hematologic<br />

relapse of chronic myelogenous leukemia following<br />

allogeneic bone marrow transplantation. Apparent<br />

graft-versus-leukemia effect following abrupt discontinuation<br />

of immunosuppression. Bone Marrow<br />

Transplant 1992; 10:391-5.<br />

196. Jiang YZ, Cullis JO, Kanfer EJ, et al. T-cell and NK cell<br />

mediated graft-versus-leukemia reactivity following<br />

donor buffy-coat transfusion to treat relapse after<br />

marrow transplantation for chronic myeloid leukemia.<br />

Bone Marrow Transplant 1993; 11:133-8.<br />

197. Mittermuller J, Kolb HJ, Gerhartz HH, et al. In vivo<br />

differentiation of leukemic blasts and effect of low<br />

dose ARA-C in a marrow grafted patient with leukemic<br />

relapse. Br J Haematol 1986; 62:757-62.<br />

198. Caux C, Dezutter-Dambuyant C, Schmitt D, et al.<br />

GM-CSF and TNF-a cooperate in the generation of<br />

dendritic Langerhans cells. Nature 1992; 360:258-61.<br />

199. Romani N, Gruner S, Brang D, et al. Proliferating den-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of allogeneic hematopoietic stem cells<br />

91<br />

dritic cell progenitors in human blood. J Exp Med<br />

1994; 180:83-93.<br />

200. Huang AYC, Golumbeck P, Ahmadzadeh M, et al.<br />

Role of bone marrow-derived cells presenting MHC<br />

class I-restricted tumor antigens. Science 1994;<br />

264:961-5.<br />

201. Rapanotti MC, Arcese W, Buffolino S, et al. Sequential<br />

molecular monitoring of chimerism in chronic<br />

myeloid leukemia patients receiving donor lymphocyte<br />

transfusion for relapse after bone marrow transplantation.<br />

Bone Marrow Transplant 1997; 19:703-7.<br />

202. Keil F, Haas OA, Fritsch G, et al. Donor leukocyte infusion<br />

for leukemic relapse after allogeneic marrow<br />

transplantation: lack of residual donor hematopoiesis<br />

predicts aplasia. Blood 1997; 89:3113-7.<br />

203. Sica S, Di Mario A, Salutari P, et al. Chemotherapy<br />

and recombinant human granulocyte colony-stimulating<br />

factor primed donor leukocyte infusion for<br />

treatment of relapse after allogeneic bone marrow<br />

transplantation. Bone Marrow Transplant 1995;<br />

16:483-5.<br />

204. Metha J, Powles R, Shighal S, et al. Cytokine-mediated<br />

immunotherapy with or without donor leukocytes<br />

for poor risk acute myeloid leukemia relapsing after<br />

allogeneic bone marrow transplantation. Bone Marrow<br />

Transplant 1995; 16:133-7.<br />

205. Gurman G, Arslan O, Koc H, Akan H. Donor leukocyte<br />

infusion for relapsed ANLL after allogeneic BMT<br />

and the use of interferon alpha to induce graft-versusleukemia<br />

effect. Bone Marrow Transplant 1996;<br />

18:825-6.<br />

206. Carella AM, Frassoni F, Melo J, et al. New insights in<br />

biology and current therapeutic options for patients<br />

with chronic myelogenous leukemia. <strong>Haematologica</strong><br />

haematologica vol. 85(suppl. to n. 12):December 2000


Self-renewal and differentiation<br />

Marrow and blood hematopoietic cells are heterogeneous<br />

and belong to different lineages at different<br />

stages of maturity. The structural and functional<br />

integrity of the hematopoietic system is maintained by<br />

stem cells that, by definition, comprise a relatively small<br />

cell population, located mainly in the bone marrow,<br />

which can (i) undergo self-renewal to produce stem<br />

cells or (ii) differentiate to produce progeny which is<br />

progressively unable to self-renew, irreversibly committed<br />

to one or other of the various hematopoietic linreview<br />

Ex vivo expansion of hematopoietic<br />

cells and their clinical use<br />

haematologica 2000; 85(supplement to no. 12):92-116<br />

Correspondence: Sante Tura, M.D., Istituto di Ematologia ed<br />

Oncologia Medica "L. & A. Seragnoli", Policlinico S. Orsola,<br />

via Massarenti 9, 40138 Bologna, Italy<br />

MASSIMO AGLIETTA,* FRANCESCO BERTOLINI,° CARMELO<br />

CARLO-STELLA, # ARMANDO DE VINCENTIIS, @ LUIGI LANATA,^<br />

ROBERTO M. LEMOLI, § ATTILIO OLIVIERI,** SALVATORE<br />

SIENA,°° PAOLA ZANON,^ SANTE TURA §<br />

*Hematology Oncology, Ospedale Mauriziano, University of<br />

Turin, Turin; °Department of Oncology, IRCCS Fondazione<br />

Maugeri, Pavia; # Hematology and Bone Marrow Transplantation,<br />

University of Parma, Parma; @ Dompé Biotec SpA, Milan;<br />

^Amgen Italia SpA, Milan; § Institute of Hematology and Medical<br />

Oncology "L. & A. Seragnoli", University of Bologna,<br />

Bologna; **Division of Hematology, University of Ancona,<br />

Ancona; and °°Istituto Clinico Humanitas, Milan, Italy<br />

Background and Objectives. Hematopoietic stem cells<br />

are being increasingly used for treatment of malignant<br />

and nonmalignant disorders. Various attempts have<br />

been made in recent years to expand and manipulate<br />

these cells in order to increase their therapeutic potential.<br />

A Working Group on Hematopoietic Cells has analyzed<br />

the most recent advances in this field.<br />

Evidence and Information Sources. The method used<br />

for preparing this review was an informal consensus<br />

development. Members of the Working Group met three<br />

times, and the participants at these meetings examined<br />

a list of problems previously prepared by the<br />

chairman. They discussed the single points in order to<br />

achieve an agreement on different judgments, and<br />

eventually approved the final manuscript. Some<br />

authors of the present review have been working in the<br />

field of stem cell biology, processing and transplantation,<br />

and have contributed original papers in peerreviewed<br />

journals. In addition, the material examined in<br />

the present review includes articles and abstracts published<br />

in journals covered by the Science Citation<br />

Index® and Medline®.<br />

State of Art. Over the last decade, recombinant DNA<br />

technology has allowed the large scale production of<br />

cytokines controlling the proliferation and differentiation<br />

of hemo-lymphopoietic cells. Thus, in principle, ex<br />

vivo manipulation of hemopoiesis has become feasible.<br />

The present review covers three major area of interest<br />

in experimental and clinical hematology: manipulation<br />

of hematopoietic stem/progenitor cells, cytotoxic effector<br />

cells and antigen presenting dendritic cells. Preliminary<br />

data demonstrate the possibility of using, in a<br />

clinical setting, ex vivo expanded hematopoietic cells<br />

with the aim of reducing, and perhaps abrogating, the<br />

myelosuppression after high-dose chemotherapy. Concurrently,<br />

other important potential applications for ex<br />

vivo manipulation of hematopoietic cells have recently<br />

been investigated such as the generation and expansion<br />

of cytotoxic cells for cancer immunotherapy, and<br />

the large scale production of professional antigen presenting<br />

cells capable of initiating the process of<br />

immune response.<br />

Conclusions and Perspectives. Present and future challenges<br />

in this field are represented by the expansion of<br />

true human stem cells without maturation, to extend<br />

this strategy to allogeneic stem cell transplantation as<br />

well as the manipulation of cycling of primitive progenitors<br />

for gene therapy programs. The selective outgrowth<br />

of normal progenitor cells over neoplastic cells<br />

to achieve tumor-free autografts may ameliorate the<br />

results of autologous transplantation. The selective production<br />

of cellular subsets to manipulate the graft versus-host<br />

and graft versus-tumor effects and anti-tumor<br />

vaccination strategies may be important to improve<br />

cellular adoptive immunotherapy.<br />

©1998, Ferrata Storti Foundation<br />

Key words: hematopoietic stem cells, bone marrow, cord blood,<br />

dendritic cells, peripheral blood, allogeneic transplantation<br />

Hematopoietic stem cells<br />

Following the discovery that bone marrow transplantation<br />

could be used to rescue irradiated mice, the<br />

identification and characterization of the hematopoietic<br />

stem cell has become essential in order to achieve<br />

new developments in stem cell expansion and transplantation<br />

(SCT). 1 The potential for using stem cells as<br />

vehicles for gene therapy has further increased the<br />

efforts of a number of research groups working on stem<br />

cell identification, characterization, cloning and manipulation.<br />

2<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

93<br />

eages, and able to generate clones of up to 10 5 lineagerestricted<br />

cells that mature into specialized cells. 3<br />

Although in recent years, (i) the development of in vitro<br />

and in vivo assays for hematopoiesis, (ii) the identification<br />

and characterization of hematopoietic growth<br />

factors, and (iii) the development of strategies for<br />

enriching hematopoietic cells have expanded our knowledge<br />

and understanding of hematopoiesis, the definition<br />

of the stem cell, originally proposed by Lajtha 4 and<br />

McCulloch, 5 has not substantially changed.<br />

In addition to self-renewal and differentiation, a number<br />

of properties are ascribed to hematopoietic stem<br />

cells, including a high migratory potential, the ability to<br />

undergo asymmetric cell divisions, the capacity to exist<br />

in a mitotically quiescent form and extensively regenerate<br />

the different cell types that constitute the tissue<br />

in which they exist. 6<br />

The issues of asymmetrical and symmetrical cell divisions<br />

and the regulation of self-renewal/differentiation<br />

process are crucial when analyzing stem cell behavior<br />

and the potential for stem cell manipulation. Asymmetric<br />

cell divisions produce one differentiated daughter<br />

(progenitor cell) and another daughter that is still a stem<br />

cell (Figure 1A). When all cell divisions are necessarily<br />

asymmetric and controlled by cell-intrinsic mechanisms,<br />

no amplification of the stem cell size is possible. 7 Asymmetric<br />

divisions are referred to unequally distributed<br />

transcription factors in daughter cells, 8,9 and have been<br />

shown to be possible in hematopoietic progenitors by<br />

clone-splitting experiments. 10 Symmetric cell divisions<br />

produce either two progenitor cells or two stem cells<br />

according to a 0.5 probability of self-renewing versus<br />

differentiative divisions (Figure 1B). In this case, it can be<br />

assumed that the size of the stem cell pool can be modified<br />

by factors affecting the 0.5 probability value, i.e.,<br />

factors that control the probability of self-renewing versus<br />

differentiative divisions. 7 A third model postulates<br />

that individual cell divisions can be, but not necessarily<br />

are, asymmetric with respect to daughter cell fate (Figure<br />

1C). This model also implies that daughters behave<br />

differently due to different local environments. Although<br />

it is not known whether a single cell can switch from an<br />

asymmetric to a symmetric mode of cell division, available<br />

evidence in the hematopoietic stem cell system<br />

favors a predominance of symmetric cell divisions.<br />

The decision of a stem cell to either self-renew or differentiate<br />

as well as the selection of a specific differentiation<br />

lineage by a multipotent progenitor during commitment<br />

have been proposed to be regulated according<br />

to either stochastic or deterministic (inductive) models.<br />

Based on computer simulation and the distributions of<br />

colony-forming units in spleen (CFU-S) in individual<br />

spleen colonies, stochastic models postulate that the<br />

decision of a stem cell to self-renew (birth) or to differentiate<br />

(death) is randomly regulated by a probability<br />

parameter "p" which is equal to 0.5 in steady-state conditions.<br />

11 Deterministic models postulate the existence<br />

of lineage-specific anatomic niches that direct the differentiation<br />

of uncommitted progenitors. 12 There is experimental<br />

evidence suggesting that the hematopoietic system<br />

may employ both stochastic and deterministic<br />

strategies, probably depending upon the stage of lineage<br />

differentiation.<br />

Based on a number of studies performed in the last<br />

three decades, the regulation of self-renewal, commitment,<br />

proliferation, maturation, and survival can be<br />

assumed to reflect highly integrated processes under control<br />

of extracellular mechanisms, including regulatory<br />

molecules and microenvironment, as well as intracellular<br />

mechanisms, including protooncogenes, cell cycle regulators,<br />

tumor suppressor genes, transcription factors.<br />

Regulatory molecules include positive (growth factors)<br />

and negative (interferons, TGF-, MIP-1) factors which<br />

interact in complex ways (synergism, recruitment, antagonism).<br />

13,14 Molecules that maintain the stem cell state<br />

are beginning to be identified. These include ligands of<br />

the Notch family receptors that act from outside the cell<br />

as regulators of proliferation or maintenance of the<br />

undifferentiated state, 15 and factors like PIE-1 that act<br />

from within the cell. 16 However, despite the efforts which<br />

have been devoted to elucidating the issue of self-renewal<br />

control, no factors have yet been identified that are<br />

Figure 1. Possible patterns<br />

of stem cell division. “S”<br />

indicates a stem cell; “C”<br />

indicates a committed progenitor<br />

cell.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


94<br />

M. Aglietta et al.<br />

capable of maintaining self-renewing divisions and the<br />

molecular basis of self-renewal capacity remains to be<br />

elucidated. Growth factors so far identified more probably<br />

act as regulators of proliferation and survival. Theoretically,<br />

growth factors and cell-cell interactions can<br />

influence the outcome of fate decisions by stem and<br />

progenitor cells in a selective or instructive manner.<br />

According to selective mechanisms, the stem cell commits<br />

to a particular lineage independently of the growth<br />

factors and the factors act to control survival and proliferation<br />

of committed progenitors. In instructive mechanisms,<br />

growth factors cause the stem cell to choose one<br />

lineage at the expense of others. The relative contribution<br />

of these two mechanisms to hematopoietic regulation<br />

remains controversial, but experimental evidence<br />

suggests that at least some subsets of stem/progenitor<br />

cells can be instructed by growth factors to choose one<br />

differentiation pathway at the expense of others. 17 In the<br />

absence of still unidentified instructive signals, it can be<br />

hypothesized that environmental signals may act by<br />

increasing or decreasing the probability of choosing a<br />

particular fate, rather than promoting or repressing it in<br />

an all-or-none manner. 2<br />

Although growth factors play a key role in stem/progenitor<br />

cell proliferation and differentiation it seems<br />

improbable that hematopoiesis is regulated by a random<br />

mix of growth factors and responsive cells. Indeed, it is<br />

likely that regulatory molecules and localization phenomena<br />

within marrow stroma are required to sustain<br />

and regulate hematopoietic function. 18 Stromal cells of<br />

the hematopoietic microenvironment provide the physical<br />

framework within which hematopoiesis occurs. They<br />

play a role in directing the processes by synthesizing,<br />

sequestering or presenting growth-stimulatory and<br />

growth-inhibitory factors, and also express a broad repertoire<br />

of adhesion molecules which mediate specific interactions<br />

with hematopoietic stem/progenitor cells. 19 Differential<br />

expression of adhesion molecules could cause<br />

different stem cell subsets to home to different marrow<br />

microenvironments capable of differently affecting selfrenewal.<br />

While much progress has been made in identifying<br />

cytokines and stromal factors, little is known about intracellular<br />

mechanisms regulating hematopoietic stem/progenitor<br />

cells self-renewal and differentiation. 20 Structure-function<br />

analysis of growth factor receptors as well<br />

as identification of novel signal transduction molecules<br />

have provided new insights into the processes involved in<br />

signal transmission pathways. Post-translational modifications<br />

of pre-existing proteins, in particular tyrosine<br />

phosphorylation, play a key role in transmitting signals<br />

and thereby linking extracellular signals to the activation<br />

of nuclear effector molecules which govern gene expression.<br />

20 Accumulating evidence points to transcription factors<br />

such as AML-1, Ikaros, SCL/Tal-1, Rbtn-2, Tan-1,<br />

GATA-2, and HOX homeobox genes as important regulators<br />

of these processes. Overexpression of HOXB4 in<br />

murine bone marrow cells markedly increases the regenerative<br />

potential of long-term repopulating cells and<br />

causes an expansion in clonogenic progenitor cell numbers,<br />

without altering their ability to differentiate normally<br />

into mature myeloid, erythroid and lymphoid cells. 21<br />

In contrast, overexpression of HOXB3 causes defective<br />

lymphoid differentiation and progressive myeloproliferation.<br />

22 The glucocorticoid receptor, in combination with<br />

an activated receptor tyrosine kinase, seems to be a key<br />

regulator of erythroid self-renewal. 23 Shc overexpression<br />

increases GM-CSF sensitivity and prevents apoptosis of<br />

the GM-CSF-dependent acute myeloid leukemia cell line<br />

GF-D8, thus suggesting that Shc is an important regulator<br />

of cell survival and proliferation. 24 Different levels of<br />

protein kinase C modulate progenitor cell phenotype by<br />

favoring myelomonocytic or eosinophil differentiation. 25<br />

Recently, it has been shown that telomerase expression<br />

correlates with hematopoietic self-renewal potential.<br />

Hematopoietic stem cells show decreasing telomere<br />

length with increasing age. 26 Thus, telomerase may regulate<br />

self-renewal capacity by reducing the rate of DNA<br />

shortening. Overall, intracellular mechanisms of hematopoietic<br />

control result in the repression or de-repression<br />

of lineage-specific genes regulating growth factor<br />

responsiveness and/or proliferation potential. 27 The exact<br />

knowledge of these mechanisms will greatly modify our<br />

approach to stem/progenitor cell manipulation.<br />

In summary, stem and progenitor cell behavior is the<br />

result of highly integrated phenomena based on extracellular<br />

signals triggering intracellular transduction phenomena.<br />

The properties of self-renewal and differentiation<br />

give stem cells their remarkable ability to repopulate<br />

the hematopoietic tissue of lethally irradiated or<br />

genetically defective recipients. Understanding the<br />

interplay between extracellular and intracellular regulatory<br />

factors in controlling lineage determination<br />

remains an important challenge for the future clinical<br />

use of hematopoietic cells.<br />

Stem cell antigen(s)<br />

CD34 is a surface glycophosphoprotein expressed on<br />

early lympho-hematopoietic stem and progenitor cells,<br />

small-vessel endothelial cells, as well as embryonic<br />

fibroblasts. 28 CD34 + hematopoietic cells are morphologically<br />

and immunologically heterogeneous and functionally<br />

characterized by the in vitro capability to generate<br />

clonal aggregates derived from early and late<br />

progenitors and the in vivo capacity to reconstitute the<br />

myelo-lymphopoietic system in a myeloablated host. 29<br />

The CD34 + cell population contains virtually all the<br />

myeloid and lymphoid progenitors as well as a small<br />

subset of cells that can initiate and maintain stromal<br />

cell-supported long-term cultures. Expression of the<br />

CD34 marker has dominated attempts to isolate, purify<br />

and characterize human hematopoietic stem cells by a<br />

variety of immunologic means.<br />

Several monoclonal antibodies (MoAbs) assigned to<br />

the CD34 cluster identify a transmembrane glycoprotein<br />

antigen of 105-120 kd expressed on 0.5-2% normal BM<br />

cells, 0.01-0.1% peripheral blood cells and 0.1-0.4%<br />

cord blood cells. 30 The function of the CD34 antigen is<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

95<br />

not yet known, although it seems that CD34 is involved<br />

in stem/progenitor cell localization/adhesion in the marrow.<br />

31 CD34 antigen expression is associated with the<br />

concomitant expression of several markers, including<br />

the lineage non-specific markers Thy1, CD38, HLA-DR,<br />

CD45RA, CD71 as well as T-lymphoid, B-lymphoid,<br />

myeloid and megakaryocytic differentiation markers. 30<br />

Analysis of the expression of CD38, Thy-1, CD71, the<br />

isoforms of CD45, and uptake of rhodamine-123 have<br />

resulted in a consensus stem cell phenotype which is<br />

CD34 bright , Thy-1 + , CD38 – , CD45RA – , rh-123 dull , Hoechst<br />

33342 dull , Lin – . CD34 + cells also express receptors for a<br />

number of growth factors classified as tyrosine kinase<br />

receptors, such as the stem cell factor receptor (SCF-R)<br />

or the stem cell tyrosine kinase receptors (STK), and<br />

hematopoietic receptors, not containing a tyrosine<br />

kinase domain. 32 Tyrosine kinase receptors are of particular<br />

relevance since their ligands might represent new<br />

factors able to selectively control stem cell self-renewal,<br />

proliferation and differentiation. 33 Recently, CD34 –<br />

cells have been shown to have functional characteristics<br />

associated with stem cells and differentiate, in vivo,<br />

to CD34 + cells. 34<br />

Stem/progenitor assays to evaluate<br />

engraftment potential<br />

Different types of progenitors can be measured directly<br />

by multiparameter phenotyping of CD34 + cells or subpopulations.<br />

Although this approach has the significant<br />

advantage that the results may be quickly available and<br />

can be used to guide clinical decisions, correlations<br />

between progenitor cell phenotype and functional activity<br />

are not yet refined enough to be clinically applicable.<br />

In addition, although CD34 antigen is expressed by virtually<br />

all progenitor cells, the percentage of CD34 + cells<br />

with clonogenic activity in vitro ranges from 10 to 50%.<br />

Non-clonogenic CD34 + cells include lymphoid progenitors<br />

as well as subsets of cells which are unresponsive to conventional<br />

growth factors and might require the presence<br />

of still unknown factors able to activate stem cell specific<br />

genes. 14 Figure 2 shows a schema of the cellular organization<br />

of hematopoiesis based on in vitro and in vivo<br />

functional properties of progenitor cells.<br />

Recently, a new human hematopoietic cell, termed the<br />

SCID repopulating cell (SRC), that is capable of extensive<br />

proliferation and multilineage repopulation of the bone<br />

marrow of non-obese diabetic (NOD)/SCID mice has been<br />

identified. 35,36 The SRCs which are detected exclusively in<br />

the CD34 + CD38 – cell fraction have been shown to be<br />

biologically distinct from CFC and most LTC-IC. 35,36 With<br />

the exception of transplantation of human cells into<br />

immunodeficient mice, the identification of putative<br />

human stem cells has essentially relied on in vitro assays.<br />

Short-term in vitro assay systems require appropriate<br />

nutrients and growth factors and are particularly suitable<br />

for measuring quantitative changes of the different<br />

progenitor cell types as well as for evaluating growth<br />

factor responsiveness or investigating differential effects<br />

of regulatory molecules on progenitors at different<br />

stages of differentiation or on different hematopoietic<br />

pathways. 37 Short-term assays are not suitable for analyzing<br />

self-renewal or interactions of hematopoietic<br />

progenitors with stromal cells.<br />

By using the long-term culture (LTC) technique, a sustained<br />

production of myeloid cells can be readily achieved<br />

in vitro, provided that a stromal layer is present, when<br />

marrow (or blood) is placed in liquid culture at relatively<br />

high cell concentration, with appropriate supplements,<br />

temperature and feeding conditions. 37<br />

The LTC system, based on the re-establishment in vitro<br />

of the essential cell types and mechanism responsible<br />

for the localized and sustained production of hematopoietic<br />

cells in the marrow in vivo, offers an approach able<br />

to investigate not only the proliferative and differentiative<br />

events but also self-renewal of any clonogenic cell<br />

types. A 5- to 8-week time period between initiating<br />

cultures and assessing clonogenic progenitor numbers<br />

allows a very primitive, self-renewing human cell, the<br />

Figure 2. Cellular organization<br />

of hematopoiesis.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


96<br />

M. Aglietta et al.<br />

so-called long-term culture-initiating cell (LTC-IC) to be<br />

quantified. 38,39 Limiting dilution assays allow the frequency<br />

of LTC-IC and their proliferative potential (number<br />

of CFU-GM generated by each LTC-IC) to be calculated.<br />

Another assay system, the cobblestone area-forming<br />

cell (CAFC), uses a pre-formed stroma as a support<br />

for hematopoiesis. 40 In this system, the primitive cells<br />

are measured directly by their ability to form characteristic<br />

colonies of cells resembling cobblestones.<br />

With the possibility of studying not only the differentiation<br />

but also the self-renewal of primitive progenitors,<br />

the LTC system will play an increasingly important role in<br />

the design and assessment of new strategies involving<br />

the genetic engineering of hematopoietic cells and marrow<br />

stromal cells. Hematopoietic cells that can generate<br />

active hematopoiesis for weeks in vitro or months in vivo<br />

after transplantation are considered stem cells. This seems<br />

a clinically useful criteria, because it characterizes those<br />

cells which are important for sustained hematopoietic<br />

recovery following SCT. However, it reflects an oversimplification<br />

of the rather complex process of hematopoietic<br />

function. In fact, the ability of a cell to provide longterm<br />

hematopoietic activity can either be due to a long<br />

period of quiescence after the initiation of the culture or<br />

be a function of the probability of stem cell self-renewal<br />

which influences the long-term survival of stem cell<br />

clones. 41 Thus, the number of primitive cells measured in<br />

an LTC assay will be the product of the number of stem<br />

cells present at the onset of the culture and the probability<br />

of stem cell self-renewal. Although LTC assays will<br />

likely predict the in vivo repopulating activity of the graft,<br />

the clinical definition of a stem cell does not consider<br />

those stem cells that differentiate and die soon after<br />

transplantation or initiation of a culture.<br />

Preparation of hematopoietic cells for<br />

ex vivo expansion<br />

In the vast majority of cell culture systems, the presence<br />

of inhibitory mature and accessory cells limits the<br />

degree of ex vivo expansion of the progenitor cell compartment.<br />

Thus, a higher production of total cells, clonogenic<br />

cells and more immature hematopoietic progenitors<br />

has been observed when purified progenitor cells<br />

(namely CD34 + cells) rather than the whole BM, cord<br />

blood. 42 or peripheral blood stem cell (PBSC) collections 43<br />

are cultured ex vivo. Haylock et al. 43 selected and cultured<br />

1,000 CD34 + cells in presence of a 10-fold excess<br />

of contaminating CD34 – , CD3 – , CD14 – cells and found no<br />

differences in total cell production after 14 days of culture,<br />

as compared to the production of 1,000 CD34 + cells<br />

grown alone. However, when CD34 + cells were mixed<br />

with increasing concentrations of CD3 + CD14 + cells, a<br />

marked decrease in the total cell output was observed<br />

after two weeks of culture suggesting an inhibitory activity<br />

of monocytes and T-cells. Despite the lack of information<br />

on CFU-C production, these results point out that<br />

the purity of the starting population is an important variable<br />

and it was recommended that at least 50% of cells<br />

should be CD34 + in the initial cellular input. 43 There are<br />

Table 1. Hematopoietic cell separation systems.<br />

Recovery Purity Clinical grade<br />

(% of initial cells) (% CD34+ cells) device<br />

Immunomagnetic beads<br />

Negative selection 20-50 20-60 Yes<br />

Positive selection 30-60 50-90 Yes<br />

MACS >80 >90 Under<br />

evaluation<br />

Panning 30-50 40-70 No<br />

Avidin-biotin 40-60 50-90 Yes<br />

immunoabsorption<br />

FACS (high-speed cell sorting) 30-50 >95 Yes<br />

also practical advantages in using a purified stem cell<br />

population as starting material for ex vivo manipulation,<br />

such as the ease of cell handling and the amount of<br />

cytokines consumed in the culture. Other variables that<br />

play an important role in stem cell expansion, which will<br />

be discussed in detail in the following paragraphs are:<br />

initial cell density, different cytokines used and their concentration,<br />

the presence of stromal cells, the composition<br />

of the culture medium and the refeeding schedule.<br />

Conversely, it has been recently demonstrated 44 that<br />

CD34 + cells can be safely and efficiently processed after<br />

cryopreservation suggesting that the availability of fresh<br />

hematopoietic cells may not be an essential prerequisite<br />

for ex vivo expansion. Moreover, whereas the majority of<br />

preclinical and clinical studies 45 have attempted to optimize<br />

the expansion of highly enriched CD34 + cells, under<br />

certain circumstances it may be advisable to select earlier<br />

subfractions of progenitor cells such as CD34 + DR –<br />

cells in chronic myelogenous leukemia (CML) or CD34 +<br />

Thy-1 + lin – cells in multiple myeloma (MM). In these diseases<br />

the CD34 + cell population is still contaminated, to<br />

various degrees, by malignant cells; 46-49 therefore, isolation<br />

of primitive progenitors prior ex vivo expansion may<br />

provide a starting cell population with a high proliferative<br />

potential free of tumor cells.<br />

Methods for hematopoietic progenitor cell<br />

(HPC) enrichment<br />

Several methods have been proposed for purification of<br />

HPC (Table 1). Their final target is a cell population with<br />

optimal purity, viability and high proliferative potential<br />

obtained by means of a low cost, rapid and simple separation<br />

technique. Early attempts toward the purification<br />

of HPC were based on the cell physical properties. Density-gradient<br />

centrifugation, velocity sedimentation and<br />

elutriation are methods that separate cells based on cell<br />

size and buoyant density. More recently, immunologic<br />

selection techniques which take advantage of the expression<br />

of specific antigens on HPC membrane, have allowed<br />

a much better degree of enrichment. Specifically, the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

97<br />

demonstration of the presence of the CD34 antigen on<br />

HPC 28 has led to a number of positive selection systems<br />

which use MoAbs to purify hematopoietic precursors.<br />

Alternatively, CD34 + cells can be enriched by depletion<br />

of CD34 – accessory and mature cells.<br />

Fluorescence activated cell sorter (FACS)<br />

Flow cytometry can be used to separate HPC from a<br />

heterogeneous population after incubation of cells with<br />

fluorochrome-conjugated MoAbs directed to cell-surface<br />

markers. Moreover, multiparameter enrichment can<br />

be obtained by combining physical properties such as<br />

cell size and cytoplasmic granularity and intracellular<br />

characteristics indicating cellular function (e.g. propidium<br />

iodide to determine cell viability; rhodamine-123 to<br />

assess metabolic quiescence and nucleic acid dyes to<br />

evaluate cycling status). This cell sorting technique can<br />

yield a highly purified (> 99%) cell population combining<br />

positive and negative markers for HPC using clinically-graded<br />

MoAbs. The main criticisms to the use of<br />

flow cytometry for selecting large numbers of cells are<br />

the low recovery of target cells and the length of time<br />

required to process the whole BM harvest or the leukapheresis<br />

products. The development of multiparameter<br />

high-speed cell sorting has been described and recently<br />

upgraded for clinical use. 50,51 Viable cells have been<br />

sorted at rates as high as 40,000 cells/sec as compared<br />

to 2,000-5,000 cells/sec of commercially available cell<br />

sorters. Thus, the sample processing time can now be<br />

reduced to 8-12 hours. The sorted cell fraction also<br />

maintains its hematopoietic potential based on the presence<br />

of CFU-C, more immature CAFC and long-term<br />

repopulating cells in mice. 51 Presently, selection of HPC<br />

(i.e. CD34 + Thy-1 + lin – cells) from clinical samples is<br />

directed toward the purification of cell populations<br />

highly enriched for HPC free of contaminating malignant<br />

cells in myeloma patients. 47<br />

Panning<br />

Anti-CD34 MoAbs bound to the bottom of cell culture<br />

flasks have been used to select CD34 + cells. 52 The<br />

target cell population present in a heterogeneous cell<br />

suspension is blocked on the plastic surface while CD34 –<br />

cells remain in the supernatant and can be easily eliminated.<br />

Despite the good results reported, the availability<br />

of more efficient methods of cell separation have<br />

made this technique largely redundant.<br />

Immunomagnetic systems<br />

A variety of magnetic cell-separation methods have<br />

been described. 53 Some of these systems are commercially<br />

available and have been used in clinical trials. The<br />

main differences between the currently used magnetic<br />

cell-separation methods are the composition and size of<br />

the magnetic particles used for labeling the cells and the<br />

separation process. Superparamagnetic beads can be<br />

equally used for negative and positive cell separation<br />

depending on the specificity of MoAbs. The rosetted target<br />

cells can be easily isolated from unlabeled cells by<br />

a magnet applied on the outer wall of the test tube or<br />

Table 2. High efficiency of Mini-MACS separation system for<br />

the enrichment of BM or circulating CD34 + cells.<br />

Enrichment<br />

Source Pre Post Recovery CE* LTC-IC<br />

(%) (%) (%) (x10 4 CD34 + )<br />

BM 2.3±1 97±3 88±9 3.6±0.3 62.5±54<br />

PB 0.7±0.4 98.9±1 90±8 3.9±0.4 48.2±35<br />

*Abbreviations: CE, clonogenic efficiency; BM, bone marrow; PB, peripheral blood.<br />

The results are expressed as mean±SD.<br />

blood bag. Large magnetic beads (diameter >0.5 µm)<br />

have been used clinically for the purging of neuroblastoma<br />

and lymphoma cells from stem cell harvests prior<br />

to autologous transplantation. 53,54 Immunomagnetic<br />

beads coupled with anti-CD34 MoAb can be used for<br />

positive selection of HPC. 55 However, before the clinical<br />

use of the enriched cell fraction, the cell-bound particles<br />

must be removed to avoid damage to the cells<br />

and/or toxic events to the patient. Beads can be released<br />

using chymopapain or a peptide competing with the<br />

CD34 Ab. In alternative, HPC can be enriched by negative<br />

depletion of mature and accessory cells targeting<br />

lineage-specific antigens.<br />

More recently, the magnetic cell-sorting (MACS) system<br />

has been proposed as an efficient and more manageable<br />

alternative to flow cytometry for cell separation.<br />

56 It uses colloidal-sized superparamagnetic particles<br />

made of dextran and iron oxide with 60 nm of<br />

diameter. The use of very small beads minimizes unspecific<br />

binding and allows the efficient isolation of rare<br />

cells. In addition, the magnetic particles are readily<br />

internalized by the labeled cells without affecting their<br />

physical, phenotypic and functional capacity. 57 Table 2<br />

reports the results of a large number of experiments<br />

(=14) comparing the efficiency of the Mini-MACS system<br />

for selecting CD34 + cells from two different cellular<br />

sources (R.M.L., unpublished observations).<br />

High-affinity chromatography based on<br />

avidin-biotin immunoabsorption<br />

This technique relies on the high affinity between the<br />

protein avidin and the vitamin biotin whose interaction<br />

has an extremely high dissociation constant (= 10 -15 M).<br />

In this system, a heterogeneous cell population is incubated<br />

with a biotinylated antibody to the CD34 antigen.<br />

The cell mixture is then passed through a disposable column<br />

containing avidin-coated polyacrylamide beads.<br />

CD34 + cells are retained on the column due to the high<br />

affinity binding of biotin to avidin while negative cells<br />

are washed away. Target cells are then recovered by<br />

mechanical agitation of the column which disrupts the<br />

antibody-antigen link. Thus, bound cells are eluted from<br />

the column mainly free of antibody. An automated version<br />

of the device controlled by a computer which guar-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


98<br />

M. Aglietta et al.<br />

antees reproducibility and reduces risks of operator errors<br />

has been developed and used in the setting of autologous<br />

and allogeneic stem cell transplantation.<br />

46, 58<br />

Dynamic systems<br />

In addition to positive or negative selection of CD34 +<br />

cells, effective ex vivo expansion of hematopoietic progenitor<br />

cells and, perhaps, putative stem cells can also be<br />

achieved in the presence of unselected cell populations.<br />

In this case the use of dynamic perfusion cultures is<br />

strictly required. As stated above, the production of<br />

inhibitory factor(s) by mature and accessory cells rather<br />

than the availability of growth promoting factors, is<br />

probably the main limitation to successful stem cell<br />

expansion. For instance, it is well known that suboptimal<br />

cell expansion occurs when CD34 + cells are cultured<br />

at concentrations exceeding 10 4 cells/mL. Moreover, the<br />

presence of stromal feeder-layer cells seems to be<br />

important for effective BM stem cell expansion induced<br />

by exogenous cytokines. Therefore, an artificial capillary-perfusion<br />

system (Bioreactor) in which nutrients<br />

consumed by proliferating cells are continuously replenished<br />

by exchange of the nutrient-depleted medium<br />

with fresh culture medium, has been tested for ex vivo<br />

expansion of hematopoietic cells co-cultivated with<br />

stroma cells or stromal cell lines. 59 Cytokines such as IL-<br />

3, IL-6 and GM-CSF have been added to the culture to<br />

optimize cell expansion and to provide growth factors<br />

which are not produced by stromal cells (i.e. IL-3). In this<br />

system, cultured cells are confined in a small compartment<br />

separated from a large medium reservoire. Medium<br />

exchange is optimized when there is a maximal<br />

membrane surface area per unit volume across which<br />

medium and nutrients can pass by diffusion. This is best<br />

achieved by a capillary-perfusion module. Three important<br />

requirements for optimal expansion of hematopoietic<br />

cells are: the capillary porous size, the capacity of<br />

supporting the attachment of stromal cells by the module<br />

and the minimal cell activation by the materials of<br />

the module. In fact, mature myeloid cells such as macrophages<br />

release ,upon surface activation, tumor necrosis<br />

factor(s) (TNFs), interferon(s) (IFNs) and other substances<br />

which negatively affect stem cell expansion.<br />

Bioreactors have induced a remarkable expansion of<br />

committed hematopoietic progenitor cells coupled with<br />

a modest increase in the number of LTC-IC, 59 a population<br />

of primitive cells which correlates most positively<br />

with the long-term reconstituting capacity of autologous<br />

and allogeneic grafts.<br />

Ex vivo expansion of myeloid<br />

progenitor cells<br />

Ex vivo expansion of hemopoietic progenitors might<br />

result in:<br />

• amplification of the population of committed progenitors<br />

due to an extensive, although controlled, differentiation<br />

process;<br />

• amplification of the stem cell pool through extensive<br />

self-renewal of the early progenitor cell population.<br />

Obviously, both processes can take place simultaneously<br />

mimicking, in vitro, the complex interplay of regulatory<br />

mechanisms that allows hemopoiesis in vivo. The<br />

latter situation, so far, has never been obtained in vitro,<br />

whereas different approaches have permitted the first<br />

goal to be reached (at least, to a certain extent) and<br />

some recent data suggest that relevant self-maintaining<br />

processes can be triggered.<br />

The clinical relevance of extended differentiation versus<br />

self-renewal is obviously different. The induction of<br />

an increased in vitro production of committed progenitors<br />

might hasten the early phases of hemopoietic reconstitution<br />

which occur after myeloablative treatment and<br />

stem cell transplantation. Moreover an increased number<br />

of infused cells might modulate graft versus-host<br />

disease (GVHD) intensity in the allogeneic setting. 60<br />

Although relevant, the potential clinical benefit of<br />

techniques allowing only committed progenitor cell<br />

expansion is outweighed by the possibility of triggering<br />

the self maintenance, and perhaps amplification, of early<br />

hemopoietic progenitors. In this situation, starting<br />

from a limited number of progenitors, long-term reconstitution<br />

of hematopoiesis might become feasible.<br />

Moreover, ex vivo manipulation of primitive hemopoietic<br />

cells could be performed under experimental conditions<br />

suboptimal for the growth of neoplastic cell contaminating<br />

autologous grafts. Thus, a purging effect<br />

could be obtained.<br />

Several attempts of in vitro expansion of hemopoietic<br />

progenitors have been published in the last years.<br />

Recent reviews 61, 62 summarize early experiences.<br />

The first studies on ex vivo generation of hematopoietic<br />

progenitors involved liquid culture in the presence<br />

of cytokines such as SCF, IL-1, IL-3, IL-6, G-CSF, GM-CSF<br />

and Epo. These experiences showed that a relevant<br />

increase (from 10 to 1,000 fold) of CD34 + cells and of<br />

committed progenitors can be obtained. The expansion<br />

of committed progenitors does not mean, however, that<br />

the long-term reconstitution of hemopoiesis is achievable.<br />

Indeed, several data support the concept that an<br />

uncontrolled commitment decreases the stem cell pool.<br />

Yonemura et al. 63 have reported that IL-3 or IL-1 abrogates<br />

the reconstituting ability of hematopoietic stem<br />

cells. Furthermore, Peters et al. 64 have demonstrated<br />

that ex vivo expansion of murine marrow cells with IL-<br />

3, IL-6, IL-11 and SCF leads to impaired engraftment in<br />

irradiated hosts.<br />

As indicated by Traycoff et al. 65 ex vivo expansion of<br />

hematopoietic cells using SCF, IL-1, IL-3 and IL-6 generates<br />

classes of cells possessing different levels of BM<br />

repopulating potential based on their cycling status.<br />

Along this line, Young et al. 66 correlated a higher proliferative<br />

capacity with a quiescent status after ex vivo<br />

expansion in the presence of SCF, IL-3, IL-6 and LIF.<br />

Taken together, these data suggest that cultures in the<br />

presence of cytokine combinations based upon SCF, IL-<br />

1 and IL-3 involve differentiation of BM or mobilized<br />

CD34 + cells entering the S phase. Different results were<br />

obtained by Di Giusto et al., 67 who found that ex vivo<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

99<br />

expanded cord blood CD34 + cells repopulated the marrow<br />

of immunodeficient mice as well as non-expanded<br />

cells. However, it must be remembered that cord blood<br />

is rich in hemopoietic progenitors 68 that have an<br />

increased proliferation potential. 69<br />

New strategies to induce the expansion of CD34 + cells<br />

with little (or no) differentiation might involve different<br />

approaches. The use of stirred suspension 70 or hollow<br />

fiber 71 bioreactors has been proposed in order to grow<br />

cells in a more physiologic environment, and inhibitors<br />

such as TGF- and MIP-1 have been the object of<br />

intense studies. Recently, MIP-1 has been found to<br />

exert a weak inhibitory effect on CD34 + CD38 – cells and<br />

to enhance the proliferation of CD34 + CD38 + cells,<br />

whereas TGF- strongly inhibits both cell populations.<br />

72,73 The most promising results, however, have<br />

been obtained with the recent introduction of FL and<br />

Tpo. FL, a recently discovered member of the class III<br />

tyrosine kinase receptor family, 74 is able to induce proliferation<br />

of very early hematopoietic progenitors that<br />

are non-responsive to other early acting cytokines, and<br />

to improve the maintainance of progenitors in vitro. 75-77<br />

This is also supported by the finding that FL significantly<br />

reduced the number of cultured cells undergoing<br />

apoptosis. 78 Analysis of the effects of 16 cytokines on<br />

CD34 + CD38 – cells showed that FL, SCF and IL-3 produced<br />

a 30-fold amplification of the input of LTC-IC. 79<br />

Yonemura et al. 80 compared FL- and SCF-driven ex vivo<br />

expansion. They reported that both cytokines, in combination<br />

with IL-11, enhanced the production of progenitors,<br />

but with different kinetics. In fact, the maximal<br />

expansion by FL required a longer incubation than with<br />

SCF. Interestingly, in these studies the combination of<br />

SCF/IL-11, together with IL-3, reduced the ability of cultured<br />

cells to reconstitute hematopoiesis in irradiated<br />

hosts. 80 Other recent data have compared the effect of<br />

FL and SCF. FL acts as a self-renewal or proliferation/expansion<br />

signal for CD34 + -low cells while the<br />

effect of SCF is more likely to transduce a differentiation<br />

signal, resulting in more rapid repopulation at the<br />

expense of cell expansion. 81 Gene transfer studies in<br />

mice have also demonstrated that FL maintains the ability<br />

of human CD34 + cells to sustain long-term hematopoiesis.<br />

In fact, incubation of CD34 + cells with FL before<br />

transduction was associated with long-term provirus<br />

expression, whereas provirus expression declined in<br />

recipients of CD34 + cells transduced in the absence of<br />

FL. 82 The expansion ex vivo of early progenitors seems to<br />

be affected at the single cell level by changes in cytokine<br />

concentrations. In a recent paper by the Vancouver<br />

group, 83 maximal LTC-IC expansion was obtained in the<br />

presence of 30 times more FL, SCF, IL-3, IL-6 and G-CSF<br />

than could concomitantly stimulate the near-maximal<br />

amplification of CFC.<br />

Tpo, the ligand of the mpl receptor expressed on both<br />

early and committed hematopoietic progenitors, 84 is<br />

known to support megakaryocytopoiesis. 85 Moreover, it<br />

has been shown to be capable of enhancing ex vivo<br />

expansion of early/committed progenitor cells. As single<br />

factors, FL and Tpo stimulated a net increase of LTC-IC<br />

generated from CD34 + CD38 – cells within 10 days. 79 Furthermore,<br />

as demonstrated in mice recipients of BM<br />

cells transduced with the mpl receptor, 86 Tpo does not<br />

induce lineage-restricted commitment of mpl-receptor<br />

positive pluripotent progenitors but permits their complete<br />

erythroid and megakaryocytic differentiation. Tpo<br />

has also been found to increase the multilineage growth<br />

of CD34 + CD38 – cells from 3%, in absence of the<br />

cytokine, up to 40% when Tpo is added to SCF and FL. 87<br />

The presence of additional cytokines such as IL-3, IL-6<br />

and Epo does not significantly enhance clonal growth<br />

above that observed in response to Tpo, SCF and FL. 87<br />

lnterestingly, the soluble form of Tpo receptor and G-CSF<br />

receptor directly stimulate the proliferation of primitive<br />

hematopoietic progenitors of mice in synergy with SCF<br />

and FL. 88<br />

A step toward extensive ex vivo amplification of early<br />

human progenitor cells has been reported by Piacibello<br />

et al. 89 They first demonstrated that IL-3 induces<br />

an early production of committed progenitors but is not<br />

able to sustain true self maintenance of hemopoietic<br />

stem cells even in the presence of other early acting<br />

cytokines (FL, Tpo, SCF). Afterwards, several combinations<br />

of early acting cytokines were tested for their ability<br />

to sustain long-term hematopoiesis in stroma free<br />

cultures. Among the various combinations tested on<br />

purified cord blood CD34 + cells, the mixture of Tpo + FL<br />

was found to be able to maintain early progenitors up<br />

to six months. 89 These data indicate the enormous<br />

potential of cord blood progenitors and the key role of<br />

Tpo and FL in the regulation of early hematopoiesis.<br />

However, several issues remain to be clarified:<br />

• is it true self-renewal or a slow differentiation of cord<br />

blood cells, which are rich in immature progenitors?<br />

• what is the in vivo repopulating capacity of ex vivo<br />

expanded cells?<br />

• is such expansion possible using CD34 + cells obtained<br />

from the marrow or peripheral blood of adult subjects?<br />

In this view, while a number of papers have already<br />

reported that committed progenitors can be generated<br />

and safely administered to transplant recipients, there<br />

are no reports on expansion of cells with long-term<br />

repopulating capacity in humans.<br />

Brugger et al. 45 reported the successful reconstitution<br />

of hematopoiesis in ten cancer patients transplanted<br />

with autologous cells generated from CD34 + cells cultured<br />

in the presence of SCF, IL-1, IL-3, IL-6 and Epo.<br />

However, the conditioning regimen given to these<br />

patients was not fully myeloablative, and this study<br />

offered no insight into the long-term engraftment<br />

potential of cells generated in this fashion. A similar<br />

approach was followed by Alcorn et al. 44 In ten patients<br />

with malignancy, an aliquot of the PBSC harvest was<br />

recovered from liquid nitrogen and CD34 were selected.<br />

Cells were cultured for 8 days in the presence of the<br />

same cytokine combination. A mean of 379×10 6 expanded<br />

cells were reinfused in addition to unmanipulated<br />

haematologica vol. 85(suppl. to n. 12):December 2000


100<br />

M. Aglietta et al.<br />

cells. The authors reported that the total LTC-IC number<br />

was not increased and most of the CD34 + cells were<br />

differentiated in front of an average 15-fold CFU-GM<br />

expansion (range 4-39). Similarly, averages of 71 (range<br />

27-151)-fold megakaryocytic cell expansion and 1,040<br />

(19-16.000)-fold erythroid cell expansion were reported.<br />

Although adverse reactions were not reported, no<br />

difference in the kinetics of engraftment was observed<br />

in comparison with historical controls.<br />

Different results come from preclinical studies where<br />

the infusion of large numbers of ex vivo expanded committed<br />

hematopoietic progenitors, together with unmanipulated<br />

cells might speed engraftment after<br />

chemotherapy and/or total body irradiation (TBI). Data<br />

from Uchida et al. 90 have suggested in the past that most<br />

of the short- as well as the long-term engraftment<br />

potential resides in uncommitted progenitors. More<br />

recently, Szilvassy et al. 91 have demonstrated that partially<br />

differentiated ex vivo expanded cells accelerate<br />

hematologic recovery in myeloablated mice transplanted<br />

with highly enriched long-term repopulating stem<br />

cells. In humans, Williams et al. 92 reinfused 9 breast cancer<br />

patients with unmanipulated apheresis products<br />

together with a mean of 44×10 6 /kg mature CD15 + cells<br />

generated ex vivo by CD34 + cells cultured in the presence<br />

of PIXY-321. No toxicity was observed after reinfusion,<br />

and time of white cell recovery was similar to<br />

that observed in the retrospective control group. In a<br />

more recent study, 78 megakaryocytic progenitors (MP)<br />

were obtained from CD34 + cells cultured in serum-free<br />

medium in the presence of Tpo, FL, SCF, IL-3, -6, -11 and<br />

MIP-1. Proliferation peaked on day 7 in culture, and a<br />

8±5-fold expansion of CD34 + /CD61 + cells, a 17±5-fold<br />

expansion of CFU-MK and a 58±14-fold expansion of<br />

the total number of CD61 + cells was obtained. Ten cancer<br />

patients undergoing autologous PBPC transplant<br />

received MP generated ex vivo (range 1-21 CD61 + cells<br />

×10 5 /kg) together with unmanipulated PBSC. Platelet<br />

transfusion support was not needed in 2 out of the 4<br />

patients receiving the highest dose of cultured MP and<br />

this result compared favorably with a retrospective control<br />

group of 14 patients, all requiring platelet transfusion<br />

support. A major concern is the potential expansion<br />

of contaminating tumor cells along with hematopoietic<br />

progenitors. In fact, it has been demonstrated that<br />

CD34 + cell selection decreases (but does not abrogate)<br />

neoplastic cell contamination from aphereses of myeloma<br />

patients. 46 For instance, in the majority of B cell lymphoma<br />

patients CD34 + cell selection does not eliminate<br />

contaminating t(14;18) + cells. However, during ex vivo<br />

expansion residual lymphoma cells do not proliferate<br />

and become undetectable by molecular analysis in the<br />

majority of cases. 93 Similarly, Vogel et al. recently indicated<br />

that exogenously mixed epithelial tumor cell lines<br />

might have a relative disadvantage over CD34 + cells during<br />

ex vivo expansion. 94<br />

Future directions<br />

Future challenges in this field are represented by the<br />

expansion of true human stem cells without maturation,<br />

to extend this strategy to allogeneic stem cell<br />

transplantation, and especially cord blood allograft, as<br />

well as the manipulation of cycling of primitive progenitors<br />

for gene therapy programs.<br />

Selective amplification of specific myeloid lineages (e.g.<br />

platelets or granulocytes) may improve the results of<br />

autologous transplantation. Moreover, although early<br />

results need confirmation, the amplification of early/committed<br />

hematopoietic cells coupled with the removal of<br />

neoplastic cells contaminating autologous grafts appears<br />

to be feasible.<br />

Expansion of cytotoxic effectors<br />

Human cytotoxic effector (CE) cells can be divided in<br />

two major groups:<br />

1. cells requiring prior antigen sensitization, which recognize<br />

their target in the context of the major histocompatibility<br />

complex (MHC) molecules;<br />

2. cells not requiring prior antigen sensitization being<br />

spontaneously cytotoxic against tumor target cells<br />

(e.g. K-562 cell line) in a non-MHC restricted setting.<br />

While the first group includes only some subsets of T-<br />

lymphocytes (CD8 + or CD4 + cells), the second one is<br />

more heterogeneous and includes both T-cells and natural<br />

killer (NK) cells, expressing the CD56 antigen. 95<br />

The so-called antibody-dependent cellular cytotoxicity<br />

(ADCC) can be mediated by cells expressing the Fc<br />

receptor II and the Fc receptor III (e.g. NK cells and<br />

CD3 + /CD16 + cells). Although this activity is not exhibited<br />

by non MHC-restricted cells, it cannot be considered<br />

aspecific and it is also exerted by monocytes.<br />

The lymphokine-activated killer cells (LAK) are capable<br />

of killing NK-resistant cellular targets (e.g. Daudi cell<br />

line). Although some tissue-resident lymphocytes may<br />

have spontaneous LAK activity, normal blood mononuclear<br />

cells do not show any LAK activity, which can be<br />

acquired after incubation with Interleukin-2 (IL-2). 96<br />

Therefore the LAK assay is a measure of the capacity<br />

of T and NK cells to become activated and to express<br />

cytolytic function.<br />

Killing mechanisms of cytolytic effectors<br />

Cytotoxic T-Lymphocytes (CTL) and NK cells possess at<br />

least two distinct, fast-acting, lytic mechanisms:<br />

97, 98<br />

1. the granule exocytosis pathway involves the secretion<br />

of perforin and granzymes which penetrate<br />

throughout the target cell pores, inducing cell death;<br />

2. a non-secretory mechanism which is mediated by<br />

the interaction between the Fas-ligand, expressed<br />

by the killer-cell and Fas (CD95) which triggers a<br />

cascade of proteolytic enzymes leading to apoptosis<br />

of both the killer and the target cell.<br />

A third cytolytic pathway, involving TNF, has recently<br />

been described. 99<br />

A series of apoptosis-resistant clones of human lym-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

101<br />

phoma cells has been described. These cells express<br />

fas/APO-1 receptors lacking the intracytoplasmic signaling<br />

domain. 100<br />

NK cells, as well as CTL, can recognize MHC class I<br />

molecules. However, recognition of MHC on target cells<br />

downregulates the NK cell function, suggesting the<br />

presence of inhibitory receptors. 101<br />

LAK cells are not MHC-restricted and are also capable<br />

of killing freshly isolated tumor cells. Similarly to<br />

CTL and NK cells, their activity is mediated by both lytic<br />

pathways (perforin/granzyme) and Fas-mediated<br />

apoptosis.<br />

Cytokines involved in CE function<br />

Several cytokines affect CTL and NK cell response. In<br />

particular, IL-2 expands the pool of alloreactive CTL precursors<br />

and IL-15 (produced by monocytes) mimics IL-<br />

2 action by inducing -IFN production, the activation of<br />

memory T cells and CTL proliferation. 102<br />

In addition, GM-CSF can affect certain T-lymphocyte<br />

functions by enhancing their cytotoxicity and -IFN<br />

production. This multifunctional cytokine can also augment<br />

NK cell function and the expression of adhesion<br />

molecules on the surface of leukemic cells. 103,104 Moreover,<br />

it has been hypothesized that the association of<br />

GM-CSF/IL-2 can also be useful for the activation of<br />

cytotoxic effectors by circulating progenitors, preserving<br />

the clonogenic potential of normal hemopoietic precursors.<br />

105<br />

Figure 3. NK cells differentiation pathway.<br />

Modified from Miller et al, Blood 1994; 83: 2594-601.<br />

IL-12 elicits the production of -IFN by CTL thus<br />

enhancing their antineoplastic efficacy and promotes the<br />

differentiation of T-helper-1. 106,107 Finally, IL-7 seems to<br />

be critical for the development of CTL and for a fast<br />

immune reconstitution after bone marrow transplantation<br />

(BMT). 108<br />

In conclusion, these cytokines play a pivotal role in the<br />

immune response against tumor cells by expanding, activating<br />

and recruiting CE and secondary effector cells<br />

(macrophages) or by directly inhibiting tumor cell growth.<br />

Role of cytotoxic effectors in immunosurveillance<br />

There are several in vitro and in vivo data supporting<br />

the role of immunosurveillance in tumor growth control.<br />

109 The graft-versus-leukemia (GVL) effect has been<br />

demonstrated to play a critical role in the eradication of<br />

minimal residual disease (MRD) after allogeneic<br />

trasplantation and there is evidence supporting the role<br />

of both T cells and NK cells in preventing disease<br />

relapse. 110<br />

Today, there is no doubt that CTL have a major role in<br />

killing allogeneic tumor cells in a MHC-restricted manner.<br />

For example, in CML, the higher relapse incidence<br />

after T-cell depleted allogeneic BMT 111 and the dramatic<br />

effect of donor T-lymphocyte infusion after relapse<br />

following BMT, 112 strongly support the importance of<br />

MHC-restricted GVL.<br />

Unfortunately, a selective GVL effect (separated from<br />

GVHD) can be obtained very rarely in patients receiving<br />

allogeneic BMT. Moreover, although animal models indicate<br />

that autologous GVHD exists and could generate a<br />

significant antitumor effect, the high incidence of relapse<br />

in patients receiving autologous BMT demonstrates that<br />

autologous GVL is often clinically ineffective. 113<br />

Rationale for cytotoxic effectors’<br />

expansion<br />

There are many important reasons to increase the<br />

number, the efficacy and the specificity of cytolytic<br />

effectors both in the allogeneic and in the autologous<br />

setting. The main clinical goals are the following:<br />

1. to reduce the relapse-incidence after autologous<br />

BMT, which is still relevant in acute leukemia, lymphoma<br />

and breast cancer;<br />

2. to cure diseases in which autologous BMT can only<br />

prolong the survival (multiple myeloma, CML, metastatic<br />

chemosensitive cancers);<br />

3. to reduce the incidence of relapse after allogeneic<br />

BMT especially in patients transplanted with great<br />

tumor burden;<br />

4. to accelerate the immune reconstitution after BMT,<br />

in order to reduce the morbidity and transplantrelated-mortality<br />

caused by serious infections (e.g.<br />

CMV and systemic mycosis).<br />

The CE which are investigated for in vivo or ex vivo<br />

expansion are mainly NK cells in the autologous setting<br />

and CTL in the allogeneic setting.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


102<br />

M. Aglietta et al.<br />

Development and expansion of NK cells<br />

NK cells belong to the naive part of the immune system<br />

and begin to appear into PB early after allo and<br />

auto-BMT. These cells express the N-CAM homologous<br />

CD56 antigen, but lack T-cell receptor / complexes.<br />

They are also characterized by low affinity receptors for<br />

IgG (CD16) 114-116 and their binding to malignant cells is<br />

mediated by CD18 molecule. 117 The most important<br />

organ for NK differentiation is the BM. Several data suggest<br />

that a common precursor of T cells and NK cells<br />

does exist. Fetal NK cells express the TCR , , , subunits<br />

while fetal B precursors do not express TCR subunits<br />

(Figure 3).<br />

These bipotential T/NK precursors do not have TCR<br />

rearrangement and share CD34/CD33/CD7 antigens.<br />

They differentiate to NK in the presence of SCF, IL-7, IL-<br />

2 and stromal feeder cells. The first step of NK differentiation<br />

is stroma-dependent while the second step of NK<br />

maturation is stroma-independent and is strongly<br />

potentiated by the association of IL-2 with IL-7. 118 In all<br />

studies IL-2 is required for NK cell differentiation from<br />

CD34 + cells. This finding suggests that a fraction of<br />

CD34 + cells expresses IL-2 receptors and that activated<br />

T-cells (as IL-2 source) should be detectable in the cellular<br />

milieu. However, T-cell deficient mice have normal<br />

NK cell development and humans lacking the -chain<br />

subunit of IL-2R lack NK activity. These unexpected<br />

observations have recently been supported by the<br />

demonstration that IL-15, produced by BM stromal cells,<br />

can directly induce CD34 + cells to differentiate into<br />

CD3 – /56 + NK cells in the absence of IL-2. 119<br />

The last step of NK development, after the expression<br />

of CD16, is characterized by the appearance of the CD56<br />

molecule. The intensity of CD56 expression directly correlates<br />

with the proliferative potential and the killing<br />

ability of NK cells. 120<br />

There is clear evidence that mature NK elements have<br />

a clonally-distributed ability to recognize their target<br />

cell by class I MHC alleles and a precise correlation has<br />

been established between the expression of p58 receptors<br />

on NK cell surface and class I MHC alleles. These<br />

receptors transduce an inhibitory signal upon interaction<br />

with MHC class I antigens, to prevent NK cells from<br />

killing target cells expressing certain (self) HLA alleles.<br />

These findings are consistent with a self-tolerance<br />

mechanism exerted by the NK population which can be<br />

disrupted as a consequence of tumor transformation or<br />

viral infection or any other events inducing (or masking)<br />

class I molecules. 121,122<br />

After incubation with IL-2, NK cells become LAK cells<br />

capable of killing otherwise NK-resistant target cells.<br />

These (NK) activated cells express new markers such as<br />

CD25, MHC class II and fibronectin which can be useful<br />

for the evaluation of their functional state. The NK cell<br />

compartment is heterogeneous and distinct subsets<br />

have been characterized. The most informative functional<br />

differences are based on relative CD56 fluorescence:<br />

only CD56 +bright , but not CD56 +dim NK cells, express<br />

the high-affinity IL-2 receptor. As a consequence, they<br />

respond to low concentrations of IL-2 and expand 10<br />

times more than CD56 +dim . This subset seems to be significantly<br />

reduced in leukemic patients. A remarkable<br />

reduction of CD56 +bright NK cells has been observed in<br />

CML patients coupled with a significant decrease of<br />

their spontaneous cytotoxicity against the K-562 line.<br />

However, this defect was corrected by 18 hours incubation<br />

with 1000 U/mL of recombinant IL-2. 120 These<br />

data strongly suggest that during tumor progression,<br />

the NK compartment (and particularly the small fraction<br />

of NK-CD56 +bright with high proliferative ability) is progressively<br />

suppressed even though the exogenous<br />

administration of IL-2 can partially reverse this phenomenon.<br />

The strong correlation between functional<br />

capacity of the NK cell compartment and tumor progression<br />

has often been reported as well as the efficacy<br />

of the administration of LAK cells plus IL-2 in restoring<br />

an anti-tumor response. 123<br />

Along this line, more than 90% of patients with acute<br />

leukemia in complete remission do not show spontaneous<br />

cytotoxicity against autologous blast cells. However,<br />

ex vivo treatment with IL-2 restores cytolytic activity<br />

in 37.5% of these patients. 124<br />

The first attempts to generate and expand LAK activity<br />

either in vivo or in vitro (after ex vivo incubation with<br />

IL-2) have been clinically disappointing especially in<br />

patients autotransplanted for acute lymphoblastic<br />

leukemia (ALL). 125 Nevertheless, there is now a renewed<br />

interest in the use of activated NK cells in hematologic<br />

malignancies, based on the optimization of different<br />

approaches:<br />

• administration of IL-2 in vivo to expand functionally<br />

active CE in patients with low tumor-burden, in<br />

order to reach an optimal effector/target ratio;<br />

• harvesting and culturing large amount of NK cells for<br />

additional ex vivo expansion/activation with IL-2.<br />

Expanded cells should be reinfused in the early phase<br />

after BMT;<br />

• sequential combination of both techniques (Figure<br />

4). 126<br />

The systemic administration of IL-2 for in vivo expansion<br />

and activation of the NK compartment may theoretically<br />

have some advantages:<br />

1. high number of CE precursors in the body;<br />

2. possibility of activating CE residing in the tumor<br />

bulk;<br />

3. more feasible and cheaper strategy than the generation<br />

and administration of LAK cells.<br />

On the other hand, the main drawbacks of this<br />

approach are the high toxicity of systemic administration<br />

of IL-2 and the high variability of anti-tumor<br />

response.<br />

Sources of cytotoxic effectors and<br />

modalities of NK cell expansion<br />

Human NK progenitor cells can normally be found in<br />

the BM, 127,128 and originate from CD34 + hematopoietic<br />

progenitors. 129 So far, the generation of NK cells from<br />

CD34 + precursors has been described on a small scale<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

103<br />

A<br />

B<br />

Figure 4. Strategies for generation and activation of NK cells in vitro and in vivo. Modified from Klingemann et al., Exp Hematol<br />

1993; 21:1263-70.<br />

basis but not in large scale experiments. 130<br />

High numbers of functionally active NK cells can be<br />

easily demonstrated in mobilized cells from patients<br />

receiving chemotherapy plus G-CSF. Silva et al. 131 have<br />

shown a 5.4-fold expansion of NK cells from leukapheresis<br />

products incubated in the presence of IL-2 for<br />

6-8 days without affecting the CD34 + cell content.<br />

However, decreased function of NK cells has recently<br />

been described in the PB of normal donors after G-CSF<br />

administration. 132 Circulating NK progenitors showed a<br />

decreased killing capacity and diminished proliferative<br />

ability in response to IL-2, as compared to their<br />

unprimed BM counterpart.<br />

Based on previously published LAK trials, 123, 133 it can be<br />

estimated that about 10 10 -10 11 activated NK cells are<br />

needed to stimulate an anti-tumor response. A 100-fold<br />

ex vivo expansion of these cells from a standard leukapheresis<br />

collection would, therefore be required to obtain<br />

such a high number of effectors.<br />

Beaujean et al. 134 reinfused autologous BM cells incubated<br />

for 10 days with IL-2 in 5 ALL patients following<br />

a myeloablative treatment. This procedure resulted in<br />

an important loss of hemopoietic progenitors with<br />

delayed engraftment. Moreover, in spite of this attempt<br />

to induce an autologous GVL, all patients eventually<br />

relapsed. 134<br />

Wong et al. 135 compared the ability of IL-2 alone or<br />

combined with IL-7 or IL-12 to stimulate NK activity in<br />

BM or PB samples. They found that IL-2/IL-12-activated<br />

blood cells suppressed the growth of the leukemia cell<br />

line K-562 about eight-fold more efficiently than BM<br />

cells. They also found that cryopreservation and subsequent<br />

stimulation of BM and PB cells did not significantly<br />

decrease the activity of NK cells. Finally, the combination<br />

of IL-2 and IL-12 showed a synergistic effect on both<br />

BM and PB elements. 135 Large-scale ex vivo expansion<br />

of NK cells for adoptive immunotherapy not only<br />

requires an optimal source of precursors, but also clinically<br />

approved materials and procedures. In this context,<br />

Miller et al. 136 obtained a 21-fold expansion of NK<br />

cells using a 21-day large scale NK culture performed in<br />

gas-permeable bags. Pierson et al. 137 observed a 352-<br />

fold expansion of NK cells after 33 days of incubation<br />

in a bioreactor. Their starting population was NK precursors<br />

enriched by negative panning with anti-CD5 and<br />

anti-CD8 antibodies. The activated NK population was<br />

highly purified (>90%) in CD56 + /CD3 – cells and maintained<br />

a powerful cytotoxicity against K-562 cells.<br />

The use of a homogeneous NK cell fraction for celltherapy<br />

programs seems to be advantageous because<br />

activated NK cells have more specific lytic activity than<br />

heterogeneous LAK populations. 138 However, creating<br />

such a fraction requires a first step of enrichment (e.g.<br />

by eliminating CD8 + /CD5 + cells) and long-term cultures<br />

carry the risk of fungal or bacterial contamination.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


104<br />

M. Aglietta et al.<br />

Cytokine induced killer cells (CIK)<br />

expansion<br />

In 1986 Lanier described a subset of CD3 + T cells coexpressing<br />

the CD56 antigen which is a typical NK marker.<br />

139 A remarkable expansion of this cellular subset has<br />

recently been obtained by Schmidt et al. 140 following a<br />

16-day incubation in the presence of IFN, IL-1, IL-2<br />

and a monoclonal antibody against CD3 as the mitogenic<br />

stimulus. The ability of CIK cells to deplete<br />

leukemic cells from CML marrow was then investigated<br />

by the same group. 141 While standard LAK cells were, in<br />

most cases, unable to lyse CML cells, CIK cells were toxic<br />

to both autologous and allogeneic CML blasts without<br />

affecting normal hemopoietic progenitors.<br />

CTL expansion for adoptive therapy<br />

It is well known that the GVL effect can be transferred<br />

with donor-buffy-coat (BC) lymphocytes. 142 The<br />

antitumor effect of the CTL contained in the BC has<br />

been shown to be more potent than that induced by NK<br />

cells, even though NK cells exert a GVL activity different<br />

from GVHD. 143<br />

In a murine model, a single dose of 210 7 CTLs in<br />

tumor bearing mice (DBA/2) resulted in the eradication<br />

of primary cancer and metastases without causing<br />

severe GVHD. 144 Unfortunately, this GVL effect cannot be<br />

easily separated from GVHD in humans. As a matter of<br />

fact, in clinical studies the beneficial effect of CTL<br />

administration is often offset by the severity of GVHD or<br />

marrow aplasia.<br />

Although leukemic cells share common antigens with<br />

other tissues of the host, there is also the chance that<br />

distinct leukemic antigens may be recognized by specific<br />

allogeneic CTL. 145<br />

Leukemia-specific T-cell clones have been isolated<br />

from HLA-identical siblings 146 and this finding may<br />

explain the high incidence of CR, without GVHD, in<br />

patients with CML relapsed after allo-BMT and treated<br />

with donor BC. 147<br />

The subset of donor-lymphocytes involved in the GVL<br />

effect is not entirely defined. Both CD4 + and CD8 + GVL<br />

effectors have been described in animal models. Recent<br />

studies in man suggest a prominent role of CD8 + cells<br />

in acute leukemia and CD4 + cells in CML. 148 The therapeutic<br />

index of this approach may be increased by<br />

treating the donor lymphocytes, previously activated<br />

with recipient PHA-stimulated blast, with anti-CD25<br />

ricin-conjugated antibodies. 149 This procedure gives origin<br />

to a CTL population which retains over 75% of its<br />

antileukemic activity with only 10% of the initial<br />

responsiveness against the non-leukemic cells of the<br />

recipient.<br />

Strategies for generation and expansion of<br />

specific CTL<br />

A very attractive system for generating and expanding<br />

CTL is based on the selection and isolation of tumorspecific<br />

peptides (e.g. those encoded by bcr-abl or PML-<br />

RAR fusion genes) and to presenting them to T-cells to<br />

stimulate a specific T-cell response. The responding T-<br />

clones can then be amplified and selected by limiting<br />

dilution techniques. Unfortunately, in many cases the<br />

tumor-specific peptide is not presented by leukemic cells<br />

making the generation of peptide-specific CTL useless. 150<br />

In this regard, the transfection in tumor cells of DNA<br />

sequences encoding for co-stimulatory molecules (B7-1)<br />

or cytokines such as GM-CSF has greatly enhanced the<br />

anti-tumor response of T-cells. Alternatively, the use of<br />

professional antigen presenting cells (APC), primed with<br />

tumor specific antigens (e.g. tumor specific idiotype in<br />

low-grade B-cell lymphoma) has proved to be effective<br />

for the generation of tumor-specific CTL clones in vivo<br />

capable of inducinga measurable anti-tumor response. 151<br />

Allo-CTL have also been used against EBV-related lymphoma<br />

developed in allograft recipients and HIV patients.<br />

EBV infection is controlled in normal individuals by specific<br />

CTL which lyse EBV-infected B-cells upon recognition<br />

of viral peptides presented on the cell membrane in<br />

association with MHC class I molecules.<br />

EBV-specific CTL have been isolated from normal<br />

donor leucocytes and expanded ex vivo by Rooney et<br />

al. 152 Following the reinfusion of 1.2×10 8 CTL/m 2 into an<br />

allograft recipient, the complete resolution of an EBVrelated<br />

immunoblastic lymphoma was observed.<br />

Ex vivo expanded CTL can also be used to restore CMVspecific<br />

responses in immunodeficient individuals receiving<br />

allogeneic BMT. Walter et al. 153 treated 14 patients<br />

with infusions of CD8 + CTL directed to CMV proteins<br />

obtained from bone marrow donors. In this study, CMVspecific<br />

CTL were expanded by stimulation with anti-CD3<br />

antibodies coupled with autologous CMV-infected<br />

fibroblasts in IL-2-containing culture. This approach to<br />

adoptive immunotherapy was well tolerated by the recipients<br />

and not associated with severe GVHD.<br />

Future directions<br />

Preliminary clinical data suggest that the efficacy of<br />

donor BC infusion for the treatment of leukemic relapse<br />

can be significantly improved by the administration of IL-<br />

2 in vivo after reinfusion and by a brief incubation of BC<br />

with IL-2 before the reinfusion. 154 The antitumor efficacy<br />

of T-lymphocytes can also be enhanced by transfection<br />

of cytokine genes or new receptors. An interesting<br />

approach is represented by the binding of TCR to a specific<br />

anti-tumor antibody Fab fragment or the use of<br />

bispecific (anti-tumor and anti-CD3) antibodies capable<br />

of recruiting and expanding tumor-specific CTL at the<br />

tumor site. 155 Recently, a significant autologous GVHD<br />

effect has been obtained by the addition of -IFN to<br />

cyclosporin-A in order to upregulate MHC class II molecules<br />

. 113 Alternatively, GM-CSF seems to enhance the<br />

anti-tumor response by stimulating professional APC<br />

(see below). 156<br />

In conclusion, there is clear evidence that CTL exert<br />

their cytotoxic effect through the recognition of minor<br />

HLA antigens. 157 However, in some patients a very low<br />

frequency of specific antileukemic CTL, responsible for<br />

a GVL effect distinct from GVHD, have been isolated.<br />

Moreover, genetic approaches, such as the transduction<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

105<br />

of donor lymphocytes with a suicide gene, have been<br />

proposed to control GVHD occurring after CTL administration.<br />

158<br />

Ex vivo generation of human dendritic APC<br />

Clinical investigators are keenly interested in the role<br />

of APC in the initiation of immune responses because of<br />

the potential to exploit these cells for immunotherapy<br />

of cancer and viral diseases. Pioneer studies in mammals<br />

by Steinman et al. 159 have demonstrated that the specialized<br />

system of APC is constituted by BM-derived<br />

dendritic cells (DC). DC are distinguished by their unique<br />

potency and ability to capture, process, and present<br />

antigens into peptide-HLA complexes to naive T lymphocytes<br />

and to deliver the co-stimulatory signals necessary<br />

for T lymphocyte activation and proliferation.<br />

Here, we summarize the main experimental evidence<br />

supporting the working hypothesis that individuals vaccinated<br />

with DC expanded ex vivo and engineered to<br />

present tumor associated antigen(s) can mount tumorspecific<br />

humoral and cellular responses. This can lead to<br />

tumor regression as well as protective immunity against<br />

tumor growth in vivo. 159-162<br />

Identification of dendritic cells<br />

DC are leukocytes derived from hematopoietic stem<br />

cells along the myeloid differentiation pathway (Figure<br />

5). The differentiation of DC is a stepwise process: 163 originating<br />

from myeloid progenitors in the BM, immature DC<br />

distribute via blood to tissues where they have the capacity<br />

to take up and to process antigens. As migratory DC,<br />

they transit through the lymph or blood to lymphoid<br />

organs, where they become mature DC, which lose antigen-processing<br />

ability and acquire superior antigen-presenting<br />

capacity for T lymphocytes. 163 In humans, DC at<br />

different developmental stages circulate in PB and they<br />

are found in virtually all tissues of the body where,<br />

depending on the location, they are referred to as interstitial<br />

DC (heart, kidney, gut, and lung), Langerhans cells<br />

(skin, mucous membranes), interdigitating DC (thymic<br />

medulla, secondary lymphoid tissue); or veiled cells (lymph,<br />

blood). 163 DC are regarded as distinct from monocytes/macrophages,<br />

although they share a common progenitor<br />

after the CFU-GM stage. However, this has been<br />

questioned as mixed colonies of dendritic cells and<br />

macrophages are generated in vitro from single CD34 +<br />

hematopoietic progenitors more commonly 163-175 than<br />

pure DC colonies. 176<br />

DC can be distinguished from other APC by a) morphology;<br />

b) cell-surface membrane phenotype; and c)<br />

the strong capacity to present antigens to T cells, usually<br />

assessed in the allogeneic mixed leukocyte culture<br />

(reviewed in ref. #163).<br />

Cutaneous DC, as well as most of the DC generated ex<br />

vivo from human CD34 + progenitors cells express high<br />

levels of the surface membrane CD1a antigen (Figure 6).<br />

Although CD1a antigen can be found on cortical thymocytes<br />

and some B lymphocytes, its presence (noted by<br />

immunofluorescence and flow cytometry) is the most<br />

useful way to quantify the ex vivo generation of DC from<br />

early precursors. In addition to the CD1a antigen, DC<br />

express peculiarly high levels of class I and class II histocompatibility<br />

complex structures, co-stimulatory molecules<br />

for T-lymphocytes such as B7-1 (CD80) and B7-<br />

2 (CD86), and adhesion molecules such as ICAM-1<br />

(CD54) and ICAM-3 (CD50) which are involved in DC-<br />

Figure 5. Scheme of DC<br />

development in BM, PB<br />

and thymus.<br />

Abbreviations: ly, lymphocyte;<br />

NK, natural killer<br />

cells; NSE, non-specific<br />

esterase; PPSC, pluripotent<br />

stem cells; CFU-DL,<br />

dendritic/Langherans cell<br />

colony forming unit. Modified<br />

from Reid CA, Br J<br />

Haematol 1997; 96:217-<br />

33.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


106<br />

M. Aglietta et al.<br />

Figure 6. Flow cytometry evaluation of cell surface phenotype of DC derived ex vivo from CD34 + progenitors on day 12 of culture<br />

in the presence of GM-CSF, TNF-, SCF and FL.<br />

dependent T-lymphocyte proliferation. DC lack monocyte/macrophage-<br />

and lymphocyte-lineage-restricted<br />

antigens with the exception of the CD4 antigen. 170 As<br />

shown in Table 3, relevant co-stimulatory (B7-1 and B7-<br />

2) and adhesion molecules (ICAM-1) are expressed on all<br />

CD1a + cells derived from CD34 + progenitors, but on fewer<br />

CD1a + cells derived from monocytes. 177,178 More<br />

recently, the CD83 cell surface antigen has been recognized<br />

as a valuable tool for detecting blood DC. 179<br />

Ex vivo expansion of dendritic cells<br />

Although DC circulate in the PB and are found in virtually<br />

all tissues of the body, it is difficult to obtain<br />

enough cells for ex vivo manipulation because of their<br />

scattered locations and low number in the blood where<br />

they account for approximately 0.1% of all leukocytes. 163<br />

For this reason, it has been of crucial interest to know<br />

that: a) TNF- cooperates with GM-CSF to generate DC<br />

from CD34 + hematopoietic progenitors from BM, cord<br />

blood or PB; 166, 168-176 and b) IL-4 cooperates with GM-<br />

CSF in the development of DC from circulating monocytes.<br />

169,180 A detailed description of the methods utilized<br />

to obtain human DC from myeloid precursors has been<br />

recently reported. 163 However, in evaluating these methods<br />

in view of a clinical trial, at least three issues should<br />

be taken into consideration:<br />

a) the type of DC generated either from monocytes<br />

(monocyte-derived DCs) or from CD34 + hematopoietic<br />

progenitors (CD34 + derived DC);<br />

b) the source of serum for DC growth in culture;<br />

c) the combination of cytokines required for optimal ex<br />

vivo expansion of functional immunostimulatory DC.<br />

Monocyte-derived DC are being employed in patients<br />

with advanced stage malignancies in phase I-II clinical trials.<br />

The trials are particularly aimed at evaluating toxicity<br />

and immune responses after subcutaneous administration<br />

following DC pulsing ex vivo with either melanoma<br />

tumor-associated peptides 181-183 or with B-cell lymphoma<br />

and myeloma idiotype proteins from autologous<br />

serum. 151,161 Early reports from clinical studies, in patients<br />

with melanoma who are HLA-A1 positive and whose<br />

malignant cells express the MAGE-1 gene, show that in<br />

vivo immunization with autologous monocyte-derived<br />

DC pulsed with MAGE-1 gene coded nonapeptide, is not<br />

toxic and can induce peptide-specific autologous<br />

melanoma reactive CD8 + cytotoxic T-lymphocyte<br />

responses in situ at the vaccination site and at distant<br />

tumor sites 181 as well as in PB. 182 From a technical point<br />

of view, in these studies the generation of DC from PB<br />

mononuclear cells is dependent on a culture medium<br />

necessarily containing GM-CSF without serum or with<br />

human pooled donor serum. Under these conditions the<br />

production of DC is quite scarce in comparison with that<br />

achieved with fetal calf serum, as reported in early studies.<br />

169 However, the presence of fetal calf serum in the<br />

culture medium induces undesired DC-mediated immune<br />

responses to xenogenic proteins as observed in murine 184<br />

and human preclinical studies. 170 In this regard, experimental<br />

data suggest that CD34 + cell-derived DC can also<br />

be generated in the absence of serum if the culture<br />

medium containing GM-CSF and TNF- is supplemented<br />

with TGF-1. 185<br />

Modalities for the large-scale procurement of functional<br />

DC from CD34 + hematopoietic progenitors in<br />

patients with cancer have been evaluated. 170 It was<br />

found that mobilized PB progenitors currently utilized in<br />

phase III trials 186 include a fraction of CD34 + DC precursors<br />

which give origin ex vivo to a progeny with the char-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

107<br />

acteristics of professional APC i.e., typical DC morphology<br />

and immunophenotype undistinguishable from cutaneous<br />

Langerhans cells and DC from cord blood and BM<br />

CD34 + cells. Most importantly, these ex vivo generated<br />

DC retained the capacity to process and present antigens<br />

to T lymphocytes as demonstrated by elicitation of HLA<br />

class II and class I-restricted activation of CD4 + and CD8 +<br />

autologous T lymphocytes in response to xenogenic antigens<br />

of fetal calf serum 170 or melanoma tumor-associated<br />

antigen peptides, 177, 178 respectively. Quantification<br />

of progenitors of DC by limiting dilution analysis of<br />

CD34 + cells sorted from blood cell autografts showed<br />

that they are approximately 140-fold more numerous<br />

than in steady-state control autograft. To obtain this<br />

favorable result, blood cell autografts were collected at<br />

the time of maximal mobilization of CD34 + cells into PB<br />

as occurs after treatment with high-dose cyclophosphamide<br />

and cytokines.<br />

In a systematic search for culture conditions capable<br />

of ameliorating the ex vivo generation of DC dendritic<br />

cells, a variety of exogenous stimuli have been evaluated<br />

as well as monocyte-derived versus CD34 + cell-derived<br />

DC. 170,177,178 In this respect, it has been shown that GM-<br />

CSF plus TNF--dependent generation of DC from mobilized<br />

CD34 + cells is 2.5 fold enhanced by either FL or SCF,<br />

and 5-fold enhanced by a combination of these growth<br />

factors. In addition, autologous high-dose chemotherapy<br />

recovery phase serum rather than fetal calf serum or<br />

human donor pooled AB serum has been shown to be the<br />

optimal serum for the generation of DC. Regardless of<br />

the precise mechanism of action of FL and SCF in association<br />

with GM-CSF and TNF- on the enhancement of<br />

DC differentiation and proliferation, these findings have<br />

provided new advantageous tools for the large-scale generation<br />

of DC from mobilized CD34 + cells in patients<br />

undergoing cancer treatment. In fact, the stimulation of<br />

CD34 + cells from an average blood cell autograft should<br />

permit the generation of a median of 0.610 9 /kg DC from<br />

Table 3. Phenotype of CD1a + dendritic cells generated from<br />

mobilized CD34 + progenitors or from monocytes.<br />

CD1a + cells derived from<br />

Antigens CD34 + progenitors Monocytes<br />

CD14 2% 3%<br />

CD80 100% 84%<br />

CD86 100% 67%<br />

CD54 100% 67%<br />

MHC Class I<br />

HLA-A0201 100% 100%<br />

MHC Class II<br />

HLA-DR 100% 100%<br />

HLADQ 60% 89%<br />

Modified from Mortarini et al., Cancer Res, in press.<br />

an average 65 kg individual, i.e., almost 4010 9 DC. In<br />

contrast, differentiation of DC from monocytes in the<br />

presence of autologous high-dose chemotherapy recovery<br />

phase serum plus GM-CSF and IL-4 is not associated<br />

with a comparably high outgrowth of DC. 177,178 These<br />

observations, together with the weaker expression of costimulatory<br />

molecules in monocyte-derived DC in comparison<br />

with CD34 + cell-derived DC 187 may favor the utilization<br />

of the latter source of APC for the development<br />

of active immunization programs involving DC in humans.<br />

The comparative efficiency as APC of DC derived from<br />

monocytes versus CD34 + hematopoietic progenitors has<br />

recently been studied with DC isolated from blood of<br />

patients with melanoma. In particular, it has been shown<br />

that DC derived from G-CSF-mobilized CD34 + cells are<br />

more efficient than those derived from monocytes in<br />

inducing melanoma tumor-associated antigen peptide<br />

specific activation of autologous CD8 + cytotoxic T-lymphocytes.<br />

Interestingly, in the same experiments the latter<br />

cells were also capable of lysing a panel of melanoma<br />

cell lines sharing the same HLA class I alleles with the<br />

patients from whom CD8 + cytotoxic T-lymphocytes were<br />

generated with tumor-associated antigen peptide pulsed<br />

autologous DC. 177,178 Moreover, CD34 + cells mobilized into<br />

PB by G-CSF were shown to be capable of generating a<br />

higher number of mature and fully functional DC than<br />

their BM counterparts. 188 However, it should be pointed<br />

out that, at present, clinical studies utilizing DC as vehicles<br />

for anti-tumor vaccination have been carried out<br />

with monocyte-derived DC either freshly isolated from<br />

PB 152 or cultured ex vivo. 161, 181-183 In addition, culture conditions<br />

which allow the large scale production of terminally<br />

differentiated and fully functional monocytederived<br />

DC have recently been described. 189<br />

Dendritic cells for antitumor cell therapy<br />

An extensive review on the clinical use of DC is beyond<br />

the scope of this paper, however, a few remarks in this<br />

regard are needed.<br />

The goal of vaccination is the induction of protective<br />

immunity. Originally, vaccinations were used in the setting<br />

of infectious diseases, but are now expanding to<br />

include the treatment of allergy, autoimmune diseases,<br />

and tumors. A rational approach to vaccination involves<br />

3 steps: a) the identification of the protective effector<br />

mechanisms, b) the choice of an antigen that can induce<br />

a response in all individuals, and c) the use of an appropriate<br />

way to deliver the vaccine to induce the proper<br />

type of response. 159-162<br />

It has now been demonstrated that certain tumor cells<br />

are antigenic by expressing tumor-associated antigens<br />

that can be recognized by T lymphocytes in a syngeneic<br />

host. However, they are often poorly immunogenic, at<br />

least in part because they lack the cellular armamentarium<br />

for specific T-lymphocyte recognition, activation,<br />

and co-stimulation typical of APC especially DC. 190,191<br />

Different mechanisms may account for the ability of<br />

tumor cells to evade immune responses. Tumor cells may<br />

haematologica vol. 85(suppl. to n. 12):December 2000


108<br />

M. Aglietta et al.<br />

display low immunogenicity through low MHC and/or<br />

tumor-speciifc antigen expression or may downregulate<br />

Fas and constitutively express Fas-Ligand, which binds to<br />

Fas on cytotoxic T-lymphocytes, resulting in apoptosis of<br />

the latter. 192 Furthermore, it has recently been shown<br />

that vascular endothelial growth factor produced by<br />

tumors inhibits the functional maturation of CD34 + cellderived<br />

DC. 193 When considering the use of the unique<br />

antigen-presenting capacity of DC to prime specific antitumor<br />

T-lymphocytes, this phenomenon should be taken<br />

into account, as it may result in poor recovery and<br />

function of DC directly recovered from the blood of cancer<br />

patients. In contrast, dendritic APC expanded ex vivo<br />

in the presence of cytokines and in the absence of<br />

inhibitory factors released by tumors would probably be<br />

functional. 193-195 However, it should also be considered<br />

that published data demonstrate that whereas in vivo<br />

administration of DC loaded with low doses of tumor<br />

antigen enhances antitumor immunity, DC pulsed with<br />

high doses of antigen or high numbers of tumor antigenpulsed<br />

DC may inhibit development of immunity. 196 This<br />

finding supports the notion that stimulation of DC-mediated<br />

antigen presentation in vivo may act in a tolerogenic<br />

or immunogenic fashion depending on a variety of<br />

partially understood factors. 197<br />

Basically, there are at least two approaches to tumor<br />

vaccination (Figure 7). The first is to identify a tumorassociated<br />

antigen to be used as a vaccine, the second<br />

is to increase the immunogenicity of tumor cells and let<br />

the immune system decide which antigen to target.<br />

Indeed, in experimental models with the appropriate<br />

manipulation exploiting the physiologic function of<br />

antigen-presenting DC, the immune system can be<br />

induced to mount responses that can kill tumor cells<br />

and also protect animals from subsequent challenge<br />

even with a poorly immunogenic tumor. 185,198-203<br />

Given the richness of recently identified tumor associated<br />

antigens and their corresponding peptide epitopes<br />

recognized by MHC-restricted CD8 + or CD4 + T<br />

lymphocytes (Table 4), investigators are currently evaluating<br />

the clinical efficacy of specific tumor-associated<br />

antigen-based vaccines for the treatment of various<br />

malignancies. Recently, in a cooperative clinical trial it<br />

was observed that partial tumor regressions can occur<br />

in HLA-A1 + patients with melanoma treated with a<br />

naked MAGE 3 peptide epitope vaccine even in the<br />

absence of any engineering of antigen-presenting cells<br />

or adjuvant cytokine(s). 204 This clinical evidence induces<br />

the belief that the effectiveness of peptide-based vaccines<br />

is likely to benefit further from administration of<br />

appropriate cytokines 156,205,206 or cellular adjuvants (e.g.<br />

DC) capable of promoting cellular immunity.<br />

Among hematologic malignancies, CML is being<br />

intensively evaluated as a possible target of dendritic<br />

APC-based immunotherapy. It is well known that CML<br />

is characterized by a specific translocation of the c-abl<br />

oncogene (9q34) to the bcr region on chromosome 22<br />

(22q11). Alternative recombination sites involving either<br />

the second or third exon of the bcr gene splicing to exon<br />

Table 4. Tumor antigens capable of eliciting T-lymphocyte<br />

responses.<br />

Activated oncogene products<br />

Mutated:<br />

Rearranged:<br />

Overexpressed:<br />

Tumor suppressor gene products<br />

p53 mutations<br />

Position 12 point mutation of 21 ras<br />

bcr-abl (b3a2 peptide)<br />

HER-2/neu<br />

Reactivated embryonic gene products<br />

MAGE family (at least 12 genes)<br />

BAGE<br />

GAGE<br />

Melanocyte differentiation antigens<br />

Tyrosinase protein<br />

Melan-A/MART1<br />

gp100<br />

gp75<br />

Viral gene products<br />

Idiotype epitopes<br />

Human papilloma virus antigens (E6, E7)<br />

EBV EBNA-1 gene products<br />

Ig and TCR hypervariable regions<br />

2 of the abl gene yield two potential fusion gene transcripts,<br />

b2a2 and b3a2, respectively. The translated 210-<br />

kd bcr-abl fusion protein, which has abnormal tyrosine<br />

kinase activity, includes a new potentially antigenic<br />

sequence at the fusion site: a new amino acid is generated<br />

at the junctional site by the fusion event; in the<br />

b2a2 fusion a glutamic acid (E) is encoded, whereas in<br />

the b3a2 recombination event a lysine (K) is generated.<br />

Interestingly, a bcr-abl peptide from the b3a2 fusion<br />

region has been found to be immunogenic in mice. 207 In<br />

humans, binding of b3a2 peptides to various HLA class<br />

I alleles 208 and priming of CD8 + cytotoxic T-lymphocytes<br />

in vitro has been described although the capacity of<br />

these peptide-specific CD8 + T-lymphocytes to lyse CML<br />

cells has not been determined. 209 In contrast, in a recent<br />

study it has been demonstrated that CD4 + T-lymphocytes<br />

can be identified that proliferate in an HLA class-<br />

II restricted manner in response to a 11mer<br />

(GFKQSSKALQR) b3a2 peptide especially when the latter<br />

is presented by purified CMRF-44 + blood DC. 210 In the<br />

same study the peptide-specific CD4 + T-lymphocytes<br />

were able to respond to the whole protein in crude<br />

extract from CML cells. Intriguingly, dendritic antigenpresenting<br />

cells in CML patients can be derived from<br />

the malignant clone and these malignant dendritic cells<br />

can induce antileukemic reactivity in autologous T lymphocytes<br />

without the necessity of additional exogenous<br />

antigens. 211<br />

Although, the above observations cannot be extrapolated<br />

a priori to other malignancies carrying specific<br />

translocations and corresponding fusion genes and<br />

products, 212 further investigations on the possible clin-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

109<br />

ical application of bcr-abl peptide(s) presented by autologous<br />

dendritic antigen-presenting cells are warranted.<br />

The translation into the clinical setting of the above<br />

experimental hints favoring the use of DC pulsed ex vivo<br />

with synthetic tumor-associated antigen peptides for<br />

effective cell therapy in humans is likely to be hampered<br />

by: a) the limited availability of patients with HLA typing<br />

compatible for the utilized tumor-associated antigen<br />

peptide(s); b) the occurrence of independent mechanisms<br />

of tumor escape in vivo such as loss of expression<br />

of tumor-associated antigens or of HLA class I during<br />

tumor progression; and c) the short duration of the<br />

immune responses thus requiring annoying boost vaccinations.<br />

It has been suggested 213 that these limitations<br />

may be overcome by transduction of genes encoding<br />

relevant proteins into DC or their progenitors so that<br />

DC could tailor peptides to their own HLA molecules<br />

thus obviating the need to synthesize tumor-specific<br />

peptides most of which have stringent HLA restrictions.<br />

A further advantage of the transduction approach may<br />

be the stable long-term expression of the antigen by<br />

the DC, which would allow its presentation to the<br />

immune system for longer periods without the concerns<br />

about the turnover of preformed peptide/HLA complexes<br />

in vivo after immunization. 214<br />

Based on the above experimental body of evidence, a<br />

pioneer clinical trial has evaluated the ability of autologous<br />

monocyte-derived DC pulsed ex vivo with non<br />

Hodgkin’s lymphoma-specific idiotype protein to stimulate<br />

host immunity when infused as a vaccine. 151 In this<br />

study active immunotherapy of patients with B-cell lymphoma<br />

against idiotypic determinants led to anti-tumor<br />

immunity that correlated with improved clinical outcome<br />

in some patients.<br />

Regardless of the type of cell manipulation (Figure 7)<br />

(ex vivo pulse with tumor-associated antigen peptides<br />

versus transduction with tumor associated antigen<br />

genes versus immunization with fusions of dendritic and<br />

carcinoma cells 215 ) that will be successful in clinical<br />

applications the necessity of methods of generating<br />

large numbers of functional DC is implicit for the evolution<br />

of such studies.<br />

Future directions<br />

Since DC have been shown to be intimately involved<br />

in the generation of CD4 + and CD8 + T-lymphocyte mediated<br />

tumor-specific immunity, it is attractive to speculate<br />

that vaccination with DC pulsed or engineered ex<br />

vivo to present tumor antigen(s) may be effective in generating<br />

tumor immunity in vivo. Among the recently<br />

prospected sources of DC, namely BM, neonatal cord<br />

blood, and adult PB, the last is certainly the richest and<br />

most accessible in all patients with cancer, although it<br />

remains to be confirmed whether functional differences<br />

will favor the utilization of monocyte- versus CD34 + cellderived<br />

DC. Thus, in the clinical setting of adoptive<br />

immunotherapy for patients with malignancies, a therapeutic<br />

protocol could be envisioned involving the mobilization<br />

of CD34 + cells into PB with hematopoietic<br />

growth factors, with or without prior intensive chemotherapy.<br />

Thus, enrichment of hematopoietic progenitor<br />

cells could be followed by ex vivo generation of DC<br />

pulsed or engineered to present tumor antigen(s), to be<br />

reinfused as a potential secondary tumor-specific immunotherapy<br />

or vaccination. It remains to be established<br />

whether the latter effect could be further enhanced by<br />

in vivo adjuvant cytokines such as GM-CSF and/or FL, as<br />

occurs in murine models.<br />

A potential advantage of ex vivo immune cell therapy<br />

over direct in vivo immune intervention, is the lack<br />

of functional inhibition that may occur in vivo. This<br />

hypothesis is based on a recently proposed mechanism<br />

of tumor escape/resistance from the host immune system,<br />

in which cancer cells produce a vascular endothelial<br />

growth factor that impairs antigen presentation<br />

required to induce specific antitumor immune responses<br />

in vivo. 193<br />

Figure 7. Sources of tumor antigens<br />

for DC-based cancer vaccines.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


110<br />

M. Aglietta et al.<br />

Contributions and Acknowledgments<br />

All the authors equally contributed to the manuscript<br />

and they are listed in alphabetical order.<br />

Disclosures<br />

Conflict of interest. This review article was prepared by<br />

a group of experts designated by <strong>Haematologica</strong> and by<br />

representatives of two pharmaceutical companies, Amgen<br />

Italia SpA and Dompé Biotec SpA, both from Milan, Italy.<br />

This co-operation between a medical journal and pharmaceutical<br />

companies is based on the common aim of<br />

achieving optimal use of new therapeutic procedures in<br />

medical practice. In agreement with the Journal’s Conflict<br />

of Interest policy, the reader is given the following<br />

information. The preparation of this manuscript was supported<br />

by educational grants from the two companies.<br />

Dompé Biotec SpA sells G-CSF and rHuEpo in Italy, and<br />

Amgen Italia SpA has a stake in Dompé Biotec SpA.<br />

Manuscript processing<br />

Manuscript received March 11, 1998; accepted June<br />

10, 1998.<br />

References<br />

1. Scott MA, Gordon MY. In search of the haemopoietic<br />

stem cell. Br J Haematol 1995; 90:738-43.<br />

2. Morrison SJ, Shah NM, Anderson DJ. Regulatory<br />

mechanisms in stem cell biology. Cell 1997; 88:287-<br />

98.<br />

3. Metcalf D. The molecular control of cell division, differentiation<br />

commitment and maturation in haemopoietic<br />

cells. Nature 1989; 339:27-30.<br />

4. Lajtha LG. Haemopoietic stem cells. Br J Haematol<br />

1975; 29:529-35.<br />

5. McCulloch EA. Control of hematopoiesis at the cellular<br />

level. In: Gordon AS, ed. Regulation of hematopoiesis<br />

(vol. 1). New York: Appleton, 1970. p 133-54.<br />

6. Gordon MY. Physiological mechanisms in BMT and<br />

haemopoiesis - revisited. Bone Marrow Transplant<br />

1993; 11:193-7.<br />

7. Potten CS, Loeffler M. Stem cells: attributes, cycles,<br />

spirals, pitfalls and uncertainties. Lessons for and from<br />

the crypt. Development 1990; 110:1001-20.<br />

8. Verdi JM, Schmandt R, Bashirullah A, et al. Mammalian<br />

numb is an evolutionary conserved signaling<br />

adapter protein that specifies cell fate. Curr Biol 1996;<br />

6:1134-45.<br />

9. Zhong WM, Feder JN, Jiang MM, Jan LY, Jan YN. Asymmetric<br />

localization of a mammalian numb homolog<br />

during mouse cortical neurogenesis. Neuron 1996;<br />

17:43-53.<br />

10. Mayani H, Dragowska W, Lansdorp PM. Lineage commitment<br />

in human hemopoiesis involves asymmetric<br />

cell division of multipotent progenitors and does not<br />

appear to be influenced by cytokines. J Cell Physiol<br />

1993; 157:576-9.<br />

11. Till JE, McCulloch EA, Siminovitch L. A stochastic<br />

model of stem cell proliferation, based on the growth<br />

of spleen colony-forming cells. Proc Natl Acad Sci USA<br />

1964; 51:29-33.<br />

12. Trentin JJ. Influence of hematopoietic organ stroma<br />

(hematopoietic inductive microenvironments) on stem<br />

cell differentiation. In: Gordon AS. ed. Regulation of<br />

Hematopoiesis. vol. 1. New York: Appleton, 1970. p.<br />

161-85.<br />

13. Ogawa M. Differentiation and proliferation of<br />

hematopoietic stem cells. Blood 1993; 81:2844-53.<br />

14. Metcalf D. Hematopoietic regulators: redundancy or<br />

subtlety? Blood 1993; 82:3515-23.<br />

15. Ellisen LW, Bird J, West DC, Soreng AL, Reynolds TC,<br />

Smith SD, Sklar J. TAN-1, the human homologue of<br />

the Drosophila notch gene, is broken by chromosomal<br />

translocations in T lymphoblastic neoplasms. Cell<br />

1991; 66:649-61.<br />

16. Seydoux G, Mello CC, Pettitt J, Wood WB, Priess JR,<br />

Fire A. Repression of gene expression in the embryonic<br />

germ lineage of C. elegans. Nature 1996; 382:713-<br />

6.<br />

17. Metcalf D. Lineage commitment of hemopoietic progenitor<br />

cells in developing blast cell colonies: influence<br />

of colony-stimulating factors. Proc Natl Acad Sci USA<br />

1991; 88:11310-4.<br />

18. Pawson T. Protein modules and signalling networks.<br />

Nature 1995; 373:573-80<br />

19. Bedi A, Sharkis SJ. Mechanisms of cell commitment in<br />

myeloid cell differentiation. Curr Opin Hematol 1995;<br />

2:12-21.<br />

20. Williamson EA, Boswell SH. Signal transduction during<br />

myeloid cell differentiation. Curr Opin Hematol<br />

1995; 2:29-40.<br />

21. Sauvageau G, Thorsteinsdottir U, Eaves CJ, et al. Overexpression<br />

of HOXB4 in hematopoietic cells causes the<br />

selective expansion of more primitive populations in<br />

vitro and in vivo. Gens Dev 1995; 9:1753-65.<br />

22. Sauvageau G, Thorsteinsdottir U, Hough MR, et al.<br />

Overexpression of HOXB3 in hematopoietic cells causes<br />

defective lymphoid development and progressive<br />

myeloproliferation. Immunity 1997; 6:13-22.<br />

23. Wessely O, Deiner E-M, Beug H, von Lindern M. The<br />

gluocorticoid receptor is a key regulator of the decision<br />

between self-renewal and differentiation in erythroid<br />

progenitors. EMBO J 1997; 16:267-80.<br />

24. Dotti GP, Carlo-Stella C, Spinelli O, et al. Shc overexpression<br />

induces selective hypersensitivity to GM-CSF<br />

and prevents apoptosis of the GM-CSF-dependent<br />

acute myelogenous leukemia cell line GF-D8. In:<br />

Hematology and blood transfusion. New York: Springer-Verlag,<br />

1997. In press.<br />

25. Rossi F, McNagny KM, Smith G, Frampton J, Graf T.<br />

Lineage commitment of transformed haematopoietic<br />

progenitors is determined by the level of PKC activity.<br />

EMBO J 1996; 15:1894-901.<br />

26. Vaziri H, Dragowska W, Allsopp RC, Thomas TE, Harley<br />

CB, Lansdorp PM. Evidence for a mitotic clock in<br />

human hematopoietic stem cells: loss of telomeric DNA<br />

with age. Proc Natl Acad Sci USA 1994; 91:9857-60.<br />

27. Morrison SJ, Uchida N, Weissman IL. The biology of<br />

hematopoietic stem cells. Annu Rev Cell Dev Biol<br />

1994; 11:35-71.<br />

28. Carlo-Stella C, Cazzola M, De Fabritiis P, et al. CD34-<br />

positive cells: biology and clinical relevance. <strong>Haematologica</strong><br />

1995; 80:367-87.<br />

29. Krause DS, Fackler MJ, Civin CI, Stratford May W.<br />

CD34: structure, biology, and clinical utility. Blood<br />

1996; 87:1-13.<br />

30. Civin CI, Gore SD. Antigenic analysis of hematopoiesis:<br />

a review. J Hematother 1993; 2:137-44.<br />

31. Baumhueter S, Singer MS, Henzel W, et al. Binding of<br />

L-selectin to the vascular sialomucin CD34. Science<br />

1993; 262:436-41.<br />

32. Gunji Y, Nakamura M, Osawa H, et al. Human primitive<br />

hematopoietic progenitor cells are more enriched<br />

in KIT low cells than in KIT high cells. Blood 1993;<br />

82:3283-9.<br />

33. Small D, Levenstein M, Kim E, et al. STK-1, the human<br />

homologue of Flk-2/Flt-3, is selectively expressed in<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

111<br />

CD34+ human bone marrow cells and is involved in<br />

the proliferation of early progenitor/stem cells. Proc<br />

Natl Acad Sci USA 1994; 91:459-63.<br />

34. Zanjani ED, Almeida-Porada G, Leary AG, Ogawa M.<br />

Human bone marrow CD34- cells engraft in vivo and<br />

undergo multilineage expression including giving rise to<br />

CD34+ cells. Blood 1997; 90:252a.<br />

35. Larochelle A, Vormoor J, Hanenberg H, et al. Identification<br />

of primitive hematopoietic cells capable of<br />

repopulating NOD/SCID mice: implications for gene<br />

therapy. Nature Med 1996; 2:1329-37.<br />

36. Bhatia M, Wang JCY, Kapp U, Bonnet D, Dick JE.<br />

Purification of primitive human hematopoietic cells<br />

capable of repopulating immunodeficient mice. Proc<br />

Natl Acad Sci USA 1997; 94:5320-5.<br />

37. Eaves CJ, Cashman JD, Eaves AC. Methodology of<br />

long-term culture of human hematopoietic cells. J Tiss<br />

Cult Methods 1991; 13:55-62.<br />

38. Sutherland HJ, Eaves CJ, Eaves AJ, Dragowskas W,<br />

Lansdorp PM. Characterization and partial purification<br />

of human marrow cells capable of initiating longterm<br />

hematopoiesis in vitro. Blood 1989; 74:1563-70.<br />

39. Petzer AL, Hogge DE, Lansdorp PM, Reid DS, Eaves CJ.<br />

Self-renewal of primitive human hematopoietic cells<br />

(long-term-culture-initiating cells) in vitro and their<br />

expansion in defined medium. Proc Natl Acad Sci USA<br />

1996; 93:1470-4.<br />

40. Breems DA, Blokland EAW, Neben S, Ploemacher RE.<br />

Frequency analysis of human primitive haematopoietic<br />

stem cell subsets using a cobblestone area forming<br />

cell assay. Leukemia 1994; 8:1095-104.<br />

41. Gordon MY, Amos TAS. Stochastic effects in hemopoiesis.<br />

Stem Cells 1994; 12:175-9.<br />

42. Briddell RA, Kern BP, Zilm KL, Stoney GB, McNiece<br />

IK. Purification of CD34+ cells is essential for optimal<br />

ex vivo expansion of umbilical cord blood cells. J<br />

Hematother 1997; 6:145-50.<br />

43. Haylock D, Simmons P, To LB, Juttner C. Growth factors<br />

and ex vivo expansion of hemopoietic progenitor<br />

cells. In: Morstyn G, Sheridan W. eds. Cell Therapy.<br />

Cambridge: Cambridge University Press, 1996. p. 221-<br />

37.<br />

44. Alcorn MJ, Holyoake TL, Richmond L, et al. CD34-<br />

positive cells isolated from cryopreserved peripheralblood<br />

progenitor cells can be expanded ex vivo and<br />

used for transplantation with little or no toxicity. J CIin<br />

Oncol 1996; 14:1839-47.<br />

45. Brugger W, Heimfeld S, Berenson RJ, Mertelsmann R,<br />

Kanz L. Reconstitution of hematopoiesis after highdose<br />

chemotherapy by autologous progenitor cells<br />

generated ex-vivo. N Engl J Med 1995; 333:283-7.<br />

46. Lemoli RM, Fortuna A, Motta MR, et al. Concomitant<br />

mobilization of plasma cells and hematopoietic progenitors<br />

into peripheral blood of multiple myeloma<br />

patients: Positive selection and transplantation of<br />

enriched CD34+ cells to remove circulating tumor<br />

cells. Blood 1996; 87:1625-32.<br />

47. Gazitt Y, Reading C, Hoffman R, et al. Purified CD34+<br />

Lin- Thy+ stem cells do not contain clonal myeloma<br />

cells. Blood 1995; 86:381-9.<br />

48. Maguer-Satta V, Petzer AL, Eaves AC, Eaves CJ. BCR-<br />

ABL expression in different subpopulations of functionally<br />

characterized Ph+ CD34+ cells from patients<br />

with chronic myeloid leukemia. Blood 1996; 88:1796-<br />

804.<br />

49. Fogli M, Amabile M, Martinelli G, et al. Selective<br />

expansion of normal haemopoietic progenitors from<br />

chronic myelogenous leukaemia marrow. Br J Haematol<br />

1998; 101:119-29.<br />

50. Peters D, Branscomb E, Dean P, et al. The LLNL highspeed<br />

sorter: design, features, operational characteristics<br />

and biological utility. Cytometry 1985; 6:290-<br />

301.<br />

51. Tsukamoto A, Sasaki D, Chen BP, Hoffman R. Characterization<br />

and isolation of mobilized peripheral<br />

blood stem cells using a high-speed cell sorter. In:<br />

Morstyn G, Sheridan W, eds. Cell Therapy. Cambridge:<br />

Cambridge University Press, 1996. p. 183-98.<br />

52. Okarma T, Lebkowski J, Schain L, et al. The AIS collector:<br />

a new technique for stem cell purification. In: Worthington-White<br />

DA, Gee AP, Gross S, eds. Advances in<br />

bone marrow purging and processing. New York: Wiley<br />

Liss, 1992. p. 449-59.<br />

53. Kemshead JT, Ugelstad J. Magnetic separation techniques:<br />

their application to medicine. Mol Cell Biochem<br />

1985; 67:11-8.<br />

54. Kvalheim G, Sorensen O, Fodstad O, et al. Immunomagnetic<br />

removal of B-lymphoma cells from human<br />

bone marrow: A procedure for clinical use. Bone Marrow<br />

Transpl 1988; 3:31-41.<br />

55. Civin CI, Strauss LC, Facker MJ, Trismann TM, Wiley<br />

JM, Loken RL. Positive stem cell selection. Basic science.<br />

In: Gross S, Gee A, Worthington-White DA, eds.<br />

Bone marrow purging and processing. New York.<br />

Wiley-Liss, 1990. p. 387-402.<br />

56. Milteny S, Mueller W, Weichel W, Radbruch A. High<br />

gradient magnetic cell separation with MACS. Cytometry<br />

1990; 11:231-8.<br />

57. Lemoli RM, Tafuri A, Fortuna A, et al. Cycling status<br />

of CD34+ cells mobilized into peripheral blood of<br />

healthy donors by recombinant human granulocyte<br />

colony-stimulating factor. Blood 1997; 89:1189-96.<br />

58. Link H, Arseniev L, Bahre O, et al. Combined transplantation<br />

of allogeneic bone marrow and CD34+<br />

blood cells. Blood 1995; 86:2500-8.<br />

59. Koller MR, Emerson SG, Palsson BO. Large scale<br />

expansion of human stem and progenitor cells from<br />

bone marrow mononuclear cells in continuous perfusion<br />

cultures. Blood 1993; 82:378-84.<br />

60. Reisner Y, Martelli MF. Bone marrow transplantation<br />

across HLA barriers by increasing the number of transplanted<br />

cells. Immunol Today 1995; 16:437-40.<br />

61. Moore MAS: Expansion of myeloid stem cells in culture.<br />

Semin Hematol 1995; 32:183-92.<br />

62. Emerson GE. Ex vivo expansion of hematopoietic precursors,<br />

progenitors and stem cells: the next generation<br />

of cellular therapeutics. Blood 1996; 87:3082-8.<br />

63. Yonemura Y, Ku H, Hirayama F, Souza LM, Ogawa M.<br />

Interleukin 3 or interleukin 1 abrogates the reconstituting<br />

ability of hematopoietic stem cells. Proc Natl<br />

Acad Sci USA 1995; 93:4040-4.<br />

64. Peters SO, Kittler ELW, Ramshaw HS, Quesenberry PJ.<br />

Ex vivo expansion of murine marrow cells with IL-3, IL-<br />

6, IL-11 and SCF leads to impaired engraftment in irradiated<br />

hosts. Blood 1996; 87:30-7.<br />

65. Traycoff CM, Cornetta K, Yoder MC, Davidson A,<br />

Srour EF. Ex vivo expansion of murine hematopoietic<br />

progenitor cells generates classes of cells possessing,<br />

different leveIs of bone marrow repopulating potential.<br />

Exp Hematol 1990; 24:299-306.<br />

66. Young JC, Varma A, DiGiusto D, Backer MP. Retention<br />

of quiescent hematopoietic cells with high proliferative<br />

potential during ex vivo stem cell culture. Blood 1996;<br />

87:545-56.<br />

67. Di Giusto DL, Lee R, Moon K, et al. Hematopoietic<br />

potential of cryopreserved and ex vivo manipulated<br />

umbilical cord blood progenitor cells evaluated in vitro<br />

and in vivo. Blood 1996; 87:1261-71.<br />

68. Gabutti V, Foà R, Mussa F, Aglietta M. Behaviour of<br />

human haematopoietic stem cells in cord and neonatal<br />

blood. <strong>Haematologica</strong> 1975; 60:4.<br />

69. Lansdorp PM, Dragowska W, Mayani H. Ontogenyrelated<br />

changes in proliferative potential of human<br />

hematopoietic celIs. J Exp Med 1993; 178:787-91.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


112<br />

M. Aglietta et al.<br />

70. Zandstra PW, Eaves CJ, Piret JM. Expansion of<br />

hematopoietic progenitor cell populations in stirred<br />

suspension bioreactors of normal human bone marrow<br />

cells. Biotechnology 1994; 12:909-14.<br />

71. Bertolini F, Battaglia M, Corsini C, et al. Engineered<br />

stromal layers and continuous flow culture enhance<br />

multidrug resistance gene transfer in hematopoietic<br />

progenitors. Cancer Res 1996; 56:2566-72.<br />

72. Keller JR, Bartelmez SH, Sitnicka E, et al. Distinct and<br />

overlapping direct effects of macrophage inflammatory<br />

protein 1 and transforming growth factor- on<br />

hematopoietic progenitor/stem cell growth. Blood<br />

1994; 84:2175-81.<br />

73. Van Reenst PCF, Snoeck HW, Lardon F, et al. TGF-<br />

and MIP-1 extert their main inhibitory activity on very<br />

primitive CD34 ++ CD38 – cells but show opposite effects<br />

on more mature CD34 + CD38 + human progenitors.<br />

Exp Hematol 1997; 24:1509-15.<br />

74. Matthews W, Jordan CT, Wiegand GW, Pardoll D,<br />

Lemischka IR. A receptor tyrosine kinase specific to<br />

hemopoietic stem cell-enriched populations. Cell<br />

1991; 65:1143-52.<br />

75. Shah AJ, Smogorzewska EM, Hannum C, Crooks GM.<br />

Flt3 ligand induces proliferation of quiescent human<br />

bone marrow CD34+CD38- cells and maintains progenitor<br />

cells in vitro. Blood 1996; 87:3563-70.<br />

76. Piacibello W, Fubini L, Sanavio F, et al. Effect of<br />

human FLT3 ligand on myeloid leukemia cells growth:<br />

heterogeneity in response and synergy with other<br />

hematopoietic growth factors. Blood 1995; 86:4105-<br />

14.<br />

77. Piacibello W, Garetto L, Sanavio F, et al. The effects of<br />

human FLT3 ligand on in vitro human megakaryocytopoiesis.<br />

Exp Hematol 1996; 24:340-6.<br />

78. Bertolini F, Battaglia M, Pedrazzoli P, et al. Megakaryocytic<br />

progenitors can be generated ex vivo and safely<br />

administered to autologous peripheral blood progenitor<br />

cell transplant recipients. Blood 1997; 89:2679-<br />

88.<br />

79. Petzer Al, Zandstra PW, Piret JM, Eaves CJ. Differential<br />

cytokine effects on primitive (CD34+CD38-) human<br />

hematopoietic cells: Novel responses to Flt3-ligand<br />

and thrombopoietin. J Exp Med 1996; 183:2551-8.<br />

80. Yonemura Y, Ku H, Lyman SD, Ogawa M. In vitro<br />

expansion of hematopoietic progenitors and maintenance<br />

ot stem cells: comparison between FLT3/FLK-2<br />

ligand and Kit ligand. Blood 1997; 89:1915-21.<br />

81. Moore TA, Zlotnik A. Differential effects of Flk2/Flt-3<br />

ligand and stem cell factor on murine thymic progenitor<br />

cells. J Immunol 1997; 158:4187-92.<br />

82. Dao MA, Hannum CH, Kohn DB, Nolta JA. FLT3 ligand<br />

preserves the ability of human CD34+ progenitors<br />

to sustain long-term hematopoiesis in immune-deficient<br />

mice after ex vivo retroviral mediated transduction.<br />

Blood 1997; 89:446-56.<br />

83. Zandstra PW, Conneally E, Petzer AL, Piret JM, Eaves<br />

CJ. Cytokine manipulation of primitive human hematopoietic<br />

cell self-renewal. Proc Natl Acad Sci USA<br />

1997; 94:4698-703.<br />

84. Kaushansky K. Thrombopoietin: The primary regulator<br />

of platelet production. Blood 1995; 86:419-31.<br />

85. Kaushanski K, Broudy VC, Lin N, et al. Thrombopoietin,<br />

the mpl ligand, is essential for full megakaryocyte<br />

development. Proc Natl Acad Sci USA 1995; 92:3234-<br />

38.<br />

86. Goncalves F, Lacout O, Villeval JL, Wendling F,<br />

Vainchenker W, Dumenil D. Thrombopoietin does not<br />

induce lineage-restricted commitment of Mpl-R<br />

expressing pluripotent progenitors but permits their<br />

complete erythroid and megakaryocytic differentiation.<br />

Blood 1997; 89:3544-53.<br />

87. Ramsfjell V, Borge OJ, Cui L, Jacobsen SEW. Thrombopoietin<br />

directly and potently stimulates multilineage<br />

growth and progenitor cell expansion from primitive<br />

(CD34+CD38-) human bone marrow progenitor cells.<br />

J Immunol 1997; 158:5169-77.<br />

88. Ku H, Hirayama F, Kato I, et al. Soluble thrombopoietin<br />

receptor and granulocyte colony-stimulating factor<br />

receptor directly stimulate proliferation of primitive<br />

hematopoietic progenitors of mice in synergy with steel<br />

factor or the ligand for FIt3/Flk2. Blood 1996; 88:<br />

4124-31.<br />

89. Piacibello W, Sanavio F, Garetto L, et al. Extensive<br />

amplification and self-renewal of human primitive<br />

hematopoietic stem cells from cord blood. Blood<br />

1997; 89:2644-53.<br />

90. Uchida N, Aguila HL, Fleming WH, Jerahek I, Weissman<br />

IL. Rapid and sustained hematopoietic recover in<br />

Iethally irradiated mice transplanted with purified<br />

Thy1.Low, Lin-Sca-1+ hematopoietic stem cells. Blood<br />

1994; 83:3758-79.<br />

91. Szilvassy SJ, et al. Partially differentiated ex vivo expanded<br />

celIs accelerate hematologic recovery in myeloablated<br />

mice transplanted with highly enriched long-term<br />

repopulating stem cells. Blood 1996; 88:3642-53.<br />

92. Williams SF, Lee WJ, Bender JG, et al. Selection and<br />

expansion of peripheral blood CD34+ cells in autologous<br />

stem cell transplantation for breast cancer. Blood<br />

1996; 87:1687-91.<br />

93. Widmer L, Pichert G, Jost LM, Stahel RA. Fate of contaminating<br />

t(14,18)+ lymphoma cells during ex vivo<br />

expansion of CD34-selected hematopoietic progeritor<br />

cells. Blood 1996; 88:3166-75.<br />

94. Vogel W, Behringer D, Scheding S, Kanz L , Brugger W.<br />

Ex vivo expansion of CD34+ peripheral blood progenitor<br />

cells: Implications for the expansion of contaminating<br />

epithelial tumor cells. Blood 1996; 88:2707-<br />

13.<br />

95. Whiteside TL, Rinaldo CR jr, Herberman RB. Cytolytic<br />

cell functions. On "Manual of Clinical Laboratory<br />

Immunology". 4 th ed. Am Soc Microbiol Washington<br />

D.C., 1992. p. 220-30.<br />

96. Torpey DJ III, Lindsley MD, Rinaldo CR Jr. HLA-restricted<br />

lysis of herpes simplex virus-infected monocytes and<br />

macrophages mediated by CD4+ and CD8+ T lymphocytes.<br />

J Immunol 1989; 142:1325-32.<br />

97. Kagi D, Vignaux F, Ledermann B, et al. Fas and perforin<br />

pathways as major mechanisms of T cell-mediated<br />

cytotoxicity. Science 1994, 265:528-30.<br />

98. Montel AH, Bochan MR, Hobbs JA, Lynch DH, Brahmi<br />

Z. Fas involvement in cytotoxicity mediated by<br />

human NK cells. Cell Immunol 1995, 166:236-46.<br />

99. Braun MY, Lowin B, French L, Acha Orbea H, Tschopp<br />

J. Cytotoxic T cell deficient in both functional Fas ligand<br />

and perforin show residual cytolytic activity yet<br />

lose their capacity to induce lethal acute graft versus<br />

host disease. J Exp Med 1996, 183:657-61.<br />

100. Cascino I, Papoff G, De Maria R, Testi R, Ruberti G.<br />

Fas/Apo-1 (CD-95) receptor lacking the intracytoplasmic<br />

signaling domain protects tumor cells from<br />

Fas-mediated apoptosis. J Immunol 1996; 156:13-7.<br />

101. Lanier LL, Phillips JH. Inhibitory MHC class I receptors<br />

on NK cells and T cells. Immunol Today 1996; 17:86-<br />

92.<br />

102. Kanegane H, Tosato G. Activation of naive and memory<br />

T cells by Interleukin-15. Blood 1996; 88:230-5.<br />

103. Richard C, Alsar MJ, Calavia J, et al. Recombinant<br />

human GM-CSF enhances T cell mediated cytotoxic<br />

function after ABMT for hematological malignancies.<br />

Bone Marrow Trasplant, 1993; 11:473-8.<br />

104. Bendall LJ, Kortlepel, Gottlieb DJ. GM-CSF enhances<br />

IL-2-activated natural killer cell lysis of clonogenic AML<br />

cells by upregulating target cell expression of ICAM-1.<br />

Leukemia 1995; 9:677-84.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

113<br />

105. Cantori I, Olivieri A, Rupoli S, et al. The association of<br />

IL-2 plus GM-CSF has protective effects and reduces<br />

the apoptosis in liquid culture. Blood 1996; 88:112a.<br />

106. Trinchieri G. Interleukin-12: a cytokine produced by<br />

antigen presenting cells with immunoregulatory functions<br />

in the generation of T-helper cells type 1 and<br />

cytotoxic lymphocytes. Blood 1994, 84:4008-27.<br />

107. Gerosa F, Paganin C, Peritt D, et al. Intereleukin-12<br />

primes human CD4 and CD 8 T cell clones for high<br />

production of both interferon- and interleukin-10. J<br />

Exp Med 1996, 183:2559-69.<br />

108. Abdul-Hai A, Or R, Slavis S, et al. Stimulation of<br />

immune reconstitution by interleukin-7 after syngeneic<br />

bone marrow transplantation in mice. Exp Hematol,<br />

1996; 24:1416-22.<br />

109. Mavroudis D, Barret J. The graft-versus-leukemia<br />

effect. Current Opinion Hematol 1996: 3:423-29.<br />

110. Glass B, Uharek L, Zeis M, et al. Graft-versus-leukemia<br />

activity can be predicted by natural cytotoxicity against<br />

leukemia cells. Br J Haematol 1996; 93:412-20.<br />

111. Goldman JM., Gale RP., Horowitz MM, et al. Bone<br />

marrow transplantation for chronic myelogenous<br />

leukemia in chronic phase: increased risk for relapse<br />

associated with T cell depletion. Ann Intern Med 1988;<br />

108:806-14.<br />

112. Kolb HJ, Schattenberg A, Goldman JM, et al. Graftversus-leukemia<br />

effect of donor lymphocyte transfusion<br />

in marrow grafted patients. Blood 1995; 86:2041-<br />

50.<br />

113. Hess AD, Bright EC, Thoburn C, Vogelsang GB, Jones<br />

RJ, Kennedy MJ. Specificity of effector T lymphocytes<br />

in autologous graft-versus-host disease: Role of the<br />

major histocompatibility complex class II invariant<br />

chain peptide. Blood, 1997; 89:2203-9.<br />

114. Hokland M, Jacobsen N, Ellegaard J, Hokland P. Natural<br />

killer function following allogeneic bone marrow<br />

transplantation. Transplantation 1988; 45:1080-4.<br />

115. Bengtsson M, Totterman TH, Smedmyr B, Festin R,<br />

Simonseen B. Regeneration of functional and activated<br />

NK and T subset cells in the marrow and blood<br />

after autologous bone marrow transplantation. A<br />

prospective phenotypic study with 2/3-color FACS<br />

analysis. Leukemia 1989; 3:68-75.<br />

116. Jorgensen H, Hokland P, Jensen T, Basse P, Hokland<br />

M. Natural killer cell in peripheral blood after autologous<br />

bone marrow transplantation. Nature Immun<br />

1995; 14:164-72.<br />

117. Timonen T, Maenpaa A, Helander T, Malygin A,<br />

Jaaskelainen J. Adhesion molecules in the binding and<br />

infiltration of human natural killer cells. In: Gahmberg<br />

CG, Mandrup-Poulsen T, Wognsen Bach L, Hokfelt B,<br />

eds. Leukocyte adhesion: basic and clinical aspects.<br />

proc 6 th Novo Nordisk Foundation Symposium "Leukocyte<br />

adhesion" 1992. p. 353-60.<br />

118. Miller JS, Alley KA, McGlave PB. Differentiation of natural<br />

killer cells from human primitive marrow progenitors<br />

in a stroma based long term culture system: identification<br />

of a CD34+/CD7+ NK progenitor. Blood<br />

1994; 83:2594-601.<br />

119. Mrozek E, Anderson P, Caligiuri MA. Role of interleukin-15<br />

in the development of human CD56+ natural<br />

killer cells from CD34+ hematopoietic progenitor<br />

cells. Blood 1996; 87:2632-40.<br />

120. Pierson BA, Miller JS. CD56+bright and CD56+dim<br />

natural killer cells in patients with chronic myelogenous<br />

leukemia progessively decrease in number,<br />

respond less to stimuli that recruit clonogenic natural<br />

killer cells, and exhibit decreased proliferation on a per<br />

cell basis. Blood 1996; 88:2279-87.<br />

121. Daniels B, Daniels B, Karlhofer FM, Seaman WE,<br />

Yokoama W. A natural killer cell receptor specific for<br />

a major histocompatibility complex class I molecule. J<br />

Exp Med 1994; 180:687-92.<br />

122. Moretta A, Vitale M, Sivori S, et al. Human natural<br />

killer cell receptors for HLA-class I molecules. Evidence<br />

that the kp43 (CD94) molecule functions as receptor<br />

for HLA-B alleles. J Exp Med 1994; 180:545-55.<br />

123. Rosenberg SA, Lotze MT, Muul LM, et al. A progress<br />

report on the treatment of 157 patients with advanced<br />

cancer using lymphokine-activated killer cells and interleukin-2<br />

or high-dose interleukin-2 alone. N Engl J Med<br />

1987; 316:889-97.<br />

124. Parrado A, Rodriguez-Fernadez JM, Casares S, et al.<br />

Generation of LAK cells in vitro in patients with acute<br />

leukemia. Leukemia 1993; 7:1344-8.<br />

125. Attal M, Blaise D, Marit G, et al. Consolidation treatment<br />

of adult acute lymphoblastic leukemia: a<br />

prospective, randomized trial comparing allogeneic versus<br />

autologous bone marrow transplantation and testing<br />

the impact of recombinant interleukin-2 after autologous<br />

bone marrow transplantation. Blood 1995;<br />

86:1619-28.<br />

126. Klingemann HG, Deal H, Reid D, Eaves CJ. Design and<br />

validation of a clinically applicable culture procedure<br />

for the generation of interleukin-2 activated natural<br />

killer cells in human bone marrow autografts. Exp<br />

Haematol 1993; 21:1263-70.<br />

127. Koo GC, Peppard JR, Latime EC. Characterization of<br />

cytotoxic cells generated from bone marrow culture.<br />

Cell Immunol 1986; 98:172-80.<br />

128. Silva MRG, Hoffman R, Srour EF, Ascensao JL. Generation<br />

of human natural killer cells from immature<br />

progenitors does not require marrow stromal cells.<br />

Blood 1994; 84:841-6.<br />

129. Lotzova E, Savary CA, Champlin RE. Genesis of human<br />

oncolytic natural killer cells from primitive CD34+<br />

CD33- bone marrow progenitors. J Immunol 1993;<br />

150:5263-9.<br />

130. Miller JS, Verfaille C, McGlave PB. The generation of<br />

human natural killer cells from CD34+/DR- primitive<br />

progenitors in long-term bone marrow culture. Blood<br />

1992; 80:2182-7.<br />

131. Silva MRG, Parreira A, Ascensao JL. Natural killer cell<br />

numbers and activity in mobilized peripheral blood<br />

stem cell grafts: conditions for in vitro expansion. Exp<br />

Hematol, 1995; 23:1676-81.<br />

132. Miller JS, Prosper F, MC Cullar V. Natural Killer cells<br />

are functionally abnormal and NK cell progenitors are<br />

diminished in granulocyte colony-stimulating factormobilized<br />

peripheral blood progenitor cell collections.<br />

Blood 1997; 90:3098-105.<br />

133. Hercend T, Farace F, Baume D, et al. Immunotherapy<br />

with lymphokine-activated natural killer cells and<br />

recombinant interleukin-2: a feasibility trial in metastatic<br />

renal cell carcinoma. J Biol 1990; 9:546-55.<br />

134. Beaujean F, Bernaudin F, Kuentz M, et al. Successful<br />

engraftment after autologous transplantation of 10-<br />

day cultured bone marrow activated by interleukin 2 in<br />

patients with acute lymphoblastic leukemia. Bone<br />

Marrow Transplant 1995; 15:691-6.<br />

135. Wong EK, Eaves C, Klingemann HG. Comparison of<br />

natural killer activity of human bone marrow and<br />

blood cells in culture containing IL-2, IL-7 and IL-12.<br />

Bone Marrow Transplant 1996; 18:63-71.<br />

136. Miller JS, Klingsporn S, Lund J, et al. Large-scale ex vivo<br />

expansion and activation of human natural killer cells<br />

for autologous therapy. Bone Marrow Transplant<br />

1994; 14: 555-62.<br />

137. Pierson BA, Europa AF, Hu WS, Miller JF. Production<br />

of human natural killer cells for adoptive immunotherapy<br />

using a computer-controlled stirred-tank bioreactor.<br />

J Hematother 1996; 5:475-83.<br />

138. Vujanovic NL, Yasumura S, Hirabayashi H, et al. Antitumor<br />

activities of human IL-2 activated natural killer<br />

haematologica vol. 85(suppl. to n. 12):December 2000


114<br />

M. Aglietta et al.<br />

cells in solid tumor tissues. J Immunol 1995; 154:281-<br />

9.<br />

139. Lanier LL, Civin CI , Loken MR, Phillips JH. The relationship<br />

of CD16 (Leu-11) and Leu-19 (NKH-1) antigen<br />

expression on human peripheral blood NK cells<br />

and cytotoxic T lymphocytes. J Immunol 1986; 136:<br />

4480-6.<br />

140. Schmidt-Wolf IGH, Lefterova P, Johnston V, Huhn D,<br />

Blume KG, Negrin RS. propagation of large numbers<br />

of T cells with natural killer cell markers. B J Haematol<br />

1994; 87:453-8.<br />

141. Scheffold C, Brandt K, Johnston V, et al. Potential of<br />

autologous immunologic effector cells for bone marrow<br />

purging in patients with chronic myeloid leukemia.<br />

Bone Marrow Transplant 1995; 15:33-9.<br />

142. Slavin S, Ackerstein A, Weiss L, Nagler A, Reuven O,<br />

Naparstek E. Immunotherapy of minimal residual disease<br />

by immunocompetent lymphocytes and their activation<br />

by cytokines. Cancer Invest 1992; 10:221-7.<br />

143. Glass B, Uhrek L, Zeis M, Loefler H, Mueller-Ruchholtz<br />

W, Gassmann W. Graft-versus-leukaemia activity can<br />

be predicted by natural cytotoxicity against leukaemia<br />

cells. Br J Haematol 1996; 93:412-20.<br />

144. Rocha M, Umansky V, Lee KH, Hacker HJ, Benner A,<br />

Schirrmacher V. Difference between graft-versusleukemia<br />

and graft-versus-host reactivity. I. Interaction<br />

of donor immune T cells with tumor and/or Host cells.<br />

Blood 1997; 89:2189-202.<br />

145. Van der Harst D, Goulmy E, Falkenburg JHF, et al.<br />

Recognition of minor histocompatibility antigens on<br />

lymphocytic and myeloid leukemic cells by cytotoxic<br />

T-cell clones. Blood 1994; 83:1060-6.<br />

146. Hoffmann T, Theobald M, Bunjes D, Weiss M, Heimpel<br />

H, Heit W. Frequency of bone marrow T cells<br />

responding to HLA-identical non-leukemic and<br />

leukemic stimulator cells. Bone Marrow Transplant<br />

1993; 12:1-8.<br />

147. Kolb HJ, Mittermueller J, Clemm CH, et al. Donor<br />

leukocyte trasfusions for treatment of recurrent chronic<br />

myelogenous leukemia in marrow transplant<br />

patients. Blood 1990; 76:2462-5.<br />

148. Champlin R. Separation of graft-vs.-host disease and<br />

graft-vs-leukemia effect against chronic myelogenous<br />

leukemia. Exp Hematol 1995; 23:1148-51.<br />

149. Datta AR, Barrett AJ, Jang YZ, et al. Distinct T cell populations<br />

distinguish chronic myeloid leukaemia cells<br />

from lymphocytes in the same individual: a model for<br />

separating GVHD from GVL reactions. Bone Marrow<br />

Transplant 1994; 14:517-24.<br />

150. Oettel KR, Wesly OH, Albertini MR, et al. Allogeneic<br />

T-cell clones able to selectively destroy Philadelphia<br />

chromosome-bearing (Ph1+) human leukemia lines<br />

can also recognize Ph1- cells from the same patient.<br />

Blood 1994; 83:3390-402.<br />

151. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of<br />

patients with B-cell lymphoma using autologous antigen-pulsed<br />

dendritic cells. Nature Med 1996; 2:52-8.<br />

152. Rooney CM, Smith CA, Loftin S, et al. Use of genemodified<br />

virus-specific T lymphocytes to control Epstein-Barr-virus-related<br />

lymphoproliferation. Lancet<br />

1995; 345:9-13.<br />

153. Walter EA, Greenberg PD, Gilbert MJ, et al. Reconstitution<br />

of cellular immunity against cytomegalovirus in<br />

recipients of allogenic bone marrow by transfer of T-<br />

cell clones from the donor. N Engl J Med 1995; 333:<br />

1038-44.<br />

154. Slavin S, Naparstek E, Nagler A, et al. Allogeneic cell<br />

therapy with donor peripheral blood cells and recombinant<br />

human interleukin-2 to treat leukemia relapse<br />

after allogeneic bone marrow transplantation. Blood<br />

1996; 87:2195-204.<br />

155. Weiner GJ, De Gast GC. Bispecific monoclonal antibody<br />

therapy of b-cell malignancy. Leuk Lymphoma<br />

1995; 16:199-207.<br />

156. Disis ML, Bernhard H, Shiota FM, et al. Granulocyte<br />

macrophage colony-stimulating factor: an effective<br />

adjuvant for protein and peptide-based vaccines.<br />

Blood 1996; 88:202-10.<br />

157. Faber LM, Van Luxemburg-Heijs SAP, Veenhof WFJ,<br />

Willemze R, Falkenburg JHF. Generation of CD4+ cytotoxic<br />

T-lymphocyte clones from patient with severe<br />

graft-versus-host disease after allogenic bone marrow<br />

transplantation: implication for graft-versus-leukemia<br />

reactivity. Blood 1995; 86:2821-8.<br />

158. Bonini C, Verzelletti S, Servida P, et al. Transfer of the<br />

HSV-TK gene into donor peripheral blood lymphocytes<br />

for in vivo immunomodulation of donor antitumor<br />

immunity after alloBMT. Blood 1994; 84:110a.<br />

159. Steinman RM. Dendritic cells and immune-based therapies.<br />

Exp Hematol 1996; 24:859-62.<br />

160. Lanzavecchia A. Identifying strategies for immune<br />

intervention. Science 1993; 260:937-44.<br />

161. Wong J. Dendritic cells reach out to the clinic. Nature<br />

Med 1997; 3:129.<br />

162. Young JW, Inaba K. Dendritic as adjuvants for class I<br />

major histocompatibility complex-restricted antitumor<br />

immunity. J Exp Med 1996; 183:7-11.<br />

163. Reid CDL. The dendritic cell lineage in haemopoiesis.<br />

Br J Haematol 1997; 96:217-33.<br />

164. Galy A, Travis M, Cen D, Chen B. Human T, B, natural<br />

killer, and dendritic cells arise from a common bone<br />

marrow progenitor cell subset. Immunity 1995; 3:459-<br />

73.<br />

165. Austyn JM. New insights into the mobilization and<br />

phagocytic activity of dendritic cells. J Exp Med 1996;<br />

183:1287-92.<br />

166. Gluckman JC, Canque B, Chapuis F, Rosenzwajg. In<br />

vitro generation of human dendritic cells and cell therapy.<br />

Cytokin Cell Mol Ther 1997; 3:187-96.<br />

167. Reid CDL, Stackpole A, Meager A, Tikerpae J. Interactions<br />

of tumor necrosis factor with granulocytemacrophage<br />

colony-stimulating factor and other<br />

cytokines in the regulation of dendritic cell growth in<br />

vitro from early bipotent CD34+ progenitors in human<br />

bone marrow. J Immunol 1992; 149:2681-8.<br />

168. Caux C, Dezutter-Dambuyant C, Schmitt D, Bancherau<br />

J. GM-CSF and TNF- cooperate in the generation<br />

of dendritic Langerhans cells. Nature 1992; 360:258-<br />

61.<br />

169. Romani N, Gruner S, Brang D, et al. Proliferating dendritic<br />

cell progenitors in human blood. J Exp Med<br />

1994; 180: 83-93.<br />

170. Siena S, Di Nicola M, Bregni M, et al. Massive ex-vivo<br />

generation of functional dendritic cells from mobilized<br />

CD34+ blood progenitors for anticancer therapy. Exp<br />

Hematol 1995; 23:1463-71.<br />

171. Szabolcs P, Moore MAS, Young JW. Expansion of<br />

immunostimulatory dendritic cells among the myeloid<br />

progeny of human CD34+ bone marrow precursors<br />

cultured with c-kit ligand, granulocyte-macrophage<br />

colony-stimulating factor, and TNF-. J Immunol<br />

1995; 154:5851-61.<br />

172. Bernhard H, Disis ML, Heimfeld S, Hand S, Gralow<br />

JR, Cheever JR. Generation of immunostimulatory dendritic<br />

cells from human CD34+ hematopoietic progenitor<br />

cells of the bone marrow and peripheral blood.<br />

Cancer Res 1995; 55:1099-104.<br />

173. Mackensen A, Herbst B, Kohler G, et al. Delineation of<br />

the dendritic cell lineage by generation of large numbers<br />

of Birbeck granule-positive Langerhans cells from<br />

human peripheral blood progenitors cells in vitro.<br />

Blood 1995; 86:2699-707.<br />

174. Rosenzwajg M, Canque B, Gluckman JC. Human dendritic<br />

cell differentiation pathway from CD34+ hema-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Clinical use of hematopoietic stem cells<br />

115<br />

topoietic precursor cells. Blood 1996; 87:535-44.<br />

175. Fisch P, Kohler G, Garbe A, et al. Generation of antigen-presenting<br />

cells for soluble protein antigens exvivo<br />

from peripheral blood CD34+ hematopoietic<br />

progenitor cells in cancer patients. Eur J Immunol<br />

1996; 26:595-600.<br />

176. Szabolcs P, Avigan D, Gezelter S, et al. Dendritic cells<br />

and macrophages can mature independently from a<br />

human bone marrow derived post-colony-forming unit<br />

intermediate. Blood 1996; 87:4520-30.<br />

177. Mortarini R, Di Nicola M, Siena S, et al. Cytokine<br />

requirements for eliciting melanoma specific cytotoxic<br />

T lymphocytes by melanoma antigen peptides<br />

loaded on CD34+ cell-derived versus monocytederived<br />

autologous dendritic cells. <strong>Haematologica</strong><br />

1996; 81 (suppl. to 5):83.<br />

178. Mortarini R, Anichini A, Di Nicola M, et al. Autologous<br />

dendritic cells derived from CD34+ progenitors and<br />

from monocytes are not functionally equivalent antigen<br />

presenting cells in the induction of Melan-<br />

A/MART-127-35-specific cytotoxic T-lymphocytes<br />

from peripheral blood lymphocytes of melanoma<br />

patients with low frequency of cytotoxic T-lymphocyte<br />

precursors. Cancer Res 1998; in press.<br />

179. Thurner M, Papesh C, Ramoner R, et al. In vitro generation<br />

of CD83+ human blood dendritic cells for<br />

active tumor immunotherapy. Exp Hematol 1997;<br />

25:232-7.<br />

180. Sallusto F, Lanzavecchia A. Efficient presentation of<br />

soluble antigen by cultured human dendritic cells is<br />

mantained by granulocyte/macrophage colony-stimulating<br />

factor plus interleukin 4 and downregulated by<br />

tumor necrosis-. J Exp Med 1994; 179:1109-18.<br />

181. Mukherji B, Chakraborty NG, Yamasaki S, et al. Induction<br />

of antigen-specific cytolytic T cells in-situ in<br />

human melanoma by immunization with synthetic<br />

peptide-pulsed autologous antigen presenting cells.<br />

Proc Natl Acad Sci USA 1995; 92:8078-82.<br />

182. Hu X, Chakraborty NG, Sporn JR. Enhancement of<br />

cytolytic T lymphocyte precursor frequency in melanoma<br />

patients following immunization with MAGE-1<br />

peptide loaded antigen presenting cell-based vaccine.<br />

Cancer Res 1996; 56:2479-83.<br />

183. Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of<br />

melanoma patients with peptide-or tumor lysatepulsed<br />

dendritic cells. Nature Med 1998; 4:328-32.<br />

184. Porgador A, Gilboa E. Bone marrow generated dendritic<br />

cells pulsed a class I-restricted peptide are potent<br />

inducers of cytotoxic T-lymphocytes. J Exp Med 1995;<br />

182:255-60.<br />

185. Strobl H, Riedl E, Scheinecker C, et al. TGF-1 promotes<br />

in vitro development of dendritic cells from<br />

CD34+ hemopoietic progenitors. J Immunol 1996;<br />

157:1499-507.<br />

186. Gianni AM, Bregni M, Siena S, et al. High-dose chemotherapy<br />

and autologous bone marrow transplantation<br />

compared with MACOP-B in aggressive B-cell lymphoma.<br />

N Engl J Med 1997; 336:1290-7.<br />

187. Siena S, Di Nicola M, Mortarini R, et al. Expansion of<br />

immunostimulatory dendritic cells from peripheral<br />

blood of patients with cancer. The Oncologist 1997;<br />

2:65-9.<br />

188. Ratta M, Rondelli D, Fortuna A, et al. Generation and<br />

functional characterization of human dendritic cells<br />

derived from CD34+ cells mobilized into peripheral<br />

blood: Comparison with bone marrow CD34+ cells. Br<br />

J Haematol 1998; in press.<br />

189. Romani N, Reider D, Heuer M, et al. Generation of<br />

mature dendritic cells from human blood. An improved<br />

method with special regard to clinical applicability. J<br />

Immunol Methods 1996; 196:137-51.<br />

190. Grabbe S, Beissert S, Schwarz T, Gransetin RD. Dendritic<br />

cells as initiators of tumor immune responses: A<br />

possible strategy for tumor immunotherapy? Immunol<br />

Today 1995; 16:117-21.<br />

191. Knight SC. Dendritic cells as initiators of tumor immunity.<br />

Immunol Today 1995; 16:547.<br />

192. Nagata S. Fas ligand and immune evasion. Nature<br />

Med 1996; 2:1306-7.<br />

193. Gabrilovich DI, Chen HL, Girgis KR, et al. Production<br />

of vascular endothelial growth factor by human<br />

tumors inhibits the functional maturation of dendritic<br />

cells. Nature Med 1996; 2:1096-103.<br />

194. Gabrilovich DI, Ciernik IF, Carbone DP. Dendritic cells<br />

in antitumor immune responses. 1. Defective antigen<br />

presentation in tumor-bearing hosts. Cell Immunol<br />

1996; 170:101-10.<br />

195. Gabrilovich DI, Nadaf S, Corak J. Dendritic cells in<br />

antitumor immune responses. 2. Dendritic cells grown<br />

from bone marrow precursors, but not mature DCs<br />

from tumor-bearing mice, are effective antigen carriers<br />

in the therapy of established tumors. Cell Immunol<br />

1996; 170:111-9.<br />

196. Steptoe RJ, Thomson AW. Dendritic cells and tolerance<br />

induction. Exp Immunol 1996; 105:397-402.<br />

197. Finkelman FD, Lees AL, Gause WC, Morris SC. Dendritic<br />

cells can present antigen in vivo in a tolerogenic<br />

or immunogenic fashion. J Immunol 1996; 157:1406-<br />

14.<br />

198. Grabbe S, Bruvers S, Gallo RL, Knisely TL, Nazareno R,<br />

Granstein RD. Tumor antigen presentation by murine<br />

epidermal cells. J Immunol 1991; 146:3656-61.<br />

199. Cohen PJ, Cohen PA, Rosenberg SA, Katz SI, Mul JJ.<br />

Murine epidermal Langerhans cells and splenic dendritic<br />

cells present tumor-associated antigens to<br />

primed T cells. Eur J Cancer 1994; 24:315-9.<br />

200. Flamand V, Sornasse T, Demanet C, et al. Murine dendritic<br />

cells pulsed in-vitro with tumor antigen induce<br />

tumor resistance in-vivo. Eur J Immunol 1994; 24:605-<br />

10.<br />

201. Mayordomo JI, Zorina T, Storkus WJ, et al. Bone marrow-derived<br />

dendritic cells pulsed with synthetic tumor<br />

peptides elicit protective and therapeutic antitumor<br />

immunity. Nature Med 1995; 12: 1297-302.<br />

202. Celluzzi CM, Mayordomo JI, Storkus WJ, Lotze MT,<br />

Falo LD. Peptide pulsed dendritic cells induce antigenspeciifc,<br />

CTL-mediated protective tumor immunity. J<br />

Exp Med 1996; 183:283-7.<br />

203. Paglia P, Chiodoni C, Rodolfo M, Colombo MP.<br />

Murine dendritic cells loaded in vitro with soluble protein<br />

prime CTL against tumor antigen in vivo. J Exp<br />

Med 1996; 183:317-22.<br />

204. Marchand M, Weynants P, Rankin E, et al. Tumor<br />

regression responses in melanoma patients treated<br />

with a peptide encoded by gene MAGE-3. Int J Cancer<br />

1995; 63:883-5.<br />

205. Maraskovsky E, Brasel K, Teepe M, et al. Dramatic<br />

increase in the numbers of functionally mature dendritic<br />

cells in flt3 ligand-treated mice: Multiple dendritic<br />

cell subpopulations identified. J Exp Med 1996;<br />

184:1953-62.<br />

206. Lynch DH, Andreasen A, Maraskovsky E, Whitmore J,<br />

Miller RE, Schuh JCL. Flt3 ligand induces tumor regression<br />

and antitumor immune responses in vivo. Nature<br />

Med 1997; 6:625-31.<br />

207. Chen W, Peace DJ, Rovira DK, You S, Cheever MA. T<br />

cell immunity to the joining region of p210 bcr-abl<br />

protein. Proc Natl Acad Sci USA 1992; 89:1468-73.<br />

208. Bocchia M, Wentworth PA, Southwood S, et al. Specific<br />

binding of leukemia oncogene fusion protein peptides<br />

to HLA class I molecules. Blood 1995; 85:2680.<br />

209. Greco G, Fruci D, Accapezzato D, et al. Two bcr-abl<br />

junction peptides bind HLA-A3 molecules and allow<br />

specific induction of human cytotoxic T lymphocytes.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


116<br />

M. Aglietta et al.<br />

Leukemia 1996; 10:693-9.<br />

210. Mannering SI, McKenzie JL, Fearnley DB, Hart DNJ.<br />

HLA-DR1-Restricted bcr-abl (b3a2)-specific CD4+ T<br />

lymphocytes respond to dendritic cells pulsed with<br />

b3a2 peptide and antigen-presenting cells exposed to<br />

b3a2 containing cell lysates. Blood 1997; 90:290-7.<br />

211. Choudhury A, Gajewski JL, Liang JC, et al. Use of<br />

leukemic dendritic cells for the generation of antileukemic<br />

cellular cytotoxicity against Philadelphia<br />

chromosome-positive chronic myelogeneous leukemia.<br />

Blood 1997; 89:1133-42.<br />

212. Dermine S, Bertazzoli C, Marchesi E, et al. Lack of T-<br />

cell mediated recognition of the fusion region of the<br />

pml/RAR- hybrid protein by lymphocytes of acute<br />

promyelocytic leukemia patients. Clin Cancer Res<br />

1996; 2:593.<br />

213. Henderson RA, Nimgaonkar MT, Watkins SC, Finn O:<br />

Human dendritic cells genetically engineered to express<br />

high levels of the human epithelial tumor antigen<br />

mucin (MUC-1). Cancer Res 1996; 56:3763-70.<br />

214. Aicher A, Westermann J, Cayeux S, et al. Successful<br />

retroviral mediated transduction of a reporter gene in<br />

human dendritic cells: Feasibility of therapy with gene<br />

modified antigen presenting cells. Exp Hematol 1997;<br />

25:39-44.<br />

215. Gong J, Chen D, Kashiwaba M, Kufe D. Induction of<br />

antitumor activity by immunization with fusions of<br />

dendritic and carcinoma cells. Nature Med 1997;<br />

5:558-61.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


eview<br />

Cell therapy: achievements and<br />

perspectives<br />

haematologica 2000; 85(supplement to no. 12):117-155<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia e Oncologia<br />

Seragnoli, Policlinico S. Orsola, Via Massarenti 9 40138 Bologna, Italy<br />

Phone: +39-051-390413 – Fax: +39-051-398973 – E-mail:<br />

tura@orsola-malpighi.med.unibo.it<br />

CLAUDIO BORDIGNON,* CARMELO CARLO-STELLA,° MARIO PAOLO<br />

COLOMBO, @ ARMANDO DE VINCENTIIS,^ LUIGI LANATA,^ ROBERTO<br />

MASSIMO LEMOLI, § FRANCO LOCATELLI, ** ATTILIO OLIVIERI,°°<br />

DAMIANO RONDELLI, § PAOLA ZANON,^ SANTE TURA §<br />

*Institute of Hematology, S. Raffaele Hospital, Milan;<br />

°Division of Oncology, Istituto Nazionale dei Tumori, Milan;<br />

@<br />

Experimental Oncology D, Istituto Nazionale dei Tumori,<br />

Milan; ^Dompé Biotec, Milan; § Institute of Hematology and<br />

Medical Oncology, University of Bologna and Policlinico S.<br />

Orsola Bologna; °°Division of Hematology, University of<br />

Ancona, Ancona; ** Department of Pediatrics, University of<br />

Pavia Medical School and IRCCS Policlinico S. Matteo, Pavia;<br />

and Amgen Italia, Milan, Italy<br />

Background and Objectives. Cell therapy can be considered<br />

as a strategy aimed at replacing, repairing, or<br />

enhancing the biological function of a damaged tissue or<br />

system by means of autologous or allogeneic cells. There<br />

have been major advances in this field in the last few<br />

years. This has prompted the Working Group on<br />

Hematopoietic Cells to examine the current utilization of<br />

this therapy in clinical hematology.<br />

Evidence and Information Sources. The method employed<br />

for preparing this review was that of informal consensus<br />

development. Members of the Working Group met three<br />

times, and the participants at these meetings examined a<br />

list of problems previously prepared by the chairman. They<br />

discussed the single points in order to reach an agreement<br />

on different opinions and eventually approved the<br />

final manuscript. Some of the authors of the present<br />

review have been working in the field of cell therapy and<br />

have contributed original papers in peer-reviewed journals.<br />

In addition, the material examined in the present<br />

review includes articles and abstracts published in journals<br />

covered by the Science Citation Index and Medline.<br />

State of the Art. Lymphokine-activated killer (LAK) and<br />

tumor-infiltrating lymphocytes (TIL) have been used since<br />

the ’70s mainly in end-stage patients with solid tumors,<br />

but the clinical benefits of these treatments has not been<br />

clearly documented. TIL are more specific and potent<br />

cytotoxic effectors than LAK, but only in few patients<br />

(mainly in those with solid tumors such as melanoma and<br />

glioblastoma) can their clinical use be considered potentially<br />

useful. Adoptive immunotherapy with donor lymphocyte<br />

infusions has proved to be effective, particularly in<br />

patients with chronic myeloid leukemia, in restoring a<br />

state of hematologic remission after leukemia relapse<br />

occurring following an allograft. The infusion of donor T-<br />

cells can also have a role in the treatment of patients with<br />

Epstein-Barr virus (EBV)-induced post-transplant lymphoproliferative<br />

disorders. However, in this regard, generation<br />

and infusion of donor-derived, virus specific T-cell lines or<br />

clones represents a more sophisticated and safer<br />

approach for treatment of viral complications occurring in<br />

immunocompromized patients. Whereas too few clinical<br />

trials have been performed so far to draw any firm conclusion,<br />

based on animal studies dendritic cell-based<br />

immunotherapy holds promises of exerting an effective<br />

anti-tumor activity. Despite leukemic cells not being<br />

immunogenic, induction on their surface of co-stimulatory<br />

molecules or generation of leukemic dendritic cells may<br />

induce antileukemic cytotoxic T-cell responses. Tumor<br />

cells express a variety of antigens and can be genetically<br />

manipulated to become immunogenic. The main in vitro<br />

and in vivo functional characteristics of marrow mesenchymal<br />

stem cells (MSCs) with particular emphasis on<br />

their hematopoietic regulatory role are reviewed. In addition,<br />

prerequisites for clinical applications using cultureexpanded<br />

mesenchymal cells are discussed<br />

Perspectives. The opportuneness of using LAK cells or<br />

activated natural killer (NK) cells in hematologic patients<br />

with low tumor burden (e.g. after stem cell transplantation)<br />

should be further explored. Moreover the role of new<br />

cytokines in enhancing the antineoplastic activity of NK<br />

cells and the infusion of selected NK in alternative to CTL<br />

for graft versus leukemia (GVL) disease (avoiding graft<br />

versus host disease (GvHD) seems very promising. Separation<br />

of GVL from GvHD through generation and infusion<br />

of leukemia-specific T-cell clones or lines is one of the<br />

most intriguing and promising fields of investigations for<br />

the future. Likewise, strategies devised to improve<br />

immune-reconstitution and restore specific anti-infectious<br />

functions through either induction of unresponsiveness to<br />

recipient alloantigens or removal of alloreactive donor T-<br />

cells might increase the applicability and success of<br />

hematopoietic stem cell transplantation. Cellular<br />

immunotherapy with DC must be standardized and several<br />

critical points, discussed in the chapter, have to be<br />

properly addressed with specific clinical studies. Stimulation<br />

of leukemic cells via CD40 receptor and transduction<br />

of tumor cells with co-stimulatory molecules and/or<br />

cytokines may be useful to prevent a tumor escaping<br />

immune surveillance. Tumor cells can be genetically modified<br />

to interact directly with dendritic cells in vivo or<br />

recombinant antigen can be delivered to dendritic cells<br />

using attenuated bacterial vectors for oral vaccination.<br />

MSCs represent an attractive therapeutic tool capable of<br />

playing a role in a wide range of clinical applications in<br />

the context of both cell and gene therapy strategies.<br />

©1999, Ferrata Storti Foundation<br />

Key words: cell therapy<br />

haematologica vol. 85(suppl. to n. 12):December 2000


118<br />

C. Bordignon et al.<br />

The role of lymphoid cells in rejecting solid tumors<br />

transplanted into animal models was strongly suggested<br />

in the first decades of this century by J.B.<br />

Murphy (1926) 1 who, nonetheless, did not demonstrate<br />

it formally. Following his revolutionary findings on the<br />

immunologic mechanisms of allogeneic skin tolerance<br />

and rejection, in 1958 P.B. Medawar 2 coined the term<br />

“immunologically competent cell” to define a cell that is<br />

“fully qualified to undertake an immunological<br />

response”. Forty years after Medawar’s definition, the<br />

development of molecular and biological research has<br />

enormously improved our understanding of the complex<br />

regulatory mechanisms of proliferation, differentiation<br />

and function of the cells involved in the immune<br />

response. The concomitant evolution of biotechnology<br />

has also progressively given new opportunities to isolate<br />

and/or expand cell subsets, or to develop new molecules,<br />

in order to amplify or modify specific cell functions.<br />

Thus, the possibility of exploiting a specific cell<br />

function, in vivo or ex vivo, to obtain a therapeutic<br />

effect, such as an anti-tumor cytotoxic activity, or complete<br />

immune reconstitution, is part of the definition of<br />

cell therapy that is herein reviewed.<br />

In a general context, cell therapy can be considered as<br />

a strategy aimed at replacing, repairing, or enhancing the<br />

biological function of a damaged tissue or system by<br />

means of autologous or allogeneic cells. For instance, in<br />

the hematopoietic system cell therapy may include: a)<br />

removal or enrichment of various cell populations; b)<br />

expansion of hematopoietic cell subsets; c) expansion or<br />

activation of lymphocytes for immunotherapy; and d)<br />

genetic modification of lymphoid or hematopoietic cells,<br />

when these cells are intended to engraft permanently or<br />

transiently in the recipient and/or be used in the treatment<br />

of a disease.<br />

This review contains extensive considerations on the<br />

clinical use of lymphocytes and/or natural killer (NK) cells<br />

as a strategic weapon in preventing or curing the neoplastic<br />

relapse after chemotherapy and/or hematopoietic<br />

stem cell transplantation, the infusion of T-cell clones or<br />

lines able to restore a specific anti-viral activity, the in<br />

vivo and ex vivo potential use of dendritic cells to generate<br />

a tumor-specific cytotoxic activity, and the innovative<br />

use of donor stromal cells in conjunction with stem<br />

cell transplantation. Even tumor cells engineered to<br />

express cytokine or co-stimulatory molecules and representing<br />

the entire antigenic repertoire of a certain neoplasia<br />

can be used as a cancer vaccine. On the other hand,<br />

a broad definition of cell therapy at this time should<br />

include autologous and allogeneic transplants of purified<br />

hematopoietic stem cells, which, however, have been<br />

extensively reviewed in previously published reports. 3-5<br />

Tumor escape from immune surveillance<br />

Although several mechanisms allowing tumor cells to<br />

escape the host immune protection have been recently<br />

described, it is conceivable that others remain still<br />

undiscovered. However, tumor cells often fail to induce<br />

specific immune responses because of their inability to<br />

function as competent antigen presenting cells (APC).<br />

Professional APC, in fact, are fully capable of delivering<br />

two signals to T cells: 6 the first is antigen (Ag) specific<br />

and is mediated by the interaction of MHC molecules<br />

carrying antigenic peptides with the T-cell receptor<br />

(TCR), and the second signal, or co-stimulatory signal, is<br />

not Ag-specific and is principally mediated by members<br />

of the B7 family, namely B7-1 (CD80) and B7-2 (CD86),<br />

via their T-cell receptors CD28 and CTLA-4, and/or by<br />

CD40 via CD40L binding. 7-8<br />

The lack of a suitable tumor-associated antigen (TAA), 910<br />

or defective antigen processing, 11 or production of<br />

immunologic inhibitors, 12 or lack of co-stimulatory signaling<br />

by tumor cells, 13 as other mechanisms, can all contribute<br />

to prevent or abrogate an anti-tumor immune<br />

response. Moreover, neoplastic cells within the same<br />

tumor may show different reactivity with monoclonal<br />

antibodies (mAbs), cytotoxic T-lymphocyte (CTL) clones<br />

and lymphokine-activated killer (LAK) or tumor infiltrating<br />

lymphocyte (TIL) populations. Furthermore, despite<br />

many tumors having TAA and potentially being capable<br />

of stimulating T cells, in some cases they fail to induce<br />

an adequate CTL frequency in vitro. In other cases the<br />

antigen loss can be one of the mechanisms for escaping<br />

immune protection. 14 Private TAA often result from<br />

mutated gene products 15 and are potentially useful for<br />

developing tumor vaccines. These Ags, however, can be<br />

down-regulated or modified by point mutations, inducing<br />

a consistent reduction or the abrogation of peptidebinding<br />

by specific CTLs. Another critical issue for preventing<br />

immune responses is the absence, or the downregulation<br />

of MHC molecules on neoplastic cells, as<br />

shown in animal models, 16 or in human lung cancer. 17<br />

The pivotal role of B7 molecules in the immune<br />

response has been demonstrated in a variety of experimental<br />

models showing that after TCR signaling, binding<br />

of CD28 induces T-cell interleukin-2 (IL-2) secretion,<br />

proliferation and effector function, whereas presentation<br />

of the antigen in the absence of co-stimuli<br />

induces T-cell unresponsiveness either by anergy or<br />

clonal deletion. Therefore, since most neoplastic cells<br />

lack co-stimulatory molecules, it is likely that they can<br />

deliver the first signal through the MHC:TCR binding,<br />

but not the second one, thus driving host T-cells to tolerate<br />

the tumor. 18 Potential strategies to prevent or to<br />

reverse T-cell tolerance by CD28 or CD40L stimulation,<br />

or IL-2 receptor triggering, are under investigation.<br />

Further mechanisms impairing immunologic responses<br />

include the suppression of cytotoxic activity by the<br />

release of soluble factors or by direct cell-contact. In<br />

fact, tumor cells may secrete cytokines, such as MIP-1,<br />

or TGF-, or IL-10, that may be capable of inhibiting T<br />

cell activity. 12 Alternatively, tumor cells may induce T-<br />

cell apoptotic clonal deletion by increasing Fas:Fas-L<br />

ligation. 19 A schematic example of the main defects<br />

described in the tumor cell: T cell interaction is shown<br />

in Figure 1.<br />

Finally, since normal lymphocytes can bind to venular<br />

endothelial cells through adhesion receptors, such as<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

119<br />

L-selectin or / integrins, and then by rolling out they<br />

can reach tissues, lack of adhesion receptors on tumor<br />

vessels might prevent lymphocytic infiltration and contact<br />

with neoplastic cells. 20 In this case even the best<br />

strategies aimed at modifying the immunogenicity of<br />

tumor cells may not be successful at overcoming the<br />

lack of an antitumor immune surveillance.<br />

Figure 1. Main mechanisms for tumor escape of immune<br />

surveillance.<br />

Lymphokine-activated killer and tumorinfiltrating<br />

lymphocytes:<br />

past and present<br />

Natural killer cells and lymphokine-activated<br />

killer phenomenon<br />

Since 1970 NK cells have been recognized as a functionally<br />

distinct subset of cytotoxic effectors (Table 1).<br />

NK cells from rodent or from human peripheral blood kill<br />

a wide range of tumor cells and virus-transformed cells<br />

without the need for prior sensitization.<br />

In 1975 Heberman et al. 21 described a phenomenon of<br />

normal unstimulated lymphoid cells lysing cultured<br />

tumor-cell lines in a short in vitro assay. This cytolytic<br />

activity was subsequently shown to be neither MHC<br />

restricted nor mediated by the T-cell receptor complex.<br />

Such ability to eliminate tumor cells, but not normal<br />

tissues suggests that NK cells are not only involved in<br />

the control of cancer, but also that their presence and<br />

state of activation are important in the outcome of the<br />

disease and finally in the treatment of tumors. 22<br />

Mature NK have a clonally-distributed ability to recognize<br />

their target cell by class I MHC alleles. Karre et<br />

al. 23 demonstrated in a murine model that leukemia cell<br />

Table 1. Characteristics of cytotoxic effectors useful for adoptive immunotherapy of cancer.<br />

Effector type CTLs TILs NK cells LAK cells CIKs<br />

Source Peripheral Metastatic Peripheral blood NK cells Subset of<br />

blood lymphocytes lymph nodes and bone marrow and CTL activated T-lymphocytes<br />

by IL-2<br />

activated by cytokines<br />

Culture conditions:<br />

Tumor stimulation Yes None None None None<br />

need of IL-2 for response ++++ ++++ ++++ (CD56 dim ) - ++++,IFN-,<br />

+ (CD56 bright ) IL-12, anti-CD3 antibody<br />

Duration of culture 6 weeks 4 weeks 2-3 weeks 2-5 days 2-5 weeks<br />

target cells in vitro allogeneic cells autologous K-562 Raji, Daudi autologous and<br />

tumoral cells<br />

allogeneic tumoral cells<br />

In vitro cytolytic activity :<br />

MHC restricted to none: spontaneous none: lyse cytotoxic activity<br />

allogeneic cells lysis of virus-infected cells, a wide spectrum of superior to LAK;<br />

Specificity Restricted to autologous tumoral cells, tumor cells lyse whether<br />

MHC not restricted, autologous tumor allogeneic tumoral cells including cells CML autologous<br />

toward opsonized (MHC and/or that are or allogeneic blasts<br />

cells (ADCC) tumor associated) antibody-dependent resistant to NK; but do not lyse<br />

antigens) cell-mediated cytotoxicity<br />

(ADCC) specificity<br />

normal hematopoietic<br />

progenitors<br />

Effector phenotype -CD3+/4+ ,CD3+/8+ CD3+/8+/56+ CD3-/CD16+/CD56+ CD56+ CD3+/56+<br />

-CD3+/8+/16+.<br />

CD25+<br />

CTL: cytotoxic T-lymphocytes; TIL: tumor inflitrating lymphocytes; NK: natural killer; LAK: lymphokine-activated killer; CIK: citokine-activated killer.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


120<br />

C. Bordignon et al.<br />

lines lacking certain MHC class I molecules were killed<br />

by NK cells, while parental H-2 bearing line were not. In<br />

humans both NK and a subset of cytotoxic T-lymphocytes<br />

express receptors for MHC HLA class I molecules<br />

which exert an inhibitory effect on cell-mediated cytotoxicity.<br />

These surface molecules, belonging to the<br />

immunoglobulin superfamily, have been termed killercell<br />

inhibitory receptors (KIRs). Two distinct KIR families<br />

have been described: a) KIRs with IG-like domains, recognizing<br />

HLA-A, B and C alleles; and b) the CD94/NKG2A<br />

subtype, with a lectin domain, recognizing peptides<br />

related to the HLA-E class I system. 24 The interaction<br />

between KIRs and the corresponding MHC class I antigens<br />

prevent NK from killing target cells expressing self<br />

HLA alleles. 25 In addition some NK also express receptors<br />

that induce lysis of target cells expressing foreign HLA<br />

class I alleles. 26<br />

These findings explain the mechanism of self-tolerance<br />

in the NK population, which can be disrupted as a<br />

consequence of tumor transformation or viral infection<br />

or any other events inducing a loss or a substantial modification<br />

of class I molecules. These transformed cells<br />

can easily escape detection by T-lymphocytes by down<br />

regulating MHC antigens, but are normally destroyed<br />

by autologous NK cells. 27<br />

The NK cell compartment is heterogeneous and distinct<br />

NK subsets have been characterized. The most<br />

informative functional differences are based on relative<br />

CD56 fluorescence: only CD56 +bright , but not CD56 +dim NK,<br />

express the high-affinity IL-2 receptor, and respond to<br />

the low IL-2 concentration. They also expand 10 times<br />

more than CD56 +dim . 28<br />

NK progenitors differentiate into immature NK in<br />

presence of SCF, IL-7, IL-2 and bone marrow stromal<br />

cells producing IL-15. This last cytokine can directly<br />

induce CD34 + cells to differentiate into NK cells in the<br />

absence of IL-2. 29 The second step of NK maturation is<br />

stroma-independent and is characterized by the appearance<br />

of CD56 molecules: the intensity of CD56 expression<br />

reflects the proliferative potential and the killing<br />

ability of the NK. 30<br />

The effects of IL-2 on NK precursors appears to be<br />

stage-specific, confirming that, while mature NK precursors<br />

readily respond to IL-2, more immature progenitors<br />

need complete mixtures of cytokines and stromal<br />

cells. NK cells, after incubation with IL-2, become lymphokine-activated<br />

killer cells: LAK cells kill NK-resistant<br />

cell targets (e.g. Daudi cell line) and a wide spectrum of<br />

different fresh tumor cells in both autologous and allogeneic<br />

settings, while fresh normal tissues are resistant<br />

to LAK-mediated lysis. 31<br />

Although some tissue-resident lymphocytes may have<br />

spontaneous LAK activity, normal blood mononuclear<br />

cells (MNC) do not show any LAK activity, which can be<br />

acquired only after incubation with interleukin-2. 32<br />

These NK activated cells express new markers such as<br />

CD25, MHC class II antigens and fibronectin. 33 LAK activity<br />

can be generated not only in peripheral blood MNC,<br />

but also in the thymus, spleen, bone marrow and in MNC<br />

from lymph nodes. Many experimental data suggest that<br />

most LAK precursors are present in the null lymphocyte<br />

population.<br />

In humans LAK activity was much more evident in the<br />

MNC population after depletion of macrophages, T and<br />

B-cells. Residual MNC were CD16 + and did not show T-<br />

cell markers. 34<br />

LAK cells: experimental observations and<br />

clinical trials<br />

In animal models the combined administration of IL-<br />

2 and LAK has proved to be more efficacious than either<br />

component alone. In murine models the administration<br />

of high-dose IL-2 alone or in conjunction with LAK cells<br />

induced the regression of lung, liver and subdermal<br />

metastases. The antitumor effect correlated both with<br />

the IL-2 dose and the number of LAK cells administered;<br />

finally at different doses of IL-2, the concomitant<br />

administration of LAK cells resulted in increased reduction<br />

in established metastases. 35,36<br />

LAK cells are capable of inhibiting acute myeloid<br />

leukemia (AML) progenitor growth, and leukemia incidence<br />

is higher in people with deficiency of NK cells. 37<br />

In the large majority of patients at diagnosis or in relapse<br />

blasts appear resistant to lysis by autologous LAK cells.<br />

Moreover, about 90% of patients with acute leukemia in<br />

complete remission do not show spontaneous cytotoxicity<br />

against autologous blast cells, but ex vivo treatment<br />

with IL-2 restores cytolytic activity in 37.5% of these<br />

patients. 38 In a population of 42 patients with AML in<br />

complete remission, LAK cytotoxicity against autologous<br />

leukemic blasts was not significantly different from LAK<br />

of normal subjects. 39 However, multivariate analysis for<br />

prognostic factors showed that patients whose LAK had<br />

more lytic activity on leukemic blasts had significantly<br />

less risk of relapse than patients with poor LAK activity.<br />

In the first National Cancer Institute trial endstage<br />

cancer patients received high-dose bolus IL-2 therapy<br />

for 3 to 5 days. 35 Lymphocytes harvested during the systemic<br />

treatment with IL-2 were cultured in the presence<br />

of IL-2 for 2 to 4 days, in order to expand the LAK cell<br />

number; autologous LAK cells were then reinfused into<br />

patients in combination with the high-dose intravenous<br />

bolus IL-2 administration. Of 72 patients with renal cancer<br />

who were treated, 33% obtained an overall response,<br />

8 with complete response (CR) and 17 with partial<br />

response (PR); of 48 patients with metastatic melanoma<br />

21% responded with 4 CR and 6 PR; responses were<br />

also observed in patients with colorectal carcinoma and<br />

non-Hodgkin’s lymphoma. 40 The ILWG used the same<br />

strategy, obtaining an overall response rate of 19% in<br />

patients with melanoma and 16% in those with renal<br />

carcinoma. 41 After these initial trials the original schema<br />

of the National Cancer Institute was modified with the<br />

use of IL-2 in continuous infusion rather than bolus<br />

injection in order to reduce the systemic toxicity. 42<br />

The first randomized study, comparing IL-2 alone to<br />

IL-2 plus LAK cells, was published by McCabe. 43 This trial<br />

included patients with either renal carcinoma or<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

121<br />

melanoma; no significant difference in response rate<br />

between the two groups was reported. A second randomized<br />

study at the National Cancer Institute followed<br />

these pioneering experiences, comparing IL-2 alone to<br />

IL-2/LAK cells: 44 181 patients were enrolled in this study<br />

(90 in the IL-2 plus LAK arm and 91 in the IL-2 alone).<br />

A total of 10 CR were observed in the IL-2/LAK arm as<br />

compared to only 3 in the IL-2 alone arm. The overall<br />

response rates were similar, but there was a survival<br />

trend (p=0.07) in favor of the IL-2/LAK arm: the actuarial<br />

survival for patients receiving IL-2/LAK was 31%<br />

compared to 17% for those receiving IL-2 alone. Toxicity<br />

was virtually equivalent in both arms and the majority<br />

of toxic effects were due to IL-2 administration,<br />

while the only complication associated with LAK therapy<br />

was transient hepatitis A, due to contamination of<br />

the culture medium.<br />

A third randomized trial, comparing IL-2 alone versus<br />

IL-2/LAK therapy was published in 1995. 45 In this study<br />

only patients with advanced renal carcinoma were<br />

treated and IL-2 was administered as a continuous infusion<br />

rather than bolus injection. Seventy-one patients<br />

entered (36 vs. 35) this trial and only 6% overall<br />

obtained a major response, with a median survival of 13<br />

months; the difference between the two groups was not<br />

significant. Therefore it may be concluded that LAK cells<br />

did not improve the activity of IL-2 in patients with<br />

advanced renal carcinoma.<br />

The last randomized trial published was conducted in<br />

174 primary lung carcinoma patients after surgery, comparing<br />

the adjuvant treatment with IL-2 plus LAK (for<br />

two years) with conventional treatment. 46 The 5- and 9-<br />

year survival rates were significantly superior in patients<br />

receiving IL-2/LAK therapy, but no comparison was<br />

planned between Il-2 alone and IL-2/LAK therapy. The<br />

impressive results obtained in terms of overall survival<br />

also in non-curative cases after surgery (OS: 52% at 5<br />

years) should probably be interpreted as due to fact that<br />

in this study patients received the immunotherapy after<br />

consistent tumor debulking.<br />

Other clinical trials (non-randomized) were conducted<br />

with IL-2 with or without LAK cells, and the overall<br />

response rate was similar for both the immunotherapy<br />

modalities. 47,48 The detailed review of other (non-ran-<br />

Table 2. Clinical trials with LAK cells.<br />

Treatment schedule<br />

Author Year Patients Kind of tumor IL-2 (dose and schedule) LAK cells Response<br />

Rosenberg 1987 157 Melanoma Randomize: IL-2 vs. IL-2+LAK CR: 2.2% vs 7.5%<br />

PR: 10.9% vs 14.2%<br />

mR: 2.2% vs 9.4%<br />

West 1987 40 Miscellaneous 1-7x10 6 U/m 2 /day CI CR+PR: 22-28%<br />

Yoshida 1988 23 Brain tumor Direct injection of LAK into recurrent tumor Regression: 26%<br />

cavity + IL-2 (50-400 U); multiple treat<br />

Fisher 1988 29v Renal carcinoma 12.9 MIU/kg (median 10 doses) 7x10 10 cells OR: 16%<br />

West 1989 30 Renal carcinoma 3x10 6 U/m 2 /day CI NR 22-28%<br />

Dutcher 1989 32 100,000 U/kg q8h 8.9x10 10 CR+PR: 19%<br />

Paciucci 1989 24 Miscellaneous 1-5x10 6 U/m 2 /day CI 5.6x10 9 CR+PR: 20.8%<br />

Neqrier 1989 51 Renal carcinoma 3x10 6 U/m 2 /day CI 1.2x10 10 CR+PR: 27%<br />

Stahel 1989 23 Miscellaneous 3x10 4 U/kg q8h 5.1x10 10 CR+PR: 17%<br />

Rosenberg 1993 181 Metastatic cancer Randomized: IL-2 vs IL-2+LAK CR: 5% vs 11.76%<br />

PR: 15.2% vs 16.5%<br />

OS (3 yrs): 17% vs 31%<br />

(p2=0.089)<br />

Bajorin 49 Renal carcinoma Randomized: IL-2 vs IL-2+LAK (73x10 9 ) No difference<br />

(3 MU/m 2 )<br />

Keilholz 1994 9 Liver metastic carcinoma IL-2 CI into the splenic artery LAK transfer into the CR+PR: 33%<br />

or intravenous infusion<br />

portal vein or the<br />

hepatic artery<br />

Murray Law 1995 66 Renal carcinoma Randomized: IL-2 vs IL-2+LAK (NR) CR+PR: 9% vs 3%<br />

(3x10 6 U/m 2 /day) (p=0.61)<br />

Kimura 1997 82,788 Resected lung carcinoma Randomized: IL-2+LAK vs. Standard therapy OS (5 yrs): 54.4% vs 52%<br />

(7x10 5 U/day x 3 days) (1-5x10 9 cell) OS (9 yrs): 33.4% vs 24.2%<br />

haematologica vol. 85(suppl. to n. 12):December 2000


122<br />

C. Bordignon et al.<br />

domized) experiences using these two different<br />

immunotherapies suggests that LAK cell reinfusion<br />

slightly increased the number of CR and the duration of<br />

response, especially in patients with metastatic<br />

melanoma (Table 2). 49,50<br />

In hematologic malignancies the first attempts to generate<br />

and expand LAK activity by using IL-2 in vivo were<br />

clinically disappointing especially in patients autotransplanted<br />

for ALL; after transplantation patients were randomly<br />

assigned to treatment with systemic IL-2 (without<br />

LAK cell administration) or no treatment, but the<br />

disease-free survival was similar in the two arms. 51 The<br />

use of LAK cells has also been proposed after autologous<br />

transplantation for hematological malignancies, but the<br />

very small series of patients reported does not allow any<br />

definitive conclusion to be drawn about its clinical benefit.<br />

52 Beaujean et al. 53 reinfused, after myeloablative<br />

therapy, BM incubated with IL-2 into 5 ALL patients,<br />

observing a very marked delay of the engraftment and<br />

the recurrence of disease in all patients. Recently there<br />

has been a report of 61 women with breast cancer autotransplanted<br />

with IL-2 activated PBPC and treated with<br />

low dose IL-2 starting from PBPC reinfusion, without<br />

graft failures or major toxicity; there are no data concerning<br />

the outcome of patients and this experience only<br />

confirms the feasibility of the approach. 54<br />

In a very preliminary experience a sustained major<br />

cytogenetic response to immunotherapy with GM-<br />

CSF+IL-2 and LAK infusion was observed in chronic<br />

myeloid leukemia (CML) patients after autologous transplantation.<br />

55 However, a renewed interest in this<br />

approach has led to new research pursuing different<br />

directions:<br />

a. selection of patients with low tumor-burden and<br />

with significant in vitro LAK activity against autologous<br />

tumor cells, in order to reach an optimal effector/target<br />

ratio;<br />

b. harvest of large amounts of NK cells (for additional<br />

ex vivo expansion/activation with IL-2) to be reinfused<br />

in the early phase after BMT; 56<br />

c. direct activation of leukapheresis, after priming with<br />

chemotherapy followed by cytokines, in order to<br />

reinfuse, after HDT, a product richer in cytotoxic<br />

effectors and probably less contaminated; 57<br />

d. identification and selection of more efficient NK<br />

progenitors (e.g. adherent NK) by eliminating undesired<br />

accessory cells which could inhibit their killing<br />

and proliferative ability;<br />

58, 59<br />

e. generation and expansion of other CE subsets with<br />

more powerful activity against autologous tumor<br />

cells, e.g. cytokine-induced killer cells (CIK); 60<br />

f. use of other cytokines in association with IL-2, in<br />

order to potentiate the activity and/or improve the<br />

selectivity of activated peripheral blood MNC.<br />

Tumor infiltrating lymphocytes<br />

The disappointing results of adoptive immunotherapy<br />

with blood-derived LAK cells led to a search for more<br />

specific CE cells. Tumor infiltrating lymphocytes (TIL) are<br />

T-lymphocytes with unique tumor activity that infiltrate<br />

some tumors and can be expanded by long-term culture<br />

with IL-2 at low-intermediate concentrations. 61 In<br />

murine models TIL have exhibited a stronger anti-tumor<br />

effect than LAK cells on a per-cell basis; in humans TIL<br />

have been isolated with variable frequency from different<br />

solid tumors and very often (about 30% of cases)<br />

from patients with melanoma. Phenotypic analysis<br />

showed that TIL consisted mainly of CD4 + cells in colon,<br />

breast and urothelial tumors, while in melanoma CD8 +<br />

cells are prevalent. 62,63 CD3 – CD16 + NK cells have also<br />

been isolated from several tumors, confirming the large<br />

heterogeneity of tumor infiltrates. 64 The mechanism of<br />

the antitumor action of TIL is unknown; there is some<br />

evidence that these cells secrete cytotoxins and<br />

cytokines which are capable of killing tumor cells and<br />

recruiting other CE.<br />

Experimental models and clinical trials<br />

Mice carrying spontaneous metastases, treated with<br />

IL-2 plus tumor-derived T-cells, obtained from splenocytes<br />

after mixed lymphocyte-tumor cultures, had a better<br />

survival than those treated with LAK cells; previous<br />

tumor debulking (with chemotherapy and/or radiotherapy)<br />

was needed to maximize the efficacy of TIL-therapy.<br />

65<br />

Unfortunately large amounts of TIL can be collected<br />

very rarely, and the large scale expansion of this population<br />

is crucial in order to obtain relevant clinical<br />

responses; this step of ex vivo manipulation is not<br />

always successful, because the need for prolonged culture<br />

of TIL (from 6 to 8 weeks with IL-2) may abrogate<br />

the selectivity against the tumor; moreover only a small<br />

fraction of the readministered human TIL is able to concentrate<br />

in the tumor sites. 66<br />

Wong et al. 67 showed in a mouse model that TIL preferentially<br />

localize in the liver and lungs. In contrast trafficking<br />

studies employing TIL radiolabeled with In 111 ,<br />

have shown that TIL do traffic to tumor sites; 68 this homing<br />

property should produce high concentrations of TIL,<br />

and probably their permanence, in the area of a tumor.<br />

Human TIL transfected in vitro with the neomycinresistance<br />

gene and reinfused intravenously, have been<br />

detected by polymerase chain reaction (PCR) techniques<br />

from 6 to 60 days in patients affected by metastatic<br />

melanoma. 69 Aebersold et al. 70 observed a strong correlation<br />

between the tested tumor cytotoxicity in vitro and<br />

the in vivo response, in a small cohort of patients with<br />

metastatic melanoma. A similar relationship was<br />

observed in a murine model in which the in vivo therapeutic<br />

effect of TIL correlated with secretion of IFN and<br />

tumor necrosis factor (TFN). 71<br />

In order to increase their specificity and potency, TIL<br />

have been engineered with genes encoding cytokines or<br />

cytotoxins such as TNF or IFN- or IL-2. 69,72 However,<br />

some experimental observations suggest that these high<br />

concentrations of cytokines can cause systemic toxicity<br />

and in some cases could even make the tumor more<br />

aggressive. 73,74<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

123<br />

Table 3. Clinical trials with LAK cells and IL-2.<br />

Treatment schedule<br />

Author Year Patients Kind of tumor IL-2(dose and schedule) TIL cells Response<br />

Rosenberg 1988 20 metastatic melanoma 110 5 U/kg every 8h; CPM 25 mg/kg 20.510 10 cell Regress: 60%<br />

Kradin 1989 38 miscellaneous 1-310 6 U/m 2 CIx 24h OR: 26%<br />

Rosenberg 1990 5 metastatic melanoma TIL gene modified<br />

Aoki 1991 10 advanced or recurrent OR: 70%<br />

ovarian cancer TIL after single CI CPM Long term: 57%<br />

Dillman 1991 21v metastatic melanoma 1810 6 IU/m 2 /day CI 10 11 cell OR: 24%<br />

expensive, difficult<br />

Arienti 1993 12v metastatic melanoma 13010 6 IU/m 2 /day CI 6.810 9 cell RR:33%<br />

Belldegrun 1993 10v metastatic renal 210 6 IU/m 2 /day in 96h (IL-2) TIL CR: 30%<br />

cell carcinoma<br />

610 6 IU/m 2 /day (IFN-)<br />

Schwartz– 1994 41 melanoma IL-2 TIL CR+PR: 21.9%<br />

entruber<br />

Pockaj 1994 38 metastatic melanoma 7.210 5 IU/kg every 8h 1.3-2.210 11 cell OR: 38.5%<br />

and CPM 25 mg/kg<br />

Chang 1997 20v advanced melanoma and IL-2 anti-CD3 vaccine primed OR:33.3%<br />

renal cell cancer lymph node cells PR:9.1%<br />

activated<br />

Curti 1998 solid tumor and NHL 9x10 6 UI/m 2 /day x 7 days CI T CD4+ cell+ anti CD3 some tumor regression<br />

Ridolfi 1998 32 miscellaneous 12-6 MIU/ day 5.8x10 10 TIL no response in patients<br />

(West's schedule)<br />

with advanced cancer<br />

ev: evaluable; PR: partial response; OR: overall response.<br />

In addition to their potential therapeutic use as<br />

cytolytic effectors, the ability of some TIL to recognize<br />

unique antigens on tumor cells has made the study of the<br />

biologic characteristics of these antigens more feasible.<br />

Melanomas from different patients who share MHC antigens<br />

are often cross-recognized by allogeneic TIL, as<br />

could be expected for an MHC-restricted T-cell response;<br />

the presence of shared antigens in different patients with<br />

melanoma suggests the possibility of using these antigens<br />

in an active immunization program for this disease. 75<br />

When adoptively transferred into patients, TIL showed<br />

significant therapeutic efficacy in patients with advanced<br />

melanoma, but not in renal carcinoma patients. In a<br />

phase II trial patients with malignant melanoma were<br />

treated with IL-2 and TIL following chemotherapy: 76 39%<br />

of them achieved some sort of response, including those<br />

who had previously experienced a failure of IL-2 therapy.<br />

Kradin et al. 77 treated some patients with a combination<br />

of chemotherapy, IL-2 and TIL, obtaining 23% of<br />

responses in those affected by melanoma and 29% in<br />

those with renal carcinoma, but none in patients with<br />

non-small cell lung carcinoma.<br />

A summary of most clinically relevant clinical trials<br />

with TIL is given in Table 3.<br />

The lack of important clinical trials with TIL is probably<br />

due to the difficulties in finding TIL at diagnosis and<br />

especially because the techniques for TIL priming and<br />

expansion are time-consuming and not completely<br />

standardized. TIL therapy is still young, but its very interesting<br />

potential has not yet been thoroughly investigated.<br />

New approaches with LAK or TIL cells<br />

Allogeneic setting. Whereas it is widely accepted that<br />

graft-versus-host disease (GvHD) is initiated by donor T<br />

cells recognizing foreign host antigens, other factors<br />

including toxicity of conditioning regimens and cytokine<br />

dysregulation are involved in the pathogenesis of<br />

GvHD. 78,79 Data from murine experiments show that NK<br />

cells play an active role both in GvHD and in garft-versus-leukemia<br />

(GVL) events: in a recently published model<br />

100% of SCID mice bearing human leukemic cells,<br />

and transplanted with NK + T-cells, died of acute GvHD;<br />

but while animals which received only T-cells developed<br />

clinical GVL associated with relevant chronic GvHD, NKtransplanted<br />

animals showed the same degree of protection<br />

from leukemia, experiencing only mild-moderate<br />

acute GvHD without chronic GvHD. 80 These data<br />

suggest that in order to optimize the GVL effect while<br />

minimizing the severity of acute GvHD, donor grafts<br />

haematologica vol. 85(suppl. to n. 12):December 2000


124<br />

C. Bordignon et al.<br />

should be manipulated by adding a moderate dose of T-<br />

cells in the early phase and using purified NK cells in the<br />

late phase after transplantation.<br />

Preliminary data suggest that in normal donors, after<br />

G-CSF mobilization, NK progenitors have decreased<br />

killing capacity and diminished proliferative ability in<br />

response to IL-2, compared to the unprimed bone marrow<br />

counterpart. 81 In contrast, after an HLA incompatible<br />

transplant, a progressive expansion of NK and CTL<br />

with NK like function (CD3 + /CD56 + ) has been observed;<br />

recipients received the T-cell depleted graft without<br />

developing GvHD, but in most cases a significant GVL<br />

effect could be demonstrated both in vitro and in vivo;<br />

these data support the critical role of CTL KIR + in this<br />

particular subset of transplanted patients. 82<br />

Concerning the expanding role of cord blood transplantation,<br />

even though the content of NK in this source<br />

seems normal, the decreased IL-12 production by cord<br />

blood MNC, reducing IFN- stimulation, may contribute<br />

to reduce NK and LAK cytotoxicity; these data suggest<br />

one possible explanation for cord blood immaturity and<br />

their clinical implications such as decreased GvHD and<br />

GVL, which could be enhanced by IL-12 administration. 83<br />

Autologous setting. Considering the impressive results<br />

observed in the allogeneic setting using donor-buffy coat<br />

lymphocytes for treatment of relapse, CML seems an<br />

attractive field for testing the efficacy of adoptive<br />

immunotherapy in the autologous setting too; some<br />

experimental data support this hypothesis. The MNC of<br />

patients with CML contain a population of benign NK<br />

cells which can be expanded and activated by IL-2, generating<br />

a CE population capable of killing both NK-sensitive<br />

and NK-resistant tumor targets. 84 Both number<br />

and functional activity of activated NK (ANK) in CML<br />

patients decrease with the progression of the disease. 85<br />

In vitro data show that autologous ANK inhibit both<br />

committed and very early Philadelphia positive progenitors<br />

in a MHC-unrestricted manner. 86 In these experiments<br />

CML progenitor cell killing by autologous and allogeneic<br />

ANK (after T-cell depletion) was comparable.<br />

Finally the CML blast killing was not dependent of soluble<br />

factors because it was abrogated by a transwell<br />

membrane, but was mediated by cell-to-cell contact<br />

being significantly blocked by anti-integrin antibodies. 87<br />

In 1986 Lanier and Phillips described a subset of CD3 +<br />

T cells co-expressing the CD56 antigen which is a typical<br />

NK marker (CIK). 88 More recently Schmidt-Wolf et al. 89<br />

obtained large expansion of this subset in a 16-day liquid<br />

culture containing IFN-, IL-1, IL-2 and a monoclonal<br />

antibody against CD3 as the mitogenic stimulus. The<br />

same group tested the ability of this population to purge<br />

bone marrow in patients with CML; they found that while<br />

standard LAK cells were in most cases unable to lyse CML<br />

cells, CIK cells were able to lyse both autologous and allogeneic<br />

CML blasts, without affecting normal hemopoietic<br />

progenitors. 90 Recently it has been reported that CIK<br />

administration in SCID mice bearing human CML induced<br />

the disappearance of Ph’+ cells in the spleen of 12/14<br />

animals. 91<br />

Another interesting potential application of autologous<br />

LAK is the treatment of EBV-related lymphomas<br />

arising in organ-transplanted patients; a preliminary<br />

description of four complete responses after treatment<br />

with autologous peripheral MNC incubated with IL-2<br />

seems very promising. 92 Recently in thyroid cancer<br />

patients Katsumoto et al. 93 generated cytotoxic CD4 +<br />

lymphocytes from TIL after non-specific in vivo stimulation<br />

with OK-432 (which induces severe local inflammation<br />

in the draining lymph nodes) and low-dose IL-2,<br />

obtaining large amounts of cytotoxic CD4 + (Th1) cells,<br />

producing high levels of IFN- and TNF- in the supernatants.<br />

These CE lysed a wide spectrum of tumor cell<br />

lines; anti-TCR antibodies did not inhibit their killing<br />

activity, which was in favor of a non-MHC restricted<br />

lysis, while antibodies anti-ICAM-1 completely inhibited<br />

the activity.<br />

Tsurushima et al. 94 induced autologous CTLs directly<br />

from peripheral blood MNC by preparing a co-culture of<br />

minced tissue fragments of glioblastoma multiforme<br />

with a mixture of cytokines (IL-1, 2, 4, 6 and IFN-) for<br />

2 weeks. At the end of culture the population contained<br />

mainly CD4 + and CD8 + lymphocytes able to kill 82 to<br />

100% of the glioblastoma cells while normal LAK cells<br />

killed only 33%.<br />

Finally, in follicular lymphomas freshly isolated TIL,<br />

normally lacking tumor-specific cytotoxicity, were stimulated<br />

with lymphoma cells, in the presence of IL-2 and<br />

CD40 ligand; these T-TIL were capable of proliferating<br />

in response to follicular lymphoma cells; moreover TIL<br />

could be further expanded in the presence of IL-4, IL-7<br />

and IFN-. 95<br />

The potential role of new cytokines<br />

Several cytokines affect CTL and NK response: first of<br />

all IL-2 which expands the precursor pool of alloreactive<br />

CTL; IL-15 (producted by monocytes) mimics IL-2<br />

action by inducing IFN- production, T-cell memory<br />

activation and CTL proliferation. 96 IL-12 shares certain<br />

functional properties with IL-2, but using a different,<br />

IL-2 independent pathway. 97 In addition IL-12 enhances<br />

the lytic activity of human peripheral blood MNC against<br />

a wide spectrum of tumors. 98,99 Recently it has been<br />

observed that the combination of IL-2 and IL-12 is capable<br />

of inducing lysis of blasts resistant to IL-2-activated<br />

effectors, even in the autologous setting. 100<br />

Therefore the association of IL-2 plus IL-12 could potentially<br />

become an important tool to increase the antitumor<br />

efficacy both ex vivo, by generating large amounts of<br />

CIK, 101 and in vivo, by systemic administration.<br />

GM-CSF is a cytokine capable of inducing a pleiotropic<br />

immunostimulatory effect and also increases the immunogenicity<br />

of tumors; in a model for ex vivo expansion of LAK<br />

cells from leukaphereses in order to obtain contemporaneously<br />

a decontaminated harvest and a large amount of<br />

CE to reinfuse after myeloablative therapy, the association<br />

GM-CSF+IL-2 obtained a 5-fold expansion of the NK compartment<br />

while sparing the clonogenic potential of hemopoietic<br />

progenitors. 102<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

125<br />

Biodistribution and targeting of LAK and TIL<br />

At present adoptive immunotherapy with LAK or IL-2<br />

activated TIL has had limited success in patients with<br />

advanced cancer. Although a well-defined mechanism<br />

remains to be established, numerous in vitro findings and<br />

in vivo data suggest that the cancer-specific cytotoxicity<br />

of CE is obtained in multiple steps; a prerequisite,<br />

however, is optimal delivery of CE to the target tissues<br />

while minimizing systemic cytotoxicity. Two major areas<br />

currently requiring investigation are the survival and<br />

localization of adoptively transferred CE in the tumorbearing<br />

host, and the detailed mechanism of tumor<br />

regression. The major goals in this area concern the optimal<br />

administration of systemic cytokines together with<br />

CE, and (finally) the ways to enhance localization and<br />

transcapillary migration of the infused cells.<br />

Experimental evidence together with theoretical considerations<br />

based on CE functions indicate that the ability<br />

of adoptive immunotherapy to eradicate an established<br />

tumor is quantitatively determined by the initial<br />

tumor burden, growth pattern, and the magnitude of<br />

immunologic response generated by CE and other accessory<br />

cells at the site of the tumor. 103,104 Thus, to achieve<br />

tumor eradication and minimize systemic toxicity, the<br />

explanation of the mechanisms underlying lymphocyte<br />

biodistribution and the factors governing effector cell<br />

uptake in tumor sites is critical, but unfortunately data<br />

about CE biodistribution in humans are scarce.<br />

Although a physiologically based kinetic modeling<br />

approach has been applied to the pharmacokinetics of<br />

drugs and antibodies, there has been no effort to extend<br />

this approach to cell biodistribution, probably because<br />

of its complexity.<br />

One interesting attempt to apply this method to adoptive<br />

immunotherapy has, however, recently been published.<br />

105 The importance of lymphocyte infiltration from<br />

surrounding normal tissues into tumor tissue was found<br />

to depend on lymphocyte migration rate, tumor size,<br />

and host organ.<br />

It is likely that therapy with CE has not been as effective<br />

as originally promised, in part because of the very<br />

low CE concentration in the systemic circulation; this<br />

was mainly due to lung entrapment. Reducing this phenomenon<br />

by decreasing the attachment rate or adhesion<br />

site density in the lung by 50%, the tumor uptake could<br />

be increased by 40% for TIL to 60% for adherent NK<br />

cells.<br />

Theoretical models indicate that intra-arterial administration<br />

has a dramatic advantage over intravenous<br />

delivery, with more than a 1,000-fold higher CE accumulation<br />

in the tumor site. Indeed experiments in murine<br />

models show that it is possible to eliminate liver metastasis<br />

by loco-regional administration of human IL-2 ANK<br />

or by systemic adoptive transfer. 106<br />

Finally the differences in biodistribution between different<br />

lymphocyte populations, mainly due to the different<br />

attachment rates in the tumor and the lung,<br />

should be carefully considered. ANK cells are more easily<br />

trapped than CTL in lung vessels due to their larger<br />

diameter and greater rigidity. 107 A greater accumulation<br />

of TIL was expected in the spleen as a result of their<br />

stronger adhesion at this site through the lymphocyte<br />

homing receptor. 108 Although this model has limitations<br />

related to the sensitivity of analysis of parameters such<br />

as adhesion-site density, lymphocyte attachment and<br />

arrest rate, it could be considered a useful basis for<br />

designing new experimental models to increase the concentration<br />

and recirculation of CE in tumor sites, reducing<br />

effector cell rigidity or blocking adhesion molecules.<br />

The so-called antibody-dependent cellular cytotoxicity<br />

(ADCC) could be mediated by cells expressing Fc<br />

receptor II and Fc receptor III (e.g. NK cells and<br />

CD3 + /CD16 + cells). This kind of cytotoxicity, even though<br />

exhibited by non MHC-restricted cells, cannot be considered<br />

aspecific and is also exhibited by monocytes.<br />

LAK cells are extremely potent mediators of ADCC 109<br />

and thus the use of LAK plus IL-2 in combination with<br />

monoclonal antibodies will probably become a powerful<br />

tool for treating some immunogenic tumors. This<br />

approach has been tested in patients with colorectal<br />

cancer, 110 but could be also proposed for treating some<br />

immunogeneic hematologic malignancies such as follicular<br />

lymphoma or multiple myeloma.<br />

Donor lymphocyte infusion for treatment<br />

of leukemia relapse and as a means for<br />

accelerating immunologic reconstitution<br />

in patients given transplantation of<br />

hematopoietic progenitors<br />

Manipulation of the immune system after hematopoietic<br />

stem cell transplantation (HSCT) to reverse<br />

leukemia relapse or to reduce its incidence remains one<br />

of the most fascinating, even though difficult, challenges<br />

for successful cure of patients with hematologic<br />

malignancies. In fact, over the last 10-15 years, evidence<br />

has emerged from clinical transplantations to<br />

suggest that the anti-leukemia effect of allogeneic HSCT<br />

cannot merely be ascribed to the myeloablative therapy<br />

employed during the preparative regimen, donor lymphocytes<br />

playing a pivotal role in the eradication of<br />

malignant cells. Adoptive immunotherapy with donor<br />

lymphocyte infusion (DLI) in patients relapsing after<br />

HSCT has provided one of the most effective demonstrations<br />

of the importance of the graft-versus-leukemia<br />

effect in the cure of patients with hematologic malignancies.<br />

111,112<br />

Even though DLI may sometimes be burdened by complications<br />

that endanger the patient's life, mainly myelosuppression<br />

and GvHD, in individuals with CML experiencing<br />

relapse in chronic phase after an allograft approximately<br />

70% complete remissions can be obtained with<br />

this treatment. 112-116 Most of these remissions are sustained<br />

over time, this proving the capacity of DLI to eradicate<br />

clonogenic leukemia cells or control their regrowth.<br />

DLI has also been extensively employed to<br />

reverse relapse in patients with acute leukemia, non-<br />

Hodgkin’s lymphoma and multiple myeloma. However,<br />

haematologica vol. 85(suppl. to n. 12):December 2000


126<br />

C. Bordignon et al.<br />

the response rate of patients with other hematologic<br />

malignancies, especially acute leukemia, is significantly<br />

lower. 115, 116 In fact, only 20-30% of patients with AML<br />

achieve a hematologic remission after DLI and the value<br />

for patients with ALL is even lower. Patients with<br />

acute leukemia experiencing recurrence following an<br />

allograft have a higher probability of response with DLI<br />

if treated after having achieved a state of complete<br />

remission with chemotherapy, that is in a condition characterized<br />

by a limited tumor burden. 117<br />

The most important factor predicting response to DLI<br />

in patients with CML is the type of relapse. In fact, as<br />

already mentioned, patients suffering from cytogenetic<br />

relapse or hematologic relapse in chronic phase have a<br />

high probability of response to DLI, while patients with<br />

more advanced disease (accelerated phase or blast crisis)<br />

respond less frequently (20-25% of cases). 112-116<br />

Relapse occurring in the first 1-2 years after allograft, 115<br />

little or no acute and chronic GvHD after transplantation<br />

or removal of T-lymphocytes before HSCT 116 are also<br />

associated with a higher probability of benefitting from<br />

DLI. In patients with CML responding to DLI, the median<br />

time to obtain hematologic remission has been reported<br />

to be about 6-8 weeks, 115 whereas a longer time (in<br />

the order of 11 months) is needed for molecular remission,<br />

this documenting that clearance of leukemia cells<br />

is a dynamic, progressive phenomenon. 118 The number of<br />

T-cells to be infused and the best schedule of DLI for<br />

optimal response without concurrent development of<br />

severe GvHD are still to be conclusively established since<br />

they depend on several variables, such as degree of HLAcompatibility<br />

between donor and recipient, original disorder,<br />

and type of relapse. 112 Some authors have claimed<br />

that infusion of no more than 110 7 donor-derived T-<br />

cells per kg of recipient body weight or CD8-depleted<br />

lymphocytes can induce a state of remission and substantially<br />

prevent GvHD occurrence. 119 However, recently,<br />

the Hammersmith Hospital group reported that the<br />

response in CML patients relapsing after HSCT and given<br />

graded increments of donor lymphocytes seems to be<br />

less sustained over time than that observed after infusion<br />

of a larger number (i.e. >110 8 /kg of recipient body<br />

weight) of T-cells (Dazzi F, personal communication,<br />

1999). Support to the importance of the number of cells<br />

infused is also given by the results of Lokhorst et al., 120<br />

who observed that, in multiple myeloma, patients given<br />

more than 110 8 T-cells/kg had the highest probability<br />

of benefitting from DLI. In some of these patients, the<br />

response was complete with disappearance of myeloma<br />

proteins.<br />

The two major complications occurring after DLI are<br />

myelosuppression and GvHD. Myelosuppression is experienced<br />

by approximately 50% of the patients treated<br />

with DLI for CML in hematologic relapse, while it occurs<br />

much less frequently in patients with cytogenetic recurrence,<br />

112 this indicating that such a complication is<br />

observed in situations characterized by a predominance<br />

of host-type hematopoiesis. Therefore, myelosuppression<br />

can be explained by a direct effect of the transfused<br />

donor lymphocytes on hematopoietic cells of the recipient,<br />

similarly to that observed in transfusion-associated<br />

GvHD. The majority of patients experiencing myelosuppression<br />

after DLI recover a normal blood cell count<br />

spontaneously: nevertheless, myelosuppression may be<br />

fatal in approximately 10% of patients, with death being<br />

caused by infection or bleeding. 115,116 Infusion of a huge<br />

number of donor-derived peripheral blood hematopoietic<br />

progenitors, mobilized through hematopoietic<br />

growth factors such as granulocyte colony-stimulating<br />

factor (G-CSF), can alleviate the problem of pancytopenia<br />

in some selected cases, hastening the recovery of<br />

neutrophil and platelet counts.<br />

Grade II-IV acute GvHD develops in almost half of<br />

patients given DLI, 115,116 the highest incidence being<br />

observed when the donor is an unrelated volunteer. 121<br />

Incidence and severity of GvHD after DLI does not appear<br />

to correlate with GvHD after the original transplant and<br />

it may occur with a high incidence since donor lymphocyte<br />

therapy involves the infusion of large numbers of T-<br />

cells, whose immunocompetence is not usually modulated<br />

by cyclosporin A and/or methotrexate. Even though<br />

GvHD occurring after DLI is well-correlated with disease<br />

response as proved by the observation that most patients<br />

obtaining a hematologic remission after this treatment<br />

developed acute and/or chronic GvHD, GvHD may not be<br />

sufficient to induce GVL. Moreover, some patients not<br />

experiencing GvHD after DLI achieve hematologic remission,<br />

this indicating the existence of a GVL effect separate<br />

from development of GvHD. 113,116,117,122<br />

GVL effect occurring after HSCT and DLI is considered<br />

to be mediated by HLA-unrestricted NK or LAK cells or by<br />

T-lymphocytes that recognize leukemia cells in an HLArestricted<br />

fashion. 123,124 In particular, when patient and<br />

donor are HLA-identical, it is believed that recipient non-<br />

MHC-encoded minor histocompatibility antigens (mHAg)<br />

are recognized by donor CTL. While widely distributed<br />

mHAg account for the GVL effect associated to GvHD, tissue<br />

restricted or leukemia-specific antigens can elicit a<br />

specific GVL reaction 108-113 and it has been demonstrated<br />

that both CD4 + and CD8 + CTL recognizing mHAg in a classical<br />

MHC-restricted fashion can be generated in vitro.<br />

124,125 In particular, mHAg-specific CD8 + CTL can display<br />

strong lysis of mature leukemia cells, as well as suppress,<br />

together with CD4 + mHAg-specific CTL, the growth<br />

of clonogenic leukemia precursor cells. 126,127 Production<br />

of cytokines (such as -interferon and tumour necrosis<br />

factor ) able to induce the apoptotic death of leukemia<br />

cells can also contribute to the GVL effect. 128,129 This said,<br />

it is not surprising that several efforts have been directed<br />

towards the identification of strategies capable of<br />

selecting and/or amplifying specific GVL response, not<br />

associated with development of GvHD. Since it has been<br />

documented in humans that CTL directed against allogeneic<br />

leukemic blasts can be detected in the peripheral<br />

blood of healthy donors 130 and that CTL specifically reactive<br />

towards recipient leukemic blasts can emerge and<br />

persist over time in children given allogeneic HSCT 131 a<br />

possible intriguing approach is that of generating and<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

127<br />

expanding clones or cell lines that are leukemia-reactive.<br />

The first elegant demonstration of the feasibility and efficacy<br />

of this sophisticated strategy has been recently<br />

reported by Falkenburg et al., 132 who, through the infusion<br />

of donor-derived in vitro cultured CTL specifically recognizing<br />

leukemia progenitor cells, induced a complete<br />

hematologic and cytogenetic response in a patient with<br />

CML who had relapsed after an allograft and was resistant<br />

to DLI treatment.<br />

A diverse, but equally elegant, approach proposed to<br />

abrogate the DLI-associated GvHD and its relevant morbidity<br />

and mortality is the infusion of thymidine kinase<br />

gene-transduced DLI followed by treatment of the recipient<br />

with ganciclovir if GvHD occurs. 133 In a study reported<br />

by Bonini et al. 133 this strategy proved to be able to<br />

control GvHD in 3 patients experiencing this complication<br />

after DLI; two of them, who had achieved a complete<br />

hematologic remission before ganciclovir administration,<br />

remained in full remission after disappearance of<br />

the transduced lymphocytes. If confirmed in a larger<br />

number of patients with a longer follow-up, genetic<br />

manipulation of donor lymphocytes, through the transfer<br />

of a suicide gene for specific and selective elimination<br />

of effector cells responsible for GvHD, could demonstrate<br />

the possibility of separating GvHD from GVL effect,<br />

thus sparing the anti-leukemia activity of DLI.<br />

One of the most important, still unsolved problem of<br />

DLI is that concerning the much lower efficacy of GVL in<br />

patients with acute leukemia than in those with CML. An<br />

immediate explanation for this observation may be that<br />

the more rapid growth kinetics of blast cells, which<br />

occurs in patients with acute leukemia during the lag<br />

period between leukocyte infusion and GVL development,<br />

may hamper the immune-mediated effect played by<br />

donor lymphocytes in controlling disease progression. In<br />

fact, response to DLI occurs after weeks and hence the<br />

exponential expansion of leukemia cells in vivo may<br />

exceed the immune response. 112,113,124 The more encouraging<br />

results obtained when DLI is used as consolidation<br />

therapy for patients who have obtained a complete<br />

remission after chemotherapy provide support for this<br />

interpretation. However, other hypotheses, involving different<br />

intrinsic susceptibility of acute leukemia to adoptive<br />

immunotherapy must be considered. In particular,<br />

since patients with ALL have the lowest chance both of<br />

responding to DLI and of benefitting from the GVL effect<br />

after bone marrow transplantation, 134 a peculiar resistance<br />

of lymphoid leukemia to immunotherapy cannot<br />

be excluded.<br />

As peptides differentially expressed within the<br />

hematopoietic system can trigger and act as a target of<br />

the GVL reaction, 112,124 it could be hypothesized that, for<br />

example, the presence of these antigens on myeloid<br />

blasts, but not on lymphoid leukemia cells accounts for<br />

the low response of ALL to donor lymphocytes. The<br />

reported demonstration of CTL response directed<br />

towards peptides derived from proteinase 3, which is<br />

expressed by myeloid cells (including blast cells), 135 is a<br />

typical example of the possible differential susceptibility<br />

to the immune-mediated anti-leukemia effect of different<br />

types of hematologic malignancies.<br />

Several other possibilities exist to explain why acute<br />

leukemia (and in particular ALL) can escape the GVL<br />

effect. For example, leukemia cells may have defective<br />

expression of HLA-class I or II molecules on their surface<br />

such that they do not present antigens or, alternatively,<br />

the mechanisms of antigen processing and transport may<br />

be impaired. 112,129 Moreover, leukemia blasts may product<br />

cytokines (such as transforming growth factor , IL-<br />

10) capable of suppressing T-cell activation, expansion<br />

and effector function or may express on their cell surface<br />

molecules, such as FAS ligand, able to mediate T-<br />

cell apoptosis. 112,129 One of the most interesting fields of<br />

investigation for explaining why in some patients a sustained<br />

anti-leukemia response in vivo fails to be induced<br />

is that of co-stimulatory molecules. As previously<br />

described, full activation of T-cells requires two distinct<br />

but synergistic signals. 136 In fact, in the absence of costimulatory<br />

signals, a T cell encountering an antigen<br />

becomes unresponsive to the appropriate stimulation<br />

(anergic) 137 or undergoes programmed cell death (apoptosis).<br />

138 Leukemia cells lacking these co-stimulatory<br />

molecules have a poor capacity of inducing a T-cell specific<br />

immune response and induction of CD80 and CD86,<br />

by signalling through the CD40 molecule, is able to<br />

restore T-cell co-stimulation via CD28 and to generate<br />

both allogeneic and autologous CTL, which could contribute<br />

to inducing or maintaining a state of hematologic<br />

remission. 139,140<br />

Some clinical strategies have been devised to improve<br />

the efficacy of adoptive immunotherapy in patients with<br />

acute leukemia. An approach for ameliorating the efficacy<br />

of DLI which has produced interesting results is that<br />

recently reported by Slavin et al., 106 who documented that<br />

the success rate of this adoptive immune therapy may be<br />

increased in patients with both acute and chronic<br />

leukemia by activation of donor peripheral blood lymphocytes<br />

with IL-2 both in vivo and/or in vitro. In particular,<br />

a relevant proportion of patients who had not<br />

responded to DLI were induced into remission only after<br />

in vivo administration of IL-2 or in vitro activation of<br />

donor lymphocytes. If further confirmed, the results<br />

obtained make it possible to hypothesize that this strategy<br />

could be employed as first-line treatment of patients<br />

with acute leukemia relapsing after an allograft, since<br />

ALL and to a lesser extent AML patients do not greatly<br />

benefit from DLI alone. Another reasonable attempt for<br />

improving the response to DLI in patients with acute<br />

leukemia is to use this adoptive immunotherapy in individuals<br />

with minimal residual disease, as determined by<br />

cytogenetic investigations or sensitive molecular tools,<br />

that is in conditions characterized by a limited tumor<br />

burden, in which the GVL effect has demonstrated its<br />

greatest efficacy.<br />

Unmanipulated DLI may also provide a means of compensatory<br />

T-cell repletion for the prevention of leukemia<br />

recurrence in patients given a T-cell depleted marrow<br />

transplantation from a relative. This approach has been<br />

haematologica vol. 85(suppl. to n. 12):December 2000


128<br />

C. Bordignon et al.<br />

recently proposed 141 and studies enrolling larger cohorts<br />

of patients are necessary to define whether this strategy<br />

can be useful to prevent the increased risk of relapse<br />

associated with the removal of donor T-cells. However,<br />

the main indication of adoptive infusion of donor<br />

immune cells to accelerate immune reconstruction after<br />

HSCT is transplants from HLA-disparate family donors.<br />

Infusion of a high number of T-cell depleted, peripheral<br />

blood hematopoietic progenitors from these donors<br />

has been demonstrated to be associated with a high<br />

chance (>95%) of donor hematopoietic engraftment. 142<br />

The significant delay in immune reconstitution, due<br />

mainly to removal of mature T cells from donor marrow<br />

and HLA disparity between donor and recipient, remains<br />

the major problem of HSCT from HLA-disparate donors.<br />

In fact, it is responsible for the dramatic incidence of<br />

leukemia relapse and life-threatening viral and fungal<br />

infections observed after this type of HSCT. A possible<br />

strategy to improve the process of immune recovery is<br />

to infuse donor T-lymphocytes selectively rendered nonreactive<br />

towards alloantigens of the recipient, but maintaining<br />

the capacity to generate an immune response<br />

against viruses, fungi and leukemia cells. In this regard,<br />

as previously mentioned, the manipulation of co-stimulatory<br />

molecules is an extremely promising field of<br />

investigation, since the absence of a second signal<br />

induces anergy rather than activation of T lymphocytes.<br />

Drugs and monoclonal antibodies blocking co-stimulatory<br />

pathways have been demonstrated to be able to<br />

prevent T-cell activation in response to alloantigens and<br />

to induce a state of anergy. 143 In particular, it was<br />

recently documented that the combination of monoclonal<br />

antibodies blocking CD80/CD86 molecules and<br />

cyclosporin A was able to generate a state of selective<br />

in vitro unresponsiveness of T cells towards allo-antigens,<br />

not reversed by adding IL-2. 144 Since the induction<br />

of this state of unresponsiveness was associated with<br />

the maintenance of in vitro capacity to respond toward<br />

virus antigens and leukemia cells, 145 the relevance of<br />

this approach is evident for strategies of donor T-cell<br />

add-back after T-cell depleted transplant of hematopoietic<br />

progenitors from HLA-partially matched donors<br />

aimed at accelerating the process of immune reconstitution.<br />

A different, but equally promising, method of deletion<br />

of unwanted alloresponses is based on the elimination<br />

of alloreactive T-cells after specific activation through<br />

their killing 146 or fluorescence-activated cell sorting, 147<br />

while sparing T cells with other functions. In a human<br />

pre-clinical study, it was demonstrated that allospecific<br />

T-cell depletion by using an immunotoxin directed<br />

against the p55 chain of IL-2 receptor, was feasible and<br />

specific. 146 The spared T-cells were still able to proliferate<br />

against third-party cells, Candida and cytomegalovirus<br />

antigens, 148 as well as to kill both leukemia blasts<br />

and autologous EBV-B lymphoblastoid cell lines. 149<br />

Moreover, in vivo studies in a murine animal model<br />

showed that this particular T-cell depletion was efficient,<br />

at least partially, in preventing both graft rejection<br />

and GvHD in a complete haplotype mismatched<br />

combination. 150<br />

Finally a brief mention should be made of the generation<br />

and infusion of T cells with suppressive and regulatory<br />

activity. A particular subset of these cells called<br />

Tr1 has recently been described by Groux et al., 151 who<br />

in an animal model demonstrated the ability of this population<br />

to prevent, through their activity on naive cells,<br />

the occurrence of ovo-albumin induced inflammatory<br />

bowel disease. Whether these cells will have a role in<br />

promoting a true state of tolerance in transplant of<br />

hematopoietic progenitors or solid organs (in which the<br />

immune response to alloantigens is mainly sustained by<br />

memory cells) remains to be proved in specific pre-clinical<br />

and clinical studies currently underway.<br />

Adoptive immunotherapy for the treatment<br />

of viral infections in immunocompromised<br />

patients<br />

Prevention or treatment of viral infections in immunecompromised<br />

patients through the infusion of specific T-<br />

cell lines or clones is one of the most sophisticated<br />

examples of adoptive immunotherapy approaches. 152 In<br />

fact, it implies the elaboration of true cellular-engineering<br />

strategies able to generate, select and expand lymphocyte<br />

subsets, which display a specific function. The<br />

first study in humans to evaluate the efficacy of adoptively<br />

transferred T-cell clones for reconstitution of specific<br />

immunity was performed in recipients of allogeneic<br />

HSCT at risk of developing human cytomegalovirus<br />

(HCMV) infection and/or disease. 153 Even though preemptive<br />

therapy of HCMV infection based on monitoring<br />

of antigenemia 154 and prophylaxis of seropositive<br />

HSCT recipients using antiviral drugs (i.e. ganciclovir and<br />

foscarnet) 155 have significantly reduced the number of<br />

patients experiencing HCMV disease, this viral infection<br />

still represents a major life-threatening complication of<br />

stem cell allograft. The capacity to recover from a severe<br />

HCMV infection in transplanted patients is directly correlated<br />

with the ability of the host to generate virusspecific<br />

class I HLA-restricted CD8 + cytotoxic cells and<br />

during the first 100 days after HSCT approximately 50%<br />

of patients are persistently deficient in CD8 + cytotoxic T-<br />

lymphocytes specific for HCMV. 156,157 It is not surprising<br />

that, to evaluate the efficacy of adoptive immunotherapy<br />

in this viral infection, HCMV-specific CD8 + T-cell<br />

clones of donor origin were generated and infused in<br />

HSCT recipients. 153,158 These cells, generated through a<br />

highly complex expansion strategy using irradiated<br />

donor-origin skin fibroblasts infected with a strain of<br />

HCMV, proved to be efficient in the prophylaxis against<br />

HCMV infections that can complicate allogeneic HSCT.<br />

Moreover, the cloning strategy allowed selection of T<br />

cells which lacked significant alloreactive capacity and,<br />

thus, did not cause clinically relevant GvHD or toxicity.<br />

These clones, directed towards either pp65 or pp150 (two<br />

abundant viral tegument proteins presented for recognition<br />

by cytotoxic T-lymphocytes), restored HCMV-spe-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

129<br />

cific cytotoxicity, which persisted for several weeks. 158 In<br />

fact, through a PCR technique able to detect the V and<br />

V T-cell receptor rearrangements specific for the donor<br />

clones, it was possible to prove the donor origin of these<br />

cells formally and to document the persistence of the<br />

adoptively transferred HCMV-specific T-cells for at least<br />

12 weeks. Unfortunately, these clones persisted in the<br />

circulation at high levels only in patients experiencing an<br />

endogenous recovery of CD4 + virus-specific cells. 158 By<br />

contrast, in patient lacking this spontaneous recovery of<br />

HCMV-specific CD4 + lymphocyte, the donor-origin,<br />

adoptively transferred cytotoxic T-cell activity progressively<br />

declined and eventually disappeared. This observation<br />

emphasizes the importance of CD4 + lymphocytes<br />

in promoting sustained restoration of antigen-specific<br />

immunity and suggests that the use of polyclonal T-cell<br />

lines containing both CD4 + and CD8 + cells could be<br />

preferable to the infusion of cytotoxic T-cell clones.<br />

In this regard, the use of T-cell lines for prevention<br />

and/or treatment of Epstein-Barr virus-induced lymphoproliferative<br />

disorders (LPD) has represented a further,<br />

equally sophisticated, evolution of the approaches<br />

of adoptive immunotherapy for the restoration of<br />

virus-specific immunity. EBV-LPD have emerged as a<br />

significant complication for both HSCT and solid organ<br />

transplant recipients. 159-161 In the former cohort, the use<br />

of HLA-partially matched family and unrelated donors,<br />

as well as selective procedures of T-cell depletion sparing<br />

B-lymphocytes, are risk factors for the development<br />

of EBV-LPD. 160-162 In HSCT recipients these disorders are<br />

of donor origin and usually present in the first 4-6<br />

months after transplantation, whereas in patients given<br />

a solid organ allograft they usually develop from the<br />

recipient B-lymphocytes months to years after transplantation.<br />

160,161 High levels of EBV-DNA in blood and in<br />

vitro spontaneous growth of EBV-lymphoblastoid cell<br />

lines predict development of these lymphoproliferative<br />

disorders. 163 They often present as high-grade diffuse<br />

large cell B-cell lymphomas, which are oligoclonal or<br />

monoclonal and express the full array of EBV antigens<br />

including EBNA-1 through EBNA-6 and the latency<br />

membrane proteins LMP-1 and LMP-2. 161 The lymphomas<br />

which develop in immunocompromised hosts<br />

not only invade the hematopoietic system, but also the<br />

lung, nasopharynx and central nervous system. The therapeutic<br />

approaches proposed to date (i.e. discontinuation<br />

of immunosuppression, -IFN, antiviral agents and<br />

cytotoxic chemotherapy) have been applied with varying,<br />

but overall unsatisfactory, results; moreover, graft<br />

rejection, GvHD and toxicity are frequent complications<br />

of these strategies, and mortality rate due to EBV-LPD<br />

remains high. 160,161<br />

Normal EBV seropositive individuals have a high frequency<br />

of circulating virus-specific cytotoxic T-lymphocytes<br />

precursors, which control outgrowth of EBV-infected<br />

B-cells. Since EBV-LPD in immunocompromised hosts<br />

appears to stem from a deficiency of virus-specific cytotoxic<br />

activity, it is reasonable to hypothesize that an<br />

adoptive immunotherapy approach with donor-derived<br />

T-lymphocytes could be able to prevent unchecked lymphoproliferation<br />

and eradicate established disease. In<br />

1994, the Sloan Kettering group first demonstrated that,<br />

through the infusion of unselected peripheral blood<br />

mononuclear cells from a donor, 5 patients given HSCT<br />

with post-transplant EBV-LPD obtained remission of the<br />

disease. 164 However, this treatment was associated with<br />

development of clinically relevant GvHD and 2 patients<br />

of inflammatory-mediated lung damage, leading to respiratory<br />

failure.<br />

A further refinement of this approach was achieved by<br />

Rooney and colleagues, who generated EBV-specific T-<br />

cell lines from donor lymphocytes and infused them as<br />

prophylaxis against EBV-LPD in patients given T-cell<br />

depleted HSCT from HLA-disparate family or unrelated<br />

donors, and, thus, considered at high risk for this disease.<br />

165 The infusion of these polyclonal T-cell lines<br />

proved to be safe and effective in the prevention of EBV-<br />

LPD. Moreover, these cytotoxic cells may also have a<br />

role in the treatment of established disease. 165 The most<br />

recent update of this experience confirms that the infusion<br />

of EBV-specific T-cell lines is highly effective for the<br />

prevention of EBV-LPD, since none of 39 patients given<br />

a T-cell depleted allograft and treated with this adoptive<br />

immunotherapy developed the disease, as compared<br />

to 7 out 61 transplanted patients not receiving the prophylactic<br />

treatment. 166 Gene marking studies have<br />

shown the persistence of these donor-derived EBV-specific<br />

cell cytotoxic lines in patient’s peripheral blood for<br />

months after infusion and their re-appearance after<br />

periods of apparent non-identifiability during episodes<br />

of viral reactivation, this further stressing the importance<br />

of helper T-cell function in the persistence of<br />

transferred CD8 + cells. 167<br />

The profound immunosuppression necessary for graft<br />

survival carries a well-recognized predisposition to the<br />

development of viral complications, in particular EBV-<br />

LPD, also in recipients of solid organ transplantation. 159<br />

An immunotherapy approach to EBV-LPD using autologous<br />

in vitro generated EBV-specific cytotoxic lines could<br />

be an appealing strategy in this cohort of patients. Support<br />

for this hypothesis is given by the recently described,<br />

although not unexpected, possibility of generating, from<br />

pre-transplantation blood samples of EBV-seropositive<br />

solid organ transplant recipients, virus-specific T-cell<br />

lines which are effective in controlling EBV replication<br />

post-transplantation. 168 However, generation and storage<br />

of cytotoxic lines for each patient undergoing solid<br />

organ transplantation requires enormous, unavailable<br />

levels of funding, laboratory facilities and workforce. A<br />

more rational strategy is to generate, expand and infuse<br />

autologous EBV-specific cytotoxic lines from the peripheral<br />

blood of organ transplant patients presenting<br />

increased EBV-DNA levels after transplantation, which, as<br />

previously mentioned, are a risk factor for EBV-LPD development.<br />

The feasibility of generating autologous EBVspecific<br />

cytotoxic lines from the peripheral blood of organ<br />

transplant patients receiving in vivo immunosuppression<br />

for prevention of graft rejection has been recently<br />

haematologica vol. 85(suppl. to n. 12):December 2000


130<br />

C. Bordignon et al.<br />

proved. 169 Moreover, these cytotoxic T-lymphocytes were<br />

demonstrated to be able to display EBV-specific killing in<br />

vivo, as proved by prompt viral DNA clearance, without<br />

augmenting the probability of graft rejection. A peculiar<br />

problem, fortunately not particularly common, is that of<br />

EBV-seronegative patients, who develop primary EBV<br />

infection after solid organ transplantation. In fact, in<br />

these patients, in vitro generation of virus-specific T-cell<br />

lines able to control EBV-driven B-cell proliferation can<br />

be particularly complicated, time-consuming and sometimes<br />

unsuccessful.<br />

Autologous EBV-specific cytotoxic lines with demonstrated<br />

anti-viral activity in vitro and in vivo may also<br />

have a role in the treatment of other EBV-associated primary<br />

malignancies: for example, 40-50% of patients<br />

with Hodgkin’s disease tumor cells are EBV-antigen positive<br />

and may therefore be suitable targets for virus specific<br />

cytotoxic lymphocytes. 160,170 A recently reported<br />

study provides further support for this possibility, documenting<br />

that, although more complicated than in normal<br />

donors, generation of EBV-specific cytotoxic lines is<br />

feasible in a relevant proportion of patients with EBVpositive<br />

Hodgkin’s disease. 171 These lines retained their<br />

potent antiviral effects in vivo and persisted for more<br />

than 13 weeks in patients with relapsed Hodgkin’s disease.<br />

171 Whether this approach of adoptive immunotherapy<br />

will become an adjunctive treatment option for<br />

patients failing to gain benefit from conventional<br />

chemotherapy remains to be proved in prospective clinical<br />

trials.<br />

Finally, it should be mentioned that adoptive transfer<br />

of cytotoxic T-cell response could be of value also in<br />

the prevention or treatment of other viral infections that<br />

cause morbidity and mortality in immunocompromised<br />

patients. In this regard, pre-clinical studies are underway<br />

to establish systems for generating cytotoxic T-cell<br />

responses to adenovirus. 160,172<br />

Genetically engineered donor lymphocyte<br />

infusion for treatment of leukemia<br />

relapse and as a means of accelerating<br />

immunologic reconstitution in patients<br />

given transplantation of hematopoietic<br />

progenitors<br />

Tumor recurrence is the major cause of treatment failure<br />

of autologous bone marrow transplantation. 173,174<br />

Indeed, the rate of tumor relapse is lower when transplantation<br />

is performed between matched unrelated or<br />

mismatched family member donor and recipients. It is<br />

now established that the curative potential of allo-BMT<br />

is represented by the additional effect of high dose<br />

chemo-radiotherapy in addition to the presence of allogeneic<br />

T-lymphocytes that are responsible for the<br />

GVL. 175,176 However, the therapeutic impact of allogeneic<br />

BMT is limited by the inevitable occurrence of<br />

GvHD. 177 Severe GvHD can be circumvented by the in vitro<br />

removal of T-lymphocytes from the BMT. 175 However,<br />

recipients of depleted marrow have delayed immune<br />

recovery, and increased incidences of viral infections<br />

and tumor relapse. 178,179<br />

Recent studies have shown the clinical efficacy of the<br />

adoptive transfer of immune effectors specific for viral<br />

antigens 153,195,167 in patients who underwent BMT. In this<br />

context gene transfer of a marker gene provides a means<br />

of evaluating the survival, homing and efficacy of the<br />

infused cells.<br />

In marrow-transplanted recipients, lymphoproliferative<br />

disorders associated with EBV, a human herpes virus<br />

that normally replicates in epithelial cells of the oropharyngeal<br />

tract, occurs in 5-30% of the treated patients.<br />

EBV-LPD are usually malignant B-cell lymphomas of<br />

donor origin, which may be either polyclonal or monoclonal.<br />

The latter have a rapidly progressive, fulminating<br />

and fatal course. 180,181 The transformed B cells express<br />

virus-encoded latent cycle nuclear antigens, latent membrane<br />

proteins, and a number of cell adhesion molecules.<br />

Most of these viral proteins are recognized as antigens<br />

by the immune system of a normal individual. 182 In the<br />

normal host, in fact, EBV-induced lymphoid proliferation<br />

is controlled by EBV-specific and MHC-restricted T-<br />

lymphocytes, MHC-unrestricted effectors and by antibodies<br />

directed toward specific viral antigens. Since a<br />

limited number of specific cytotoxic T-lymphocytes is<br />

required for controlling EBV-transformed B-lymphocytes<br />

in normal individuals, the administration of donor lymphocytes<br />

for the occurrence of EBV-LPD in recipients of<br />

T-cell depleted bone marrow transplantation could control<br />

this severe complication by providing the patient<br />

with donor immunity against EBV. 164,183 Successful<br />

regression of the disease, documented histologically and<br />

by full clinical remission, has been achieved by the infusion<br />

of unmanipulated donor leukocytes. 164 However,<br />

acute or mild chronic GvHD developed in all the patients<br />

who responded to the treatment. 164<br />

To prevent GvHD, Brenner’s group has evaluated the<br />

use of EBV-specific CTL rather than unmanipulated T<br />

cells. Donor derived EBV-specific CTL have been generated<br />

in vitro by stimulation with irradiated donorderived<br />

EBV-infected lymphoblastoid cell lines (LCL). 184<br />

The polyclonal effector populations were predominantly<br />

CD8 + with a varying number of CD4 and showed specific<br />

cytotoxic activity toward the EBV-infected target<br />

cells. In order to investigate the long-lasting survival of<br />

the injected cells, the anti-EBV effectors were marked<br />

with the neo-gene before administration.<br />

Neo-marked cells were detected in circulation for at<br />

least 10 weeks after the injections. 165 Moreover, the<br />

infusions allowed the establishment of a population of<br />

CTL precursors that could be activated to proliferate by<br />

in vivo or in vitro challenge with the virus. 166 The authors<br />

showed that EBV-specific CTL lines expressing the neomarker,<br />

could be derived from patient's peripheral blood<br />

lymphocytes (PBL) for up to 18 months, by in vitro restimulation<br />

with the autologous EBV-lines. 166<br />

These findings support a more widespread use of antigen-specific<br />

CTL in the treatment of infections and cancer.<br />

Their use may extend in the near future to other dis-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

131<br />

eases which express well-known antigens that could<br />

serve as target of CTL therapy (e.g. Hodgkin’s disease and<br />

nasopharyngeal carcinoma).<br />

The adoptive transfer of in vitro stimulated effectors<br />

achieves clinical results without causing the appearance<br />

of GvHD. However, the application of this strategy<br />

to a large number of allo-BMT treated patients, especially<br />

in prophylaxis protocols has some limitations<br />

related to the in vitro manipulation necessary for the<br />

generation of specific effectors (e.g. availability of<br />

donor-EBV lines; in vitro stimulation and expansion of<br />

antigen-specific effectors). An alternative approach was<br />

proposed in 1994 by the S. Raffaele Hospital group. 185,186<br />

Their protocol was aimed at maintaining the potential<br />

of the infusion of polyclonal cell lines while providing a<br />

specific means to control acute GvHD. To this aim they<br />

transduced donor lymphocytes by a retroviral vector<br />

containing a suicide gene for in vivo selective elimination<br />

of the infused lymphocytes.<br />

It was previously shown that introduction of a gene<br />

encoding for a susceptibility factor, a so-called suicide<br />

gene, makes transduced cells sensitive to a drug not<br />

ordinarily toxic. 187,188 A series of retroviral vectors carrying<br />

a suicide gene for ganciclovir-mediated in vivo selective<br />

elimination of the infused lymphocytes was<br />

designed. The vectors carried either an HSV-thymidinekinase-neo<br />

(Tk-neo) fusion gene, coding for a chimeric<br />

protein for both negative and positive selection, or the<br />

HSV-Tk gene alone. 189<br />

A crucial prerequisite for the application of this strategy<br />

in the clinical context is the transduction of all<br />

infused donor lymphocytes. For this purpose, the<br />

designed retroviral vectors also carried a gene encoding<br />

a modified (non-functional) cell surface marker not<br />

expressed on human lymphocytes. Positive immunoselection<br />

of the transduced cells 190 by the use of the<br />

cell surface marker resulted in virtually 100% genemodified<br />

lymphocytes.<br />

Based upon the preclinical data described above, a<br />

clinical protocol was developed 186 for the use of donor<br />

lymphocytes transduced by the SFCMM-2 retroviral vector<br />

for transfer and expression of two genes: the HSV-<br />

Tk gene that confers to the transduced PBL in vivo sensitivity<br />

to the drug ganciclovir, for in vivo specific elimination<br />

of cells potentially responsible for GvHD; and a<br />

modified (non-functional) form of the low affinity<br />

receptor for the nerve growth factor gene (∆LNGFr), for<br />

in vitro selection of transduced cells and for in vivo follow-up<br />

of the infused donor lymphocytes.<br />

Increasing doses (beginning at 110 6 /kg) of donor<br />

PBL were infused into several patients affected by<br />

hematologic malignancies who developed severe complications<br />

following a T-cell depleted BMT from their<br />

HLA-identical related donors. After the infusion, the<br />

transduced lymphocytes could be detected in the blood<br />

of patients by cytofluorimetric and PCR analyses. In particular<br />

one patient affected by an EBV-LPD, showed a<br />

progressive increase in the number (up to 13.4% of the<br />

total PBL) of infused marked lymphocytes that was<br />

accompanied by a complete clinical response. However,<br />

signs of acute GvHD, confirmed by skin biopsy, were<br />

observed approximately four weeks after the infusion<br />

of the transduced-donor lymphocytes. The intravenous<br />

(i.v.) administration of two doses of ganciclovir (10<br />

mg/kg/day) quickly resulted in elimination of marked<br />

donor PBL, and near resolution of all clinical and biochemical<br />

signs of acute GvHD. 187<br />

As mentioned before, when comparable preparative<br />

regimens are employed, the rate of tumor recurrences<br />

after autologous BMT is significantly higher than the<br />

rate observed after allogeneic BMT. GvHD develops in<br />

50-70% of patients undergoing allogeneic BMT. The<br />

effectors of such response are thought to be mature<br />

donor lymphocytes from the marrow graft that respond<br />

to the foreign major and/or minor histocompatibility<br />

antigens of the recipient and also recognize and destroy<br />

the tumor cells. In fact, patients who underwent mature<br />

T-cell-depleted allogeneic BMT have a lower rate of<br />

GvHD but also a higher rate of leukemia relapses. 178,179<br />

The infusion of donor lymphocytes, early after T-celldepleted<br />

allogeneic BMT, increases the incidence of<br />

GvHD without improving the control of leukemia. 191<br />

However, a delayed transfusion of donor lymphocytes,<br />

when graft tolerance is established, seems to be more<br />

effective in preventing and treating tumor relapses.<br />

Indeed the delayed administration of donor lymphocytes<br />

has recently become a new tool for treating<br />

leukemic relapse after BMT. Patients affected by post-<br />

BMT recurrence of chronic myelogenous leukemia, acute<br />

leukemia, lymphoma, and multiple myeloma could<br />

achieve complete remission after the infusion of donor<br />

leukocytes without requiring cytoreductive chemotherapy<br />

or radiotherapy, 106,192-194 even though the response<br />

rate of patients with acute leukemia, non-Hodgkin’s<br />

lymphoma and multiple myeloma is significantly lower<br />

than that of patients affected by chronic myelogenous<br />

leukemia. Although the delay in the administration of T<br />

lymphocytes is expected to reduce the risk of GvHD, this<br />

risk is still present at higher doses of donor T-cells. 116<br />

Therefore, as described above, a clinical protocol was<br />

developed, for the use of donor lymphocytes transduced<br />

by the SFCMM-2 retroviral vectors 186 for transfer and<br />

expression of the HSV-Tk gene, and the cell surface<br />

marker ∆LNGFr, for in vitro selection of 100% transduced<br />

cells and for in vivo follow-up of the infused<br />

donor lymphocytes. 190<br />

In a phase I-II study, eight patients affected by hematologic<br />

malignancies who developed severe complications<br />

following an allogeneic T-cell depleted BMT,<br />

received escalating doses of donor PBL transduced by<br />

the described retroviral vector. 133 After gene transfer,<br />

transduced cells were selected for the expression of the<br />

cell surface marker ∆LNGFr by the use of specific<br />

immunobeads and the proportion of transduced cells<br />

was assessed by cytofluorimetric analysis. 190 In this study,<br />

we made the following observations: 1) transduced cells<br />

survived long-term in vivo and were detectable by cytofluorimetric<br />

analysis and PCR in high proportions (up to<br />

haematologica vol. 85(suppl. to n. 12):December 2000


132<br />

C. Bordignon et al.<br />

13.4% of circulating PBL) and long-term (up to 6<br />

months); 2) three patients showed complete response,<br />

three patients had partial response, one progressed with<br />

no response, and one patient could not be evaluated; 3)<br />

three patients developed GvHD that required ganciclovir<br />

treatment; 4) ganciclovir-mediated elimination of transduced<br />

cells resulted in near resolution of all clinical and<br />

biochemical signs of acute GvHD. Data from this study 133<br />

indicate that genetically modified cells maintain their in<br />

vivo potential to develop both anti-tumor and GvHD<br />

effect, and may represent a new potent tool for exploiting<br />

anti-tumor and anti-host immunity, while providing<br />

a specific means for eliminating acute GvHD, in the<br />

absence of any immunosuppressive drug.<br />

A potential limitation of the clinical approaches<br />

described could be the development of a specific<br />

immune response against vector-encoded proteins,<br />

which might allow the selective elimination of the<br />

transduced cells by the host immune system. For some<br />

gene products, such as the hygromycin-thymidine<br />

kinase (Hy-Tk) fusion protein, a specific immune<br />

response, able to eliminate large numbers of transduced<br />

cells in less than 48 hours, has been described in HIVpatients.<br />

195<br />

We observed that immune recognition and killing of<br />

cells transduced by retroviral vectors is a more general<br />

phenomenon related to the foreign nature of the proteins<br />

expressed by the injected cells. Indeed, cells<br />

expressing the widely used marker gene neo and the<br />

HSV-Tk gene are targets of a strong immune response,<br />

while the endogenous proteins (e.g. the cell surface<br />

marker ∆LNGFr) are not recognized, even if ectopically<br />

expressed in a context which is otherwise extremely<br />

immunogenic. 196 The relative immunogenicity detected<br />

for the three vector-encoded components (none by<br />

∆LNGFr, low by HSV-Tk, high by neo) clearly outlined<br />

the modifications of this type of gene therapy. Since neo<br />

is the only component not-strictly necessary for the<br />

strategy and can be efficaciously replaced by the surface<br />

marker for all in vitro handling and selection, 189,197 the<br />

immunogenicity of the new neo-less vectors should be<br />

reduced.<br />

The clinical results obtained with gene modified donor<br />

lymphocytes, for the treatment of hematologic relapses<br />

and EBV-lymphoproliferative disorders, suggest the<br />

potential use of this approach. 133 The transfer of a suicide<br />

gene, that allows selective and specific elimination<br />

of effector cells of GvHD may allow full advantage to be<br />

taken of the beneficial effect of allogeneic lymphocytes<br />

with the possibility of eliminating all unwanted effects<br />

of GvHD in the absence of toxic side effects. A large<br />

scale application of this strategy will increase the number<br />

of patients who could potentially benefit from allogeneic<br />

BMT by allowing the use of less compatible marrow<br />

donors.<br />

With regard to the immune recovery associated with<br />

the genetically-engineered donor lymphocytes, our group<br />

has recently obtained in vitro data demonstrating that<br />

genetically-engineered donor T-cells maintain a normal<br />

TCR V immune repertoire and retain antigen-specific<br />

lytic activity against an allogeneic target or an autologous<br />

EBV cell line at cytotoxic T-cell precursor frequencies<br />

comparable to unmodified lymphocytes. In the light<br />

of this in vitro evidence, and our previous clinical application,<br />

133 a clinical trial, based on the prophylactic infusion<br />

of 110 7 /kg HSV-Tk transduced T-cells six weeks<br />

after T-cell-depleted bone marrow transplantation, was<br />

developed. In the first five treated patients we documented<br />

the presence of various proportions of transduced<br />

cells in the peripheral blood. In particular, geneticallyengineered<br />

donor lymphocytes were responsible for antiviral<br />

immune reconstitution in one patient. CD3 + lymphocytes<br />

began to appear in the circulation of this<br />

patient two weeks after the infusion of HSV-Tk T-cells. All<br />

the CD3 + lymphocytes were genetically engineered as<br />

documented by the expression of the cell surface marker<br />

∆LNGFr. These cells retained a polyclonal TCR repertoire<br />

and were probably responsible for the clearance of<br />

a persistent CMV antigenemia. Indeed, the CMV antigenemia<br />

dropped below levels which could be detected<br />

by PCR shortly after the appearance of circulating genetically-engineered<br />

CD3 + T cells in the absence of any<br />

antiviral drug therapy. 198 These data, if confirmed in a<br />

larger number of patients with longer follow-up, suggest<br />

that in addition to the anti-tumor activity, the infusion<br />

of genetically-engineered donor lymphocytes may play a<br />

role in restoring immunity against opportunistic infections<br />

early after allogeneic BMT.<br />

Dendritic cells as natural adjuvants in<br />

cancer immunotherapy<br />

Among professional antigen presenting cells (APC),<br />

dendritic cells (DC) are specialized in capturing and processing<br />

antigens into peptide fragments that bind to<br />

major histocompatibility complex molecules. DC are the<br />

most potent stimulators of T-cell responses and they<br />

are unique in that they stimulate not only memory but<br />

also naive T-lymphocytes. Thus, DC appear critical<br />

(nature adjuvants) for the induction of B-and T-cellmediated<br />

immune responses. Recent evidence in experimental<br />

models supports the role of DC for immunization<br />

strategies aimed at stimulating specific anti-tumor<br />

immunity.<br />

In this section we will briefly review:<br />

1. the biological characterization of DC;<br />

2. different strategies for ex vivo generation of DC;<br />

3. methods for the efficient delivery of tumor associated<br />

antigens (TAA) to DC;<br />

4. the use of DC for cellular immunotherapy.<br />

Biological characterization of dendritic<br />

cells<br />

DC are widely distributed in the body and are particulary<br />

abundant in tissues that interface the environment<br />

(i.e. Langerhans cells in the skin and mucous membranes)<br />

and in lymphoid organs (interdigitating DC)<br />

where they act as sentinels for incoming pathogens.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

133<br />

Inflammatory signals such as TNF- and IL-1 as well<br />

as bacteria, bacterial products (LPS) and viruses induce<br />

migration of antigen-loaded DC from the peripheral tissues<br />

to secondary lymphoid organs. During migration,<br />

DC mature and upregulate MHC, adhesion and co-stimulatory<br />

molecules, thus strongly augmenting their ability<br />

to prime T-cells. 199-204<br />

The functional activity of DC derives from a number<br />

of properties of these cells (Figure 2). Their dendritic<br />

shape, along with the high level of expression of certain<br />

adhesion molecules and integrins (LFA-3, ICAM-1,<br />

ICAM-3), increases the area of contact with the effector<br />

cells of the immune system. 205 DC strongly express<br />

the HLA class II molecules -RD, -DQ and -DP and costimulatory<br />

molecules (CD80, CD86 and CD40) which<br />

activate their ligands on T-cells (CD28, CTLA-4 and<br />

CD40L), thus providing the second signal strictly necessary<br />

to induce a proliferative response, rather than tolerance,<br />

upon antigen recognition. 199 In addition, DC produce<br />

a number of cytokines including IL-12 which promotes<br />

a cytotoxic immune response by inducing the differentiation<br />

of TH0 cells to IFN- and IL-2-producing<br />

TH1 cells. 206,207 It has recently been demonstrated that<br />

upon Ag recognition, T-helper cells activate DC via<br />

CD40-CD40L interaction and activated DC are then able<br />

to trigger a cytotoxic response from T-killer cells. 208-210<br />

However, DC are present in peripheral tissues in an<br />

immature state unable to prime T-cells. At this stage of<br />

differentiation, they can very efficiently take up soluble<br />

antigens, particles and micro-organisms by phagocytosis,<br />

macropinocytosis or by the macrophage mannose receptor,<br />

Fc and Fc receptors, 211 but they lack all the accessory<br />

signals for T-cell activation. Antigen uptake induces<br />

DC to maturation by up-regulating MHC and co-stimulatory<br />

molecules as well as DC-associated Ag (e.g. CD83<br />

and p55) whereas the capacity to capture and process Ag<br />

is lost. However, full activation of DC is dependent upon<br />

the contact with T-cells by the CD40-CD40L interaction<br />

which induces the production of IL-12. Thus, the key<br />

functions of DC (antigen uptake, T-cell stimulation) are<br />

strictly segregated to subsequent stages of differentiation<br />

(Figure 3). It is noteworthy that IL-10 212 and vascular<br />

endothelial growth factor (VEGF), secreted by cancer<br />

cells, 213 prevent the maturation of DC thus inhibiting the<br />

efficient priming of T-cells.<br />

Different strategies for the generation of<br />

DC ex vivo<br />

Circulating CD14 + monocytes represent the most readily<br />

available source of DC if incubated with appropriate<br />

cytokines such as GM-CSF, IL-4 and TNF-. 214, 215 Moreover,<br />

DC precursors have been isolated within the CD34 +<br />

cell fraction in bone marrow, cord blood and steady state<br />

or mobilized peripheral blood. 216-221 Also in this case the<br />

differentiation of CD34 + cells into fully functional DC is<br />

strictly dependent upon stimulation with certain<br />

cytokines such as GM-CSF, TNF-, SCF, FLT3-L and IL-4.<br />

Figure 2. Phenotypic and functional characteristics of dendritic cells. Modified from ref. #199 (Bancherau and Steinman,<br />

Nature, 1998).<br />

haematologica vol. 85(suppl. to n. 12):December 2000


134<br />

C. Bordignon et al.<br />

An extensive review of the different types of human DC<br />

and their ex vivo generation is beyond the scope of this<br />

chapter. However, in view of the clinical use of DC a few<br />

critical points should be stressed. GM-CSF and IL-4<br />

induce the differentiation of non-proliferating CD14 +<br />

monocytes to immature DC with a low level of expression<br />

of CD83 and p55 Ag and are largely incapable of priming<br />

naive T-cells. These immature DC are not fully differentiated<br />

and revert to an adherent state if the cytokines<br />

are removed from the culture medium. 222,223 The addition<br />

of inflammatory cytokines such as TNF-, IL-1 or PGE2<br />

for 1-2 days to the medium containing GM-CSF and IL-<br />

4 promotes the maturation of DC and increases the ability<br />

of stimulating T-cells. A potential bias toward the<br />

clinical use of this culture system is the requirement of<br />

fetal calf serum (FCS), a xenogenic protein that is contraindicated<br />

for human use. An innovative culture system<br />

for the generation of mature and functional DC from circulating<br />

monocytes that uses FCS-free conditions has<br />

recently been described. 222,223 In this system, adherent<br />

peripheral blood (PB) cells are cultured for 6-7 days with<br />

GM-CSF and IL-4 in the presence of FCS, which is then<br />

washed out, and subsequently exposed to macrophageconditioned<br />

medium (Mo-CM) and 1-5% autologous<br />

plasma for 1-3 days. Mo-CM is very efficient in inducing<br />

the terminal maturation of DC and is prepared by growing<br />

T-cell-depleted PB cells on immunoglobulin (Ig)-coated<br />

Petri dishes for 24 hours.<br />

Taken together, these findings lead to the conclusion<br />

that immature DC generated from CD14 + cells in the<br />

presence of GM-CSF and IL-4 are well equipped for capturing<br />

and processing soluble TAA. However, they do<br />

require a further maturation stimulus (Mo-CM, TNF-)<br />

to exert their stimulatory effect on T-cells. Immature DC<br />

are the ideal targets for genetic manipulation using viral<br />

or bacterial vectors which infect non-replicating cells<br />

(see below). In this case, the modified pathogens can<br />

induce by themselves the full maturation of DC. In alternative,<br />

mature DC could be used in vaccination protocols<br />

involving TA peptides as DC also prime T-cells to foreign<br />

Ag that bind directly to MHC molecules without prior<br />

processing. 224<br />

As reported above, CD34 + cells can be induced to differentiate<br />

into fully functional DC which resemble cutaneous<br />

Langherans cells. 218 The issue of the large scale<br />

production of DC from CD34 + precursors has been discussed<br />

in detail elsewhere. 5 However, very recently the<br />

phenotypic and functional characteristics of DC derived<br />

from CD34 + cells mobilized into PB or from BM progenitors<br />

have been formally compared. 225 The published<br />

results indicate that G-CSF mobilizes DC precursors (CFU-<br />

DC) with an increased frequency and a higher proliferative<br />

capacity than their BM counterparts. This finding<br />

translates into a higher number of mature DC generated<br />

in liquid culture. Despite pre-treatment with G-CSF, these<br />

cells maintain the same functional capacity of stimulating<br />

allogeneic T-cells as BM-derived DC. CD34 + cellderived<br />

DC are also capable of processing and presenting<br />

soluble Ag to autologous T-cells for both primary and<br />

secondary immune responses. The potential clinical usefulness<br />

of autologous serum in place of FCS 220 was also<br />

confirmed in the same study. Of note, IL-4 was shown to<br />

be capable of modulating DC differentiation from bipotent<br />

CD34 + cells during the later stages of the culture as<br />

previously demonstrated for monocyte-derived DC. 226<br />

Thus, mobilized CD34 + cells may represent the optimal<br />

source for the generation of DC for cancer immunotherapy<br />

rather than BM precursors. Very recent data indicate<br />

the mobilization of large numbers of DC precursors<br />

by GM-CSF 227 and FLT-3L. 228 However, it remains to be<br />

Figure 3. Functional proporties<br />

of dendritic cells at different<br />

stages of differentiation.<br />

Pathogens or inflammatory<br />

cytokines induce the<br />

maturation of dendritic cells<br />

which become activated<br />

upon interaction with T-cells<br />

via CD40-CD40L.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

135<br />

established whether circulating CD34 + elements are an<br />

equivalent source of DC to CD14 + monocytes. In this view,<br />

it has recently been demonstrated 229 that CD34 + cellderived<br />

DC are more efficient than monocyte-derived DC,<br />

from the same patients, in stimulating a specific CTL<br />

response to Melan-A/Mart-1 peptides.<br />

Delivery of TAA to DC<br />

Several methods for the efficient delivery of TAA to DC<br />

have been described so far (Figure 4). Their rationale is<br />

based on the finding that tumor cells are often poorly<br />

immunogenic due to the lack of T-cell recognition, activation<br />

and co-stimulation typical of professional APC. To<br />

this end, Gong et al. 230 fused murine DC with the carcinoma<br />

cell line MC38 to provide tumor cells with the<br />

functional characteristics of DC. The fusion cells showed<br />

all the phenotypic features of DC and were shown to be<br />

capable of preventing tumor growth when the mice<br />

were challenged with the cell line. Moreover, treatment<br />

with fusion cells induced the rejection of pulmonary<br />

metastates.<br />

Several TA peptides which are presented to T-cells in<br />

association with HLA class I molecules have been<br />

recently identified and proved to be useful in stimulating<br />

an autologous CTL response in vitro and in vivo.<br />

However, pulsing DC with peptides may not be optimal<br />

for clinical application because of the strict MHC restriction<br />

of the immune response and their limited stability.<br />

In addition, pulsing with peptides may not induce a T-<br />

cell response directed toward tumor cells expressing the<br />

relevant Ag. Although DC can be loaded with a cocktail<br />

of peptides from different Ag derived from the same<br />

type of cancer (see below), this vaccination approach is<br />

likely to limit patient selection on the basis of HLA phenotype.<br />

An attractive alternative is the use of unfractionated<br />

tumor-derived proteins, when available (see<br />

below), apoptotic cells 231 or tumor lysates. In the last<br />

case the obvious disadvantage is the possibility of inducing<br />

immune responses against self-Ag expressed in tissues<br />

other than tumor cells.<br />

A further possibility is the transduction of DC with<br />

expression vectors encoding for TAA genes (Figure 4). DC<br />

can be engineered by different means which differ in<br />

their capacity of targeting quiescent cells, stable integration<br />

in the genome, infection efficiency and stimulation<br />

of anti-tumor immunity (Figure 5). Retrovirallytransduced<br />

DC constitutively express the relevant<br />

sequence and are potent stimulators of a specific T-cell<br />

response. 232 However, retroviral vectors have a relatively<br />

low efficiency of transduction, they can only infect<br />

actively replicating cells and carry the theoretical risk of<br />

oncogenic transformation of target cells. Conversely, adenoviruses<br />

infect both quiescent and proliferating cells<br />

and do not integrate into DNA. 233 Moreover, supernatants<br />

with a high titer of the virus can be easily obtained.<br />

Recently, DC have been transduced with an adenovirus<br />

combined with cationic liposomes showing an infection<br />

efficiency close to 100%. 234 The major limitation to the<br />

clinical use of adenoviruses is their high immunogenicity<br />

which induces the production of neutralizing antibodies<br />

and the rapid development of CTL directed at infected<br />

cells.<br />

Vaccinia virus vectors are not oncogenic, do not integrate<br />

into genome and can be manipulated to carry<br />

large fragments of heterologous DNA. 235 However, these<br />

viruses are toxic for target cells and the viability of DC<br />

is approximately 50%. Nonetheless, antigen-specific<br />

inhibition of tumor growth has been observed in murine<br />

models using vaccinia vectors encoding for CEA and<br />

Mucin-1. 236,237 Two phase I clinical trials have been conducted<br />

to assess the safety of vaccinia virus vectors<br />

engineered to express HPV and CEA genes and to asses<br />

their capacity of stimulating an immune response. 238,239<br />

More recently, maturation of DC with neo-biosynthesis,<br />

translocation and stabilization of MHC molecules on<br />

the cell surface and efficient induction of both CD4 and<br />

CD8 T-cell activation has been induced by infection with<br />

bacterial vectors. 240 As a result, a model Ag (ovalbumin)<br />

expressed on the surface of recombinant Streptococco<br />

Figure 4.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


136<br />

C. Bordignon et al.<br />

Gordonii, is processed and presented on MHC class I<br />

molecules 10 6 times more efficiently than soluble OVA<br />

protein. Therefore, bacterial vectors are potentially useful<br />

means of delivering exogenous Ag to DC for stimulating<br />

a tumor-specific CTL response. A different<br />

approach has been taken by Boczkowsky et al. 241 who<br />

transfected DC with the total RNA extracted from tumor<br />

cells and combined it with cationic lipid to enhance the<br />

infection efficiency. Similarly to the use of tumor lysates,<br />

this strategy can be applied in those situations in which<br />

a tumor-specific antigenic marker is lacking; the major<br />

concern is the increased risk of autoimmune reactivity.<br />

DC for cellular immunotherapy<br />

The central role of DC in stimulating a tumor-specific<br />

immune response is well established in vitro and in<br />

vivo in animal models. 232,241-246 Whereas murine DC<br />

pulsed with TA-proteins or peptides or transduced with<br />

TAA genes have induced both the rejection of challenge<br />

tumor cells and the regression of established cancers, it<br />

remains to be determined which of the several strategies<br />

proposed for cellular immunotherapy is the most<br />

efficient. It may well be that different tumors require<br />

different approaches.<br />

In humans, initial studies were performed in patients<br />

with melanoma using DC pulsed with MAGE peptide.<br />

247,248 The infusion of loaded DC induced the migration<br />

of MAGE-specific CTL to the site of injection and<br />

increased the frequency of circulating tumor-specific<br />

CTL. More recently, Nestle et al. 249 have treated advance<br />

stage melanoma patients with intranodal injection of<br />

peptides or tumor lysates-pulsed DC according to the<br />

HLA profile of the patient. The authors reported the<br />

stimulation of a peptide-specific T-cell response in all<br />

cases. Moreover, in 5/16 patients an objective clinical<br />

response was observed. In this study, DC were generated<br />

ex vivo from monocyte precursors in the presence of<br />

IL-4 and GM-CSF and directly injected into an inguinal<br />

lymph node to reach T-cell rich areas.<br />

Tumor-specific peptides (fragments of prostate specific<br />

antigen, PSA) have also been used to pulse autologous<br />

DC in prostate cancer patients refractory to hormone-therapy.<br />

250 Seven out of 51 patients showed a partial<br />

response while none of the patients in the control<br />

group, injected with peptides alone, showed any clinical<br />

benefit. In B-cell malignancies, the patient-specific idiotype<br />

(Id) gene sequence and its protein product represent<br />

the optimal targets for vaccination strategies as previously<br />

shown in murine models 251,252 and humans. 253 Hsu<br />

et al. 254 have reported on the treatment of 4 patients<br />

with low-grade non-Hodgkin’s lymphoma (NHL), resistant<br />

to conventional chemotherapy or who had relapsed,<br />

with DC pulsed with the Id as soluble antigen. A tumorspecific<br />

T-cell-response was observed in all cases coupled,<br />

in one case, with the regression of tumor burden.<br />

At the time of writing, 16 patients have been treated<br />

and a tumor-specific cellular response has been found in<br />

8 individuals (R. Levy, personal communication). The<br />

same strategy of targeting the Id has been proposed by<br />

the same group for inducing a T-cell immune response<br />

in multiple myeloma patients. 255<br />

In contrast to the strategy used by Nestle et al. 249 in this<br />

preliminary trial DC were freshly isolated from the PB by<br />

subsequent enrichment steps and were reinfused intravenously.<br />

Although a much larger number of DC were<br />

injected in NHL patients compared to melanoma patients<br />

(3-2010 6 DC vs 110 6 ), this approach raises concerns<br />

about both the efficacy of uncultured PB DC of efficiently<br />

Figure 5.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

137<br />

stimulating T-cells and the capacity of Id-loaded APC to<br />

reach secondary lymphoid organs to prime T-cells, escaping<br />

the entrapment of the pulmonary apparatus.<br />

Future directions<br />

The few clinical data available so far have barely provided<br />

the proof of principle that autologous DC generated<br />

ex vivo and reinfused into cancer patients are<br />

effective in stimulating an anti-tumor immune response.<br />

This is the result of the complexity of the interplay<br />

between different cellular populations involved in tumor<br />

immunity. In addition, cellular immunotherapy with DC<br />

has yet to be standardized. As mentioned above, crucial<br />

issues such as 1) the choice of the most suitable TAA to<br />

stimulate an immune response; 2) the use of soluble<br />

proteins/peptides or DC engineered with expression vectors;<br />

3) the optimal source for the generation of DC and<br />

the number of APC needed to promote a clinical effect;<br />

and 4) the most effective route of administration of DC,<br />

are points which still need to be solved. At this stage,<br />

relying for the most part on animal studies, we can only<br />

conclude that DC-based immunotherapy holds promises<br />

of exerting a potent anti-tumor effect in humans.<br />

Oral vaccination by in vivo targeting of DC<br />

A simple approach to targeting APC in vivo is to use<br />

attenuated bacterial vectors, such as those commonly<br />

developed to control infectious diseases. They usually<br />

enter the host through the oral route and selectively<br />

replicate within macrophages and DC. Listeria monocytogenes<br />

is a promising vaccine carrier that naturally<br />

infects APC, and may deliver immunogens to both MHC-<br />

I and II pathways of antigen processing and presentation.<br />

256 Furthermore, this bacterium may constitute per<br />

se an excellent danger signal for the immune system,<br />

since it stimulates the innate immune response to produce<br />

cytokines (e.g. IL-12) and mediators (e.g. nitric<br />

oxide) that enhance antigen presentation. In addition, it<br />

promotes a TH1-type cellular response, which is mainly<br />

associated with the eradication of tumors and intracellular<br />

parasites. Most of these features are also shared<br />

by Salmonella typhimurium-based carriers.<br />

The ideal vaccine carrier should maintain its immunogenicity<br />

intact, being attenuated enough to allow its<br />

use in humans. However, the safety profile of a vaccine<br />

destined for human use also requires the absolute stability<br />

of the mutant phenotype, which can only be guaranteed<br />

by the generation of chromosomal deletion<br />

mutants. Furthermore, the release of recombinant microorganisms<br />

under uncontrolled conditions makes the lack<br />

of antibiotic resistance markers essential. Mutation of<br />

genes involved in bacterial spread and survival are the<br />

best targets for attenuation.<br />

The recent progress in Listeria and Salmonella genetic<br />

manipulation and the availability of suitable in vitro<br />

and in vivo models, make these micro-organisms very<br />

attractive vaccine delivery systems.<br />

For example, attenuated Listeria monocytogenes carrier<br />

strains expressing the -galactosidase (-gal) model<br />

antigen can prevent outgrowth of an experimental<br />

tumor in BALB/c mice by inducing a specific immune<br />

response against the -gal TAA. 257 Similarly, a live attenuated<br />

AroA- auxotrophic mutant of Salmonella typhimurium<br />

(SL7207) has been used as a carrier for the<br />

pCMV vector that contains the -gal gene under the<br />

control of the immediate early promoter of cytomegalovirus<br />

(CMV). After a primary immunization and three<br />

orally administered boosts at 15-day intervals, a Salmonella-based<br />

vaccine induced both cell-mediated and<br />

systemic humoral responses to -gal. These experiments<br />

suggested that insertion of a plasmid containing an<br />

expression cassette into a Salmonella-carrier allowed<br />

DNA immunization and specific targeting of antigen<br />

expression to APC, in vivo, through oral immunization.<br />

To prove that the transgene was actually expressed by<br />

APC cells as a function of a eukaryotic promoter the<br />

green fluorescent protein (GFP) was placed under the<br />

control of either the eukaryotic CMV or a prokaryotic<br />

promoter and spleen cells from treated mice were analyzed<br />

by cytofluorometric analysis.<br />

GFP was detectable in both macrophages and DC, but<br />

not in other splenocytes, of mice treated with Salmonella<br />

containing the CMV-plasmid, 28 days after the<br />

first vaccine administration, whereas it was undetectable<br />

in spleen cells of mice receiving the Salmonella<br />

containing the constitutive prokaryotic promoter<br />

which directs GFP synthesis only within the carrier. 258<br />

GFP expression in DC highlights the possibility of loading<br />

DC without the need for ex vivo manipulations and<br />

opens up the possibility of administering a cancer vaccine<br />

orally. Oral vaccination is viewed as an easier and<br />

more acceptable strategy for patients especially in a<br />

phase in which they are disease-free.<br />

Leukemic cells as antigen presenting<br />

cells<br />

Tumors may escape immune detection and killing<br />

through a variety of mechanisms affecting the capacity<br />

of either presenting tumor antigens or fully activating T-<br />

cells. 258,260 In particular, tumor cells are likely to prevent<br />

a clinically evident cytotoxic T-cell response because of<br />

the absence of a specific antigenic tumor peptide, or<br />

because they lack HLA molecules, or co-stimulatory molecules<br />

on their surface. In this last case the patient’s T-<br />

cells might become anergic and tolerate tumor cells.<br />

Alternatively, neoplastic antigens may induce a clonal<br />

deletion of thymocytes, 261 or tumor cells expressing Fas<br />

molecule may be responsible for an apoptotic T-cell deletion<br />

through Fas:FasL interaction. 262 So far, different<br />

immunologic strategies aimed at overcoming these<br />

defects by inducing or improving the antigen presenting<br />

function of tumor cells have been demonstrated in experimental<br />

models, 263,264 and the hypothesis that leukemic<br />

cells may become efficient APC by changing their phenotype<br />

or by differentiating into DC-like cells has been<br />

tested. A first example was shown in B-cell neoplasms<br />

since it is well known that normal B-cells may present<br />

antigen to T-cells 265 and that cognate interactions<br />

between B- and T-cells may induce either a T-cell prolif-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


138<br />

C. Bordignon et al.<br />

eration and an enhanced T-helper activity to cytotoxic T-<br />

cells, 266 or T-cell clonal unresponsiveness. 267 The triggering<br />

of the CD40 receptor on the surface of APC increases<br />

the expression of adhesion and co-stimulatory molecules<br />

both in vitro and in vivo. 269,269 Thus, the possibility<br />

of modifying the phenotype and the APC function of<br />

CD40 + -chronic lymphocytic leukemia B (CLL-B) cells<br />

through the CD40:CD40L interaction was demonstrated<br />

showing that this pathway induces the upregulation of<br />

CD80 and CD86 on CLL-B cells and the triggering of a T-<br />

cell proliferative response. 270,271 These results support the<br />

idea that induction of B7 molecules on CLL-B cells, either<br />

by T-cell-contact and growth factors, 270, 272 or by gene<br />

transfer methods 273 may be a potential clinical vaccinetherapy<br />

capable of eliciting efficient anti-leukemic<br />

immune responses. Similar approaches may also apply to<br />

B non-Hodgkin’s lymphomas (B-NHL). Studies in experimental<br />

models indicated that CD40 stimulation may<br />

result in the inhibition of lymphoma cell growth in vivo, 274<br />

and in the up-regulation of adhesion receptors and costimulatory<br />

molecules on lymphoma cells in vitro. 275,276<br />

Interestingly, follicular B-NHL cells which express CD40<br />

and low levels of B7-2 fail to present alloantigen, but<br />

after activation via CD40 they express higher levels of B7-<br />

1 and LFA-3 and alloreactive T-cells respond to tumor<br />

cells efficiently. 276 Finally, encouraging results have also<br />

been obtained in pre-B acute lymphoblastic leukemia 139<br />

in which approximately 50% of the cases blast cells have<br />

been reported to express CD86 but not to induce tumor<br />

rejection, and B7-blasts determine an immunologic tolerance<br />

of the tumor. Nonetheless, this study showed that<br />

pre-activation of blast cells via CD40, or cross-linking<br />

CD28, or signaling through the common chain of the<br />

IL-2 receptor on T-cells can prevent T-cell tolerance. The<br />

authors hypothesize at least two possible mechanisms to<br />

explain the induction of lymphocyte unresponsiveness:<br />

first, they propose that at the time of initial transformation,<br />

clonogenic pre-B acute leukemia cells may not<br />

express CD86 thus inducing a T-cell anergy that could not<br />

be reversed by following expression of CD86 on a blast<br />

cell fraction; second, they suggest that marrow microenviroment<br />

may play a role in modulating T-cell immunity<br />

by secreting negative regulators, as previously shown in<br />

experimental models. 277,278 However, after co-stimulation<br />

by either B7 transfectants or professional APC, autologous<br />

antileukemic cytotoxic marrow T cells can be generated<br />

upon contact with CD40-stimulated pre-B acute<br />

leukemia cells. 140<br />

All these data on B-cell neoplasms strongly suggest<br />

that poor tumor immunogenicity may depend on both<br />

the quality and the quantity of accessory molecules<br />

required for T-cell stimulation. However, future therapeutic<br />

strategies aimed at stimulating the CD40 receptor,<br />

or at directly transducing B7 molecules on chronic or<br />

acute leukemia B-cells will facilitate the ex vivo expansion<br />

of specific anti-tumor cytototoxic T-cells. Normal<br />

myeloid CD34 + progenitors include a small subset of<br />

APC279, 280<br />

that are committed precursors of the<br />

macrophage/dendritic lineage. 281 In fact, both marrow<br />

and peripheral blood CD34 + cells, and circulating monocytes<br />

can be utilized to obtain large numbers of dendritic<br />

cells in vitro. Due to the relevance of co-stimulatory<br />

molecules on tumor cells for the generation of anti-tumor<br />

immune responses, the hypothesis of whether even acute<br />

or chronic myelogenous leukemic cells might differentiate<br />

into dendritic cells in vitro and become immunogenic<br />

has been addressed by several groups. Alternatively,<br />

transduction of co-stimulatory molecules on leukemic<br />

myeloblasts has been attempted in experimental models<br />

to generate specific cytotoxic responses. Both these<br />

approaches require that TAA are expressed and exposed<br />

on HLA molecules, and it is likely that genetic alterations,<br />

such as chromosomic translocations, might result in the<br />

appearance of pathologic peptides, specific for each<br />

acute or chronic leukemia and potentially immunogenic.<br />

Chronic myelogenous leukemia may represent an optimal<br />

candidate for antitumor vaccine strategies since several<br />

reports have shown that the bcr-abl fusion protein can<br />

bind to defined HLA class I and class II molecules 282-286<br />

and also that dendritic cells generated in vitro from CML<br />

patients still carry the t(9;22). 287,288 In this latter study, in<br />

fact, CML cells that were incubated with GM-CSF, IL-4<br />

and TNF- developed DC phenotypic and functional characteristics<br />

inducing autologous cytotoxic T-cells capable<br />

of directly lysing leukemic cells and of inhibiting CML<br />

colony growth in vitro. Further studies suggested that<br />

CML DC-stimulated anti-leukemic T-cell reactivity is due<br />

to an oligoclonal T-cell response and develops in an HLArestricted<br />

manner. 289 Dendritic cells can be generated<br />

even from CD34 + CML marrow progenitors in the presence<br />

of GM-CSF, TNF- and IL-4, and after 7-10 days of<br />

culture they are Ph + , express high levels of HLA molecules<br />

and co-stimulatory receptors and induce a T-cell<br />

proliferation 10-30 fold higher than unprocessed marrow<br />

cells. 290 Nonetheless, it is likely that different culture systems<br />

may be required for efficient in vitro generation of<br />

DC when using CML-CD34 + cells rather than normal<br />

progenitors, since the former show a lower DC clonogenic<br />

activity but both their expansion and their differentiation<br />

can be significantly improved by prolonging the<br />

duration of culture in the presence of specific growth<br />

factors. 291<br />

When a neoplastic event affects undifferentiated or<br />

more mature progenitors of the granulocytic and/or<br />

macrophage lineage an AML develops, and we can distinguish<br />

different subtypes of AML on the basis of morphologic<br />

and phenotypic characteristics. The identification<br />

of AML cells with some phenotypic affinities to DC,<br />

such as the expression of the CD1a marker, 292 or deriving<br />

from a monocytic/dendritic cell progenitor, 293 has been<br />

attempted in the past. Indeed in this latter study, cells<br />

from an AML, FAB M2 patient were shown to differentiate<br />

into terminal DC with potent alloantigen presenting<br />

capacity after in vitro culture with GM-CSF, TNF-, SCF<br />

and IL-6. Similar results were achieved by culturing freshly<br />

isolated AML cells with GM-CSF, IL-4 and IL-13 for 7<br />

days. 294 Alternatively, restoration of anti-tumor immune<br />

control can be attempted by identifying peptides, such as<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

139<br />

PR-1 derived from proteinase 3, 135 that could be capable<br />

of inducing HLA-restricted cytotoxic T-lymphocytes to<br />

lyse fresh leukemic cells, or by engineeing leukemic cells<br />

to induce either the expression of co-stimulatory molecules<br />

or the production of cytokines. The role of B7-1 in<br />

developing protective immunity was initially tested in a<br />

mouse model in which the injection of a myeloid cell line<br />

transfected with the bcr/abl gene was rapidly lethal,<br />

while prolonged survival was observed only in mice that<br />

received the cell line co-transfected with the B7-1<br />

gene. 295 Moreover, the same model was used to test the<br />

role of both B7-1 and B7-2, suggesting that B7-1 may be<br />

more effective than B7-2 in obtaining an efficient in vivo<br />

anti-leukemic response. 296 The potential advantage of B7-<br />

transduced blasts was confirmed by using primary AML<br />

cells instead of a cell line; a CD8 + T-cell dependent and<br />

B7:CD28-mediated anti-leukemia activity was documented.<br />

297 A recent study compared the in vitro immunogenic<br />

activity of human AML cells cultured with GM-CSF,<br />

IL-4 and TNF-, or transfected with CD80. 298 Both these<br />

approaches resulted in an enhanced T-cell response in a<br />

mismatched primary MLR, however, only B7-1 transduced<br />

AML cells stimulated a strong immune response of<br />

T-cells from an HLA identical bone marrow donor, and<br />

generated leukemia reactive CD4 + T-cell lines and clones.<br />

Interestingly, this model allowed the authors to observe<br />

CD80 + AML-mediated T-cell responses that can be directed<br />

against the patient’s minor histocompatibility antigens<br />

or tumor-specific antigens.<br />

Although B7-1 and B7-2-engineered tumor cells<br />

could play a pivotal role in anti-leukemia immunotherapy<br />

strategies, there is evidence that transduction<br />

of other receptors 299 or cytokines 300-303 might, at least,<br />

co-operate with B7 molecules in the antigen presenting<br />

capacity of neoplastic cells.<br />

Genetically modified cells as vaccine<br />

for the active immunotherapy of cancer<br />

Non-specific approaches to cancer immunotherapy<br />

probably date back to the beginning of the 18 th century<br />

and originated from the observation of sporadic,<br />

spontaneous remission of tumors in patients who suffered<br />

severe bacterial infection. This observation<br />

prompted Dr. William B. Coley to begin, in 1891, to treat<br />

patients with soft tissue sarcoma with a mixture of<br />

Gram positive and negative bacteria: Coley’s toxins.<br />

This empirical approach was enforced by Shear’s discovery<br />

that endotoxins were active components responsible<br />

for tumor hemorrhagic necrosis. Furthermore, the<br />

finding that bacillus Calmette-Guérin (BCG) increased<br />

resistance to tumor transplants in mice led to clinical<br />

application of BCG which, together with Streptococcusderived<br />

OK-432, is a strategy used to this day.<br />

The anti-tumor effects obtained by treatment with BCG<br />

and derivatives are largely dependent on indiscriminate<br />

necrosis of tissues containing mycobacterium (the Koch<br />

phenomenon). The discovery of cytokines explained most of<br />

the phenomena induced by microbial products and<br />

cytokines were then used with the initial hope of copying<br />

the positive effects of such bacterial products while avoid-<br />

Known, limited<br />

repertoire of TAA<br />

Large repertoire of<br />

unknown antigens<br />

Gene<br />

Gene, protein/peptide<br />

Cell fragments RNA<br />

Whole tumor cells<br />

Loading DC<br />

Ex vivo<br />

Directly in vivo<br />

DNA vaccination<br />

Needs adjuvant:<br />

plasmid sequences<br />

muscle injury<br />

Bacterial vectors<br />

Active immunization for cancer:<br />

strategies are based on whether tumor antigens<br />

have been molecularly defined.<br />

Figure 6.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


140<br />

C. Bordignon et al.<br />

ing the negative ones.<br />

More recently, the discovery of Th1 and Th2 distinct<br />

pathways of T-cell maturation helped to explain protective<br />

and non-protective BCG-induced cell-mediated<br />

immune reactions in tuberculosis, phenomena that have<br />

correlates with protection against cancer. In the presence<br />

of a Th1 deflected immune response, the effect of<br />

TNF- is not that of large necrosis which is, rather, the<br />

characteristic of inflamed tissues of a Th2 type of<br />

response, in this case extremely sensitive to TNF-. 304<br />

Cytokines deflecting the immune response to a Th1 or<br />

Th2 type of response may drive the type of immune<br />

response to cancer cells, escaping the simple definition of<br />

Th1 promoting and Th2 inhibiting anti-tumor immunity.<br />

Rather, a strong Th1 as well as a strong Th2 response<br />

may induce tumor destruction and immune memory with<br />

the same efficacy although through different mechanisms<br />

(see below). Moreover, genetic background may<br />

influence the ability to mount a Th1 or Th2 response, as<br />

shown in murine models.<br />

Microbial products have mainly local effects which<br />

may be reproduced and improved by local injection of<br />

recombinant cytokines. Experiments in non-tumor systems<br />

have shown that IL-2 offsets antigen recognition<br />

and overcomes tolerance. Thus cytokines could be used<br />

not only to stimulate tumor destruction but also to<br />

impair tolerance and activate effective and specific<br />

immune recognition of TAA.<br />

Identification and cloning of the long elusive TAA,<br />

especially from human melanomas, 305 pointed tumor<br />

immunotherapy to a general systemic response and, thus,<br />

the use of cytokines shifted from that of being responsible<br />

for local tumor debulking to that of being an aid to<br />

triggering and boosting the immune response to TAA.<br />

In addition to antigens triggering the T-cell-receptor<br />

(TCR) of T-lymphocytes, optimal T-cell response also<br />

requires co-stimulatory molecules, as detailed above.<br />

Cytokines, co-stimulatory molecules and several<br />

cloned TAA are now available: how can we use them to<br />

provide an effective immunotherapeutic approach to<br />

cancer patients?<br />

Two major strategies are envisaged (see Figure 6): one,<br />

already described, takes advantage of antigen availability<br />

in the forms of genes, proteins or peptides and of<br />

the standardized methods of obtaining DC from peripheral<br />

blood in large quantities to be loaded with the antigen<br />

and reinfused in vivo; the other strategy still considers<br />

the tumor cells representative of the entire antigenic<br />

repertoire of a certain neoplasia; such cells, genetically<br />

modified to produce cytokines and/or co-stimulatory<br />

genes, could be injected into patients as a cellular<br />

vaccine. In the latter case a pool of cell lines derived<br />

from different patients with the same type of tumor<br />

could increase the antigenic repertoire and avoid<br />

immunoselection that certain antigens may have<br />

encountered in some patients. Unmatched MHA are not<br />

a problem in terms of antigen presentation since injected<br />

cells are destroyed and represented by host APC.<br />

Moreover if different sets of alloantigens are selected<br />

from different pools, the risk of repeated alloimmunization<br />

during booster vaccination would probably be<br />

avoided. The background and prospectives of genetically<br />

modified tumor cell vaccines are presented below.<br />

Cytokines at the tumor site<br />

In initial studies recombinant cytokines were injected<br />

at the tumor site or cytokine genes were inserted<br />

into somatic cells to be injected at the tumor site. All<br />

these studies collectively established that most of the<br />

cytokines accumulated at the tumor site were able to<br />

induce tumor destruction and the reaction they induced<br />

was sometimes strong enough to eradicate a tumor<br />

antigenically unrelated to the cytokine-releasing cells.<br />

The obtained tumor debulking was often followed by a<br />

systemic tumor-specific immune memory. It should be<br />

underlined, however, that tumor debulking may occur<br />

through non-specific immune reactions or so fast as to<br />

prevent efficient T-cell priming, this being reminiscent<br />

of the dichotomy described for BCG: indiscriminate<br />

necrosis versus protective immunity.<br />

Engineered tumor cell vaccines<br />

Engineering of tumor cells with the gene of a particular<br />

cytokine is an efficient way of ensuring that this<br />

cytokine will be durably present at the tumor site.<br />

Repeated local injections would, of course, have the<br />

same effect. Bolus administration, however, does not<br />

provide a constant supply of cytokine. Its effects are<br />

much less evident than those achieved by the injection<br />

of engineered tumor cells 306 that can ensure the provision<br />

of antigen and continued local accumulation of the<br />

cytokine until a physiologic or a pharmacologic threshold<br />

is reached, and the biological activity of the cytokine<br />

can begin.<br />

The immunogenicity that tumor cells can acquire<br />

upon cytokine-gene transduction may stem from<br />

recruitment by released cytokines, of particular repertoires<br />

of inflammatory cells, whose differing abilities to<br />

influence TAA presentation and secrete secondary<br />

cytokines may shape both immunogenicity and deflection<br />

of the ensuing immune memory towards a Th1 or<br />

Th2 type of response. A cytokine may be simultaneously<br />

involved in tumor rejection, leukocyte recruitment<br />

and activation of memory mechanisms.<br />

Many experimental studies have been performed in<br />

mice over the last seven years and cytokine genes from<br />

IL-1 to IL-18 have been tested. Most of those studies<br />

described whether a certain cytokine gene, upon transduction,<br />

can inhibit tumor growth in vivo; some also<br />

described whether the cytokine induced protective<br />

immunization against challenge by parental cells whereas<br />

only a few studies described efficacy in a therapeutic<br />

setting. It is clear that the way cytokines modify<br />

tumor oncogenicity, immunogenicity and curative effect<br />

is not only dependent on the cytokine employed but also<br />

on the tumor model utilized. The immune mechanisms<br />

responsible for inhibition of tumor growth may not be<br />

the same as those required for immune memory or those<br />

necessary for eradication of an established tumor.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

141<br />

Translation of animal studies into a clinical setting<br />

faces a substantial difference, that is the fast growth of<br />

transplanted tumors and therefore the short time window<br />

in which immunization can be performed before<br />

the animal’s death. In murine models, the so-called<br />

established tumor is a tumor that has been injected one<br />

to three days before the beginning of vaccination. This<br />

contrasts with phase I/II clinical studies in which enrolled<br />

patients have advanced disease. Clear evidence of therapeutic<br />

effects is not expected in these patients, therefore<br />

tumors with antigens whose genes have been cloned<br />

and are recognized by CTL should be used to allow, at<br />

least, an immunologic follow-up that could prove the<br />

effect of vaccination. This confines the choice to those<br />

carrying the MAGE, GAGE and BAGE family genes and to<br />

melanomas, which also express antigens of the melanocyte<br />

lineage, such as tyrosinase, gp100 and MART-<br />

1/Melan-A. 305 The choice is further restricted by the difficulty<br />

of obtaining cells and cell lines from tumors that<br />

are not melanomas to be transduced and then employed<br />

for immunologic evaluation. Melanoma is thus the tumor<br />

most frequently chosen for vaccination studies.<br />

Nevertheless, vaccination with cytokine-transduced,<br />

freshly isolated cells, which should retain the tumorantigen<br />

repertoire, could be a way of generating tumorspecific<br />

T-lymphocyte lines and clones with which to<br />

identify antigens expressed by tumors other than<br />

melanomas.<br />

In a few cases only, the antigens associated with the<br />

murine tumors employed in pre-clinical studies were<br />

characterized; the majority of studies designed to discover<br />

the immunologic mechanisms associated with<br />

tumor rejection utilized proteins not classifiable as<br />

tumor-associated antigens, such as -galactosidase 244<br />

and influenza nucleoprotein. 307 Most of these animal<br />

studies were carried out in the syngeneic system, that<br />

in humans corresponds to the autologous situation, in<br />

which a tumor cell line was both the cell vaccine and the<br />

tumor to be cured. Autologous application is actually<br />

difficult, since it requires tumor cell cultures from every<br />

patient for both gene transduction and immunologic<br />

follow-up. Each patient’s cell vaccine should then be<br />

checked for safety, and a great variability in terms of<br />

cytokine production other than adhesion molecules and<br />

antigenic phenotypes may exist between cell vaccines.<br />

The use of allogeneic cell lines, on the other hand, has<br />

the advantage of employing vaccines well-characterized<br />

in terms of tumor antigen, MHC and adhesion molecules,<br />

as well as the constant amount of cytokine<br />

released; these parameters in combination may provide<br />

a standard reagent for clinical studies.<br />

Both syngeneic and allogeneic tumor cells expressing<br />

a common TAA are processed by host APC such that TAA<br />

derived peptides are presented in association with host<br />

MHC in either case. 307 Nevertheless, in most clinical protocols<br />

the expression of the MHC class I allele, which<br />

presents TAA derived peptide(s), on the immunizing<br />

tumor cells is preferred. If cross-priming occurs efficiently,<br />

this should not be necessary, but it is still unclear<br />

whether vaccination with transduced tumor cells actually<br />

primes the host or boosts already present activated<br />

T-lymphocytes. This observation indicates that co-stimulatory<br />

molecules, such as B7, in addition to cytokines<br />

may be transduced in cell vaccines in order to amplify<br />

the boosting effect, since is not clear whether B7 transduced<br />

cells prime the host directly.<br />

Clinical vaccination protocols using IL-2 or IL-4 genetransduced<br />

allogeneic melanoma cells have been performed<br />

at the Istituto Nazionale Tumori in Milan, Italy.<br />

An HLA-A2 melanoma cell line expressing Melan-<br />

A/MART-1, tyrosinase, gp100 and MAGE-3 has been<br />

transduced and irradiated before the treatment of<br />

advanced HLA-A2+ melanoma patients. 308 In the first<br />

protocol, patients were injected subcutaneously on days<br />

1, 13, and 26 with IL-2 gene-transduced and irradiated<br />

melanoma cells at doses of 5 (3 patients) and 15 (4<br />

patients) 10 7 cells. Mixed lymphocyte-tumor cultures<br />

(MLTC) and limiting dilution analyses were performed to<br />

compare pre- and post-vaccination PBL. While MLTC<br />

revealed an increased but MHC-unrestricted cytotoxicity,<br />

in two cases the frequencies of melanoma-specific<br />

CTL precursors were clearly augmented by vaccination.<br />

In one patient, HLA class II-restricted effectors were<br />

found to be involved in the recognition of autologous<br />

tumor. Which antigen(s) was involved in the recognition<br />

by PBL of vaccinated patients remains unclear. In 3 out<br />

of 5 cases studied, pre- and post-vaccination PBL could<br />

not recognize any melanoma peptide tested or known to<br />

be restricted by HLA-A2 allele. 308 Among other possible<br />

explanations, this might be due to a tumor associated<br />

antigenic repertoire that exceeds the limited number of<br />

antigens whose genes have been cloned so far.<br />

This indicates that vaccination with cell lines is advantageous<br />

because the cell lines stimulate the host with<br />

the entire repertoire of known and unknown antigens. In<br />

the allogeneic system it is then easy to rotate the transduced<br />

cell line within the protocols and so maximize the<br />

chances that a relevant tumor antigen is present in the<br />

vaccine. Some antigens, in fact, may be negatively<br />

selected and lost in one patient-derived line, but not in<br />

others. In addition, selection of allogeneic cell lines displaying<br />

various MHC reduces the interference of repeated<br />

boosting with strong alloantigens. Indeed vaccination<br />

with a pool of three melanoma cell lines commenced<br />

before the cloning of known melanoma associated antigens,<br />

resulted in increased survival correlated with the<br />

level of antibody against the GM2 ganglioside, indicating<br />

possible involvement of a humoral response; correlation<br />

with the CTL response was not investigated. 309<br />

Going back to animal studies in which vaccination<br />

therapy with cytokine-transduced tumor cells was successful,<br />

it should be underlined that it was not clear<br />

which of the measured immune responses was responsible<br />

for the therapeutic effect since, generally, induction<br />

of cytotoxic T-lymphocytes was, per se, insufficient<br />

to produce a cure. In keeping with this statement, vaccination<br />

of 13 evaluable patients with MAGE-3.A1 peptide<br />

resulted in 3 clinical regressions, although no CTL<br />

haematologica vol. 85(suppl. to n. 12):December 2000


142<br />

C. Bordignon et al.<br />

precursors were found in the PBL of these responders. 310<br />

Refined animal studies performed to identify which<br />

immune responses correlate with the therapeutic activity<br />

indicated that both T- and B-cells should be properly<br />

activated.<br />

311, 312<br />

These observations may suggest that while a patient<br />

could be immunized against a tumor, the immunity thus<br />

induced might be insufficient to fight the established<br />

tumor growing within its own stroma. The combination<br />

of poor immune function and large tumor burden makes<br />

patients with advanced disease dubious predictors of<br />

clinical response.<br />

The general idea is that cytokine engineered tumor<br />

cells should be used as vaccines in minimal disease settings.<br />

313 A new form of treatment would thus be available<br />

for combination with conventional management<br />

of patients after surgical removal of their tumor,<br />

patients with minimal residual disease, or patients<br />

expected to manifest tumor recurrence after a significant<br />

apparently disease-free interval. When compared<br />

with conventional forms of management, vaccination is<br />

a soft, non-invasive treatment, unlikely to cause particular<br />

distress or side-effects, and could be administered<br />

after resection of a primary tumor when recurrence is<br />

expected.<br />

Use of mesenchymal cells for treatment of<br />

neoplastic and non-neoplastic disorders<br />

In addition to hematopoietic stem cells which can differentiate<br />

to produce progenitors committed to terminal<br />

maturation, 314 human bone marrow also contains<br />

stem cells of non-hematopoietic tissues which are currently<br />

referred to as mesenchymal stem cells (MSC),<br />

because of their ability to differentiate into cells that<br />

can roughly be defined as mesenchymal, or as marrow<br />

stromal cells because they appear to arise from the complex<br />

array of supporting structures found in marrow. 315<br />

Stromal cells of the marrow microenvironment include<br />

fibroblasts, endothelial cells, reticular cells, adipocytes,<br />

osteoblasts and macrophages, the last, although of<br />

hematopoietic origin, being considered functional components<br />

of the regulatory stroma. 316 The heterogeneous<br />

populations of mesenchymal cells and their associated<br />

biosynthetic products have the unique capacity to regulate<br />

hematopoiesis. 317<br />

Environmental components can modify the proliferative<br />

and differentiative behavior of hematopoietic cells by<br />

means of (i) cell-to-cell interactions, (ii) interactions of<br />

cells with extracellular matrix molecules, and (iii) interactions<br />

of cells with soluble growth regulatory molecules.<br />

316 All these regulatory modalities participate in<br />

stromal cell-mediated regulation of hematopoiesis. In<br />

fact, marrow stromal cells provide the physical framework<br />

within which hematopoiesis occurs, play a role in directing<br />

the processes by synthesizing, sequestering or presenting<br />

growth-stimulatory and growth-inhibitory factors,<br />

and also produce numerous extracellular matrix proteins<br />

and express a broad repertoire of adhesion molecules<br />

that serve to mediate specific interactions with<br />

hematopoietic stem/progenitor cells of both myeloid and<br />

lymphoid origin. 318, 319 Although growth factors play key<br />

roles in stem/progenitor cell proliferation and differentiation<br />

it seems improbable that hematopoiesis is regulated<br />

only by a random mix of growth factors and responsive<br />

cells. Rather, it is likely that regulatory molecules<br />

and localization phenomena within marrow stroma are<br />

required to sustain and regulate the function of the<br />

hematopoietic system. 320<br />

Although it is commonly accepted that stem cells are<br />

capable of homing to the marrow and docking at specific<br />

sites, the exact role of microenvironmental cells, adhesion<br />

molecules and extracellular matrix molecules in regulating<br />

the localization and spatial organization of<br />

hematopoietic stem cells in the marrow and driving<br />

myeloid and lymphoid regeneration following stem cell<br />

transplantation remains a matter of hypothesis. 321 Studies<br />

in animals demonstrated that stem and progenitor<br />

cells have different distributions across the femoral marrow<br />

cavity of mice, thus suggesting that marrow stroma<br />

is organized into functionally discrete environments, such<br />

as primary microenvironmental and secondary microenvironmental<br />

areas, allowing distinct differentiation patterns<br />

of hematopoietic stem cells. 322 The stem cell niche<br />

hypothesis, proposed by Schofield 323 suggested that certain<br />

microenvironmental cells of the marrow stroma<br />

could maintain the stem cells in a primitive, quiescent<br />

state. Another mechanism supporting the concept of specialized<br />

microenvironmental areas is stroma-mediated,<br />

compartimentalized growth factor production. Growth<br />

factor produced locally by stromal cells may bind to the<br />

extracellular matrix and be presented to immobilized target<br />

cells which recognize each growth factor through<br />

specific receptors. 320 This mechanism may provide the<br />

opportunity for localizing distinct growth factors at relatively<br />

high concentrations to discrete sites. As yet, relatively<br />

little is known of the nature of the factor(s) produced<br />

by different stromal cell types which modulate lineage<br />

development. However, a growing body of evidence<br />

suggests that marrow stroma is involved not only in regulating<br />

myeloid cell growth, but also in T- and B-cell lymphopoietic<br />

development. 324-327 Distinct adhesion molecules<br />

and cytokines are known to regulate stroma-dependent<br />

T- and B-lymphopoiesis, 328, 329 suggesting that marrow<br />

stroma may function as a site of T- as well as B-cell<br />

lymphopoiesis.<br />

The existence of self-renewing MSC is supported by<br />

several in vitro and in vivo data. 330 At the functional level,<br />

MSC residing within marrow microenvironment, establish<br />

marrow stroma both in vitro and in vivo and have<br />

multilineage differentiation capacity, being capable of<br />

generating progenitors with restricted development<br />

potential which include fibroblast, osteoblast, adipocyte,<br />

chondrocyte and myoblast progenitors (Figure 7). 331-333<br />

Putative stromal cell progenitors have been identified in<br />

human marrow by their ability to generate colonies of<br />

fibroblast-like cells originating from single clonogenic<br />

progenitors termed fibroblast colony-forming units (CFU-<br />

F). 334 These progenitors, which belong to the osteogenic<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

143<br />

Figure 7.<br />

stromal lineage, play a central role in establishing the<br />

marrow microenvironment both in vitro and in vivo. 335-337<br />

Under appropriate culture conditions and supplementation<br />

with specific stimuli, a proportion of marrow CFU-F<br />

can be induced to either adipogenesis 333 or osteoblastogenesis.<br />

338 Studies involving ectopic transplantation of<br />

individual fibroblastic clones grown in vitro from mouse<br />

marrow beneath the renal capsule of syngeneic hosts<br />

demonstrated that approximately 15% produced a marrow<br />

organ containing the full spectrum of stromal cell<br />

types of hematopoietic microenvironment, thus suggesting<br />

that CFU-F have multilineage differentiation capacity<br />

and supporting the stromal stem cell hypothesis. 339<br />

Based on these findings, CFU-F can be identified as multipotent<br />

stromal progenitors rather than lineage-restricted<br />

fibroblast progenitors.<br />

CFU-F can be enriched from adult bone marrow by<br />

means of the STRO-1 monoclonal antibody that identifies<br />

essentially all assayable marrow CFU-F. 340 STRO-1 +<br />

cells do not express the CD34 antigen and fail to generate<br />

hematopoietic progenitors, thus facilitating a clean<br />

separation between hematopoietic and stromal progenitors.<br />

341 Flow-sorted STRO-1+ cells grown under longterm<br />

culture conditions generate adherent stromal layers<br />

consisting of fibroblasts, osteoblasts, smooth muscle<br />

cells and adipocytes. 340 These stromal layers are capable<br />

of supporting hematopoiesis in long-term cultures initiated<br />

with CD34 + cells. In addition to STRO-1, other monoclonal<br />

antibodies, such as SH-2, have been described<br />

which specifically detect mesenchymal progenitors. 342<br />

In vivo data generated in animal models support the<br />

functional regulatory role of the marrow microenvironment.<br />

In the fetal sheep model of in utero stem cell<br />

transplantation, co-transplantation of stem cells with<br />

marrow stromal cells has been shown to improve levels<br />

of donor cell engraftment. 343 In the NOD/SCID mouse<br />

model of in utero stem cell transplantation, fetal stem<br />

cells have a nine times greater engraftment potential<br />

but this advantage is abrogated if the recipients are irradiated<br />

prior to transplant, indicating that the marrow<br />

microenvironment is important in driving myeloid and<br />

lymphoid engraftment. 344<br />

The importance of stromal cells in hematopoiesis has<br />

also been demonstrated by several studies in humans.<br />

Despite normal peripheral blood counts, levels of primitive<br />

and committed progenitors in the bone marrow of<br />

patients who have received allogeneic stem cell transplantation<br />

remain subnormal for many years. 345 Furthermore,<br />

cultured stromal cells from patients who have<br />

received allogeneic stem cell transplant (SCT) show significant<br />

impairment in their ability to support the growth<br />

of hematopoietic progenitors from normal marrow. 346<br />

Decreased CFU-GM production and defective stroma production<br />

have been demonstrated following autologous<br />

SCT 347 as well as after induction chemotherapy. 348<br />

The role of marrow stroma in hematopoietic regulation<br />

and the peculiar functional characteristics of stromal<br />

cells raise the possibility that the delivery of ex vivo<br />

expanded marrow MSC into a hematopoietically-compromised<br />

marrow might promote hematopoiesis. Bone<br />

marrow stromal cells are a quiescent, non-cycling population<br />

with low cell turn-over, as demonstrated by the<br />

resistance to irradiation. Based on these characteristics,<br />

methods have been developed which allow for gene<br />

delivery into stromal cells. 349 Since stromal cells are metabolically<br />

active they also provide a suitable means of<br />

secreting therapeutic proteins, including coagulation factors<br />

or adenosine deaminase. 350 Recent data showing that<br />

MSC suppress allogeneic T-cell responses in vitro suggest<br />

a role for stromal cells in modulating allogeneic<br />

haematologica vol. 85(suppl. to n. 12):December 2000


144<br />

C. Bordignon et al.<br />

transplant rejection and graft-versus-host disease. 351<br />

It must be emphasized that because of the limited<br />

knowledge of MSC biology, clinical applications of stromal<br />

cells, although exciting, essentially remain a matter<br />

of hypothesis to be carefully tested in the appropriate<br />

clinical setting. Essential prerequisites for clinical<br />

applications using culture-expanded mesenchymal cells<br />

as a supplement for hematopoietic SCT are (i) the possibility<br />

of isolating mesenchymal progenitors and<br />

manipulating their growth under defined in vitro culture<br />

conditions 352 and (ii) the demonstration of the possibility<br />

of efficiently introducing cultured stromal cells back<br />

into patients.<br />

Studies in rodents and dogs have clearly demonstrated<br />

that if sufficient stromal cells are reinfused, they not<br />

only seed the bone marrow but also enhance<br />

hematopoietic recovery. 353-357 Although demonstrated<br />

in several mouse models, the transplantability of marrow<br />

stromal elements remains a controversial issue in<br />

humans. 358, 359 The majority of data so far generated in<br />

recipients of HLA-identical marrow transplants has<br />

failed to demonstrate any contribution of donor cells to<br />

marrow stroma regeneration. 358 Although many factors<br />

may affect the transplantability of stromal elements,<br />

the low frequency of stromal progenitors in conventional<br />

marrow harvests may explain the failure of mesenchymal<br />

cell transplanttion in humans.<br />

Indeed, during the last decade, SCT methodology has<br />

changed substantially, particularly as a result of the<br />

increasing use of peripheral blood transplants. The existence<br />

of a circulating stromal progenitor has been<br />

demonstrated by using a NOD/SCID model and this is<br />

extremely relevant to stromal cell therapy. 360 By using<br />

the X-linked human androgen receptor (HUMARA) gene<br />

and fluorescent in situ hybridization analysis for the Y<br />

chromosome, the transplantability of stromal progenitors<br />

in a proportion of recipients of haploidentical HLAmismatched<br />

T-cell-depleted allografts reinfused with a<br />

combination of bone marrow and mobilized peripheral<br />

blood cells has recently been demonstrated (Carlo-Stella<br />

and Tabilio, unpublished observations, 1999). Taken<br />

together, these findings allow the hypothesis that MSC<br />

are transplantable in man provided that an adequate,<br />

but as yet unidentified, number of CFU-F is reinfused. In<br />

addition, these data allow the planning of clinical studies<br />

using culture-expanded, gene-marked mesenchymal<br />

cells in order to investigate a number of issues, including<br />

(i) dose of marrow stromal progenitors necessary to<br />

achieve a transplant; (ii) duration of post-transplant<br />

marrow stromal cell function; (iii) role of stromal cells<br />

in myeloid, B- and T-lymphoid reconstitution following<br />

SCT.<br />

A limited number of clinical trials using ex vivo generated<br />

MSC are currently underway. So far, the only<br />

published phase I clinical trial using MSC reported that<br />

the systemic infusion of autologous MSC appears to be<br />

well tolerated. 361 MSC can be explored as vehicles for<br />

both cell therapy and gene therapy (Table 4). MSC could<br />

be used to replace marrow microenvironment damaged<br />

Table 4. Potential clinical applications of mesenchymal<br />

stem cells.<br />

• Replacement of chemotherapy-damaged stroma<br />

• Enhancement of myeloid recovery following hematopoietic stem cell<br />

transplantation<br />

• Enhancement of T- and B-cell reconstitution following allogeneic<br />

stem cell transplantation<br />

• Compartimentalized growth factor/cytokine production<br />

• Modulation of GvHD<br />

• Delivery of exogenous gene products<br />

by high-dose chemotherapy in order to either improve<br />

hematopoietic recovery from myeloablative chemotherapy<br />

or to treat late graft failures or delayed platelet<br />

engraftment. Based on their functional characteristics,<br />

MSC are attractive vehicles for gene therapy in that they<br />

are expected not to be lost through differentiation as<br />

rapidly as hematopoietic progenitors. Examples of diseases<br />

in which stromal cell-mediated gene therapy<br />

might be appropriate include factor VIII and factor IX<br />

deficiencies and the various lysosomal storage diseases.<br />

Interestingly, compared to skin fibroblasts or leukocytes,<br />

marrow-derived mesenchymal cells produce significantly<br />

higher levels of -iduronidase, an enzyme<br />

involved in type II mucopolysaccharidoses (Danesino and<br />

Carlo-Stella, unpublished data). In addition, stromal cells<br />

might also be transduced with cDNA of various<br />

hematopoietic growth factors or cytokines. This<br />

approach might allow high levels of compartimentalized<br />

growth factor production and might be used (i) to stimulate<br />

hematopoiesis in patients with congenital or<br />

acquired hematopoietic defects, (ii) to improve B- and<br />

T-cell recovery following allogeneic SCT, (iii) to accelerate<br />

myeloid reconstitution in recipients of cord blood<br />

transplants.<br />

In conclusion, MSC appear to be an attractive therapeutic<br />

tool capable of playing a role in a wide range of<br />

clinical applications in the context of both cell and gene<br />

therapy strategies. However, a number of fundamental<br />

questions about MSC still need to be resolved before<br />

they can be used for safe and effective cell and gene<br />

therapy.<br />

Conclusions<br />

Although most of the new therapeutic approaches of<br />

cell therapy are experimental and have not yet been validated<br />

by phase III clinical trials, they appear to hold a<br />

high therapeutic potential. Separation of GVL from GvHD<br />

through generation and infusion of leukemia-specific T-<br />

cell clones or lines is one of the most intriguing and<br />

promising fields of investigations for the future. Likewise,<br />

strategies devised to improve immune reconstitution<br />

and restore specific anti-infectious functions<br />

through either induction of unresponsiveness to recipient<br />

alloantigens or removal of alloreactive donor T-cells<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

145<br />

might increase the applicability and success of<br />

hematopoietic stem cell transplantation. Cellular<br />

immunotherapy with DC must be standardized and several<br />

critical points, discussed in this review article must<br />

be properly addressed with specific clinical studies. Stimulation<br />

of leukemic cells via CD40 receptors and transduction<br />

of tumor cells with co-stimulatory molecules<br />

and/or cytokines may be useful in preventing tumor<br />

escape from immune surveillance. Tumor cells can be<br />

genetically modified to interact directly with dendritic<br />

cells in vivo or recombinant antigens can be delivered to<br />

dendritic cells using attenuated bacterial vectors by oral<br />

vaccination. MSC represent an attractive therapeutic<br />

tool capable of playing a role in a wide range of clinical<br />

applications in the context of both cell and gene therapy<br />

strategies.<br />

Contributions and Acknowledgments<br />

All authors gave substantial contributions to analysis<br />

and interpretation of literature data and drafting the<br />

article or revising it critically. CB was primarily responsible<br />

for the section on genetically engineered donor lymphocyte<br />

infusion for treatment of leukemia relapse, CCS<br />

for the section on mesenchymal cells, MPC for the sections<br />

on oral vaccination and engineered tumor vaccines;<br />

RML for the section on dendritic cells DR. FL was primarily<br />

responsible for sections on adoptive immunotherapy,<br />

AO for the sections on LAK and TIL and DR for the section<br />

on tumor escape from immune surveillance and on<br />

leukemic cells as APC. The authors are listed in alphabetical<br />

order.<br />

Disclosures<br />

Conflict of interest. This review article was prepared by<br />

request from <strong>Haematologica</strong>. The authors were a group of<br />

experts and representatives of two pharmaceutical companies,<br />

Amgen Italia SpA and Dompé Biotec SpA, both<br />

from Milan, Italy. This co-operation between a medical<br />

journal and pharmaceutical companies is based on the<br />

common aim of achieving optimal use of new therapeutic<br />

procedures in medical practice. In agreement with the<br />

Journal’s Conflict of Interest policy, the reader is given<br />

the following information. The preparation of this manuscript<br />

was supported by educational grants from the two<br />

companies. Dompé Biotec SpA sells G-CSF and rHuEpo in<br />

Italy, and Amgen Italia SpA has a stake in Dompé Biotec<br />

SpA.<br />

Redundant publications: no overlapping with previous<br />

papers.<br />

Manuscript processing<br />

Manuscript received May 10, 1999; accepted August<br />

30, 1999.<br />

References<br />

1. Murphy JB. Monography. Rockefeller Institute of<br />

Medical Research 1926; 21:1-168.<br />

2. Medawar PB. Croonian Lecture. Proc Royal Soc B<br />

1958; 148:159-61.<br />

3. Bertolini F, de Vincentiis A, Lanata L, et al. Allogeneic<br />

hematopoietic stem cells from sources other than<br />

bone marrow: biological and technical aspects. <strong>Haematologica</strong><br />

1997; 82:220-38.<br />

4. Arcese W, Aversa F, Bandini G, et al Clinical use of<br />

allogeneic hematopoietic stem cells from sources other<br />

than bone marrow. <strong>Haematologica</strong> 1998; 83:159-<br />

82.<br />

5. Aglietta M, Bertolini F, Carlo-Stella C, et al. Ex-vivo<br />

expansion of hematopoietic cells and their clinical use.<br />

<strong>Haematologica</strong> 1998; 83:824-48.<br />

6. Janeway CA Jr, Bottomly K. Signals and signs for lymphocyte<br />

responses. Cell 1994; 76:275-85.<br />

7. Thompson CB. Distinct roles for the costimulatory<br />

ligands B7-1 and B7-2 in T helper cell differentiation?<br />

Cell 1995; 81:979-82.<br />

8. van Kooten C, Banchereau J. Functions of CD40 on B<br />

cells, dendritic cells and other cells. Curr Opin<br />

Immunol 1997; 9:330-7.<br />

9. Boon T, DePlaen E, Traversari C, et al. Identification<br />

of tumor rejection antigens recognized by T lymphocytes.<br />

Cancer Surv 1992; 13:23-37.<br />

10. van der Bruggen P, Traversari C, Chomez P, et al. A<br />

gene encoding an antigen recognized by cytolytic T<br />

lymphocytes on human melanoma. Science 1991;<br />

254:1643-7.<br />

11. Restifo NP, Esquivel F, Kawakami Y, et al. Identification<br />

of human cancers deficient in antigen processing.<br />

J Exp Med 1993; 177:265-72.<br />

12. Merogi AJ, Marrogi AJ, Ramesh R, Robinson WR, Fermin<br />

CD, Freeman SM. Tumor-host interaction: analysis<br />

of cytokines, growth factors, and tumor-infiltrating<br />

lymphocytes in ovarian carcinomas. Hum Pathol<br />

1997; 28:321-31.<br />

13. Guinan EC, Gribben JG, Boussiotis VA, Freeman GJ,<br />

Nadler LM. Pivotal role of the B7: CD28 pathway in<br />

transplantation tolerance and tumor immunity. Blood<br />

1994; 84:3261-82.<br />

14. Maeurer MJ, Gollin SM, Martin D, et al. Tumor escape<br />

from immune recognition. J Clin Invest 1996; 98:<br />

1633-41.<br />

15. Wolfel T, Hauer M, Schneider J, et al. A p16 INK4a -insensitive<br />

CDK4 mutant targeted by cytolytic T lymphocytes<br />

in a human melanoma. Science 1995; 269:<br />

1281-4.<br />

16. Tanaka K, Yoshioka T, Bieberich C, Jay C. Role of<br />

major histocompatibility complex class I antigens in<br />

tumor growth and metastasis. Annu Rev Immunol<br />

1988; 6:359-80.<br />

17. Korkolopoulou P, Kaklamanis L, Pezzella F, Harris AL,<br />

Gatter KC. Loss of antigen-presenting molecules<br />

(MHC class I and TAP-1) in lung cancer. Br J Cancer<br />

1996; 97:148-53.<br />

18. Schultze J, Nadler LM, Gribben JG. B7-mediated costimulation<br />

and the immune response. Blood Rev<br />

1996; 10:111-27.<br />

19. Strand S, Galle PR. Immune evasion by tumours:<br />

involvement of the CD95 (APO-1/Fas) system and its<br />

clinical implications. Mol Med Today 1998; 4:63-8.<br />

20. Onrust SV, Harti PM, Rosen SD, Hanahan D. Modulation<br />

of L-selection ligand expression during an<br />

immune response accompanying tumorigenesis in<br />

transgenic mice. J Clin Invest 1996; 97:54-64.<br />

21. Herberman RB, Nunn ME, Lavrin DH. Natural cytotoxic<br />

reactivity of mouse lymphoid cells against syngeneic<br />

and allogeneic tumors. I. Distribution of reactivity<br />

and specificity. Int J Cancer 1975; 16: 216-29.<br />

22. Barlozzari T, Reynolds CW, Herberman RB. In vivo<br />

role of natural killer cells: involvement of large granular<br />

lymphocytes in the clearance of tumor cells in antiasialo<br />

GM1-treated rats. J Immunol 1983; 131:1024-<br />

7.<br />

23. Karre K, Ljunggren H-G, Piontek G, Kiessling R. Selec-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


146<br />

C. Bordignon et al.<br />

tive rejection of H2-deficient lymphoma variants suggests<br />

alternative immune defense strategy. Nature<br />

1986; 319:675-8.<br />

24. Braud VM, Allan DS, O’Callaghan CA, et al. HLA-E<br />

binds to natural killer cell receptors CD94/NKG2A, B<br />

and C. Nature 1998; 391; 795-9.<br />

25. Ciccone E, Grossi CE, Velardi A. Opposing functions<br />

of activatory T-cell receptors and inhibitory NK-cell<br />

receptors on cytotoxic T cells. Immunol Today 1996;<br />

17:451-3.<br />

26. Colonna M. Immunology: unmasking the killer's<br />

accomplice. Nature 1998; 391:642-3.<br />

27. Daniels B, Karlhofer FM, Seaman WE, Yokoyama<br />

WM. A natural killer cell receptor specific for a major<br />

histocompatibility complex class I molecule. J Ex Med<br />

1994; 180:687-92.<br />

28. Miller JS, Alley KA, McGlave PB. Differentiation of natural<br />

killer cells from human primitive marrow progenitors<br />

in a stroma based long term culture system:<br />

identification of a CD34+/CD7+ NK progenitor.<br />

Blood 1994; 83:2594-601.<br />

29. Mrozek E, Anderson P, Caligiuri MA. Role of interleukin-15<br />

in the development of human CD56+ natural<br />

killer cells from CD34+ hematopoietic progenitor<br />

cells. Blood 1996; 87:2632-40.<br />

30. Pierson BA, Miller JS. CD56+ bright and CD56+ dim natural<br />

killer cells in patients with chronic myelogenous<br />

leukemia progessively decrease in number, respond<br />

less to stimuli that recruit clonogenic natural killer<br />

cells, and exhibit decreased proliferation on a per cell<br />

basis. Blood 1996; 88:2279-87.<br />

31. Rayner AA, Grimm EA, Lotze MT, et al. Lymphokineactivated<br />

killer (LAK) cell phenomenon. IV. Lysis by<br />

LAK cell clones of fresh human tumor cells from autologous<br />

and multiple allogeneic tumors. J Natl Cancer<br />

Inst 1985; 75:67-75.<br />

32. Torpey DJ 3rd, Lindsley MD, Rinaldo CR Jr. HLArestricted<br />

lysis of herpes simplex virus-infected monocytes<br />

and macrophages mediated by CD4+ and CD8+<br />

T lymphocytes. J Immunol 1989; 142:1325-32.<br />

33. Jorgensen H, Hokland P, Jensen T, et al. Natural killer<br />

cells in peripheral blood after autologous bone marrow<br />

transplantation: a combined phenotypic and<br />

functional study. Nat Immunol 1995; 14:164-72.<br />

34. Roberts K, Lotze MT, Rosenberg SA. Separation and<br />

functional studies of the human lymphokine-activated<br />

killer cell. Cancer Res 1987; 47: 4366-71.<br />

35. Rosenberg SA. Lymphokine-activated killer cells: a new<br />

approach to immunotherapy of cancer. J Natl Cancer<br />

Inst 1985; 75:595-603.<br />

36. Papa MZ, Mulé JJ, Rosenberg SA. The antitumor efficacy<br />

of lymphokine activated killer cells and recombinant<br />

IL-2 in vivo: successful immunotherapy of established<br />

pulmonary metastases from weakly immunogenic<br />

and non-immunogenic murine tumors of three distinct<br />

histological types. Cancer Res 1986; 46:4973-8.<br />

37. Lotzova E, Savary CA, Herberman RB. Inhibition of<br />

clonogenic growth of fresh leukemia cells by unstimulated<br />

and IL-2 stimulated NK cells of normal donors.<br />

Leukemia Res 1987; 11:1059-66.<br />

38. Parrado A, Rodriguez-Fernadez JM, Casares S, et al.<br />

Generation of LAK cells in vitro in patients with acute<br />

leukemia. Leukemia 1993; 7:1344-8.<br />

39. Archimbaud E, Bailly M, Doré JF. Inducibility of lymphokine<br />

activated killer (LAK) cells in patients with<br />

acute myelogenous leukaemia in complete remission<br />

and its clinical relevance. Br J Haematol 1991; 77:328-<br />

34.<br />

40. Rosenberg SA, Lotze MT, Muul LM, et al. Observations<br />

on the systemic administration of autologous<br />

lymphokine-activated killer cells and recombinant<br />

interleukin-2 to patients with metastatic cancer. N<br />

Engl J Med 1985; 313:1485-92.<br />

41. Hawkins MJ. PPO Update IL2/LAK. Princ Prac Oncol<br />

1989; 3:1-4.<br />

42. Weiss GR, Margolin KA, Aronson FR, et al. A randomized<br />

phase II trial of conttinuous infusion interleukin-2<br />

or bolus injection interleukin-2 plus lymphokine-activated<br />

killer cells for advanced renal cell<br />

carcinoma. J Clin Oncol 1992; 10:275-81.<br />

43. McCabe MS, Stablein D, Hawkins MJ. The modified<br />

group C experience-phase III randomized trials of IL-<br />

2 versus IL-2/ LAK in advanced renal cell carcinoma<br />

and advanced melanoma [abstract]. Proceedings of<br />

American Society of Clinical Oncology; 1991; 10:<br />

213a.<br />

44. Rosenberg SA, Lotze MT, Yang JC, et al. Prospective<br />

randomized trial of high-dose interleukin-2 alone or in<br />

conjunction with lymphokine-activated killer cells for<br />

the treatment of patients with advanced cancer. J Natl<br />

Cancer Inst 1993; 85:622-32.<br />

45. Murray Law T, Motzer RJ, Mazumdar M, et al. Phase<br />

III randomized trial of interleukin-2 with or without<br />

lymphokine activated killer cells in the treatment of<br />

patients with advanced renal cell carcinoma. Cancer<br />

1995; 76: 824-32.<br />

46. Kimura H, Yamaguchi Y: A phase III randomized study<br />

of interleukin-2 lymphokine-activated killer cell<br />

immunotherapy combined with chemotherapy or<br />

radiotherapy after curative or noncurative resection<br />

of primary lung carcinoma. Cancer 1997; 80:42-9.<br />

47. Boldt DH, Mills BJ, Gemlo BT, et al. Laboratory correlates<br />

of adoptive immunotherapy with recombinant<br />

interleukin-2 and lymphokine-activated killer cells in<br />

humans. Cancer Res 1988; 48:4409-16.<br />

48. Margolin KA, Rayner AA, Hawkins MJ, et al. Interleukin-2<br />

and lymphokine-activated killer cell therapy<br />

of solid tumors: analysis of toxicity and management<br />

guidelines. J Clin Oncol 1989; 7:486-98.<br />

49. Négrier S, Philip T, Stoter G, et al. Interleukin-2 with<br />

or without LAK cells in metastatic renal cell carcinoma:<br />

a report of a European multicentre study. Eur J<br />

Cancer Clin Oncol 1989; 25(suppl 3):S21-S28.<br />

50. Escudier B, Farace F, Droz JP, et al. Abstract Proceedings<br />

of American Society of Clinical Oncology<br />

1991; 10:527a.<br />

51. Attal M, Blaise D, Marit G, et al. Consolidation treatment<br />

of adult acute lymphoblastic leukemia: a<br />

prospective, randomized trial comparing allogeneic<br />

versus autologous bone marrow trasplantation and<br />

testing the impact of recombinant interleukin-2 after<br />

autologous bone marrow transplantation. Blood<br />

1995; 86:1619-28.<br />

52. Fefer A, Benyunes M, Higuchi C, et al. IL-2+/- lymphokine<br />

activated killer cells as consolidative immunotherapy<br />

after autologous bone marrow transplantation<br />

for hematologic malignancies. Acta Hematol<br />

1993; 89:2-7.<br />

53. Beaujean F, Bernaudin F, Kuentz M, et al. Successful<br />

engraftment after autologous transplantation of 10-<br />

day cultured bone marrow activated by interleukin 2<br />

in patients with acute lymphoblastic leukemia. Bone<br />

Marrow Transplant 1995; 15:691-6.<br />

54. Mechan KR, Verma UN, Cahill R, et al. Interleukin-2-<br />

activated hematopoietic stem cell transplantation for<br />

breast cancer: investigation of dose level with clinical<br />

correlates. Bone Marrow Transplant 1997; 20:643-51.<br />

55. Olivieri A, Cantori I, Montanari M, et al. GM-CSF plus<br />

IL-2 administration associated with multiple autologous<br />

LAK reinfusions can induce a major cytogenetic<br />

response in early relapsed CML after autologous<br />

transplantation: a case report [abstract]. Bone Marrow<br />

Transplant 1998; 21:S65.<br />

56. Klingemann HG, Deal H, Reid D, Eaves CJ. Design<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

147<br />

and validation of a clinically applicable culture procedure<br />

for the generation of interleukin-2 activated<br />

natural killer cells in human bone marrow autografts.<br />

Exp Hematol 1993; 21:1263-70.<br />

57. Silva MRG, Parreira A, Ascensao JL. Natural killer cell<br />

numbers and activity in mobilized peripheral blood<br />

stem cell grafts: conditions for in vitro expansion. Exp<br />

Hematol 1995; 23:1676-81.<br />

58. Miller JS, Klingsporn S, Lund J, et al. Large-scale ex<br />

vivo expansion and activation of human natural killer<br />

cells for autologous therapy. Bone Marrow Transplant<br />

1994; 14:555-62.<br />

59. Vujanovic NL, Rabinowich H, Lee YJ Jost L, Herberman<br />

RB, Whiteside TL. Distinct phenotypes and functional<br />

characteristics of human natural killer cells obtained<br />

by rapid interleukin-induced adherence to plastic. Cell<br />

Immunol 1993; 151:133-57.<br />

60. Sheffold C, Brandt K, Johnston V. Potential of autologous<br />

immunologic effector cells for bone marrow<br />

purging in patients with chronic myeloid leukemia.<br />

Bone Marrow Transplant 1995; 15:33-9.<br />

61. Spiess PJ, Yang JC, Rosenberg SA. In vivo antitumor<br />

activity of tumor-infiltrating lymphocytes expanded in<br />

recombinant IL-2. J Natl Cancer Inst 1987; 79:1067-<br />

75.<br />

62. Balch CM, Riley LB, Bae YJ, et al. Patterns of human<br />

tumour-infiltrating lymphocytes in 120 human cancers.<br />

Arch Surg 1990; 12:200-5.<br />

63. Haas GP Solomon D, Rosenberg SA. Tumour-infiltrating<br />

lymphocytes from nonrenal urological malignancies.<br />

Cancer Immunol Immunother 1990; 30:342-<br />

50.<br />

64. Heo DS Whiteside TL, Johnson JT, Chen KN, Barnes<br />

EL, Herberman RB. Long term interleukin-2-dependent<br />

growth and cytotoxic activity of tumour-infiltrating<br />

lymphocytes from human squamous cell carcinoma<br />

of the head and neck. Cancer Res 1987; 47:<br />

6353-62.<br />

65. Rodolfo M, Salvi C, Bassi C, Parmiani G. Adoptive<br />

immunotherapy of a mouse colon carcinoma with<br />

recombinant interleukin-2 alone or combined with<br />

lymphokine-activated killer cells or tumor-immune<br />

lymphocytes. Survival benefit of adjuvant post-surgical<br />

treatments and comparison with experimental<br />

metastases model. Cancer Immunol Immunother<br />

1990; 31:28-36.<br />

66. Griffin JD. Hemopoietins in oncology: factoring out<br />

myelosuppression. J Clin Oncol 1989; 7:151-5.<br />

67. Wong RA, Alexander RB, Puri RK, Rosenberg SA. In<br />

vivo proliferation of adoptively transferred tumor-infiltrating<br />

lymphocytes in mice. J Immunother 1991; 10:<br />

120-30.<br />

68. Fisher B, Packard BS, Read EJ, et al. Tumor localization<br />

of adoptively transferred indium-111 labeled<br />

tumor infiltrating lymphocytes in patients with<br />

metastatic melanoma. J Clin Oncol 1989; 7:250-61.<br />

69. Rosenberg SA, Aebersold P, Cornetta K, et al. Gene<br />

transfer into humans: immunotherapy of patients<br />

with advanced melanoma using tumor infiltrating<br />

lymphocytes modified by retroviral gene transfer. N<br />

Engl J Med 1990; 323:570-8.<br />

70. Aebersold P, Hyatt C, Johnson S, et al. Lysis of autologous<br />

melanoma cells by tumor-infiltrating lymphocytes:<br />

association with clinical response. J Natl Cancer<br />

Inst 1991; 83:932-7.<br />

71. Barth RJ, Mulé JJ, Spiess PJ, Rosenberg SA. Interferon<br />

gamma and tumor necrosis factor have a role in<br />

tumor regressions mediated by murine CD8+ tumorinfiltrating<br />

lymphocytes. J Exp Med 1991; 173:647-<br />

58.<br />

72. Treisman J, Hwu P, Minamoto S, et al. Interleukin-2<br />

transduced lymphocytes grow in an autocrine fashion<br />

and remain responsive to antigen. Blood 1995; 85:<br />

139-45.<br />

73. Bani MR, Garofalo A, Scanziani E, Giavazzi R. Effect<br />

of interleukin-1- on metastases formation in different<br />

tumor systems. J Natl Cancer Inst 1991; 83:119-<br />

23.<br />

74. Malik STA, Naylor S, EAST N, Oliff A, Balkwill FR.<br />

Cells secreting tumour necrosis factor show enhanced<br />

metastases in nude mice. Eur J Cancer 1990; 26:<br />

1031-4.<br />

75. Robbins PF, Kawakami Y. Human tumor antigens recognized<br />

by T-cells. Curr Opin Immunol 1996; 8:628-<br />

36.<br />

76. Rosenberg SA, Packard BS, Aebersold PM, et al. Use<br />

of tumor-infiltrating lymphocytes and interleukin-2 in<br />

the immunotherapy of patients with metastatic<br />

melanoma. A preliminary report. N Engl J Med 1988;<br />

319:1676-80.<br />

77. Kradin RL, Lazarus DS, Dubinett SM, et al. Tumourinfiltrating<br />

lymphocytes and interleukin-2 in treatment<br />

of advanced cancer. Lancet 1989; 1:577-9.<br />

78. Deeg HJ, Spitzer TR, Cootler-Fox M, et al. Conditioning-related<br />

toxicity and acute graft-versus-host disease<br />

in patients given methotrexate/cyclosporine prophylaxis.<br />

Bone Marrow Transplant 1991; 7:193-8.<br />

79. Vogelsang GB, Hess AD. Graft-versus-host disease:<br />

new directions for a persistent problem. Blood 1994;<br />

84:2061-7.<br />

80. Xun CQ, Thompson JS, Jennings CD, et al. The effect<br />

of human IL-2-activated natural killer and T-cells on<br />

graft-versus-host disease and graft-versus-leukemia in<br />

SCID mice bearing human leukemic cells. Transplantation<br />

1995; 6:821-7.<br />

81. Miller JS, Prosper F, Mc Cullar V. Natural killer cells are<br />

functionally abnormal and NK cell progenitors are<br />

diminished in granulocyte colony-stimulating factormobilized<br />

peripheral blood progenitor cell collections.<br />

Blood 1997; 90:3098-105.<br />

82. Albi N, Ruggeri L, Aversa F, et al. Natural killer (NK)<br />

function and antileukemic activity of a large population<br />

of CD3+/CD8+ T cells expressing NK receptors<br />

for major histocompatibility complex class I after<br />

three-loci HLA-incompatible bone marrow transplantation.<br />

Blood 1996; 87:3993-4000.<br />

83. Lee S, Suen Y, Chang L, et al. Decreased interleukin-<br />

12 from activated cord versus adult peripheral blood<br />

mononuclear cells and upregulation of interferon-,<br />

natural killer, and lymphokine activated killer activity<br />

by IL-12 in cord blood mononuclear cells. Blood<br />

1996; 88:945-54.<br />

84. Verfaillie C, Miller W, Kay N, McGlave PB. Adherent<br />

lymphokine activated killer (ALAK) cells in chronic<br />

myelogenous leukemia: a benign cell population with<br />

potent cytotoxic activity. Blood 1989; 74:793-7.<br />

85. Hauch M, Gazzola MV, Small T, et al. Anti-leukemia<br />

potential of interleukin-2 activated natural killer cells<br />

after bone marrow transplantation for chronic myelogenous<br />

leukemia. Blood 1990; 75:2250-62.<br />

86. Miller JS, Verfaillie C, McGlave P. Expansion and activation<br />

of human natural killer cells for autologous<br />

therapy. J Hematother 1994; 3:71-4.<br />

87. Cervantes F, Pierson BA, McGlave PB, Verfaillie CM,<br />

Miller JS. Autologous activated natural killer cells suppress<br />

primitive chronic myelogenous leukemia progenitors<br />

in long-term culture. Blood 1996; 87:2476-<br />

85.<br />

88. Lanier LL, Phillips JH. Inhibitory MHC class I receptors<br />

on NK cells and T cells. Immunol Today 1986; 17:86-<br />

92.<br />

89. Schmidt-Wolf IGH, Lefterova P, Johnston V. Propagation<br />

of large numbers of T cells with natural killer<br />

cell markers. Br J Haematol 1994; 87:453-8.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


148<br />

C. Bordignon et al.<br />

90. Scheffold C, Brandt K, Johnston V. Potential of autologous<br />

immunologic effector cells for bone marrow<br />

purging in patients with chronic myeloid leukemia.<br />

Bone Marrow Transplant 1995; 15:33-9.<br />

91. Hoyle C, Bangs CD, Chang P, Kamel O, Metta B,<br />

Negrin RS. Expansion of Philadelphia chromosomenegative<br />

CD3+ CD56+ cytotoxic cells from chronic<br />

myeloid leukemia patients: in vitro and in vivo efficacy<br />

in severe combined immunodeficiency disease mice.<br />

Blood 1998; 92:3318-27.<br />

92. Nalesnik MA, Rao AS, Furukawa H, et al. Autologous<br />

lymphokine-activated killer cell therapy of Epstein-<br />

Barr virus-positive and -negative lymphoproliferative<br />

disorders arising in organ transplant recipients. Transplantation<br />

1997; 63:1200-5.<br />

93. Katsumoto Y, Monden T, Takeda T, et al. Analysis of<br />

cytotoxic activity of the CD4+ T lymphocytes generated<br />

by local immunotherapy. Br J Cancer 1996; 73:<br />

110-6.<br />

94. Tsurushima H, Qin Liu S, Tsuboi K, Yoshii Y, Nose T,<br />

Ohno T. Induction of human autologous cytotoxic T<br />

lymphocytes against minced tissues of glioblastoma<br />

multiforme. J Neurosurg 1996; 84:258-63.<br />

95. Schultze JL, Seamon MJ, Michalak S, Gribben JG,<br />

Nadler LM. Autologous tumor-infiltrating T cells cytotoxic<br />

for follicular lymphoma cells can be expanded in<br />

vitro. Blood 1997; 89:3806-16.<br />

96. Kanegane H, Tosato G. Activation of naive and memory<br />

T cells by interleukin-15. Blood 1996; 88:230-5.<br />

97. Robertson M, Soffier R, Wolf S, et al. Response of<br />

human natural killer (NK) cell to NK cell stimulatory<br />

factor (NKSF): cytolytic activity and proliferation of<br />

NK cells are differentially regulated by NKSF. J Exp<br />

Med 1992; 175:779-88.<br />

98. Rossi AR, Pericle F, Rashleigh S, Janiec J, Djeu J. Lysis<br />

of neuroblastoma cell lines by human natural killer<br />

cells activated by interleukin-12. Blood 1994; 83:<br />

1323-8.<br />

99. Kusher DI, Rashlegh SR, Endicott JN, Djeu J. Interleukin-2<br />

and interleukin-12 activate killer cell cytolytic<br />

response of peripheral blood mononuclear cells<br />

from patients with advanced head and neck squamous<br />

cell carcinoma [abstract]. Proceedings of the<br />

American Association of Cancer Research 1994; 35:<br />

3119a.<br />

100. Vitale A, Guarini A, Latagliata R, Cignetti A, Foa R.<br />

Cytotoxic effectors activated by low-dose IL-2 plus IL-<br />

12 lyse IL-2-resistant autologous acute myeloid<br />

leukemia blasts. Br J Haematol 1998; 101:150-7.<br />

101. Satoh M, Seki S, Hashimoto W, et al. Cytotoxic or<br />

T cells with a natural killer cell marker, CD56,<br />

induced from human peripheral blood lymphocytes<br />

by a combination of IL-12 and IL-2. J Immunol 1996;<br />

157:3886-92.<br />

102. Olivieri A, Cantori I, Provinciali M, et al. The association<br />

of GM-CSF plus IL-2 for cytotoxic cell expansion:<br />

influence of monocyte and GM-CSF concentration<br />

[abstract]. Blood 1997; 90:4302a.<br />

103. Basse P, Goldfarb RH. Localization of immune effector<br />

cells to tumor metastases. In: Goldfarb R, Whiteside<br />

T, eds. Tumor Immunology and Cancer Therapy,<br />

New York: Marcel Dekker, Inc., 1994. p. 149-58.<br />

104. Kuznetsov VA, Makalkin IA, Taylor MA, Perelson AS.<br />

Nonlinear dynamics of immunogenic tumors: parameter<br />

estimation and global bifurcation analysis. Bull<br />

Math Biol 1994; 56:295-321.<br />

105. Zhu H, Melder RJ, Baxter LT, Jain RK. Physiologically<br />

based kinetic model of effector cell biodistribution in<br />

mammals: implications for adoptive immunotherapy.<br />

Cancer Res 1996; 56:3771-81.<br />

106. Okada K, Nannmark U, Vujanovic N, et al. Elimination<br />

of established liver metastases by human interleukin-2-activated<br />

natural killer cells after locoregional<br />

or systemic adoptive transfer. Cancer Res 1996;<br />

56:1599-608.<br />

107. Sasaki A, Jain RK, Maghazachi AA, Goldfarb RH, Herberman<br />

RB. Low deformability of lymphokine-activated<br />

killer cells as a possible determinant of in vivo<br />

biodistribution. Cancer Res 1989; 49:3742-6.<br />

108. Melder RJ, Jain RK. Kinetics of interleukin-2 induced<br />

changes in rigidity of human natural killer cells. Cell<br />

Biophys 1992; 20:161-76.<br />

109. Shiloni E, Eisenthal A, Sachs D, et al. Antibody-dependent<br />

cellular cytotoxicity mediated by murine lymphocytes<br />

activated in recombinant interleukin-2. J<br />

Immunol 1987; 6:1992-8.<br />

110. Eisenthal A, Cameron RB, Uppenkamp I, et al. Effect<br />

of combined therapy with lymphokine-activated killer<br />

cells, interleukin-2 and specific monoclonal antibody<br />

on established B16 melanoma lung metastases. Cancer<br />

Res 1988; 48:7140-5.<br />

111. Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte<br />

transfusions for treatment of recurrent chronic<br />

myelogenous leukemia in marrow transplant patients.<br />

Blood 1990; 76:2462-5.<br />

112. Locatelli F. The role of repeat transplantation of haematopoietic<br />

stem cells and adoptive immunotherapy<br />

in treatment of leukaemia relapsing following allogeneic<br />

transplantation. Br J Haematol 1998; 102:633-<br />

8.<br />

113. Mackinnon S, Papadopoulos EB, Carabasi MH, et al.<br />

Adoptive immunotherapy evaluating escalating doses<br />

of donor leukocytes for relapse of chronic myeloid<br />

leukemia after bone marrow transplantation: separation<br />

of graft-versus-leukemia responses from graft-versus-host<br />

disease. Blood 1995; 86:1261-68.<br />

114. van Rhee F, Lin F, Cullis JO, et al. Relapse of chronic<br />

myeloid leukemia after allogeneic bone marrow transplant:<br />

the case for giving donor leukocyte transfusions<br />

before the onset of hematologic relapse. Blood 1994;<br />

83:3377-83.<br />

115. Collins-RH J, Shpilberg O, Drobyski WR, et al. Donor<br />

leukocyte infusions in 140 patients with relapsed<br />

malignancy after allogeneic bone marrow transplantation.<br />

J Clin Oncol 1997; 15:433-44.<br />

116. Kolb HJ, Schattenberg A, Goldman JM, et al. Graftversus-leukemia<br />

effect of donor lymphocyte transfusions<br />

in marrow grafted patients. European Group for<br />

Blood and Marrow Transplantation Working Party<br />

Chronic Leukemia. Blood 1995; 86:2041-50.<br />

117. Mehta J, Powles R, Treleaven J, et al. Outcome of<br />

acute leukemia relapsing after bone marrow transplantation:<br />

utility of second transplants and adoptive<br />

immunotherapy. Bone Marrow Transplant 1997; 19:<br />

709-19.<br />

118. Raanani P, Dazzi F, Sohal J, et al. The rate and kinetics<br />

of molecular response to donor leucocyte transfusions<br />

in chronic myeloid leukaemia patients treated<br />

for relapse after allogeneic bone marrow transplantation.<br />

Br J Haematol 1997; 99:945-50.<br />

119. Giralt S, Hester J, Huh Y, et al. CD8-depleted donor<br />

lymphocyte infusion as treatment for relapsed chronic<br />

myelogenous leukemia after allogeneic bone marrow<br />

transplantation. Blood 1995; 86:337-43.<br />

120. Lokhorst HM, Schattenberg A, Cornelissen JJ, Thomas<br />

LLM, Verdonck LF. Donor leukocyte infusions are<br />

effective in relapsed multiple myeloma after allogeneic<br />

bone marrow transplantation. Blood 1997; 90:<br />

4206-11.<br />

121. Dazzi F, Raanani P, van Rhee P, et al. Donor lymphocyte<br />

infusion (DLI) for relapse of CML after allo-BMT:<br />

comparison of two regimens. Bone Marrow Transplant<br />

1998; 21(Suppl. 1):S70.<br />

122. Slavin S, Naparstek E, Nagler A, et al. Allogeneic cell<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

149<br />

therapy with donor peripheral blood cells and recombinant<br />

human interleukin-2 to treat leukaemia relapse<br />

after allogeneic bone marrow transplantation. Blood<br />

1996; 87:2195-204.<br />

123. Murphy WJ, Longo DL.The potential role of NK cells<br />

in the separation of graft-versus-tumor effects from<br />

graft-versus-host disease after allogeneic bone marrow<br />

transplantation. Immunol Rev 1997; 157:167-<br />

76.<br />

124. Falkenburg, JH, Smit WM, Willemze R. Cytotoxic T-<br />

lymphocyte (CTL) responses against acute or chronic<br />

myeloid leukemia. Immunol Rev 1997; 157:223-30<br />

125. Goulmy E. Human minor histocompatibility antigens:<br />

new concepts for marrow transplantation and adoptive<br />

immunotherapy. Immunol Rev 1997; 157:125-<br />

40.<br />

126. Faber LM, van der Hoeven J, Goulmy E, et al. Recognition<br />

of clonogenic leukemic cells, remission bone<br />

marrow and HLA identical donor bone marrow by<br />

CD8+ or CD4+ minor histocompatibility antigen-specific<br />

cytotoxic T lymphocytes. J Clin Invest 1995; 96:<br />

877-83.<br />

127. Faber LM, van Luxemburg-Heijs SAP, Veenhof WFJ,<br />

Willemze R, Falkenburg JHF. Generation of CD4+<br />

cytotoxic T-lymphocytes clones from a patient with<br />

severe graft-versus-host disease after allogeneic bone<br />

marrow transplantation: implications for graft-versusleukemia<br />

reactivity. Blood 1995; 86:2821-8.<br />

128. Jiang JYZ, Mavroudis DA, Dermine S, Molldrem J,<br />

Hensel NF, Barrett AJ. Preferential usage of T cell<br />

receptor (TCR) V by allogeneic T cells recognizing<br />

myeloid leukemia cells: implications for separating<br />

graft-versus-leukemia effect from graft-versus-host disease.<br />

Bone Marrow Transplant 1997; 19:899-903.<br />

129. Barrett AJ, Malkovska V. Graft-versus-leukemia:<br />

understanding and using the alloimmune response to<br />

treat haematological malignancies. Br J Haematol<br />

1996; 93:754-61.<br />

130. Faber LM, van-Luxemburg Heijs SA, Willemze R,<br />

Falkenburg, JH. Generation of leukemia-reactive cytotoxic<br />

T lymphocyte clones from the HLA-identical<br />

bone marrow donor of a patient with leukemia. J Exp<br />

Med 1992; 176: 1283-9.<br />

131. Montagna D, Locatelli F, Calcaterra V, et al. Does the<br />

emergence and persistence of donor derived leukemiareactive<br />

cytotoxic T-lymphocytes protect patients given<br />

an allogeneic BMT from recurrence? Results of a<br />

preliminary study. Bone Marrow Transplant 1998; 22:<br />

743-50.<br />

132. Falkenburg JH, Wafelman AR, van Bergen CAM, et al.<br />

Leukemia-reactive cytotoxic T lymphocytes (CTL)<br />

induce complete remission in a patient with refractory<br />

accelerated phase chronic myeloid leukemia (CML).<br />

Blood 1997; 90(Suppl. 1):589a.<br />

133. Bonini C, Ferrari G, Verzelletti S, et al. HSV-TK gene<br />

transfer into donor-lymphocytes for control of allogeneic<br />

graft-versus-leukaemia. Science 1997; 276:<br />

1719-24.<br />

134. Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia<br />

reactions after bone marrow transplantation.<br />

Blood 1990; 75:555-62.<br />

135. Molldrem J, Dermime S, Parker K, et al. Targeted T-cell<br />

therapy for human leukemia: cytotoxic T lymphocytes<br />

specific for a peptide derived from proteinase 3 preferentially<br />

lyse human myeloid leukemia cells. Blood<br />

1996; 88:2450-7.<br />

136. Janeway CA Jr, Bottomly K. Signals and signs for lymphocyte<br />

responses. Cell 1994; 76:275-85.<br />

137. Gimmi CD, Freeman GJ, Gribben JG, Gray G, Nadler<br />

LM. Human T-cell clonal anergy is induced by antigen<br />

presentation in the absence of B7 costimulation. Proc<br />

Natl Acad Sci USA 1993; 90:6586-90.<br />

138. Noel PJ, Boise LH, Green JM, Thompson CB. CD28<br />

costimulation prevents cell death during primary T cell<br />

activation. J Immunol 1996; 157:636-42.<br />

139. Cardoso AA, Schultze JL, Boussiotis VA, et al. Pre-B<br />

acute lymphoblastic leukemia cells may induce T-cell<br />

anergy to alloantigen. Blood 1996; 88:41-8.<br />

140. Cardoso AA, Seamon MJ, Afonso HM, et al. Ex vivo<br />

generation of human anti-pre-B leukemia-specific<br />

autologous cytolytic T cells. Blood 1997; 90:549-61.<br />

141. Barrett AJ, Mavroudis D, Molldrem J, et al. Optimizing<br />

the dose and timing of lymphocytes add-back in<br />

T-cell depleted BMT between HLA-identical siblings.<br />

Blood 1996; 88 (Suppl. 1): 460a.<br />

142. Aversa F, Tabilio A, Terenzi A, et al. Successful engraftment<br />

of T-cell-depleted haploidentical “three-loci”<br />

incompatible transplants in leukemia patients by<br />

addition of recombinant human granulocyte colonystimulating<br />

factor-mobilized peripheral blood progenitor<br />

cells to bone marrow inoculum. Blood 1994;<br />

84: 3948-55.<br />

143. Gribben JG, Guinan EC, Boussiotis VA, et al. Complete<br />

blockade of B7 family-mediated costimulation is<br />

necessary to induce human alloantigen-specific anergy:<br />

a method to ameliorate graft-versus-host disease<br />

and extend the donor pool. Blood 1996; 87:4887-93.<br />

144. Comoli P, Montagna D, Moretta A, Zecca M, Locatelli<br />

F, Maccario R. Alloantigen-induced human lymphocytes<br />

rendered nonresponsive by a combination of<br />

anti-CD80 monoclonal antibodies and cyclosporin-A<br />

suppress mixed lymphocyte reaction in vitro. J<br />

Immunol 1995; 155:5506-11.<br />

145. Comoli P, Locatelli F, Montagna D, et al. Induction of<br />

alloantigen-specific anergy do not impair cytolytic<br />

activity of leukemia-reactive human T cells. Blood<br />

1997; 90(suppl. 1):535a.<br />

146. Cavazzana-Calvo M, Fromont C, Le Deist F, et al. Specific<br />

elimination of alloreactive T cells by an anti-interleukin-2<br />

receptor B chain-specific immunotoxin.<br />

Transplantation 1990; 50:1-7.<br />

147. Mickey B, Kohn C, Lowdell MW, Prentice HG. Selective<br />

removal of alloreactive lymphocytes from peripheral<br />

blood mononuclear cell preparations. Blood<br />

1996; 88(suppl 1):253a.<br />

148. Valteau-Couanet D, Cavazzana-Calvo M, Le Deist F,<br />

Fromont C, Fisher A. Functional study of residual T<br />

lymphocytes after specific elimination of alloreactive<br />

T cells by a specific anti-interleukin-2 receptor B chain<br />

immunotoxin. Transplantation 1993; 56:1574-6.<br />

149. Montagna D, Yvon E, Calcaterra V, et al. Depletion of<br />

alloreactive T cells by a specific anti-interleukin-2<br />

receptor p55 chain immunotoxin does not impair in<br />

vitro antileukemia and antiviral activity. Blood 1999;<br />

93:3550-7.<br />

150. Cavazzana-Calvo M, Stephan JL, Sarnacki S, et al.<br />

Attenuation of graft-versus-host disease and graft<br />

rejection by ex vivo immunotoxin elimination of alloreactive<br />

T cells in an H-2 haplotype disparate mouse<br />

combination. Blood 1994; 83:288-98.<br />

151. Groux H, O’Garra A, Bigler M, et al. A CD4+ T-cell<br />

subset inhibits antigen specific T-cell responses and<br />

prevents colitis. Nature 1997; 389:737-42.<br />

152. Riddell SR, Greenberg PD. Principles for adoptive T<br />

cell therapy of human viral disease. Annu Rev<br />

Immunol 1995; 13:545-86.<br />

153. Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha<br />

ME, Greenberg PD. Restoration of viral immunity in<br />

immunodeficient humans by the adoptive transfer of<br />

T cell clones. Science 1992; 257:238-41.<br />

154. Locatelli F, Percivalle E, Comoli P, et al. Human<br />

cytomegalovirus infection in pediatric patients given<br />

allogeneic bone marrow transplantation: role of early<br />

treatment of antigenemia on patients’ outcome. Br<br />

haematologica vol. 85(suppl. to n. 12):December 2000


150<br />

C. Bordignon et al.<br />

J Haematol 1994; 88:64-71.<br />

155. Goodrich JM, Bowden RA, Fisher L, Keller C, Schoch<br />

G, Meyers JD. Ganciclovir prophylaxis to prevent<br />

cytomegalovirus disease afetr allogeneic marrow<br />

transplant. Ann Intern Med 1993; 118:173-8.<br />

156. Reusser P, Riddell SR, Meyers JD, Greenberg PD. Cytotoxic<br />

T-lymphocyte response to cytomegalovirus after<br />

human allogeneic bone marrow transplantation: pattern<br />

of recovery and correlation with cytomegalovirus<br />

infection and disease. Blood 1991; 78:1373-80.<br />

157. Li CR, Greenberg PD, Gilbert MJ, Goodrich JM, Riddell<br />

SR. Recovery of HLA-restricted cytomegalovirus<br />

(CMV) specific T cell responses after allogeneic bone<br />

marrow transplantation: correlation with CMV disease<br />

and effect of ganciclovir prophylaxis. Blood 1994;<br />

83:1971-9.<br />

158. Walter EA, Greenberg PJ, Gilbert MJ, et al. Reconstitution<br />

of cellular immunity against cytomegalovirus<br />

in recipients of allogeneic bone marrow by transfer of<br />

T-cell clones from the donor. N Engl J Med 1995; 333:<br />

1038-44.<br />

159. Nalesnik MA. Posttransplantation lymphoproliferative<br />

disorders (PTLD): current perspectives. Semin<br />

Thor Cardiov Surg 1996; 8:139-48.<br />

160. Heslop EE, Rooney CM. Adoptive cellular immunotherapy<br />

for EBV lymphoproliferative diseases.<br />

Immunol Rev 1997; 157:217-22.<br />

161. O’Reilly R, Small TN, Papadopulos E, Lucas K, Lacerda<br />

J, Koulova L. Biology and adoptive cell therapy of<br />

Epstein-Barr-virus associated lymphoproliferative disorders<br />

in recipients of marrow allografts. Immunol<br />

Rev 1997; 157:195-216.<br />

162. Hale G, Waldmann H. Risks of developing Epstein<br />

Barr virus-related lymphoproliferative disorders after<br />

T cell depleted marrow transplants. Blood 1998; 91:<br />

3079-83.<br />

163. Rooney CM, Loftin SK, Holladay MS, Brenner MK,<br />

Krance RA, Heslop HE. Early identification of Epstein-<br />

Barr virus associated post-transplant lymphoproliferative<br />

disorders. Br J Haematol 1995; 89:98-103.<br />

164. Papadopoulos EB, Ladanyi M, Emanuel D, et al. Infusions<br />

of donor leukocytes as treatment of Epstein-Barr<br />

virus associated lymphoprolipherative disorders complicating<br />

allogeneic marrow transplantation. N Engl J<br />

Med 1994; 330:1185-91.<br />

165. Rooney CM, Smith CA, Ng CY, et al. Use of gene-modified<br />

virus-specific T lymphocytes to control Epstein-<br />

Barr-virus-related lymphoproliferation. Lancet 1995;<br />

345:9-13.<br />

166. Rooney CM, Smith CA, Ng CYC, et al. Infusion of<br />

cytotoxic T cells for the prevention and treatment of<br />

Epstein Barr virus-induced lymphoma in allogeneic<br />

transplant recipients. Blood 1998; 92:1549-55.<br />

167. Heslop HE, Ng CY, Li C, et al. Long-term restoration<br />

of immunity against Epstein-Barr virus infection by<br />

adoptive transfer of gene-modified virus-specific T<br />

lymphocytes. Nature Med 1996; 2:551-5.<br />

168. Haque T, Amlot PL, Helling N, et al. Reconstitution of<br />

EBV specific T cell immunity in solid organ transplant<br />

recipients. J Immunol 1998; 160:6204-9.<br />

169. Comoli P, Locatelli F, Gerna G, Grossi P, Viganò M,<br />

Maccario R. Autologous EBV-specific cytotoxic T cells<br />

to treat EBV-associated post-transplant lymphoproliferative<br />

disease (PTLD) [abstract]. Blood 1997; 90<br />

(Suppl.1):249a.<br />

170. Sing AP, Ambinder RF, Hong DJ, et al. Isolation of<br />

Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes<br />

that lyse Reed-Sternberg cells: implications for<br />

immune-mediated therapy of EBV+ Hodgkin disease.<br />

Blood 1997; 89:1978-86.<br />

171. Roskrow MA, Suzuki N, Gan Y, et al. Epstein-Barr virus<br />

(EBV)-specific cytotoxic T lymphocytes for the treatment<br />

of patients with EBV positive relapsed Hodgkin’s<br />

disease. Blood 1998; 91:2925-34.<br />

172. Smith CA, Woodruff LS, Kitchingman GR, Rooney<br />

CM. Adenovirus-pulsed dendritic cells stimulate<br />

human virus-specific T-cell responses in vitro. J Virol<br />

1996; 70:6733-40.<br />

173. Spitzer G, Velasquez W, Dunphy FR, Spencer V. Autologous<br />

bone marrow transplantation in solid tumors.<br />

Curr Opin Oncol 1992; 4:272-8.<br />

174. Jones RJ. Autologous bone marrow transplantation.<br />

Curr Opin Oncol 1993; 5:270-5.<br />

175. O'Reilly R. Bone marrow transplantation. Curr Opin<br />

Hematol 1993; 1:221-2.<br />

176. Thomas ED, Clift RA, Fefer A, et al. Marrow transplantation<br />

for the treatment of chronic myelogenous<br />

leukemia. Ann Intern Med 1986; 104:155-63.<br />

177. Santos GW, Hess AD, Volgelsang GB. Graft-versushost<br />

reactions and disease. Immunol Rev 1985; 88:<br />

169-92.<br />

178. Kernan NA, Bordignon C, Collins NH, et al. Bone marrow<br />

failure in HLA-identical T-cell depleted allogeneic<br />

transplants for leukemia: I Clinical aspects. Blood<br />

1989; 74:2227-36.<br />

179. Goldman JM, Gale RP, Horowitz MM, et al. Bone<br />

marrow transplantation for chronic myelogenous<br />

leukemia in chronic phase. Ann Intern Med 1988;<br />

108: 806-14.<br />

180. Zutter MM, Martin PJ, Sale GE, et al. Epstein-Barr<br />

virus lymphoproliferation after bone marrow transplantation.<br />

Blood 1988; 72:520-9.<br />

181. Shapiro RS, McClain K, Frizzera G, et al. Epstein-Barr<br />

virus associated lymphoproliferative disorders following<br />

bone marrow transplantation. Blood 1988; 71:<br />

1234-43.<br />

182. Tosato G. The Epstein-Barr virus and the immune system.<br />

Adv Cancer Res 1987; 49:75-125.<br />

183. Heslop HE, Brenner MK, Rooney CM. Donor T cells<br />

to treat EBV-associated lymphoma [letter]. N Engl J<br />

Med 1994; 331:679-80.<br />

184. Smith CA, Heslop E, Hollyday MS, et al. Production<br />

of genetically modified EBV-specific cytotoxic T cells<br />

for adoptive transfer to patient at high risk of EBVassociated<br />

lymphoproliferative disease. J Hematother<br />

1995; 4:473-9.<br />

185. Servida P, Rossini S, Traversari C, et al. Gene transfer<br />

into peripheral blood lymphocytes for in vivo immunomodulation<br />

of donor anti-tumor immunity in a<br />

patient affected by EBV-induced lymphoma [abstract].<br />

Blood 1993; 82:214a.<br />

186. Bordignon C, Bonini C, Verzeletti S, et al. Transfer of<br />

the HSV-tk gene into donor peripheral blood lymphocytes<br />

for in vivo modulation of donor anti-tumor<br />

immunity after allogeneic bone marrow transplantation.<br />

Hum Gene Ther 1995; 6:813-9.<br />

187. Moolten FL. Drug sensitivity (“suicide”) genes for<br />

selective cancer chemotherapy. Cancer Gene Ther<br />

1994; 1:279-87.<br />

188. Tiberghien P, Reynolds CW, Keller J, et al. Ganciclovir<br />

treatment of herpes simplex thymidine kinase-transduced<br />

primary T lymphocytes: an approach for specific<br />

in vivo donor T-cell depletion after bone marrow<br />

transplantation? Blood 1994; 84:1333-41.<br />

189. Verzeletti S, Bonini C, Traversari C, et al. Transfer of<br />

the HSV-tk gene into donor peripheral blood lymphocytes<br />

for in vivo immunomodulation of donor<br />

anti-tumor immunity after allo-BMT. Hum Gene Ther<br />

1998; 6:813-9.<br />

190. Mavilio F, Ferrari G, Rossini S, et al. Peripheral blood<br />

lymphocytes as target cells of retroviral vector-mediated<br />

gene transfer. Blood 1994; 83:1988-97.<br />

191. Sullivan KM, Storb R, Buckner CD, et al. Graft-versushost<br />

disease as adoptive immunotherapy in patients<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

151<br />

with advance hematologic neoplasms. N Engl J Med<br />

1989; 320:828-34.<br />

192. Porter DL, Roth MS, Mc Garigle C, Ferrara JLM, Antin<br />

JH. Induction of a graft-versus-host disease as immunotherapy<br />

for relapsed chronic myeloid leukemia. N<br />

Engl J Med 1994; 330:100-6.<br />

193. Drobyski WR, Keever CA, Roth MS, et al. Salvage<br />

immunotherapy using donor leukocyte infusions as<br />

treatment for relapsed chronic myelogenous leukemia<br />

after allogeneic bone marrow transplantation: efficacy<br />

and toxicity of a defined T-cell dose. Blood 1993;<br />

82:2310-8.<br />

194. Tricot G, Vesole DH, Jagganath S, Hilton J, Munshi N,<br />

Barlogie B. Graft-versus-myeloma effect: proof of principle.<br />

Blood 1996; 87:1196-8.<br />

195. Riddell SR, Elliott M, Lewinsohn DA, et al. T-cell mediated<br />

rejection of gene-modified HIV-specific cytotoxic<br />

lymphocytes in HIV-infected patients. Nature Med<br />

1996; 2:216-23.<br />

196. Bonini C, Verzelletti S, Servida P, et al. Immunity<br />

against the transgene product may limit efficacy of<br />

HSV-tk-transduced donor peripheral blood lymphocytes<br />

after allo-BMT [abstract]. Blood 1995; 84:628a.<br />

197. Phillips K, Gentry T, McCowange G, Gilboa E, Smith<br />

C. Cell-surface markers for assessing gene transfer into<br />

human hematopoietic cells. Nature Med 1996; 2:<br />

1154-6.<br />

198. Ciceri F, Marktel S, Bonini C, et al. HSV-TK genetically<br />

engineered donor lymphocytes restore anti-viral<br />

immunity early after T-depleted BMT [abstract].<br />

Blood 1998; 92:667a.<br />

199. Bancherau J, Steinman, RM. Dendritic cells and the<br />

control of immunity. Nature 1998; 392:245-52.<br />

200. Guery JC, Adorini L. Dendritic cells are the most efficient<br />

in presenting endogenous naturally processed<br />

self-epitopes to class II-restricted T cells. J Immunol<br />

1995; 154:536-44.<br />

201. Inaba K, Metlay JP, Crowley MT, Steinman RM. Dendritic<br />

cells pulsed with protein antigens in vitro can<br />

prime antigen-specific MHC-restricted T cells in situ.<br />

J Exp Med 1990; 172:631-40.<br />

202. Young JW, Steiman RM. Dendritic cells stimulate primary<br />

human cytolytic lymphocyte responses in the<br />

absence of CD4+ helper T cells. J Exp Med 1990; 171:<br />

1315-20.<br />

203. Bhardwaj N, Bender A, Gonzales N, et al. Influenza<br />

virus-infected dendritic cells stimulate strong proliferative<br />

and cytolytic responses from human CD8+ T<br />

cells. J Clin Invest 1994; 94:797-801.<br />

204. Metha Damani A, Markowicz S, Engleman EG. Generation<br />

of antigen-specific CD8+ CTLs from naive precursors.<br />

J Immunol 1994; 153:996-1003.<br />

205. Winzler C, Rovere P, Rescigno M, et al. Maturation<br />

stages of mouse dendritic cells in growth factordependent<br />

long-term cultures. J Exp Med 1997; 185:<br />

317-28.<br />

206. Macatonia SE, Hosken NA, Litton M, et al. Dendritic<br />

cells produce IL-12 and direct the development of<br />

TH1 cells from the naive CD4+ T cells. J Immunol<br />

1995; 154:5071-9.<br />

207. Cella M, Scheidegger D, Palmer-Lehmann K, et al. Ligation<br />

of CD40 on dendritic cells triggers production<br />

of high levels of interleukin-12 and enhances T cell<br />

stimulatory capacity: T-T help via APC activation. J<br />

Exp Med 1996; 184:747-52.<br />

208. Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic<br />

cell can be a temporal bridge between a CD4+<br />

T-helper and a T-killer cell. Nature 1998; 394:474-8.<br />

209. Bennet SRM, Carbone FR, Karamalis F, Flavell RA,<br />

Miller JFAP, Hearth WR. Help for cytotoxic T-cell<br />

responses is mediated by CD40 signalling. Nature<br />

1998; 394: 478-80.<br />

210. Schoenberger SP, Toes REM, van der Voort EIH,<br />

Offringa R, Melief CJM. T-cell help for cytotoxic T lymphocytes<br />

is mediated by CD40-CD40L interactions.<br />

Nature 1998; 394:480-3.<br />

211. Steinman RM, Swanson J. The endocytic activity of<br />

dendritic cells. J Exp Med 1995; 182:283-8.<br />

212. Koch F, Stanzl U, Jennewein P, et al. High level IL-12<br />

production by murine dendritic cells: upregulation via<br />

MHC class II and CD40 molecules and down regulation<br />

by IL-4 and IL-10. J Exp Med 1996; 184:741-7.<br />

213. Gabrilovich DI, Chen HL, Girgis KR , et al. Production<br />

of vascular endothelial growth factor by human<br />

tumors inhibits the functional maturation of dendritic<br />

cells. Nature Med 1996; 2:1096-103.<br />

214. Romani N, Gruner S, Brang D, et al. Proliferating dendritic<br />

cell progenitors in human blood. J Exp Med<br />

1994; 180:83-93.<br />

215. Sallusto F, Lanzavecchia A. Efficient presentation of<br />

soluble antigen by cultured human dendritic cells is<br />

maintained by granulocyte/macrophage colony-stimulating<br />

factor plus interleukin-4 and down regulated<br />

by tumor necrosis factor alpha. J Exp Med 1994; 179:<br />

1109-18.<br />

216. Reid CDL, Stackpole A, Meager A, Tikepae J. Interaction<br />

of tumor necrosis factor with granulocytemacrophage<br />

colony-stimulating factor and other<br />

cytokines in the regulation of dendritic cell growth in<br />

vitro from early bipotent CD34+ progenitors in<br />

human bone marrow. J Immunol 1992; 149:2681-8.<br />

217. Caux C, Dezutter-Dambuyant S, Schmitt D, Bancherau<br />

J. GM-CSF and TNF- cooperate in the generation<br />

of dendritic Langherans cells. Nature 1992; 360:258-<br />

61.<br />

218. Strunk D, Rappersberger K, Egger C, et al. Generation<br />

of human dendritic cells/Langherans cells from circulating<br />

CD34+ hematopoietic progenitor cells. Blood<br />

1996; 87:1292-302.<br />

219. Szabolcs P, Moore MAS, Young JW. Expansion of<br />

immunostimulatory dendritic cells among the myeloid<br />

progeny of human CD34+ bone marrow precursors<br />

cultured with c-kit ligand, granulocyte-macrophage<br />

colony-stimulating factor, and TNF-. J Immunol<br />

1995; 154:5851-61.<br />

220. Siena S, Di Nicola M, Bregni M, et al. Massive ex vivo<br />

generation of functional dendritic cells from mobilized<br />

CD34+ blood progenitors for anticancer therapy.<br />

Exp Hematol 1995; 23:1463-71.<br />

221. Fisch P, Kohler G, Garbe A, et al. Generation of antigen-presenting<br />

cells for soluble protein antigens ex<br />

vivo from peripheral blood CD34+ cells hematopoietic<br />

progenitor cells in cancer patients. Eur J Immunol<br />

1996; 26:595-600.<br />

222. Romani N, Reider D, Heuer M, et al. Generation of<br />

mature dendritic cells from human blood. An<br />

improved method with special regard to clinical<br />

applicability. J Immunol Methods 1996; 196:137-51.<br />

223. Bender A, Sapp M, Schuler G, Steinman RM, Bhardwaj<br />

N. Improved methods for the generation of dendritic<br />

cells from non-proliferating progenitors in human<br />

blood. J Immunol Methods 1996; 196:121-35.<br />

224. Bhardwaj N, Young JW, Nisanian AJ, Biggers J, Steinman<br />

RM. Small amounts of superantigen, when presented<br />

on dendritic cells, are sufficient to initiate T<br />

cell responses. J Exp Med 1993; 178:633-42.<br />

225. Ratta M, Rondelli D, Fortuna A, et al. Generation and<br />

functional characterization of human dendritic cells<br />

derived from CD34+ mobilized into peripheral blood:<br />

comparison with bone marrow CD34+ cells. Br J<br />

Haematol 1998;101:756-65.<br />

226. Rosenzwajg M, Canque B, Gluckman JC. Human dendritic<br />

cell differentiation pathway from CD34+ hematopoietic<br />

precursor cells. Blood 1996; 87:535-44.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


152<br />

C. Bordignon et al.<br />

227. Haug JS, Todd G, Bremer R, Link D, Brown R, DiPersio<br />

JF. Mobilization of CD80+ dendritic cells into the<br />

peripheral circulation by GM-CSF but not G-CSF<br />

[abstract]. Blood 1998; 92 (Suppl 1):444a.<br />

228. Lebsack ME, Maraskowsky E, Roux E, et al. Increased<br />

circulating dendritic cells in healthy human volunteers<br />

following administration of FLT3 ligand alone or in<br />

combination with GM-CSF or G-CSF [abstract].<br />

Blood 1998; 92 (Suppl 1):507a.<br />

229. Mortarini R, Anichini A, Di Nicola M, et al. Autologous<br />

dendritic cells derived from CD34+ progenitors<br />

and monocytes are not functionally equivalent APC in<br />

the induction of Melan-A/Mart-27-35-specific CTL<br />

from PBL of melanoma patients with low frequency of<br />

CTL precursors. Cancer Res 1997; 57:5534-41.<br />

230. Gong J, Chen D, Kashiwaba M, Kufe D. Induction of<br />

antitumor activity by immunization with fusions of<br />

dendritic and carcinoma cells. Nature Med 1997; 3:<br />

558-61.<br />

231. Albert LM, Sauter B, Bhardwaj N. Dendritic cells<br />

acquire antigen from apoptotic cells and induce class<br />

I-restricted CTLs. Nature 1998; 392:86-9.<br />

232. Reeves ME, Royal RE, Lam JS, Rosenberg SA, Hwu P.<br />

Retroviral transduction of human dendritic cells with<br />

a tumor-associated antigen gene. Cancer Res 1996;<br />

56:5672-7.<br />

233. Gong J, Chen L, Chen D, et al. Induction of antigenspecific<br />

antitumor immunity with adenovirus-transduced<br />

dendritic cells. Gene Ther 1998; 4:1023-8.<br />

234. Dietz AB, Vuk-Pavlovic S. High efficiency adenovirusmediated<br />

gene transfer to human dendritic cells.<br />

Blood 1998; 91:392-8.<br />

235. Di Nicola M, Siena S, Bregni M, et al. Gene transfer<br />

into human dendritic antigen-presenting cells by vaccinia<br />

virus and adenovirus vectors. Cancer Gene Ther<br />

1998; 5:350-6.<br />

236. Akagi J, Hodge JW, McLaughlin JP, et al. Therapeutic<br />

antitumor response after immunization with an<br />

admixture of recombinant vaccinia viruses expressing<br />

a modified MUC1 gene and the murine T-cell costimulatory<br />

molecule B-7. J Immunother 1997; 20:38-47.<br />

237. McLaughlin JP, Schlom J, Kantor JA, et al. Improved<br />

immunotherapy of a recombinant carcinoembryonic<br />

antigen vaccinia vaccine when given in combination<br />

with interleukin-2. Cancer Res 1997; 56:2361-7.<br />

238. Borysiewicz LK, Fiander A, Nimako M, et al. A recombinant<br />

vaccinia virus encoding human papillomavirus<br />

types 16 and 18, E6 and E7 proteins as immunotherapy<br />

for cervical cancer. Lancet 1996; 347:1523-7.<br />

239. McAneny D, Ryan CA, Beazley RM, et al. Results of a<br />

phase I trial of a recombinant vaccinia virus that<br />

expresses carcinoembryonic antigen in patients with<br />

advanced colorectal cancer. Ann Surg Oncol 1996; 3:<br />

495-500.<br />

240. Rescigno M, Citterio S, Thery C, et al. Bacteriainduced<br />

neo-biosynthesis, stabilization, and surface<br />

expression of functional class I molecules in mouse<br />

dendritic cells. Proc Natl Acad Sci USA 1998; 95:<br />

5229-34.<br />

241. Boczkowski D, Nair KS, Snyder D, Gilboa E. Dendritic<br />

cells pulsed with RNA are potent antigen-presenting<br />

cells in vitro and in vivo. J Exp Med 1996; 184:465-<br />

72.<br />

242. Mayordomo JL, Zorina T, Storkus WJ, et al. Bone marrow-derived<br />

dendritic cells pulsed with synthetic<br />

tumor peptides elicit protective and therapeutic antitumor<br />

immunity. Nature Med 1995; 1:1297-302.<br />

243. Celluzzi CM, Mayordomo JL, Storkus WJ, Lotze MT,<br />

Falo LD. Peptide-pulsed dendritic cells induce antigen-specific<br />

CTL-mediated protective tumor immunity.<br />

J Exp Med 1996; 183:283-7.<br />

244. Paglia P, Chiodoni C, Rodolfo M, Colombo MP.<br />

Murine dendritic cells loaded in vitro with soluble protein<br />

prime cytotoxic T lymphocytes against tumor antigen<br />

in vivo. J Expl Med 1996; 183:317-22.<br />

245. Porgador A, Snyder D, Gilboa E. Induction of antitumor<br />

immunity using bone marrow-generated dendritic<br />

cells. J Immunol 1996; 156:2918-26.<br />

246. Flamand V, Sornasse T, Thielemans K, et al. Murine<br />

dendritic cells pulsed in vitro with tumor antigen<br />

induce tumor resistance in vivo. Eur J Immunol 1994;<br />

24:605-10.<br />

247. Mukherji B, Charkraborty NG, Yamasaki S, et al.<br />

Induction of antigen specific cytolytic T cells in situ in<br />

human melanoma by immunization with synthetic<br />

peptide-pulsed autologous antigen presenting cells.<br />

Proc Natl Acad Sci USA 1995; 92:8078-82.<br />

248. Hu X, Charkraborty NG, Sporn JR, et al. Enhancement<br />

of cytolytic T lymphocyte precursors frequency in<br />

melanoma patients following immunization with the<br />

MAGE-1 peptide loaded antigen presented-based vaccine.<br />

Cancer Res 1996; 56:2479-83.<br />

249. Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of<br />

melanoma patients with peptide-or tumor lysatepulsed<br />

dendritic cells. Nature Med 1998; 4:328-32.<br />

250. Salgaller ML, Tjoa BA, Lodge PA, et al. Dendritic cellbased<br />

immunotherapy of prostate cancer. Crit Rev<br />

Immunol 1998; 18:109-19.<br />

251. Tao MH, Levy R. Idiotype/granulocyte-macrophage<br />

colony-stimulating factor fusion protein as a vaccine<br />

for B-cell lymphoma. Nature 1993; 362:755-8.<br />

252. Campbell MJ, Esserman L, Byars NE, et al. Idiotype<br />

vaccination against murine B cell lymphoma.<br />

Humoral and cellular requirements for the full expression<br />

of antitumor immunity. J Immunol 1990;<br />

145:1029-36.<br />

253. Hsu FJ, Caspar CB, Czerwinsky D, et al. Tumor-specific<br />

idiotype vaccines in the treatment of patients with<br />

B-cell lymphoma. Long-term results of a clinical study.<br />

Blood 1997; 89:3129-35.<br />

254. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of<br />

patients with B-cell lymphoma using autologous antigen-pulsed<br />

dendritic cells. Nature Med 1996; 2:52-8.<br />

255. Liso A, Stockerl-Goldstein KE, Reichardt VL, et al. Idiotype<br />

vaccination using dendritic cells after autologous<br />

peripheral blood progenitor cell transplantation for<br />

multiple myeloma [abstract]. Blood 1998; 92(Suppl.<br />

1):105a.<br />

256. Kaufmann SHE. Immunity to intracellular bacteria.<br />

Annu Rev Immunol 1993; 11:129-63.<br />

257. Paglia P, Arioli I, Frahm N, Chakraborty T, Colombo<br />

MP, Guzman CA. The defined attenuated Listeria<br />

monocytogenes ∆mp12 mutant is an effective oral vaccine<br />

carrier to trigger a long-lasting immune response<br />

against a mouse fibrosarcoma. Eur J Immunol 1997;<br />

27:1570-5.<br />

258. Paglia P, Medina E, Arioli I, Guzman CA, Colombo<br />

MP. Oral DNA vaccination with Salmonella<br />

typhimurium mediates gene transfer to antigen presenting<br />

cells and results in protective immunity against<br />

a murine fibrosarcoma. Blood 1998; 92:3172-6.<br />

259. Lanzavecchia A. Identifying strategies for immune<br />

intervention. Science 1993; 260:937-43.<br />

260. Hellstrom KE, Hellstrom I, Chen L. Can co-stimulated<br />

tumor immunity be therapeutically efficacious?<br />

Immunol Rev 1995; 145:123-45.<br />

261. Lauritzsen GF, Hofgaard PO, Scenck K, Bogen B. Clonal<br />

deletion of thymocytes as a tumor escape mechanism.<br />

Int J Cancer 1998; 78:216-22.<br />

262. Nagata S. Fas ligand and immune evasion. Nat Med<br />

1996; 2:1306-17.<br />

263. Chen L, Ashe S, Brady WA, et al. Costimulation of antitumor<br />

immunity by the B7 counter-receptor for the T<br />

lymphocyte molecules CD28 and CTLA-4. Cell 1992;<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

153<br />

71:1093-102.<br />

264. Townsend SE, Allison JP. Tumor rejection after direct<br />

costimulation of CD8+ T cells by B7-transfected melanoma<br />

cells. Science 1993; 259:368-70.<br />

265. Lanzavecchia A. Antigen-specific interactions between<br />

T and B cells. Nature 1985; 314:537-9.<br />

266. Kurt-Jones EA, Liano D, Hayglass KA, Benacerraf B,<br />

MS Sy, Abbas AK. The role of antigen-presenting B<br />

cells in T cell priming in vivo. Studies of B cell-deficient<br />

mice. J Immunol 1988; 140:3773-8.<br />

267. Schwartz RH. Models of T cell anergy: is there a common<br />

molecular mechanism? J Exp Med 1996;184:1-8.<br />

268. Noelle RJ. CD40 and its ligand in host defense. Immunity<br />

1996; 4:415-9.<br />

269. Yang Y, Wilson JM. CD40 ligand-dependent T cell activation:<br />

requirement of B7-CD28 signaling through<br />

CD40. Science 1996; 273:1864-7.<br />

270. Ranheim EA, Kipps TJ. Activated T cells induce expression<br />

of B7/BB1 on normal or leukemic B cells through<br />

a CD40-dependent signal. J Exp Med 1993; 177:925-<br />

35.<br />

271. Yellin MJ, Sinning J, Covey LR, et al. T lymphocyte T<br />

cell-B cell-activating molecule/CD40-L molecules<br />

induce normal B cells to express CD80 (B7/BB-1) and<br />

enhance their costimulatory activity. J Immunol 1994;<br />

153:666-74.<br />

272. Ranheim EA, Kipps TJ. Tumor necrosis factor-alpha<br />

facilitates induction of CD80 (B7-1) and CD54 on<br />

human B cells by activated T cells: complex regulation<br />

by IL-4, IL-10, and CD40L. Cell Immunol 1995; 161:<br />

226-35.<br />

273. Cantwell MJ, Sharma S, Friedmann T, Kipps TJ. Adenovirus<br />

vector infection of chronic lymphocytic<br />

leukemia B cells. Blood 1996; 88:4676-83.<br />

274. Funakoshi S, Longo DL, Beckwith M, et al. Inhibition<br />

of human B-cell lymphoma growth by CD40 stimulation.<br />

Blood 1994; 83:2787-94.<br />

275. Vyth-Dreese FA, Dellemijn TAM, van Oostveen JW,<br />

Feltkamp CA, Hekman A. Functional expression of<br />

adhesion receptors and costimulatory molecules by<br />

fresh and immortalized B-cell non-Hodgkin lymphoma<br />

cells. Blood 1995; 85:2802-12.<br />

276. Schultze JL, Cardoso AA, Freeman GJ, et al. Follicular<br />

lymphomas can be induced to present alloantigen efficiently:<br />

a conceptual model to improve their tumor<br />

immunogenicity. Proc Natl Acad Sci USA 1995; 92:<br />

8200-4.<br />

277. Inge TH, Hoover SK, Susskind BM, Barret SK, Bear<br />

HD. Inhibition of tumor-specific cytotoxic T-lymphocyte<br />

responses by transforming growth factor beta 1.<br />

Cancer Res 1992; 52:1386-92.<br />

278. Harada M, Matsunaga K, Oguchi Y, et al. The involvement<br />

of transforming growth factor beta in impaired<br />

antitumor T-cell response at the gut-associated lymphoid<br />

tissue (GALT). Cancer Res 1995;55:6146-51.<br />

279. Rondelli D, Andrews RG, Hansen JA, Ryncarz R, Faerber<br />

MA, Anasetti C. Alloantigen presenting function of<br />

normal human CD34+ hematopoietic cells. Blood<br />

1996; 88:2619-25.<br />

280. Rondelli D, Anasetti C, Fortuna A, et al. T cell alloreactivity<br />

induced by normal G-CSF-mobilized CD34+<br />

blood cells. Bone Marrow Transplant 1998; 21:1183-<br />

91.<br />

281. Ryncarz R, Anasetti C. Expression of CD86 on human<br />

marrow CD34+ cells identifies immunocompetent<br />

committed precursors of macrophages and dendritic<br />

cells. Blood 1998; 91:3892-900.<br />

282. Bocchia M, Wentworth PA, Southwood S, et al. Specific<br />

binding of leukemia fusion protein peptides to<br />

HLA class I molecules. Blood 1995; 85:2680-4.<br />

283. Bocchia M, Korontsvit T, Xu Q, et al. Specific human<br />

cellular immunity to bcr-abl oncogene-derived peptides.<br />

Blood 1996; 87:3587-92.<br />

284. Pawelec G, Max H, Halder T, et al. BCR/ABL leukemia<br />

oncogene fusion peptides selectively bind to certain<br />

HLA-DR alleles and can be recognized by T cells found<br />

at low frequency in the repertoire of normal donors.<br />

Blood 1996; 88:2118-24.<br />

285. Mannering SI, McKenzie JL, Fearnley DB, Hart DNJ.<br />

HLA-DR-restricted bcr-abl (b3a2)-specific CD4+ T<br />

lymphocytes respond to dendritic cells pulsed with<br />

b3a2 peptide and antigen-presenting cells exposed to<br />

b3a2 containing cell lysates. Blood 1997; 90:290-7.<br />

286. Papadopoulos KP, Suciu-Foca N, Hesdorffer CS,<br />

Tugulea S, Maffei A, Harris PE. Naturally processed<br />

tissue- and differentiation stage-specific autologous<br />

peptides bound by HLA class I and II molecules of<br />

chronic myeloid leukemia blasts. Blood 1997; 90:<br />

4938-46.<br />

287. Eibl B, Ebner S, Duba C, et al. Dendritic cells generated<br />

from blood precursors of chronic myelogenous<br />

leukemia patients carry the Philadelphia translocation<br />

and can induce a CML-specific primary cytotoxic T-<br />

cell response. Genes Chromosomes Cancer 1997;<br />

20:215-23.<br />

288. Choudhury A, Gajewski JL, Liang JC, et al. Use of<br />

leukemic dendritic cells for the generation of antileukemic<br />

cellular cytotoxicity against Philadelphia<br />

chromosome-positive chronic myelogenous leukemia.<br />

Blood 1997;89:1133-42.<br />

289. Choudhury A, Toubert A, Sutaria S, Charron D,<br />

Champlin RE, Claxton DF. Human leukemia-derived<br />

dendritic cells: ex-vivo development of specific<br />

antileukemic cytotoxicity. Crit Rev Immunol 1998; 18:<br />

121-31.<br />

290. Smit WM, Rijnbeek M, van Bergen CAM, et al. Generation<br />

of dendritic cells expressing bcr-abl from<br />

CD34-positive chronic myeloid leukemia precursor<br />

cells. Hum Immunol 1997; 53:216-23.<br />

291. Carlo-Stella C, Garau D, Regazzi E, et al. Generation<br />

of BCR/ABL positive dendritic cells from chronic myelogenous<br />

leukemia CD34+ cells [abstract]. Blood<br />

1998;92 (Suppl.1):2590.<br />

292. Misery L, Campos L, Dezutter-Dambuyant C, et al.<br />

CD1-reactive leukemic cells in bone marrow: presence<br />

of Langherans cell marker on leukemic monocytic<br />

cells. Eur J Haematol 1992; 48:27-32.<br />

293. Santiago-Schwarz F, Coppock DL, Hindenburg AA,<br />

Kern J. Identification of a malignant counterpart of<br />

the monocyte-dendritic cell progenitor in an acute<br />

myeloid leukemia. Blood 1994; 84:3054-62.<br />

294. Strunk D, Linkesch W. Acute myelogenous leukemia<br />

cells can be differentiated into dendritic cells in vitro<br />

[abstract]. Blood 1997; 90 (Suppl.1):829.<br />

295. Matulonis UA, Dosiou C, Lamont C, et al. Role of B7-<br />

1 in mediating an immune response to myeloid<br />

leukemia cells. Blood 1995; 85:2507-15.<br />

296. Matulonis UA, Dosiou C, Freeman G, et al. B7-1 is<br />

superior to B7-2 costimulation in the induction and<br />

maintenance of T-cell mediated anti-leukemia immunity.<br />

Further evidence that B7-1 and B7-2 are functionally<br />

distinct. J Immunol 1996; 156:1126-31.<br />

297. Dunussi-Joannopoulos K, Weistein HJ, Nickerson PW,<br />

et al. Irradiated B7-1 transduced primary acute myelogenous<br />

leukemia (AML) cells can be used as therapeutic<br />

vaccines in murine AML. Blood 1996; 87:2938-<br />

46.<br />

298. Mutis T, Schrama E, Melief CJM, Goulmy E. CD80-<br />

transfected acute myeloid leukemia cells induce primary<br />

allogeneic T-cell responses directed at patient specific<br />

minor histocompatibility antigens and leukemiaassociated<br />

antigens. Blood 1998; 92:1677-84.<br />

299. Li Y, Hellstrom KE, Newby SA, Chen L. Costimulation<br />

by CD48 and B7-1 induces immunity against poorly<br />

haematologica vol. 85(suppl. to n. 12):December 2000


154<br />

C. Bordignon et al.<br />

immunogenic tumors. J Exp Med 1996; 183:639-44.<br />

300. Colombo MP, Ferrari G, Stoppacciaro A, et al. Granulocyte<br />

colony-stimulating factor gene transfer suppresses<br />

tumorigenicity of a murine adenocarcinoma in<br />

vivo. J Exp Med 1991; 173:889-97.<br />

301. Zilocchi C, Stoppacciaro A, Chiodoni C, Parenza M,<br />

Terrazzini N, Colombo MP. Interferon gamma-independent<br />

rejection of interleukin 12-transduced carcinoma<br />

cells requires CD4+ T cells and granulocyte/macrophage<br />

colony-stimulating factor. J Exp<br />

Med 1998; 188:133-43.<br />

302. Zitvogel L, Robbins PD, Storkus WJ, et al. Interleukin-<br />

12 and B7.1 co-stimulation cooperate in the induction<br />

of effective antitumor immunity and therapy of<br />

established tumors. Eur J Immunol 1996; 26:1335-<br />

41.<br />

303. Dunussi-Joannopoulos K, Dranoff G, Weinstein HJ,<br />

Ferrara JL, Bierer BE, Croop JM. Gene immunotherapy<br />

in murine acute myeloid leukemia: granulocytemacrophage<br />

colony-stimulating factor tumor cell vaccines<br />

elicit more potent antitumor immunity compared<br />

with B7 family and other cytokine vaccines.<br />

Blood 1998; 91:222-30.<br />

304. Grange JM, Stanford JL, Rook GA. Tuberculosis and<br />

cancer: parallels in host responses and therapeutic<br />

approaches? Lancet 1995; 345:1350-2.<br />

305. Boon T, van der Bruggen P. Human tumor antigens<br />

recognized by T lymphocytes. J Exp Med 1996; 183:<br />

725-30.<br />

306. Forni G, Giovarelli M, Cavallo F, et al. Cytokine<br />

induced tumor immunogenicity: from exogenous<br />

cytokines to gene therapy. J Immunother 1993; 14:<br />

253-7.<br />

307. Huang YC, Golumbeck P, Ahmadzadeh M, Jaffee E,<br />

Pardoll D, Levitsky H. Role of bone-marrow derived<br />

cells in presenting MHC class I-restricted tumor antigens.<br />

Science 1994; 264:961-5.<br />

308. Arienti F, Sulé-Suso J, Belli F, et al Limited antitumor<br />

T cell response in melanoma patients vaccinated with<br />

interleukin-2 gene-transduced allogeneic melanoma<br />

cells. Hum Gene Ther 1996; 7:1955-63.<br />

309. Morton DL, Foshag LJ, Hoon DSB, et al. Prolongation<br />

of survival in metastatic melanoma after active specific<br />

immunotherapy with a new polyvalent melanoma vaccine.<br />

Ann Surg 1992; 216:463-82.<br />

310. Marchand M, Weynants P, Rankin E.Tumor regression<br />

responses in melanoma patients treated with a<br />

peptide encoded by gene MAGE-3. Int J Cancer 1995;<br />

63:883-5.<br />

311. Rodolfo M, Melani C, Zilocchi C, et al. IgG2a induced<br />

by IL-12-producing tumor cell vaccines but not IgG1<br />

induced by IL-4 vaccine are associated with the eradication<br />

of experimental metastases. Cancer Res 1998;<br />

58:5812-7.<br />

312. Rodolfo M, Zilocchi C, Cappetti B, Parmiani G,<br />

Melani C, Colombo MP. Eradication of experimental<br />

metastases by IL-12-transduced tumor vaccine is associated<br />

with GM-CSF producing CD8 lymphocytes recognizing<br />

tumor antigens that are not immunoselected.<br />

Gene Therapy 1999, in press.<br />

313. Colombo MP, Forni G. Cytokine gene transfer in<br />

tumor inhibition and tentative tumor therapy: Where<br />

are we now? Immunol Today 1994; 15:48-51.<br />

314. Scott MA, Gordon MY. In search of the haemopoietic<br />

stem cell. Br J Haematol 1995; 90:738-43.<br />

315. Gronthos S, Simmons PJ. The biology and application<br />

of human bone marrow stromal cell precursors.<br />

J Hematother 1996; 5:15-23.<br />

316. Dexter TM. Regulation of hemopoietic cell growth and<br />

development: experimental and clinical studies.<br />

Leukemia 1989; 3:469-74.<br />

317. Morrison SJ, Shah NM, Anderson DJ. Regulatory<br />

mechanisms in stem cell biology. Cell 1997; 88:287-<br />

98.<br />

318. Trentin JJ. Influence of hematopoietic organ stroma<br />

(hematopoietic inductive microenvironments) on<br />

stem cell differentiation. In: Gordon AS, ed. Regulation<br />

of hematopoiesis. vol 1. New York: Appleton,<br />

1970. p. 161-185.<br />

319. Simmons PJ, Zannettino A, Gronthos S, Leavesley D.<br />

Potential adhesion mechanisms for localisation of<br />

haemopoietic progenitors to bone marrow stroma.<br />

Leuk Lymphoma 1994; 12:353-63.<br />

320. Gordon MY, Riley GP, Watt SM, Greaves MF. Compartimentalization<br />

of a haemopoietic growth factor<br />

(GM-CSF) by glycosaminoglycans in the bone marrow<br />

microenvironment. Nature 1987; 326:403-5.<br />

321. Carlo-Stella C, Tabilio A. Stem cells and stem cell<br />

transplantation. <strong>Haematologica</strong> 1996; 81:573-87.<br />

322. Gordon MY. Physiological mechanisms in BMT and<br />

haemopoiesis - revisited. Bone Marrow Transplant<br />

1993; 11:193-7.<br />

323. Schofield R. The relationship between the haemopoietic<br />

stem cell and the spleen colony-forming cell: a<br />

hypothesis. Blood Cells 1978; 4:7-25.<br />

324. McGinnes K, Quesniaux V, Hitzler J, Paige C. Human<br />

B-lymphopoiesis is supported by bone marrowderived<br />

stromal cells. Exp Hematol 1991; 19:294-303.<br />

325. Landreth KS, Dorshkind K. Pre-B cell generation<br />

potentiated by soluble factors from a bone marrow<br />

stromal cell line. J Immunol 1988; 140:845-52.<br />

326. Kierney PC, Dorshkind K. B lymphocyte precursors<br />

and myeloid progenitors survive in diffusion chamber<br />

cultures but B cell differentiation requires close association<br />

with stromal cells. Blood 1987; 70:1418-24.<br />

327. Touw I, Löwenberg B. Production of T lymphocyte<br />

colony-forming units from precursors in human longterm<br />

bone marrow cultures. Blood 1984; 64:656-61.<br />

328. Dorshkind K, Johnson A, Collins L, Keller GM, Phillips<br />

RA. Generation of purified stromal cell cultures that<br />

support lymphoid and myeloid precursors. J Immunol<br />

Methods 1986; 89:37-47.<br />

329. Barda-Saad M, Rozenszajn LA, Globerson A, Zhang<br />

AS, Zipori D. Selective adhesion of immature thymocytes<br />

to bone marrow stromal cells: relevance to T cell<br />

lymphopoiesis. Exp Hematol 1996; 24:386-91.<br />

330. Prockop DJ. Marrow stromal cells as stem cells for<br />

nonhematopoietic tissues. Science 1997; 276:71-4.<br />

331. Owen ME, Cave J, Joyner CJ. Clonal analysis in vitro of<br />

osteogenic differentiation of CFU-F. J Cell Sci 1987;<br />

87:731-9.<br />

332. Owen ME, Friedenstein AJ. Stromal stem cells: marrow-derived<br />

osteogenic precursors. CIBA Found Symp<br />

1988; 136:42-60.<br />

333. Bennet JH, Joyner CJ, Triffitt JT, Owen ME. Adipocyte<br />

cells cultured from marrow have osteogenic potential.<br />

J Cell Sci 1991; 99:131-9.<br />

334. Castro-Malaspina H, Gay RE, Resnick G, et al. Characterization<br />

of human bone marrow fibroblast<br />

colony-forming cells (CFU-F) and their progeny. Blood<br />

1980; 56:289-301.<br />

335. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The<br />

development of fibroblast colonies in monolayer cultures<br />

of guinea-pig bone marrow and spleen cells. Cell<br />

Tissue Kinet 1970; 3:393-403.<br />

336. Ashton BA, Allen TD, Howlett CR, et al. Formation of<br />

bone and cartilage by marrow stromal cells in diffusion<br />

chambers in vivo. Clin Orthop 1980; 151:294-<br />

307.<br />

337. Friedenstein AJ, Chailakhyan RK, Latsinik NV, et al.<br />

Stromal cells responsible for transferring the microenvironment<br />

of the hemopoietic tissues. Cloning in vitro<br />

and retransplantation in vivo. Transplantation<br />

1974; 17:331-40.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Cell therapy<br />

155<br />

338. Gronthos S, Graves SE, Ohta S, Simmons PJ. The<br />

STRO-1+ fraction of adult human bone marrow contains<br />

the osteogenic precursors. Blood 1994; 84:<br />

4164-73.<br />

339. Owen M. Lineage of osteogenic cells and their relationship<br />

to the stromal system. J Bone Miner Res<br />

1985; 3:1-12.<br />

340. Simmons PJ, Torok-Storb B. Identification of stromal<br />

cell precursors in human bone marrow by a novel<br />

monoclonal antibody, STRO-1. Blood 1991; 78:55-<br />

62.<br />

341. Simmons PJ, Torok-Storb B. CD34 expression by stromal<br />

precursors in normal human adult bone marrow.<br />

Blood 1991; 78:2848-53.<br />

342. Majumdar MK, Thiede MA, Mosca JD, Moorman M,<br />

Gerson SL. Phenotypic and functional comparison of<br />

cultures of marrow-derived mesenchymal stem cells<br />

(MSCs) and stromal cells. J Cell Physiol 1998; 176:57-<br />

66.<br />

343. Almeida-Porada G, Ascensao JL, Zanjani ED. The role<br />

of sheep stroma in human haemopoiesis in the<br />

human/sheep chimaeras. Br J Haematol 1996; 93:<br />

795-802.<br />

344. Gan OI, Murdoch B, Larochelle A, Dick JE. Differential<br />

maintenance of primitive human SCID-repopulating<br />

cells, clonogenic progenitors, and long-term<br />

culture-initiating cells after incubation on human<br />

bone marrow stromal cells. Blood 1997; 90:641-50.<br />

345. Li S, Champlin R, Fitchen JH, Gale RP. Abnormalities<br />

of myeloid progenitor cells after "successful" bone<br />

marrow transplantation. J Clin Invest 1984; 75:234-<br />

41.<br />

346. O'Flaherty E, Sparrow R, Szer J. Bone marrow stromal<br />

function from patients after bone marrow transplantation.<br />

Bone Marrow Transplant 1995; 15:207-<br />

12.<br />

347. Domenech J, Gihana E, Dayan A, et al. Haemopoiesis<br />

of transplanted patients with autologous marrows<br />

assessed by long-term marrow culture. Br J Haematol<br />

1994; 88:488-96.<br />

348. Carlo-Stella C, Tabilio A, Regazzi E, et al. Effect of<br />

chemotherapy for acute myelogenous leukemia on<br />

hematopoietic and fibroblast marrow progenitors.<br />

Bone Marrow Transplant 1997; 20:465-71.<br />

349. Keating A, Horsfall W, Hawley RG, Toneguzzo F.<br />

Effect of different promoters on expression of genes<br />

introduced into hematopoietic and marrow stromal<br />

cells by electroporation. Exp Hematol 1990; 18:99-<br />

102.<br />

350. van Beusechem VW, Kukler A, Heidt PJ, Valerio D.<br />

Long-term expression of human adenosine deaminase<br />

in rhesus monkeys transplanted with retrovirus-infected<br />

bone-marrow cells. Proc Natl Acad Sci U S A 1992;<br />

89:7640-4.<br />

351. Klyushnenkova E, Mosca JD, McIntosh KR, Thiede<br />

MA. Human mesenchymal stem cells suppress allogeneic<br />

T cell responses in vitro: implications for allogeneic<br />

transplantation [abstract]. Blood 1998; 92<br />

(suppl 1):2652.<br />

352. Gronthos S, Simmons PJ. The growth factor requirements<br />

of STRO-1-positive human bone marrow stromal<br />

precursors under serum-deprived conditions in<br />

vitro. Blood 1995; 85: 929-40.<br />

353. Perkins S, Fleischman RA. Hematopoietic microenvironment:<br />

origin, lineage, and transplantability of the<br />

stromal cells in long-term bone marrow cultures from<br />

chimeric mice. J Clin Invest 1988; 81:1072-7.<br />

354. Anklesaria P, Kase K, Glowacki J, et al. Engraftment of<br />

a clonal bone marrow stromal cell line in vivo stimulates<br />

hematopoietic recovery from total body irradiation.<br />

Proc Natl Acad Sci USA 1987; 84:7681-5.<br />

355. El-Badri NS, Wang BY, Cherry, Good RA. Osteoblasts<br />

promote engraftment of allogeneic hematopoietic<br />

stem cells. Exp Hematol 1998; 26:110-6.<br />

356. Nolta JA, Hanley MB, Kohn DB. Sustained human<br />

hematopoiesis in immunodeficient mice by co-transplantation<br />

of marrow stroma expressing human interleukin-3:<br />

analysis of gene transduction of long-lived<br />

progenitors. Blood 1994; 83:3041-51.<br />

357. Anklesaria P, Fitzgerald TJ, Kase K, Ohara A, Greenberger<br />

JS. Improved hematopoiesis in anemic Sl/Sld<br />

mice by splenectomy and therapeutic transplantation<br />

of a hematopoietic microenvironment. Blood 1989;<br />

74:1144-51.<br />

358. Simmons PJ, Przepiorka D, Thomas ED, Torok-Storb<br />

B. Host origin of marrow stromal cells following allogeneic<br />

bone marrow transplantation. Nature 1987;<br />

328:429-32.<br />

359. Keating A, Singer JW, Killen PD, et al. Donor origin of<br />

the in vitro haemopoietic microenvironment after<br />

marrow transplantation in man. Nature 1982;<br />

298:280-3.<br />

360. Henschler R, Junghahn I, Fichtner I, Becker M, Goan<br />

SR. Donor fibroblasts from human blood engraft in<br />

immunodeficient mice [abstract]. Blood 1998; 92<br />

(suppl 1): 2417.<br />

361. Lazarus HM, Haynesworth SE, Gerson SL, Rosenthal<br />

NS, Caplan A. Ex vivo expansion and subsequent infusion<br />

of human bone marrow-derived stromal progenitor<br />

cells (mesenchymal progenitor cells): implications<br />

for therapeutic use. Bone Marrow Transplant 1995;<br />

16:557-64.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


eview<br />

Antitumor vaccination:<br />

where we stand<br />

haematologica 2000; 85(supplement to no. 12):156-187<br />

Correspondence: Prof. Sante Tura, Istituto di Ematologia e Oncologia<br />

Medica “Seràgnoli”, Università di Bologna, Policlinico S.Orsola-<br />

Malpighi, via Massarenti 9, 40138 Bologna, Italy. Phone: international<br />

+39-051-6363680 – Fax: international-39-051-6364037 –<br />

E-mail: tura@orsola-malpighi.med.unibo.it<br />

MONICA BOCCHIA,* VINCENZO BRONTE,° MARIO P. COLOMBO, #<br />

ARMANDO DE VINCENTIIS, @ MASSIMO DI NICOLA,^<br />

GUIDO FORNI, § LUIGI LANATA,** ROBERTO M. LEMOLI,°°<br />

MASSIMO MASSAIA, ## DAMIANO RONDELLI,°° PAOLA ZANON,**<br />

SANTE TURA°°<br />

*Department of Hematology, University of Siena; °Department<br />

of Oncology, University of Padova; # Division of Oncology, Istituto<br />

Nazionale dei Tumori, Milan; @ Dompè-Biotec, Milan; ^Bone<br />

Marrow Transplant Unit, Istituto Nazionale dei Tumori, Milan;<br />

§<br />

Department of Clinical and Biological Sciences, University of<br />

Turin; **Amgen Italia, Milan; °°Institute of Hematology and<br />

Medical Oncology “Seràgnoli”, University of Bologna; ## Division<br />

of Hematology, University of Turin, Italy<br />

Background and Objectives. Vaccination is an effective<br />

medical procedure of preventive medicine based on<br />

the induction of a long-lasting immunologic memory<br />

characterized by mechanisms endowed with high<br />

destructive potential and specificity. In the last few<br />

years, identification of tumor-associated antigens (TAA)<br />

has prompted the development of different strategies<br />

for antitumor vaccination, aimed at inducing specific<br />

recognition of TAA in order to elicit a persistent<br />

immune memory that may eliminate residual tumor<br />

cells and protect recipients from relapses. In this<br />

review characterization of TAA, different potential<br />

means of vaccination in experimental models and preliminary<br />

data from clinical trials in humans have been<br />

examined by the Working Group on Hematopoietic<br />

Cells.<br />

Evidence and Information Sources. The method<br />

employed for preparing this review was that of informal<br />

consensus development. Members of the Working<br />

Group met four times and discussed the single points,<br />

previously assigned by the chairman, in order to<br />

achieve an agreement on different opinions and<br />

approve the final manuscript. Some of the authors of<br />

the present review have been working in the field of<br />

antitumor immunotherapy and have contributed original<br />

papers to peer-reviewed journals. In addition, the<br />

material examined in the present review includes articles<br />

and abstracts published in journals covered by the<br />

Science Citation Index and Medline.<br />

State of the art. The cellular basis of antitumor<br />

immune memory consists in the generation and extended<br />

persistence of expanded populations of T- and B-<br />

lymphocytes that specifically recognize and react<br />

against TAA. The efficacy of the memory can be modulated<br />

by compounds, called "adjuvants", such as certain<br />

bacterial products and mineral oils, cytokines,<br />

chemokines, by monoclonal antibodies triggering costimulatory<br />

receptors. Strategies that have been shown<br />

in preclinical models to be efficient in protecting from<br />

tumor engraftment, or in preventing a tumor rechallenge,<br />

include vaccination by means of soluble proteins<br />

or peptides, recombinant viruses or bacteria as TAA<br />

genes vectors, DNA injection, tumor cells genetically<br />

modified to express co-stimulatory molecules and/or<br />

cytokines. The use of professional antigen-presenting<br />

cells, namely dendritic cells, either pulsed with TAA or<br />

transduced with tumor-specific genes, provides a useful<br />

alternative for inducing antitumor cytotoxic activity.<br />

Some of these approaches have been tested in phase<br />

I/II clinical trials in hematologic malignancies, such as<br />

lymphoproliferative diseases or chronic myeloid<br />

leukemia, and in solid tumors, such as melanoma,<br />

colon cancer, prostate cancer and renal cell carcinoma.<br />

Different types of vaccines, use of adjuvants, timing of<br />

vaccination as well as selection of patients eligible for<br />

this procedure are discussed in this review.<br />

Perspectives. Experimental models demonstrate the<br />

possibility of curing cancer through the active induction<br />

of a specific immune response to TAA. However, while<br />

pre-clinical research has identified several possible targets<br />

and strategies for tumor vaccination the clinical<br />

scenario is far more complex for a number of possible<br />

reasons. Since experimental data suggest that vaccination<br />

is more likely to be effective on small tumor burden,<br />

such as a minimal residual disease after conventional<br />

treatments, or tumors at an early stage of disease,<br />

better selection of patients will allow more reliable<br />

clinical results to be obtained. Moreover, a poor<br />

correlation is frequently observed between the ability of<br />

TAA to induce a T-cell response in vitro and clinical<br />

responses. Controversial findings may also be due to<br />

the techniques used for monitoring the immune status.<br />

Therefore, the development of reliable assays for efficient<br />

monitoring of the state of immunization of cancer<br />

patients against TAA is an important goal that will<br />

markedly improve the progress of antitumor vaccines.<br />

Finally, given the promising results, identification of<br />

new or mutated genes involved in neoplastic events<br />

might provide the opportunity to vaccinate susceptible<br />

subjects against their foreseeable cancer in the next<br />

future.<br />

©2000, Ferrata Storti Foundation<br />

Key words: antitumor vaccination.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

157<br />

Antitumor vaccines: the meaning<br />

As illustrated in a previous paper of this series, many<br />

strategies are being used to try to cure cancer, each one<br />

based on different theoretical and experimental<br />

grounds. 1 Active immunotherapy strategies elicit specific<br />

or non-specific antitumor reactions by stimulating the<br />

patient’s immune system. Alternatively, lymphocytes<br />

collected from patients are stimulated in vitro and reinjected<br />

into the patient (adoptive immunotherapy).<br />

Lastly, passive immunotherapy consists in the administration<br />

of antitumor antibodies to the patient. However,<br />

dealing with such a dramatic issue as is cancer, emotional<br />

empiricism spurs the adoption of distinct strategies<br />

or their mixing in an apprehensive pursuit of efficacy.<br />

Indeed, emotional empiricism has been and still is<br />

a deadly sin of tumor immunology. In the long run, only<br />

rational considerations lead to clinical progress.<br />

The issue<br />

Vaccination is an effective medical procedure characterized<br />

by being a) predominantly a maneuver of preventive<br />

medicine; b) based on the induction of a longlasting<br />

immunologic memory that is c) characterized by<br />

mechanisms endowed with high destructive potential<br />

and specificity. 2 It rests on an artificial encounter of a<br />

sham pathogen with the immune system. The sham<br />

pathogen elicits a strong host reaction and leaves a persistent<br />

memory of this first artificial fight. If the real<br />

pathogen enters the immunized organism it becomes the<br />

target of a much stronger and precise reaction than that<br />

put up by a non-immunized organism. If the pathogen<br />

had a small possibility of escaping the reaction of a naive<br />

immune system, very seldom would it evade a memory<br />

reaction. 3 The cellular basis of immune memory consists<br />

in the extended persistence of expanded populations of<br />

T- and B- lymphocytes that specifically recognize and<br />

react against the pathogen. Memory lymphocytes are also<br />

experienced veterans, able to detect a pathogen promptly<br />

and fight effectively against it.<br />

There is a large universe of sham pathogens that are<br />

used for vaccination, named antigens or immunogens.<br />

A killed or inactivated pathogen, or a non-pathogenic<br />

organism sharing critical molecules with the real thing<br />

can be a good immunogen. Memory can also be induced<br />

by a protein from the pathogen. In addition, a virus engineered<br />

with the DNA coding for a protein of the<br />

pathogen or even the mere naked DNA induces an effective<br />

memory. 4<br />

The efficacy of the memory is modulated by numerous<br />

compounds, called adjuvants and danger signals. 5,6<br />

These provide additional activation signals, recruit reactive<br />

leukocytes at the immunization site and delay antigen<br />

catabolism. Bacterial products and mineral oils are<br />

typical conventional adjuvants. Cytokines and chemokines<br />

also act as adjuvants. As will be discussed in detail,<br />

their use allows the induction of selective mechanisms<br />

of immune memory. 7<br />

Antitumor vaccination has a defined goal: to provoke<br />

specific recognition of tumor-associated antigens (TAA)<br />

in order to elicit a persistent immune memory. Many<br />

experimental data have shown that following immunization,<br />

the growth of tumor cells expressing the same<br />

TAA as the vaccine can be impaired. In patients, the<br />

immune memory elicited by vaccines is sometimes fast<br />

and strong enough to hamper the growth of their<br />

tumor. 8<br />

A brief history<br />

Interest in antitumor vaccination arose around 1900<br />

when a series of microbial vaccines proved to be effective.<br />

The idea was straightforward: to apply the same<br />

intervention to tumor. «If .........it is possible to protect<br />

small laboratory animals in an easy and safe way against<br />

infectious and highly aggressive neoplastic specimens,<br />

then it will be possible to do the same for human<br />

patients». These words of 1897 by Paul Ehrlich 9 ignited<br />

a series of studies with transplantable mouse tumors.<br />

However the underlying issue turned out to be more<br />

complex than had first been presumed. More than one<br />

century was required to elucidate its molecular and<br />

genetic features.<br />

The first outcome was not a progress in anti-tumor<br />

vaccination, but instead the definition of a few rules of<br />

allograft rejection. Transplanted tumors were rejected by<br />

immunized host not because they expressed a particular<br />

TAA but because they were from histoincompatible<br />

mice and displayed normal allogeneic histocompatibility<br />

antigens. 10 Later studies with syngeneic mouse<br />

strains showed the feasibility of immunizing a mouse<br />

against a subsequent tumor challenge. 11 However, the<br />

suspicion that residual unnoticed histocompatibility differences<br />

were involved in these vaccination-rejection<br />

studies was not ruled out until experiments by George<br />

Klein in 1960. 12 Carcinoma was induced by methylcholanthrene<br />

in syngeneic mice. The carcinoma was<br />

then surgically removed, and its cells were cultured in<br />

vitro and used to repeatedly immunize the mouse in<br />

which the tumor had arisen. Finally the immunized<br />

mouse and a few control syngeneic mice were challenged<br />

with the carcinoma cells of the original tumor<br />

maintained in vitro. While these cells gave rise to a carcinoma<br />

in control mice, they were rejected by the immunized<br />

mouse in which the carcinoma had arisen originally.<br />

This evidence of the possibility of immunizing<br />

against a lethal dose of own tumor was of seminal<br />

importance and had notable consequences. A very large<br />

series of subsequent studies established a few basic<br />

foundations of tumor vaccination. 13<br />

Looking back at tumor immunology over the last 20-<br />

30 years, the importance of models in influencing<br />

immunologic beliefs is strikingly evident. Inappropriate<br />

use of an experimental model may produce wrong<br />

beliefs, from which it is then very hard to escape. Virusand<br />

chemically-induced tumors form highly immunogenic<br />

models. Since they are easy to handle, these were<br />

used to establish the rules of tumor vaccination. However,<br />

it was disputable whether the information from<br />

these models was relevant to the situation of patients<br />

haematologica vol. 85(suppl. to n. 12):December 2000


158<br />

M. Bocchia et al.<br />

with cancer. Using a series of murine spontaneous<br />

tumors, Hewitt concluded that it was not possible to<br />

immunize against these tumors. 14 This observation had<br />

crucial importance in shaping subsequent studies. The<br />

possibility that the experimental work done with high<br />

immunogenic transplantable tumors had little relevance<br />

to human tumors was a dark shadow that hindered the<br />

progress of tumor immunology. Later, more careful use<br />

of these spontaneous tumors and more refined immunization<br />

techniques showed that Hewitt's conclusions<br />

were wrong. 15<br />

Boon led the genetic and molecular identification of<br />

a large series of TAA. Initially his studies were performed<br />

using conventional transplantable mouse tumors. 16<br />

Then, tumor antigens were detected on the same spontanous<br />

tumors that had previously been classified as<br />

non-immunogenic by Hewitt. 15 Now many antigens<br />

associated with human tumors have been identified. 17,18<br />

The targets<br />

Boon and others 18,19 provided an unambiguous definition<br />

of TAA, an important finding that definitively laid<br />

to rest the doubts on the foundations of tumor<br />

immunology in man. 20 In many cases TAA are peptides<br />

presented by class I and class II glycoproteins of the<br />

major histocompatibility complex (MHC). Things that<br />

may give rise to these tumor-associated peptides are<br />

enhanced or diminished expression of some normal proteins<br />

and the new expression of altered or normally<br />

repressed molecules. Less frequently these antigens are<br />

tumor-specific as they derive from mutated proteins.<br />

Lastly, various TAA are shared by tumors with distinct<br />

histology and origin (Table 1). Telomerase catalytic subunit<br />

looks like another widely expressed TAA recognized<br />

by T-lymphocytes. It is markedly activated in most<br />

human tumors while it is silent in normal tissues. 21<br />

Why?<br />

The central tenet of antitumor vaccination is that the<br />

immune system is able to destroy tumor cells and to<br />

retain a long-lasting memory provided that TAA are first<br />

efficiently recognized. While the studies aimed at the<br />

definition of TAA progressed quickly, investigations of<br />

lymphocyte receptors and their idiotype network, costimulatory<br />

molecules, and cytokines were leading to a<br />

more exact description of the requirements for the<br />

induction of an immune response. 22 Finally, technical<br />

refinement of genetic engineering is making the development<br />

of new cancer vaccines easier . 23 The convergence<br />

of these issues is once again placing antitumor<br />

vaccination at the cutting edge of biological research.<br />

A survey by Science 24 indicates that antitumor vaccination<br />

is expected soon to become an established therapeutic<br />

option.<br />

When?<br />

Whereas individuals are immunized with microbial<br />

vaccines prior to encountering the pathogen, cancer<br />

patients have to be immunized when a tumor has been<br />

already detected. It is not yet possible to predict which<br />

combination of gene mutations will give rise to cancer.<br />

Therefore, the common clinical setting is elicitation of<br />

an immune response in a tumor-bearing patient, rather<br />

than prior to tumor development. The very concept of<br />

vaccine is somewhat distorted since it has moved from<br />

being preventive to being therapeutic. 23<br />

The kind of patients who should be considered eligible<br />

for tumor vaccination is not a minor issue. In many<br />

trials patients with advanced diseases are enrolled both<br />

for compassionate reasons and because of the constraints<br />

imposed by ethical considerations. But, do experimental<br />

data suggest that vaccination could be effective<br />

in advanced stages of neoplastic progression? The experimental<br />

data provide an unambiguous picture of the<br />

potentials and limits of vaccination. This picture is not,<br />

however, generally taken into account. Perhaps unconscious<br />

reasons lead to experimental data being assessed<br />

with optimistic superficiality. 25 Many experimental studies<br />

have shown that an antitumor response can be elicited<br />

by new vaccines. This results in strong resistance to<br />

a subsequent tumor challenge and inhibition of minimal<br />

residual disease remaining after convention therapy. The<br />

pitfall hidden in the evaluation of these vaccine-re-challenge<br />

experiments is that successful immunization of<br />

healthy mice against a subsequent re-challenge with<br />

tumor cells does not demonstrate a true therapeutic<br />

effect. 26 Examination of more realistic studies of the ability<br />

of vaccines to cure existing tumors shows that only<br />

a minority of tumor-bearing mice could be cured. Furthermore,<br />

the limited therapeutic efficacy of vaccines<br />

was lost when they were not given in the first few days<br />

after the implantation of tumor cells. 26<br />

A similar picture is emerging from phase I studies on<br />

vaccination of cancer patients. The vaccination is safe,<br />

but the results suggest that only a minority of patients<br />

(about 10%) display an objective response. The immunologic<br />

performance status of these patients is obviously<br />

suboptimal for this type of therapy. Even so, one would<br />

have expected a greater number of responders to support<br />

the promise of new sophisticated vaccines. 23<br />

Therapeutic vaccination has not had much success in<br />

the management of infectious diseases. Its use against<br />

the progression of an established tumor is very challenging,<br />

since it must secure an effective immune<br />

response capable of getting the better of a well-established,<br />

proliferating tumor. Of the several objectives that<br />

Table 1. Cross-expression of some tumor-associated antigens<br />

among histologically different human tumors from distinct<br />

organs.<br />

bladder BAGE GAGE<br />

breast BAGE MAGE CEA p53 ras MUC-1<br />

colon CEA p53 ras<br />

lung BAGE CEA p53<br />

melanoma BAGE GAGE MAGE ras<br />

pancreas CEA p53 ras MUC-1<br />

sarcomas<br />

GAGE<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

159<br />

have been made approachable by antitumor vaccines,<br />

the cure of advanced tumors is both the most difficult<br />

and at the same time the most common in clinical trials.<br />

The strong emotions kindled by cancer suffering provide<br />

the main justification for these attempts. 27 Perhaps,<br />

major improvements in antitumor vaccination will make<br />

this goal approachable. However, the data reviewed<br />

show that this is far from the present reality.<br />

Nevertheless it should be considered that most tumor<br />

lethality depends on a few neoplastic cells remaining<br />

after surgical excision of a tumor mass or after having<br />

escaped direct killing by chemo- and radiotherapy. Many<br />

experimental findings 28-30 suggest that a stage of minimal<br />

residual disease is one in which it is possible to<br />

foresee a significant cure by immunization. After successful<br />

conventional management the tumor burden<br />

may be low, and the tumor may reappear after a long<br />

dormancy. This is a realistic setting in which vaccination<br />

could lead to the induction of anti-tumor immunity<br />

capable of extending the survival of patients. As there<br />

are grounds for believing that antitumor vaccines could<br />

be used as an effective anticancer tool, the purpose of<br />

this review is to describe the types of vaccines that are<br />

being experimented, emerging clinical results and the<br />

new perspectives opened by this scientific endeavor.<br />

Common tools<br />

Cytokines and cellular signals<br />

The immunologic attempt of the immune system to<br />

prevent the development of a neoplastic disease may<br />

be ineffective due to either a lack of immunogenicity of<br />

tumor cells, or to a weak reaction unable to contrast the<br />

neoplastic proliferation. In both cases, it is likely that<br />

most of the physiologic mechanisms of priming of the<br />

immunologic effector cells may be impaired or absent.<br />

In fact, initiation of immune responses requires that<br />

professional antigen-presenting cells (APC) deliver a first<br />

signal to T lymphocytes through the binding of the T-cell<br />

receptor by the peptide enclosed in the HLA molecule,<br />

that is responsible for the specificity of the immune<br />

response, and a second or co-stimulatory signal that is<br />

not antigen-specific but it is required for T-cell activation<br />

31,32 mainly through CD80 (B7-1) and CD86 (B7-2)<br />

binding to CD28 receptor, or the CD40:CD40L pathway.<br />

Moreover, the capacity of dendritic cells (DC) to activate<br />

natural killer (NK) cells by ligation of the CD40 molecule<br />

with its counter-receptor has recently been demonstrated.<br />

33,34 Immunocompetent cells may also determine<br />

the type of immune response by the expression of<br />

chemokines and by the release of pro-inflammatory, or<br />

anti-inflammatory cytokines which drive T-cells to different<br />

activities or even to suppression. 35<br />

Therefore, given the complex network of regulatory<br />

signals by professional APC and naive and memory lymphocytes<br />

occurring in antigen-specific immune responses,<br />

it is not surprising that tumor cells may fail to induce<br />

efficient humoral and cellular immune reactions even<br />

when a well known TAA is present. In this review, several<br />

strategies to overcome the immune escape mechanisms<br />

of tumor cells will be considered, such as the<br />

direct use of TAA to elicit specific reactions, the use of<br />

dendritic cells to present TAA in order to enhance the<br />

immune response, and the use of tumor cells genetically<br />

modified to function as professional APC or to release<br />

soluble factors. Animal models have been widely used<br />

for many years to demonstrate the effect of different<br />

cytokines, added to or secreted by tumor cell-based vaccines,<br />

in increasing the in vitro and in vivo cytotoxicity<br />

against tumor challenge. The role of the main cytokines<br />

involved in activation of humoral and cellular immune<br />

responses is represented in Figure 1. On the basis of<br />

cytokine functions it has been previously shown in<br />

experimental models that: the number of APC in the<br />

site of tumor infiltration can be increased by cytokines<br />

such as granulocyte-macrophage colony-stimulating<br />

factor (GM-CSF) and interleukin (IL)-4, which should<br />

allow also the differentiation of DC precursors; B- and<br />

T-cell responses are potently increased by IL-2, IL-12, or<br />

GM-CSF; 36 and in particular T-cell cytotoxicity is<br />

enhanced by GM-CSF, IL-2, IL-12, interferon (IFN-γ),<br />

tumor necrosis factor (TNF-α), while NK activity is<br />

enhanced by IL-12 or FLT 3-L. 36,37 However, different<br />

models and different TAA resulted in controversial findings.<br />

Furthermore, since these cytokines are likely to be<br />

more effective when released within the tumor area,<br />

the transduction of cytokine genes into tumor cells and<br />

their use as cellular vaccines after irradiation has been<br />

tested in animal models and in humans. 38-40<br />

Initial clinical experiences in patients with advanced<br />

melanoma or renal cell carcinoma suggest that tumor<br />

cell-based vaccines, either engineered to produce GM-<br />

CSF or IL-12 or with exogenous GM-CSF, may facilitate<br />

marked infiltration of DC and CD4 + and CD8 + T-lymphocytes<br />

into tumor lesions, potentially improving the<br />

antitumor effect. These data provided evidence of the<br />

feasibility of the approach but were unlikely to be able<br />

to address the point of efficacy, due to the large tumor<br />

burden of these patients. Future immunotherapy<br />

attempts should, in fact, focus on the possibility of eradicating<br />

minimal residual disease. More recent data<br />

demonstrated the role of GM-CSF as a useful adjuvant<br />

in peptide-based vaccines in ovarian and breast cancer<br />

and in follicular lymphoma, as will be described later in<br />

this review.<br />

Figure 1 also shows that cell-to-cell contact via<br />

CD40:CD40L plays a pivotal role in activating specific T-<br />

cell, B-cell and NK-cell responses. On the other hand, T-<br />

cell tolerance can be obtained by blockade of CD40L<br />

receptor in non-human primates undergoing solid organ<br />

allogeneic transplantation, and in mice receiving either<br />

allogeneic bone marrow or solid organ transplantation.<br />

41-43<br />

Recent experiments demonstrated that stimulation, via<br />

an activating anti-CD40 antibody, resulted in the activation<br />

of host APC and could convert lymphocytes of<br />

mice treated with a tolerogenic peptide vaccine into<br />

cytotoxic T- cells. Moreover, this stimulation induced the<br />

haematologica vol. 85(suppl. to n. 12):December 2000


160<br />

M. Bocchia et al.<br />

regression of established tumors that had not been<br />

affected by previous vaccination alone, 44,45 thus showing<br />

that triggering the CD40 molecule may both overcome T-<br />

cell tolerance in a tumor-bearing animal and greatly<br />

potentiate a peptide-based vaccine. Moreover, gene<br />

transfer by an adenovirus vector of CD40L in human B-<br />

cell chronic lymphocytic leukemia (B-CLL) allowed the<br />

activation of bystander non-infected B-CLL cells that<br />

upregulated co-stimulatory molecules such as CD80 and<br />

CD86 and stimulated autologous cytotoxic T-cells. 46<br />

Another important issue concerns the way T-cells are<br />

turned off by CTLA-4 receptor following activation via<br />

CD28. In fact, both CD28 and CTLA-4 bind with high<br />

affinity to CD80 and CD86 and CTLA-4 physiologically<br />

blocks T-cell activation. In the case of antitumor T-cell<br />

response it has been demonstrated that blockade of negative<br />

regulatory signals by an anti-CTLA-4 monoclonal<br />

antibody may retard tumor growth in experimental systems.<br />

47,48 More recent studies in mice suggested that this<br />

molecule was extremely efficient in causing tumor<br />

regression when used in combination with subtherapeutic<br />

doses of melphalan, or with a GM-CSF-expressing<br />

tumor. 49,50<br />

Altogether, these studies strongly support the role of<br />

cytokines or immunomodulatory molecules in anticancer<br />

vaccine strategies. However, they do not clarify<br />

whether a strict T-helper (Th1) response is required to<br />

achieve tumor killing, or whether a humoral response<br />

induced by anti-inflammatory cytokines should also be<br />

pursued. Finally, future directions of anticancer vaccine<br />

research are likely to deal with monoclonal antibodies<br />

enhancing or blocking specific receptors.<br />

Dendritic cells as initiators of immune<br />

response<br />

Dendritic cells (DC) represent a heterogeneous population<br />

of leukocytes defined by morphologic, phenotypic<br />

and functional criteria which distinguish them from<br />

monocytes and macrophages. 32 From among the professional<br />

APC, DC are the most potent stimulators of T-<br />

cell responses and play a crucial role in the initiation of<br />

primary immune responses. 32<br />

The DC system comprises at least three distinct subsets,<br />

including two within the myeloid or non-lymphoid<br />

lineage, and a third represented by lymphoid DC. 32,51<br />

There is also a continuum of differentiation within each<br />

of these subsets, from precursors circulating through<br />

blood and lymphatics, to immature DC resident in<br />

peripheral tissues, to mature or maturing forms in the<br />

thymus and secondary lymphoid organs. Recent studies<br />

have focused on the different roles of lymphoid and<br />

myeloid DC: more resident lymphoid DC induce tolerance<br />

to self, whereas migratory myeloid DC, including<br />

Langherans cells, are activated by foreign antigens in the<br />

periphery and move to lymphoid organs to initiate an<br />

immune response. 32<br />

DC have always been described as having two distinct<br />

functional stages: 1) immature, with high antigen<br />

uptake and processing ability, and poor T-cell stimulatory<br />

function; 2) mature, with high stimulatory function<br />

and poor antigen uptake and processing ability. Bacterial<br />

products such as lipopolysaccharides, and inflammatory<br />

cytokines such as IL-1, TNF-α, type I interferons<br />

(IFN α or β) and prostaglandin E2 (PGE2) stimulate DC<br />

maturation, whereas IL-10 inhibits it. 52 Interestingly,<br />

Figure 1. Principal cytokines<br />

involved in the antitumor immune<br />

response.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

161<br />

human and murine DC upregulate the synthesis of HLA<br />

class I and II molecules, and B7-1, B7-2 and CD40 molecules,<br />

after ingestion of bacteria or bacterial products,<br />

such as lipopolysaccharides, can prime naive T-cells.<br />

An emerging concept is that APC activate T-helper (Th)<br />

cells not only with antigen and co-stimulatory signals,<br />

but also with a polarizing signal (signal 3). This signal<br />

can be mediated by many APC-derived factors, but IL-12<br />

and PGE2 seem to be of major importance. As for Th cells,<br />

APC can be functionally polarized. In vitro experiments<br />

with monocyte-derived DC showed that the presence of<br />

IFN-γ during activation of immature DC induces mature<br />

DC with the ability to produce large quantities of IL-12<br />

and, consequently, a Th1-driving capacity (APC1 or DC1).<br />

In contrast, PGE2 primes for a low IL-12 production ability<br />

and Th2-driving capacity (APC2 or DC2). 32,53 DC-stimulated<br />

CD4 + cells upregulate CD40L/ CD154 that reciprocally<br />

activates DC via CD40. This renders DC more<br />

potent stimulators of CD8 + cytotoxic T-cell (CTL). 54 This<br />

novel concept is in contrast to simultaneous stimulation<br />

of CD4 + and CD8 + T-cells by DC, whereby the CD4 + T-<br />

cells secrete helper lymphokines in support of CD8 + CTL<br />

development. 55 Together with CD40 L and CD40, two different<br />

groups have discovered another paired member of<br />

the TNF-TNF receptor family. This factor, termed TNFrelated<br />

activation induced cytokine (TRANCE) or receptor<br />

activator of NF-kB ligand (RANK-L), is expressed by T<br />

cells. 56 Its corresponding receptor, receptor activator of<br />

NF-kB (RANK7) or TRANCE R, is expressed by mature DCs<br />

but not on freshly isolated B cells, macrophages, or T<br />

cells. 57 Ligation to this receptor causes either activation<br />

of T-lymphocytes or enhancement of DC survival. In addition,<br />

IL-12 is a critical mediator of DC-supported differentiation<br />

of naive, but not memory, B-cells, 58 indicating<br />

that direct interactions occur between DC and B cells,<br />

apart from those that occur via cognate CD4 + T-cell help.<br />

Lastly, it should be mentioned that the primary and<br />

secondary B cell follicles contain another population of<br />

DC, the follicular DC (FDC). The origin of these cells is<br />

not clear, and most investigators believe that they are<br />

not leukocytes. FDC trap and retain intact native antigen<br />

as immune complexes for long periods of time, present<br />

it to B cells and are likely to be involved in the affinity<br />

maturation of antibodies, the generation of immune<br />

memory and the maintenance of humoral immune<br />

responses. 59<br />

In conclusion, there is a general agreement in considering<br />

DC as very important players in the game of<br />

immune responses against foreign antigens either of<br />

infectious agents or of neoplastic cells. Many developing<br />

immunotherapeutic strategies against danger antigens<br />

are aimed at exploiting the powerful antigen-presenting<br />

properties of DC by an in vivo or ex vivo engineering<br />

of the DC system. In fact, subcutaneous or intramuscular<br />

injection of antigens relies on the local recruitment<br />

and activation of DC to capture and present antigens to<br />

the immune system. Although the techniques for targeting<br />

tumor antigens to DC in situ might eventually obviate<br />

the need for ex vivo manipulation of DC, novel methods<br />

for ex vivo generation and activation of large<br />

amounts of human DC have been developed.<br />

Antitumor vaccination: types<br />

and formulations<br />

Killing for priming and killing to destroy<br />

The way cell vaccines die when injected in vivo influences<br />

DC loading. The initial activity of cytokines transduced<br />

into the cell vaccine is to select the leukocyte type<br />

recruited and stimulated at the site of injection. Tumor<br />

cells are killed by effectors of the innate response; NK<br />

and polymorphonuclear cells also produce secondary<br />

cytokines that set up a local inflammation recruiting DC,<br />

whereas T-cell response is activated later. Of note, gene<br />

engineering allows the manipulation of the first phase of<br />

the process through the choice of cytokine and/or cofactor<br />

and by deciding the level of cytokine to be produced.<br />

Once the infiltrate leukocytes are activated, their<br />

response to the triggering cytokine is physiologic and<br />

independent. They produce other cytokines thus amplifying<br />

the magnitude and the complexity of response. The<br />

initial stage is finalized to T- and B-cell activation, killing<br />

of cell vaccine is to provide the antigen(s) and the<br />

inflammatory response should provide the right environment<br />

for such activation. 60<br />

The final stage is aimed at the destruction of existing<br />

tumor. Specific immune response is often insufficient<br />

to fight solid tumor nodules. Among the possible causes,<br />

immunosuppression, low effector-target ratio, MHC<br />

downregulation on tumor cells are the most common.<br />

Animal studies have shown that these problems can be<br />

overcome by general inflammation associated with neutrophil<br />

influx. This combination may destroy the tumorassociated<br />

blood vessels 61 in a way that may resemble<br />

tissue damage in vasculitis. In this setting a specific<br />

immune response is not directly responsible for tumor<br />

elimination but should be strong enough to at least<br />

begin and direct the inflammatory response to the tumor<br />

site. In this perspective, tumor vessels are the main target<br />

of a non-specific immune response, their functional<br />

impairment increasing the effect of either T- or B-<br />

cell-mediated specific immune responses.<br />

An add to DC-common link?<br />

Although DC have been indicated as central in alerting<br />

and activating the immune system, it is now clear that<br />

certain peptides are not and can not be presented by<br />

mature DC. This observation, made by Van den Eynde and<br />

colleagues, 62 concerns autoantigens and T lymphocytes<br />

that recognize them without being deleted in the thymus<br />

and normally without provoking autoimmunity. In fact,<br />

APC differ from other cell types by the proteosome that<br />

digests protein into immunogenic peptides to fit the MHC<br />

groove. APC immunopreoteosomes have three catalytic<br />

subunits substituted by those induced by IFN-γ, thus generating<br />

slightly different peptides from those generated by<br />

non-APC cells.<br />

Several self-antigens identified as tumor-associated<br />

because of CTL recognition may not be processed by<br />

haematologica vol. 85(suppl. to n. 12):December 2000


162<br />

M. Bocchia et al.<br />

immunoproteosome. The implication is that such CTL were<br />

not generated through DC presentation or at least not<br />

through DC processing unless this happened during transition<br />

from immature to mature DC. 63 Perhaps free peptides<br />

can be captured on the surface of DC for presentation,<br />

or perhaps other as yet unknown mechanisms are<br />

involved.<br />

Genetically modified tumor cell vaccines<br />

Old and recent discoveries confirm the possibility of a<br />

cancer vaccine made of tumor cells.<br />

An empirical approach, such as the use of allogeneic<br />

whole-cell vaccine composed of 3 allogeneic melanoma<br />

cell lines established in vitro, allowed a 3-fold increase of<br />

the five-year survival of patients with stage IV melanoma<br />

as reported by Morton et al. 64 The most active component<br />

of Morton’s vaccine has been identified by Livingston and<br />

colleagues 65 to be a ganglioside (GM2). Patients who<br />

develop antibody response to ganglioside showed a significant<br />

survival advantage. Whether a CTL response was<br />

also activated has not been investigated but does probably<br />

exist. However, this finding prompted a phase III study<br />

in patients with stage III disease.<br />

In the autologous setting, irradiated melanoma cells<br />

were modified with dinitrophenyl and used to treat<br />

patients with metastatic disease. Clinical evidence of<br />

inflammatory response to superficial metastases was<br />

reported. The same treatment administered to phase III<br />

patients who remain tumor-free after resection of lymph<br />

node metastases has resulted in 50 and 60% 4-year<br />

relapse-free and overall survival, respectively. 66 Immunostaining,<br />

TCR repertoire analysis and functional data of<br />

node-metastases post-vaccination have shown that treatment<br />

with autologous dinitrophenyl-modified melanoma<br />

cells can expand certain T-cell clones at the tumor site. 67<br />

The above results bring up two issues: autologous versus<br />

allogeneic tumor cells and chemical or genetic (see<br />

below) modifications of tumor cells to be used as cancer<br />

vaccines.<br />

Before discussing these issues we should, however,<br />

address a more basic question, that is how to use cancer<br />

cells as vaccine. Well before identification of tumor antigens,<br />

their existence was inferred in melanoma on the<br />

basis of expansion and characterization of cytotoxic T-<br />

lymphocytes recognizing autologous tumor, 68 whereas<br />

antibodies against a variety of tumor types were isolated<br />

from patient sera. Antigens recognized by CTL can be<br />

tumor-specific and even restricted to the autologous<br />

tumor or cross-react among different neoplasms of the<br />

same or different tissue origin depending on the mechanism<br />

generating such an antigen: point mutation, incorrect<br />

splicing, over-expression, translocation and other (see<br />

Table 2). The number of tumor antigens is always increasing<br />

thus suggesting that our knowledge of the antigenic<br />

repertoire of tumor cells is partial. In addition, which antigen<br />

among those already characterized should be used in<br />

a vaccine?<br />

These problems can be solved altogether by using tumor<br />

cells that would represent the entire antigenic repertoire<br />

with a single drawback, that is immunoselection of certain<br />

antigens. Tumor cells from different patients may<br />

undergo different processes of immunoselection and<br />

therefore a pool of these tumors would be ideal for preparing<br />

an allogeneic vaccine. The finding that antigens<br />

belonging to allogeneic cells are processed by host antigen-presenting<br />

cells, 69 so that they can be recognized by<br />

host T-lymphocytes (a phenomenon called cross-priming),<br />

removes any conceptual obstacles to the use of allogeneic<br />

cells. Allovaccines have several advantages over autologous<br />

vaccines: cell lines that are extensively characterized<br />

in vitro can be used to treat all the patients included<br />

in a clinical study. Genetic modifications of tumor cells<br />

have been widely studied as a way to increase immunogenicity.<br />

These modifications can be easily made to a cell<br />

line, thus avoiding the need to isolate cells from every<br />

tumor. The rate of success in culturing tumor cells from a<br />

primary tumor varies according to the type of tumor and<br />

experience of the operator. Transduced cell lines can be<br />

selected for production of a certain amount of transgene<br />

thus assuring that the same vaccine will be given to all<br />

patients. Which modification is most efficient in inducing<br />

tumor immunity has not been unequivocally determined<br />

since variability among different tumors has been<br />

described. Chemical modification, also called haptenization,<br />

aims at adding helper determinant to tumor cells, 70<br />

although the exact mechanism and the downstream pathways<br />

activated in this way are not clearly understood.<br />

Genetic modification is now preferred since the effect of<br />

several cytokines and co-stimulatory molecules are known<br />

at molecular level. Their genes can be transduced into<br />

tumor cells that acquire new immunoregulatory functions.<br />

In this way a desired immune response can be fine-tuned<br />

through gene dosage, recruitment of certain cell types,<br />

deflection of Th1 or Th2 type of response and several others<br />

mechanisms. 60<br />

To summarize the most recent and promising<br />

approaches we may consider two strategies aimed at<br />

favoring vaccine interaction with DC or directly with T<br />

lymphocytes. The two approaches can be viewed for<br />

priming or boosting (Figure 2). A combination of<br />

cytokine and co-stimulatory factors is likely to be synergistic<br />

as generally occurs when soluble and cell-contact<br />

signals are given together. Tumor cells transduced<br />

with both GM-CSF and CD40L have been shown to be<br />

heavily infiltrated by DC. GM-CSF induces proliferation<br />

and maturation of hematopoietic cells, and has been<br />

shown to stimulate DC accessory properties and<br />

enhance the immune response initiated by these cells. 71<br />

The CD40/CD40L interaction plays a critical role in<br />

cell-mediated immunity, and in proliferation and activation<br />

of APC, as shown for B cells, monocytes and, more<br />

recently, DC. 72 Ligation of CD40 on monocytes and DC<br />

results in the secretion of several cytokines, including IL-<br />

1, IL-6, IL-12, and TNF-α. The murine tumor transduced<br />

with both GM-CSF and CD40L genes showed that tumor<br />

infiltrating DC can take up cellular antigen from the<br />

tumor and can present it to T lymphocytes in vitro. 73 In<br />

this setting, DC bridges cell vaccine-T-lymphocytes<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

163<br />

interaction and can be envisaged as the way to prime<br />

patients against weaker antigens otherwise ignored by<br />

the immune system. A complementary approach would<br />

consider the possibility of boosting the primed or the<br />

existing immune response. In this case DC that reencounter<br />

activated T-cells can be lysed and may not be<br />

appropriate for boosting. Boosting can be done using<br />

tumor cells transduced with IL-2 and B7-1 such as to<br />

provide both cell expansion and co-stimulation from<br />

the tumor cell vaccine directly to T lymphocytes (Figure<br />

2). The way cellular antigen can be captured by DC for<br />

priming of T-lymphocytes depends on how tumor cells<br />

die after injection. Irradiation of the cell vaccine induces<br />

apoptotic cell death and apoptotic bodies can be captured<br />

by DC. 74 Others suggest that peptides of cellular<br />

protein complexed to heat shock protein (HSP), the natural<br />

chaperon of peptide from proteosome to membrane-associated<br />

TAP, leave the cells upon necrosis to<br />

be taken up by DC. 75,76 DC loading must be followed by<br />

DC maturation and migration to the lymph node to<br />

ensure correct antigen presentation. 32<br />

The way cytokines and co-signals modulate vaccinehost<br />

interactions may determine the extent and the efficacy<br />

of treatment. Systemic activation of the immune<br />

response (T-cell cytotoxicity, antibodies) is easy to measure<br />

but can not be used as a read-out system to predict<br />

whether the tumor will be rejected. Tumor nodules might<br />

be reached by circulating lymphocytes which, however,<br />

may be neutralized because of either immunosuppression<br />

or peripheral tolerance. The former is likely dependent<br />

on tumor size and is less expected when a small<br />

tumor, minimal residual disease or prevention of recurrence<br />

is the target to be treated. The latter could be surmounted<br />

by appropriate co-signals, for example CD40L. 45<br />

Soluble proteins as immunogens<br />

Soluble proteins derived from autologous cancer cells<br />

are not as immunogenic as proteins derived from infectious<br />

agents. The degree of foreignness, which depends<br />

on the reciprocal distribution between epitopes subject<br />

and not subject to self-tolerance, is low in TAA. Moreover,<br />

TAA do not have the particulate nature of infectious<br />

agents that is a key factor in increasing their<br />

immunogenicity. Finally, infectious agents are rich in<br />

molecules that are able to raise immune responsiveness<br />

without being themselves immunogenic. A significant<br />

effort has been made over the last few years to translate<br />

the immunogenic properties of infectious agents<br />

into cancer vaccines for clinical use. The most suitable<br />

TAA should be directly involved in the malignant behavior<br />

of tumor cells; contain multiple immunodominant B<br />

cell and T cell epitopes, including both helper and cytotoxic<br />

T-cell epitopes; contain a very high degree of foreignness;<br />

be unprotected by self-tolerance mechanisms.<br />

Of course, HLA haplotype remains a major constraint in<br />

determining whether TAA-derived immunodominant<br />

peptides can elicit tumor-specific immune responses in<br />

a given individual (see also below).<br />

Table 2a. Tumor-associated antigens recognized in class I<br />

HLA restriction.<br />

Antigen defined Type of antigen Type of tumor HLA-restriction<br />

and distribution<br />

allele<br />

gp 100 Melanocyte diff. Melanoma A2, A3, A24, Cw8<br />

Melanocortin receptor 1 Melanocyte diff. Melanoma<br />

MART-1/MelanA Melanocyte diff. Melanoma A2, B45<br />

Tyrosinase Melanocyte diff. Melanoma A1, A2, A24, B44<br />

TRP-1 (gp 75) Melanocyte diff. Melanoma A31<br />

TRP-2 Melanocyte diff. Melanoma A2, A31, Cw8<br />

p15 Widely expressed Melanoma A24<br />

SART-1 Widely expressed Lung carcinoma A2601<br />

PRAME Widely expressed Melanoma, renal A24<br />

NAG-V* Melanoma sheared Melanoma A2.1<br />

β catenin Unique tumor specific Melanoma A24<br />

CDK4-Kinase Unique tumor specific Melanoma A2<br />

MUM-1 Unique tumor specific Melanoma B44<br />

TRP2/INT2 Unique tumor specific Melanoma A6801, Cw8<br />

CASPASE-8 Unique tumor specific Head/neck cancer B35<br />

HLA-A*201 mutated Unique tumor specific Renal cancer A2.1<br />

KIAA0205 Unique tumor specific Bladder cancer B44*03<br />

BAGE Cancer/ testis Melanoma Cw 1601<br />

GAGE-1/2 Cancer/ testis Melanoma Cw 6<br />

MAGE-1 Cancer/ testis Melanoma A1, Cw16<br />

MAGE-3 Cancer/ testis Melanoma A1, A2, B44<br />

RAGE Cancer/ testis Renal cancer B7<br />

NY-ESO-1 Cancer/ testis Melanoma, ovarian, A2<br />

esophageal cancer<br />

K-RAS-D13 mutated Shared tumor-specific Colon cancer A2.1<br />

p53 mutated Shared tumor-specific Colon and lung A2.1<br />

Bcr/abl Shared tumor-specific CML A2.1, A3,<br />

A11, B8<br />

MUC-1 Shared tumor-specific Breast, colon, A11<br />

pancreatic cancer<br />

HPV16E7 Viral related Cervical cancer A2.1<br />

Table 2b. Human tumor antigens recognized by HLA class<br />

II-restricted CD4 + T cells.<br />

Antigen Tissue distribution Class II restriction<br />

Tyrosinase Melanoma/melanocytes DRb1*0401<br />

Tyrosinase Melanoma/melanocytes DRb1*1501<br />

Triosephosphate isomerase Melanoma, unique DRb1*0101<br />

mutated form<br />

MAGE-3 Melanoma and other tumors, testis DRb1*1301<br />

MAGE-1, -2 or -6 Melanoma and other tumors, testis DRb1*1301<br />

MAGE-3 Melanoma and other tumors, testis DRb1*1101<br />

RAS-D12 mutated Colon and pancreatic cancer DR1<br />

HER-2/neu Breast and ovarian cancer DR11<br />

PML/RARα Acute promyelocytic leukemia DR2<br />

Bcr/abl Chronic myeloid leukemia DR1, DR4, DR11<br />

CDC27 Melanoma DR4<br />

*N-acetylglucosaminyl transferaseV. CML: chronic myeloid leukemia.<br />

Building up immunogenicity of soluble proteins<br />

Even the most suitable TAA is not immunogenic unless<br />

it is processed and presented by professional APC to the<br />

host immune system. To this aim, TAA should interact<br />

with APC directly. The route of administration and adjuvants<br />

are key factors in determining the final outcome<br />

haematologica vol. 85(suppl. to n. 12):December 2000


164<br />

M. Bocchia et al.<br />

Figure 2. Priming of DC and boosting of T-cell responses by transduced tumor cells as vaccine.<br />

of immunization. Intravenous injection of soluble antigens<br />

induces tolerance, whereas subcutaneous or intramuscular<br />

injection of the same antigens results in immunity<br />

because of the interaction with epidermal Langerhans<br />

cells or dermal dendritic cells. Accordingly, delivery<br />

of antigens via mucosal surfaces may result in immunity<br />

because antigens may interact with the numerous DC<br />

located just beneath the epithelium of mucosal lymphoid<br />

organs. The association between soluble antigens and<br />

APC is made much more intense by adjuvants. Adjuvants<br />

act via different principles and pathways, but the common<br />

goals are to prolong the interaction with APC by<br />

promoting a slow antigen release, and functionally activate<br />

APC themselves by delivering danger signals.<br />

Cytokines have also emerged as potent immunoadjuvants<br />

since they can influence the immune responses at<br />

different levels (see above).<br />

Particulation of soluble proteins<br />

Precipitation with aluminium hydroxide or aluminium<br />

phosphate has been used to particulate antigens in diphtheria,<br />

tetanus, hepatitis B, and other vaccines. These<br />

types of vaccines induce antibody formation, but very little<br />

delayed cutaneous hypersensitivity (DTH) or cell-mediated<br />

cytotoxicity. In a pilot study, five stage I-III patients<br />

with multiple myeloma were immunized with autologous<br />

idiotype (Id) precipitated in aluminium phosphate suspension.<br />

Three patients developed idiotype-specific T-<br />

and B- cell responses, but these responses were transient<br />

and their amplitude was low. 77<br />

The use of immunostaining complexes (ISCOMs) is<br />

another strategy that has been used to particulate antigens.<br />

ISCOMs are cagelike structures made of cholesterol,<br />

saponin, phospholipid, and viral envelope proteins<br />

to which other proteins can be associated. Saponin is a<br />

plant derivative that is critical to the efficacy of ISCOMs.<br />

QS21 is the most effective fraction of saponin and is<br />

currently under clinical investigation in several trials. 78<br />

ISCOMs may reach the endocytic pathway and induce<br />

DTH and CTL responses other than antibody production.<br />

Liposomes, virosomes, and proteasomes are alternative<br />

strategies to ISCOMS. Experimental data have recently<br />

shown in the 38C13 mouse B-cell tumor that liposomal<br />

formulation of autologous Id converts this weak selfantigen<br />

into a potent tumor rejection antigen. 79<br />

Promoting slow release of soluble antigens<br />

A slow release of antigen is one of the major goals of<br />

adjuvants. Antigen polymerization and emulsifying agents<br />

have been put together to achieve this goal. Polymerization<br />

can be obtained by association with non-ionic block<br />

polymers or by association with carbohydrate polymers.<br />

Non-ionic block polymers have been used as components<br />

of water-in-oil or oil-in-water emulsions. SAF-1 (Syntex<br />

Adjuvant Formulation-1) is an oil-in-water adjuvant formulation<br />

containing non-ionic block polymers, squalene,<br />

and Tween 80. SAF-1 has been used by Kwak et al. in their<br />

pioneering study on idiotype vaccination in follicular lym-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

165<br />

phoma patients. 80 Chemical immunomodulators such as<br />

derivatives of muramyl dipeptide, the smallest subunit of<br />

the mycobacterial cell wall that retains immunoadjuvant<br />

activity, can be added to oil-in-water adjuvants. 81 This<br />

approach was used in the study by Hsu et al. in which idiotype/KLH<br />

conjugates were delivered to follicular lymphoma<br />

patients after mixing with SAF-165 that contained<br />

muramyl dipeptide as immunomodulator. 82 Lipid components<br />

of bacteria have also been used as adjuvants in<br />

experimental models. These are portions of the LPS endotoxins<br />

of Gram-negative bacteria. These molecules are,<br />

however, too toxic and chemically modified derivatives<br />

have been developed for clinical use. So far, monophosphoryl<br />

lipid A is the least toxic derivative capable of promoting<br />

cell-mediated immunity. 83<br />

Polysaccharide polymers have also been used as a sustained-release<br />

vehicle for TAA. Poly-N-acetyl glucosamine<br />

is a highly purified, biocompatible polysaccharide<br />

matrix that has recently become available for<br />

this purpose. 84<br />

Peptide vaccines<br />

T lymphocytes recognize small peptides that represent<br />

the degradation products of a complex intracellular<br />

process and are presented on the cell surface complexed<br />

to 1 of 2 types of histocompatibility leukocyte<br />

antigen molecules (HLA classes I or II). CTLs (CD8 + ) mainly<br />

recognize peptides of 8 to 10 amino acids derived<br />

from intracellular or endogenous proteins and complexed<br />

to HLA class I molecules. 85-89 CD4 + T- lymphocytes<br />

recognize exogenous proteins which are ingested<br />

by APC, degraded to peptides of 12-24 amino acids and<br />

complexed to HLA class II molecules. 90,91<br />

Class I and Class II peptides that are presented on the<br />

cell surface, although randomly derived from the original<br />

protein, must contain specific amino acids in 1 or 2<br />

critical positions in order to be able to bind the appropriate<br />

HLA molecules. Thus, peptide binding is HLArestricted.<br />

The amino acid motifs responsible for the<br />

specific peptide-binding to HLA class I and class II molecules<br />

have been determined for the common HLA types<br />

by analyzing acid-eluted naturally processed peptides<br />

and by using cell lines defective in intracellular peptide<br />

loading and processing. 92-94<br />

Several tumor-specific and some leukemia-specific<br />

peptides have so far been identified, and studies aimed<br />

at evaluating the potential clinical benefit of peptide vaccines<br />

in cancer patients have begun. Among the reasons<br />

that make a peptide vaccine strategy interesting are several<br />

unique advantages that peptide immunization offers<br />

over other vaccine approaches: 1) peptide vaccines permit<br />

specific targeting of the immune response against 1<br />

or 2 unique antigens (thus limiting the potential autoimmune<br />

cross-reactivity or immunosuppressive activity<br />

often observed with more complex immunogens); 2)<br />

emerging technology has made it simple, rapid and inexpensive<br />

to sequence and prepare larger quantities of<br />

tumor antigen peptides for both laboratory and clinical<br />

use; 3) use of synthetic peptides greatly reduces the possible<br />

risk of bacterial or viral contamination that might<br />

derive from autologous or allogeneic tissue for immunization.<br />

On the down side, the main disadvantages of<br />

peptide immunization are: 1) lack of universal applicability<br />

as each peptide is restricted to a single HLA molecule;<br />

2) poor immunogenicity of most native peptides; 3)<br />

risk of inducing antigenic tolerance. Successful attempts<br />

to enhance HLA binding affinity have been based on synthetically<br />

generating peptides with amino acid deletions<br />

or substitutions while maintaining antigen specificity. 95,96<br />

In initial studies, synthetic substituted peptides appeared<br />

to enhance immunogenicity and also to overcome the<br />

host immune tolerance that exists to native peptides. 97,98<br />

Conversely it has been reported that changes in the fine<br />

specificity of modified peptide-reactive T-cells following<br />

vaccination may occur with subsequent loss of tumor cell<br />

recognition. 99<br />

A peptide vaccine approach involves several steps<br />

which are aimed firstly at identifying the appropriate<br />

peptide, secondly at checking for its immunogenicity<br />

and relevance as a tumor-associated antigen (TAA) in<br />

vitro, and thirdly at formulating a safe product to be<br />

used clinically.<br />

Identification of the appropriate peptide<br />

1) From protein to peptide. This approach involves the<br />

screening of potentially HLA-binding peptides within the<br />

sequence of a known tumor-specific protein by using HLA<br />

anchor motifs and epitope selection. Peptides derived by<br />

mut RAS, 100 melanoma-associated MAGE protein, 101<br />

prostate specific antigen 102 and chronic myelogenous<br />

leukemia (CML) specific P210 were identified by this<br />

approach. 103,104 In CML, for example, a possible total of 76<br />

peptides, 8 to 11 amino acids in length, spanning the b2a2<br />

and b3a2 junctional regions of bcr-abl were screened for<br />

HLA class I- binding motifs and tested for the effective<br />

binding property to purified HLA molecules. Four of them,<br />

all derived from the b3a2 breakpoint, were found to be<br />

able to bind with either intermediate or high affinity to<br />

purified HLA A3, A11 and B8. 103 A similar approach<br />

allowed identification of class II b3a2 breakpoint peptides<br />

capable of binding HLA DR11, 105 DR4 106 and DR1 107 This<br />

method for identifying tumor-specific peptides is relatively<br />

simple, fast and suitable for any known intracellular<br />

protein that may be a potential TAA. Nevertheless, it<br />

bears the disadvantage that it cannot, by itself, predict<br />

whether the identified peptide is found on HLA molecules<br />

of the leukemia or cancer cells that contain the parent<br />

protein.<br />

2) From peptide to protein. Another strategy used to<br />

identify suitable peptides for cancer vaccines involves the<br />

structural analysis of naturally processed peptides (NPPs)<br />

bound to HLA class I and class II molecules of cancer<br />

cells. NPPs were first isolated and sequenced by acidelution<br />

from immunoaffinity purified HLA molecules and<br />

subsequently compared with existing protein<br />

sequences. 108 Alternative approaches are to obtain NPPs<br />

by mechanically destroying and acid-treating whole<br />

tumor cells and/or by exposing living tumor cells to rapid<br />

haematologica vol. 85(suppl. to n. 12):December 2000


166<br />

M. Bocchia et al.<br />

acid treatment. 109 These procedures should characterize<br />

tumor-, differentiation stage- and tissue-specific selfantigen<br />

MHC-bound peptides as well as the naturally<br />

processed proteins from which they are derived and use<br />

them as tools for immunotherapy. The main disadvantage<br />

of this approach is that many tumor cells express low<br />

levels of HLA molecules and the yield of NPPs can be<br />

scarce. Nevertheless some immunogenic peptides derived<br />

from wild-type p53 protein, melanoma associated MART-<br />

1 and gp100 proteins were identified by these methods,<br />

110,111 and naturally processed peptides from acute<br />

myeloblastic leukemia cells and CML blasts are now<br />

under evaluation. 112,113 Advantages and disadvantages of<br />

synthetic versus natural tumor peptides have been<br />

recently reviewed. 114<br />

3) From tumor infiltrating lymphocytes to peptide.<br />

Probably the most clinically relevant tumor peptides are<br />

those identified from the epitope analysis of tumor infiltrating<br />

lymphocytes (TIL). 115-118 In preliminary experiments<br />

HLA class I restricted CTL lines were derived by repetitive<br />

in vitro stimulation of TIL with autologous tumor cells.<br />

Subsequently, by transfection of a tumor cDNA library<br />

and in vitro sensitization assays, the peptide sequences<br />

recognized by the tumor-specific CTLs were identified as<br />

were the parent proteins (i.e. peptides derived from<br />

melanoma associated gp100, tyrosinase and the MAGE<br />

family). Most TIL-derived tumor peptides found in the<br />

past few years are MHC class I-restricted; however, a<br />

novel melanoma antigen resulting from a chromosomal<br />

rearrangement and recognized by a HLA-DR1-restricted<br />

CD4 + -TIL has recently been identified. 119<br />

Checking for peptide immunogenicity<br />

Except for TIL-derived peptides, all other tumor-specific,<br />

HLA binding, synthetic or naturally expressed peptides<br />

still need to be tested for immunogenicity. The<br />

ability of inducing CTL or specific CD4 + proliferation has<br />

been evaluated for all tumor-specific peptides that were<br />

subsequently used in clinical trials. P210-derived peptides,<br />

for example, were able to elicit peptide-specific T-<br />

cell immunity both in normal donors, 105,120 and CML<br />

patients. 121 Their relevance as TAAs has been further<br />

confirmed by observing peptide-specific HLA restricted<br />

CTLs and CD4 + cells able to mediated killing of b3a2-<br />

CML cells and proliferation in the presence of b3a2 containing<br />

cell lysates, respectively. 106,107 The latter findings<br />

were the indirect proof of natural processing of P210<br />

and of HLA presentation of breakpoint-derived peptides.<br />

Although strong peptide-specific CTL and CD4 + responses<br />

have been shown in vitro for most tumor peptides so<br />

far identified, few data on T-cell induced immunity after<br />

peptide vaccination in patients have been generated.<br />

100,122,123 Thus, strategies to improve peptide immunogenicity,<br />

by using different adjuvants and delivery systems,<br />

are currently under evaluation.<br />

Peptide vaccine formulation<br />

The goal of experimental clinical protocols using peptide<br />

antigens for active vaccination is to induce a strong<br />

CTL response against the immunizing antigen and thereby<br />

against tumor cells expressing the antigen. The mode<br />

of peptide-based cancer vaccination critically affects the<br />

clinical outcome. The synthesis of a peptide on a large<br />

scale, its purification and testing for common Quality<br />

Control/Quality Assurance compliance are simple and<br />

fast procedures but the choice of an effective delivery<br />

system for the peptide is crucial. Peptide vaccination<br />

strategies currently being evaluated include: 1) direct<br />

peptide vaccination with immunologic adjuvants and/or<br />

cytokines; 124 2) lipopeptide conjugates; 125 3) peptide<br />

loading onto splenocytes or DC; 126 4) lysosomal complexes.<br />

127 Recently, a specific formulation of the polysaccharide<br />

poly-N-acetyl glucosamine has been found to<br />

be an effective vehicle for sustained peptide delivery in<br />

a murine vaccine model able to generate a primary CTL<br />

response with a minimal peptide dose. 84 Finally, triggering<br />

CD40 in vivo with an activating antibody considerably<br />

improved the efficacy of peptide-based anti-tumor<br />

vaccines in mice, converting a peptide with minimal<br />

immunogenicity into a strong CTL inducer. 44<br />

Recombinant viruses<br />

The molecular identification of the antigens on human<br />

tumors recognized by T- and B-lymphocytes offers the<br />

opportunity to design novel cancer vaccines based on<br />

recombinant forms of TAA. Genes coding for TAA can be<br />

inserted into the genome of attenuated micro-organisms<br />

such as bacteria and viruses. Viruses are among<br />

the most interesting vectors since they are able to<br />

induce antibody, Th, and CTL responses in the absence<br />

of co-stimulation. 128 Their long-lasting cohabitation<br />

with human beings has likely favored the evolution of<br />

specific patterns recognized by the innate immune system<br />

which create an immunostimulatory environment<br />

for optimal immune responses. The vector choice, however,<br />

is limited by the possible disadvantages of recombinant<br />

virus utilization, such as recombination with<br />

wild-type viruses, oncogenic potential, or virus-induced<br />

immunosuppression.<br />

Vaccinia virus (VV) belongs to the Poxviridae family,<br />

and its worldwide use in the smallpox eradication campaign<br />

demonstrated that it was safe and very effective.<br />

To date, no other large-scale vaccination program has<br />

had such an impact on human diseases, because smallpox<br />

has been virtually eliminated from the world population.<br />

Large amounts of foreign DNA can be stably<br />

inserted into the VV genome by homologous recombination.<br />

129 VV employs a built-in transcriptional and posttranslational<br />

apparatus to produce large amounts of the<br />

protein encoded by the inserted gene. VV sojourns within<br />

the host cell cytoplasm and does not integrate nor is<br />

it oncogenic. 129 The induction of potent cellular and<br />

humoral immune responses with recombinant (r)VV was<br />

observed in several tumor systems. 130-132 Preclinical studies<br />

in models of pulmonary metastatization caused by<br />

tumors bearing a prototype TAA have revealed some features<br />

of rVV that are important in determining successful<br />

therapy. In particular, TAA gene must be expressed<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

167<br />

under the control of a strong, early promoter which<br />

allows its expression in professional APC such as DC. 133<br />

Moreover, while prevention from tumor challenge<br />

requires only the synthesis of the TAA by the recombinant<br />

virus, genes encoding immunostimulatory molecules<br />

inserted in the recombinant poxvirus, 134 exogenous<br />

cytokines, 131,135 or their combination 136 are required to<br />

induce eradication of established tumors.<br />

Encouraging results have also been obtained in mouse<br />

models more relevant to the therapy of human cancer.<br />

Immunization of mice transgenic for the human HLA-<br />

A*0201 allele with an rVV encoding a form of the<br />

melanoma antigen gp100, which had been modified to<br />

increase epitope binding to the restricting class I molecule,<br />

elicited CD8 + T-lymphocytes specific for the epitope<br />

that is naturally presented on the surface of an HLA-<br />

A*0201-expressing mouse melanoma. 137 Repeated inoculations<br />

of an rVV encoding the mouse tyrosinase-related<br />

protein-1 (TRP-1/gp75) caused autoimmune attack of<br />

normal melanocytes manifested by hair depigmentation<br />

(vitiligo) and CD4 + -mediated melanoma destruction in<br />

mice. 138 In a clinical trial, administration of VV encoding<br />

human carcinoembryonic antigen (CEA) proved effective<br />

in inducing both humoral and cellular immune responses<br />

in patients with colorectal cancer. 139<br />

Poxviruses are not the only choice for tumor immunologists.<br />

Adenoviruses in which critical genes that<br />

enable viral replication have been deleted and replaced<br />

by genes encoding heterologous antigens, have been<br />

generally used in gene therapy studies, but have also<br />

provided antitumor activity when employed as immunogens.<br />

140 Liver toxicity was described following systemic<br />

administration of high titers of first generation E1-<br />

deleted Adenovirus vectors optimized for gene therapy.<br />

141 These side effects, which certainly raise some concerns<br />

about Adenovirus administration in patients,<br />

might not restrain their use in cancer immunotherapy<br />

since systemic delivery of high viral titers would certainly<br />

not be the favored immunization route.<br />

Initial clinical trials have unveiled some of the intrinsic<br />

limitations of recombinant viruses. Many patients, in<br />

fact, have high neutralizing antibodies against VV as a<br />

consequence of its use as a vaccine for smallpox prevention,<br />

and against Adenoviruses, which cause upper respiratory<br />

tract infections throughout life. High doses of<br />

recombinant adenoviruses expressing the human<br />

melanoma antigens MART-1 and gp100 could be safely<br />

administered to cancer patients, but the high levels of<br />

neutralizing antibodies present in their sera likely impair<br />

the ability of these viruses to immunize against the<br />

melanoma antigens. 142<br />

These results, largely expected, have not put an end<br />

to the clinical use of recombinant viruses, as various<br />

strategies have been exploited to overcome pre-existing<br />

immunity. Several groups have engineered non-replicating<br />

viruses which normally do not infect human<br />

beings. The Avipoxviridae family comprises viruses, such<br />

as fowlpox and canarypox, which can productively infect<br />

avian but not mammalian cells, and are not cross-reactive<br />

with VV. 143 A recombinant fowlpox virus expressing<br />

a model TAA was able to cure established tumors in<br />

mice; 144 an important aspect of this study was the observation<br />

that prior exposure to VV did not abrogate the<br />

immune responses induced by the recombinant fowlpox<br />

virus. A different non-replicating virus, canarypox virus<br />

(ALVAC), has also been employed to elicit immune<br />

responses against a variety of antigens. 145,146<br />

A highly attenuated strain of VV, known as modified VV<br />

Ankara (MVA), has been inoculated as smallpox vaccine<br />

into more than 120,000 recipients without causing any<br />

significant side effect. 147 Replication of MVA is blocked at<br />

the step of virion assembly and for this reason the MVA<br />

vectors produce recombinant proteins expressed under<br />

the control of both early and late viral promoters, thus<br />

mimicking the expression in wild-type virus. An MVA vector,<br />

and a fowlpox virus vector expressing a model TAA<br />

showed better therapeutic effects on pulmonary metastasis<br />

than a VV encoding the same TAA. 148 MVA is a very<br />

promising vector for the development of recombinant<br />

vaccines for cancer, and can be efficiently used in combination<br />

with DNA vaccines. 149<br />

As an alternative approach, the mucosal route of<br />

administration was recently shown to overcome preexisting<br />

immunity to VV. Intrarectal immunization of<br />

vaccinia-immune mice with rVV expressing HIV gp160<br />

induced specific serum antibody and strong HIV-specific<br />

CTL responses in both mucosal and systemic lymphoid<br />

tissue, whereas systemic immunization was ineffective<br />

under these circumstances. 150<br />

Direct immunization of mice with recombinant adenoviruses<br />

resulted in the induction of high titers of neutralizing<br />

antibodies, which precluded a boost of CTL<br />

responses after repeated inoculations. The presence of<br />

neutralizing antibodies did not, however, affect the<br />

immunogenicity of infected DC, as repeated administration<br />

of virus-infected DC boosted the CTL response even<br />

in mice previously infected with the recombinant vector.<br />

151 It was also shown that protective immunity against<br />

mouse melanoma “self” antigens, gp100 and TRP-2, could<br />

be obtained by DC transduced with Adenovirus vector<br />

encoding the antigen. Importantly, immunization with<br />

Adenovirus-transduced DC was not impaired in mice that<br />

had been pre-immunized with Adenovirus. 152 With the<br />

help of these novel strategies, recombinant vectors could<br />

be used in the general population, including those individuals<br />

previously exposed to the viruses. Gene transfer<br />

to different human DC subpopulations by vaccinia and<br />

adenovirus vectors is a conceivable strategy for TAA loading.<br />

153<br />

DNA vaccines<br />

Following the first and somewhat shocking demonstration<br />

that the intramuscular injection of naked DNA<br />

(i.e., DNA devoid of a viral coat) encoding the influenza<br />

A nucleoprotein could induce nuceloprotein-specific<br />

CTL, and protect mice from challenge with heterologous<br />

influenza strains, 154 DNA immunization has become a<br />

rapidly developing technology. This vaccination method<br />

haematologica vol. 85(suppl. to n. 12):December 2000


168<br />

M. Bocchia et al.<br />

provides a stable and long-lasting source of antigen,<br />

and elicits both antibody- and cell-mediated immune<br />

responses. Compared to recombinant viruses, DNA vaccines<br />

offer a number of potential advantages because<br />

they are cheap, easy to produce, and do not require special<br />

storage or handling. DNA vaccines express virtually<br />

only the heterologous gene, therefore, they should<br />

induce an immune response selective for the antigen<br />

and not the vector, thus supplying a source of antigen<br />

suitable for repeated boosting. DNA vaccination has<br />

proven to be a generally applicable approach to various<br />

preclinical animal models of infectious and non-infectious<br />

diseases, 155 and several DNA vaccines have now<br />

entered phase I/II human clinical trials. Although the<br />

clinical application of DNA vaccines is a very young<br />

practice, some trials have already demonstrated that is<br />

possible to elicit a specific CTL response against malaria<br />

and HIV proteins in human volunteers. 156,157<br />

This novel vaccination approach involves different<br />

steps: 1) cloning of a heterologous gene under the control<br />

of a viral promoter (ordinarily derived from the CMV<br />

immediate early region); 2) purification of the endotoxin-free<br />

DNA plasmid from bacteria factories; 3) administration<br />

of the expression vectors by direct intramuscular<br />

or intradermal injection with a hypodermic needle<br />

or using a helium-driven, gene gun to shoot the skin<br />

with DNA-coated gold beads. Heterologous DNA can<br />

also be introduced into recombinant Salmonella, 158,159<br />

or Listeria strains 160 that can be thus administered by a<br />

mucosal route (Figure 3). In addition to these classic<br />

routes of DNA delivery, plasmid-based gene expression<br />

vectors have also been admixed with polymers and<br />

administered with a needle-free injection device,<br />

achieving high and sustained levels of antigen-specific<br />

antibodies. 161 The route of DNA delivery can profoundly<br />

influence the type of immune response by preferentially<br />

activating different Th populations: gene gun bombardment<br />

elicits a Th2 response, while intramuscular<br />

inoculation induces Th1 activation, even though the<br />

antigen form (i.e., membrane-bound vs secreted) can<br />

also exert some effect. 162,163<br />

The immunostimulatory activity of DNA vaccines has<br />

been associated with the prokaryotic-derived portion of<br />

the plasmid, which contains a central CpG motif in the<br />

sequence PuPuCpGPyPy. 164,165 In their unmethylated<br />

form, these hexamers stimulate monocytes and macrophages<br />

to produce different cytokines with a Th1 promoting<br />

activity including IL-12, TNF-α, and IFN-γ. 166,167<br />

A plasmid that incorporated several CpG islands in the<br />

prokaryotic ampicillin-resistance gene induced a<br />

stronger immune response when compared to a second<br />

plasmid carrying the kanamycin-resistance gene which<br />

possesses none. 168 To date, it is not known whether the<br />

CpG motifs will have the same immunostimulatory<br />

properties when applied to vaccination of human beings.<br />

However, it was recently reported that CpG motifs can<br />

activate in vitro subsets of freshly isolated human DC to<br />

promote Th1 immune responses. 169<br />

The mechanism of DNA-induced immunization has not<br />

yet been fully elucidated. An exclusive role for the direct<br />

transfection of normal tissue cells, such as myocytes or<br />

keratinocytes, has been debated because surgical ablation<br />

of the injected muscle within 1 minute of DNA inoculation<br />

did not affect the magnitude and longevity of DNAinduced<br />

antibodies. 170 Moreover, studies with bone-marrow<br />

chimeras clearly indicated that bone-marrowderived<br />

APC, either transfected by the DNA plasmid or<br />

able to capture the antigen expressed by other transfected<br />

cells, were necessary to prime T- and B-lymphocyte<br />

responses. 171 Indeed, more recent evidence suggests<br />

that Th and CTL are activated by DC directly transfected<br />

in vivo following DNA immunization. 172,173<br />

The first applications of DNA immunization to preclinical<br />

models of tumor growth revealed some interesting<br />

aspects. In general, the potency of naked DNA does not<br />

equal that of recombinant viruses, probably because DNA<br />

does not undergo a replicative amplification in the transfected<br />

cells, which in turn limits the amount of heterologous<br />

antigen produced. Inflammatory responses caused<br />

by DNA inoculation are more contained than those occurring<br />

during infection with viruses; for this reason, repeated<br />

inoculations of plasmid DNA, or the use of adjuvants<br />

such as cardiotoxin are generally required for the induction<br />

of an optimal response. Another emerging issue is<br />

that the efficacy of the vaccination approach depends<br />

more on the type of antigen than on the route of administration<br />

(Table 3). Vaccines based on shared viral antigens,<br />

or model TAA artificially introduced in the experimental<br />

tumors can be used to induce a strong, and often<br />

therapeutic immune response. Using a gene gun for DNA<br />

immunization, Irvine et al. 174 observed effective treatment<br />

of established pulmonary metastases, but recombinant<br />

cytokines were necessary to enhance the therapeutic<br />

effects. Unlike viral and model TAA, self TAA fail to induce<br />

sterilizing immunity since therapy of established tumors<br />

has been rarely reported, and prevention from challenge<br />

is often partial. 175-178 Central and peripheral tolerance to<br />

self antigen has thus emerged as the main limitation to<br />

the successful application of DNA vaccines to the therapy<br />

of cancer. This conclusion seems to apply to several<br />

mouse melanocyte differentiation antigens, a class of<br />

molecules that is expressed in both melanomas and<br />

melanocytes and includes tyrosinase, TRP-1/gp75, TRP-2,<br />

and gp100/pmel 17. However, tolerance can be broken by<br />

the use of a xenogeneic source of TAA. While immunization<br />

with mouse TRP-1/gp75 or TRP-2 antigens failed to<br />

induce a detectable immune response, vaccination with<br />

a plasmid DNA encoding the human homologous antigens<br />

elicited autoantibodies and CTL in C56BL/6<br />

mice: 178,179 immunized mice rejected metastatic melanoma<br />

and developed patchy depigmentation of their coats<br />

(vitiligo). This “obligatory” association of vitiligo and antitumor<br />

response was recently questioned by a study showing<br />

that immunization with mouse TRP-2-encoding plasmid<br />

could protect CB6 F1 mice in the absence of overt<br />

vitiligo, suggesting a role for the genetic background in<br />

controlling both the extent and the consequences of the<br />

immune activation against self TAA. 180<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

169<br />

A novel vaccine that combines the properties of viruses<br />

and DNA is based on antigen production in the context<br />

of an alphavirus replicon. These new vectors rely on<br />

the ability of the alphavirus RNA replicase to drive the<br />

replication of its own gene, as well as of subgenomic<br />

RNA encoding the heterologous antigen. This loop of<br />

self-replication results in a several-fold amplification of<br />

protein production in infected cells. 181 Compared with<br />

traditional DNA vaccine strategies, in which vectors are<br />

persistent and the expression constitutive, the expression<br />

mediated by the alphaviral vector is transient and<br />

lytic, resulting in a decrease of biosafety risks as well as<br />

the risk of inducing immunologic tolerance due to longlasting<br />

antigen expression. A single intramuscular injection<br />

of a self-replicating RNA immunogen at doses lower<br />

than those required for standard DNA-based vaccines<br />

elicited antibodies, CD8 + T-cell responses, and prolonged<br />

the survival of mice with established tumors. 182 Interestingly,<br />

the enhanced immunogenicity of these vectors<br />

correlated with the apoptotic death of transfected cells,<br />

which facilitated their uptake and presentation by DC.<br />

Methodology for ex vivo generation of DC<br />

Investigators working in human and murine systems<br />

have discovered culture conditions that use hematopoietic<br />

cytokines to support the growth, differentiation, and<br />

maturation of large amounts of DC. Therefore, DC can<br />

also be purified from peripheral blood after removal of<br />

other defined T, B, NK and monocyte populations by using<br />

antibodies and magnetic beads or a cell-sorter. 183 However,<br />

the very low frequencies of DC in accessible body<br />

samples, especially blood, limits the use of these DC for<br />

vaccination protocol. The ex vivo differentiation of DC<br />

progenitors can be traced easily by monitoring changes<br />

in some key surface molecules such as CD1a (acquired by<br />

DC) and CD14 (expressed by monocytes and lost by DC).<br />

Furthermore expression of co-stimulatory molecules such<br />

as CD40, CD80, CD86, as well as HLA antigens, can be<br />

used to evaluate the stage of differentiation and the<br />

degree of maturation of DC during in vitro culture. In<br />

addition, two new markers, CD83 and p55, have been<br />

shown to be selectively expressed by a small subset of<br />

mature DC differentiated in in vitro culture. 184,185 According<br />

to the knowledge of DC ontogeny, two major strategies<br />

are used. The first is based on the ability of CD34 +<br />

progenitors isolated from bone marrow, 186 peripheral<br />

blood, 187 or neonatal cord blood 188 to differentiate ex vivo<br />

within 12-14 days into mature CD1a + /CD83± HLA-DR +<br />

DC in the presence GM-CSF and TNF-α. Both stem cell<br />

factor (SCF) and FLT3 ligand are able to augment the DC<br />

yield if these key factors are present in the culture. 189<br />

The maturation of DC from progenitors is influenced<br />

not only by cytokines, but also by extracellular matrix<br />

(ECM) proteins, such as fibronectin which has been<br />

reported to enhance DC maturation by mediating a specific<br />

adhesion through the a5b1 integrin receptor. 190 The<br />

choice of culture conditions, especially the cytokine combination,<br />

will influence DC purity, maturation and function,<br />

and this is a consideration of prime importance<br />

before starting a DC-based immunotherapy strategy. A<br />

more practical approach is the production of DC from<br />

CD14 + monocytes, in the presence of GM-CSF and IL-4.<br />

A future strategy for easy achievement of large<br />

amounts of DC is the in vivo injection of the same<br />

cytokines utilized for ex vivo DC generation. In fact, the<br />

administration of FLT3 ligand either in animals 191 or in<br />

humans 192 results in a reversible accumulation of functionally<br />

active DC in both lymphoid and non-lymphoid<br />

tissues. Therefore, in murine models it has been demonstrated<br />

that FLT3 ligand caused the regression of various<br />

tumors supporting the suggestion that DC may be directly<br />

involved in the antitumor effect of FLT3 ligand. 193,194<br />

Strategies for delivery of TAA into DC<br />

Several approaches for delivery of TAA into DC have<br />

been utilized. To date, in the clinical protocol of vaccination<br />

by DC both synthetic peptides corresponding to<br />

known tumor antigens and tumor-eluted peptides have<br />

been used for DC-mediated antigen presentation. 195<br />

While synthetic peptides represent only the limited antigenic<br />

repertoire of the presently known tumor antigens,<br />

tumor-eluted peptides, though originating from<br />

unknown proteins, may reflect a wider antigenic spectrum.<br />

Another potential disadvantage of using defined<br />

synthetic peptides to activate tumor-reactive T-cells is<br />

that the generated peptide-specific T-cells may not recognize<br />

autologous tumor cells expressing the antigen of<br />

interest. Loading DC with cocktails of different synthetic<br />

peptides, corresponding to different tumor antigens<br />

expressed by the same tumor, has been demonstrated to<br />

be a clinically effective procedure. Nevertheless it is possible<br />

that the synthetic peptide-approach will limit<br />

patient selection, on the basis of the HLA phenotype,<br />

and will prevent the possibility of activating both CD4<br />

and CD8 T-cells directed to different epitopes of the<br />

same antigen. To by-pass these disadvantages, several<br />

alternative methodologies using a mix of TAA have been<br />

developed. DC are able to internalize complete tumor<br />

lysates or apoptotic cells and to present derived antigen<br />

in an HLA I-restricted manner. 196 In addition, DC secrete<br />

antigen-presenting vesicles, called exosomes, which<br />

express functional HLA class I and class II, and T-cell<br />

co-stimulatory molecules. Tumor peptide-pulsed DCderived<br />

exosomes prime specific cytotoxic T-lymphocytes<br />

in vivo and eradicate or suppress growth of established<br />

murine tumors in a T-cell-dependent manner. 197<br />

However, a possible limitation of these approaches is<br />

the need for large numbers of primary samples or tumor<br />

cell lines and the complete lack of control of the nature<br />

of the antigens that are being presented by the DC. The<br />

use of RNA instead of protein could constitute a good<br />

alternative since it could be amplified in vitro. In addition,<br />

substractive hybridization could allow the enrichment<br />

of tumor-specific RNA, thus limiting immune<br />

response against self antigen. 198 A further possibility is<br />

the engineering of DC with expression vectors carrying<br />

TAA genes. Among the viral vectors, retroviral, adenoviral,<br />

and vaccinia vectors have been widely utilized to<br />

haematologica vol. 85(suppl. to n. 12):December 2000


170<br />

M. Bocchia et al.<br />

transduce either monocyte or CD34 + cell-derived DC.<br />

Many authors have chosen retroviral vectors because<br />

retroviral transduced-DC should be able to constitutively<br />

express and process TAA to produce long-term<br />

antigen presentation in vivo. Specific CTLs against transduced-TAA<br />

are elicited by retrovirus-engineered DC. 199<br />

However, their low efficiency of transduction limits the<br />

clinical use of retroviruses.<br />

In contrast, adenoviral vectors infect replicating and<br />

non-replicating cells, are easy to handle, and supernatant<br />

with clinical grade high titer is readily achievable. Both<br />

monocyte and CD34 + cell-derived DC can be transduced<br />

with high efficiency by adenovirus combined with polycations.<br />

200,201 Moreover, DC transduced by adenovirus<br />

maintain their APC functions. 202 However, the use of adenovirus<br />

vectors is hampered by their immunogenicity<br />

which causes the rapid development of a CTL response<br />

that eliminates virus-infected cells and generation of<br />

neutralizing antibodies in recipients. Moreover, vaccinia<br />

virus which is a member of the poxvirus family, is not<br />

oncogenic, does not integrate into the host genome, is<br />

easy to manipulate genetically and is capable of accepting<br />

large fragments of heterologous DNA. 203 The transduction<br />

of CD34 + cell-derived DC is feasible but their<br />

use is limited by the narrow therapeutic index between<br />

optimal transduction and target cell viability. 153 The presence<br />

of acquired genetic abnormalities in myeloid hematologic<br />

malignancies might represent the basis for innovative<br />

immune-based anti-leukemic strategies. In fact,<br />

intracellular proteins can be processed and presented on<br />

the cell surface by HLA molecules indicating the possibility<br />

that leukemia-specific genetic abnormalities may<br />

be targets for cytotoxic T-cells. The generation of functional<br />

monocyte-derived DC carrying the specific genetic<br />

lesion has been reported for both acute myeloid and<br />

lymphoid leukemia (AML), 204,205 as well as chronic myelogenous<br />

leukemia (CML). 206 Current protocols for ex vivo<br />

DC generation from CD34 + cells does not allow large<br />

numbers of leukemic DC to be produced, probably<br />

because of a defective proliferative and/or maturative<br />

capacity of transformed CD34 + cells.<br />

Recently, a protocol which allows the optimal generation<br />

of BCR/ABL-positive DC from CML-derived CD34 +<br />

cells has been reported. 207<br />

Antitumor vaccination:<br />

emerging clinical results<br />

Clinical trials in solid tumors<br />

Despite the fact that the large number of ongoing clinical<br />

trials which can be derived from the Physician Data<br />

Query (PDQ) of NCI (Figure 4) suggests a diffuse interest<br />

in immunotherapy, there is still a strong need to define<br />

the clinical impact of immunotherapy in the treatment<br />

of solid tumors. Table 4 summarizes the already published<br />

vaccination trials carried out using a) autologous<br />

or allogeneic neoplastic cells, b) synthetic peptides corresponding<br />

to defined TAA, alone or pulsed on autologous<br />

monocytes or DC.<br />

Melanoma<br />

Melanoma is the most striking example of a non-virusinduced<br />

immunogenic tumor in man that is able to elic-<br />

Figure 3. DNA immunization protocols.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

171<br />

it T-cell-mediated antitumor immunity. The majority of<br />

tumor antigens defined by T-cells have been identified<br />

utilizing patients’ T-lymphocytes as effector cells and<br />

tumor cells obtained from autologous (metastatic) tumor<br />

deposits as the target. 68 Several investigators have isolated<br />

cross-reactive tumor-specific CTLs from peripheral<br />

blood, lymphocytes, or tumor-infiltrating lymphocytes of<br />

melanoma patients, and these CTLs are able to recognize<br />

common tumor antigen expressed in melanomas that<br />

share the restricting HLA class I allele. 19 Thus, large numbers<br />

of ongoing or published clinical trials have been carried<br />

out on patients with metastatic melanoma. Experimental<br />

strategies are encouraged since with current<br />

standard therapies the prognosis of patients with<br />

metastatic melanoma is poor with a median survival of<br />

about 6 months. 208 Several approaches to induce antitumor<br />

immune response have been reported.<br />

Irradiated tumor cells. The initial anti-melanoma vaccines<br />

were similar in their preparation to the vaccines<br />

against infectious diseases. Crude preparations of<br />

homogenized tumor cells were mixed with immunestimulating<br />

adjuvants such as viral or bacterial particles.<br />

Some of these early vaccines were tested in clinical trials<br />

and induced objective clinical responses in about<br />

25% of the cancer patients. 209 The most impressive trial<br />

was reported by Morton et al. 64 who administered, to<br />

136 stage IIIA and IV (American Joint Commitee on Cancer,<br />

AJCC) melanoma patients, a polyvalent melanoma<br />

cell vaccine (MVC) comprising 3 allogeneic melanoma<br />

cell lines. Of 40 patients with evaluable disease, 9 (23%)<br />

had regressions (3 complete). Induction of cell-mediated<br />

and humoral immune responses to common<br />

melanoma-associated antigens present on autologous<br />

melanoma cells was observed in patients receiving the<br />

vaccine. Survival correlated significantly with DTH and<br />

antibody response to MCV and there was a 3-fold<br />

increase in 5-year survival of patients with stage IV<br />

melanoma. Livingston et al. 65 randomized 122 stage III<br />

melanoma patients free of disease after surgery to<br />

receive treatment with the ganglioside GM2/BCG vaccine<br />

or with BCG alone. All patients were pretreated with<br />

low-dose cyclophosphamide. In most patients vaccinated<br />

with GM2/BCG, an antibody production against ganglioside<br />

was demonstrated and this was associated with<br />

a prolonged disease-free interval and survival, although<br />

the improvement did not reach statistical significance.<br />

Gene-modified tumor cell vaccine. Autologous or allogeneic<br />

tumor or fibroblast cells have been modified to<br />

express cytokines and/or co-stimulatory molecules and/or<br />

a suicide compound. This genetic engineering is mainly<br />

performed ex vivo using retroviral vectors. In the published<br />

human trials, tumor cells have been transduced to<br />

express several cytokines. Arienti et al. 210 described 12<br />

stage IV melanoma patients who underwent vaccination<br />

with HLA-A2-compatible allogeneic human melanoma<br />

cells (5x10 7 or 15x10 7 cells) engineered to release IL-2.<br />

Little toxicity with three mixed clinical responses was<br />

recorded. Among the nine patients immunologically evaluated,<br />

peripheral blood lymphocytes from three patients<br />

displayed enhanced non-HLA-restricted cytotoxicity, and<br />

two of those individuals had an increased reactivity<br />

against tyrosinase peptide or gp-100 peptide after immu-<br />

Table 3. DNA vaccines in active immunotherapy of experimental mouse tumors.<br />

TAA Experimental tumor Route of inoculation Adjuvant Vaccine effects Reference<br />

Beta-galactosidase adenocarcinoma gene gun bombardment cytokines treatment of 2-day-old (47)<br />

(model TAA) followed by cytokine i.p. (IL-2, IL-12) pulmonary metastases<br />

gag from M-MuLV leukemia i.m., 3 inoculations every 10 days none complete protection (56)<br />

from challenge<br />

HPV-E7 sarcoma 3 gene gun bombardments, none complete protection (48)<br />

every 2 weeks<br />

from challenge<br />

Neu spontaneous mammalian i.m., 4 weekly inoculations IL-2-encoding plasmid partial protection (50)<br />

tumor<br />

from challenge<br />

P1A mastocytoma i.m., 3 inoculations every 10 days none partial protection (49)<br />

from challenge<br />

Idiotype/GM-CSF B-cell lymphoma i.m., 3 inoculation every 3 weeks none partial protection (57)<br />

fusion protein<br />

from challenge<br />

mouse TRP-2 melanoma 3 gene gun bombardments, IL-12-encoding plasmid partial protection (48)<br />

every 2 weeks<br />

from challenge<br />

human TRP-1 melanoma 5 gene gun bombardments, weekly none reduction of pulmonary (52)<br />

metastases upon challenge;<br />

vitiligo<br />

human TRP-2 melanoma 1-4 gene gun bombardments GM-CSF (in therapy setting) reduction of pulmonary (51)<br />

metastases (prevention<br />

and therapy); vitiligo<br />

mouse TRP-2 melanoma i.m. cardio-toxin protection from challenge; (53)<br />

no vitiligo<br />

haematologica vol. 85(suppl. to n. 12):December 2000


172<br />

M. Bocchia et al.<br />

nization. In the only patient for whom the autologous<br />

melanoma line was available, and following in vitro stimulation<br />

of the PBLs after vaccination, the frequency of<br />

CTL precursor (CTLp) was significantly enhanced. Other<br />

groups utilized a similar approach transducing different<br />

cytokines, i.e. IL-12, 40 IL-7, 211 and GM-CSF. 39 Despite documented<br />

specific antitumor reactivity with an increased<br />

frequency of anti-melanoma cytolytic precursor cells,<br />

negligible clinical results were demonstrated. To summarize,<br />

the advantage of a genetically modified autologous<br />

cell vaccine is that it contains the whole collection of<br />

tumor proteins and therefore has the greatest chance of<br />

inducing an immune response against relevant tumor<br />

antigens. However, growing autologous tumor cells in vitro<br />

to establish tumor cell lines is time-consuming and<br />

often unsuccessful. These studies are relevant since they<br />

show that injection of gene-modified cells into a patient<br />

a) is safe; b) is followed by efficient and variable transduction<br />

rate of host tissues; c) is associated with transgene<br />

expression in the patient; d) is associated with biological<br />

activity of the transgene product in most instances.<br />

Synthetic and natural peptides. Phase I clinical trials<br />

have been carried out using synthetic peptides corresponding<br />

to defined TAA. The clinical trial using<br />

melanoma differentiation antigen (MAGE-3.A1) by<br />

Marchand et al., 123 enrolling 39 chemoresistant stage IV<br />

melanoma patients, was encouraging because monthly<br />

injection of 100-300 µg peptide alone was associated<br />

with tumor regression in 7 out of 26 patients who<br />

received the complete treatment. All but one of these<br />

regressions involved cutaneous metastases. No evidence<br />

for CTL response was found in the blood of the 4 patients<br />

who were analyzed, including 2 who displayed complete<br />

tumor regression. In contrast, Jager et al. 212 immunized<br />

similar patients with gp100 peptide along with GM-CSF<br />

and, in some patients, were able to document an<br />

increase in the specific CTL activity against the immunizing<br />

peptide. Rosenberg et al. 213 reported that 31<br />

metastatic melanoma patients were immunized with<br />

the modified g209-2M peptide in incomplete Freund’s<br />

adjuvant (IFA) along with IL-2 obtaining tumor regression<br />

in 42% of patients. Peripheral blood mononuclear<br />

cells harvested from these patients after, but not before,<br />

immunization exhibited a high degree of reactivity<br />

against the native g209-217 peptide, as well as against<br />

HLA-A2+ melanoma cells.<br />

These studies indicate that vaccination with synthetic<br />

peptides is well tolerated, with occasional occurrence<br />

of mild fever and inflammation at the site of injection.<br />

Nonetheless, it should be pointed out that there is usually<br />

a poor correlation between induction of specific T-<br />

cells and the clinical response. The reasons for this discrepancy<br />

might be the selection of the patients enrolled<br />

in the trials since the majority of patients were in stage<br />

IV with large amounts of disease.<br />

Dendritic cells. Autologous DC generated from peripheral<br />

blood monocytes have been utilized as antigen-presenting<br />

cells after their loading with specific melanoma<br />

antigens. Chakraborty et al. 214 found that intradermal<br />

administration of DC pulsed with a MAGE-1 HLA class<br />

I-restricted peptide could elicit peptide and autologous<br />

melanoma reactive-CTLs in patients with advanced<br />

melanoma. However, despite the presence of these CTLp<br />

in the vaccination site, peripheral blood, and distant<br />

tumor sites, no significant therapeutic responses were<br />

seen.<br />

Nestle et al. 195 recently described the immunization of<br />

16 melanoma patients using DC loaded with melanoma<br />

peptides or tumor lysates. DC were pulsed with a cocktail<br />

of gp100, MART-1, tyrosinase, MAGE-1, or MAGE-3<br />

peptides chosen to suit the individual patient class I HLA<br />

molecules. Four patients whose HLA haplotype was inappropriate<br />

for peptide pulsing received DC pulsed with<br />

autologous tumor lysate. Keyhole limpet hemocyanin<br />

(KLH) was included during antigen pulsing. DC were<br />

administered by direct injection into uninvolved lymph<br />

nodes via ultrasound guidance to facilitate entry into the<br />

lymphatics and to minimize DC loss. Patients received 6-<br />

10 injections of 1x10 6 cells every 1-4 weeks. Toxicity was<br />

limited to mild local reactions at the injection sites.<br />

Immunologic monitoring revealed DTH skin reactions to<br />

peptides in 11 cases, and peptide-specific CTLs could be<br />

recovered from the skin biopsies of some patients.<br />

Regression of tumor was seen in 5 out 16 patients,<br />

including 2 complete responses lasting over 15 months.<br />

Responding tumor sites included skin, lung, soft tissue,<br />

bone, and pancreas. Importantly, two of the responding<br />

patients received only tumor lysate-pulsed DC, suggesting<br />

an approach applicable to cancers lacking defined<br />

tumor antigens.<br />

In a similar but smaller trial at the University of Pittsburgh,<br />

215 6 HLA-A2 + patients with metastatic melanoma<br />

received four weekly intravenous injections of 1-3x10 6<br />

monocyte-derived DC pulsed with HLA-A2-restricted<br />

peptides derived from MART-1, gp100, and tyrosinase.<br />

Complete regression of a subcutaneous mass lasting<br />

more than one year has been observed in one patient.<br />

Colon cancer<br />

Colon cancer is potentially curable by surgery; the<br />

cure rate is, however, moderate to poor depending on<br />

the extent of disease. Adjuvant chemotherapy with 5-<br />

fluorouracil plus levamisole or folinic acid is the standard<br />

treatment for stage III colon cancer based on the<br />

results of numerous co-operative and intergroup clinical<br />

studies. In contrast, adjuvant chemotherapy for stage<br />

II disease has no benefit. 216 Despite several immunotherapeutic<br />

approaches having been tested for colon cancer<br />

patients, only one study has reported clinical results.<br />

In a prospective randomized study, 217 254 patients with<br />

stage II or III post-surgery colon cancer were randomly<br />

assigned to receive active specific immunotherapy,<br />

namely autologous tumor cell-bacille Calmette-Guèrin<br />

(BCG) or no adjuvant treatment. The immunotherapy<br />

program comprised three weekly intradermal injections<br />

starting 4 weeks after surgery, with a booster vaccination<br />

at 6 months with 10 7 irradiated autologous tumor<br />

cells. The first vaccination contained 10 7 BCG organ-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

173<br />

isms. The 5-year median follow-up showed a 44%<br />

reduction of risk of recurrence in all patients receiving<br />

the vaccinations. The major impact of immunotherapy<br />

was evident in patients with stage II disease, who had<br />

a significantly longer disease-free period and 61% risk<br />

reduction. In addition, no patient discontinued treatment<br />

early because of side effects.<br />

Recently, Foon et al. 218 generated anti-idiotype antibody,<br />

designated CeaVac, that is an internal image of<br />

CEA. Thirty-two patients with resected Dukes’ B, C, and<br />

D, and incompletely resected Dukes’ D disease were<br />

treated with 2 mg of CeaVac every other week for four<br />

injections and then monthly until tumor recurrence or<br />

progression. Fourteen patients were treated concurrently<br />

with a 5-FU chemotherapy regimen. All 32<br />

patients entered into this trial generated a potent anti-<br />

CEA humoral and cellular immune response. Interesting,<br />

the 5-FU regimen did not affect the immune response.<br />

A phase III trial for patients with resected colon cancer<br />

is ongoing.<br />

Prostate cancer<br />

Several prostate-tissue-associated antigens, including<br />

prostatic alkaline phosphatase (PAP), prostate-specific<br />

membrane antigen (PSMA), and prostate-specific antigen<br />

(PSA), are now being explored as targets for prostate<br />

cancer immunotherapy. 219-221 Valone et al. 221 have carried<br />

out a dose-escalation trial of peripheral blood DC pulsed<br />

with recombinant PAP protein in 12 patients with<br />

advanced prostate cancer. Intravenous administration<br />

of 0.3, 0.6, and 1.2x10 9 pulsed cells/m 2 monthly for three<br />

months resulted in T-cell proliferative responses against<br />

PAP in all patients, the magnitude of which was related<br />

to cell dosage. Toxicity was limited to myalgias in<br />

three patients. Clinical outcomes have not been reported.<br />

Salgallar et al. 222 have recently updated the results of<br />

another trial 223 in which monocyte-derived DC pulsed<br />

with HLA-A2-binding peptides derived from PSMA were<br />

administered to 82 patients with advanced, hormonerefractory<br />

prostate cancer. Six infusions of up to 2x10 7<br />

DC were administered every six weeks, with half of the<br />

patients also receiving systemic GM-CSF. The treatment<br />

was well tolerated. However, only two patients mounted<br />

T-cell responses against the PSMA peptides as measured<br />

by enzyme-linked immunospot (ELISPOT) and DTH<br />

skin testing. Although four patients had reductions in<br />

serum tumor markers following vaccination, the concurrent<br />

administration of radiotherapy and hormonal<br />

therapy makes interpretation of the vaccine’s effects<br />

difficult.<br />

Renal cell carcinoma<br />

Renal cell carcinoma accounts for 2% of all malignancies<br />

and many patients have metastatic disease at<br />

diagnosis and the prognosis is unfavorable. At present,<br />

neither chemotherapy nor radiation therapy has any significant<br />

influence on the course of disease or the survival<br />

time. Immunotherapy using recombinant IL-2<br />

alone 224 or combined with interferon-α 225 is currently<br />

the standard therapy for metatastic renal cell carcinoma.<br />

Cellular immunotherapy includes the adoptive<br />

transfer of in vitro expanded tumor infiltrating lymphocytes<br />

226 as well as active immunotherapy with an autologous<br />

tumor cell vaccine engineered to secrete GM-<br />

CSF. 38 Although each of these attempts generated<br />

promising results neither attempt met the expectations.<br />

Recently, Holtl et al. 227 have administered, to 4 metastatic<br />

renal cell carcinoma patients, autologous monocytederived<br />

DC pulsed with autologous tumor cell lysate.<br />

Each patient received 3 monthly intravenous infusions<br />

with the immunogeneic KLH. Initial results have shown<br />

Figure 4. NCI-registered immunotherapeutic trials according to diseases and strategies.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


174<br />

M. Bocchia et al.<br />

Table 4. Phase I/II trials of vaccination in patients with solid tumor.<br />

Disease Stage Pts Vaccine<br />

Melanoma IIIA/IV 136<br />

IV 12<br />

Melanoma Cell<br />

Vaccine (MCV)<br />

IL-2 Transduced-<br />

Melanoma Cell Line<br />

Route of<br />

Admin. TA Ads Side Effects Clinical Results<br />

ID<br />

SC<br />

Human<br />

Melanoma Cell<br />

Lines<br />

Melanoma Cell<br />

Line<br />

Yes/No<br />

No<br />

Mild (Erithema,<br />

Fever)<br />

Mild (Erithema,<br />

Fever)<br />

9/40 evaluable pts<br />

(3 CR, 6 PR)<br />

3 MR; 1 SD<br />

Immunol<br />

Results<br />

↑Cell mediated and<br />

humoral responses to<br />

MCV ; ↑ Activation of<br />

TIL<br />

↑ Melanoma-Specific<br />

CTLp<br />

Authors<br />

Morton, 1992<br />

Arienti, 1996<br />

IV 10<br />

IL-7 Transduced-<br />

Autologous<br />

Melanoma Cells<br />

SC<br />

Autologous<br />

Melanoma<br />

Cells<br />

No Mild (Fever) 2 MR 5 SD<br />

↑ Melanoma-Specific<br />

CTLp<br />

Moller, 1998<br />

IV 21<br />

GM-CSF Transduced-<br />

Autologous<br />

Melanoma Cells<br />

ID and SC<br />

Autologous<br />

Melanoma<br />

Cells<br />

No<br />

Mild (Erithema,<br />

Induration, Itching)<br />

1 PR; 1 MR; 3 Minor Res<br />

↑ Melanoma-Specific<br />

CTLp; 80% Tumor<br />

Destruction into<br />

Metastases<br />

Soiffer,1998<br />

IV 31<br />

Peptide+IL-2<br />

SC<br />

modified g209-<br />

2M<br />

IL-2 and<br />

IFA<br />

Mild (Erithema) 6 PR, 3 MR, 3 SD<br />

↑ Melanoma-Specific<br />

CTLp<br />

Rosenberg, 1998<br />

IV 39<br />

Peptide<br />

SC and ID<br />

MAGE-3 (HLA-<br />

A1)<br />

No Mild 3 CR; 4 PR<br />

No Increase of anti<br />

MAGE-3 CTLs even<br />

in Responders<br />

Marchand,1999<br />

IV 16<br />

Ag Pulsed Autologous<br />

Monocyte- derived<br />

Lymph nodes<br />

DCs<br />

Peptide<br />

Cocktail or<br />

Autologous<br />

Tumor Lysate<br />

KLH Mild (Fever) 2 CR; 3 PR;1 MR<br />

DTH to KLH in 16;<br />

DTH to peptidepulsed<br />

DCs in 11<br />

Nestle, 1998<br />

IV 17<br />

Ag Pulsed Autologous<br />

Monocyte- derived<br />

DCs<br />

ID<br />

Autologous<br />

Tumor Lysate<br />

No No 1 PR<br />

DTH to Vaccine in<br />

9/17; CD8 Cells in<br />

expanded-VIL<br />

Chakraborty, 1998<br />

Colon<br />

Cancer<br />

Prostate<br />

Cancer<br />

Renal Cell<br />

Cancer<br />

II and III 254<br />

Locally<br />

Advanced<br />

82<br />

IV 12<br />

Irradiated Autologous<br />

Tumor Cells<br />

ID<br />

Ag Pulsed Autologous<br />

Monocyte- derived<br />

Lymph nodes<br />

DCs<br />

Ag Pulsed Monocyte<br />

derived-DCs<br />

IV<br />

Autologous<br />

Tumor Cells<br />

Peptide<br />

Cocktail or<br />

Autologous<br />

Tumor Lysate<br />

Autologous<br />

Tumor Lysate<br />

BCG<br />

Mild<br />

Stage II: 61% Risk<br />

Reduction for Recurrences;<br />

Stage III: no Significant<br />

Benefits<br />

KLH Mild (Fever) 2 CR; 3 PR; 1 MR<br />

KLH Mild (Fever) 1 PR<br />

DTH+: ≥90% Pts<br />

DTH to KLH in 16;<br />

DTH to peptidepulsed<br />

DCs in 11<br />

DTH to KLH after 1°<br />

and 2° Vaccination<br />

Vermorken,1999<br />

Salgallar, 1998<br />

Holt, 1999<br />

Pts: Patients; TA: Tumor Antigens; Ads: Adjuvants; SC: Subcutaneous; MR: Mixed Response; SD: Stable Disease; CTLp: Cytotoxic T Lymphocyte precursor; ID: Intradermic; PR: Partial Response;<br />

TIL: Tumor-Infiltrating LymphocytesIFA: Incomplte Freund's Adjuvant; KLH: Keyhole Lympocianin; DTH: Delayed Type Hypersensitivity; IV: Intravenous<br />

that a potent immunologic response to KLH and, most<br />

importantly, against cell lysate could be measured in<br />

vitro after the vaccinations. In addition, the treatment<br />

was well tolerated with moderate fever as the only side<br />

effect. In contrast, only one partial response after 2 vaccinations<br />

was observed.<br />

Recently, Kugler et al. 228 vaccinated 17 patients with<br />

metastatic renal cell carcinoma using hybrids of autologous<br />

tumor and allogeneic DC generated by an electrofusion<br />

technique. After vaccination, and with a mean<br />

follow-up time of 13 months, four patients completely<br />

rejected all metastatic tumor lesions, one presented a<br />

mixed response, and two had a tumor mass reduction of<br />

greater 50%. These promising data indicate that hybrid<br />

cell vaccination is a safe and effective therapy for renal<br />

cell carcinoma and may provide a broadly applicable<br />

strategy for other malignancies with unknown antigens.<br />

Hematologic malignancies<br />

Tumor vaccines in B-cell lymphoproliferative<br />

disorders<br />

Multiple myeloma (MM) and low-grade non-Hodgkin’s<br />

lymphomas (NHL) are clonal expansions of lymphoid cells<br />

that have rearranged immunoglobulin (Ig) genes. Early<br />

during development, pre-B-cells become committed to<br />

the expression of a heavy and light chain Ig variable<br />

region. The heavy chain derives from the recombination<br />

of variable (V) with diversity (D) and joining (J) region<br />

genes with a constant region (C). The V-D-J joins occur<br />

with a variable number of nucleotide insertions or deletions<br />

resulting in a unique sequence which creates the<br />

third hypervariable region (CDR III) and contributes to<br />

the antigen-binding site. These antigenic regions (idiotype;<br />

Id) are characteristic for any given Ig-producing<br />

tumor (e.g. MM and NHL) and can be recognized by an<br />

immune response consisting of anti-Id antibodies and/or<br />

by Id reactive T-cells. 229-235 The tumor-derived Id is a self<br />

protein which, in most circumstances, is poorly immunogenic.<br />

However, haptens and adjuvants, including<br />

cytokines, have been used in several animal models to<br />

increase Id immunogenicity and establish protective anti-<br />

Id-immunity. 236 Lately, Id vaccines have come into medical<br />

use in patients with lymphoma and MM.<br />

Idiotype vaccination in human lymphoma<br />

A pioneering study was carried out in 9 lymphoma<br />

patients in CR or partial remission. They were immunized<br />

with subcutaneous injections of autologous Id, conjugated<br />

to KLH and emulsified in an oil-in-water emulsion<br />

containing non-ionic block polymers. 80 Specific anti-Id<br />

humoral and/or cellular responses were observed in 7/9<br />

patients. Two patients with measurable disease showed<br />

a clinical improvement. These results have been con-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

175<br />

firmed in a larger series of patients. 82 Following standard<br />

chemotherapy, 41 patients with B-cell lymphoma<br />

received subcutaneous injections of autologous Id-KLH<br />

conjugates mixed with an oil-in-water emulsion containing<br />

non-ionic block polymers and threonyl-muramyl<br />

dipeptide. Approximately 50% of the patients generated<br />

specific anti-Id responses and isolated tumor regressions<br />

were observed. In particular, 11/16 patients had a significant<br />

increase in the frequency of tumor-specific cytotoxic<br />

T-lymphocytes precursor (CTLp). 237 The median<br />

duration of freedom from cancer-progression was significantly<br />

prolonged and this resulted in a survival advantage,<br />

especially in patients who generated cell-mediated<br />

anti-Id immunity.<br />

These pioneering studies did not prospectively investigate<br />

the effect of Id vaccines on tumor burden, since<br />

most patients were already in clinical remission, and<br />

standard tumor regression criteria could not be used. A<br />

recent study has directly evaluated the ability of Id vaccines<br />

to eradicate residual t(14;18) + lymphoma cells in<br />

20 patients in first remission after ProMACE-based<br />

chemotherapy. 238 These patients received multiple injections<br />

of Id /KLH conjugates in the presence of GM-CSF.<br />

Eight of eleven patients with detectable translocations<br />

in the peripheral blood converted to a PCR negative status<br />

after vaccination. Tumor-specific cytotoxic CD8+ and<br />

CD4 + T-cells were uniformly seen in most patients. Antibodies<br />

to autologous Id were also detected, but they<br />

were apparently not required for molecular remission<br />

since the latter was achieved in some patients without<br />

a detectable antibody response. Clinical monitoring indicates<br />

a 90% disease-free survival after a median followup<br />

of 3 years. This is encouraging compared with the<br />

44% disease-free survival (after a median follow-up of<br />

3 years) in another series of patients treated at the same<br />

Institution with anti-B4-blocked ricin in first remission<br />

after ProMACE-based chemotherapy. The encouraging<br />

results obtained in lymphoma patients have provided the<br />

rationale for exploring the use of Id vaccination in MM.<br />

Id vaccination in human MM<br />

MM has several biological features that can advantageously<br />

be exploited in the setting of active specific<br />

immunotherapy. Among others, pre-existing tumor-specific<br />

T-cell immunity, 229-232 the possibility of using clonal<br />

markers to track the fate of residual tumor cells, 239<br />

and the preserved susceptibility of chemoresistant<br />

myeloma cells to the effector mechanisms of cytolytic<br />

T-cells 240 may, together, represent a favorable basis for<br />

the efficacy of Id vaccines.<br />

In a pilot study, five MM patients were repeatedly<br />

immunized with autologous Id precipitated in aluminium<br />

phosphate suspension. 77 Four patients were previously<br />

untreated and one patient was in stable-partial<br />

remission following chemotherapy. Three patients developed<br />

specific anti-Id T- and B-cell responses, but these<br />

responses were transient and their magnitude was low.<br />

A more effective immunization schedule was developed<br />

with the goal of achieving long-lasting T-cell anti-Id<br />

immunity. This effort was focused on early stage MM,<br />

based on the assumption that Id-specific T-cells are present<br />

at higher frequency mainly in patients with early<br />

stage MM or MGUS. 231 Most of these Id-reactive T-cells<br />

are Th1-type cells in early stage MM, whereas they are<br />

predominantly Th2-type in patients with advanced disease.<br />

Thus, a more effective antitumor T-cell immune<br />

response may be expected if vaccines are delivered when<br />

the frequency of T-cells with the potential to develop<br />

cytotoxic activity is higher. In this series, patients<br />

received subcutaneous injections of autologous Id precipitated<br />

in aluminium phosphate suspension, together<br />

with free GM-CSF. 241 Long-lasting Id-specific T-cell<br />

responses were induced in all five immunized patients.<br />

Moreover, one patient showed a decrease of circulating<br />

Id upon immunization.<br />

A vaccination trial in which Id/KLH conjugates and<br />

GM-CSF were administered subcutaneously as a maintenance<br />

treatment after high-dose chemotherapy and<br />

PBPC infusion has recently been published. 242 Most<br />

patients generated Id-specific DTH reactions. DTH specificity<br />

was confirmed in one patient by investigating the<br />

reactivity to synthetic peptides derived from the VDJ<br />

sequence of the tumor-specific Ig heavy chain. In 3<br />

patients with minimal residual disease, the DTH skin<br />

tests remained positive up to two years after the last<br />

immunization, but residual tumor cells were not eliminated<br />

by these long-lasting immune responses. Nevertheless,<br />

these patients remained in clinical remission<br />

without any further maintenance treatment. Thus, it is<br />

possible to generate anti-Id immune responses that are<br />

not potent enough to eliminate residual tumor cells, but<br />

are sufficient to hold the disease in check for extended<br />

periods.<br />

These results have been confirmed in a preliminary<br />

report of 18 patients with MM receiving Id/KLH conjugates<br />

and GM-CSF in first remission after high-dose<br />

chemotherapy and tandem transplantation followed by<br />

PBPC infusions. In particular, 50% of the patients generated<br />

positive DTH reactions and 2 patients, in partial<br />

remission at the time of vaccination, achieved a complete<br />

remission following vaccination. A retrospective<br />

pair-mate analysis has shown a trend for a better clinical<br />

outcome in MM patients receiving Id vaccines compared<br />

to those receiving IFN-α alone. This tendency is<br />

particularly evident in patients who generated positive<br />

Id-specific T-cell responses. Although preliminary and<br />

retrospective, this is the first study providing clinical evidence<br />

that the generation of T-cell immune response<br />

against tumor cells may positively influence the clinical<br />

outcome of MM patients in the remission phase (N.<br />

Munshi and L. Kwak , personal communications).<br />

DC-based anti-Id vaccination<br />

As previously discussed, a proportion of lymphoma<br />

and MM patients enrolled in clinical trials mounted an<br />

Id-restricted antibody and T-cell response and some of<br />

them showed tumor regression. However, there are<br />

important differences between lymphoma cells and MM<br />

haematologica vol. 85(suppl. to n. 12):December 2000


176<br />

M. Bocchia et al.<br />

plasma cells. In the case of lymphomas, the cells are<br />

characterized by high surface expression and little antibody<br />

secretion whereas myeloma cells have very low<br />

levels of cell surface Ig with high levels of antibody<br />

secretion. Thus, it is unlikely that the sole generation of<br />

an antibody-based anti-Id immune response will be<br />

beneficial for MM patients. In fact, anti-Id antibodies<br />

may be blocked from reaching the tumor cells by the<br />

high levels of circulating Id. Moreover, despite the existence<br />

of a pre-plasma cell stem cell compartment in<br />

MM with a higher expression of surface Ig, it is possible<br />

that tumor cells would not express enough target<br />

protein for the antibodies to be effective. Conversely,<br />

an Id specific T-cell response would not need to bind to<br />

cell surface Ig to be active. T-cells do not recognize<br />

intact protein, but are specific for processed peptide<br />

fragments of the Id expressed on class I or II molecules.<br />

The advantage of a cytotoxic T-cell response is that it<br />

would not be blocked by free circulating paraprotein<br />

and would not depend on the expression of the native<br />

protein on the surface of tumor cells. Moreover, B-cells,<br />

including putative myeloma stem cells, are known to<br />

process and present peptides of the Ig on their membrane<br />

associated with class I and II molecules. Therefore,<br />

optimal strategies for Id vaccination may require the<br />

induction of a T-cell-mediated immune response which<br />

is best achieved by the use of APC. In this view, the rapid<br />

generation of a T-cell immunity in healthy volunteers<br />

after a single injection of mature DC has recently been<br />

described. 243 Nine normal subjects were given subcutaneous<br />

injections of monocyte-derived mature DC<br />

unpulsed or pulsed with KLH, tetanus toxoid (TT) or HLA-<br />

A*0201-positive restricted influenza matrix peptide.<br />

Four other individuals received these antigens without<br />

DC. Of note, administration of unpulsed DC or antigens<br />

alone failed to induce any T-cell response. Conversely, a<br />

CD4 + T-cell response was observed in 9/9 and 5/6 subjects<br />

injected with KLH or TT-pulsed DC, respectively.<br />

Moreover, a significant stimulation of effector and<br />

memory CD8 + CTLs was also reported. This feasibility trial<br />

provides the first controlled evidence of the capacity<br />

of DC to stimulate T-cell immunity.<br />

DC-based anti-Id vaccination has been reported In B-<br />

cell malignancies in a few papers. Hsu et al. have 244<br />

described 4 low-grade non-Hodgkin’s lymphoma (NHL)<br />

patients resistant to conventional chemotherapy or<br />

relapsed who were injected intravenously with Id-pulsed<br />

DC freshly isolated from PB by subsequent enrichment<br />

steps. A tumor-specific T-cell response was observed in<br />

all cases associated, in one case, with tumor regression.<br />

Sixteen patients have been treated so far and an anti-<br />

Id restricted cellular response has been observed in 8<br />

subjects (R. Levy, personal communication). The same<br />

strategy of targeting the Id has been applied by the same<br />

group to induce a T-cell immune response in MM<br />

patients. 245 Twelve patients were injected, 3 to 7 months<br />

after autologous stem cell transplantation, with Idpulsed<br />

DC followed by 5 subcutaneous boosts of Id/KLH<br />

administered with adjuvant. Whereas 11/12 patients<br />

developed a strong KLH-specific cellular proliferative<br />

response, thus suggesting immunocompetence after<br />

high-dose chemotherapy, only 2 individuals generated<br />

an anti-Id restricted T-cell proliferation and only 1/3<br />

patients showed a transient Id-specific CTLs response.<br />

This approach raises concerns about the efficacy of<br />

uncultured blood DC of stimulating efficiently T-cells,<br />

the capacity of Id-loaded DC to reach secondary lymphoid<br />

tissues to prime T-cells escaping the entrapment<br />

of the lungs and the role of Id-KLH boosts after DC<br />

administration.<br />

Wen et al. 246 reported immunization of one MM patient<br />

injected with Id-pulsed DC derived from adherent<br />

mononuclear cells in the presence of appropriate<br />

cytokines. In this paper, Id-specific T- cell proliferation<br />

and secretion of IFN-γ were reported, as were the production<br />

of anti-Id antibodies. Similar results have recently<br />

been reported by Cull et al. who treated two patients<br />

with advanced refractory myeloma with a series of four<br />

vaccinations using autologous Id-protein pulsed DC combined<br />

with adjuvant GM-CSF. 247 DC were derived from<br />

adherent mononuclear cells. Both patients generated a<br />

specific T-cell proliferative response that was associated<br />

with the production of IFN-γ, indicating a Th-1-like<br />

response. However, no Id-specific cytotoxic T-cell<br />

response could be demonstrated. Lim and Bailey-Wood<br />

have also treated 6 MM with DC generated from adherent<br />

mononuclear cells. 248 DC were pulsed with the autologous<br />

Id and KLH as a control vaccine. All patients developed<br />

both B- and T- cell responses to KLH, suggesting the<br />

integrity of the host immune system. Id-specific responses<br />

were also observed. In one patient, a modest but consistent<br />

drop in the serum Id level was observed. Lastly, a<br />

vaccine formulation based on CD34 stem cell-derived DC<br />

pulsed with Id-derived peptides has recently been used in<br />

11 MM patients with advanced disease. 249 Five patients<br />

generated Id-specific immune responses and one patient<br />

showed a decreased plasma cell infiltration in the bone<br />

marrow.<br />

New strategies in Id vaccination<br />

The generation of an effective antitumor response<br />

greatly depends on the final activation of tumor-specific<br />

cytolytic CD8 cells. This is the final event resulting from a<br />

series of cognate and non-cognate interactions occurring<br />

among tumor cells, professional APC, CD4, and CD8 cells.<br />

Each of these cell populations plays a unique role, and<br />

may represent a possible target of immune intervention to<br />

improve the efficacy of vaccination. Malignant B-cells can<br />

be modified to become efficient APCs themselves and present<br />

peptides from their own tumor-specific antigens to<br />

autologous T cells. To this end, a number of strategies are<br />

currently under preclinical and clinical evaluation. One<br />

possibility is to fuse tumor cells with dendritic cells. The<br />

fusion product will combine the functional properties of<br />

DC with the full antigenic repertoire of tumor cells. 250 As<br />

an alternative, malignant B-cells can be turned into effective<br />

APC by stimulating cell surface CD40 with its specific<br />

counter-receptor CD40 ligand. 251,252 Genetic engineering<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

177<br />

is another approach that may turn malignant B-cells into<br />

effective APC. Transfection with immunologically relevant<br />

DNA sequences coding for cytokines or co-stimulatory<br />

molecules greatly enhances the ability of malignant B-<br />

cells to activate antitumor immune responses. This<br />

approach has recently been used in MM taking advantage<br />

of the selective expression of functional adenoviral receptors<br />

on the cell surface of myeloma cells. 253<br />

Interestingly, there has been a description 254 of the<br />

immunization of a matched related donor of allogeneic<br />

bone marrow with the myeloma derived Id (conjugated<br />

with KLH) isolated pretransplant from patients. After<br />

transplantation, a CD4 + T-cell line was established from<br />

the peripheral blood of the recipient and found to be of<br />

donor origin and proliferate specifically in response to<br />

the myeloma Id. Thus, this experience demonstrates the<br />

principle of transfer of donor immunity with the advantage<br />

of immunizing a tumor naive donor who may be<br />

more likely to be able to generate an immune response<br />

against the tumor Ig without interference or suppression<br />

by the malignant cells. However, the lack of an anti-Id<br />

CTL response and ethical concerns on the immunization<br />

of healthy donors with tumor-derived products, suggest<br />

that in the future an alternative strategy based upon the<br />

generation, ex vivo, of Id-reactive CTL clones by means of<br />

APC, will be needed.<br />

The prerequisite for any vaccine-based strategy is the<br />

possibility of differentiating tumor cells from normal cells.<br />

Id is absolutely tumor-specific, but is not directly related<br />

to the malignant phenotype of myeloma cells, and is<br />

self-Ag. As such, it is protected by self-tolerance mechanisms.<br />

There is a growing list of alternative tumor-specific<br />

antigens that can be exploited as targets for active<br />

specific immunotherapy. Among others, the core protein<br />

of Muc-1, 255,256 antigens encoded by MAGE-type genes, 257<br />

overexpressed or fusion proteins resulting from chromosomal<br />

abnormalities may all represent alternative<br />

immunogens. 258,259 Compared to Id, some of these antigens<br />

may be more intrinsically related to the malignant<br />

phenotype of tumor cells.<br />

Polymorphism of the HLA molecules is a major obstacle<br />

in the outcome of vaccines aimed at triggering<br />

cytolytic T-cell responses. By combining amino acid<br />

sequencing of tumor-specific antigens and HLA typing, it<br />

is now possible to predict whether the HLA alleles of a<br />

given individual can bind tumor-derived peptides. Several<br />

groups are planning to use Id vaccination only in those<br />

patients for whom preliminary sequencing demonstrates<br />

a compatible restriction between HLA and peptides.<br />

Finally, a new generation of immunogens has been<br />

developed using DNA-based technologies. The whole<br />

tumor-specific immunoglobulin or the variable region<br />

sequences of both heavy- and light-chains have been used<br />

as immunogens or to produce recombinant proteins in<br />

bacteria. However, it has soon become clear that naked<br />

DNA is not immunogenic per se and additional sequences<br />

are required to elicit protective immune responses. 260<br />

Sequences coding for cytokines, chemokines or xenogeneic<br />

proteins have been included in these constructs<br />

and used as adjuvants. 261-264 Experimental data indicate<br />

that these fusion genes have indeed the potential to raise<br />

protective immunity in both NHL and MM.<br />

Vaccine trials in chronic myelogenous<br />

leukemia<br />

Chronic myelogenous leukemia (CML) is a biphasic<br />

neoplastic disorder with a prolonged indolent phase lasting<br />

an average of 4 years followed by an acute phase of<br />

blastic transformation which inevitably leads rapidly to<br />

death. There is no curative therapy for CML other than<br />

allogeneic bone marrow transplantation, an option that<br />

is available only to a small fraction of patients who have<br />

both a matched donor and are young enough to tolerate<br />

the procedure.<br />

Recently, IFN-α has been shown to induce hematologic<br />

remissions in most CML patients, with a relevant portion<br />

of them also experiencing several degrees of cytogenetic<br />

response and ultimately a statistically significant prolongation<br />

of their chronic phase. However, still too few CML<br />

patients are long survivors if not cured regardless of the<br />

treatment option they received. Because of the unique features<br />

of this disease, the hallmark translocation that characterizes<br />

all neoplastic cells, a therapeutic targeting<br />

approach only the Ph+ clone could be a powerful tool in<br />

the treatment of CML.<br />

The first direct evidence of the immune system’s crucial<br />

role in recognizing and eliminating Ph+ CML cells<br />

came from the demonstration that infusion of large doses<br />

of peripheral blood leukocytes from the marrow donor<br />

induced durable remission in patients with CML who had<br />

relapsed following a T cell depleted marrow allograft. 265,266<br />

This latter finding proved that in CML the graft-versustumor<br />

effect is mediated by the cellular arm of the<br />

immune system. While the nature of the response is likely<br />

to be largely allogeneic a possible role for specific anti-<br />

CML responses is suggested by the lack of this observation<br />

in patients with other myeloid leukemias undergoing<br />

the same treatment. 267<br />

The hypothesis that CML cells could be recognized by<br />

the immune system through the presentation of P210,<br />

the tumor-specific product of the bcr/abl hybrid gene,<br />

was first tested. The evidence that P210 b3a2-breakpoint<br />

peptides were able to bind HLA class I and HLA class II<br />

molecules and to elicit specific T cell responses in normal<br />

donors provided the rationale for a peptide vaccine in<br />

CML patients. 103-106 Pinilla-Ibarz et al. 124 have recently<br />

completed a phase I dose escalation trial (5 doses over 10<br />

weeks) of a multivalent peptide vaccine (5 peptides) plus<br />

QS21 in patients with CML and b3a2 breakpoint. Patient<br />

characteristics included hematologic remission, IFN-α<br />

therapy and no HLA restriction. In a preliminary report the<br />

peptide vaccine appeared safe with patients experiencing<br />

only minimal discomfort at the site of injection.<br />

With regards to the immune response, peptide-specific<br />

delayed hypersensitivity (DTH) in vivo and peptide-specific<br />

proliferation in vitro were shown but no peptidespecific<br />

CTL response was induced. Bocchia et al. are currently<br />

conducting a multicenter phase I/II trial of a pen-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


178<br />

M. Bocchia et al.<br />

tavalent peptide vaccine plus QS21 and GM-CSF in b3a2-<br />

CML patients expressing any of HLA A3, A11 B8 or DR11.<br />

Patient characteristics also include major or complete<br />

cytogenetic response with or without IFN-α maintaining<br />

therapy. The protocol comprises 6 s.c. vaccinations with<br />

peptides + QS21 at 2 weekly intervals, with GM-CSF<br />

injected at the vaccine site for 4 consecutive days starting<br />

the day before each vaccination. Goals of the study<br />

are the evaluation of the induction of peptide-specific T-<br />

cell response, of the role of GM-CSF as immunologic<br />

adjuvant in CML patients and the impact of the peptide<br />

vaccine on minimal residual disease.<br />

As in other cancers, the use of DC as powerful inducers<br />

of an active specific immune response in CML is now<br />

under in vitro evaluation. 107,268 Interestingly, most CMLderived<br />

DC carry the t(9;22) translocation and therefore<br />

could naturally present P210 derived peptides. In fact,<br />

CML derived Ph + DC were able to strongly stimulate<br />

autologous T-cells that displayed vigorous cytotoxicity<br />

activity against autologous CML cells but low reactivity<br />

to HLA-matched normal bone marrow cells or autologous<br />

remission state bone marrow mononuclear cells. 206<br />

P210-derived peptides could have a role in inducing this<br />

leukemia-specific response and a vaccine strategy which<br />

combines Ph + DC and breakpoint peptides will be investigated.<br />

Conclusions<br />

This review shows that there are many prospects of<br />

curing cancer through the active induction of a specific<br />

immune response to TAA. The terms of the matter are<br />

now defined with molecular and genetic details for<br />

melanomas. Ongoing research is aimed at defining TAA<br />

on other forms of tumors. Indeed, experimental data and<br />

very recent clinical evidence suggest that antitumor vaccines<br />

will soon be a new form of tumor treatment that<br />

will be able to be adopted for the management of<br />

defined stages of neoplastic disease, in sequential association<br />

with conventional treatments. 269<br />

Prediction of when the efficacy of antitumor vaccination<br />

will be assessed and will become a routine procedure<br />

is beyond a simple scientific evaluation. While preclinical<br />

research has identified several possible targets<br />

and strategies for tumor vaccination, the clinical scenario<br />

is far more complex and as yet no specific clue has<br />

emerged to clearly envisage a clinical development strategy<br />

which could make biotechnology investments in this<br />

area attractive enough to pharmaceutical companies.<br />

Patent issue complexity further contributes to slowing<br />

down the development of expensive clinical trial programs.<br />

A cautious, yet attentive attitude seems, at the<br />

moment, to be the general behavior of the pharmaceutical<br />

industry.<br />

At present peptide vaccination may appear of more<br />

immediate application. Several phase I clinical trials have<br />

already been carried out using synthetic peptides from<br />

defined TAA. These peptides have been administered<br />

alone 123 or combined with adjuvants, or presented by<br />

monocytes or DC. 269 Nearly all studies indicate that this<br />

form of vaccination is well tolerated, mild fever and<br />

inflammation at the site of injection being the only occasional<br />

side effects observed. Nonetheless it should be<br />

pointed out that there is usually a poor correlation<br />

between peptide ability to induce a T-cell response and<br />

clinical response. Among the several reasons that may<br />

account for this discrepancy, the choice of the peptides<br />

may have a critical importance. Most peptide-based vaccines<br />

have considered HLA class I restricted peptides<br />

only, whereas there is increasing evidence that tumorspecific<br />

CD4 + T-cells may be important in inducing an<br />

effective antitumor immunity. The addition of peptides<br />

that bind class II HLA glycoproteins to peptide vaccines<br />

could lead to an amplification of the immune response<br />

as well as to better clinical effect.<br />

A survey of the outcomes of vaccination trials shows<br />

that the poor correlation between induction of immunologic<br />

responses and the clinical results is a consistent<br />

finding, independently of the immunizing strategy<br />

adopted. Many factors may contribute to this poor correlation,<br />

e.g.:<br />

a) the selection of the patients enrolled in the trial:<br />

tumor burden, stage of disease, and others, as discussed<br />

above;<br />

b) the techniques used for the immune monitoring in<br />

vitro: most of the current studies evaluate T-cell induction<br />

through in vitro peptide stimulation of PBMC, while<br />

the use of tetrameric soluble class I-peptide complexes<br />

270 or reverse solid phase ELISPOT analysis 271 may provide<br />

complementary information;<br />

c) the assays for immune monitoring in vivo: often a<br />

positive DTH test does not correlate with evident tumor<br />

regression.<br />

Perhaps, fine needle biopsies at the site of regressing<br />

and non-regressing tumors could provide a more direct<br />

insight into the events associated with the clinical outcome.<br />

The immune pattern within the tumor should be<br />

compared to the one found in the skin. This should allow<br />

evaluation of the exact role of vaccine-induced T-cells.<br />

Which of the existing in vitro and in vivo assays correlates<br />

most accurately with clinical responses remains to<br />

be established.<br />

Several recent studies showing that the immune system<br />

recognizes TAA during tumor growth did not clarify<br />

whether such recognition was indeed associated with<br />

subsequent tumor cell destruction. The development of<br />

reliable assays for efficient monitoring of the state of<br />

immunization of cancer patients against TAA is as an<br />

important goal that will markedly affect the progress of<br />

antitumor vaccines. A major problem in testing the efficacy<br />

of antitumor vaccination in adjuvant settings<br />

depends on both the long period of time and large number<br />

of patients required. The possibility of effectively<br />

monitoring the immune response induced acquires critical<br />

importance since it may provide a much earlier surrogate<br />

end-point, predictive of the clinical outcome.<br />

The downregulation of the expression of TAA represents<br />

another crucial issue for vaccination therapies,<br />

since it may lead to the immunoselection of tumor cell<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

179<br />

clones that hide the target TAA. An ideal TAA is a protein<br />

that is essential for sustaining the malignant phenotype,<br />

and that is not stripped or downmodulated by<br />

the immune reaction. Mutations that give rise to TAA of<br />

this kind have been described. 272 However, they will be<br />

an appropriate target only for the tumors expressing<br />

these particular mutations and will not be suitable for<br />

more general cancer vaccines. Improvements in the<br />

identification of tumor-associated mutations that may<br />

be potentially recognized by the immune system may<br />

also open up the possibility of tailoring individual cancer<br />

vaccines. The recent report of the construction of<br />

fusion cells composed of autologous tumor cells and DC<br />

or TAA pulsed-DC represents a step forward towards the<br />

quick manufacture of tumor-specific and individualized<br />

vaccines. In contrast, the characterization of the telomerase<br />

catalytic subunit (hTERT) expressed in more than<br />

85% of human cancers appears to open the way to a<br />

novel strategy for a general anticancer vaccination targeting<br />

a widely distributed TAA. 273<br />

The in vivo or ex vivo introduction of TAA genes into DC<br />

through recombinant viral vectors is still hampered by<br />

the lack of an ideal viral vector and by the induction of<br />

an immune response against the viral proteins. Nonetheless,<br />

many viral vectors successfully used in animal models<br />

and currently tested in clinical trials appear to be safe<br />

vehicles for gene transfer, without any major toxicity. To<br />

control transgene expression levels better, investigators<br />

are exploiting tissue- or cell-specific regulatory elements<br />

such as cytomegalovirus promoters and enhancers that<br />

are preferentially expressed in tumor cells.<br />

Certainly the new prospects opened by antitumor vaccines<br />

are fascinating. When compared with conventional<br />

cancer management, vaccination is a soft, noninvasive<br />

treatment free from particular distress and<br />

iatrogenic side effects. Antitumor vaccines can be<br />

expected to have a considerable social impact, but a<br />

few large clinical trials enrolling the appropriate patients<br />

are now necessary to assess their efficacy.<br />

This review will end considering their use not in the<br />

treatment of cancer patients but to prevent cancer in<br />

healthy persons, a so far neglected prospect. Current<br />

studies are leading to the detection of gene mutations<br />

that predispose to cancer. 274 Identification of the gene<br />

at risk and its mutated or amplified products would provide<br />

the opportunity to vaccinate susceptible subjects<br />

against their foreseeable cancer. Molecular characterization<br />

of altered gene products predictably destined to<br />

become a tumor antigen will be the first step towards<br />

the engineering of effective vaccines to be used for this<br />

purpose. 25<br />

An unrestrained imagination may picture an even<br />

broader application of antitumor vaccines, i.e. their use<br />

to prevent tumors in the general population. Molecular<br />

and genetic data suggest that the number of TAA is not<br />

endless. Several of the TAA detected so far are shared by<br />

histologically distinct tumors arising in different organs<br />

(Table 1). The possibility of vaccinating against most<br />

common human cancers by using not many more than<br />

twenty TAA may perhaps be conceivable. Experimental<br />

data suggest that the immunity elicited by specific vaccination<br />

is much more effective in the inhibition of<br />

incipient tumors than in the cure of those that have<br />

already progressed. 275 The risk of inducing an autoimmune<br />

disease would be a major worry since not rarely<br />

antigens acting as TAA are expressed by normal tissues.<br />

276 This risk would be much harder to accept when<br />

treating healthy individuals than in the vaccination of<br />

cancer patients, where the scales of risk-benefit are<br />

biased by a short life-expectancy. On the other hand<br />

failure to intervene when a disease so diffuse and dramatic<br />

as cancer can be prevented could also be viewed<br />

as harmful. 277 Lastly, it should be considered that the<br />

same or even a higher risk of inducing autoimmune<br />

reactions is associated with many antimicrobial vaccines.<br />

Fortunately, they started to be used when this risk<br />

was not yet perceived.<br />

In conclusion, even if cancer vaccines are an old<br />

dream, 278 only recently has their design become a rational<br />

enterprise. There are now many ways of constructing<br />

vaccines able to elicit a strong protective immunity.<br />

This progress is offering ground for optimism.<br />

Contributions and Acknowledgments<br />

The authors were a group of experts and representatives<br />

of two pharmaceutical companies, Amgen Italia<br />

Spa and Dompé Biotech Spa, both from Milan, Italy. This<br />

co-operation between a medical journal and pharmaceutical<br />

companies is based on the common aim of<br />

achieving optimal use of new therapeutic procedures in<br />

medical practice.<br />

Funding<br />

The preparation of this manuscript was supported by<br />

educational grants from Dompé Biotech Spa and Amgen<br />

Italia Spa.<br />

Disclosures<br />

Conflict on interest: Dompé Biotec Spa sells G-CSF and<br />

rHuEpo in Italy, and Amgen Italia Spa has a stake in Dompé<br />

Biotec Spa.<br />

Redundant publications: no substantial overlapping<br />

with previous papers.<br />

Manuscript processing<br />

Manuscript received August 7, 2000; accepted October<br />

24, 2000.<br />

References<br />

1. Bordignon C, Carlo-Stella C, Colombo MP, et al. Cell<br />

therapy: achievements and perspectives. <strong>Haematologica</strong><br />

1999; 84:1110-49.<br />

2. Ada GL. The immunological principles of vaccination.<br />

Lancet 1990; 335:523-6.<br />

3. Sprent J. Lifespan of naive, memory and effector lymphocytes.<br />

Curr Opin Immunol 1993; 5:433-8.<br />

4. Rabinovich NR, McInnes P, Klein DL, Hall BF. Vaccine<br />

technologies: view to the future. Science 1994;<br />

265:1401-4.<br />

5. Medzhitov R, Janeway CA Jr. Innate immunity: the<br />

virtues of a nonclonal system of recognition. Cell<br />

haematologica vol. 85(suppl. to n. 12):December 2000


180<br />

M. Bocchia et al.<br />

1997; 91:295-8.<br />

6. Matzinger P. Tolerance, danger, and the extended<br />

family. Annu Rev Immunol 1994; 12:991-1045.<br />

7. Musiani P, Modesti A, Giovarelli M, et al. Cytokines,<br />

tumor-cell death and immunogenicity: a question of<br />

choice. Immunol Today 1997; 18:32-6.<br />

8. Pardoll DM. Cancer vaccines: a road map for the next<br />

decade. Curr Opin Immunol 1996; 8:619-20.<br />

9. Ehrlich P. In: Collected papers of Paul Ehrlich. London:<br />

Pergamon Press; 1960.<br />

10. Snell GD. The histocompatibility systems. Transplant<br />

Proc 1971; 3:1133-8.<br />

11. Gross L. Intradermal immunization of C3H mice<br />

against a sarcoma that originated in an animal of the<br />

same line. Cancer Res 1963; 3:326-33.<br />

12. Klein G, Sjogren HO, Klein E. Demonstration of resistance<br />

against methylcholanthrene-induced sarcomas<br />

in the primary autochthonous host. Cancer Res 1960;<br />

20:1561-72.<br />

13. Jaffe EM, Pardoll DM. Murine tumor antigens: is it<br />

worth the search? Curr Opin Immunol 1996; 8:622-<br />

7.<br />

14. Hewitt HB, Blake ER, Walder AS. A critique of the evidence<br />

for active host defence against cancer, based on<br />

personal studies of 27 murine tumors of spontaneous<br />

origin. Br J Cancer 1976; 33:241-59.<br />

15. Boon T. Antigenic tumor cell variants obtained with<br />

mutagens. Adv Cancer Res 1983; 39:121-51.<br />

16. De Plaen E, Lurquin C, Van Pel A, et al. Immunogenic<br />

(tum-) variants of mouse tumor P185: cloning the<br />

gene of tum- antigen P91A and identification of the<br />

tum- mutation. Proc Natl Acad Sci USA 1988;<br />

85:2274-8.<br />

17. Traversari C, van der Bruggen P, Luescher IF, et al. A<br />

nonapeptide encoded by human gene MAGE-1 is recognized<br />

on HLA-A1 by cytolytic T lymphocytes directed<br />

against tumor antigen MZ2-E. J Exp Med 1992;<br />

176:1453-7.<br />

18. Coulie PG. Human tumour antigens recognized by T<br />

cells: new perspectives for anti-cancer vaccines? Mol<br />

Med Today 1997; 3:261-8.<br />

19. Robbins PF, Kawakami Y. Human tumor antigens recognized<br />

by T cells. Curr Opin Immunol 1996; 8:628-<br />

35.<br />

20. Nossal GJV. The case history of Mr. T.I. - terminal<br />

patient or still curable. Immunol Today 1980; 1:5-6.<br />

21. Vonderheide RH, Hahn WC, Schultze JL, Nadler LM.<br />

The telomerase catalytic subunit is a widely expressed<br />

tumor-associated antigen recognized by cytotoxic T<br />

lymphocytes. Immunity 1999; 10:673-9.<br />

22. Lanzavecchia A. Identifying strategies for immune<br />

intervention. Science 1993; 260:937-44.<br />

23. Colombo MP, Forni G. Immunotherapy I: Cytokine<br />

gene transfer strategies. Cancer Metast Rev 1996;<br />

15:317-28.<br />

24. Immunity future, Science, http://www.aaas.org/science/immunology.<br />

25. Lollini PL, Forni G. Specific and nonspecific immunity<br />

in the prevention of spontaneous tumours.<br />

Immunol Today 1999; 20:347-50.<br />

26. Nanni P, Forni G, Lollini PL. Cytokine gene therapy:<br />

hopes and pitfalls. Ann Oncol 1999; 10:261-6.<br />

27. Colombo MP, Forni G. Cytokine gene transfer in<br />

tumor inhibition and tumor therapy: where are we<br />

now ? Immunol Today 1994; 15:48-51.<br />

28. Allione A, Consalvo M, Nanni P, et al. Immunizing<br />

and curative potential of replicating and nonreplicating<br />

murine mammary adenocarcinoma cells engineered<br />

with interleukin (IL)-2, IL-4, IL6, IL-7, IL-10,<br />

tumor necrosis factor α, granulocyte-macrophage<br />

colony-stimulating factor, and γ-interferon gene or<br />

admixed with conventional adjuvants. Cancer Res<br />

1994; 54:6022-6.<br />

29. Cavallo F, Signorelli P, Giovarelli M, et al. Antitumor<br />

efficacy of adenocarcinoma cells engineered to produce<br />

interleukin 12 (IL-12) or other cytokines compared<br />

with exogenous IL-12. J Natl Cancer I 1997;<br />

89:1049-58.<br />

30. Cavallo F, Di Carlo E, Butera M, et al. Immune events<br />

associated with the cure of established tumors and<br />

spontaneous metastases by local and systemic interleukin<br />

12. Cancer Res 1999; 59:414-21.<br />

31. Janeway CA Jr, Bottomly K. Signals and signs for lymphocyte<br />

responses. Cell 1994; 76:275-85.<br />

32. Banchereau J, Steinman RM. Dendritic cells and the<br />

control of immunity. Nature 1998; 392:245-52.<br />

33. Kitamura H, Iwakabe K, Yahata T, et al. The natural<br />

killer T (NKT) cell ligand alpha-galactosylceramide<br />

demonstrates its immunopotentiating effect by inducing<br />

interleukin (IL)-12 production by dendritic cells<br />

and IL-12 receptor expression on NKT cells. J Exp Med<br />

1999; 189:1121-8.<br />

34. Cayeux S, Richter G, Becker C, Pezzutto A, Dorken B,<br />

Blankenstein T. Direct and indirect T cell priming by<br />

dendritic cell vaccines. Eur J Immunol 1999; 29:225-<br />

34.<br />

35. Sallusto F, Lanzavecchia A, Mackay CR. Chemokines<br />

and chemokine receptors in T-cell priming and<br />

Th1/Th2-mediated responses. Immunol Today 1998;<br />

19:568-74.<br />

36. Forni G, Cavallo F, Consalvo M, et al. Molecular<br />

approaches to cancer immunotherapy. Cytokines Mol<br />

Ther 1995; 1:225-48.<br />

37. Peron JM, Esche C, Subbotin VM, Maliszewski C,<br />

Lotze MT, Shurin MR. FLT3-ligand administration<br />

inhibits liver metastases: role of NK cells. J Immunol<br />

1998; 161:6164-70.<br />

38. Simons JW, Jaffee EM, Weber CE, et al. Bioactivity of<br />

autologous irradiated renal cell carcinoma vaccines<br />

generated by ex vivo granulocyte-macrophage colonystimulating<br />

factor gene transfer. Cancer Res 1997;<br />

57:1537-46.<br />

39. Soiffer R, Lynch T, Mihm M, et al. Vaccination with<br />

irradiated autologous melanoma cells engineered to<br />

secrete human granulocyte-macrophage colony-stimulating<br />

factor generates potent antitumor immunity in<br />

patients with metastatic melanoma. Proc Natl Acad<br />

Sci USA 1998; 95:13141-6.<br />

40. Sun Y, Jurgovsky K, Moller P, et al. Vaccination with IL-<br />

12 gene-modified autologous melanoma cells: preclinical<br />

results and a first clinical phase I study. Gene<br />

Ther 1998; 5:481-90.<br />

41. Kirk AD, Burkly LC, Batty DS, et al. Treatment with<br />

humanized monoclonal antibody against CD154 prevents<br />

acute renal allograft rejection in nonhuman primates.<br />

Nat Med 1999; 5:686-93.<br />

42. Larsen CP, Elwood ET, Alexander DZ, et al. Long-term<br />

acceptance of skin and cardiac allografts after blocking<br />

CD40 and CD28 pathways. Nature 1996; 381:<br />

434-8.<br />

43. Blazar BR, Taylor PA, Noelle RJ, Vallera DA. CD4(+)<br />

T cells tolerized ex vivo to host alloantigen by anti-<br />

CD40 ligand (CD40L:CD154) antibody lose their<br />

graft-versus-host disease lethality capacity but retain<br />

nominal antigen responses. J Clin Invest 1998;<br />

102:473-82.<br />

44. Diehl L, den Boer AT, Schoenberger SP, van der Voort<br />

EI, Schumacher TN, Melief CJ. CD40 activation in vivo<br />

overcomes peptide-induced peripheral cytotoxic T-<br />

lymphocyte tolerance and augments anti-tumor vaccine<br />

efficacy. Nat Med 1999; 5:774-9.<br />

45. Sotomayor EM, Borrello I, Tubb E, et al. Conversion<br />

of tumor-specific CD4+ T-cell tolerance to T-cell priming<br />

through in vivo ligation of CD40. Nat Med 1999;<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

181<br />

5:780-7.<br />

46. Kato K, Cantwell MJ, Sharma S, Kipps TJ. Gene transfer<br />

of CD40-ligand induces autologous immune<br />

recognition of chronic lymphocytic leukemia B cells. J<br />

Clin Invest 1998; 5:1133-41.<br />

47. Leach DR, Krummel MF, Allison JP. Enhancement of<br />

antitumor immunity by CTLA-4 blockade. Science<br />

1996; 271:1734-6.<br />

48. Yang YF, Zou JP, Mu J, et al. Enhanced induction of<br />

antitumor T-cell responses by cytotoxic T lymphocyteassociated<br />

molecule-4 blockade: the effect is manifested<br />

only at the restricted tumor-bearing stages.<br />

Cancer Res 1997; 57:4036-41.<br />

49. van Elsas A, Hurwitz AA, Allison JP. Combination<br />

immunotherapy of B16 melanoma using anti-cytotoxic<br />

T lymphocyte-associated antigen 4 (CTLA-4)<br />

and granulocyte/macrophage colony-stimulating factor<br />

(GM-CSF)-producing vaccines induces rejection<br />

of subcutaneous and metastatic tumors accompanied<br />

by autoimmune depigmentation. J Exp Med 1999;<br />

190:355-66.<br />

50. Hurwitz AA, Yu TF, Leach DR, Allison JP. CTLA-4<br />

blockade synergizes with tumor-derived granulocytemacrophage<br />

colony-stimulating factor for treatment<br />

of an experimental mammary carcinoma. Proc Natl<br />

Acad Sci USA 1998; 95:10067-71.<br />

51. Young JW. Dendritic cells: expansion and differentiation<br />

with hematopoietic growth factors. Curr Opin<br />

Hematol 1999; 6:135-44.<br />

52. Rescigno M, Granucci F, Citterio S, Foti M, Ricciardi-<br />

Castagnoli P. Coordinated events during bacteriainduced<br />

DC maturation. Immunol Today 1999;<br />

20:200-3.<br />

53. Kapsenberg ML, Hilkens CM, Wierenga EA, Kalinski P.<br />

The paradigm of type 1 and type 2 antigen presenting<br />

cells. Implications for atopic allergy. Clin Exp Allergy<br />

1999; 29 (Suppl) 2:33-6.<br />

54. Cella M, Scheidegger D, Palmer-Lehmann K, Lane P,<br />

Lanzavecchia A, Alber G. Ligation of CD40 on dendritic<br />

cells triggers production of high levels of interleukin-12<br />

and enhances T cell stimulatory capacity:<br />

T-T help via APC activation. J Exp Med 1996;<br />

184:747-52.<br />

55. Rissoan MC, Soumelis V, Kadowaki N, et al. Reciprocal<br />

control of T helper cell and dendritic cell differentation.<br />

Science 1999; 283:1183-6.<br />

56. Wong BR, Josien R, Lee SY, et al. TRANCE (tumor<br />

necrosis factor [TNF]-related activation-induced<br />

cytokine), a new TNF family member predominantly<br />

expressed in T cells, is a dendritic cell-specific survival<br />

factor. J Exp Med 1997; 186:2075-80.<br />

57. Anderson DM, Maraskovsky E, Billingsley WL, et al. A<br />

homologue of the TNF receptor and its ligand<br />

enhance T-cell growth and dendritic-cell function.<br />

Nature 1997; 390:175-9.<br />

58. Dubois B, Massacrier C, Vanbervliet B, et al. Critical<br />

role of IL-12 in dendritic cell-induced differentiation of<br />

naive B lymphocytes. J Immunol 1998;161:2223-31.<br />

59. Caux C, Liu YJ, Banchereau J. Recent advances in the<br />

study of dendritic cells and follicular dendritic cells.<br />

Immunol Today 1995; 16:2-4.<br />

60. Cao L, Kulmburg P, Veelken H, et al. Cytokine gene<br />

transfer in cancer therapy. Stem Cells 1998; 16(Suppl<br />

1):251-60.<br />

61. Colombo MP, Lombardi L, Melani C, et al. Hypoxic<br />

tumor cell death and modulation of endothelial adhesion<br />

molecules in the regression of granulocyte colonystimulating<br />

factor-transduced tumors. Am J Pathol<br />

1996; 148:473-83.<br />

62. Morel S, Lévy F, Burlet-Schiltz O, et al. Processing of<br />

some antigens by the standard proteasome but not by<br />

the immunoproteasome results in poor presentation<br />

by dendritic cells. Immunity 2000; 12:107-17.<br />

63. Groettrup M, Schmidtke G. Selective proteasome<br />

inhibitors: modulators of antigen presentation ? Drug<br />

Discov Today 1999; 4:63-71.<br />

64. Morton DL, Foshag LJ, Hoon DS, et al. Prolongation<br />

of survival in metastatic melanoma after active specific<br />

immunotherapy with a new polyvalent melanoma vaccine.<br />

Ann Surg 1992; 216:463-82.<br />

65. Livingston PO, Wong GY, Adluri S, et al. Improved<br />

survival in stage III melanoma patients with GM2 antibodies:<br />

a randomized trial of adjuvant vaccination<br />

with GM2 ganglioside. J Clin Oncol 1994; 12:1036-<br />

44.<br />

66. Berd D, Maguire HC Jr, Schuchter LM, et al. Autologous<br />

hapten-modified melanoma vaccine as postsurgical<br />

adjuvant treatment after resection of nodal<br />

metastases. J Clin Oncol 1997; 15:2359-70.<br />

67. Sensi M, Farina C, Maccalli C, et al. Clonal expansion<br />

of T lymphocytes in human melanoma metastases<br />

after treatment with a hapten-modified autologous<br />

tumor vaccine. J Clin Invest 1997; 99:710-7.<br />

68. Boon T, van der Bruggen P. Human tumor antigens<br />

recognized by T lymphocytes. J Exp Med 1996; 183:<br />

725-9.<br />

69. Huang YC, Golumbeck P, Ahmadzadeh M, Jaffee E,<br />

Pardoll D, Levitsky H. Role of bone marrow-derived<br />

cells in presenting MHC class I-restricted tumor antigens.<br />

Science 1994; 264:961-5.<br />

70. Fujiwara H, Aoki H, Yoshioka T, Tomita S, Ikegami R,<br />

Hamaoka T. Establishment of a tumor-specific<br />

immunotherapy model utilizing TNP-reactive helper<br />

T cell activity and its application to the autochthonous<br />

tumor system. J Immunol 1984; 133:509-14.<br />

71. Sallusto F, Lanzavecchia A. Efficient presentation of<br />

soluble antigen by cultured human dendritic cells is<br />

maintained by granulocyte/macrophage colony-stimulating<br />

factor plus interleukin 4 and downregulated by<br />

tumor necrosis factor α. J Exp Med 1994; 179:1109-<br />

18.<br />

72. van Kooten C, Banchereau J. Functions of CD40 on B<br />

cells, dendritic cells and other cells. Curr Opin<br />

Immunol 1997; 9:330-7.<br />

73. Chiodoni C, Paglia P, Stoppacciaro A, Rodolfo M,<br />

Parenza M, Colombo MP. Dendritic cells infiltrating<br />

tumors cotransduced with granulocyte-macrophage<br />

colony-stimulating factor (GM-CSF) and CD40 ligand<br />

genes take up and present endogenous tumorassociated<br />

antigens, and prime naive mice for a cytotoxic<br />

T lymphocyte response. J Exp Med 1999;<br />

190:125-33.<br />

74. Albert ML, Bhardwaj N. Resurrecting the dead: DCs<br />

cross-present antigen derived from apoptotic cells on<br />

MHC I. The Immunologist 1998; 5:194-8.<br />

75. Melcher A, Todryk S, Hardwick N, Ford M, Jacobson<br />

M, Vile RG. Tumor immunogenicity is determined by<br />

the mechanism of cell death via induction of heat<br />

shock protein expression. Nat Med 1998; 4:581-7.<br />

76. Srivastava PK, Menoret A, Basu S, Binder RJ,<br />

McQuade KL. Heat shock proteins come of age: primitive<br />

functions acquire new roles in an adaptive world.<br />

Immunity 1998; 8:657-65.<br />

77. Bergenbrant S, Yi Q, Osterborg A, Bjorkholm M, et al.<br />

Modulation of anti-idiotypic immune response by<br />

immunization with the autologous M-component<br />

protein in multiple myeloma patients. Br J Haematol<br />

1996; 92:840-6.<br />

78. Adluri S, Gilewski T, Zhang S, Ramnath V, Ragupathi<br />

G, Livingston P. Specificity analysis of sera from breast<br />

cancer patients vaccinated with MUC1-KLH plus QS-<br />

21. Br J Cancer 1999; 79:1806-12.<br />

79. Kwak LW, Pennington R, Boni L, Ochoa AC, Robb<br />

RJ, Popescu MC. Liposomal formulation of a self lym-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


182<br />

M. Bocchia et al.<br />

phoma antigen induces protective antitumor immunity.<br />

J Immunol 1998; 160:3637-41.<br />

80. Kwak LW, Campbell MJ, Czerwinski DK, Hart S, Miller<br />

RA, Levy R. Induction of immune responses in patients<br />

with B-cell lymphoma against the surfaceimmunoglobulin<br />

idiotype expressed by their tumors.<br />

N Engl J Med 1992; 327:1209-15.<br />

81. Byars NE, Allison AC. Adjuvant formulation for use in<br />

vaccines to elicit both cell-mediated and humoral<br />

immunity. Vaccine 1987; 5:223-8.<br />

82. Hsu FJ, Caspar CB, Czerwinski D, et al. Tumor-specific<br />

idiotype vaccines in the treatment of patients with B-<br />

cell lymphoma: long-term results of a clinical trial.<br />

Blood 1997; 89:3129-35.<br />

83. Johnson AG, Tomai M, Solem L, Beck L, Ribi E. Characterization<br />

of a nontoxic monophosphoryl lipid A.<br />

Rev Infect Dis 1987; 9:S512-6.<br />

84. Maitre N, Brown JM, Demcheva M, et al. Primary T-<br />

cell and activated macrophage response associated<br />

with tumor protection using peptide/poly-N-acetyl<br />

glucosamine vaccination. Clin Cancer Res 1999;<br />

5:1173-82.<br />

85. Yewdell JW, Bennink JR. Cell biology of antigen processing<br />

and presentation to major histocompatibility<br />

complex class I molecule-restricted T-lymphocytes.<br />

Adv Immunol 1992; 52:1-123.<br />

86. Unanue ER, Cerottini JC. Antigen presentation. FASEB<br />

J 1989; 3:2496-502.<br />

87. Howard JC. Supply and transport of peptides presented<br />

by class I MHC molecules. Curr Opin Immunol<br />

1995; 7:69-76.<br />

88. Rammensee HG. Chemistry of peptides associated<br />

with MHC class I and class II molecules. Curr Opin<br />

Immunol 1995; 7:85-96.<br />

89. Braciale TJ, Braciale VL. Antigen presentation: structural<br />

themes and functional variations. Immunol<br />

Today 1991; 12:124-9.<br />

90. Germain RN. MHC-dependent antigen processing<br />

and peptide presentation: providing ligands for T lymphocyte<br />

activation. Cell 1994; 76:287-99.<br />

91. Accolla RS, Adorini L, Santoris S, Sinigaglia F, Guardiola<br />

J. MHC: orchestrating the immune response.<br />

Immunol Today 1995; 16:8-11.<br />

92. Rammensee HG, Friede T, Stevanovic S. MHC ligands<br />

and peptide motifs: first listing. Immunogenetics<br />

1995; 1:178-228.<br />

93. Falk K, Rotzschke O, Stevanovic S, Jung G, Rammensee<br />

HG. Allele-specific motifs revealed by sequencing<br />

of self-peptides eluted from MHC molecules.<br />

Nature 1991; 351:290-6.<br />

94. Stuber G, Modrow S, Hoglund P, et al. Assessment of<br />

major histocompatibility complex class I interaction<br />

with Epstein-Barr virus and human immunodeficiency<br />

virus peptides by elevation of membrane H-2 and<br />

HLA in peptide loading-deficient cells. Eur J Immunol<br />

1992; 22:2697-703.<br />

95. Sette A, Vitiello A, Reherman B, et al. The relationship<br />

between class I binding affinity and the immunogenicity<br />

of potential cytotoxic T cell epitopes. J<br />

Immunol 1994; 153:5586-92.<br />

96. Kawakami Y, Eliyahu S, Sakaguchi K, et al. Identification<br />

of the immunodominant peptides of the MART-<br />

1 human melanoma antigen recognized by the majority<br />

of HLA-A2-restricted tumor infiltrating lymphocytes.<br />

J Exp Med 1994; 180:347-52.<br />

97. Parkhurst MR, Salgaller ML, Southwood S, et al.<br />

Improved induction of melanoma-reactive CTL with<br />

peptides from the melanoma antigen gp100 modified<br />

at HLA-A*0201-binding residues. J Immunol 1996;<br />

157:2539-48.<br />

98. Valmori D, Gervois N, Rimoldi D, et al. Diversity of the<br />

fine specificity displayed by HLA-A*0201-restricted<br />

CTL specific for the immunodominant Melan-<br />

A/MART-1 antigenic peptide. J Immunol 1998;<br />

161:6956-62.<br />

99. Clay TM, Custer MC, McKee MD, et al. Changes in<br />

the fine specificity of gp100(209-217)-reactive T cells<br />

in patients following vaccination with a peptide modified<br />

at an HLA-A2.1 anchor residue. J Immunol 1999;<br />

162:1749-55.<br />

100. Khleif SN, Abrams SI, Hamilton JM, et al. A phase I<br />

vaccine trial with peptides reflecting ras oncogene<br />

mutations of solid tumors. J Immunother 1999;<br />

22:155-65.<br />

101. Manici S, Sturniolo T, Imro MA, et al. Melanoma cells<br />

present a MAGE-3 epitope to CD4(+) cytotoxic T cells<br />

in association with histocompatibility leukocyte antigen<br />

DR11. J Exp Med 1999; 189:871-6.<br />

102. Correale P, Walmsley K, Nieroda C, et al. In vitro generation<br />

of human cytotoxic T lymphocytes specific for<br />

peptides derived from prostate-specific antigen. J Natl<br />

Cancer I 1997; 89:293-300.<br />

103. Bocchia M, Wentworth PA, Southwood S, et al. Specific<br />

binding of leukemia oncogene fusion protein peptides<br />

to HLA class I molecules. Blood 1995; 85:2680-<br />

4.<br />

104. Greco G, Fruci D, Accapezzato D, et al. Two bcr-abl<br />

junction peptides bind to HLA-A3 molecules and<br />

allow specific induction of human cytotoxic T lymphocytes.<br />

Leukemia 1996; 10:693-9.<br />

105. Bocchia M, Korontsvit T, Xu Q, et al. Specific human<br />

cellular immunity to BCR-ABL oncogene-derived peptides.<br />

Blood 1996; 87:3587-92.<br />

106. Bosh GJ, Joosten AM, Kessler JH, Melief CJ, Leeksma<br />

OC. Recognition of BCR-ABL positive leukemia blasts<br />

by human CD4+ T cells elicited by primary in vitro<br />

immunization with a BCR-ABL breakpoint peptide.<br />

Blood 1996; 88:3522-7.<br />

107. Mannering SI, McKenzie JL, Fearnley DB, Hart DN.<br />

HLA-DR1-restricted bcr-abl (b3a2)-specific CD4+ T<br />

lymphocytes respond to dendritic cells pulsed with<br />

b3a2 peptide and antigen-presenting cells exposed to<br />

b3a2 containing cell lysates. Blood 1997; 90:290-7.<br />

108. Demotz S, Grey HM, Appella E, Sette A. Characterization<br />

of a naturally processed MHC class II-restricted<br />

T-cell determinant of hen egg lysozyme. Nature<br />

1989; 342:682-4.<br />

109. Storkus WJ, Zeh HD, Salter RD, Lotze MT. Identification<br />

of T-cell epitopes: rapid isolation of class-I presented<br />

peptides from viable cells by mild acid elution.<br />

J Immunother 1993; 14:94-103.<br />

110. Papadopoulos KP, Hesdorffer CS, Suciu-Foca N, Hibshoosh<br />

H, Harris PE. Wild-type p53 epitope naturally<br />

processed and presented by an HLA-B haplotype<br />

on human breast carcinoma cells. Clin Cancer Res<br />

1999; 5:2089-93.<br />

111. Skipper JC, Gulden PH, Hendrickson RC, et al. Massspectometric<br />

evaluation of HLA-A*0201-associated<br />

peptides identifies dominant naturally processed<br />

forms of CTL epitopes from MART-1 and gp100. Int<br />

J Cancer 1999; 82:669-77.<br />

112. Ostankovitch M, Buzyn A, Bonhomme D, et al.<br />

Antileukemic HLA-restricted T-cell clones generated<br />

with naturally processed peptides eluted from acute<br />

myeloblastic leukemia blasts. Blood 1998; 92:19-24.<br />

113. Papadopoulos KP, Suciu-Foca N, Hesdorffer CS, et<br />

al. Naturally processed tissue-and differentiation<br />

stage-specific autologous peptides bound by HLA<br />

class I and II molecules of chronic myeloid leukemia<br />

blasts. Blood 1997; 90:4938-46.<br />

114. Bellone M, Iezzi G, Imro MA, Protti MP. Cancer<br />

immunotherapy: synthetic and natural peptides in the<br />

balance. Immunol Today 1999; 20:457-62.<br />

115. Kawakami Y, Nishimura MI, Restifo NP, et al. T-cell<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

183<br />

recognition of human melanoma antigens. J<br />

Immunother 1993; 14:88-93.<br />

116. van der Bruggen P, Traversari C, Chomez P, et al. A<br />

gene encoding an antigen recognized by cytolytic T<br />

lymphocytes on a human melanoma. Science 1991;<br />

254:1643-7.<br />

117. Kawakami Y, Eliyahu S, Jennings C, et al. Recognition<br />

of multiple epitopes in the human melanoma antigen<br />

gp100 by tumor-infiltrating T lymphocytes associated<br />

with in vivo tumor regression. J Immunol 1995;<br />

154:3961-8.<br />

118. Kang X, Kawakami Y, el-Gamil M, et al. Identification<br />

of a tyrosinase epitope recognized by HLA-A24-<br />

restricted, tumor-infiltrating lymphocytes. J Immunol<br />

1995; 155:1343-8.<br />

119. Wang RF, Wang X, Rosenberg SA. Identification of a<br />

novel major histocompatibility complex class IIrestricted<br />

tumor antigen resulting from a chromosomal<br />

rearrangement recognized by CD4+ T cells. J Exp<br />

Med 1999; 189:1659-68.<br />

120. Pawelec G, Max H, Halder T, et al. BCR/ABL leukemia<br />

oncogene fusion peptides selectively bind to certain<br />

HLA-DR alleles and can be recognized by T cells found<br />

at low frequency in the repertoire of normal donors.<br />

Blood 1996; 88:2118-24.<br />

121. Yotnda P, Firat H, Garcia-Pons F, et al. Cytotoxic T cell<br />

response against the chimeric p210 BCR-ABL protein<br />

in patients with chronic myelogenous leukemia. J Clin<br />

Invest 1998; 101:2290-6.<br />

122. Goydos JS, Elder E, Whiteside TL, Finn OJ, Lotze MT.<br />

A phase I trial of a synthetic mucin peptide vaccine.<br />

Induction of specific immune reactivity in patients<br />

with adenocarcinoma. J Surg Res 1996; 63:298-304.<br />

123. Marchand M, van Baren N, Weynants P, et al. Tumor<br />

regressions observed in patients with metastatic<br />

melanoma treated with an antigenic peptide encoded<br />

by gene MAGE-3 and presented by HLA-A1. Int J Cancer<br />

1999; 80:219-30.<br />

124. Pinilla-Ibarz J, Cathcart K, Korontsvit T, et al. Vaccination<br />

of patients with chronic myelogenous leukemia<br />

with bcr-abl oncogene breakpoint fusion peptides<br />

generates specific immune responses. Blood 2000; 95:<br />

1781-7.<br />

125. Steller MA, Gurski KJ, Murakami M, et al. Cell-mediated<br />

immunological responses in cervical and vaginal<br />

cancer patients immunized with a lipidated epitope of<br />

human papillomavirus type 16 E7. Clin Cancer Res<br />

1998; 4:2103-9.<br />

126. Mukherji B, Chakraborty NG, Yamasaki S, et al. Induction<br />

of antigen-specific cytolytic T cells in situ in<br />

human melanoma by immunization with synthetic<br />

peptide-pulsed autologous antigen presenting cells.<br />

Proc Natl Acad Sci USA 1995; 92:8078-82.<br />

127. Shirai M, Pendleton CD, Ahlers J, et al. Helper-cytotoxic<br />

T lymphocytes (CTL) determinant linkage<br />

required for priming of anti-HIV CD8+CTL in vivo with<br />

peptide vaccine constructs. J Immunol 1994;<br />

152:549-56.<br />

128. Bachmann MF, Zinkernagel RM, Oxenius A. Immune<br />

responses in the absence of costimulation: viruses<br />

know the trick. J Immunol 1998; 161:5791-4.<br />

129. Moss B, Carroll MW, Wyatt LS, et al. Host range<br />

restricted, non-replicating vaccinia virus vectors as<br />

vaccine candidates. Adv Exp Med Biol 1996; 397:7-<br />

13.<br />

130. Hodge JW, Schlom J, Donohue SJ, et al. A recombinant<br />

vaccinia virus expressing human prostate-specific<br />

antigen (PSA): safety and immunogenicity in a nonhuman<br />

primate. Int J Cancer 1995; 63:231-7.<br />

131. Bronte V, Tsung K, Rao JB, et al. IL-2 enhances the<br />

function of recombinant poxvirus-based vaccines in<br />

the treatment of established pulmonary metastases. J<br />

Immunol 1995; 154:5282-92.<br />

132. Kantor J, Irvine K, Abrams S, Kaufman H, DiPietro J,<br />

Schlom J. Antitumor activity and immune responses<br />

induced by a recombinant carcinoembryonic antigenvaccinia<br />

virus vaccine. J Natl Cancer I 1992; 84:1084-<br />

91.<br />

133. Bronte V, Carroll MW, Goletz TJ, et al. Antigen expression<br />

by dendritic cells correlates with the therapeutic<br />

effectiveness of a model recombinant poxvirus tumor<br />

vaccine. Proc Natl Acad Sci USA 1997; 94:3183-8.<br />

134. Chamberlain RS, Carroll MW, Bronte V, et al. Costimulation<br />

enhances the active immunotherapy effect<br />

of recombinant anticancer vaccines. Cancer Res 1996;<br />

56:2832-6.<br />

135. Rao JB, Chamberlain RS, Bronte V, et al. IL-12 is an<br />

effective adjuvant to recombinant vaccinia virusbased<br />

tumor vaccines: enhancement by simultaneous<br />

B7-1 expression. J Immunol 1996; 156:3357-65.<br />

136. Carroll MW, Overwijk WW, Surman DR, Tsung K,<br />

Moss B, Restifo NP. Construction and characterization<br />

of a triple-recombinant vaccinia virus encoding<br />

B7-1, interleukin 12, and a model tumor antigen. J<br />

Natl Cancer I 1998; 90:1881-7.<br />

137. Irvine KR, Parkhurst MR, Shulman EP, et al. Recombinant<br />

virus vaccination against "self" antigens using<br />

anchor-fixed immunogens. Cancer Res 1999; 59:<br />

2536-40.<br />

138. Overwijk WW, Lee DS, Surman DR, et al. Vaccination<br />

with a recombinant vaccinia virus encoding a "self"<br />

antigen induces autoimmune vitiligo and tumor cell<br />

destruction in mice: requirement for CD4(+) T lymphocytes.<br />

Proc Natl Acad Sci USA 1999; 96:2982-7.<br />

139. Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton<br />

JM, Schlom J. Generation of human cytotoxic T<br />

cells specific for human carcinoembryonic antigen epitopes<br />

from patients immunized with recombinant vaccinia-CEA<br />

vaccine. J Natl Cancer I 1995; 87:982-90.<br />

140. Chen PW, Wang M, Bronte V, Zhai Y, Rosenberg SA,<br />

Restifo NP. Therapeutic antitumor response after<br />

immunization with a recombinant adenovirus encoding<br />

a model tumor-associated antigen. J Immunol<br />

1996; 156:224-31.<br />

141. O'Neal WK, Zhou H, Morral N, et al. Toxicological<br />

comparison of E2a-deleted and first-generation adenoviral<br />

vectors expressing alpha1-antitrypsin after systemic<br />

delivery. Hum Gene Ther 1998; 9:1587-98.<br />

142. Rosenberg SA, Zhai Y, Yang JC, et al. Immunizing<br />

patients with metastatic melanoma using recombinant<br />

adenoviruses encoding MART-1 or gp100<br />

melanoma antigens. J Natl Cancer I 1998; 90:1894-<br />

900.<br />

143. Baxby D, Paoletti E. Potential use of non-replicating<br />

vectors as recombinant vaccines. Vaccine 1992; 10: 8-<br />

9.<br />

144. Wang M, Bronte V, Chen PW, et al. Active immunotherapy<br />

of cancer with a nonreplicating recombinant<br />

fowlpox virus encoding a model tumor-associated<br />

antigen. J Immunol 1995; 154:4685-92.<br />

145. Cox WI, Tartaglia J, Paoletti E. Induction of cytotoxic<br />

T lymphocytes by recombinant canarypox (ALVAC)<br />

and attenuated vaccinia (NYVAC) viruses expressing<br />

the HIV-1 envelope glycoprotein. Virology 1993; 195:<br />

845-50.<br />

146. Perkus ME, Tartaglia J, Paoletti E. Poxvirus-based vaccine<br />

candidates for cancer, AIDS, and other infectious<br />

diseases. J Leukocyte Biol 1995; 58:1-13.<br />

147. Mahnel H, Mayr A. Experiences with immunization<br />

against orthopox viruses of humans and animals using<br />

vaccine strain MVA. Berl Munch Tierarztl 1994;<br />

107:253-6.<br />

148. Carroll MW, Overwijk WW, Chamberlain RS, Rosenberg<br />

SA, Moss B, Restifo NP. Highly attenuated mod-<br />

haematologica vol. 85(suppl. to n. 12):December 2000


184<br />

M. Bocchia et al.<br />

ified vaccinia virus Ankara (MVA) as an effective<br />

recombinant vector: a murine tumor model. Vaccine<br />

1997; 15:387-94.<br />

149. Schneider J, Gilbert SC, Blanchard TJ, et al. Enhanced<br />

immunogenicity for CD8+ T cell induction and complete<br />

protective efficacy of malaria DNA vaccination<br />

by boosting with modified vaccinia virus Ankara. Nat<br />

Med 1998; 4:397-402.<br />

150. Belyakov IM, Moss B, Strober W, Berzofsky JA.<br />

Mucosal vaccination overcomes the barrier to recombinant<br />

vaccinia immunization caused by preexisting<br />

poxvirus immunity. Proc Natl Acad Sci USA 1999;<br />

96:4512-7.<br />

151. Brossart P, Goldrath AW, Butz EA, Martin S, Bevan<br />

MJ. Virus-mediated delivery of antigenic epitopes into<br />

dendritic cells as a means to induce CTL. J Immunol<br />

1997; 158:3270-6.<br />

152. Kaplan JM, Yu Q, Piraino ST, et al. Induction of antitumor<br />

immunity with dendritic cells transduced with<br />

adenovirus vector-encoding endogenous tumor-associated<br />

antigens. J Immunol 1999; 163:699-707.<br />

153. Di Nicola M, Siena S, Bregni M, et al. Gene transfer<br />

into human dendritic antigen-presenting cells by vaccinia<br />

virus and adenovirus vectors. Cancer Gene Ther<br />

1998; 5:350-6.<br />

154. Ulmer JB, Donnelly JJ, Parker SE, et al. Heterologous<br />

protection against influenza by injection of DNA<br />

encoding a viral protein. Science 1993; 259:1745-9.<br />

155. Donnelly JJ, Ulmer JB, Shiver JW, Liu MA. DNA vaccines.<br />

Annu Rev Immunol 1997; 15:617-48.<br />

156. Wang R, Doolan DL, Le TP, et al. Induction of antigen-specific<br />

cytotoxic T lymphocytes in humans by a<br />

malaria DNA vaccine. Science 1998; 282:476-80.<br />

157. Calarota S, Bratt G, Nordlund S, et al. Cellular cytotoxic<br />

response induced by DNA vaccination in HIV-1-<br />

infected patients. Lancet 1998; 351:1320-5.<br />

158. Medina E, Guzman CA, Staendner LH, Colombo MP,<br />

Paglia P. Salmonella vaccine carrier strains: effective<br />

delivery system to trigger anti-tumor immunity by oral<br />

route. Eur J Immunol 1999; 29:693-9.<br />

159. Paglia P, Medina E, Arioli I, Guzman CA, Colombo<br />

MP. Gene transfer in dendritic cells, induced by oral<br />

DNA vaccination with Salmonella typhimurium,<br />

results in protective immunity against a murine<br />

fibrosarcoma. Blood 1998; 92:3172-6.<br />

160. Ikonomidis G, Paterson Y, Kos FJ, Portnoy DA. Delivery<br />

of a viral antigen to the class I processing and presentation<br />

pathway by Listeria monocytogenes. J Exp<br />

Med 1994; 180:2209-18.<br />

161. Anwer K, Earle KA, Shi M, et al. Synergistic effect of<br />

formulated plasmid and needle-free injection for<br />

genetic vaccines. Pharm Res 1999; 16:889-95.<br />

162. Pertmer TM, Roberts TR, Haynes JR. Influenza virus<br />

nucleoprotein-specific immunoglobulin G subclass<br />

and cytokine responses elicited by DNA vaccination<br />

are dependent on the route of vector DNA delivery. J<br />

Virol 1996; 70:6119-25.<br />

163. Feltquate DM, Heaney S, Webster RG, Robinson HL.<br />

Different T helper cell types and antibody isotypes generated<br />

by saline and gene gun DNA immunization. J<br />

Immunol 1997; 158:2278-84.<br />

164. Krieg AM, Yi AK, Matson S, et al. CpG motifs in bacterial<br />

DNA trigger direct B-cell activation. Nature<br />

1995; 374:546-9.<br />

165. Klinman DM, Yi AK, Beaucage SL, Conover J, Krieg<br />

AM. CpG motifs present in bacteria DNA rapidly<br />

induce lymphocytes to secrete interleukin 6, interleukin<br />

12, and interferon gamma. Proc Natl Acad Sci<br />

USA 1996; 93:2879-83.<br />

166. Halpern MD, Kurlander RJ, Pisetsky DS. Bacterial<br />

DNA induces murine interferon-gamma production<br />

by stimulation of interleukin-12 and tumor necrosis<br />

factor-alpha. Cell Immunol 1996; 167:72-8.<br />

167. Klinman DM, Barnhart KM, Conover J. CpG motifs as<br />

immune adjuvants. Vaccine 1999; 17:19-25.<br />

168. Sato Y, Roman M, Tighe H, et al. Immunostimulatory<br />

DNA sequences necessary for effective intradermal<br />

gene immunization. Science 1996; 273:352-4.<br />

169. Hartmann G, Weiner GJ, Krieg AM. CpG DNA: a<br />

potent signal for growth, activation, and maturation<br />

of human dendritic cells. Proc Natl Acad Sci USA<br />

1999; 96:9305-10.<br />

170. Torres CA, Iwasaki A, Barber BH, Robinson HL. Differential<br />

dependence on target site tissue for gene gun<br />

and intramuscular DNA immunizations. J Immunol<br />

1997; 158:4529-32.<br />

171. Iwasaki A, Torres CA, Ohashi PS, Robinson HL, Barber<br />

BH. The dominant role of bone marrow-derived<br />

cells in CTL induction following plasmid DNA immunization<br />

at different sites. J Immunol 1997; 159:11-4.<br />

172. Porgador A, Irvine KR, Iwasaki A, Barber BH, Restifo<br />

NP, Germain RN. Predominant role for directly transfected<br />

dendritic cells in antigen presentation to CD8+<br />

T cells after gene gun immunization. J Exp Med 1998;<br />

188:1075-82.<br />

173. Akbari O, Panjwani N, Garcia S, Tascon R, Lowrie D,<br />

Stockinger B. DNA vaccination: transfection and activation<br />

of dendritic cells as key events for immunity. J<br />

Exp Med 1999; 189:169-78.<br />

174. Irvine KR, Rao JB, Rosenberg SA, Restifo NP. Cytokine<br />

enhancement of DNA immunization leads to effective<br />

treatment of established pulmonary metastases. J<br />

Immunol 1996; 156:238-45.<br />

175. Tuting T, Gambotto A, DeLeo A, Lotze MT, Robbins<br />

PD, Storkus WJ. Induction of tumor antigen-specific<br />

immunity using plasmid DNA immunization in mice.<br />

Cancer Gene Ther 1999; 6:73-80.<br />

176. Rosato A, Zambon A, Milan G, et al. CTL response<br />

and protection against P815 tumor challenge in mice<br />

immunized with DNA expressing the tumor-specific<br />

antigen P815A. Hum Gene Ther 1997; 8:1451-8.<br />

177. Chen Y, Hu D, Eling DJ, Robbins J, Kipps TJ. DNA vaccines<br />

encoding full-length or truncated Neu induce<br />

protective immunity against Neu-expressing mammary<br />

tumors. Cancer Res 1998; 58:1965-71.<br />

178. Bowne WB, Srinivasan R, Wolchok JD, et al. Coupling<br />

and uncoupling of tumor immunity and autoimmunity.<br />

J Exp Med 1999; 190:1717-22.<br />

179. Weber LW, Bowne WB, Wolchok JD, et al. Tumor<br />

immunity and autoimmunity induced by immunization<br />

with homologous DNA. J Clin Invest 1998; 102:<br />

1258-64.<br />

180. Bronte V, Apolloni E, Ronca R, et al. Genetic vaccination<br />

with "self" tyrosinase-related protein 2 causes<br />

melanoma eradication but not vitiligo. Cancer Res<br />

2000; 60:253-8.<br />

181. Liljestrom P. Alphavirus expression systems. Curr Opin<br />

Biotechnol 1994; 5:495-500.<br />

182. Ying H, Zaks TZ, Wang RF, et al. Cancer therapy using<br />

a self-replicating RNA vaccine. Nat Med 1999; 5:823-<br />

7.<br />

183. O'Doherty U, Steinman RM, Peng M, et al. Dendritic<br />

cells freshly isolated from human blood express CD4<br />

and mature into typical immunostimulatory dendritic<br />

cells after culture in monocyte-conditioned medium.<br />

J Exp Med 1993; 178:1067-76.<br />

184. Zhou LJ, Schwarting R, Smith HM, Tedder TF. A novel<br />

cell-surface molecule expressed by human interdigitating<br />

reticulum cells, Langerhans cells, and activated<br />

lymphocytes is a new member of the IG superfamily.<br />

J Immunol 1992; 149:735-42.<br />

185. Mosialos G, Birkenbach M, Ayehunie S, et al. Circulating<br />

human dendritic cells differentially express high<br />

levels of a 55-kd actin-bundling protein. Am J Pathol<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

185<br />

1996; 148:593-600.<br />

186. Reid CD, Stackpoole A, Meager A, Tikerpae J. Interactions<br />

of tumor necrosis factor with granulocytemacrophage<br />

colony-stimulating factor and other<br />

cytokines in the regulation of dendritic cell growth in<br />

vitro from early bipotent CD34+ progenitors in<br />

human bone marrow. J Immunol 1992; 149:2681-8.<br />

187. Szabolcs P, Moore MA, Young JW. Expansion of<br />

immunostimulatory dendritic cells among the myeloid<br />

progeny of human CD34+ bone marrow precursors<br />

cultured with c-kit ligand, granulocyte-macrophage<br />

colony-stimulating factor, and TNF-alpha. J Immunol<br />

1995; 154:5851-61.<br />

188. Caux C, Vanbervliet B, Massacrier C, et al. CD34+<br />

hematopoietic progenitors from human cord blood<br />

differentiate along two independent dendritic cell<br />

pathways in response to GM-CSF+TNF alpha. J Exp<br />

Med 1996; 184:695-706.<br />

189. Siena S, Di Nicola M, Bregni M, et al. Massive ex vivo<br />

generation of functional dendritic cells from mobilized<br />

CD34+ blood progenitors for anticancer therapy.<br />

Exp Hematol 1995; 23:1463-71.<br />

190. Staquet MJ, Jacquet C, Dezutter-Dambuyant C,<br />

Schmitt D. Fibronectin upregulates in vitro generation<br />

of dendritic Langerhans cells from human cord blood<br />

CD34+ progenitors. J Invest Dermatol 1997; 109:<br />

738-43.<br />

191. Maraskovsky E, Brasel K, Teepe M, et al. Drammatic<br />

increase in the numberS of functionally mature dendritic<br />

cells in Flt3 ligand-treated mice: multiple dendritic<br />

cell subpopulations identified. J Exp Med 1996;<br />

184:1953-62.<br />

192. Lebsack ME, Maraskovsky E, Roux E, et al. Increased<br />

circulating dendritic cells in healthy human volunteers<br />

following administration of FLT3 ligand alone or in<br />

combination with GM-CSF or G-CSF. Blood 1998; 92<br />

(Suppl.1):2086a.<br />

193. Lynch DH, Andreasen A, Maraskovsky E, Whitmore J,<br />

Miller RE, Schuh JC. Flt3 ligand induces tumor regression<br />

and antitumor immune responses in vivo. Nat<br />

Med 1997; 3:625-31.<br />

194. Esche C, Subbotin VM, Maliszewski C, Lotze MT,<br />

Shurin MR. FLT3 ligand administration inhibits tumor<br />

growth in murine melanoma and lymphoma. Cancer<br />

Res 1998; 58:380-3.<br />

195. Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of<br />

melanoma patients with peptide- or tumor lysatepulsed<br />

dendritic cells. Nat Med 1998; 4:328-32.<br />

196. Albert ML, Sauter B, Bhardwaj N. Dendritic cells<br />

acquire antigen from apoptotic cells and induce class<br />

I-restricted CTLs. Nature 1998; 392:86-9.<br />

197. Zitvogel L, Regnault A, Lozier A, et al. Eradication of<br />

established murine tumors using a novel cell-free vaccine:<br />

dendritic cell-derived exosomes. Nat Med 1998;<br />

4:594-600.<br />

198. Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic<br />

cells pulsed with RNA are potent antigen-presenting<br />

cells in vitro and in vivo. J Exp Med 1996; 184:465-<br />

72.<br />

199. Reeves ME, Royal RE, Lam JS, Rosenberg SA, Hwu P.<br />

Retroviral transduction of human dendritic cells with<br />

a tumor-associated antigen gene. Cancer Res 1996;<br />

56:5672-7.<br />

200. Dietz AB, Vuk-Pavlovic S. High efficiency adenovirusmediated<br />

gene transfer to human dendritic cells.<br />

Blood 1998; 91:392-8.<br />

201. Di Nicola M, Carlo-Stella C, Milanesi M, et al. Largescale<br />

feasibility of gene transduction into human<br />

CD34+ cell derived-dendritic cells by polication/adenoviral<br />

complex. Br J Haematol 2000; (In Press).<br />

202. Zhong L, Granelli-Piperno A, Choi Y, Steinman RM.<br />

Recombinant adenovirus is an efficient and non-perturbing<br />

genetic vector for human dendritic cells. Eur J<br />

Immunol 1999; 29:964-72.<br />

203. Moss B. Vaccinia virus: a tool for research and vaccine<br />

development. Science 1991; 252:1662-7.<br />

204. Choudhury BA, Liang JC, Thomas EK, et al. Dendritic<br />

cells derived in vitro from acute myelogenous<br />

leukemia cells stimulate autologous, antileukemic T-<br />

cell responses. Blood 1999; 93:780-6.<br />

205. Cignetti A, Bryant E, Allione B, Vitale A, Foa R, Cheever<br />

MA. CD34(+) acute myeloid and lymphoid<br />

leukemic blasts can be induced to differentiate into<br />

dendritic cells. Blood 1999; 94:2048-55.<br />

206. Choudhury A, Gajewski JL, Liang JC, et al. Use of<br />

leukemic dendritic cells for the generation of<br />

antileukemic cellular cytotoxicity against Philadelphia<br />

chromosome-positive chronic myelogenous leukemia.<br />

Blood 1997; 89:1133-42.<br />

207. Carlo-Stella C, Regazzi E, Garau D, et al. Ex vivo generation<br />

of bcr/abl positive dendritic cells from CD34+<br />

chronic myeloid leukemia cells. <strong>Haematologica</strong> 1998;<br />

83 (Meeting Supplement):78-9.<br />

208. Balch CM, Reintgen DS, Kirkwood JM, et al.Cutaneous<br />

melanoma. Cancer Principles & Practice of<br />

Oncology, 5th Edition, De Vita VT, Hellman S, Rosenberg<br />

SA (Eds.), Lippincot-Raven, 1997, p. 1947-94.<br />

209. Linehan DC, Goedegebuure PS, Eberlein TJ. Vaccine<br />

therapy for cancer. Ann Surg Oncol 1996; 3:219-28.<br />

210. Arienti F, Sule-Suso J, Belli F, et al. Limited antitumor<br />

T cell response in melanoma patients vaccinated with<br />

interleukin-2 gene-transduced allogeneic melanoma<br />

cells. Hum Gene Ther 1996; 7:1955-63.<br />

211. Moller P, Sun Y, Dorbic T, et al. Vaccination with IL-<br />

7 gene-modified autologous melanoma cells can<br />

enhance the anti-melanoma lytic activity in peripheral<br />

blood of patients with a good clinical performance<br />

status: a clinical phase I study. Br J Cancer 1998;<br />

77:1907-16.<br />

212. Jager E, Ringhoffer M, Dienes HP, et al. Granulocytemacrophage-colony-stimulating<br />

factor enhances<br />

immune responses to melanoma-associated peptides<br />

in vivo. Int J Cancer 1996; 67:54-62.<br />

213. Rosenberg SA, Yang JC, Schwartzentruber DJ, et al.<br />

Immunologic and therapeutic evaluation of a synthetic<br />

peptide vaccine for the treatment of patients<br />

with metastatic melanoma. Nat Med 1998; 4:321-7.<br />

214. Chakraborty NG, Sporn JR, Tortora AF, et al. Immunization<br />

with a tumor-cell-lysate-loaded autologousantigen-presenting-cell-based<br />

vaccine in melanoma.<br />

Cancer Immunol Immun 1998; 47:58-64.<br />

215. Lotze MT, Hellerstedt B, Stolinski L, et al. The role of<br />

interleukin-2, interleukin-12, and dendritic cells in<br />

cancer therapy. Cancer J Sci Am 1997; Suppl 1:S109-<br />

14.<br />

216. Blieberg H, Rougier P, Wilke HJ. Management of colorectal<br />

cancer. Martin Dunitz Publisher, London, 1998.<br />

217. Vermorken JB, Claessen AM, van Tinteren H, et al.<br />

Active specific immunotherapy for stage II and stage<br />

III human colon cancer: a randomised trial. Lancet<br />

1999; 353:345-50.<br />

218. Foon KA, John WJ, Chakraborty M, et al. Clinical and<br />

immune responses in resected colon cancer patients<br />

treated with anti-idiotype monoclonal antibody vaccine<br />

that mimics the carcinoembryonic antigen. J Clin<br />

Oncol 1999; 17:2889-95.<br />

219. Tjoa B, Boynton A, Kenny G, Ragde H, Misrock SL,<br />

Murphy G. Presentation of prostate tumor antigens by<br />

dendritic cells stimulates T-cell proliferation and cytotoxicity.<br />

Prostate 1996; 28:65-9.<br />

220. Correale P, Walmsley K, Nieroda C, et al. In vitro generation<br />

of human cytotoxic T lymphocytes specific for<br />

peptides derived from prostate-specific antigen. J Natl<br />

Cancer I 1997; 89:293-300.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


186<br />

M. Bocchia et al.<br />

221. Valone F, Small E, Peshwa MV, et al. Phase I trial of<br />

dendritic cell-based immunotherapy with APC8015<br />

for hormone-refractory prostate cancer (HRPC). Proc<br />

Am Soc Cancer Res 1998; 39:173(Abstr).<br />

222. Salgallar M, Lodge PA, Tjoa BA, et al. Monitoring of<br />

prostate-specific membrane antigen (PSMA)-specific<br />

immune response and prostate markers in a phase II<br />

clinical trial with patients infused with dendritic cells<br />

pulsed with PSMA-derived peptides. Proc Am Soc<br />

Cancer Res 1998; 39:(Abstr).<br />

223. Murphy G, Tjoa B, Ragde H, Kenny G, Boynton A.<br />

Phase I clinical trial: T-cell therapy for prostate cancer<br />

using autologous dendritic cells pulsed with HLA-<br />

A0201-specific peptides from prostate-specific membrane<br />

antigen. Prostate 1996 ;29:371-80.<br />

224. Rosenberg SA, Lotze MT, Muul LM, et al. A progress<br />

report on the treatment of 157 patients with advanced<br />

cancer using lymphokine-activated killer cells and<br />

interleukin-2 or high-dose interleukin-2 alone. N Engl<br />

J Med 1987; 316:889-97.<br />

225. Vogelzang NJ, Lipton A, Figlin RA. Subcutaneous<br />

interleukin-2 plus interferon alfa-2a in metastatic renal<br />

cancer: an outpatient multicenter trial. J Clin Oncol<br />

1993; 11:1809-16.<br />

226. Figlin RA, Pierce WC, Kaboo R, et al. Treatment of<br />

metastatic renal cell carcinoma with nephrectomy,<br />

interleukin-2 and cytokine-primed or CD8(+) selected<br />

tumor infiltrating lymphocytes from primary tumor. J<br />

Urol 1997; 158:740-5.<br />

227. Holtl L, Rieser C, Papesh C, et al. Cellular and<br />

humoral immune responses in patients with metastatic<br />

renal cell carcinoma after vaccination with antigen<br />

pulsed dendritic cells. J Urol 1999; 161:777-82.<br />

228. Kugler A, Stuhler G, Walden P, et al. Regression of<br />

human metastatic renal cell carcinoma after vaccination<br />

with tumor cell-dendritic cell hybrids. Nat Med<br />

2000; 6:332-6.<br />

229. Dianzani U, Pileri A, Boccadoro M, et al. Activated<br />

idiotype-reactive cells in suppressor/cytotoxic subpopulations<br />

of monoclonal gammopathies: correlation<br />

with diagnosis and disease status. Blood 1988;<br />

72:1064-8.<br />

230. Osterborg A, Masucci M, Bergenbrant S, Holm G,<br />

Lefvert AK, Mellstedt H. Generation of T cell clones<br />

binding F(ab')2 fragments of the idiotypic immunoglobulin<br />

in patients with monoclonal gammopathy.<br />

Cancer Immunol Immun 1991; 34:157-62.<br />

231. Yi Q, Osterborg A, Bergenbrant S, Mellstedt H, Holm<br />

G, Lefvert AK. Idiotype-reactive T-cell subsets and<br />

tumor load in monoclonal gammopathies. Blood<br />

1995; 86:3043-9.<br />

232. Yi Q, Eriksson I, He W, Holm G, Mellstedt H, Osterborg<br />

A. Idiotype-specific T lymphocytes in monoclonal<br />

gammopathies: evidence for the presence of CD4+<br />

and CD8+ subsets. Br J Haematol 1997; 96:338-45.<br />

233. Wen YJ, Lim SH. T cells recognize the VH complementarity-determining<br />

region 3 of the idiotypic protein<br />

of B cell non-Hodgkin’s lymphoma. Eur J<br />

Immunol 1997; 27:1043-7.<br />

234. Wen YJ, Ling M, Lim SH. Immunogenicity and crossreactivity<br />

with idiotypic IgA of VH CDR3 peptide in<br />

multiple myeloma. Br J Haematol 1998; 100:464-8.<br />

235. Fagerberg J, Yi Q, Gigliotti D, et al. T-cell-epitope mapping<br />

of the idiotypic monoclonal IgG heavy and light<br />

chains in multiple myeloma. Int J Cancer 1999; 80:<br />

671-80.<br />

236. Bianchi A, Massaia M. Idiotypic vaccination in B-cell<br />

malignancies. Mol Med Today 1997; 3:435-41.<br />

237. Nelson EL, Li X, Hsu FJ, et al. Tumor-specific, cytotoxic<br />

T-lymphocyte response after idiotype vaccination for<br />

B-cell, non-Hodgkin's lymphoma. Blood 1996; 88:<br />

580-9.<br />

238. Bendandi M, Gocke CD, Kobrin CB, et al. Complete<br />

molecular remissions induced by patient-specific vaccination<br />

plus granulocyte-monocyte colony-stimulating<br />

factor against lymphoma. Nat Med 1999; 5:1171-<br />

7.<br />

239. Corradini P, Voena C, Astolfi M, et al. High-dose<br />

sequential chemoradiotherapy in multiple myeloma:<br />

residual tumor cells are detectable in bone marrow<br />

and peripheral blood cell harvests and after autografting.<br />

Blood 1995; 85:1596-602.<br />

240. Shtil A, Turner JG, Durfee J, Dalton WS, Yu H.<br />

Cytokine-based tumor cell vaccine is equally effective<br />

against parental and isogenic multidrug-resistant<br />

myeloma cells: the role of cytotoxic T lymphocytes.<br />

Blood 1999; 93:1831-7.<br />

241. Osterborg A, Yi Q, Henriksson L, et al. Idiotype immunization<br />

combined with granulocyte-macrophage<br />

colony-stimulating factor in myeloma patients<br />

induced type I, major histocompatibility complexrestricted,<br />

CD8- and CD4-specific T-cell responses.<br />

Blood 1998; 91:2459-66.<br />

242. Massaia M, Borrione P, Battaglio S, et al. Idiotype<br />

vaccination in human myeloma: generation of tumorspecific<br />

immune responses after high-dose chemotherapy.<br />

Blood 1999; 94:673-83.<br />

243. Dhodapkar MV, Steinman RM, Sapp M, et al. Rapid<br />

generation of broad T-cell immunity in humans after<br />

a single injection of mature dendritic cells. J Clin Invest<br />

1999; 104:173-80.<br />

244. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of<br />

patients with B-cell lymphoma using autologous antigen-pulsed<br />

dendritic cells. Nat Med 1996; 2:52-8.<br />

245. Reichardt VL, Okada CY, Liso A, et al. Idiotype vaccination<br />

using dendritic cells after autologous peripheral<br />

blood stem cell transplantation for multiple<br />

myeloma-a feasibility study. Blood 1999; 93:2411-9.<br />

246. Wen YJ, Ling M, Bailey-Wood R, Lim SH. Idiotypic<br />

protein-pulsed adherent peripheral blood mononuclear<br />

cell-derived dendritic cells prime the immune system<br />

in multiple myeloma. Clin Cancer Res 1998; 4:<br />

957-62.<br />

247. Cull G, Durrant L, Stainer C, Haynes A, Russell N.<br />

Generation of anti-idiotype immune responses following<br />

vaccination with idiotype-protein pulsed dendritic<br />

cells in myeloma. Br J Haematol 1999; 107:648-<br />

55.<br />

248. Lim SH, Bailey-Wood R. Idiotypic protein-pulsed dendritic<br />

cell vaccination in multiple myeloma. Int J Cancer<br />

1999; 83:215-22.<br />

249. Titzer S, Christensen O, Manzke O, et al. Vaccination<br />

of multiple myeloma patients with idiotype-pulsed<br />

dendritic cells: immunological and clinical aspects. Br<br />

J Haematol 2000; 108:805-16.<br />

250. Gong J, Chen D, Kashiwaba M, Kufe D. Induction of<br />

antitumor activity by immunization with fusions of<br />

dendritic and carcinoma cells. Nat Med 1997; 3:558-<br />

61.<br />

251. Schultze JL, Cardoso AA, Freeman GJ, et al. Follicular<br />

lymphomas can be induced to present alloantigen efficiently:<br />

a conceptual model to improve their tumor<br />

immunogenicity. Proc Natl Acad Sci USA 1995;<br />

92:8200-4.<br />

252. Schultze JL, Michalak S, Seamon MJ, et al. CD40-activated<br />

human B cells: an alternative source of highly<br />

efficient antigen presenting cells to generate autologous<br />

antigen-specific T cells for adoptive immunotherapy.<br />

J Clin Invest 1997; 100:2757-65.<br />

253. Teoh G, Chen L, Urashima M, et al. Adenovirus vector-based<br />

purging of multiple myeloma cells. Blood<br />

1998; 92:4591-601.<br />

254. Kwak LW, Taub DD, Duffey PL, et al. Transfer of<br />

myeloma idiotype-specific immunity from an actively<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Antitumor vaccination<br />

187<br />

immunised marrow donor. Lancet 1995; 345:1016-<br />

20.<br />

255. Takahashi T, Makiguchi Y, Hinoda Y, et al. Expression<br />

of MUC1 on myeloma cells and induction of HLAunrestricted<br />

CTL against MUC1 from a multiple<br />

myeloma patient. J Immunol 1994 ;153:2102-9.<br />

256. Treon SP, Mollick JA, Urashima M, et al. Muc-1 core<br />

protein is expressed on multiple myeloma cells and is<br />

induced by dexamethasone. Blood 1999; 93:1287-<br />

98.<br />

257. Van Baren N, Brasseur F, Godelaine D, et al. Genes<br />

encoding tumor-specific antigens are expressed in<br />

human myeloma cells. Blood 1999; 94:1156-64.<br />

258. Rao PH, Cigudosa JC, Ning Y, et al. Multicolor spectral<br />

karyotyping identifies new recurring breakpoints<br />

and translocations in multiple myeloma. Blood 1998;<br />

92:1743-8.<br />

259. Chesi M, Nardini E, Brents LA, et al. Frequent translocation<br />

t(4;14)(p16.3;q32.3) in multiple myeloma is<br />

associated with increased expression and activating<br />

mutations of fibroblast growth factor receptor 3. Nat<br />

Genet 1997; 16:260-4.<br />

260. Stevenson FK, Zhu D, King CA, Ashworth LJ, Kumar S,<br />

Hawkins RE. Idiotypic DNA vaccines against B-cell<br />

lymphoma. Immunol Rev 1995; 145:211-28.<br />

261. Tao MH, Levy R. Idiotype/granulocyte-macrophage<br />

colony-stimulating factor fusion protein as a vaccine<br />

for B-cell lymphoma. Nature 1993; 362:755-8.<br />

262. Syrengelas AD, Chen TT, Levy R. DNA immunization<br />

induces protective immunity against B-cell lymphoma.<br />

Nat Med 1996; 2:1038-41.<br />

263. King CA, Spellerberg MB, Zhu D, et al. DNA vaccines<br />

with single-chain Fv fused to fragment C of tetanus<br />

toxin induce protective immunity against lymphoma<br />

and myeloma. Nat Med 1998; 4:1281-6.<br />

264. Biragyn A, Tani K, Grimm MC, Weeks S, Kwak LW.<br />

Genetic fusion of chemokines to a self tumor antigen<br />

induces protective, T-cell dependent antitumor immunity.<br />

Nat Biotechnol 1999; 17:253-8.<br />

265. Kolb HJ, Mittermuller J, Clemm CH, et al. Donor<br />

leukocyte transfusions for treatment of recurrent<br />

chronic myelogenous leukemia in marrow transplant<br />

patients. Blood 1990; 76:2462-5.<br />

266. Mackinnon S, Papadopoulos EB, Carabasi MH, et al.<br />

Adoptive immunotherapy evaluating escalating doses<br />

of donor leukocytes for relapse or chronic myeloid<br />

leukemia after bone marrow transplantation: separation<br />

of graft-versus-leukemia responses from graft-versus-host<br />

disease. Blood 1995; 86:1261-8.<br />

267. Slavin S, Naparstek E, Nagler A, et al. Allogeneic cell<br />

therapy with donor peripheral blood cells and recombinant<br />

human interleukin-2 to treat leukemia relapse<br />

after allogeneic bone marrow transplantation. Blood<br />

1996; 87:2195-204.<br />

268. Nieda M, Nicol A, Kicuchi A, et al. Dendritic cells stimulate<br />

the expansion of bcr-abl specific CD8+ T cells<br />

with cytotoxic activity against leukemic cells from<br />

patients with chronic myeloid leukemia. Blood 1998;<br />

91:977-83.<br />

269. Schadendorf D. Cancer Vaccines. Once more unto the<br />

breach. The Economist 1994; 12:78-9.<br />

270. Molldrem JJ, Lee PP, Wang C, Champlin RE, Davis<br />

MM. A PR1-human leukocyte antigen-A2 tetramer<br />

can be used to isolate low-frequency cytotoxic T lymphocytes<br />

from healthy donors that selectively lyse<br />

chronic myelogenous leukemia. Cancer Res 1999; 59:<br />

2675-81.<br />

271. Pass HA, Schwarz SL, Wunderlich JR, Rosenberg SA.<br />

Immunization of patients with melanoma peptide vaccines:<br />

immunologic assessment using the ELISPOT<br />

assay. Cancer J Sci Am 1998; 4:316-23.<br />

272. Wolfel T, Hauer M, Schneider J, et al. A p16INK4ainsensitive<br />

CDK4 mutant targeted by cytolytic T lymphocytes<br />

in a human melanoma. Science 1995;<br />

269:1281-4.<br />

273. Vonderheide RH, Hahn WC, Schultze JL, Nadler LM.<br />

The telomerase catalytic subunit is a widely expressed<br />

tumor-associated antigen recognized by cytotoxic T<br />

lymphocytes. Immunity 1999; 10:673-9.<br />

274. Jonsen AR, Durfy SJ, Burke W, Motulsky AG. The<br />

advent of the 'unpatients'. Nat Med 1996; 2: 622-4.<br />

275. Nanni P, Forni G, Lollini PL. Cytokine gene therapy:<br />

hopes and pitfalls. Ann Oncol 1999; 10:261-6.<br />

276. Pardoll DM. Inducing autoimmune disease to treat<br />

cancer. Proc Natl Acad Sci USA 1999; 96: 5340-2.<br />

277. Prevention of cancer in the next millenium: Report of<br />

the Chemoprevention Working Group to the American<br />

Association for Cancer Research. Cancer Res<br />

1999; 59: 4743-58.<br />

278. Ehrlich P. Ueber den jetzigen Stand der Karzinomforschung.<br />

Ned Tijdschr Geneeskd 1909; 5:273-90.<br />

haematologica vol. 85(suppl. to n. 12):December 2000


Index of authors<br />

Aglietta Massimo, 19, 92<br />

Arcese William, 69<br />

Aversa Franco, 69<br />

Bandini Giuseppe, 69<br />

Bertolini Francesco, 49, 92<br />

Bocchia Monica, 156<br />

Bordignon Claudio, 117<br />

Bronte Vincenzo, 156<br />

Caligaris Cappio Federico, 32<br />

Carlo-Stella Carmelo, 1, 92, 117<br />

Cavo Michele, 32<br />

Cazzola Mario, 1<br />

Colombo Mario P., 117, 156<br />

De Fabritiis Paolo, 1<br />

De Vincentiis Armando, 1, 19, 32, 49, 69, 92, 117, 156<br />

Di Nicola Massimo, 156<br />

Falda Michele, 69<br />

Forni Guido, 156<br />

Gianni Alessandro Massimo 1<br />

Lanata Luigi , 19, 32, 49, 69, 92, 117, 156<br />

Lanza Francesco, 1, 19<br />

Lauria Francesco, 1<br />

Lemoli Roberto M., 1, 19, 32, 49, 69, 92, 117, 156<br />

Locatelli Franco , 69, 117<br />

Maccario Rita, 49<br />

Majolino Ignazio, 32, 49, 69<br />

Massaia Massimo, 156<br />

Menichella Giacomo, 19<br />

Olivieri Attilio, 92, 117<br />

Ponchio Luisa, 49<br />

Rondelli Damiano, 49, 117, 156<br />

Siena Salvatore, 92<br />

Tabilio Antonio, 49<br />

Tafuri Agostino, 19<br />

Tarella Corrado, 1, 32<br />

Tura Sante, 1, 19, 32, 49, 69, 92, 117, 156<br />

Zanon Paola, 1, 19, 32, 49, 69, 92, 117, 156<br />

Direttore responsabile: Prof. Edoardo Ascari<br />

Autorizzazione del Tribunale di Pavia n. 63 del 5 marzo 1955<br />

Composizione: = Medit – via gen. C.A. Dalla Chiesa, 22 – Voghera, Italy<br />

Stampa: Tipografia PI-ME – via Vigentina 136 – Pavia, Italy<br />

<br />

Printed in December 2000


In collaborazione con Dompé Biotec s.p.a.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!