06.02.2013 Views

LEUKEMIA AND LYMPHOMA East and West are Together

LEUKEMIA AND LYMPHOMA East and West are Together

LEUKEMIA AND LYMPHOMA East and West are Together

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>LEUKEMIA</strong> <strong>AND</strong> <strong>LYMPHOMA</strong><br />

<strong>East</strong> <strong>and</strong> <strong>West</strong> <strong>are</strong> <strong>Together</strong><br />

September 17–21, 2011<br />

Dubrovnik, Croatia<br />

Organizers:<br />

The University of Texas, MD Anderson Cancer Center<br />

<strong>and</strong><br />

Clinical Hospital Center Zagreb, School of Medicine<br />

University of Zagreb<br />

SUPLEMENT<br />

LIJE^ VJESN 133:1–130 4 ZAGREB, 2011.


Utemeljen 1877. Founded 1877<br />

SAVJET / ADVISORY BOARD<br />

PREDSJEDNIK / PRESIDENT<br />

Mladen Belicza<br />

TAJNIK / SECRETARY<br />

Miroslav Hromadko<br />

^LANOVI / MEMBERS<br />

Ivan Bakran (Zagreb) – Tomislav Dasovi} (Zagreb) – Hedviga Hricak (New York) – Miroslav Hromadko (Zagreb) – Mirko Jung<br />

(Zürich) – Ivica Kostovi} (Zagreb) – Mladen Kri` (Rijeka) – Ante Padjen (Montreal) – Marko Pe}ina (Zagreb) – Dinko Podrug<br />

(New York) – Miljenko Pukani} (Sydney) – Smiljan Pulji} (Upper Saddle River, New Jersey) – Petar Rei} (Split) – Berislav Tomac<br />

(Hagen) – Marko Turina (Zürich) – Ljiljana Zergollern-^upak (Zagreb) – @ivojin @agar (Zagreb)<br />

UREDNI^KI ODBOR / EDITORIAL BOARD<br />

Branimir Ani} (Zagreb) – Darko Anti~evi} (Zagreb) – Alen Babacanli (Zagreb) – @elimir Bradamante (Zagreb) – Nada ^ike{<br />

(Zagreb) – Vladimir Duga~ki (Zagreb) – @eljko Feren~i} (Zagreb) – Rudolf Gre gurek (Zagreb) – Smilja Kaleni} (Zagreb) – Ni kola<br />

M<strong>and</strong>i} (Osijek) – August Miji} (Zagreb) – Davor Mili~i} (Zagreb) – Stojan Poli} (Split) – @eljko Reiner (Zagreb) – Darko Richter<br />

(Zagreb) – Zvonko Rumboldt (Split) – Juraj Sep~i} (Rijeka) – Zdenko Sonicki (Zagreb) – Davor [timac (Rijeka)<br />

GLAVNI I ODGOVORNI UREDNIK / EDITOR-IN-CHIEF<br />

Branimir Ani}<br />

TAJNIK UREDNI^KOG ODBORA / SECRETARY OF THE EDITORIAL BOARD<br />

@eljko Feren~i}<br />

Osniva~ i izdava~ / Founder <strong>and</strong> Publisher<br />

HRVATSKI LIJE^NI^KI ZBOR<br />

Glavni i odgovorni urednik / Editor-in-Chief<br />

BRANIMIR ANI]<br />

Tajnica redakcije / Secretary of the Editorial Offi ce<br />

DRA@ENKA KONTEK<br />

Rje{enje naslovne stranice / Front Page Design<br />

BRANKO VUJANOVI]<br />

Tehni~ki urednik / Technical Editor<br />

JOSIP VLAHOVI]<br />

Slog / Typesetting<br />

»GREDICE« – Zagreb, Horva}anska 67<br />

Tisak / Printed by<br />

PRINTERA GRUPA – Zagreb<br />

Web stranica / Web page<br />

ALEN BABACANLI<br />

Zagreb 2011.<br />

Naklada 200 primjeraka<br />

Rukopis i svi ~lanci {alju se Uredni{tvu Lije~ni~kog vjesnika, Zagreb, [ubi}eva ul. 9, tel. (01) 46-93-300. ^lanarina,<br />

pretplata i sve nov~ane po{iljke {alju se Hrvatskomu lije~ni~kom zboru, Zagreb, [ubi}eva ulica 9, odnosno na `irora~un<br />

br. 2360000-1101214818 ili devizni ra~un br. 2100060384, Zagreba~ka banka d.d., Savska c. 60, Zagreb, Swift:<br />

Zaba HR 2X, IBAN HR7423600001101214818, OIB 60192951611. ^lanarina Hrvatskoga lije~ni~kog zbora iznosi<br />

200 kuna; za lije~nike pripravnike i obiteljska ~lanarina iznosi 100 kuna. Pretplata za Lije~ni~ki vjesnik je 270 kuna.<br />

Radi redovitog primanja potrebno je odmah svaku promjenu adrese i boravka javiti Uredni{tvu. ^asopis se {alje<br />

svim ~lanovima besplatno. Svaki ~lan Hrvatskoga lije~ni~kog zbora ima pravo besplatno objaviti ~lanak u Lije~ni~kom<br />

vjesniku; autori koji nisu ~lanovi Zbora moraju platiti naknadu u iznosu od 150 kuna.<br />

Lije~ni~ki vjesnik citiraju: MEDLINE/Index Medicus i EMBASE/Excerpta Medica.


MEETING CHAIRMAN:<br />

Emil Freireich (USA)<br />

Hagop Kantarjian (USA)<br />

Boris Labar (Croatia)<br />

Ranka Serventi Seiwerth (Croatia)<br />

Srdjan Verstovsek (USA)<br />

SCIENTIFIC SECRETARY:<br />

S<strong>and</strong>ra Basic Kinda (Croatia)<br />

Mirta Mikulic (Croatia)<br />

MEETING VENUE:<br />

Hotel Palace, Dubrovnik<br />

September 17–21, 2011<br />

Dear Colleagues,<br />

It is our pleasure to welcome you for 2nd Leukemia <strong>and</strong> Lymphoma Meeting to Dubrovnik,<br />

Croatia, September 17–21, 2011. The organizers <strong>are</strong> MD Anderson Cancer<br />

Center, Houston, TX, USA <strong>and</strong> University Hospital Center, Zagreb, School of Medicine<br />

University of Zagreb, Croatia.<br />

During this 5-day meeting, we will explore the most relevant <strong>and</strong> recent evidencebased<br />

data on treatment of hematological neoplasms. We will discuss the same topics as<br />

in the 1st Leukemia <strong>and</strong> Lymphoma Meeting; however, in order to encourage audience<br />

participation <strong>and</strong> interaction, we added new sessions:<br />

Meet the professor <strong>and</strong> Lunch with Experts which will break into informal discussion<br />

groups.<br />

End of the Day debate to discuss controversies in the treatment of hematological<br />

malignancies.<br />

Many distinguished invited speakers from Europe <strong>and</strong> USA will participate again.<br />

Welcome to Dubrovnik!<br />

ORGANIZERS


Golden sponsor<br />

Sponsors<br />

Contributors


Contents<br />

Scientific program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii<br />

SA01 State of the art I.: Acute leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3<br />

OP01-73 Symposia/Special Lectures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17<br />

OP01-04 Presidential Symposium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17<br />

OP05-10 Symposium: AML – Risk adapted therapy I. . . . . . . . . . . . . . . . . . . . . . . . . . 20<br />

OP11-13 Symposium: AML – Risk adapted therapy II. . . . . . . . . . . . . . . . . . . . . . . . . . 23<br />

OP14-19 Symposium: MDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23<br />

OP20-25 Symposium: Myeloproliferative Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . 31<br />

OP26-31 Symposium: CML . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35<br />

OP32-35 Symposium: Multiple Myeloma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46<br />

OP36-43 Symposium: ALL . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48<br />

OP44-48 Symposium: CLL . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53<br />

OP49-57 Symposium: Stem Cell Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . 54<br />

OP58 Special Lecture I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60<br />

OP59-66 Symposium: Non- Hodgkin Lymphoma . . . . . . . . . . . . . . . . . . . . . . . . . . . 61<br />

OP67-73 Symposium: Hodgkin Lymphoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63<br />

MP01-10 Meet the Professor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71<br />

SA02 State of the art II.: Diagnostic <strong>and</strong> Treatment Approach<br />

for Acute Myeloblastic Leukemia - Report from Central <strong>and</strong> <strong>East</strong>ern European<br />

Leukemia Group (CELG) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81<br />

PP01-64 Poster Sessions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87<br />

PP01-07 Acute leukemia I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87<br />

PP08-13 Ph + Chronic Myeloid Leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90<br />

PP14-20 Stem Cell Transplantation I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93<br />

PP21-27 Lymphoma I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97<br />

PP28-34 Myeloproliferative neoplasm <strong>and</strong> miscelaneus . . . . . . . . . . . . . . . . . . . . . . . . 101<br />

PP35-40 Multiple myeloma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104


PP41-47 Acute Leukemia II. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107<br />

PP48-54 Lymphoma II. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110<br />

PP55-60 Stem Cell Transplantation II. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113<br />

PP61-64 Miscelaneus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116<br />

SP01-13 Satellite Symposia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119<br />

Author Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127


Scientific Program<br />

Friday, 16. September 2011<br />

<strong>LEUKEMIA</strong> <strong>AND</strong> <strong>LYMPHOMA</strong><br />

<strong>East</strong> <strong>and</strong> <strong>West</strong> <strong>are</strong> <strong>Together</strong><br />

Dubrovnik, Croatia, September 17–21, 2011<br />

Organizers:<br />

The University of Texas, MD Anderson Cancer Center<br />

<strong>and</strong><br />

Clinical Hospital Center Zagreb, School of Medicine<br />

University of Zagreb<br />

Meeting Venue: Hotel Palace, Dubrovnik<br />

15.00 – 19.00 Meeting: Central <strong>and</strong> <strong>East</strong>ern European Leukemia Group<br />

15.00 – 15.45 Registration of the CELG Group<br />

15.45 – 16.30 Acute Leukemia<br />

16.30 – 17.00 Coffee break<br />

17.00 – 17.45 CML<br />

17.45 – 18.30 Myelofibrosis<br />

18.30 – 19.00 New proposals for CELG Activity<br />

Saturday, 17. September 2011.<br />

13.30 – 15.30 Lunch Satellite Symposium: Therapy of fungal infections in immunocompromised patients<br />

Chair: Boris Labar (Croatia)<br />

14.00 – 14.05 Boris Labar (Croatia)<br />

Welcome <strong>and</strong> introduction<br />

14.05 – 14.30 Lubos Drgona (Slovakia)<br />

Hematologic patients dying of IFI? Trends in IFI epidemiology (SP01)<br />

14.30 – 14.55 Michael Girschikofsky (Austria)<br />

Invasive aspergillosis in hematological patients – from empirical to early pre-emptive therapy (SP02)<br />

14.55 – 15.20 Ben De Pauw (The Netherl<strong>and</strong>s)<br />

Management of Invasive aspergillosis:guidelines at the bedside (SP03)<br />

15.20 – 15.30 Boris Labar (Croatia)<br />

Panel discussion <strong>and</strong> meeting close<br />

16.15 – 17.45 Satellite Symposium: Iron chelation therapy for MDS<br />

Chair: Ranka Serventi Seiwerth (Croatia)<br />

16.15 – 16.40 Heather A. Leitch (Canada)<br />

Role of Iron Chelation in Myelodysplastic Syndrome (SP04)<br />

16.40 – 17.05 Aristoteles Giagounidis (Germany)<br />

Which MDS patients <strong>are</strong> eligible for iron chelation therapy? (SP05)<br />

17.05 – 17.30 Theo De Witte (The Netherl<strong>and</strong>s)<br />

Deferasirox in iron-overloaded patients with transfusion-dependent MDS (SP06)<br />

17.30 – 17.45 Panel discussion <strong>and</strong> closing of the Symposium<br />

vii


Scientific Program Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

18.00 – 20.00 OPENING CEREMONY <strong>and</strong> PRESIDENTIAL SYMPOSIUM<br />

18.00 – 18.20 Opening Ceremony<br />

18.20 – 20.00 Presidential Symposium<br />

Chair: Hagop Kantarjian (USA), Boris Labar (Croatia), Srdjan Verstovsek (USA),<br />

Ranka Serventi-Seiwerth (Croatia)<br />

18.20 – 18.45 Emil Freireich (USA)<br />

White blood cell transfusion <strong>and</strong> leukemia therapy (OP01)<br />

18.45 – 19.10 Roel Willemze (The Netherl<strong>and</strong>s)<br />

Presence <strong>and</strong> future of treatment of acute myeloid leukemia in patients under the age of 60 (OP02)<br />

19.10 – 19.35 Michael Keating (USA)<br />

CLL: how close is the cure? (OP03)<br />

19.35 – 20.00 Hans-Jochem Kolb (Germany)<br />

From donor »Buffy Coat« to vaccination (OP04)<br />

20.30 – 23.00 CONCERT <strong>AND</strong> WELCOME COCKTAIL<br />

Sunday 18.09. 2011.<br />

08.30 – 12.30 SYMPOSIUM AML – Risk adapted therapy<br />

Chair: Hagop Kantarjian (USA) Sergio Amadori (Italy)<br />

08.30 – 08.45 Torsten Haferlach (Germany)<br />

AML in adults – what we need for diagnosis (OP05)<br />

08.45 – 09.00 Joop Jansen (The Netherl<strong>and</strong>s)<br />

Mutation of epigenetic regulators in AML <strong>and</strong> MDS (OP06)<br />

09.00 – 09.15 Alan Burnett (United Kingdom)<br />

Therapy for younger adults with AML (OP07)<br />

09.15 – 09.30 Sergio Amadori (Italy)<br />

Therapy for elderly AML (OP08)<br />

09.30 – 09.45 Miguel Angel Sanz (Spain)<br />

Do we need chemotherapy for APL treatment? (OP09)<br />

09.45 – 10.00 Farhad Rav<strong>and</strong>i (USA)<br />

FLT3 inhibitors – too much expectations?! (OP10, SA01)<br />

10.00 – 10.30 Coffee break<br />

Chair: Ranka Serventi-Seiwerth (Croatia), Steven Z. Pavletic (USA)<br />

10.30 – 10.45 Marry Horowitz (USA)<br />

Allogeneic SCT for AML – when to treat (OP11)<br />

10.45 – 11.00 Didier Blaise (France)<br />

Allogeneic SCT for AML with reduced intensity conditioning (OP12)<br />

11.00 – 11.15 Hagop Kantarjian (USA)<br />

Novel drugs for AML (OP13, SA01)<br />

11.15 – 11.45 Questions <strong>and</strong> Answers<br />

12.15 – 13.45 Lunch Satellite Symposium Recent innovations in leukemia <strong>and</strong> lymphoma treatment<br />

– new hope?!<br />

Chair: Arnold Ganser (Germany)<br />

12.15 – 12.35 Raphael Duarte (Spain)<br />

More patients for autologous transplantation<br />

12.35 – 12.55 Kai Huebel (Germany)<br />

Further development in Stem Cell Mobilization <strong>and</strong> transplantation<br />

12.55 – 13.15 Arnold Ganser (Germany)<br />

Future Perspectives for Adult Myeloid Leukemia<br />

viii


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Scientific Program<br />

13.15 – 13.35 Rupert H<strong>and</strong>gretinger (Germany)<br />

Allogeneic Transplant Strategies for the Treatment of Pediatric Acute Leukemias<br />

13.35 – 13.45 Panel Discussion <strong>and</strong> Closing of the Symposium<br />

14.00 – 15.15 Lunch/Poster Session<br />

Poster presentation: Session 1. Acute leukemia I.<br />

Chair: Drago Batinic (Croatia)<br />

14.00 – 14.10 Rapamycin enhances dimethyl sulfoxide-mediated growth arrest in human myelogenous<br />

leukemia cells (PP01)<br />

Lalic H, Lukinovic-Skudar V, Banfic H, Visnjic D. Department of Physiology & CIBR,<br />

School of Medicine, University of Zagreb<br />

14.10 – 14.20 Ikaros transcription factors expression in cell differentiation (PP02)<br />

Antica M1 , Paradzik M1 , Matulic M2 , Batinic D3 , Labar B3 . 1Division of Molecular Biology,<br />

Ru|er Bo{kovi} Institute, Zagreb, 2Department of Molecular Biology, Faculty of Science, Zagreb,<br />

3University Hospital Center Zagreb <strong>and</strong> School of Medicine University of Zagreb, Croatia.<br />

14.20 – 14.30 Multicenter Perforomance Evaluation of the Multifrugquant Assay Kit (PP03)<br />

Márki-Zay J1 , Tauber Jakab K1 , Sipka S2 , Nagy G2 , Baráth S2 , Gyimesi E2 , Hevessy Zs3 , Sziráki Kiss V3 ,<br />

Szabó P5 , Tõkés-Füzesi M5 , Nagy É7 , Trucza É7 , Udvardy M4 , Borbényi Z8 , Dávid M6 , Kappelmayer J3 .<br />

1 2 3 Solvo Biotechnology, Szeged, Hungary; Regional Immunological Laboratory, Department of Clinical<br />

Biochemistry <strong>and</strong> Molecular Pathology, 42nd Department of Internal Medicine, University of Debrecen-<br />

Medical <strong>and</strong> Health Science Center, Debrecen, Hungary; 5Department of Laboratory Medicine,<br />

University of Pécs-Medical School, 61st Department of Internal Medicine, Pécs, Hungary; 7Department of Laboratory Medicine, 82nd Department of Medicine <strong>and</strong> Cardiology, University of Szeged – Albert<br />

Szent-Györgyi Clinical Center, Szeged, Hungary<br />

14.30 – 14.40 NPM1 mutations in AML detected by fragment analysis <strong>and</strong> Sanger sequencing (PP04)<br />

Crncec I, Musani V, Marusic Vrsalovic M, Livun A, Pejsa V, Jaksic O, Haris V, Ajdukovic R,<br />

Stoos Veic T, Kusec R. Departments of Hematology, Divison of molecular diagnostics <strong>and</strong> genetics<br />

<strong>and</strong> Cytology, Universtiy hospital Dubrava <strong>and</strong> Zagreb School of Medicine, Division of molecular<br />

medicine, Institute Rudjer Boskovic, Zagreb, Croatia.<br />

14.40 – 14.50 NPM1 mutation A <strong>and</strong> FLT3/ITD in AML with myelodysplasia-related changes (PP05)<br />

Radic Antolic M1 , Ries S2 , Zadro R1 , Davidovic S3 , Labar B4 . 1Department of Clinical Laboratory,<br />

2 3 4 Department of Pathology, Deaprtment of Pediatric, Department of Medicine, Clinical Hospital Center<br />

<strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

14.50 – 15.00 Prognostic Significance of CD56 Antigen Expression in Patients with Acute Myeloid<br />

Leukemia (PP06)<br />

Djunic I, Virijevic M, Djurasinovic V, Novkovic A, Colovic N, Kraguljac-Kurtovic N, Vidovic A,<br />

Suvajdzic-Vukovic N, Tomin D. Clinic of hematology, Clinical Center Serbia, Belgrade, Serbia<br />

15.00 – 15.10 FLT3 internal t<strong>and</strong>em duplication is a poor risk factor in patients with AML with diverse<br />

cytogenetic features (PP07)<br />

Mikulic M1 , Zadro R2 , Davidovic S3 , Serventi Seiwerth R1 , Sertic D1 , Nemet D1 , Batinic J1 , Batinic D2 ,<br />

Gjadrov K4 , Ries S4 , Labar B1 . 1Division of Hematology, 1Department of Medicine, 2Department of<br />

Clinical Laboratory, 3Department of Pediatric, 4Department of Pathology, Clinical Hospital Center<br />

<strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

Poster presentation: Session 2. Ph+ Chronic Myeloid Leukemia<br />

Chair: Martin Mistrik (Slovakia)<br />

14.00 – 14.10 A Philadelphia-negative Chronic Myeloid Leukemia with a BCR/ABL Fusion Gene (PP08)<br />

Lasan Trcic R 1 , Kardum I 2 , Jaksic B 2 , Jaksic O 3 , Kusec R 3 , Pejsa V 3 , Zadro R 4 , Batinic D 4 , Labar B 5 ,<br />

Begovic D 1 . 1 Division of Medical Genetics, Department of Paediatrics Univesity Hospital Center<br />

Zagreb, Zagreb, Croatia. 2 Division of Hematology, Department of Internal Medicine, Clinical Hospital<br />

Merkur, Zagreb, Croatia. 3 Division of Hematology Departmrnt of Internal Medicine, Clinical Hospital<br />

Dubrava, Zagreb, Croatia. 4 Clinical Laboratory Diagnostics, University Hospital Center Zagreb,<br />

Zagreb, Croatia 5 Division of Hematology, Department of Internal Medicine Univesity Hospital Center<br />

Zagreb, Zagreb, Croatia<br />

ix


Scientific Program Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

14.10 – 14.20 Croatian CML Registry based on European LeukemiaNet CML Registry (PP09)<br />

Sertic D1 , Peric Z1 , Sincic-Petricevic J2 , Lozic D3 , M<strong>and</strong>ac-Rogulj I4 , Duletic-Nacinovic A5 ,<br />

Gveric-Krecak V6 , Corovic-Arneri E7 , Pejsa V8 , Suvic-Krizanic V9 , Skunca Z10 , Babok-Flegaric R11 ,<br />

Krmek-Zupanic D12 , Carzavec D13 , Lazic-Prodan V14 , Coha B15 , Ajdukovic R8 , Romic I7 , Zadro R1 ,<br />

Davidovic S1 , Lasan-Trcic R1 , Labar B1 . Croatian Cooperative Group for Hematologic Diseases-CML<br />

Group: 1University Hospital Center Zagreb, 2University Hospital Center Osijek, 3University Hospital<br />

Center Split, 4Clinical Hospital Merkur Zagreb, 5University Hospital Center Rijeka, 6General Hospital<br />

[ibenik, 7General Hospital Dubrovnik, 8Clinical Hospital Dubrava-Zagreb, 9General Hospital Sisak,<br />

10 11 12 13 General Hospital Zadar, General Hospital Vara`din, General Hospital Sv.Duh-Zagreb, Clinical<br />

Hospital Center Sestre milosrdnice-Zagreb, 14General Hospital Pula, 15General Hospital Slavonski Brod<br />

14.20 – 14.30 Use of Imatinib mesylate in Chronic Phase CML in Clinical Practice – Our Experience (PP10)<br />

Miljkovic E, Markovic D, Cojbasic I, Nikolic V, Marjanovic G, Govedarovic N,<br />

Macukanovic-Golubovic L, Vucic M, Pavlovic M, Vukicevic T, Tijanic I, Simonovic O. Clinic of<br />

hematology, Clinical Center Nis, Serbia<br />

14.30 – 14.40 Imatinib for CML as a frontline therapy – Zagreb Experience (PP11)<br />

Sertic D1 , Zadro R2 , Nemet D1 , Davidovic S3 , Lasan-Trcis R3 , Radic Antolic M2 , Horvat I2 ,<br />

Frani} Simic I3 , Roncevic P1 , Radman I1 , Basic-Kinda S1 , Aurer I1 , Labar B1. 1Division of Hematolofy<br />

Department of Medicine, 2Department of Clinical Laboratory, 3Department of Pediatric, Clinical<br />

Hospitral Center <strong>and</strong> School od Medicine, University of Zagreb, Croatia<br />

14.40 – 14.50 Frequency of 4 bcr-abl1 kinase domain mutations in chronic myeloid leukemia patients resistant<br />

to imatinib mesylate therapy (PP12)<br />

Horvat I1 , Radic Antolic M1, Zadro R1 , Sertic D2 , Labar B2 . 1Department of Clinical Laboratory,<br />

2Department of Medicine, Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb,<br />

Croatia<br />

14.50 – 15.00 Imatinib mesylate in Patients with CML Relapse after Allogeneic Transplantation (PP13)<br />

Serventi Seiwerth R1 , Sertic D1 , Radic Antolic M2 , Mrsic M1 , Zadro R2 , Davidovic S3 , Grubic Z2 ,<br />

Mikulic M1 , Labar B1 . 1Department of Medicine, 2Department of Clinical Laboratory, 3Department of<br />

Pediatric, Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

Poster presentation: Session 3. Stem Cell Transplantation I.<br />

Chair: Alois Gratwohl (Switzerl<strong>and</strong>)<br />

14.00 – 14.10 Long-term survival <strong>and</strong> late-onset complications after reduced-intensity conditioning (RIC)<br />

allogeneic stem cell transplantation (allo-SCT) (PP14)<br />

Peric Z, Clavert A, Chevallier P, Brissot E, Malard F, Guillaume T, Delaunay J, Ayari S, Dubruille V,<br />

Le Gouill S, Mahe B, Gastinne T, Blin N, Harousseau JL, Moreau P, Milpied N, Mohty M. Service<br />

d’Hématologie Clinique, Centre Hospitalier et Universitaire (CHU) de Nantes, Nantes, France<br />

14.10 – 14.20 Factors predicting overall survival in patients with advanced chronic graft versus host disease<br />

diagnosed by NIH consensus criteria (PP15)<br />

Grkovic L1,2 , Steinberg SM1 , Baird K1 , Williams KM1 , Cowen EW1 , Mitchell SA1 , Pulanic D1,2 ,<br />

Avila DN1 , Taylor TN1 , Wroblewski SG1 , Serventi-Seiwerth R1,2 , Gress RE1 , Pavletic SZ1 . 1National Cancer Institute, National Institutes of Health, Bethesda, USA, 2Department of Medicine, Clinical<br />

Hospital Center Zagreb, Croatia<br />

14.20 – 14.30 Secondary Systemic Immunosuppressants (SSI) after Myeloablative HLA Matched Related<br />

<strong>and</strong> Unrelated Bone Marrow Transplantation (BMT) using High Dose Cyclophosphamide (Hi Cy)<br />

as sole GVHD prophylaxis (PP16)<br />

Durakovic N1 , Bolaños Meade J1 , Zahurak M2 , Kowalski J2 , Fuchs EJ1 , Jones RJ1 , Luznik L1 . 1Oncology, Johns Hopkins University, Baltimore, Maryl<strong>and</strong>, United States <strong>and</strong> 2Oncology Biostatistics, Johns<br />

Hopkins University, Baltimore, Maryl<strong>and</strong>, United States<br />

14.30 – 14.40 Allogeneic transplantation from HLA matched unrelated donor – single Center experience (PP17)<br />

Serventi Seiwerth R1 , Mikulic M1 , Mrsic M1 , Grubic Z2 , Bojanic I3 , Durakovic N1 , Stingl K2 , Labar B1 .<br />

1 2 3 Division of Hematology, Department of Medicine, Department of Clinical Laboratory, Department of<br />

Transfusiology, Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

14.40 – 14.50 Extracorporeal Photochemotherapy in Treatment of Chronic Graft-versus-Host Disease (PP18)<br />

Bojanic I1 , Serventi Seiwerth R2 , Golubic Cepulic B1 , Mazic S1 , Lukic M1 , Raos M1 , Plenkovic F1 ,<br />

Golemovic M1 , Dubravcic K3 , Perkovic S3 , Batinic D3 , Labar B2 . 1Department of Transfusiology,<br />

2 3 Division of Hematology, Department of Medicine, Department of Clinical Laboratory, Clinical<br />

Hospital Center <strong>and</strong> School of Medicine University of Zagreb, Croatia.<br />

x


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Scientific Program<br />

14.50 – 15.00 Wernicke’s encephalopathy in an adolescent after allogeneic hematopoietic stem cell<br />

transplantation: a case report (PP19)<br />

Rajic Lj1 , Pavlovic M1 , Femenic R1 , Bilic E1 , Krnic N1 , Barisic N1 , Ozretic D1 , Tesovic G4 , Dusek D4 ,<br />

Zadro R2 , Serventi Seiwerth R3 , Mikulic M3 , Konja J1 , Labar B3 . 1Department of Pediatric, 2Department of Clinical Laboratory, 3Department of Medicine, Clinical Hospital Center Zagreb, 4Clinical Hospital<br />

for Infectious Diseases, 1,2,3,4School of Medicine University of Zagreb, Croatia<br />

15.00 – 15.10 Need + Information + Satisfaction (PP20)<br />

Vieira S, Monroe D, Librojo M, London, United Kingdom<br />

Poster presentation: Session 4. Lymphoma<br />

Chair: Vlatko Pejsa (Croatia)<br />

14.00 – 14.10 CD43 expression is associated with inferior survival within activated B-cell group of diffuse large<br />

B-cell lymphoma (PP21)<br />

Mitrovic Z1,4 , Iqbal J1 , Fu K1 , Smith LM2 , Weisenburger DD1 , Greiner T1 , Auon P1 , Bast M3 , Armitage<br />

JO3 , Vose JM3 , Chan WC1 . Departments of 1Pathology/Microbiology <strong>and</strong> 3Hematology/Oncology, 2 4 College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA, School of<br />

Medicine University of Zagreb, Croatia<br />

14.10 – 14.20 Association of Interleukin-10,TNF-Alpha <strong>and</strong> TGF-Beta Gene Polymorphisms on The Outcome<br />

of Diffuse Large B-cell Lymphoma (PP22)<br />

Tarabar O, Tukic Lj, Cikota B, Milanovic N, Aleksic A, Magic Z. Military Medical Academy, Belgrade,<br />

Serbia<br />

14.20 – 14.30 Dose Adjusted EPOCH – Rituximab as First Line Treatment for High Risk Diffuse Large B-Cell<br />

Lymphoma: A Single Center Experience (PP23)<br />

Pejsa V, Prka Z, Lucijanic M, Jaksic O, Pirsic M, Ajdukovic R, Kusec R. Division of Hematolofy,<br />

Department of Medicine, Universiti Hospital Dubrava <strong>and</strong> School of Medicine, University of Zagreb,<br />

Croatia<br />

14.30 – 14.40 Immunoblastic Morphology as a Possible Prognostic Indicator for the Outcome of the Patients<br />

with Diffuse Large B cell Lymphoma in Era of the Rituximab Based Treatment: Single Centre<br />

Experience (PP24)<br />

Trajkova S, Panovska-Stavridis I, Stojanovik A, Petrusevska G, Dukovski D, Cevreska L. Department of<br />

Hematology, Medical Faculty, Skopje, Republic of Macedonia<br />

14.40 – 14.50 Primary Mediastinal Large B-Cell Lymphoma – Results of our Center (PP25)<br />

Miljkovic E, Marjanovic G, Cojbasic I, Tijanic I, Markovic D, Nikolic V, Macukanovic-Golubovic L,<br />

Govedarovic N, Vucic M. Clinic of hematology, Clinical Center Nis, Serbia<br />

14.50 – 15.00 Dose-Intense BACOP for Treatment of High-Risk Peripheral T-Cell NHL (PP26)<br />

Aurer I, Dujmovic D, Basic-Kinda S, Nemet D, Radman I, Ilic I, Stern-Padovan R, Santek F, Sertic D,<br />

Cigrovski N, Labar B. Departments of Internal Medicine, Pathology, Radiology <strong>and</strong> Oncology,<br />

University Hospital Center Zagreb, Zagreb, Croatia<br />

15.00 – 15.10 Oral lomustine, chlorambucil, etoposide <strong>and</strong> prednisolone (CCEP) for treatment of patients<br />

with advanced lymphoma (PP27)<br />

Radman I, Vodanovic M, Mitrovic Z, Aurer I, Basic-Kinda S, Labar B. Division of Hematology,<br />

Department of Medicine, Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb,<br />

Croatia<br />

Poster presentation Session 5. Myeloproliferative neoplasm <strong>and</strong> miscelaneous<br />

Chair: Rajko Kusec (Croatia)<br />

14.00 – 14.10 Efficacy <strong>and</strong> Safety of Ruxolitinib, a JAK1 <strong>and</strong> JAK2 Inhibitor, in Patients With Myelofibrosis:<br />

Results From a R<strong>and</strong>omized, Double-Blind, Placebo-Controlled, Phase III Trial (COMFORT-I)<br />

(PP28)<br />

Verstovsek S 1 , Mesa R 2 , Gotlib J 3 , Levy R 4 , Gupta V 5 , DiPersio J 6 , Catalano J 7 , Deininger M 8 , Miller C 9 ,<br />

Silver R 10 , Talpaz M 11 , Winton E 12 , Harvey J 13 , Arcasoy M 14 , Hexner E 15 , Lyons R 16 , Paquette R 17 ,<br />

Raza A 18 , Vaddi K 4 , Erickson-Viitanen S 4 , Koumenis I 4 , Sun W 4 , S<strong>and</strong>or V 4 , Kantarjian H 1 . 1 The<br />

University of Texas MD Anderson Cancer Center, Houston, TX, USA; 2 Mayo Clinic, Scottsdale, AZ,<br />

USA; 3 Stanford Cancer Center, Stanford, CA, USA; 4 Incyte Corporation, Wilmington, DE, USA;<br />

5 Princess Marg<strong>are</strong>t Hospital, Toronto, Canada; 6 Washington University School of Medicine, St. Louis,<br />

MO, USA; 7 Frankston Hospital, Frankston, Australia; 8 University of Utah Huntsman Cancer Institute,<br />

Salt Lake City, UT, USA; 9 Saint Agnes Cancer Institute, Baltimore, MD, USA; 10 Weill Cornell Medical<br />

xi


Scientific Program Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

College, New York, NY, USA; 11 University of Michigan, Ann Arbor, MI, USA; 12 Emory University School<br />

of Medicine, Atlanta, GA, USA; 13 Birmingham Hematology & Oncology, Birmingham, AL, USA; 14 Duke<br />

University Health System, Durham, NC, USA; 15 University of Pennsylvania Health System, Philadelphia,<br />

PA, USA; 16 Cancer C<strong>are</strong> Center of South Texas, San Antonio, TX, USA; 17 UCLA Medical Hematology &<br />

Oncology, Los Angeles, CA, USA; 18 Columbia Presbyterian Medical Center, New York, NY, USA.<br />

14.10 – 14.20 Prevalence of the JAK2 V617F Mutation in Croatian Patients with Classic Ph-Negative<br />

Myeloproliferative neoplasm (PP29)<br />

Duletic Nacinovic A, Dekanic A, Hadzisejdic I, Seili I, Grahovac B, Grohovac D, Valkovic T, Host I,<br />

Petranovic D, Jonjic N. Department of Medicine <strong>and</strong> Deppartment of Pathology, Clinical Hospital<br />

Center <strong>and</strong> School of Medicine, University of Rijeka, Croatia<br />

14.20 – 14.30 Frequency <strong>and</strong> Clinical Correlates of JAK2 46/1 Haplotype in Essential Thrombocythemia:<br />

Single Center Experience (PP30)<br />

Panovska-Stavridis I, Matevska N, Ivanovski M, Trajkova S, Dukovski D, Pivkova-Veljanovska A,<br />

Cevreska L, Dimovski A. Department of Hematology, Medical Faculty, Skopje, Republic of Macedonia<br />

14.30 – 14.40 Myeloid sarcoma – diagnostic <strong>and</strong> therapeutic pitfals (PP31)<br />

Antic D, Elezovic I, Perunicic Jovanovic M, Suvajdzic N, Djunic I, Mitrovic M, Tomin D.<br />

Clinic of hematology, Clinical Center Serbia, Belgrade, Serbia<br />

14.40 – 14.50 The Risk of Hematopoetic Malignancy in Children Born after in vitro Fertilization (PP32)<br />

Bilic E, Stemberger L, Slipac J, Stojsavljevic S, Bilic E, Konjevoda P, Konja J, Femenic R, Rajic Lj.<br />

Department of Pediatrics <strong>and</strong> Departmen of Neurology, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

14.50 – 15.00 H1N1 Influenza pneumonia in patient with chronic lymphocytic leukemia – case report (PP33)<br />

Vince A 1 , Barsic B 1 , Dusek D 1 , Vrhovac R 2 , Kutlesa M 1 , Santini M 1 . 1 University Hospital for Infectious<br />

Diseases »Dr. Fran Mihaljevic«, Zagreb, Croatia, 2 Department of Medicine, Division of Hematology.<br />

University Hospital »Merkur« Zagreb Croatia.<br />

15.00 – 15.10 Treatment of paroxismal nocturnal hemoglobinuria – a case report (PP34)<br />

Boban A 1 , Serventi Seiwerth R 1 , Basic-Kinda S 1 , Peric Z 1 , Dubravcic K 2 , Batinic D 2 , Labar B 1 . 1 Division<br />

of Hematology, Department of Medicine, 2 Department of Clinical Laboratory, Clinical Hospital Center<br />

<strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

15.30 – 17.15 SYMPOSIUM MDS<br />

Chair: David Steensma (USA), Pierre Wijermans (The Netherl<strong>and</strong>s)<br />

15.30 – 15.45 Ross Levine (USA)<br />

Molecular pathogenesis of myelodysplastic syndromes (OP14)<br />

15.45 – 16.00 David Steensma (USA)<br />

Risk models in MDS (OP15)<br />

16.00 – 16.15 Luca Malcovati (Italy)<br />

Transfusion iron overload <strong>and</strong> iron chelation therapy in MDS (OP16)<br />

16.15 – 16.30 Pierre Wijermans (The Netherl<strong>and</strong>s)<br />

Progress with epigenetic therapy in MDS (OP17)<br />

16.30 – 16.45 Theo de Witte (The Netherl<strong>and</strong>s)<br />

Hematopoietic cell transplantation for MDS: New Developments (OP18)<br />

16.45 – 17.00 Hagop Kantarjian (USA)<br />

Novel Agents in MDS (OP19)<br />

17.00 – 17.15 Questions <strong>and</strong> Answers<br />

17.15 – 17.45 Coffee Break<br />

17.45 – 19.15 MEET THE PROFESSOR<br />

Martin Mistik (Slovakia)<br />

Chronic myeloid leukemia (MP01)<br />

Petro Petrides (Germany)<br />

Essential Thrombocythaemia (MP02)<br />

17.45 – 19.15 END OF THE DAY DEBATE<br />

Moderator: Theo de Witte (The Netherl<strong>and</strong>s)<br />

AML vs MDS – what is similar<br />

Participants: Luca Malcovati (Italy)<br />

David Steensma (USA)<br />

xii


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Scientific Program<br />

Monday 19.09. 2011.<br />

08.30 – 10.30 MYELOPROLIFERATIVE NEOPLASM<br />

Chair: Srdjan Verstovsek (USA), Lidija Cevreska (Macedonia)<br />

08.30 – 08.45 Richard Van Etten (USA)<br />

Pathophysiology of MPNs <strong>and</strong> role of JAK2 mutations (OP20)<br />

08.45 – 09.00 Srdjan Verstovsek (USA)<br />

JAK2 inhibitors (OP21)<br />

09.00 – 09.15 Petro E. Petrides (Germany)<br />

Individualized MPN management–long term experience with anagrelide (OP22)<br />

09.15 – 09.30 Richard Silver (USA)<br />

Interferon as therapy for PV (OP23)<br />

09.30 – 09.45 Nicolaus Kröger (Germany)<br />

Allogeneic Hematopoietic Stem Cell Transplantation for Myelofibrosis (OP24)<br />

09.45 – 10.00 Vesna Najfeld (USA)<br />

Jumping 1q Translocations <strong>are</strong> Clonal Markers of Myeloid Malignancies Associated<br />

with Transformation to AML. (OP25)<br />

10.00 – 10.30 Question <strong>and</strong> Answers<br />

10.30 – 11.00 Coffee Break<br />

11.00 – 13.00 SYMPOSIUM CML<br />

Chair: Dubravka Sertic (Croatia), Elias Jabbour (USA)<br />

11.00 – 11.15 Rudiger Hehlmann (Germany)<br />

Prognostic tools in CML (OP26)<br />

11.15 – 11.30 David Marin (United Kingdom)<br />

Imatinib, still the best first line therapy?(OP27)<br />

11.30 – 11.45 Elias Jabbour (USA)<br />

Second line TKI – when, which one? (OP28)<br />

11.45 – 12.00 Gianantonio Rosti (Italy)<br />

Second line TKI – the best first line therapy? (OP29)<br />

12.00 – 12.15 Elias Jabbour (USA)<br />

How to monitor CML therapy (OP30)<br />

12.15 – 12.30 Alois Gratwohl (Switzerl<strong>and</strong>)<br />

Stem cell transplants for chronic phase CML (OP31)<br />

12.30 – 13.00 Questions <strong>and</strong> Answers<br />

13.15 – 14.45 Lunch Satellite Symposium: The optimal management of CML<br />

Chair: Gianantonio Rosti (Italy) Boris Labar (Croatia)<br />

13.15 – 13.25 Gianantonio Rosti (Italy)<br />

Introduction<br />

13.25 – 13.45 Giuseppe Saglio (Italy)<br />

Nilotinib versus Imatinib for Newly Diagnosed CML patients: ENESTnd study results (SP07)<br />

13.45 – 14.05 Francis Giles (Irel<strong>and</strong>)<br />

Optimal 1st line treatment for CML (SP08)<br />

14.05 – 14.25 Martin M. Mueller (Germany)<br />

The importance of molecular monitoring in CML patients (SP09)<br />

14.25 – 14.45 Boris Labar (Croatia)<br />

Panel discussion <strong>and</strong> conclusion<br />

xiii


Scientific Program Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

15.00 – 16.15 Poster Session<br />

Poster presentation: Session 6. Multiple myeloma<br />

Chair: Joan Bladé (Spain)<br />

15.00 – 15.10 Anti-proliferative <strong>and</strong> pro-apoptotic action of the combined cytostatic/immunosuppressive<br />

treatment of myeloma cell lines in vitro (PP35)<br />

Zelic A1 , Ivcevic S1 , Kovacic N1 , Kusec R2 , Grcevi} D1 . 1Zagreb University School of Medicine;<br />

2Dubrava University Hospital; Zagreb, Croatia<br />

15.10 – 15.20 T<strong>and</strong>em Transplantation in Multiple Myeloma – Single Center Experience (PP36)<br />

Batinic J1 , Durakovic N1 , Sertic D1 , Bojanic I2 , Batinic D3 , Golubic-Cepulic B2 ,<br />

Mazic S2 , Radman Livaja I1 , Basic Kinda S1 , Aurer I1 , Labar B1 , Nemet1 . 1Division of Hematology, Department of Medicine, 2Department of Transfusiology, 3Department of Clinical<br />

Laboratory, Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

15.20 – 15.30 The Role of Thalidomide as Maintenance Therapy in Multiple Myeloma (PP37)<br />

Bila J1,2 , Bodrozic J2 , Todorovic M1,2 , Sefer D2 , Kraguljac N2 , Antic D2 , Andjelic B2 , Elezovic I1,2 ,<br />

Tomin D1,2 , Gotic M1,2 , Mihaljevic B1,2 . 1Medical Faculty, University of Belgrade, 2Clinic of Hematology,<br />

CCS, Belgrade<br />

15.30 – 15.40 Modificated dosing of the VMP /Velcade + melphalan + prednisone/ regime in multiple myeloma<br />

/MM/ patients (PP38)<br />

Masarova K, Stefanikova Z, Mistrik M. Department of Hematology <strong>and</strong> Transfusiology, University<br />

Hospital, Bratislava, Slovakia<br />

15.40 – 15.50 Tevagrastim <strong>and</strong> Plerixafor as first line mobilizing regimen in patients with lymphoma<br />

<strong>and</strong> Multiple Myeloma (PP39)<br />

Andreola G, Babic A, Negri M, Drera M, Martinelli G, Laszlo D. Department of Haematology,<br />

European Institute of Oncology, Milan, Italy<br />

15.50 – 16.00 Does Prelixafor do the trick in mobilising Stem Cells? (PP40)<br />

Vieira S, Monroe D, Librojo M. London, United Kingdom<br />

Poster presentation: Session 7. Acute leukemia<br />

Chair: Dragica Tomin (Serbia)<br />

15.00 – 15.10 Multidrug Resistance: From Research to Clinics (PP41)<br />

Tauber Jakab K1 , Márki-Zay J1 , Kis E1 , Udvardy M2 , Borbényi Z3 , Dávid M4 , Krajcsi P1 . 1Solvo Biotechnology, Szeged, Hungary; 22nd Department of Internal Medicine, University of Debrecen-<br />

Medical <strong>and</strong> Health Science Center, Debrecen, Hungary; 32nd Department of Medicine <strong>and</strong> Cardiology,<br />

University of Szeged – Albert Szent-Györgyi Clinical Center, Szeged, Hungary; 41st Department of<br />

Internal Medicine, Pécs, Hungary<br />

15.10 – 15.20 P-glycoprotein activity <strong>and</strong> Flt3 internal t<strong>and</strong>em duplication in 39 patients with acute myeloid<br />

leukemia (PP42)<br />

Batinic D1 , Perkovic S1 , Zadro R1 , Labar B2 . 1Department of Clinical Laboratory, 2Division of Hematology, Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

15.20 – 15.30 The pretreatment risk factors <strong>and</strong> importance of comorbidity index for overall survival,<br />

complete remission <strong>and</strong> early death in patients with acute myeloid leukemia (PP43)<br />

Djunic I, Virijevic M, Novkovic A, Djurasinovic V, Colovic N, Vidovic A, Suvajdzic-Vukovic N,<br />

Tomin D, Clinic of hematology, Clinical Center Serbia, Belgrade, Serbia<br />

15.30 – 15.40 Treatment of Adolescents with Acute Lymphoblastic Leukemia (PP44)<br />

Konja J, Rajic Lj, Bilic E, Femenic R, Anicic M. Department of Pediatric, Clinical Hospital Center<br />

<strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

15.40 – 15.50 FLAG-IDA as the Salvage Regime for Relapsed <strong>and</strong> Refractory Acute Leukemias (PP45)<br />

Virijevic M, Suvajdzic N, Djunic I, Novkovic A, Djurasinovic V, Colovic N, Vidovic A, Tomin D.<br />

Clinic for hematology, Clinical Center Serbia, Belgrade, Serbia<br />

15.50 – 16.00 Clinical outcome <strong>and</strong> prognosis related factor in acute promyelocytic leukemia patients treating<br />

with PETHEMA LPA 099 protocol (PP46)<br />

Mitrovic M, Suvajdzic N, Bogdanovic A, Novkovic A, Kraguljac N, Sretenovic A, Antic D, Djunic I,<br />

Vidovic A, Colovic N, Virijevic M, Djurasinovic V, Elezovic I, Tomin D. Clinic of hematology,<br />

Clinical Center Serbia, Belgrade, Serbia<br />

xiv


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Scientific Program<br />

16.00 – 16.10 Acute myeloid leukemia with t(8;21)(q22;q22); RUNX1-RUNX1T1 – case report (PP47)<br />

Radic Antolic M, Rajic Lj, Femenic R, Bilic E, Pavlovic M, Perkovic S, Lasan R,<br />

Markovic-Glamocak M, Zadro R. Department of Clinical Laboratory <strong>and</strong> Department of Pediatrics,<br />

Clinical Hospital Center <strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

Poster presentation: Session 8. Lymphoma<br />

Chair: Igor Aurer (Croatia)<br />

15.00 – 15.10 Successful treatment of B-cell prolymphocytic leukemia (B-PLL) with FCR-Lite protocol (PP48)<br />

Batar P, Telek B, Udvardy M. Department of Hematology, University of Debrecen Health Sciences<br />

Center, Debrecen, Hungary<br />

15.10 – 15.20 Combination of rituximab <strong>and</strong> high-dose chlorambucil in therapy of small lymphocyte lymphoma<br />

/ chronic lymphocytic leukemia (PP49)<br />

Aurer I, Hude I, Basic Kinda S, Dujmovi} D, Nemet D, Radman I, Ilic I, Stern-Padovan R, Santek F,<br />

Sertic D, Labar B. Departments of Internal Medicine, Pathology, Radiology <strong>and</strong> Oncology, University<br />

Hospital Center Zagreb, Zagreb, Croatia<br />

15.20 – 15.30 Atypical Blastoid Variant of Hairy Cell Leukemia: a Case Report (PP50)<br />

Hadji-Pecova L, Petrusevska G, Panovska I, Stojanovski Z, Stojanovic A. Department of Hematology,<br />

Medical Faculty, Skopje, Republic of Macedonia<br />

15.30 – 15.40 Intravascular T-cell lymphoma presented as a subcutaneous panniculitis-like lymphoma:<br />

A case report (PP51)<br />

Ilic I(1), Basic Kinda S (2), Bosnic D (2), Aurer I 2(2), Dotlic S (2), Gasparovic V (1).<br />

1 2 Department of Pathology, Department of Medicine, Clinical Hospital Center <strong>and</strong> School of Medicine,<br />

University of Zagreb, Croatia<br />

15.40 – 15.50 Primarily Localized non Hodgkin Lymphoma of Testes with Relapse Localized in Buccal Area<br />

– Case Report (PP52)<br />

Hotic-Laz<strong>are</strong>vic S, Kezic Lj, M<strong>and</strong>ic D, Stojcic B, Malinovic J, Mrdja J. Division of Hematology,<br />

Department of Internal Medicine, Clinical Center Banja Luka, Banja Luka, Bosnia <strong>and</strong> Herzegovina<br />

15.50 – 16.00 A 48 Year Old Woman with a R<strong>are</strong> Case of a Midline Destructive Disease Presenting<br />

as Having both Midline <strong>and</strong> Nonmidline Lesions – a Case Report (PP53)<br />

Fern<strong>and</strong>ez S, Lapus F, B<strong>are</strong>z MY. Davao Medical Center, Davao City, Philippines<br />

16.00 – 16.10 Successful mobilisation with Plerixafor – clinical report (PP54)<br />

Babic A. Istituto Europeo di Oncologie, Divisione di Ematologia, Milan, Italy<br />

Poster presentation: Session 9. Stem Cell Transplantation II.<br />

Chair: Mirta Mikulic (Croatia)<br />

15.00 – 15.10 Isolation <strong>and</strong> in vitro cell culture of mesenchymal stem cells from human umbilical cord<br />

blood (PP55)<br />

Mazic S (1), Golemovic M (1), Skific M (1), Bojanic I (1), Humar I (2), Golubic Cepulic B (1).<br />

1 2 Department of Transfusiology, Deaprtment of Clinical Laboratory, Cliniacal Hospital Center, Zagreb,<br />

Croatia<br />

15.10 – 15.20 Biological properties of human mesenchymal stem cells exp<strong>and</strong>ed in vitro in media supplemented<br />

with human platelet lysate (PP56)<br />

Skific M (1), Golemovic M (1), Mazic S (1), Bojanic I (1), Humar I (2), Davidovic S (3),<br />

Crkvenac Gornik K (3), Ilic I (4), Durakovic N (5), Mikulic M (5), Serventi-Seiwerth R (5), Labar B (5),<br />

Golubic Cepulic B (1). 1Department of Transfusiology, 2Department of Clinical Laboratory, 3Department of Pediatric, 4Department of Pathology, 5Department of Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, Universiuty of Zagreb, Croatia<br />

15.20 – 15.30 Croatian Cord Blood Bank: the first 4 years (PP57)<br />

Mazic S (1), Bojanic I (1), Zlopasa G (2), Mrsic M (3), Plenkovic F (1), Lukic M (1), Raos M (1),<br />

Gojceta K (1), Golemovic M (1), Skific M (1), Tomac G (1), Burnac IL (1), Novoselac J (1), Vidovic I<br />

(1), Biskup M (1), Labar B (3), Golubic Cepulic B (1). 1Department of Transfusiology, 2Department of<br />

Gynecology <strong>and</strong> Obstetrics, 3Department of Medicine, Clinical Hospital Center <strong>and</strong> School of Medicine,<br />

Universiuty of Zagreb, Croatia<br />

15.30 – 15.40 Italian nurses group in mobilisation <strong>and</strong> apheresis (GIIMA): ideation <strong>and</strong> activity (PP58)<br />

Babic A, Laszlo D, Galgano L, Marchi E, Orl<strong>and</strong>o L, Martinelli G. Istituto Europeo di Oncologia,<br />

Divisione di Ematologia, Milan, Italy<br />

xv


Scientific Program Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

15.40 – 15.50 Unrelated Donor Search Experience in the Croatian Hematopoietic Stem Cell Transplantation<br />

Program (PP59)<br />

Grubic Z1 , Stingl K1 , Serventi Seiwerth R2 , Labar B2 , Zunec R1 . 1Tissue Typing Centre, 2Department of<br />

hematology, Clinical Hospital Centre Zagreb, Croatia<br />

15.50 – 16.00 Following the Chimerism Status of Patients after Hematopoietic Stem Cell Transplantation<br />

– Ten Years Experience (PP60)<br />

Stingl K1 , Grubic Z1 , Serventi Seiwerth R2 , Labar B2 , Rajic Lj3 , Vrhovac R4 , Zunec R1 . 1Tissue Typing<br />

Centre, 2Department of Medicine, 3Clinic of pediatrics, Clinical Hospital Centre, 4Department of<br />

Medicine, Clinical Hospital Merkur, Zagreb, Croatia<br />

Poster presentation: Session 10. Miscalenous<br />

Chair: Ivo Radman (Croatia)<br />

15.00 – 15.10 Venous access for Apheresis <strong>and</strong> Photopheresis procedures (PP61)<br />

Babic A. Istituto Europeo di Oncologia, Divisione di Ematologia, Milan, Italy<br />

15.10 – 15.20 Pure red cell aplasia preceding a DLBCL with 6 years (PP62)<br />

Barca G, Marin S, Nada S, Dragan C, Barbu D, Angelescu S, Tevet M, Saguna C, Bizu I, Lupu AR.<br />

Coltea Clinical Hospital, Hematology Clinic, Buch<strong>are</strong>st, Romania<br />

15.20 – 15.30 Can Platelet be Unnecessary (PP63)<br />

Vieira S, Monroe D, Librojo M, London, United Kingdom<br />

15.30 – 15.40 Transfuse or Not to Transfuse? (PP64)<br />

Vieira S, Monroe D, Librojo M, London, United Kingdom<br />

16.15 – 17.30 SYMPOSIUM MULTIPLE MYELOMA <strong>AND</strong> RELATED DISORDERS<br />

Chair: Bart Barlogie (USA), Damir Nemet (Croatia),<br />

16.15 – 16.30 Christoph J. Heuck (USA)<br />

Pathophysiology of Myeloma (OP32)<br />

16.30 – 16.45 Joan Blade (Spain)<br />

Bortezomib in the Treatment of Multiple Myeloma (OP33)<br />

16.45 – 17.00 Robert P. Gale (USA)<br />

IMIDs in Myeloma (OP34)<br />

17.00 – 17.15 Bart Barlogie (USA)<br />

Curing Multiple myeloma – the facts behind the claim (OP 35)<br />

17.15 – 17.30 Questions <strong>and</strong> Answers<br />

17.45 – 19.15 End of the Day Debate<br />

Moderator: Damir Nemet (Croatia)<br />

SCT in Myeloma – for all young patients?<br />

Participants: Bart Barlogie (USA)<br />

Joan Blade (Spain)<br />

Tuesday 20. 09. 2011.<br />

07.00 – 08.15 Satellite Symposium: Optimal management of PNH<br />

Chair: Peter Hillmen (United Kingdom) Boris Labar (Croatia)<br />

07.00 – 07.15 Boris Labar (Croatia)<br />

PNH – Morbidities & Mortality (SP10)<br />

07.15 – 07.40 Peter Hillmen (United Kingdom)<br />

Shifting the Paradigm in PNH Management (SP11)<br />

07.40 – 07.50 Jaroslav Cermak<br />

Acute renal failure <strong>and</strong> PNH – Case report (SP12)<br />

07.50 – 08.00 Ana Boban (Croatia)<br />

Treatment of Paroxismal nocturnal hemoglobinuria – a case report (SP13)<br />

08.00 – 08.15 Q & A <strong>and</strong> Summary<br />

xvi


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Scientific Program<br />

08.30 – 10.30 SYMPOSIUM ALL<br />

Chair: Jean Pierre Marie (France), Susan O’Brien (USA)<br />

08.30 – 08.45 Dieter Hoelzer (Germany)<br />

Adult ALL treatment approach for B-ALL (OP36)<br />

08.45 – 09.00 Stefan Faderl (USA)<br />

T-ALL current treatment (OP37, SA01)<br />

09.00 – 09.15 Jean Pierre Marie (France)<br />

Young adult with ALL – how to treat (OP38)<br />

09.15 – 09.30 Oliver Ottman (Germany)<br />

Ph+ALL – Current treatment options (OP39)<br />

09.30 – 09.45 Deborah Thomas (USA)<br />

MRD – criteria for postremission therapy (OP40)<br />

09.45 – 10.00 Mohamad Mohty (France)<br />

SCT for ALL: st<strong>and</strong>ard vs mini-allo (OP41)<br />

10.00 – 10.15 Sebastian Giebel (Pol<strong>and</strong>)<br />

Autologous SCT for ALL – stil the chalange (OP42)<br />

10.15 – 10.30 Susan O’Brien (USA)<br />

Novel drugs for ALL (OP43, SA01)<br />

10.30 – 11.00 Questions <strong>and</strong> Answers<br />

11.00 – 11.30 Coffee Break<br />

11.30 – 13.15 SYMPOSIUM CLL<br />

Chair: Aless<strong>and</strong>ra Ferrajoli (USA) Branimir Jaksic (Croatia)<br />

11.30 – 11.45 William Wierda (USA)<br />

Prognostic impact of Biologic factors in CLL (OP44)<br />

11.45 – 12.00 Susan O’Brien<br />

Frontline therapy for CLL (OP45)<br />

12.00 – 12.15 Aless<strong>and</strong>ra Ferrajoli (USA)<br />

Antibodies for CLL – alone or in combinations (OP46)<br />

12.15 – 12. 30 Peter Hillmen (United Kingdom)<br />

New <strong>and</strong> old drugs in CLL – we can do better (OP47)<br />

12.30 – 12.45 Issa Khouri (USA)<br />

SCT for CLL – when? (OP48)<br />

12.45 – 13.15 Questions <strong>and</strong> Answers<br />

13.30 – 15.00 Meet the Professor<br />

– Jiri Hollowiecki (Pol<strong>and</strong>) Acute myeloid leukemia (MP03)<br />

– Boris Labar (Croatia) Stem cell transplantation (MP04)<br />

– Deborah Thomas (USA) Acute lymphoblastic leukemia (MP05)<br />

– Petra Muus (The Netherl<strong>and</strong>s) Myelodysplastic syndrome (MP06)<br />

– Branimir Jaksic (Croatia) Chronic lymphocytic leukemia (MP07)<br />

– Srdjan Verstovsek (USA) Myelofibrosis (MP08)<br />

– Igor Aurer (Croatia) Non-Hodgkin Lymphoma (MP09)<br />

– Damir Nemet (Croatia) Multiple Myeloma (MP10)<br />

13.30 – 15.00 Lunch with Experts (1 expert on 10 registered particip.)<br />

Hagop Kantarijan (USA) Acute Myeloid Leukemia<br />

Dieter Hoelzer (Germany) Acute lymphoblastic Leukemia<br />

David Steensma (USA) Myelodysplastic Syndrome<br />

Rudiger Hehlmann (Germany) Chronic myeloid leukemia<br />

Peter Hillmen (United Kingdom) Chronic lymphocytic leukemia<br />

Bart Barlogie (USA) Multiple myeloma<br />

Patrice Carde (France) Hodgkin lymphoma<br />

xvii


Scientific Program Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

15.15 – 18.45 SYMPOSIUM STEM CELL TRANSPLANTATION<br />

Moderator: Boris Labar (Croatia)<br />

15.15 – 15.30 Introduction<br />

15.30 – 15.45 Alois Gratwohl (Switzerl<strong>and</strong>)<br />

Prognostic score for SCT (OP49)<br />

15.45 – 16.00 Mary Horowitz (USA)<br />

Consideration in choosing the optimal stem cell source (OP50)<br />

16.00 – 16.15 Mohamad Mohty (France)<br />

Source of stem cells in the allogeneic transplant setting: bone marrow versus peripheral blood stem<br />

cells (OP51)<br />

16.15 – 16.45 Dietger Niederwieser (Germany), John Barrett (USA)<br />

Conditioning for SCT – the impact of disease?! (OP52) (OP53)<br />

16.45 – 17.15 Coffee break<br />

17.15 – 18.15 Complications – anything new in prevention/treatment<br />

17.15 – 17.30 Finn Bo Petersen (USA)<br />

Infectious Complications of Stem Cell Transplantation (OP54)<br />

17.30 – 17.45 Leo Luznik (USA)<br />

Old drug, new tricks in prevention of graft-versus-host disease (GVHD): Development <strong>and</strong> clinical<br />

applicability of high-dose cyclophosphamide (OP55)<br />

17.45 – 18.00 Steven Z. Pavletic (USA)<br />

Chronic Graft-versus-Host Disease – Anything New in Prevention <strong>and</strong> Treatment? (OP56)<br />

18.00 – 18.15 Michael Bishop (USA)<br />

Relapse after Allogeneic Hematopoietic Stem Cell Transplantation – The Necessity for New Strategies<br />

<strong>and</strong> Treatments (OP57)<br />

18.15 – 18.45 Round table Discussion <strong>and</strong> Conclusion:<br />

Perspective of SCT<br />

19.00 – 19.20 SPECIAL LECTURE:<br />

Chair: Roel Willemze (The Netherl<strong>and</strong>s)<br />

19.00 – 19.20 Stefan Suciu (Belgium)<br />

Design, conduct <strong>and</strong> analysis of clinical trials in acute leukemia: the keys of success (OP58)<br />

20.30 – 24.00 F<strong>are</strong>well Dinner<br />

Wednesday 21. 09. 2011.<br />

08.30 – 11.45 SYMPOSIUM NON-HODGKIN <strong>LYMPHOMA</strong><br />

Chair: Nathan H. Fowler (USA) Igor Aurer (Croatia)<br />

08.30 – 08.45 Nathan H. Fowler (USA)<br />

Front line therapy for indolent B-cell lymphoma (OP59)<br />

08.45 – 09.00 Ofer Sphilberg (Israel)<br />

Rituximab maintenance treatment in indolent lymphoma (OP60)<br />

09.00 – 09.15 M<strong>are</strong>k Trneny (Czech Republic)<br />

Risk adapted treatment for patients with B-DLBCL (OP61)<br />

09.15 – 09.30 Piere Luiggi Zinzani<br />

Therapy for peripheral T-cell lymphoma (OP62)<br />

09.30 – 09.45 Christian H. Geisler (Denmark)<br />

Therapy of Mantle cell Lymphoma (OP63)<br />

19.45 – 10.00 Piere Luiggi Zinzani (Italy)<br />

Is there a role of radioimmunotherapy in NHL (OP64)<br />

10.00 – 10.30 Coffee Break<br />

10.30 – 10.45 Anas Younes (USA)<br />

Therapy for relapsed/refractory aggressive NHL (OP65)<br />

xviii


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Scientific Program<br />

10.45 – 11.00 Luis Fayad (USA)<br />

Alternative monoclonal antibodies for the treatment of NHL (OP66)<br />

11.00 – 11.30 Questions <strong>and</strong> Answers<br />

12.00 – 14.30 Symposium/Central <strong>and</strong> <strong>East</strong>ern European Leukemia Group<br />

CELG-AML-1 Study<br />

Chair: Zita Borbenyi (Hungary), Agnieszka Wierzbowska (Pol<strong>and</strong>), Georgi Mihajlov (Bulgaria)<br />

12.00 – 12.15 Boris Labar (Croatia)<br />

Design <strong>and</strong> Objective of the CELG-AML-1 Study<br />

12.15 – 12.30 Dragica Tomin (Serbia)<br />

Cytomorphology for diagnosis of AML (CELG-AML-1 Study)<br />

12.30 – 12.45 Zita Borbenyi (Hungary)<br />

Immunophenotyping for diagnosis of AML (CELG-AML-1 Study)<br />

12.45 – 13.05 Agnieszka Wierzbowska (Pol<strong>and</strong>)<br />

Cytogenetic <strong>and</strong> Molecular diagnostic for diagnosis of AML (CELG-AML-1 Study)<br />

13.05 – 13.25 Georgi Mihajlov (Bulgaria)<br />

CELG-AML-1 Study – Induction therapy <strong>and</strong> Consolidation<br />

13.25 – 13.40 Lidija Cevreska (Macedonia), Irina Panovska (Macedonia)<br />

CELG-AML-1 Study – Autologous SCT<br />

13.40 – 13.55 Labar B. (Croatia)<br />

CELG-AML-1 Study – Allogeneic SCT<br />

13.55 – 14.10 Panel Discussion <strong>and</strong> Conclusion<br />

Plerixafor for Stem Cell Mobilization<br />

Chair: Ines Bojanic (Croatia)<br />

14.10 – 14.30 Grzegorz Wladyslaw Basak<br />

Novel aspects of stem cell mobilization with plerixafor <strong>and</strong> G-CSF based on CELG data<br />

15.00 – 17.15 SYMPOSIUM HODGKIN <strong>LYMPHOMA</strong><br />

Patrice Carde (France), S<strong>and</strong>ra Basic-Kinda (Croatia)<br />

15.00 – 15.15 Patrice Carde (France)<br />

Early favorable <strong>and</strong> unfavorable Hodgkin lymphoma in adults, st<strong>and</strong>ards <strong>and</strong> directions (OP67)<br />

15.15 – 15.30 Berthe Aleman (The Netherl<strong>and</strong>s)<br />

Radiation therapy for Hodgkin’s lymphoma (OP68)<br />

15.30 – 15.45 Bastian von Tresckow (Germany)<br />

Improving Outcome in High Risk Hodgkin’s lymphoma (OP69)<br />

15.45 – 16.00 Martin Hutchings (Denmark)<br />

What is the role of PET in Hodgkin’s lymphoma (OP70)<br />

16.00 – 16.15 Igor Aurer (Croatia)<br />

Treatment of Relapsed/Refractory Hodgkin’s lymphoma (OP71)<br />

16.15 – 16.30 Ana Sureda (Spain)<br />

Allogeneic SCT for Hodkin’s lymphoma (OP72)<br />

16.30 – 16.45 Anas Younes (USA)<br />

New Drugs in the treatment of Hodkin’s lymphoma (OP73)<br />

16.45 – 17.15 Questions <strong>and</strong> Answers<br />

17.15 – 17.30 Adjourn<br />

xix


<strong>LEUKEMIA</strong> <strong>AND</strong> <strong>LYMPHOMA</strong><br />

<strong>East</strong> <strong>and</strong> <strong>West</strong> <strong>are</strong> <strong>Together</strong>


State of the Art I.<br />

SA01 Acute Leukemias<br />

Elias Jabbour, Susan O’Brien, Stefan Faderl,<br />

Farhad Rav<strong>and</strong>i, Jorge Cortes, Hagop Kantarjian,<br />

Department of Leukemia, U.T. M.D. Anderson Cancer<br />

Center, Houston, TX, USA<br />

Abstract: Acute leukemias <strong>are</strong> clonal malignant hematopoietic<br />

disorders resulting from genetic alterations in normal<br />

hematopoietic stem cells. These alterations induce differentiation<br />

arrest <strong>and</strong>/or excessive proliferation of abnormal<br />

»leukemic« cells or »blasts«. Acute leukemias <strong>are</strong> classified<br />

as those of myeloid or lymphoid lineage. Over the<br />

past several decades, improvements in chemotherapeutic<br />

regimens have improved outcomes in both acute myeloid<br />

leukemia (AML) <strong>and</strong> acute lymphoblastic leukemia (ALL).<br />

Better underst<strong>and</strong>ing of the biology of both AML <strong>and</strong> ALL<br />

has resulted in the identification of new therapeutic targets.<br />

Despite current optimism, most patients with AML still die<br />

of their disease, <strong>and</strong> only about 30–40% of adults achieve<br />

long-term disease-free survival in ALL. With better molecular<br />

definition <strong>and</strong> elucidation of the physiopathology of<br />

AML <strong>and</strong> ALL subtypes, <strong>and</strong> development of new <strong>and</strong> targeted<br />

therapies, a better outcome for acute leukemias may<br />

be achievable in the future.<br />

Epidemiology <strong>and</strong> Etiology<br />

The incidence of acute myeloid leukemia (AML) is 2.7 per<br />

100,000 while acute lymphoblastic leukemia (ALL) occurs<br />

at a rate of approximately 1 to 1.5 per 100,000 persons1–4 The median age at presentation for AML is 65 years. Along<br />

with its precursor myelodysplasia, AML appears to be rising,<br />

particularly in the population over the age of 60 <strong>and</strong> represents<br />

the most common type of acute leukemia in adults. ALL<br />

exhibits a bimodal distribution, with an early peak in children<br />

4 to 5 years old (4 to 5 per 100,000 persons), followed<br />

by a second peak at approximately 50 years of age (2 per<br />

100,000 persons). 4 ALL represents the most common childhood<br />

acute leukemia representing about 80% of acute leukemias,<br />

while it comprises only 20% of adult leukemias. 4,5<br />

The incidence of AML is slightly higher in males <strong>and</strong> in<br />

populations of European descent. Acute promyelocytic leukemia<br />

(APL), a distinct subtype of AML, is more common<br />

among populations of Latino or Hispanic background. 6,7 An<br />

increased incidence of AML is seen in patients with disorders<br />

associated with excessive chromatin fragility such as<br />

Bloom’s syndrome, Fanconi’s anemia, Kostmann’s syndrome,<br />

<strong>and</strong> with Wiskott-Aldrich syndrome or ataxia-telangiectasia.<br />

Other syndromes like Down’s (trisomy of chromosome<br />

21), Klinefelter (XXY <strong>and</strong> variants) <strong>and</strong> Patau<br />

(trisomy of chromosome 13) have also been associated with<br />

a higher incidence of AML. 8–11<br />

Survivors of the atomic bombs in Japan had an increased<br />

incidence of myeloid leukemias that peaked 5 to 7 years<br />

following exposure. 8 Therapeutic radiation increases AML<br />

risk, particularly if given with alkylating agents. There <strong>are</strong><br />

two main types of therapy-related AML. The »classic« alkylating-agent<br />

type (e.g. cyclophosphamide, melphalan,<br />

nitrogen mustard) has a latency period of 4 to 8 years, <strong>and</strong> is<br />

often associated with abnormalities of chromosome 5 <strong>and</strong>/<br />

or 7. Exposure to agents that inhibit the DNA repair enzyme<br />

topoisomerase II (e.g. etoposide, teniposide) <strong>are</strong> associated<br />

with secondary AML with a shorter latency period, usually<br />

1 to 3 years, with chromosome 11q23 at the location of the<br />

MLL gene, <strong>and</strong> with an M4 or M5 morphology. 8,12,13 Drugs<br />

such as chloramphenicol, phenylbutazone, chloroquine, <strong>and</strong><br />

methoxypsoralen can induce marrow damage that may later<br />

evolve into AML. Benzene, smoking, dyes, herbicides <strong>and</strong><br />

pesticides have been implicated as potential risk factors for<br />

development of AML. 9,10<br />

AML may also be secondary to progression of a myelodysplastic<br />

process or of a chronic bone marrow »stem cell«<br />

disorder, such as polycythemia vera, chronic myelogenous<br />

leukemia, primary thrombocytosis, or paroxysmal nocturnal<br />

hemoglobinuria. 10–14<br />

The etiology of ALL remains unknown. Chromosomal<br />

translocations occurring in utero during fetal hematopoiesis<br />

have been suggested as the primary cause for pediatric ALL,<br />

while postnatal genetic events <strong>are</strong> suggested as secondary<br />

contributors. A higher incidence of ALL is noted among<br />

mono <strong>and</strong> di-zygotic twins of patients with ALL, reflecting<br />

possible genetic predisposition. 5 Patients with trisomy 21,<br />

Klinefelter’s syndrome <strong>and</strong> inherited diseases with excessive<br />

chromosomal fragility such as Fanconi’s anemia,<br />

Bloom’s syndrome, <strong>and</strong> ataxia-telangiectasia have a higher<br />

risk of developing ALL. Implications have also been made<br />

towards infectious etiologies. 15–17 Associations between human<br />

T-cell lymphotrophic virus type 1 <strong>and</strong> adult T-cell<br />

leukemia/lymphoma, as well as HIV <strong>and</strong> lymphoproliferative<br />

disorders have been established. In addition, associations<br />

with varicella <strong>and</strong> influenza viruses have also been<br />

suggested.<br />

Clinical Presentation<br />

Fatigue, bruising or bleeding, fever <strong>and</strong> infection, reflecting<br />

a state of bone marrow failure, <strong>are</strong> common in both<br />

AML <strong>and</strong> ALL. Only 10% of AML patients present with<br />

white blood cell (WBC) counts greater than 100 x 109 /L. 2,18<br />

These patients <strong>are</strong> at higher risk of tumor lysis syndrome<br />

<strong>and</strong> leukostasis, both of which <strong>are</strong> considered oncologic<br />

emergencies <strong>and</strong> require prompt recognition <strong>and</strong> management.<br />

Tumor lysis syndrome can be due to spontaneous or<br />

treatment-mediated cell destruction <strong>and</strong> is characterized by<br />

hyperuricemia, renal failure, acidosis, hypocalcemia <strong>and</strong><br />

3


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

hyperphosphatemia. 19 Manifestations of leukostasis include<br />

dyspnea, chest pain, headache, altered mental status, cranial<br />

nerve palsies, <strong>and</strong>/or priapism. In addition, physical findings<br />

in AML may include organomegaly, lymphadenopathy,<br />

sternal tenderness, <strong>and</strong> retinal hemorrhages. Monocytic<br />

variants, M4 or M5, commonly display infiltration of gingivae,<br />

skin, soft tissues or meninges. 3 Disseminated intravascular<br />

coagulopathy (DIC) with bleeding diathesis is a common<br />

presentation in APL<br />

Whereas symptoms related to hyperleukocytosis can occur<br />

in AML, they <strong>are</strong> r<strong>are</strong> in ALL, even in the presence of<br />

high white cell counts. »B-symptoms« such as fever, night<br />

sweats, or weight loss can occur in ALL. Central nervous<br />

system (CNS) involvement (cranial neuropathies, meningeal<br />

infiltration) at presentation, common in mature B-ALL<br />

(Burkitt leukemia) occurs in 5% to 8% of patients. 20 Abdominal<br />

masses <strong>and</strong> significant spontaneous tumor lysis<br />

syndrome <strong>are</strong> more likely with mature B ALL. Lymphadenopathy<br />

<strong>and</strong> hepatosplenomegaly, r<strong>are</strong>ly symptomatic, <strong>are</strong><br />

noted in 20% of the patients, with a higher incidence in T<br />

<strong>and</strong> mature B cell ALL. The combination of hypercalcemia<br />

<strong>and</strong> lytic bone lesions <strong>are</strong> suggestive of adult T cell leukemia/lymphoma<br />

(ATLL). Chin numbness (mental nerve involvement),<br />

when elicited in the history or exam, is suggestive<br />

or mature B-cell ALL (Burkitt’s).<br />

Diagnosis <strong>and</strong> Classification<br />

The diagnosis of leukemia is often demonstrated by increased<br />

number of blasts in the bone marrow, or peripheral<br />

blood. By the French-American-British (FAB) Cooperative<br />

Group criteria, acute leukemia is diagnosed when a<br />

200-cell differential reveals the presence of 30% or more<br />

blasts in a marrow aspirate. 21 The minimal criterion has<br />

recently been changed to 20% by the World Health Organization<br />

(WHO). 22 Patients with the cytogenetic abnormalities<br />

t(8;21)(q22;q22), inversion(16)(p13q22) or<br />

t(16;16)(p13;q22), <strong>and</strong> t(15;17)(q22;q12) should be considered<br />

to have AML regardless of the blast percentage. After<br />

establishing the diagnosis, the blasts lineage (myeloid,<br />

lymphoid, or undifferentiated) is determined. The distinction<br />

is important <strong>and</strong> dictates specific therapy. Lineage determination<br />

is made using cytochemical stains (Figure 1). If<br />

3% or more blasts stain positive for myeloperoxidase (MPO)<br />

or Sudan Black B, the diagnosis is AML. If the blasts <strong>are</strong><br />

peroxidase negative, but stain for butyrate or non-specific<br />

esterase, acute monocytic leukemia is diagnosed. 1 If not,<br />

lineage determination is based on the blasts expressing surface<br />

antigens associated with the myeloid (equivalently<br />

monocytic, erythroid, or megakaryocytic), or lymphoid immunophenotype.<br />

Myeloid antigens <strong>are</strong> CD13, CD33, c-kit,<br />

CD 14, CD64 (the latter 2 <strong>are</strong> monocytic markers), glycophorin<br />

A (an erythroid marker), <strong>and</strong> CD41 (a megakaryocytic<br />

marker). Lymphoid markers <strong>are</strong> CD10, CD19, CD20<br />

(pre-B or B cells) <strong>and</strong> CD2, CD3, CD4, CD5, <strong>and</strong> CD8<br />

(T-cells). 23,24 A minority of MPO negative <strong>and</strong> Sudan Black<br />

B-negative cases <strong>are</strong> AML, <strong>and</strong> may include minimally differentiated<br />

(M0), <strong>and</strong> megakaryocytic leukemias (M7) that<br />

require flow cytometry for characterization. 1,25,26<br />

If the blasts <strong>are</strong> peroxidase <strong>and</strong> butyrate negative <strong>and</strong> express<br />

none of the myeloid or lymphoid antigens noted<br />

4<br />

Figure 1. Acute leukemia diagnostic algorithm<br />

above, the diagnosis is acute undifferentiated leukemia,<br />

which is treated like AML. The initial marrow aspirate may<br />

be a »dry tap«, reflecting marrow fibrosis. In this case, a<br />

biopsy is needed to exclude acute megakaryocytic leukemia<br />

or hairy cell leukemia. 26,27 If the marrow contains >50% normoblasts<br />

<strong>and</strong> pronormoblasts, the blast percent is based<br />

only on the non-erythroid cells; in these cases, the diagnosis<br />

is typically acute erythroid leukemia (M6), which can be<br />

confirmed if glycophorin A expression on the surface of the<br />

blasts.<br />

The WHO classification incorporates molecular, cytogenetic,<br />

<strong>and</strong> clinical features (prior hematological disorder) to<br />

the morphologic characteristics to better recognize the diversity<br />

of the disease, <strong>and</strong> its response to therapy. For example,<br />

AML with multilineage dysplasia by the WHO classification<br />

has no exact counterpart in the FAB classification.<br />

Several chromosomal abnormalities <strong>and</strong> mutations have<br />

been identified: The t (8;21), t(16;16) <strong>and</strong> inv 16 mutations<br />

<strong>are</strong> associated with a better prognosis, while alterations of<br />

11q23, leading to the MLL rearrangement. (mixed-lineage<br />

leukemia) portends a worse outcome (Table 1). The mutation<br />

in FLT3 (tyrosine kinase receptor) is the most common<br />

mutation in AML <strong>and</strong> is associated with poor clinical outcome<br />

(Table 2). T(15;17) is always associated with acute<br />

promyelocytic leukemia (APL), <strong>and</strong> leads to a mutation in<br />

retinoic acid receptor implicated in hematopoeitic differentiation.<br />

The FAB Cooperative Group distinguishes three ALL<br />

groups (L1 to L3) based on morphologic criteria (cell size,<br />

cytoplasm, nucleoli, basophilia, vacuolation). 21 The morphologic<br />

distinction between L1 <strong>and</strong> L2 has lost its prognostic<br />

significance. L3 morphology is associated with mature<br />

B-cell ALL (Burkitt’s leukemia). The WHO proposed<br />

new guidelines for the diagnosis of Neoplastic Diseases of<br />

Hematopoietic <strong>and</strong> Lymphoid Tissues or Lymphomas. 28 In<br />

addition to lowering the blast count to ≥20% as sufficient<br />

for an ALL diagnosis, the morphologic distinction of L1,<br />

L2, <strong>and</strong> L3 morphologies is ab<strong>and</strong>oned as no longer relevant.<br />

Both FAB <strong>and</strong> WHO classification systems continue<br />

to rely heavily on morphological assessment. 29 Identification<br />

of the immunophenotype has become a major part of


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

Karyotype % Frequency<br />

Table 1. AML cytogenetic risk group.<br />

% Complete<br />

Remission<br />

% Event-free<br />

Survival<br />

Favorable<br />

t(8;21) 5–10 90 60–70<br />

inv(16) 5–10 90 60–70<br />

t(15;17)<br />

Intermediate<br />

5–10 80–90 70<br />

Diploid, -Y<br />

Unfavorable<br />

40–50 70–80 30–40<br />

–5 / –7 20–30 50 5–10<br />

+8 10 60 10–20<br />

11q23, 20q- , other 10–20 60 10<br />

Table 2. Prognostic factors in AML.<br />

Relapse Rate Survival<br />

↑ BAACL NS ↓<br />

FLT3 ITD/mutation ↑ ↓<br />

MLL PTD ↑ NS<br />

↑ BCL2 <strong>and</strong> WT1 mRNA ↑ ↓<br />

↑ EVI1 mRNA ↑ ↓<br />

p53 mutation NS ↓<br />

CEBPA mutation ↓ ↑<br />

c-kit mutation ↑ ↓<br />

BAACL: Brain And Acute Leukemia Cytoplasmic; FLT3: Fms-like tyrosine<br />

kinase 3; MLL: Mixed lineage leukemia; BCL2: B-cell CLL lymphoma 2;<br />

WT1: Wilms tumor 1;<br />

EVI1: Ecotropic viral integration site 1; CEBPA: CCAAT/enhancer binding<br />

protein-alpha; NS: Not significant.<br />

Table 3. Immunophenotypic classification of ALL 30,33<br />

B Lineage T Lineage<br />

CD19/CD79a/CD22 CD3 (surface/cytoplasmic)<br />

Pre-pre-B ALL – Precursor T ALL CD1a, CD2, CD5,<br />

CD7, CD8, cCD3<br />

Common ALL CD10 (CALLA) Mature T ALL Surface CD3<br />

(plus any other<br />

T cell markers)<br />

Pre-B ALL Cytoplasmic IgM<br />

Mature B ALL Cytoplasmic or<br />

surface Igκ or λ<br />

ALL diagnosis (Table 3). 30–33 Three broad groups can be distinguished:<br />

precursor B-cell, mature B-cell, <strong>and</strong> T-cell ALL.<br />

Treatment (AML)<br />

Achievement of a complete remission (CR) significantly<br />

improves survival in AML, therefore the objective of therapy<br />

is to produce <strong>and</strong> maintain CR. 34 Criteria for CR include:<br />

platelet count ≥100 x 109 /l, neutrophil count ≥1 x<br />

109 /l, <strong>and</strong> ≤5% blasts present in the bone marrow. 35 The<br />

probability of AML recurrence sharply declines to less than<br />

10% after 3 years in CR, <strong>and</strong> patients in continuous CR for<br />

3 or more years can be considered »potentially cured«. 36 In<br />

recent years additional response criteria have been proposed,<br />

such as »CR with incomplete platelet recovery«<br />

(CRp), defined as CR with platelet count greater than 30 x<br />

109 /l, but less than 100 x 109 /l. 37<br />

Once AML is diagnosed <strong>and</strong> the decision to treat is made,<br />

the need for emergency therapy must be assessed. Emergency<br />

treatment is required in cases of APL, if the circulating<br />

blast count is > 50–100 x 10 9 /l, in the presence of DIC<br />

or organ dysfunction (especially pulmonary) attributed to<br />

leukemic infiltration (most seen in patients with > 10 x 10 9 /l<br />

circulating blasts <strong>and</strong>/or M4 or M5 FAB morphology). In<br />

the latter situation, it is important to initiate immediate chemotherapy.<br />

Leukapheresis for severe leukocytosis <strong>and</strong>/or<br />

leukostasis should be considered.<br />

Conventional treatment for AML, divided into remission<br />

induction <strong>and</strong> post-remission therapy, has been with combinations<br />

of anthracyclines <strong>and</strong> cytarabine. Traditionally, the<br />

anthracycline (i.e. idarubicin, daunorubicin) is usually administered<br />

daily for 3 days, <strong>and</strong> cytarabine is given at 100–<br />

200 mg/ m 2 daily for 7 days by continuous infusion (3+7<br />

regimen). In clinical practice, a bone marrow aspirate is<br />

usually obtained 2–3 weeks after beginning therapy. A biopsy<br />

is only needed if the quality of the aspirate does not<br />

permit determination of cellularity. If the marrow continues<br />

to show blasts <strong>and</strong> is cellular, a second course of the same<br />

therapy is often given, sometimes at a reduced total dose (2<br />

days anthracycline + 5 days cytarabine). If the day 14 or 21<br />

marrow is hypoplastic, therapy is usually delayed until it is<br />

clear that leukemia has reappe<strong>are</strong>d, at which time the second<br />

course begins. A second repeated course of therapy can<br />

produce remissions, but these <strong>are</strong> usually of shorter duration<br />

than remissions produced after one course of therapy, although<br />

this is controversial. 38 The timing of a second course<br />

with persistent AML is controversial. Several cooperative<br />

group studies advocated starting a second course if there is<br />

persistent AML on day 10–15 of chemotherapy. With highdose<br />

cytarabine (HDAC), a delay of a second course with<br />

persistent disease on day 21–28 may be indicated if the<br />

blasts <strong>are</strong> decreasing because most (90%) CRs <strong>are</strong> obtained<br />

after the first course, <strong>and</strong> response to a second course is<br />

poor. It is important to recognize that the initial marrow obtained<br />

after a period of hypoplasia may demonstrate up to<br />

30–50% blasts as a reflection of the regeneration of normal,<br />

not »leukemic,« marrow recovery. In this circumstance, follow-up<br />

(e.g. at 1–2 week intervals) marrows show reduction<br />

in blast percentages concomitant with a rise in neutrophils<br />

<strong>and</strong> platelets. Administration of a colony-stimulating factor<br />

may have an impact on the bone marrow findings.<br />

Three types of post-remission therapy can be distinguished.<br />

Maintenance therapy is usually defined as therapy<br />

less myelosuppressive than that used to produce remission.<br />

The doses used in consolidation therapy usually approach,<br />

whereas those in intensification therapy may surpass, the<br />

doses used during induction. Typically, once in remission<br />

after treatment with the »3 + 7« regimen, patients receive<br />

maintenance therapy, with the same drugs given during induction<br />

administered at approximately monthly intervals<br />

for 4–12 months. The need for a prolonged duration of<br />

maintenance therapy may depend on the intensity of induction<br />

<strong>and</strong> post-remission therapy. 39,40 For example, a r<strong>and</strong>omized<br />

German AML Cooperative Group (AMLCG) trial noted<br />

that addition of 3 years of maintenance improved the<br />

probability of 3-year relapse-free survival (RFS) from 7%<br />

to 30%. However, a similarly r<strong>and</strong>omized trial by the same<br />

5


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

group found no difference in RFS or survival when patients<br />

in the no maintenance arm received a more intensive induction<br />

treatment <strong>and</strong> one intense post-remission course .41–43<br />

Some maintenance therapy may be needed if intensive postremission<br />

therapy is not contemplated. However, any benefit<br />

from traditional maintenance after the administration of<br />

3–4 months of consolidation is relatively small, <strong>and</strong> perhaps<br />

nonexistent, with respect to survival. 44<br />

R<strong>and</strong>omized trials have attempted to identify which anthracycline<br />

(e.g. idarubicin [IDA], daunorubicin [DNR],<br />

mitoxantrone [MTZ], aclarubicin) is better. 45–47 Based on<br />

several such r<strong>and</strong>omized studies, IDA (12 mg/m 2 daily days<br />

1 through 3) has been proposed as the anthracycline of<br />

choice. 48 However, this may be misleading since the anthracyclines<br />

(IDA versus DNR) were not administered at equally<br />

toxic doses. 46 In several studies, st<strong>and</strong>ard-dose cytarabine<br />

plus MTZ 12 mg/m 2 daily x 3 has been comp<strong>are</strong>d with cytarabine<br />

plus DNR at 45 mg/m 2 daily x 3. Overall, the CR<br />

rates with the two regimens were similar, <strong>and</strong> there were no<br />

significant differences in the incidence or the severity of<br />

toxicities. In a three-arm r<strong>and</strong>omized study, there was no<br />

advantage when comparing DNR, IDA, <strong>and</strong> MTZ as part of<br />

the induction regimen for patients over 55 years of age. 49 In<br />

contrast, the three-arm r<strong>and</strong>omized trial, conducted by the<br />

EORTC <strong>and</strong> GIMEMA, comparing DNR to MTZ <strong>and</strong> IDA,<br />

reported that 5-year disease free survival (DFS) <strong>and</strong> overall<br />

survival (OS) were significantly better for patients receiving<br />

IDA <strong>and</strong> MTZ (p=0.03 <strong>and</strong> 0.02, respectively). Of note, the<br />

recovery time was longer with IDA <strong>and</strong> MTZ (p


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

appear to be intrinsically more resistant to st<strong>and</strong>ard chemotherapy<br />

<strong>and</strong> the presence of comorbid conditions <strong>and</strong> decreased<br />

marrow reserve contribute to the poor outcomes.<br />

The approximate CR rate of 45% in patients over age 60 is<br />

considerably lower than younger patients. In addition, elderly<br />

patients experience shorter remissions, minimal longterm<br />

<strong>and</strong> overall survival, <strong>and</strong> considerable treatment related<br />

mortality. Factors contributing to worse outcome in elderly<br />

AML <strong>are</strong>: age > 75 years, unfavorable karyotype (mostly<br />

complex), poor performance status <strong>and</strong> organ dysfunction,<br />

<strong>and</strong> longer duration of AHD. Treatment should be individualized<br />

<strong>and</strong> may include st<strong>and</strong>ard intensive therapy for patients<br />

with none or at most one adverse factor, or low-intensity<br />

investigational therapies for those with multiple adverse<br />

factors. Targeted therapies, low dose cytarabine, nucleoside<br />

analogs, <strong>and</strong> palliative regimens may be considered as lowintensity<br />

options. Investigational treatment or palliative c<strong>are</strong><br />

options become more plausible in poorer-prognosis elderly<br />

AML.<br />

Relapsed/Refractory AML<br />

Recurrent AML is common with approximately 10–50%<br />

of newly-diagnosed patients not achieving CR (»primary<br />

refractory«). The most important predictive factor for outcome<br />

of therapy in relapsed/refractory AML (salvage therapy)<br />

is duration of first CR. 64 Salvage therapy produces CR<br />

rates of 40% to 60% in patients with duration of first CR of<br />

one year or longer, but only 10% to 15% in those with shorter<br />

first CR duration. Allogeneic transplant appears superior<br />

to HDAC or idarubicin <strong>and</strong> cytarabine containing regimens<br />

in patients with first CR duration less than 1 year; the great<br />

majority of these transplants were from HLA-matched sibling<br />

donor. 65–68 However, since very few patients <strong>are</strong> cured<br />

with conventional therapy, all patients with relapsed or refractory<br />

AML should be treated in clinical trials.<br />

Mechanisms to lengthen remission duration require further<br />

evaluation. Beneficial effects of T-cells <strong>and</strong> natural<br />

killer cells on preventing relapse of AML have been recognized.<br />

69,70 However, the addition of recombinant interleukin-<br />

2 (rIL-2) during maintenance therapy has not led to improved<br />

outcomes. 54<br />

Investigational Agents<br />

Investigational therapies should be considered when expectations<br />

with st<strong>and</strong>ard therapy <strong>are</strong> low, either because of<br />

high mortality or high relapse rates. These include: 1) poor<br />

performance status 3–4; 2) age 80 years or older; 3) poor<br />

risk cytogenetics; or 4) intermediate prognosis cytogenetic<br />

group, but with an AHD, FLT3 mutation or (MDR)1 positivity.<br />

If investigational therapy is not feasible, palliative<br />

c<strong>are</strong> or low-dose cytarabine alone or with hydroxyurea may<br />

be considered in patients age 65 or older with poor performance<br />

status 3–4, or in patients 80 years or older regardless<br />

of performance status.<br />

Current investigational approaches include targeted therapy<br />

against surface antigens with monoclonal antibodies<br />

such as GO, signal transduction targeting (FLT3, farnesyl<br />

transferase inhibitors [FTIs], methylation, angiogenesis,<br />

inhibition of bcl-2 with antisense therapy) <strong>and</strong> new formu-<br />

lations of established drugs (e.g. liposomal formulations,<br />

pegylated formulations). Multidrug resistance (MDR)-reversing<br />

agents or nucleoside analogs like clofarabine, decitabine<br />

<strong>and</strong> azacitidine in different regimens <strong>are</strong> also of<br />

interest. 71–73<br />

Clofarabine, a nucleoside analog, has demonstrated activity<br />

with acceptable toxicity in elderly patients. 74 In newly<br />

diagnosed patients with 2 or more poor prognostic factors,<br />

39% of patients 70 years of age or older achieved CR with<br />

clofarabine 30 mg/m 2 administered intravenously daily for<br />

5 days. 75 Induction related mortality was reduced to 10%<br />

with clofarabine in comparison to conventional chemotherapy<br />

regimens for older AML patients that report 30 <strong>and</strong> 60<br />

day mortality ranging from 27% to above 60%. 76,77 In another<br />

study, a lower dose of clofarabine (20 mg/m 2 administered<br />

intravenously daily for 5 days) in combination with<br />

subcutaneously administered cytarabine led to higher rates<br />

of CR (59%), with similar induction mortality (9%). 78 Survival<br />

was longer with this combination (11.3 months) versus<br />

idarubicin <strong>and</strong> cytarabine (8.7 months), though this did<br />

not reach statistical significance. 79<br />

Decitabine, a pyrimidine analogue with significant antileukemic<br />

activity, induced an overall response rate of 32%<br />

(9 CRs <strong>and</strong> 7 PRs) among 50 patients with recurrent-refractory<br />

hematologic malignancies treated in a phase I study. 80<br />

In combination with valproic acid, the response rate was<br />

22%. 81 Decitabine has also displayed favorable response<br />

when used in combination with vorinostat. 82 A similar agent,<br />

azacitidine, has also been evaluated for elderly patients unfit<br />

for intensive chemotherapy. In a front-line investigation<br />

with or without valproic acid, CR of 17% <strong>and</strong> improvements<br />

in cytopenias were reported with azacitidine, both translating<br />

to survival benefit. 83<br />

Early clinical trials have demonstrated the activity of<br />

FLT3 inhibitors, particularly in patients who harbor FLT3<br />

mutations in their leukemic blasts. 84 Their role in combination<br />

with other agents is undergoing further investigation.<br />

CR was achieved in 88% (22/25) of newly diagnosed AML<br />

patients under the age of 65 years with the addition of<br />

sorafenib, an oral multi-kinase inhibitor, to idarubicin <strong>and</strong><br />

cytarabine. 85 A second generation FLT3 inhibitor, AC220,<br />

has demonstrated responses in a Phase 1 study, with phase<br />

II investigations ongoing. 86<br />

Another novel treatment for AML is the nucleoside analogue,<br />

sapacitabine, which displayed activity against AML<br />

<strong>and</strong> MDS patients in phase I study. Current evaluations have<br />

targeted the drug for patients 70 years of age or older with<br />

newly diagnosed AML or in first relapse. 87 Different dosing<br />

regimens have led to overall response rates ranging from<br />

25–45%, with mild to moderate toxicities reported that include<br />

fatigue, nausea, diarrhea, anemia, febrile neutropenia,<br />

thrombocytopenia, <strong>and</strong> peripheral edema.<br />

Acute promyelocytic leukemia (AML)<br />

This is a distinct subtype of AML accounting for 5% to<br />

15% of cases, with unique clinical, morphologic, <strong>and</strong> cytogenetic<br />

features. APL results from translocations between<br />

the retinoic acid α (RARα) locus on chromosome 17 <strong>and</strong><br />

the »promyelocytic leukemia« protein (PML) locus located<br />

7


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

on chromosome 15, demonstrable in 95–100% of cases. 88,89<br />

Independent risk factors for a diagnosis of APL in a patient<br />

with AML <strong>are</strong> younger age, Hispanic background, <strong>and</strong> obesity.<br />

7 The main clinical presentation is bleeding diathesis<br />

resulting both from plasmin-dependent primary fibrinolysis<br />

<strong>and</strong> disseminated intravascular coagulation. 90 Cognition of<br />

APL is crucial because appropriate treatment is different<br />

than for other types of AML <strong>and</strong> is curative in most patients<br />

with APL. 91,92<br />

Anthracyclines were historically the first effective treatment<br />

inducing a cure rate of 30–40% in APL. Addition of<br />

ATRA, 45 mg/m 2 daily, to chemotherapy (idarubicin) increases<br />

CR rate, <strong>and</strong> more dramatically the cure rate from<br />

40% to 70%. 93–95 Data suggests ATRA should be used together<br />

with anthracyclines during induction <strong>and</strong>, probably,<br />

during post-remission therapy. The major toxicity of ATRA<br />

is a potentially fatal leukocytosis syndrome (APL differentiation<br />

syndrome) characterized by fever <strong>and</strong> leakage of<br />

fluid into the extravascular space producing fluid retention,<br />

effusions, dyspnea, <strong>and</strong> hypotension. It is effectively treated<br />

with dexamethasone. 96 Use of frequent transfusions of platelets,<br />

cryoprecipitate, <strong>and</strong> fresh frozen plasma makes the use<br />

of heparin (historically recommended) unnecessary. The<br />

comparison of ATRA, arsenic trioxide (As 2 O 3 ) , <strong>and</strong> ATRA +<br />

As 2 O 3 in 61 patients with untreated APL (all patients also<br />

received consolidation chemotherapy <strong>and</strong> maintenance) displayed<br />

favorable response with combination therapy. 97<br />

ATRA <strong>and</strong> As 2 O 3 for induction <strong>and</strong> consolidation (without<br />

chemotherapy) induced a CR rate of 86%, molecular remission<br />

rate of 94% <strong>and</strong> 2-year event-free survival of 85%<br />

among 37 newly diagnosed APL patients. 98 The results of<br />

several studies support the role of As 2 O 3 in front-line therapy.<br />

99,100<br />

A stratification system has been developed that distinguishes<br />

newly-diagnosed patients with APL into low, intermediate,<br />

or high risk. Low risk patients present with WBC<br />

count less than 10 x 10 9 /l <strong>and</strong> platelet count above 40 x 10 9 /<br />

l; a WBC count above 10 x 10 9 /l identifies high-risk patients.<br />

Others <strong>are</strong> at intermediate risk. Anticipated cure rates<br />

<strong>are</strong> close to 100%, 90%, <strong>and</strong> 70%, respectively. Low-risk<br />

patients may only require 3–4 courses of post-remission<br />

therapy, following which PCR tests can be done every 3<br />

months for 1 year. Intermediate risk patients may require a<br />

longer duration of post-remission treatment (e.g. 6 months).<br />

High-risk patients have both a higher early death rate from<br />

hemorrhage <strong>and</strong> a higher risk of relapse. These patients may<br />

benefit from more intense post-remission therapy. More frequent<br />

PCR testing (e.g. monthly in CR) may also be advisable.<br />

Treatment (ALL)<br />

Therapy of ALL remains among the most complex therapies<br />

of anticancer programs. Multiple drugs <strong>are</strong> molded into<br />

regimen-specific sequences of dose- <strong>and</strong> time-intensity with<br />

the goal to reconstitute normal hematopoiesis, prevent<br />

emergence of resistant subclones, provide adequate prophylaxis<br />

of sanctuary sites (e.g. central nervous system (CNS),<br />

testicles), <strong>and</strong> eliminate minimal residual disease through<br />

post-remission consolidation <strong>and</strong> maintenance. 5 Three dis-<br />

8<br />

tinct phases <strong>and</strong> 4 components <strong>are</strong> distinguished: induction,<br />

intensified consolidation, maintenance, <strong>and</strong> CNS prophylaxis<br />

in the fourth component that accompanies induction<br />

<strong>and</strong> consolidation.<br />

The combination of vincristine, corticosteroids, <strong>and</strong> anthracyclines<br />

represents the backbone of ALL induction regimens.<br />

This combination achieves complete remission (CR)<br />

rates of 72% to 92% with a median remission duration of<br />

around 18 months. 101 Dexamethasone is often substituted<br />

for prednisone because of better in vitro antileukemic activity<br />

<strong>and</strong> achievement of higher drug levels in the cerebrospinal<br />

fluid (CSF). 102,103 While L-asparaginase is an important<br />

agent in the treatment of pediatric ALL, its role in adult<br />

ALL is not entirely well defined. Hematopoietic growth factors<br />

during induction accelerate recovery from myelosuppression<br />

<strong>and</strong> allow timely administration of dose-intense<br />

treatment regimens. 104 Consolidation represents a repetition<br />

of a modified induction schedule, rotational consolidation<br />

programs, or stem cell transplantation. Novel strategies try<br />

to emphasize subtype- <strong>and</strong> risk-oriented approaches of consolidation<br />

programs.<br />

Daily 6-mercaptopurine, weekly methotrexate, <strong>and</strong> monthly<br />

pulses of vincristine <strong>and</strong> prednisone, given over 2 to 3<br />

years <strong>are</strong> the mainstay of maintenance therapy. Extension of<br />

maintenance beyond 3 years, as well as omission or shortening<br />

of therapy is not beneficial. No maintenance therapy<br />

is given in mature B-cell ALL as these patients have a high<br />

cure rate with short-term dose-intense regimens, <strong>and</strong> relapses<br />

beyond the first year in remission <strong>are</strong> r<strong>are</strong>. The best maintenance<br />

for patients with Ph-positive ALL remains disputed,<br />

but should incorporate effective BCR-ABL tyrosine kinase<br />

inhibitors.<br />

Although CNS disease is uncommon at diagnosis (


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

liposomal cytarabine, <strong>and</strong> thiotepa. In the absence of IT<br />

therapy, isolated CNS recurrence can account for 10% to<br />

16% of relapses, warranting the inclusion of IT chemotherapy<br />

in CNS prophylactic regimens. The use of IT chemotherapy<br />

in combination with the hyper-CVAD regimen (hyperfractionated<br />

cyclophosphamide, vincristine, doxorubicin,<br />

dexamethasone alternating with methotrexate <strong>and</strong> highdose<br />

cytarabine), reduced the incidence of CNS relapse to<br />

4%. 105 The number of IT injections depend on the risk of<br />

CNS relapse (two IT treatments per course). Mature B-cell<br />

ALL, serum lactate dehydrogenase (LDH) levels, <strong>and</strong> a high<br />

proportion of bone marrow cells in a proliferative state<br />

(>14% of cells in S+G2M phase of the cell cycle) have been<br />

associated with a higher risk of CNS disease in adults. 118<br />

Patients with low-risk CNS disease receive six IT treatments,<br />

those with undetermined risk eight, <strong>and</strong> those with<br />

high-risk disease 16 IT treatments including all patients<br />

with mature B cell ALL. 119<br />

Philadelphia (Ph) chromosome-positive ALL<br />

Philadelphia chromosome (Ph) positivity is the most<br />

common cytogenetic abnormality in adults with ALL, occurring<br />

in 20–30% of patients. The outcome of patients with<br />

Ph-positive ALL with conventional chemotherapy remains<br />

poor with long-term disease-free survival (DFS) rates of<br />


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

Minimal residual disease (MRD) in adult ALL<br />

MRD describes the presence of disease below the threshold<br />

of detection by conventional methods (light microscopy<br />

<strong>and</strong> cytochemical stains). Different methodologies <strong>are</strong> available<br />

to detect <strong>and</strong> monitor MRD, including fluorescence in<br />

situ hybridization (FISH), multicolor flow cytometry, <strong>and</strong><br />

polymerase chain reaction (PCR) assays, especially real<br />

time quantitative (RQ) PCR. Multicolor flow cytometry <strong>and</strong><br />

PCR techniques take advantage of either fusion transcripts<br />

resulting from chromosome abnormalities (e.g. BCR-ABL,<br />

MLL-AF4, TEL-AML1) or patient-specific junctional regions<br />

of rearranged immunoglobulin <strong>and</strong> T cell receptor<br />

genes.<br />

Measurement of MRD in ALL has become an increasingly<br />

important prognostic factor for assessing risk of relapse.<br />

To which extent MRD needs to be controlled or eliminated<br />

remains uncertain. Several questions remain: i) what<br />

<strong>are</strong> the most appropriate time points for measurement of<br />

MRD. Persistence of MRD early in CR does typically not<br />

have the same significance as detection of residual disease<br />

in later stages of therapy; ii) Is there a definable threshold of<br />

residual disease by quantitative assays that would predict<br />

high relapse probability; iii) is it necessary to revise response<br />

criteria <strong>and</strong> introduce the concept of a »molecular<br />

relapse«. It should be emphasized that molecular relapse<br />

does not always predict clinical relapse, <strong>and</strong> intensification<br />

of therapy under these circumstances may not be appropriate;<br />

iv) can quantitation of MRD over time lead to a new<br />

risk classification with impact on choice of consolidation<br />

<strong>and</strong> maintenance strategies. Most of our current knowledge<br />

about MRD <strong>and</strong> its kinetics derive from studies in childhood<br />

ALL <strong>and</strong> differences in pattern <strong>and</strong> dynamics of clearance<br />

of MRD between adult <strong>and</strong> childhood ALL. Marrow<br />

samples from 33 adults <strong>and</strong> 21 children were analyzed by<br />

PCR for immunoglobulin heavy chain gene rearrangements<br />

at specific time points after diagnosis. 131 Among patients<br />

who remained in CR, a decrease in the MRD positivity occurred<br />

during the first 12 months. The proportion of positive<br />

tests decreased faster in children than in adults, suggesting<br />

more rapid resolution of MRD, particularly in the first 6<br />

months of CR.<br />

Salvage therapy<br />

Outcome of salvage therapy in adult ALL remains poor.<br />

CR rates r<strong>are</strong>ly exceed 50% <strong>and</strong> long-term DSF is r<strong>are</strong>. Independent<br />

prognostic factors associated with achieving CR<br />

include duration of first CR <strong>and</strong> platelet count. 132 Short duration<br />

of first CR, thrombocytopenia, elevated percent bone<br />

marrow blasts, <strong>and</strong> low albumin level represent factors associated<br />

with poor survival rates (Table 4). Salvage regimens<br />

<strong>are</strong> structured according to outcomes from frontline<br />

therapy. Newer agents have been approved in the relapsed<br />

setting, while the role of novel formulations of conventional<br />

active agents <strong>are</strong> being further investigated.<br />

Clofarabine demonstrates activity in acute leukemias. In<br />

phase 2 trials of clofarabine in relapsed or refractory pediatric<br />

leukemias, 31% of patients with ALL responded including<br />

12% CR, 8% CRp, <strong>and</strong> 10% partial response. 133 Based<br />

on the response rate in pediatric relapsed ALL, clofarabine<br />

10<br />

Table 4. Prognostic factors in relapsed ALL 123<br />

Poor prognostic factors for CR Poor prognostic factors for survival<br />

Albumin level < 3 g/L* Albumin level < 3 g/L*<br />

Duration of first CR < 36 months* Duration of first CR < 36 months*<br />

Hemoglobin level < 10 g/dL Hemoglobin level < 10 g/dL<br />

Platelet count ≤ 50 x 109 /L* Platelet count ≤ 50 x 109 /L*<br />

Percent bone marrow blasts > 50 % Percent bone marrow blasts > 50%*<br />

Peripheral blood blasts ≥ 1% Percent peripheral blood blasts ≥ 1%<br />

White blood cell count > 20 x 109 /L<br />

All prognostic factors listed significantly reduced rate of CR <strong>and</strong> survival.<br />

*Independent prognostic factors identified by multivariate analysis.<br />

was FDA approved for this indication. The maximum tolerated<br />

dose in adults with leukemia is 40 mg/m 2 /day for 5<br />

days, 20 times the MTD studied in solid tumors. 133 Unlike<br />

other purine analogs, clofarabine is not associated with neurotoxicity.<br />

Hepatotoxicity, skin rashes, drug fever, <strong>and</strong> palmoplantar<br />

erythrodyethseia have been reported. 133–136 Studies<br />

exploring the combinations of clofarabine with known<br />

active agents for ALL <strong>are</strong> ongoing.<br />

Nelarabine, an araguanosine analogue, has been shown to<br />

be very active as a single agent in recurrent T-cell ALL with<br />

a response rate more than 50% in first bone marrow relapse.<br />

When administered on an alternate day schedule of 1.5 mg/<br />

m 2 /day (day 1, 3, <strong>and</strong> 5) to relapsed/refractory T-cell ALL,<br />

31% of patients achieved CR. 129 Neurotoxicity with nelarabine<br />

has been of concern since initial studies. Concurrent<br />

intrathecal chemotherapy should be cautioned as the combination<br />

may compound the risk of neurotoxicity. Nelarabine<br />

in combination with hyper-CVAD is currently under investigation<br />

in newly diagnosed T-Cell ALL. Nelarabine given<br />

at 650 mg/m 2 daily x 5 days every 28 days for two courses<br />

as consolidation following completion of the hyper-CVAD<br />

sequence was feasible, however the true impact on survival<br />

has yet to be determined. 137<br />

Vincristine, a key component to the backbone of ALL<br />

therapy, causes peripheral neuropathy necessitating dose reductions<br />

or omission from the chemotherapy regimens. Liposomal<br />

formulations of chemotherapeutic agents generally<br />

produced reduced toxicity <strong>and</strong> increased efficacy. Activity<br />

has been observed in relapsed ALL with use of sphingosomal<br />

vincristine with minimal neurotoxicity comp<strong>are</strong>d<br />

with the conventional formulation. 138 While capping doses<br />

of conventional vincristine at 2 mg has become common<br />

practice, sphingosomal vincristine may be administered<br />

without dose capping. 139 Preliminary results <strong>are</strong> encouraging,<br />

with further studies planned.<br />

The role of pegaspargase in adult ALL requires further<br />

exploration. Although critical to DNA <strong>and</strong> RNA synthesis,<br />

asparagine can not be produced by ALL cells <strong>and</strong> it’s depletion<br />

through break down by asparaginase results in death of<br />

leukemic calls. Applying variations of pediatric regimens to<br />

adults may improve outcomes. Pegaspargase, E-Coli asparaginase<br />

liked to polyethylene glycol, minimizes reactions<br />

while maintaining the enzymatic activity of asparaginase.<br />

Combinations with pegaspargase <strong>are</strong> critical to determine<br />

the efficacy in adult ALL. When substituted for L-asparaginase<br />

in the BFM regimen (prednisone, vincristine, daunoru-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

bicin, L-asparaginase followed by cyclophosphamide, cytarabine,<br />

mercaptopurine <strong>and</strong> IT methotrexate), CR was reported<br />

at 96%. 140,141<br />

Modifications to the hyper-CVAD regimen mimicking<br />

approaches from pediatric regimens improve the outcomes<br />

in relapsed/refractory ALL. The design of the »augmented«<br />

hyper-CVAD regimen intensified the dosages of vincristine<br />

<strong>and</strong> dexamethasone, with the addition of L-asparaginase.<br />

With each course of »augmented« hyper-CVAD, vincristine<br />

dosed at 2 mg <strong>and</strong> L-asparaginase dosed at 20,000 units<br />

were administered on day 1, 8 <strong>and</strong> 15. 142,143 Additionally,<br />

dexamethasone was increased from the st<strong>and</strong>ard hyper-<br />

CVAD regimen to 80 mg daily on days 1–4 <strong>and</strong> days 15–18.<br />

Of 49 patients treated, 45% achieved CR with a median remission<br />

duration of 4.75 months. 143 L-asparaginase related<br />

toxicities were frequent with 5% of patients removed from<br />

study due to intolerance. The role of pegaspargase in combination<br />

with this regimen is currently under investigation.<br />

Specific sequencing of medications enhances antileukemic<br />

effect. For example, asparaginase increases the sensitivity<br />

of leukemic cells to methotrexate <strong>and</strong> administration<br />

24 hours after methotrexate reduces the toxicity of methotrexate.<br />

144 In previously treated ALL, combining sequential<br />

moderate-dose methotrexate (100–225 mg/m 2 ) <strong>and</strong> asparaginase<br />

with vincristine <strong>and</strong> dexamethasone (MOAD) proved<br />

beneficial in attaining remission. CR was achieved in 11 of<br />

14 (79%) previously treated patients. The median duration<br />

of CR was 7.5 months <strong>and</strong> the median survival was 11.2<br />

month.<br />

Stem cell transplant is recommended for all patients with<br />

Ph-chromosome positive ALL in first CR. For Ph-chromosome<br />

positive patients not eligible for transplant, survival is<br />

minimal. Acquired resistance to imatinib results from mutations<br />

in the kinase domain of ABL <strong>and</strong> BCR-ABL amplification.<br />

Second generation tyrosine kinase inhibitors appear<br />

promising in the management of imatinib resistant ALL.<br />

While imatinib only binds to BCR-ABL in the closed conformation,<br />

dasatinib binds in both the open <strong>and</strong> closed conformation,<br />

resulting in an increased affinity to BCR-ABL.<br />

The ability of dasatinib to inhibit src activity may provide<br />

advantage over imatinib by blocking BCR-ABL activity<br />

through a separate mechanism. Dasatinib, in combination<br />

with chemotherapy (hyper-CVAD) was evaluated in 14 patients<br />

with relapsed Ph-chromosome positive ALL or CML<br />

with lymphoid blast crisis. 145 Seventy-one percent of patients<br />

achieved CR while 29% attained complete response<br />

without platelet recovery (CRp). Major molecular response<br />

was achieved in 64%. Of four patients that had relapsed,<br />

two acquired the T315I mutation, resistant to currently<br />

available tyrosine kinase inhibitors.<br />

Stem Cell Transplant (SCT)<br />

High-dose chemotherapy with or without radiation followed<br />

by hematopoietic stem cell transplantation (SCT) is<br />

increasingly used as therapy for acute leukemia. AML in<br />

first CR, particularly in patients with poor-risk factors such<br />

as deletion of 5q <strong>and</strong> 7q, trisomy 8, t(6;9), t(9;22), <strong>and</strong> a<br />

history of myelodysplasia or AHD <strong>are</strong> c<strong>and</strong>idates for SCT.<br />

With a possible advantage of allogeneic transplantation in<br />

patients with poor-risk cytogenetic features, there <strong>are</strong> no<br />

subgroups identified in whom there is a clear advantage to a<br />

particular modality. 146 An exception is patients with inversion<br />

16, or t(8;21) who do better with chemotherapy. 147 Two<br />

other exceptions <strong>are</strong> patients younger than 20 years in whom<br />

transplant-related mortality is relatively low <strong>and</strong> who may<br />

do better with allogeneic stem cell transplantation, <strong>and</strong> patients<br />

older than 60 years in whom excessive mortality excludes<br />

the possibility of conventional myeloablative transplant.<br />

148,149 If allogeneic transplant is proposed, data suggest<br />

there is no advantage in giving consolidation chemotherapy<br />

before transplantation. 150<br />

The outcome with SCT versus continuation of chemotherapy<br />

in ALL has been debated. Although SCT is superior<br />

to chemotherapy with long-term disease-free survival rates<br />

of 20% to 40% in salvage, only 30% to 40% of patients who<br />

achieve a second CR <strong>are</strong> eligible for SCT <strong>and</strong> fewer than<br />

half have enough time prior to relapse to undergo SCT. Traditionally,<br />

SCT has been reserved for patients with Ph-positive<br />

ALL or in patients considered high risk (age greater<br />

than 35 years, B lineage with white blood cell count ≥ 100 x<br />

10 9 /L, T lineage with white blood cell count ≥ 30 x 10 9 /L). 151<br />

However, survival at 5 years was not significantly different<br />

between patients treated with a matched related donor SCT<br />

<strong>and</strong> those treated with continued chemotherapy or autologous<br />

SCT in high-risk patients. Improvements in survival at<br />

5 years were significantly greater in patients treated with a<br />

matched related donor SCT in patients not considered highrisk.<br />

SCT reduced the rate of relapse comp<strong>are</strong>d with chemotherapy<br />

or autologous SCT in both risk groups of patients.<br />

The use of chemotherapy is favored over autologous SCT<br />

for patients without a suitable donor.<br />

Conclusion<br />

The biology of both AML <strong>and</strong> ALL is now better understood.<br />

Following a period of paucity in discoveries, new<br />

strategies <strong>are</strong> finally evolving that may help patients with<br />

AML. Better definition of the complex process initiating<br />

<strong>and</strong> sustaining the leukemic process will lead to a better<br />

definition of targets for therapeutic intervention that may<br />

translate into improved cure rates. Specific attention must<br />

be given to prognostic factors that identify subsets of AML.<br />

Progress in the underst<strong>and</strong>ing of the biology <strong>and</strong> pathogenesis<br />

of adult ALL has helped improve outcome <strong>and</strong> prognosis.<br />

More intensive regimens combined with targeted therapy<br />

<strong>are</strong> being evaluated with encouraging results. As in other<br />

leukemias, the key to further improving prognosis of adult<br />

ALL lies in an appreciation <strong>and</strong> recognition of the heterogeneity<br />

of ALL. Novel strategies under investigation as monotherapy<br />

or in combination with chemotherapy provide improvements<br />

in the treatment of relapsed disease. Development<br />

of new drugs <strong>and</strong> agents tailored to subset-specific<br />

cytogenetic-molecular characteristics remains vital to the<br />

success in adult ALL. Progress in adults has been marked by<br />

utilization of intensive regimens proven beneficial in pediatric<br />

patients as well as the inclusion of new targeted therapy.<br />

Treatment remains highly successful in pediatric ALL <strong>and</strong><br />

with continuous investigation, adults may soon achieve<br />

comparable long term-survival.<br />

11


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

R E F E R E N C E S<br />

1. O’Donnell MR. Acute Leukemias. In: Pazdur R, Coia LR, Hoskins WJ,<br />

Wagman LD, eds. Cancer Management: A multidisciplinary approach.<br />

8th ed. Manhasset, NY: CMP Healthc<strong>are</strong> Media, the Oncology Group;<br />

2004:747–772.<br />

2. Foon KA, Casciato DA. Acute Leukemia. In: Casciato DA, Lowitz BB,<br />

eds. Manual of Clinical Oncology. 3rd ed. Little, Brown <strong>and</strong> Company;<br />

1995:431–445.<br />

3. Abraham J, Monahan BP. The Acute Leukemias. In: Abraham J, Allegra<br />

CJ, eds. Bethesda H<strong>and</strong>book of Clinical Oncology. 1st ed. Lippincott,<br />

Williams <strong>and</strong> Wilkins; 2001. Chapter 22:271–285.<br />

4. Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA Cancer<br />

J Clin 2004;54:8–29<br />

5. Jabbour EJ, Faderl S, Kantarjian HM. Adult acute lymphoblastic leukemia.<br />

Mayo Clin Proc 2005;80:1517–1527.<br />

6. Douer D, Preston-Martin S, Chang E, Nichols PW, Watkins KJ, Levine<br />

AM. High frequency of acute promyelocytic leukemia among Latinos<br />

with acute myeloid leukemia. Blood 1996;87:308–313.<br />

7. Estey E, Thall P, Kantarjian H, Pierce S, Kornblau S, Keating M. Association<br />

between increased body mass index <strong>and</strong> a diagnosis of acute<br />

promyelocytic leukemia in patients with acute myeloid leukemia. Leukemia<br />

1997;11:1661–1664.<br />

8. Pedersen-Bjergaard J, Christiansen DH, Andersen MK, et al. Causality<br />

of myelodysplasia <strong>and</strong> acute myeloid leukemia <strong>and</strong> their genetic abnormalities.<br />

Leukemia 2002;16:2177–2184.<br />

9. Dong F, Brynes RK, Tidow N, Welte K, Lowenberg B, Touw IP. Mutations<br />

in the gene for the granulocyte colony-stimulating-factor receptor<br />

in patients with acute myeloid leukemia preceded by severe congenital<br />

neutropenia. N Engl J Med 1995;333:487–493.<br />

10. <strong>West</strong> RR, Stafford DA, White AD, Bowen DT, Padua RA. Cytogenetic<br />

abnormalities in the myelodysplastic syndromes <strong>and</strong> occupational or<br />

environmental exposure. Blood 2000;95:2093–2097.<br />

11. Crane MM, Strom SS, Halabi S, et al. Correlation between selected<br />

environmental exposures <strong>and</strong> karyotype in acute myelocytic leukemia.<br />

Cancer Epidemiol Biomarkers Prev 1996;5:639–644.<br />

12. Ayton PM, Cleary ML. Transformation of myeloid progenitors by MLL<br />

oncoproteins is dependent on Hoxa7 <strong>and</strong> Hoxa9. Genes Dev 2003;<br />

17:2298–2307.<br />

13. Armstrong SA, Staunton JE, Silverman LB, et al. MLL translocations<br />

specify a distinct gene expression profile that distinguishes a unique<br />

leukemia. Nat Genet 2002;30:41–47.<br />

14. Haferlach T, Schoch C, Loffler H, et al. Morphologic dysplasia in de<br />

novo acute myeloid leukemia (AML) is related to unfavorable cytogenetics<br />

but has no independent prognostic relevance under the conditions<br />

of intensive induction therapy: results of a multiparameter analysis<br />

from the German AML Cooperative Group studies. J Clin Oncol<br />

2003;21:256–265.<br />

15. Mahieux R, Gessain A. HTLV-1 <strong>and</strong> associated adult T-cell leukemia/<br />

lymphoma. Rev Clin Exp Hematol 2003;7:336–361.<br />

16. Lombardi L, Newcomb EW, Dalla-Favera R. Pathogenesis of Burkitt’s<br />

lymphoma: expression of an activated c-myc oncogene causes the tumorigenic<br />

conversion of EBV-infected B lymphoblasts. Cell 1987;<br />

49:161.<br />

17. Vianna NJ, Polan AK. Childhood lymphatic leukemia: prenatal seasonality<br />

<strong>and</strong> possible association with congenital varicella. Am J Epidemiol<br />

1976;103:321–332.<br />

18. Appelbaum FR. Acute myeloid leukemia in adults. In: Abeloff M, Armitage<br />

J, Kastan M, et al, eds. Clinical Oncology, 3rd ed. Philadelphia,<br />

Pa: Elsevier science; 2004:2825–2848.<br />

19. Jabbour E, Ribrag V. Acute tumor lysis syndrome: update on therapy.<br />

Rev Med Interne. 2005;26:27–32.<br />

20. Cortes J. Central nervous system involvement in adult acute lymphocytic<br />

leukemia. Hematol Oncol Clin North Am 2001;15:145–162<br />

21. Bennett JM, Catovsky D, Daniel MT, Proposals for the classification of<br />

the acute leukaemias. French-American-British (FAB) co-operative<br />

group. Br J Haematol. 1976;33:451–458.<br />

22. Vardiman JW, Harris NL, Brunning RD. The World Health Organization<br />

(WHO) classification of the myeloid neoplasms. Blood 2002;100:<br />

2292–2302.<br />

23. Klobusicka M, Kusenda J, Babusikova O. Myeloid enzymes profile related<br />

to the immunophenotypic characteristics of blast cells from patients<br />

with acute myeloid leukemia (AML) at diagnosis. Neoplasma<br />

2005;52:211–218.<br />

12<br />

24. Wang XB, Zheng JE, Gu JX, et al. Correlation of immunophenotype to<br />

cytogenetics <strong>and</strong> clinical features of adult acute myeloid leukemia. Ai<br />

Zheng 2005;24:667–671.<br />

25. Roumier C, Eclache V, Imbert M, et al. M0 AML, clinical <strong>and</strong> biologic<br />

features of the disease, including AML1 gene mutations: a report of 59<br />

cases by the Groupe Francais d’Hematologie Cellulaire (GFHC) <strong>and</strong><br />

the Groupe Francais de Cytogenetique Hematologique (GFCH). Blood<br />

2003;101:1277–1283.<br />

26. Tallman MS, Neuberg D, Bennett JM, et al. Acute megakaryocytic leukemia:<br />

the <strong>East</strong>ern Cooperative Oncology Group experience. Blood<br />

2000;96:2405–2411.<br />

27. Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised criteria<br />

for the classification of acute myeloid leukemia. A report of the French-<br />

American-British Cooperative Group. Ann Intern Med 1985;103:<br />

620–625.<br />

28. Harris NL, Jaffe ES, Diebold J, et al. World Health Organization Classification<br />

of Neoplastic Diseases of the Hematopoietic <strong>and</strong> Lymphoid<br />

Tissues: report of the Clinical Advisory Committee Meeting – Airlie<br />

House, Virginia, November 1997. J Clin Oncol 1999;17:3835–3849.<br />

29. Foa R, Vitale A. Towards an integrated classification of adult acute<br />

lymphoblastic leukemia. Rev Clin Exp Hematol 2002;6:181.<br />

30. Bene MC, Castoldi G, Knapp W, et al. Proposals for the immunological<br />

classification of acute leukemias. Leukemia 1995;9:1783.<br />

31. Huh YO, Ibrahim S. Immunophenotypes in adult acute lymphoblastic<br />

leukemia. Role of flow cytometry in diagnosis <strong>and</strong> monitoring of disease.<br />

Hematol Oncol Clin North Am 2000;14:1251.<br />

32. P<strong>are</strong>des-Aguilera R, Romero-Guzman L, Lopez-Santiago N, et al.<br />

Flow cytometric analysis of cell-surface <strong>and</strong> intracellular antigens in<br />

the diagnosis of acute leukemia. Am J Hematol 2001;68:69.<br />

33. Terstappen LWMM, Huang S, Picker LJ. Flow cytometric assessment<br />

of human T-cell differentiation in thymus <strong>and</strong> bone marrow. Blood<br />

1992;79:666.<br />

34. Freireich EJ, Gehan EA, Sulman D et al. The effect of chemotherapy on<br />

acute leukemia in the human. J Chron Dis 1961;14:593.<br />

35. Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations<br />

of the International Working Group for Diagnosis, St<strong>and</strong>ardization of<br />

Response Criteria, Treatment Outcomes, <strong>and</strong> Reporting St<strong>and</strong>ards for<br />

Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol 2003;<br />

21:4642–4649.<br />

36. DeLima M, Strom SS, Keating M, et al. Implications of potential cure<br />

in acute myelogenous leukemia: development of subsequent cancer <strong>and</strong><br />

return to work. Blood 1997;90:4719–4724.<br />

37. Lopez G, Giles F, Cortes J et al. Sub-types of resistant disease in patients<br />

with AML, RAEB-t, or RAEB who fail initial induction chemotherapy.<br />

Blood 2001;98:329a, (Abstract#1388).<br />

38. Rowe JM, Kim HT, Cassileth PA, et al. Time to complete remission is<br />

not a significant prognostic factor in AML: a report on 1959 consecutive<br />

patients registered to 6 studies of the eastern cooperative oncology<br />

group (ECOG). Blood 2004;104:246a, (Abstract#862).<br />

39. Preisler HD, Anderson K, Rai K et al: The frequency of long-term remission<br />

in patients with acute myelogenous leukaemia treated with<br />

conventional maintenance chemotherapy: a study of 760 patients with<br />

a minimal follow-up time of 6 years. Br J Haematol 1989;71:189–194.<br />

40. Leone E, Almqvist A, Hanninen A, et al. Comparison between four <strong>and</strong><br />

eight cycles of intensive chemotherapy in adult acute myeloid leukemia:<br />

a r<strong>and</strong>omized trial of the Finnish Leukemia Group. Leukemia<br />

1998;12:1041–1048.<br />

41. Büchner T, Hiddemann W, Wörmann B, et al. Acute Myeloid Leukemia<br />

in adults: is postconsolidation maintenance therapy necessary? Intl J<br />

Hematology 2000;72:285–289.<br />

42. Büchner T, Grbanitz D, Hiddemann W, et al. Intensified induction <strong>and</strong><br />

consolidation with or without maintenance chemotherapy for acute<br />

myeloid leukemia (AML): two multicenter studies of the German AML<br />

Cooperative Group. J Clin Oncol 1985;3:1583–1590.<br />

43. Büchner T, Hiddemann W, Berdel B. Requestioning the role of prolonged<br />

maintenance chemotherapy in AML: a r<strong>and</strong>omized trial by the<br />

German AML Cooperative Group. Blood 2001;98:462a, (Abstract<br />

#1933).<br />

44. Sauter C, Fopp M, Imbach P, et al. Acute myelogenous leukaemia:<br />

maintenance chemotherapy after early consolidation treatment does not<br />

prolong survival. Lancet 1984;8373:379–382.<br />

45. Berman E, Heller G, Santorsa J, et al. Results of a r<strong>and</strong>omized trial<br />

comparing idarubicin <strong>and</strong> cytosine arabinoside with daunorubicin <strong>and</strong><br />

cytosine arabinoside in adult patients with newly diagnosed acute myelogenous<br />

leukemia. Blood 1991;77:1666–1674.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

46. Wiernik PH, Banks PL, Case DC Jr, et al. Cytarabine plus idarubicin or<br />

daunorubicin as induction <strong>and</strong> consolidation therapy for previously<br />

untreated adult patients with acute myeloid leukemia. Blood 1992;79:<br />

313–319.<br />

47. Vogler WR, Velez-Garcia E, Weiner RS, et al. A phase III trial comparing<br />

idarubicin <strong>and</strong> daunorubicin in combination with cytarabine in<br />

acute myelogenous leukemia: a Southeastern Cancer Study Group<br />

Study. J Clin Oncol 1992;10:1103–1111.<br />

48. Wheatley K. On behalf of the AML Collaborative Group L: meta-analysis<br />

of r<strong>and</strong>omized trials of idarubicin (IDAR) or mitoxantrone (MITO)<br />

versus daunorubicin (DNR) as induction therapy for acute myeloid leukemia<br />

(AML). Blood 1995;86:434a, (Abstract#1724).<br />

49. Rowe JM, Neuberg D, Friedenberg W, et al. A phase III study of daunorubicin<br />

vs idarubicin vs. mitoxantrone for older adult patients (>55<br />

yrs) with acute myelogenous leukemia (AML): a study of the <strong>East</strong>ren<br />

Cooperative Oncology Group (E3993). Blood 1998;92:313a, (Abstract<br />

#1284).<br />

50. Vignetti M, De Witte TM, Suciu S, et al. Daunorubicin (DNR) vs Mitoxantrone<br />

(MTZ) vs Idarubicin administered during induction <strong>and</strong><br />

consolidation in acute myelogenous leukemia (AML) followed by autologous<br />

or allogeneic stem cell transplantation (SCT): results of the<br />

EORTC-GIMEMA. Blood 2003;102:175a, (Abstract#611).<br />

51. Bishop JF, Lowenthal RM, Joshua D, et al. Etoposide in acute nonlymphocytic<br />

leukemia. Australian Leukemia Study Group. Blood 1990;<br />

75:27–32.<br />

52. Fern<strong>and</strong>ez HF, Sun Z, Yao X, et al. Anthracycline dose intensification<br />

in acute myeloid leukemia. N Engl J Med 2009;361:1249–1259.<br />

53. Lowenberg B, Ossenkoppele GJ, van Putten W, et al. High-dose daunorubicin<br />

in older patients with acute myeloid leukemia. N Engl J Med<br />

2009;361:1235–1248.<br />

54. Pautas C, Merabet F, Thomas, X, et al. R<strong>and</strong>omized study of intensified<br />

anthracycline doses for induction <strong>and</strong> recombinant interleukin-2 for<br />

maintenance in patients with acute myeloid leukemia age 5- to 70 years:<br />

results of the ALFA-9801 study. J Clin Oncol 2010;28:808–814.<br />

55. Burnett AK, Hills RK, Milligan D, et al. Attempts to optimize induction<br />

<strong>and</strong> consolidation chemotherapy in patients with acute myeloid leukaemia:<br />

results of the MRC AML 15 trial. Blood 2009;113:abstract 484.<br />

56. Holowiecki J, Grosicki S, Kyrcz-Krzemien, et al. Cladribine in combination<br />

with st<strong>and</strong>ard daunorubicine <strong>and</strong> cytarabine (DAC) as a remission<br />

induction treatment improves the overall survival in untreated<br />

adults with AML aged < 60 y contrary to combination including fludarabine<br />

(DAF): a multicenter, r<strong>and</strong>omized, phase II PALG AML 1/2004<br />

DAC/DAF/DA study in 673 patients – A final update. Blood 2009;114:<br />

abstract 2055.<br />

57. Mayer RJ, Davis RB, Schiffer CA, et al. Intensive post remission chemotherapy<br />

in adults with acute myeloid leukemia. Cancer <strong>and</strong> Leukemia<br />

Group B. N Engl J Med 1994;331:896–903.<br />

58. Cassileth PA, Lynch E, Hines JD, et al. Varying intensity of post remission<br />

therapy in acute myeloid leukemia. Blood 1992;79:1924–1930.<br />

59. Weick JK, kopecky KJ, Appelbaum FR, et al. A r<strong>and</strong>omized investigation<br />

of high-dose versus st<strong>and</strong>ard-dose cytosine arabinoside with daunorubicin<br />

in patients with previously untreated acute myeloid leukemia:<br />

a Southwest Oncology Group Study. Blood 1996;88:2841–2851.<br />

60. Bishop JF, Matthews JP, Young GA, Bradstock K, Lowenthal RM. Intensified<br />

induction chemotherapy with high dose cytarabine <strong>and</strong> etoposide<br />

for acute myeloid leukemia: a review <strong>and</strong> updated results of<br />

the Australian Leukemia Study Group. Leuk Lymphoma 1998;28:<br />

315–327.<br />

61. Borthakur G, Faderl S, Verstovsek S, et al. Clinical <strong>and</strong> molecular response<br />

in core binding factor acute myelogenous leukemia with fludarabine,<br />

cytarabine, G-CSF, <strong>and</strong> gemtuzumab ozogamicin. Blood 2009;<br />

114:abstract 2056.<br />

62. Borthakur G, Jabbour E, Wang X, et al. Effect of front-line therapy<br />

with fludarabine, cytarabine, filgrastim, <strong>and</strong> gemtuzumab ozogamicin<br />

(FLAG-GO) on outcome in core-binding factor associated acute myelogenous<br />

leukemia (CBF-AML). J Clin Oncol 2010;28:abstract 6552.<br />

63. FDA 2010. US Food <strong>and</strong> Drug Administration. Gemtuzumab ozogamicin.<br />

Available at: http://www.fda.gov/AboutFDA/CentersOffices/<br />

CDER/ucm216790.htm. Accessed July 22, 2010.<br />

64. Estey EH. Treatment of relapsed <strong>and</strong> refractory acute myelogenous leukemia.<br />

Leukemia 2000;14:476–479.<br />

65. Estey E, Kornblau S, Pierce S, et al. A stratification system for evaluating<br />

<strong>and</strong> selecting therapies in patients with relapsed or primary refractory<br />

AML. Blood 1996;88:756.<br />

66. Buckner CD, S<strong>and</strong>ers J, Appelbaum FR. Allogeneic marrow transplantation<br />

for acute non-lymphoblastic leukemia: first remission versus after<br />

first relapse. Bone Marrow Transplant 1989;4:244–251.<br />

67. Forman SJ, Schmidt GM, Nademanee AP, et al.: Allogeneic bone marrow<br />

transplantation as therapy for primary, induction failure for patients<br />

with acute leukemia. J Clin Oncol 1991;9:1570–1958.<br />

68. Biggs JC, Horowitz MM, Gale RP, et al. Bone marrow transplants may<br />

cure patients with acute leukemia never achieving remission with chemotherapy.<br />

Blood 1992;80:1090–1098.<br />

69. Archimbaud E, Bailly M, Dore JF. Inducibility of lymphokine activated<br />

killer (LAK) cells in patients with acute myelogenous leukaemia in<br />

complete remission <strong>and</strong> its clinical relevance. Br J Haematol 1991;<br />

77:328–334.<br />

70. Lauria F, Raspadori D, Rondelli D, et al. In vitro susceptibility of acute<br />

leukemia cells to the cytotoxic activity of allogeneic <strong>and</strong> autologous<br />

lymphokind activated killer (LAK) effectors: Correlation with the rate<br />

<strong>and</strong> duration of complete remission <strong>and</strong> with survival. Leukemia 1994;<br />

8:724–728.<br />

71. Kantarjian HM, O’Brien S, Estey E, et al. Decitabine studies in chronic<br />

<strong>and</strong> acute myelogenous leukemia. Leukemia 1997;11:S35–36.<br />

72. Kantarjian HM, G<strong>and</strong>hi V, Kozuch P, et al. Phase I clinical <strong>and</strong> pharmacology<br />

study of clofarabine in patients with solid <strong>and</strong> hematologic cancers.<br />

J Clin Oncol 2003;21:1167–1173.<br />

73. Faderl S, G<strong>and</strong>hi V, O’Brien S, et al. Results of a phase 1–2 study of<br />

clofarabine in combination with cytarabine (ara-C) in relapsed <strong>and</strong> refractory<br />

acute leukemias. Blood 2005;105:940–947.<br />

74. Burnett AK, Russell N, Kell JW, Milligan D, Culligan D. A phase 2<br />

evaluation of single agent clofarabine as first line treatment for older<br />

patients with AML who <strong>are</strong> not considered fit for intensive chemotherapy.<br />

Blood 2004;104:248a, (Abstract #869).<br />

75. Kantarjian HM, Erba HP, Claxton D, et al. Phase II study of clofarabine<br />

monotherapy in previously untreated older adults with acute myeloid<br />

leukemia <strong>and</strong> unfavorable prognostic factors. J Clin Oncol 2010;<br />

28:549–555.<br />

76. Appelbaum FR, Gundacker H, Head DR, et al. Age <strong>and</strong> acute myeloid<br />

leukemia. Blood 2006;107:3481–3485.<br />

77. Kantarjian H, O’Brien S, Cortes J, et al. Results of intensive chemotherapy<br />

in 998 patients age 65 years or older with acute myeloid leukemia<br />

or high-rick myelodysplastic syndrome: predictive prognostic<br />

models for outcome. Cancer 2006;106:1090–1098.<br />

78. Parikh SA, Kantarjian H, Garcia-Manero G, et al. Clofarabine plus<br />

low-dose cytarabine induction followed by consolidation with clofarabine<br />

plus low-dose cytarabine alternating with decitabine as frontline<br />

therapy for patients (pts) with acute myeloid leukemia (AML) ≥60<br />

years (yrs). Blood 2009;114:abstract 2058.<br />

79. Faderl S, Rav<strong>and</strong>i Kashani R, Borthakur G, et al. Analysis of survival<br />

of older patients with frontline AML treated with clofarabine with or<br />

without low-dose cytarabine or intermediate-dose cytarabine: A<br />

matched cohort comparison. J Clin Oncol 2010;28:abstract 6551.<br />

80. Issa JP, Garcia-Manero G, Giles FJ, et al. Phase 1 study of low-dose<br />

prolonged exposure schedules of the hypomethylating agent 5-aza-2’deoxycytidine<br />

(decitabine) in hematopoietic malignancies. Blood 2004;<br />

103:1635–1640.<br />

81. Garcia-Manero, Kantarjian H, Sanchez-Gonzalez B, et al. Results of a<br />

phase I/II study of the combination of 5-aza-2’-deoxycytidine (DAC)<br />

<strong>and</strong> valproic acid (VA) in patients with leukemia. Blood 2004;104:78a,<br />

(Abstract#263).<br />

82. Kirschbaum M, Gojo I, Goldberg S, et al. Phase I study of vorinostat in<br />

combination with decitabine in patients with relapsed or newly diagnosed<br />

acute myelogenous leukemia or myelodysplastic syndrome.<br />

Blood 2008;112: Abstract 3651.<br />

83. Thepot S, Itzykson R, Seegers V, et al. Azacytidine (AZA) as first line<br />

therapy in AML: results of the French ATU program. Blood 2009;114:<br />

abstract 843.<br />

84. Jabbour EJ, Yee KW, Giles FJ. FLT-3 receptor tyrosine kinase as a drug<br />

target in leukemia. Future oncology 2005;1:323–333.<br />

85. Rav<strong>and</strong>i F, Cortes J, Faderl S, et al. Combination of sorafenib, idarubicin,<br />

<strong>and</strong> cytarabine has a high response rate in patients with newly diagnosed<br />

acute myeloid leukemia (AML) younger than 65 years. Blood<br />

2008;112: Abstract 768.<br />

86. Cortes J, Foran J, Ghirdaladze D, et al. AC220, a potent, selective, second<br />

generation FLT3 receptor tyrosine kinase (RTK) inhibitor, in a first-inhuman<br />

(FIY) phase 1 AML study. Blood 2009;114:abstract 636.<br />

13


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

87. Kantarjian H, Garcia-Manero G, Luger S, et al. A r<strong>and</strong>omized phase<br />

2 study of sapacitabine, an oral nucleoside analogue, in elderly patients<br />

with AML previously untreated or in first relapse.<br />

88. Melnick A, Licht JD. Deconstructing a disease: RARα,its fusion partners,<br />

<strong>and</strong> their roles in the pathogenesis of acute promyelocytic leukemia.<br />

Blood 1999;93:3167–3215.<br />

89. Lo Coco F, Diverio D, Falini B et al. Genetic diagnosis <strong>and</strong> molecular<br />

monitoring in management of acute promyelocytic leukemia. Blood<br />

1999;94:12–22.<br />

90. Tallman MS <strong>and</strong> Kwaan HC. Reassessing the hemostatic disorder associated<br />

with acute promyelocytic leukemia. Blood 1992;79:542–553.<br />

91. Golomb HM, Rowley JD, Vardiman JW et al. »Microgranular« acute<br />

promyelocytic leukemia: a distinct clinical, ultrastructural, <strong>and</strong> cytogenetic<br />

entity. Blood 1980;55:253–260.<br />

92. Dyck JA, Warrell RP, Evans RM et al. Rapid diagnosis of acute promyelocytic<br />

leukemia by immunohistochemical localization of PML /<br />

RAR-α protein. Blood 1995;80:862–865.<br />

93. Fenaux P, Le Deley MC, Castaigne S, et al. Effect of all trans retinoic<br />

acid in newly diagnosed acute promyelocytic leukemia. Results of a<br />

multicenter r<strong>and</strong>omized trial. European APL 91 Group. Blood 1993;<br />

82: 3241–3249.<br />

94. Fenaux P, Castaigne S, Dombret H et al: Treatment of newly diagnosed<br />

APL by all transretinoic acid (ATRA) combined with chemotherapy:<br />

the European experience. Leukemia <strong>and</strong> Lymphoma 1995;16:<br />

431–438.<br />

95. Tallman MS, Andersen JW, Schiffer CA, et al. All-trans retinoic acid<br />

APL: long-term outcome <strong>and</strong> prognostic factor analysis from the North<br />

American Intergroup protocol. Blood 2002;100:4298–4302.<br />

96. Tallman MS, Andersen JW, Schiffer CA, et al. Clinical description of<br />

44 patients with APL who developed the retinoic acid syndrome.<br />

Blood 2000;95:90–95.<br />

97. Shen ZX, Shi ZZ, Fang J, et al. All-trans retinoic acid/As2O3 combination<br />

yields a high quality remission <strong>and</strong> survival in newly diagnosed<br />

acute promyelocytic leukemia. Proc Natl Acad Sci USA 2004;<br />

101:5328–5335.<br />

98. Estey EH, Garcia-Manero G, Ferrajoli A, et al. Use of all-transretinoic<br />

acid (ATRA) + arsenic trioxide (ATO) to eliminate or minimize<br />

use of chemotherapy (CT) in untreated acute promyelocytic leukemia<br />

(APL). Blood 2004;104:115a, (Abstract#393).<br />

99. Rav<strong>and</strong>i F, Estey E, Jones D, et al. Effective treatment of acute promyelocytic<br />

leukemia with all-trans-retinoic acid, arsenic trioxide, <strong>and</strong><br />

gemtuzumab ozogamicin. J Clin Oncol 2009;27:4:504–510.<br />

100. Ghavamzadeh A, Alimoghaddam K, Ghaffari S, et al. Results of new<br />

cases of APL treatment by arsenic trioxide <strong>and</strong> long-term follow-up:<br />

Is it time for using arsenic trioxide in first-line treatment. J Clin Oncol<br />

2010;28:abstract 6545.<br />

101. Kantarjian HM, O’Brien S, Smith T, et al. Acute lymphocytic leukaemia<br />

in the elderly: characteristics <strong>and</strong> outcome with the vincristineadriamycin-dexamethasone<br />

(VAD) regimen. Br J Haematol 1994;88:<br />

94–100.<br />

102. Jones B, Freeman Aik, Shuster JJ, et al. Lower incidence of meningeal<br />

leukemia when prednisone is replaced by dexamethasone in the<br />

treatment of acute lymphocytic leukemia. Med Pediatr Oncol 1991;<br />

19:269–275.<br />

103. Hurwitz CA, Silverman LB, Schorin MA, et al. Substituting dexamethasone<br />

for prednisone complicates remission induction in children<br />

with acute lymphoblastic leukemia. Cancer 2000;88:1964–1969.<br />

104. Ottman OG, Hoelzer D, Gracien E, et al. Concomitant granulocyte<br />

colony-stimulating factor <strong>and</strong> induction chemoradiotherapy in adult<br />

acute lymphoblastic leukemia: a r<strong>and</strong>omized phase III trial. Blood<br />

1995;86:444–450.<br />

105. Kantarjian HM, O’Brien S, Smith TL, et al. Results of treatment with<br />

hyper-CVAD, a dose-intensive regimen, in adult acute lymphocytic<br />

leukemia. J Clin Oncol 2000;18:547–561<br />

106. Cortes J, O’Brien SM, Pierce S, et al. The value of high-dose systemic<br />

chemotherapy <strong>and</strong> intrathecal therapy for central nervous system<br />

prophylaxis in different risk groups of adult acute lymphoblastic<br />

leukemia. Blood 1995;86:2091–2097.<br />

107. Mahmoud DH Jr, Rivera GK, Hancock ML, et al. Low leukocyte<br />

counts with blast cells in cerebrospinal fluid of children with newly<br />

diagnosed acute lymphoblastic leukemia. N Engl J Med 1993;329:<br />

314–319.<br />

108. Laack NN <strong>and</strong> Brown PD. Cognitive sequelae of brain radiation in<br />

adults. Semin Oncol 2004;31:702–713.<br />

14<br />

109. Pui C-H, Cheng C, Leung W, et al. Extended follow-up of long-term<br />

survivors of childhood acute lymphoblastic leukemia. N Engl J Med<br />

2003;349:640–649.<br />

110. Tucker J, Prior PF, Green CR, et al. Minimal neuropsychological sequelae<br />

following prophylactic treatment of the central nervous system<br />

in adult leukemia <strong>and</strong> lymphoma. Br J Cancer 1989;60:775.<br />

111. Pui C-H. Central Nervous System disease in acute lymphoblastic leukemia:<br />

Prophylaxis <strong>and</strong> treatment. Hematology Am Soc Hematol<br />

Educ Program 2006:142–46.<br />

112. Rudnick SA, Cadman EC, Capizzi RL, Skeel RT, Bertino JR, et al.<br />

High dose cytosine arabinoside (HDARAC) in refractory acute leukemia.<br />

Cancer. 1979;44(4):1189–1193.<br />

113. Capizzi RL, Yang JL, Cheng E, et al. Alteration of the pharmacokinetics<br />

of high-dose AraC by its metabolite, high araU in patients with<br />

acute leukemia. J Clin Oncol 1983, 1:763–771.<br />

114. H<strong>and</strong>e KR, Stein RS, McDonough DA, et al. Effects of high-dose<br />

cytarabine. Clin Pharmacol Ther 1982;31:669–674.<br />

115. Ackl<strong>and</strong> SP, Schilsky RL. High-dose methotrexate: a critical reappraisal.<br />

J Clin Oncol 1987;5;2017–2031.<br />

116. Wellwood J, Taylor K. Central Nervous System prophylaxis in hematological<br />

malignancies. Int Med J 2002;32:252–258.<br />

117. Gokbuget N, Hoelzer D. High-dose methotrexate in the treatment of<br />

adult lymphoblastic leukemia. Ann Hematol 1996;72:194–201.<br />

118. Kantarjian HM, Walters RS, Smith TL, et al. Identification of risk<br />

groups for development of central nervous system leukemia in adults<br />

with acute lymphocytic leukemia. Blood 1988;72:1784–1789.<br />

119. Kantarjian H, Thomas D, O’Brien S, et al. Long-term follow-up results<br />

of hyperfractionated cyclophosphamide, vincristine, doxorubicin,<br />

<strong>and</strong> dexamethasone (Hyper-CVAD), a dose-intensive regimen, in<br />

adult acute lymphocytic leukemia. Cancer 2004;101:2788–2801.<br />

120. Thomas DA, Faderl S, Cortes J, et al. Treatment of Philadelphia chromosome-positive<br />

acute lymphocytic leukemia with hyper-CVAD <strong>and</strong><br />

imatinib mesylate. Blood 2004;103:4396–4407.<br />

121. Thomas DA, Faderl S, Cortes J, et al. Update of the Hyper-CVAD <strong>and</strong><br />

imatinib mesylate regimen in Philadelphia (Ph) positive acute lymphocytic<br />

leukemia (ALL). Blood 2004;104:748a.<br />

122. Thomas DA, O’Brien SM, Faderl S, et al. Long-term outcome after<br />

hyper-CVAD <strong>and</strong> imatinib (IM) for de novo or minimally treated<br />

Philadelphia chromosome-positive acute lymphoblastic leukemia<br />

(Ph-ALL). J Clin Oncol 2010;28:abstract 6506.<br />

123. Rav<strong>and</strong>i F, Thomas D, Kantarjian H, et al. Phase II study of combination<br />

of hyperCVAD with dasatinib in frontline therapy of patients with<br />

Philadelphia chromosome (Ph) positive acute lymphoblastic leukemia<br />

(ALL). Blood 2008;112:Abstract 2921.<br />

124. Thomas DA, Cortes J, Faderl S, et al. Hyper-CVAD <strong>and</strong> rituximab<br />

therapy in HIV-negative Burkitt (BL) or Burkitt-like (BLL) leukemia/<br />

lymphoma <strong>and</strong> mature B-cell acute lymphocytic leukemia (B-ALL).<br />

ASCO Annual Meeting Proc 2005;23:567s.<br />

125. Thomas DA, Cortes J, O’Brien S, et al: Hyper-CVAD program in<br />

Burkitt’s-type adult acute lymphoblastic leukemia. J Clin Oncol 1999;<br />

17:2461–2470.<br />

126. Thomas DA, O’Brien S, Faderl S, et al. Chemoimmunotherapy with a<br />

modified hyper-CVAD <strong>and</strong> rituximab regimen improves outcome in<br />

de novo Philadelphia chromosome-negative precursor B-lineage acute<br />

lymphoblastic leukemia. J Clin Oncol 2010;28:3880–3889.<br />

127. Thomas DA, O’Brien S, Kantarjian HM. Monoclonal antibody therapy<br />

with rituximab for acute lymphoblastic leukemia. Hematol Oncol<br />

Clin North Am 2009;23:949–971.<br />

128. Dworzak MN, Schumich A, Printz D, et al. CD20 up-regulation in<br />

pediatric B-cell precursor acute lymphoblastic leukemia during induction<br />

treatment: Setting the stage for anti-CD20 directed immunotherapy.<br />

Blood 2008;112:3982–3988.<br />

129. DeAngelo DJ, Yu D, Johnson JL, et al. Nelarabine induces complete<br />

remission in adults with relapsed or refractory T-lineage acute lymphoblastic<br />

leukemia or lymphoblastic lymphoma: Cancer <strong>and</strong> Leukemia<br />

Group B study 19801. Blood 2007;109:5136–5142.<br />

130. O,Brien S, Thomas DA, Rav<strong>and</strong>i F, Faderl S, Pierce S, Kantarjian.<br />

Results of the hyperfractionated cyclophosphamide, vincristine,<br />

doxorubicin, <strong>and</strong> dexamethasone regimen in elderly patients with<br />

acute lymphocytic leukemia. Cancer 2008;113:2097–2101.<br />

131. Foroni L, Coyle LA, Papaioannou M, et al. Molecular detection of<br />

minimal residual disease in adult <strong>and</strong> childhood acute lymphoblastic<br />

leukaemia reveals differences in treatment <strong>and</strong> response. Leukemia<br />

1997;11:1732–1741.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

132. O’Brien S, Thomas D, Rav<strong>and</strong>i F, et al. Outcome of adults with acute<br />

lymphocytic leukemia after second salvage therapy. Cancer 2008;<br />

113:3186–3191.<br />

133. Jeha S, Razzouk BI, Rytting ME, et al. Phase II trials of clofarabine in<br />

relapsed or refractory pediatric leukemia. Blood 2004;104:196a.<br />

134. Jeha S, Kantarjian H. Clofarabine for the treatment of acute lymphoblastic<br />

leukemia. Expert Rev Anticancer Ther 2007;7:113–18.<br />

135. Kantarjian HM, G<strong>and</strong>hi V, Kozuch P, et al. Phase I clinical <strong>and</strong> pharmacology<br />

study of clofarabine in patients with solid <strong>and</strong> hematologic<br />

cancers. J Clin Oncol 2003;21:1167–1173.<br />

136. Kantarjian H, G<strong>and</strong>hi V, Cortes J, et al. Phase 2 clinical <strong>and</strong> pharmacologic<br />

study of clofarabine in patients with refractory or relapsed<br />

acute leukemia. Blood 2003;102:2379–2386.<br />

137. Faderl S, Thomas DA, Koller CA, et al. Hyper-CVAD plus nelarabine:<br />

a pilot study for patients with newly diagnosed T cell acute lymphoblastic<br />

leukemia (ALL)/lymphoblastic lymphoma (LL). Blood 2008;<br />

112:Abstract 3960.<br />

138. Thomas DA, Sarris AH, Cortes J, et al. Phase II study of sphingosomal<br />

vincristine in patients with recurrent or refractory adult acute<br />

lymphocytic leukemia. Cancer 2006;106:120–127.<br />

139. O’Brien S, Damon LE, Larson ML, et al. Early signs of tolerability<br />

<strong>and</strong> activity in a phase 2 study of weekly vincristine sulfate liposomal<br />

injection (VSLI, Marqibo) in adults with Philadelphia chromosomenegative<br />

acute lymphoblastic leukemia (ALL) in second relapse or<br />

progressing following two anti-leukemia treatment lines. Blood 2008;<br />

112:Abstract 3959.<br />

140. Douer D, Yampolsky H, Cohen LJ, et al. Pharmacodynamic <strong>and</strong> safety<br />

of intravenous pegaspargase during remission induction in adults<br />

aged 55 years or younger with newly diagnosed acute lymphoblastic<br />

leukemia. Blood 2007;109:2744–2750.<br />

141. Hoelzer D, Thiel E, Loffler H, et al. Prognostic factors in a multicenter<br />

study for treatment of acute lymphoblastic leukemia in adults.<br />

Blood 1988;71:123–131.<br />

142. Faderl S, Thomas DA, Garcia-Manero G, et al. Augmented hyper-<br />

CVAD in acute lymphoblastic leukemia(ALL): the MDACC experience<br />

with intensified L-asparaginase <strong>and</strong> vincristine in adult ALL<br />

salvage. Blood 2005;106:Abstract 1840.<br />

143. Faderl S, Thomas DA, Rav<strong>and</strong>i F, et al. Intensification of hyper-cvad<br />

with L-asparaginase, vincristine, <strong>and</strong> dexamethasone (»Augmented<br />

hyper-CVAD) has activity in adult patients with relapsed/refractory<br />

acute lymphoblastic leukemia(ALL). Blood 2007;110:Abstract 4324.<br />

144. Esterhay RJ, Wiernik PH, Grove WR, Markus SD, Wesley MN. Moderate<br />

dose methotrexate, vincristine, asparaginase, <strong>and</strong> dexamethasone<br />

for treatment of adult acute lymphocytic leukemia. Blood 1982;<br />

59:334–45.<br />

145. Jabbour E, O’Brien S, Thomas DA, et al. Combination of the hyper-<br />

CVAD regimen with dasatinib is effective in patients with relapsed<br />

Philadelphia chromosome (Ph) positive acute lymphoblastic leukemia<br />

(ALL) <strong>and</strong> lymphoid blast phase chronic myeloid leukemia (CML-<br />

LB). Blood 2058;112:Abstract 2919.<br />

146. Burnett AK, Goldstone AH, Stevens RM, et al. Biologic characteristics<br />

of disease determines the outcome of allogeneic or autologous<br />

BMT in CR1. Blood 1995;86:616a.<br />

147. Burnett AK. Transplantation in first remission of acute myeloid leukemia.<br />

N Engl J Med 1998;339:1698–1700.<br />

148. Woods WG, Neudorf S, Gold S, et al. A comparison of allogeneic<br />

bone marrow transplantation, autologous bone marrow transplantation,<br />

<strong>and</strong> aggressive chemotherapy in children with acute myeloid<br />

leukemia in remission. Blood 2001;97:56–62.<br />

149. Slavin S. Treatment of myeloid leukemias with non-myelo-ablative<br />

stem cell transplantation: accomplishments <strong>and</strong> future goals. Hematology,<br />

American Society of Hematology Education Program Book<br />

2000:81–89.<br />

150. Tallman MS, Rowlings PA, Milone G, et al. Effect of post remission<br />

chemotherapy before human leukocyte antigen-identical sibling transplantation<br />

for acute myelogenous leukemia in first complete remission.<br />

Blood 2000;96:1254–1258.<br />

151. Goldstone AH, Richards SM, Lazarus HM, Tallman MS, Buck G, et<br />

al. In adults with st<strong>and</strong>ard-risk acute lymphoblastic leukemia, the<br />

greatest benefit is achieved from a matched sibling allogeneic transplantation<br />

in first complete remission, <strong>and</strong> an autologous transplantation<br />

is less effective than conventional consolidation/maintenance<br />

chemotherapy in all patients: final results of the International ALL<br />

Trial (MRC UKALL XII/ECOG 2993). Blood 2008;111:1827–1833.<br />

15


Oral Presentations<br />

OP01 White blood cell transfusion <strong>and</strong> leukemia<br />

therapy<br />

Emil Freireich, The University of Texas MD Anderson<br />

Cancer Center, Houston, Texas, USA<br />

The leading cause of morbidity <strong>and</strong> mortality in leukemia<br />

patients results from uncontrolled infection. Since the late<br />

50s, it has been clear that leucopenia is a major contributing<br />

factor to the occurrence of infection. Both the severity <strong>and</strong><br />

duration of leucopenia result in increased incidence of infection.<br />

Similarly, thrombocytopenia is the major contributing<br />

cause to hemorrhage. Transfusion of allogeneic platelets<br />

has been very effective in controlling hemorrhage, <strong>and</strong> decreasing<br />

hemorrhage as a cause of morbidity <strong>and</strong> mortality.<br />

However, for infections, replacement of leucocytes by transfusion<br />

has been frustrated by inability to technically collect<br />

a sufficient number of leucocytes to increase the number of<br />

leucocytes in the circulation, <strong>and</strong> the physiology of the leucocytes<br />

where a great majority of the cells (over 95%) <strong>are</strong><br />

located outside of the vasculature. Thus, the volume of distribution<br />

is at least 20 fold that of red cells. First efforts to<br />

overcome this limitation were the studies of the transfusion<br />

of allogeneic leucocytes collected from donors with uncontrolled<br />

chronic myeloid leukemia (CML), which allowed<br />

transfusion of between 10 <strong>and</strong> 100 billion cells to leucopenic<br />

children with acute leukemia. These studies demonstrated<br />

that with an adequate dose of these cells, even abnormal<br />

granulocytes from CML patients, doses of 50–100 billion<br />

cells was uniformly effective in controlling established infection.<br />

To reach these concentrations of normal cells required<br />

the development of an efficient <strong>and</strong> semi-automated<br />

continuous blood flow cell separator; the application of a<br />

safe <strong>and</strong> largely degradable macromolecule to increase separation<br />

efficiency of the granulocytes; <strong>and</strong> the discovery of<br />

the granulocytes stimulating colony factors to mobilize myeloid<br />

cells from the bone marrow into the blood to allow<br />

collection. With these advances, it became possible to regularly<br />

collect 30–50 billion leucocytes from a single volunteer<br />

donor <strong>and</strong> transfusion of these cells into severely leucopenic<br />

patients with existing infection resulted in significant<br />

reversal of infection in 40% of patients who received a minimum<br />

of 4 consecutive daily transfusions. These data were<br />

reported in the late 1970s, <strong>and</strong> in several centers leucocyte<br />

transfusion was practiced. In the mid 80s, following several<br />

anecdotal reports of a syndrome resembling Graft vs Host<br />

Disease (GVHD) were reported in literature. These were<br />

single or two patient reports. However, these reports result<br />

in radiating all leucocyte transfusion products from the mid<br />

80s to 2008. It is important to recognize that several metaanalyses<br />

of all the published reports of transfusion associated<br />

GVHD did not support the conclusion that such a syndrome<br />

resulted from leucocyte transfusion.<br />

While radiation of the transfusion product did not have a<br />

major effect on neutrophil function in vitro, it completely<br />

eliminated lymphoid cells <strong>and</strong> thus was considered to be<br />

free of any possibility of transfusion associated Graft vs<br />

Host Disease (TAGVHD). In vitro studies subsequently demonstrated<br />

that in addition to the effect on lymphoid cells,<br />

the mononuclear monocytes <strong>and</strong> macrophages were completely<br />

inactivated by doses of radiation administered. But<br />

perhaps most importantly since the GCSF mobilized leucocytes<br />

collections were demonstrated to have myeloid hemapoietic<br />

stem cells <strong>and</strong> a large number of undifferential<br />

myeloid cells which, in the recipient, were able to continue<br />

to mature into granulocytes. The consequence was that the<br />

half life of measured granulocytes post non-radiated transfusions<br />

was approximately 24 hours, whereas the half life of<br />

the radiated products was approximately 6 hours. In 2008<br />

we undertook a prospective r<strong>and</strong>omized double-blind study<br />

comparing unradiated granulocytes with the same product<br />

which has been radiated in vitro. The goal of this study was<br />

primarily to determine the risk of TAGVHD was, <strong>and</strong> secondly,<br />

to quantitatively measure the effect of radiation on<br />

post transfusion increments <strong>and</strong> granulocyte levels in patients.<br />

The study was closed on July 14 th 2011 <strong>and</strong> we found,<br />

after the study of 109 patients, that in the 49 patients who<br />

received irradiated granulocytes transfusion, there were no<br />

patients who developed any syndrome resembling GVHD.<br />

While the 30 day survival <strong>and</strong> the response rates <strong>and</strong> overall<br />

survival were not significantly different, the increments<br />

measured post transfusion were more than twice in the recipients<br />

of non irradiated transfusions than those in the radiated<br />

transfusions. This study led to two conclusions – first,<br />

the unradiated leucocytes transfusions have the potential of<br />

being effective in helping control infectious complications<br />

in leucopenic patients <strong>and</strong> secondly, that such transfusions<br />

<strong>are</strong> demonstrated to be safe.<br />

OP02 Presence <strong>and</strong> future of treatment of acute<br />

myeloid leukemia in patients under the age<br />

of 60<br />

Roel Willemze 1 , Stefan Suciu S, Giovanna Meloni, Boris<br />

Labar, Halkes S, Petra Muus, David Selleslag D, Marko<br />

Vignetti, Sergio Amadori, Theo de Witte T, Jean-Pierre<br />

Marie, on behalf of the EORTC <strong>and</strong> GIMEMA Leukemia<br />

Groups, 1 Department of Hematology, Leiden University<br />

Medical Center, the Netherl<strong>and</strong>s<br />

The age of the patient still determines the choice of treatment<br />

<strong>and</strong> outcome in acute myeloid leukemia (AML). In<br />

patients, younger than 60 years, 65% to 80% enter complete<br />

remission after several anthracycline-cytarabine-based combinations.<br />

Dosing of these drugs influences the prognosis of<br />

17


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

the patients. Intermediate high dose cytarabine is recently<br />

reported to lead to the same results as st<strong>and</strong>ard dose cytarabine,<br />

while high dose cytarabine may improve results comp<strong>are</strong>d<br />

to st<strong>and</strong>ard dose cytarabine. A similar phenomenon is<br />

true for different kinds of anthracyclines or dosages of anthracyclines.<br />

Although much is still unclear, it still looks<br />

like more antileukemic drug, in addition to improvements in<br />

supportive c<strong>are</strong> (new antibiotics <strong>and</strong> bloodbank support),<br />

leads to a better outcome. One consolidation course consisting<br />

of high-dose cytarabine is considered necessary although<br />

firm proof is lacking. The intensity of the induction<br />

schedule appears to influence the duration of the hematological<br />

recovery phase <strong>and</strong> the number of autologous stem<br />

cell that can be harvested after consolidation, <strong>and</strong> the relapse<br />

risk after this consolidation course. These findings<br />

may indicate that in certain patients the sensitivity of normal<br />

hematopoietic stem cells for certain antileukemic drugs<br />

parallels the sensitivity of leukemic stem cells. Further consolidation<br />

of the remission status using repeated high-dose<br />

cytarabine courses or a stem cell transplantation decreases<br />

relapse rate <strong>and</strong> increases remission duration, allogeneic<br />

stem cell transplantation in the younger patients being superior<br />

to autologous stem cell transplantation or high-dose<br />

cytarabine. Although the administration of IL-2 after autologous<br />

stem cell transplantation or intensive consolidation<br />

therapy did not affect the duration of remission in most<br />

studies, the combination of low-dose of interleukin-2 <strong>and</strong><br />

histamine as maintenance prolonged remission in one study.<br />

Outcome improved by allogeneic stem cell transplantation<br />

using sibling donors, especially in patients under the age of<br />

50 with intermediate or bad risk AML. Currently, due to<br />

improved HLA-matching techniques, the outcome of allogeneic<br />

stem cell transplantation using matched unrelated<br />

donors equals the results obtained by sibling donors. Adoptive<br />

cellular immunotherapy after allogeneic stem cell transplantation<br />

using donor lymphocytes or in vitro cultured T<br />

cells <strong>are</strong> under investigation but preliminary results look<br />

promising with respect to the control of molecular or cytogenetic<br />

relapses. Acute myeloid leukemia appears to be a<br />

high heterogenous disease where an increasing number of<br />

cytogenetic <strong>and</strong> molecular aberrations of the leukemic cells<br />

determines the prognosis. These differences led already to a<br />

subdivision of AML in good, intermediate <strong>and</strong> bad or very<br />

bad risk subgroups where different treatment strategies need<br />

to be applied. However some molecular aberrations may be<br />

present across these prognostic subtypes, complicating the<br />

study of the value of drugs specifically designed against<br />

these aberrations. Relatively few new drugs have been approved<br />

for AML in the last two decades. Gemtuzumab ozogamicin<br />

is the first example of antibody directed chemotherapy<br />

targeting the CD33 epitope. Its use in combination<br />

with chemotherapy has been denied in Europe due to the<br />

lack of benefit in overall survival. Studies targeting the<br />

FLT3 mutation have shown limited activity as monotherapy<br />

but r<strong>and</strong>omized trials in combination <strong>are</strong> underway. Targeting<br />

RAS <strong>and</strong> other farnesylated peptides <strong>and</strong> P-glycoprotein<br />

has been disappointing. Demethylating agents such as<br />

decitabine, have shown superior antileukemic activity in the<br />

past, appear useful in the management of patients with myelodysplasia,<br />

<strong>and</strong> <strong>are</strong> currently reintroduced in the treatment<br />

18<br />

of AML. A new agent, clofarabine, shows increased hematological<br />

toxicity <strong>and</strong> a high complete remission rate at the<br />

maximum tolerated dose in phase I/II trials but requires r<strong>and</strong>omized<br />

evidence to evaluate its real impact on duration of<br />

remission <strong>and</strong> overall survival.<br />

In conclusion, the outcome of AML under the age of 60<br />

has improved considerably during the last 25 years due to<br />

the use of more intensive chemotherapy <strong>and</strong> stem cell transplantation<br />

<strong>and</strong> improvements in supportive c<strong>are</strong>. Currently<br />

<strong>and</strong> in the next future the improved knowledge on tumor<br />

immunology <strong>and</strong> on molecular aberrations of leukemic cells<br />

will lead to an extreme subdivision of AML in subgroups<br />

with specific treatment requirements. In this personalised<br />

treatment era, patients available to enter into a phase III trial<br />

will become quite limited. This development necessitates<br />

worldwide cooperation of Leukemia Groups in order to collect<br />

sufficient patients of a specific subgroup of AML.<br />

OP03 CLL: how close is the cure?<br />

Michael Keating, The University of Texas M. D. Anderson<br />

Cancer Center, Houston, Texas, USA<br />

Dramatic changes have occurred in the outcome of patients<br />

with chronic lymphocytic leukemia (CLL) over the<br />

last 25 years. Using modern day criteria for response, the<br />

complete remission (CR) rate with alkylating agent regimens<br />

was approximately 5%. As fludarabine entered into<br />

frontline treatment, the CR rate increased to 20–25%, going<br />

to 30–35% for fludarabine plus cyclophsomide (F+C), <strong>and</strong><br />

more recently with the FCR regimen, adding rituximab to<br />

FC, the CR rate is 50–70%. An important part of the development<br />

of evaluation criteria were provided by the NCI<br />

working group guidelines 1 <strong>and</strong> more recently the iwCLL<br />

guidelines 2 for indications for treatment <strong>and</strong> response evaluation<br />

in CLL. In most tumors when the complete remission<br />

rate gets above 50% there is a fraction of patients who <strong>are</strong><br />

cured of their disease. This question is increasingly being<br />

asked in chronic lymphocytic leukemia.<br />

It is clear that there is a close correlation of the time to<br />

progression (TTP) of complete <strong>and</strong> partial responders (CR<br />

+ PR) according to the quality of the remission <strong>and</strong> other<br />

parameters such as the mutation status <strong>and</strong> FISH genetics.<br />

Patients who achieve CR using NCI WG criteria have a significantly<br />

longer time to progression than nodular partial<br />

remissions (NPR) who again have a longer remission duration<br />

than other partial responders. An analysis of 300 patients<br />

treated with the FCR regimen (FCR300) previously<br />

published 3 has demonstrated that there is a clear association<br />

of time to progression with the mutation status of the immunoglobulin<br />

variable sequence of the heavy chain (IgVH) 4 .<br />

The mutated patients have a very significantly greater likelihood<br />

of being 5 <strong>and</strong> 10 years in remission than patients who<br />

<strong>are</strong> unmutated. There is also evidence that patients that<br />

achieve minimal residual disease (MRD) negativity have a<br />

greater probability of being long term remissions. The technology<br />

for measuring MRD has evolved over time so that<br />

the more recent strict criteria of flow cytometry identifying<br />

1 in 10 4 cells has only recently been applied. Most of these


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

clinical trials evaluating FCR regimens or other regimens<br />

combining purine analogs with monoclonal antibodies such<br />

as rituximab <strong>are</strong> too immature to evaluate true long term<br />

remissions.<br />

We now have complete follow-up of patients treated with<br />

FCR (300). The percent of patients who <strong>are</strong> free of recurrence<br />

at 10 years <strong>are</strong> 46% for CR, 26% for NPR <strong>and</strong> 17%<br />

for other PR.<br />

The FISH evaluation was not available for the FCR 300<br />

patients <strong>and</strong> we cannot make such projections. However, it<br />

is clear that more recent CR + PR patients who <strong>are</strong> mutated<br />

have a 61% likelihood of being free of relapse at seven years<br />

but only 18% if unmutated. Within the unmated group of<br />

patients it is app<strong>are</strong>nt that shorter times to remission occur<br />

in patients with deletion of chromosome 11q.22-23 (del<br />

11q). Older patients have not f<strong>are</strong>d as well, but in the small<br />

proportion of patients 70 years of age or older who were CR<br />

or PR in the first FCR study, 36% <strong>are</strong> still alive. 53% have<br />

relapsed <strong>and</strong> 17% have died in remission. A substantial<br />

number of patients <strong>are</strong> dying of other illnesses while still in<br />

remission of their disease which is functionally a cure of<br />

their disorder.<br />

Now that subsets of patients who <strong>are</strong> at high risk of relapse<br />

can be more readily identified, post remission strategies<br />

with monoclonal antibodies, lenalidomide <strong>and</strong> stem<br />

cell transplantation <strong>are</strong> being applied. The prospects for<br />

long term survival of patients with 17p deletion or p53 mutation<br />

is still grim with stem cell transplantation the only<br />

reasonable option to give long term control. The 11q group<br />

of patients have a very high propensity of relapse, but they<br />

have a good second remission rate so that opportunities <strong>are</strong><br />

present to prolong remission by intervention during their<br />

first <strong>and</strong> later remissions. All other subsets including trisomy<br />

chromosome 12, 13q deletion <strong>and</strong> negative patients have<br />

the same likelihood of being free of disease. With the development<br />

of new therapies such as the B cell Receptor signaling<br />

antagonists such as CAL-101 <strong>and</strong> PCI-32765, improved<br />

transplant options for patients up to 75 years of age <strong>and</strong> immune<br />

modulating agents such as lenalidomide, prospects<br />

for all subsets of patients is improving.<br />

R E F E R E N C E S<br />

1. Cheson BD, Bennett JM, Grever M, Kay N, Keating MJ, O’Brien S, Rai<br />

KR. National Cancer Institute-sponsored Working Group guidelines for<br />

chronic lymphocytic leukemia: revised guidelines for diagnosis <strong>and</strong><br />

treatment. Blood 1996;87(12):4990–7.<br />

2. Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G,<br />

Döhner H, Hillmen P, Keating MJ, Montserrat E, Rai KR, Kipps TJ.<br />

Guidelines for the diagnosis <strong>and</strong> treatment of chronic lymphocytic leukemia:<br />

a report from the International Workshop on Chronic Lymphocytic<br />

Leukemia updating the National Cancer Institute-Working Group<br />

1996 guidelines. Blood 2008;111(12):5446–56.<br />

3. Tam CS, O’Brien S, Wierda W, Kantarjian H, Wen S, Do KA, Thomas<br />

DA, Cortes J, Lerner S, Keating MJ. Long-term results of the fludarabine,<br />

cyclophosphamide, <strong>and</strong> rituximab regimen as initial therapy of<br />

chronic lymphocytic leukemia. Blood 2008;112(4):975–80.<br />

4. Lin KI, Tam CS, Keating MJ, Wierda WG, O’Brien S, Lerner S,<br />

Coombes KR, Schlette E, Ferrajoli A, Barron LL, Kipps TJ, Rassenti L,<br />

Faderl S, Kantarjian H, Abruzzo LV. Relevance of the immunoglobulin<br />

VH somatic mutation status in patients with chronic lymphocytic leukemia<br />

treated with fludarabine, cyclophosphamide, <strong>and</strong> rituximab<br />

(FCR) or related chemoimmunotherapy regimens. Blood 2009;113(14):<br />

3168–71.<br />

OP04 From donor buffy coat to vaccination<br />

Hans-Jochem Kolb 1 , Helga Schmetzer 2 , Arndt Borckhardt 3 .<br />

1 3. Med. Klinik, Techn. Universitaet Muenchen,<br />

1 Helmholtz Zentrum München, National Reserch Center<br />

for Environmental Health, 2 3. Med. Klinik, Universitaet<br />

München, Germany, 3 Kinderklinik Univ. Düsseldorf,<br />

Germany<br />

Chimerism <strong>and</strong> transplantation tolerance can be achieved<br />

with allogeneic stem cell transplantation. The state of chimerism<br />

provides the chance for adoptive immunotherapy<br />

using donor lymphocytes exerting strong immune effects on<br />

leukemia <strong>and</strong> other malignancies of the host without being<br />

rejected. The risks of allogeneic lymphocytes <strong>are</strong> development<br />

of graft-versus-host disease (GVHD) as well as tolerance<br />

toward the malignant disease. Delay of donor lymphocyte<br />

transfusions after cessation of the cytokine storm of<br />

conditioning treatment <strong>and</strong> establishment of some form of<br />

peripheral tolerance <strong>and</strong> c<strong>are</strong>ful treatment in escalating doses<br />

starting at a low dose has facilitated the treatment of relapse<br />

of chronic myeloid leukemia <strong>and</strong> prevention of acute<br />

myeloid leukemia without severe GVHD. However, preemptive<br />

transfusion of donor lymphocytes has been limited<br />

to a minority of patients because of early relapses <strong>and</strong> acute<br />

GVHD. The hypothesis that myeloid leukemia respond better<br />

to donor lymphocytes because of their differentiation to<br />

dendritic cells has been supported by the demonstration of<br />

dendritic cells with the karyotype of the leukemia <strong>and</strong> the<br />

improved response following treatment with interferon-a<br />

<strong>and</strong> GM-CSF. However in the case of rapidly progressive<br />

relapse cytotoxic therapy may be necessary <strong>and</strong> subsequently<br />

donor lymphocytes may produce GVHD. Therefore we<br />

<strong>and</strong> others have studied lymphocytes of treated patients for<br />

the recognition of antigens that <strong>are</strong> overexpressed in leukemia<br />

blasts like WT-1, PR-1 <strong>and</strong> PRAME. We found overexpression<br />

of these proteins in acute myeloid leukemia. Moreover<br />

we found 4 new antigens overexpressed inAML coded<br />

by the Y-chromosome. Unfortunately the number of CD8cells<br />

with multimer staining for peptides of these antigens<br />

were few <strong>and</strong> gamma-interferon release low. We also studied<br />

splice variants of a Y-chromosome associated antigen<br />

UTY for tissue restriction. We could demonstrate a multitude<br />

of splice variants, but no specific tissue restriction.<br />

These findings <strong>are</strong> in contrast to the selection of T cells using<br />

antigenic peptides of EBV covering more than 90% of<br />

HLA-antigens; these cells could be selected <strong>and</strong> transfused<br />

inducing rapid remissions in patients with EBV-associated<br />

post-transplant lymphoma. This has been proven as a life<br />

saving cost effective procedure that would be ideal for the<br />

treatment of leukemia, if an antigen of similar strength<br />

could be defined for leukemia. Genome wide screening for<br />

SNPs using T-cells of stem cell transplant patients has<br />

shown a few new minor histocompatibility antigens. The<br />

cumulative frequency of genes for minor H-antigens may<br />

thus increase <strong>and</strong> more patients could benefit of immunotherapy<br />

directed against mHA. We have tried to test the generation<br />

of T cells in vitro against dendritic cells of leukemic<br />

origin, <strong>and</strong> we found two results: first patients with blasts<br />

differentiating into DC have a better chance to respond to<br />

19


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

door lymphocytes, secondly cytotoxic T cells generated to<br />

dendritic cells of leukemia origin in vitro have T cell receptors<br />

( Vß-chain) found in the patients post transplantation.<br />

Several Vß chain clones <strong>are</strong> represented in more than 1 sample;<br />

they may represent dominant clones that can be recognized<br />

in vitro <strong>and</strong> sometimes even in the donor. There is a<br />

good chance that they can be selected <strong>and</strong> used for immunotherapy<br />

without further analysis of the antigens against<br />

which they <strong>are</strong> directed.<br />

Major histocompatibility antigen may cause more vigorous<br />

reactions including early cellular reactions <strong>and</strong> antibody<br />

production. Of particular interest is NK reactivity against<br />

hematopoietic cells missing own HLA-antigens, transplantation<br />

across cross-reactive HLA-C groups sharing killer<br />

inhibitor receptors have a strong graft-versus-leukemia effect<br />

in acute leukemia, not in lymphoma. Maternal transplants<br />

have a superior GVL effect to paternal grafts indicating<br />

immune memory against paternal antigens. For alternate<br />

donor transplants we prefer HLA-haploidentical transplants<br />

for younger patients <strong>and</strong> cord blood transplants for elderly<br />

patients. Double cord transplants have a strong GVL activity<br />

for as yet unknown reasons. Non-inherited maternal antigens<br />

<strong>and</strong> graft-versus-graft reactions may play a role. Major<br />

histocompatibility antigens may not only be a strong<br />

target for alloimmune reactions, they may as well present<br />

peptide of autologous antigens in an immunogenic way <strong>and</strong><br />

provide a major activating stimulus to reactions against minor<br />

HA. Several recent observations support the necessity<br />

of strong costimulation in order to prevent tolerance toward<br />

leukemia-associated antigens <strong>and</strong> minor HA. Repeat vaccination<br />

with WT1 <strong>and</strong> PR1 peptides induced non-reactivity<br />

(Rezvani et al. 2011) even in the presence of memory T<br />

cells (Pospori et al, 2011). We discuss mechanisms of immune<br />

escape in leukemia <strong>and</strong> possible ways to overcome it<br />

with interferon-a, GM-CSF <strong>and</strong> bispecific antibody treatment.<br />

Future donors have not only to selected according to<br />

histocompatibility, but also according to their immune repertoire.<br />

In future immunotherapy not only target antigens<br />

<strong>and</strong> effector cells have to be considered, but also immune<br />

escape <strong>and</strong> mechanisms to maintain immune tolerance.<br />

OP05 AML in adults – what we need for diagnosis<br />

Torsten Haferlach, MLL Munich Leukemia Laboratory,<br />

Munich, Germany<br />

The diagnosis of AML is getting more <strong>and</strong> more comprehensive<br />

but also complicated. First of all, making the diagnosis<br />

does not always mean to classify AML: according to<br />

the WHO classification as published in 2008 1 , several hierarchical<br />

steps have to be fulfilled:<br />

To make it easier for application, one could first discriminate<br />

between »treatment-related AML« following radiation<br />

<strong>and</strong>/or chemotherapy <strong>and</strong> »de novo AML«. Thus, the history<br />

of the patient has to be known to the physician but also to<br />

the diagnostic laboratory.<br />

As a second step, »AML with recurrent cytogenetic aberrations«<br />

<strong>are</strong> defined by the WHO. This for sure needs chromosomal<br />

b<strong>and</strong>ing analysis, even if in some cases molecular<br />

techniques such as PCR screening may be helpful. But not<br />

20<br />

all entities subcategorized in this »AML with recurrent genetic<br />

abnormalities« can be detected by PCR, as such st<strong>and</strong>ard<br />

chromosomal b<strong>and</strong>ing analysis accompanied by FISH<br />

<strong>and</strong>/or PCR <strong>are</strong> necessary. As a matter of fact, these techniques<br />

have a turn around time of three to ten days in experienced<br />

laboratories. Therefore, the diagnosis cannot rely<br />

only <strong>and</strong> primarily on this classification aspects, chromosomal<br />

b<strong>and</strong>ing analysis.<br />

In the next step of the hierarchy, »AML with myelodysplasia-related<br />

changes« have to be taken into account.<br />

Again, these cases <strong>are</strong> based on their respective history of<br />

the patient (AML following MDS or MDS/MPN overlap)<br />

but also include in addition or only cytogenetic changes,<br />

mostly known from MDS, <strong>and</strong>/or dysplastic features, socalled<br />

multilineage dysplasia. Also in this category, as it is a<br />

combination of different methods, turn around time <strong>and</strong><br />

availability make it not easy to get this category implemented<br />

into a daily routine diagnostic workflow.<br />

Finally, those cases not fulfilling the above-mentioned<br />

criteria <strong>are</strong> classified according to the formally known morphologic<br />

criteria as defined in the FAB classification. In<br />

these cases, at least cytomorphology is important.<br />

In addition, several other aspects <strong>are</strong> included today more<br />

<strong>and</strong> more in st<strong>and</strong>ard approaches to AML diagnostics: molecular<br />

markers such as NPM1 <strong>and</strong> CEBPA have been suggested<br />

to be »provisional entities« according to WHO classification,<br />

especially within a normal karyotype, other genes<br />

should be investigated as well such as FLT3-ITD, MLL-<br />

PTD <strong>and</strong> newer markers my also be implemented in the near<br />

future such as DNMT3A. In addition, the expression of several<br />

genes such as EVI1, WT1 <strong>and</strong> BAALC have to be taken<br />

into account, which makes it even more difficult to follow<br />

up with the complete WHO scenario for diagnosis of AML<br />

in a routine setting 2 .<br />

Therefore, several guidelines have been suggested or different<br />

scenarios, the most important one was published by a<br />

cooperating initiative of the ELN 3 . The authors clearly elaborate<br />

on the specific diagnostic algorithms in AML in adults,<br />

discriminating between st<strong>and</strong>ard diagnostic approaches in<br />

general <strong>and</strong> clinical studies:<br />

From clinical perspective it seems m<strong>and</strong>atory that for patients<br />

who may undergo st<strong>and</strong>ard treatment approaches,<br />

cytomorphology (leading to the diagnosis at the same day)<br />

accompanied by immunophenotyping (same turn around<br />

time) is m<strong>and</strong>atory. This has to be performed together with<br />

st<strong>and</strong>ard cytogenetic analysis, as cytogenetic results still<br />

provide the most important prognostic factor in AML 4 . In<br />

addition, cytogenetic results <strong>are</strong> m<strong>and</strong>atory to fulfil baseline<br />

diagnostic criteria according to the WHO classification<br />

2008.<br />

If allogeneic transplantation may be an option, also the<br />

investigation of molecular markers such as NPM1, FLT3-<br />

ITD <strong>and</strong> CEBPA, especially in the normal karyotype AML<br />

subset, seems important 5 . It further has to be discussed that<br />

minimal residual disease studies in AML with molecular<br />

markers such as PML-RARA, CBFB/MYH11 but also NPM1<br />

may help to further subclassify <strong>and</strong> guide treatment in the<br />

individual AML patient.<br />

The next future will also demonstrate that new molecular<br />

markers such as DNMT3A 6 , TET2 <strong>and</strong> others may not only


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

add important diagnostic value for AML but also will guide<br />

treatment <strong>and</strong> define prognosis. However, it has to be admitted<br />

that the diagnosis of AML is still getting more complicated,<br />

labour intensive <strong>and</strong> expensive. The next few years<br />

will demonstrate, if techniques such as next-generation sequencing<br />

(NGS) will help us to better characterize AML<br />

patients not only in clinical studies but also in the daily routine<br />

setting.<br />

We have to realize that many patients still die with the<br />

diagnosis of AML <strong>and</strong> it is highly appreciated that any new<br />

diagnostic <strong>and</strong> therapeutic approach has be followed to be<br />

investigated within clinical studies <strong>and</strong> to be implemented<br />

for all patients after validation in a short timeframe.<br />

R E F E R E N C E S<br />

1. WHO Classification of Tumours of Haematopoietic <strong>and</strong> Lymphoid Tissues.<br />

Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H<br />

et al., editors. 4th. 2008. Lyon, International Agency for Research on<br />

Cancer (IARC).<br />

2. Marcucci G, Haferlach T, Döhner H. Molecular genetics of adult acute<br />

myeloid leukemia: prognostic <strong>and</strong> therapeutic implications. J Clin Oncol<br />

2011;29:475–486.<br />

3. Döhner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett<br />

AK, et al. Diagnosis <strong>and</strong> management of acute myeloid leukemia in<br />

adults: recommendations from an international expert panel, on behalf<br />

of the European LeukemiaNet. Blood 2010;115:453–474.<br />

4. Grimwade D, Hills RK, Moorman AV, Walker H, Chatters S, Goldstone<br />

AH, et al. Refinement of cytogenetic classification in acute myeloid leukemia:<br />

determination of prognostic significance of r<strong>are</strong> recurring chromosomal<br />

abnormalities among 5876 younger adult patients treated in<br />

the United Kingdom Medical Research Council trials. Blood 2010;116:<br />

354–365.<br />

5. Schlenk RF, Dohner K, Krauter J, Frohling S, Corbacioglu A, Bullinger<br />

L, et al. Mutations <strong>and</strong> treatment outcome in cytogenetically normal<br />

acute myeloid leukemia. N Engl J Med 2008;358:1909–1918.<br />

6. Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, et<br />

al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med 2010;<br />

363:2424–2433.<br />

OP06 Mutation of epigenetic regulators in AML<br />

<strong>and</strong> MDS<br />

Joop Jansen, Laboratory of Hematology, Radboud<br />

University Nijmegen Medical Centre St. Radboud <strong>and</strong><br />

Nijmegen Centre for Molecular Life Sciences, Nijmegen,<br />

The Netherl<strong>and</strong>s<br />

Aberrant epigenetic modifications have been described in<br />

many types of cancer. In myeloid malignancies aberrant<br />

methylation patterns may be observed, <strong>and</strong> it has been proposed<br />

that repression of genes that <strong>are</strong> crucial for the cessation<br />

of the cell cycle <strong>and</strong> the induction of differentiation<br />

may contribute to the malignant transformation of normal<br />

hematopoietic cells. In the past years, various genes were<br />

shown to be mutated in AML <strong>and</strong> MDS, <strong>and</strong> this has been<br />

accelerated by the application of novel technologies like<br />

high-resolution SNP-array analysis <strong>and</strong> next generation sequencing.<br />

Strikingly, several of the newly identified genes<br />

(ASXL1, UTX, TET2, EZH2, DNMT3A) function to regulate<br />

gene expression by epigenetically changing chromatin. The<br />

TET2 gene is currently the most frequently mutated gene in<br />

MDS known so far (20–25% of the cases), <strong>and</strong> is also mutated<br />

in AML (10% of the cases) <strong>and</strong> various types of MPN<br />

(including CMML, where it is mutated in 30–40% of the<br />

cases). It was shown to convert methylated cytosines (correlating<br />

with repression of gene expression) into 5-hydroxymethyl<br />

cytosine, of which the functional consequences <strong>are</strong><br />

under investigation. Mutations of the DNA-methyltransferase<br />

3A gene (DNMT3A) were found both in AML <strong>and</strong> in<br />

MDS, which also provides a direct link between gene mutations<br />

<strong>and</strong> DNA methylation. Furthermore, mutations in<br />

genes that regulate the epigenetic modifications of histones<br />

have been found, such as the polycomb-repressor complex-<br />

2 (PRC2) component EZH2. It will be interesting to see<br />

whether the patients who carry mutations in the genes that<br />

regulate epigenetic chromatin changes <strong>are</strong> responding differently<br />

to the novel epigenetic therapies (like hypomethylating<br />

agents) that <strong>are</strong> currently being investigated.<br />

OP07 Therapy for younger adults with AML<br />

Alan Burnett, Department of Haematology, Cardiff<br />

University School of Medicine, Heath Park, Cardiff,<br />

United Kingdom<br />

Survival in younger patients (arbitrarily defined as patients<br />

less than 60 years) has improved in the last 20 years<br />

such that 45 to 50% can now be cured, but may have reached<br />

the limit of the potential with »st<strong>and</strong>ard chemotherapy.<br />

St<strong>and</strong>ard of c<strong>are</strong> comprises 7+3 Ara-C + Daunorubicin for<br />

induction followed by high dose Ara-C (3 g/m 2 ) for consolidation<br />

or allogeneic transplant for non-favourable cases<br />

for who a donor is available. However higher remission<br />

rates have been consistently shown with alternative inductions<br />

such as a 10 day Ara-C schedule, but intensification of<br />

Ara-c in induction has not been consistently of benefit. Similarly<br />

comparisons of different anthracylcine or anthracyline-like<br />

agents have not shown consistent superiority over<br />

Daunorubicin once dose equivalence has been taken into<br />

account. Recently a Daunorubicin dose of 90 mg/m 2 has<br />

been shown to have improved induction <strong>and</strong> in some cases<br />

survival. However the comparator <strong>are</strong> only produced a 57%<br />

CR rate. However Idarubicin is equivalent to Daunorubicin<br />

80 mg/m 2 . Many consider that a 60 mg/m 2 the optimum<br />

dose. The addition of mylotarg to a Daunorubicin dose of<br />

45 mg was equivalent to a 60 mg dose, however in the largest<br />

trial testing the addition of mylotarg to a 3+10 schedule<br />

significantly improved survival by 10% in 70% of patients<br />

with high risk disease. Inclusion of other nucleoside analogues<br />

such as Fludarabine/ Cladrabine or clofarabine has<br />

potential to be more effective induction.<br />

High dose Ara-C is st<strong>and</strong>ard of c<strong>are</strong> in consolidation but<br />

what dose <strong>and</strong> how many courses is still under study. The<br />

MRC AML15 trial comp<strong>are</strong>d 3 g vs a 1.5 g dose <strong>and</strong> found<br />

no overall difference. Three consolidation courses was not<br />

superior to two. In this trial, two courses of FLAG-Ida in<br />

induction did not improve overall survival, although it significantly<br />

reduced the risk of relapse, but because of more<br />

myelosuppression there was less compliance with consolidation<br />

therapy. However the survival of patients who received<br />

2 courses of FLAG-Ida only was the same as for patients<br />

who received all planned four courses. The survival<br />

21


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

for patients who received FLAG-Ida X 2 <strong>and</strong> Ara-C consolidation<br />

X 2 was 64%.<br />

Stem cell transplantation is clearly superior to chemotherapy<br />

in high risk disease, although far from satisfactory<br />

for this difficult subgroup in which there has been no improvement<br />

in the last 30 years.There is continuing controversy<br />

concerning which intermediate risk patients should<br />

have SCT in CR1. This is usually initially defined by cytogenetics,<br />

however of other important prognostic factors <strong>are</strong><br />

used to identify further high risk groups (inadequate marrow<br />

response to course 1/secondary disease/ high white<br />

count/ male gender) the remaining patients may not need<br />

transplantation. Younger patients ( 10 x 10 9 /L) seem<br />

to benefit from treatments including cytarabine in the ATRA<br />

plus chemotherapy scheme, whereas st<strong>and</strong>ard risk patients<br />

can be successfully managed with less intensive regimens<br />

containing ATRA <strong>and</strong> anthracycline-based chemotherapy.<br />

More recently, arsenic trioxide (ATO), which as a single<br />

agent is the most effective drug for APL, has unquestionably<br />

emerged as the most exciting »new drug« to be included<br />

in the treatment strategy for this disease. Following the<br />

outst<strong>and</strong>ing results with ATO in the treatment of APL relapse,<br />

several clinical trials have been reported by single<br />

institutions in relatively small <strong>and</strong> non-comparative studies.<br />

Although published studies <strong>are</strong> lacking, there <strong>are</strong> currently<br />

at least two ongoing r<strong>and</strong>omized trials comparing ATObased<br />

versus chemotherapy-based approaches. One conducted<br />

by the Italian GIMEMA group for patients with WBC<br />

count < 10 x 10 9 /L <strong>and</strong> another by the British NCRI for all<br />

patients with APL Further to explore the role of ATO in<br />

front-line therapy with the aim of minimizing or even eliminating<br />

the use of chemotherapy, other clinical trials have<br />

been designed to assess the use of ATO to reinforce st<strong>and</strong>ard<br />

ATRA plus chemotherapy regimens <strong>and</strong> further improve the<br />

outcome of APL. In this regard, a recent report of a large<br />

r<strong>and</strong>omized study by the US Intergroup showed significantly<br />

better outcomes in patients receiving two courses of ATO<br />

immediately after achieving complete remission <strong>and</strong> before<br />

the st<strong>and</strong>ard consolidation with ATRA <strong>and</strong> chemotherapy.<br />

Currently other major cooperative groups, such as the<br />

French-Belgian-Swiss group <strong>and</strong> the PETHEMA-HOVON<br />

group, <strong>are</strong> also conducting studies to assess the role of ATO<br />

in consolidation therapy of ATRA plus chemotherapy regimens.<br />

The opportunities offered by the incorporation of<br />

ATO into the therapeutic armamentarium of APL, not only<br />

to minimize or even eliminate the use of chemotherapy, but<br />

also to optimize the st<strong>and</strong>ard approach, will be discussed in<br />

this presentation.<br />

OP10 FLT3 inhibitors – too much expectations?!<br />

Farhad Rav<strong>and</strong>i, University of Texas – MD Anderson<br />

Cancer Center, Houston, Texas, USA [see SA01]<br />

The FLT3 receptor kinase is highly expressed in acute<br />

leukemias. FLT3 gene mutations <strong>are</strong> among the most com-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

mon molecular abnormalities in AML occurring in about a<br />

third of patients particularly those with diploid cytogenetics<br />

<strong>and</strong> acute promyelocytic leukemia. Available evidence suggests<br />

that these molecular lesions confer a shorter diseasefree<br />

survival <strong>and</strong> overall survival in patients with intermediate-risk<br />

cytogenetics. As a result, there is substantial interest<br />

in inhibition of the FLT3 kibase as a therapeutic target <strong>and</strong><br />

several promising inhibitors that target this tyrosine kinase<br />

<strong>are</strong> in clinical development.<br />

Several reported trials of these kinase inhibitors as a single<br />

agent <strong>and</strong> incombination with traditional cytotoxic<br />

agents have clearly demonstrated their activity. However,<br />

future trials involving perhaps more potent <strong>are</strong> likely to shed<br />

further insights on the relevance of FLT3 kinase as a target,<br />

the population most likely to derive benefit, the most appropriate<br />

agents for combination, <strong>and</strong> the clinical <strong>and</strong> biological<br />

correlates predicting a benefit.<br />

OP11 Allogeneic SCT for AML – when to treat<br />

OP12 Allogeneic SCT with reduced intensity<br />

conditioning for AML<br />

Didier Blaise, Unité de Transplantation et de Thérapie<br />

Cellulaire (UTTC), département d’hématologie, Institut<br />

Paoli-Calmettes, Marseille <strong>and</strong> Université de la<br />

Méditerranée, Marseille, France.<br />

Allogeneic stem cell transplantation (allo-SCT) is the<br />

treatment exerting the most efficient antileukemic effect in<br />

acute myeloblastic leukemia (AML). In selected subset of<br />

patients, this conducts into a survival benefit as comp<strong>are</strong>d to<br />

chemotherapy strategies. However, the oldest patients can<br />

r<strong>are</strong>ly benefit from intensive treatment including st<strong>and</strong>ard<br />

myeloablative allo-SCT because of an unacceptable high<br />

risk of procedure-related toxicity. This point is especially<br />

critical when considering AML patients in first CR (CR1).<br />

Finally, most of the patients lack a human lymphocyte antigen<br />

(HLA)-identical donor further precluding an allo-SCT<br />

strategy.<br />

The development of the so-called non-ablative, or reduced-intensity<br />

conditioning (RIC) regimens appears to decrease<br />

allo-SCT-related toxicities, as promising results have<br />

already been reported in different series of patients with<br />

various hematological malignancies. In contrast to st<strong>and</strong>ard-dosed<br />

myeloablative allo-SCT, <strong>and</strong> although long term<br />

follow-up is still needed in many studies, RIC allo-SCT is<br />

relatively well tolerated by patients with high-risk clinical<br />

features such as advanced age or associated co-morbidities.<br />

Nevertheless, toxicity might represent only one face of the<br />

problem, since AML encompasses a group of chemo-sensitive<br />

diseases, raising concerns that significant reduction of<br />

the intensity of the st<strong>and</strong>ard conditioning regimens may<br />

negatively impact long-term leukemic control. This concern<br />

might be particularly app<strong>are</strong>nt in patients with high-risk<br />

leukemic features. Indeed, the importance of dose intensity<br />

has been already shown in myeloablative allo-SCT. However,<br />

the beneficial effect of more intensive conditioning<br />

that is associated with a reduced risk for relapse was offset<br />

by an increased transplant-related toxicity. The latter figures<br />

may be even more complex since the relative benefit of myeloablation<br />

as part of the conditioning regimen, may also<br />

depend on patient <strong>and</strong> disease status at time of allo-SCT<br />

(e.g. CR1 vs. beyond CR1 or advanced disease). Thus, investigators<br />

<strong>are</strong> currently faced with a dilemma on how to<br />

optimize the potential role of RIC allo-SCT in patients with<br />

AML, while delivering minimal myeloablation <strong>and</strong> maximizing<br />

allogeneic immunotherapy. To add to this debate we<br />

will review present knowledge on reduced intensity regimen<br />

approach in AML <strong>and</strong> report our experience.<br />

OP13 Novel drugs for AML<br />

Hagop Kantarjian, Department of Leukemia, U.T. M.D.<br />

Anderson Cancer Center, Houston, TX, USA [see SA01]<br />

OP14 Molecular Pathogenesis of Myelodysplastic<br />

Syndromes<br />

Omar Abdel-Wahab, Alan Shih, Ross L. Levine, Leukemia<br />

Service, Department of Medicine, Human Oncology <strong>and</strong><br />

Pathogenesis Program, Memorial Sloan Kettering Cancer<br />

Center<br />

Myelodysplastic syndromes represent one of the most<br />

common myeloid malignancies, <strong>and</strong> <strong>are</strong> associated with poor<br />

outcome overall due to progressive cytopenias/bone marrow<br />

failure <strong>and</strong> progression to acute myeloid leukemia. Importantly,<br />

clinical <strong>and</strong> cytogenetic parameters have been<br />

used to improve prognostication for MDS patients, however<br />

little insight into the molecular pathogenesis of MDS<br />

had been elucidated until recently when a series of c<strong>and</strong>idate<br />

gene <strong>and</strong> whole genome studies have identified recurrent<br />

somatic mutations in MDS patients including TET2,<br />

ASXL1, DNMT3A, EZH2, <strong>and</strong> TP53 mutations. Importantly,<br />

a subset of these genes including ASXL1, EZH2, <strong>and</strong><br />

TP53 have been associated with poor outcome, <strong>and</strong> recent<br />

studies have shown that these mutations can be used to supplement<br />

the IPSS <strong>and</strong> improve prognostication in MDS.<br />

These data will be reviewed.<br />

In addition recent functional studies have begun to elucidate<br />

how these disease alleles contribute to MDS pathogenesis.<br />

Specifically, the TET family of proteins have been<br />

shown to place a hydroxyl mark on methylated DNA <strong>and</strong><br />

lead to DNA demethylation. We have shown that TET2 mutations<br />

leads to loss of DNA hydroxymethylation <strong>and</strong> a hypermethylation<br />

phenotype in leukemia patients. In addition,<br />

in vitro <strong>and</strong> in vivo studies show that TET2 loss leads to<br />

impaired hematopoietic differentiation, increased stem cell<br />

self-renewal, <strong>and</strong> myeloid transformation in vivo. These<br />

data will be presented in detail.<br />

Finally, more recent studies have revealed a role of mutations<br />

in chromatin modifying enzymes in MDS pathogenesis,<br />

including ASXL1 <strong>and</strong> EZH2 mutations. The effects of<br />

these mutations on chromatin state, gene expression, <strong>and</strong><br />

hematopoietic function have begun to be elucidated, sug-<br />

23


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

gesting mutations in chromatin modifying enzymes coopt<br />

the epigenetic state of hematopoietic stem/progenitor cells<br />

to contribute to transformation. Data from recent in vitro<br />

<strong>and</strong> in vivo studies including novel MDS models will be<br />

presented in detail.<br />

OP15 Risk models in MDS<br />

David Steensma, Dana-Farber Cancer Institute, Harvard<br />

Medical School, Boston, MA, USA<br />

In recent years, investigators have proposed several novel<br />

risk models for the purpose of forecasting the clinical course<br />

of patients with myelodysplastic syndromes (MDS). The<br />

most widely used MDS prognostic system in clinical practice<br />

continues to be the 4-strata 1997 International Prognostic<br />

Scoring System (IPSS), but clinicians have recognized<br />

for many years that the IPSS has a number of critical limitations.<br />

These limitations include unsatisfactory applicability<br />

of the IPSS to a variety of clinical situations such as secondary,<br />

therapy-related MDS or previously treated patients;<br />

lack of sensitivity to the degree of patients’ peripheral blood<br />

cytopenias, especially severe thrombocytopenia; excessive<br />

emphasis on elevated marrow blast proportion comp<strong>are</strong>d to<br />

high-risk cytogenetics; a restricted number of included<br />

karyotypes; <strong>and</strong> failure to account for either comorbid conditions,<br />

or newer biomarkers of demonstrated prognostic<br />

value such as the presence of high-grade marrow fibrosis or<br />

elevated serum ferritin level. Collectively, these limitations<br />

make the original IPSS a less accurate <strong>and</strong> helpful tool than<br />

it might otherwise be, <strong>and</strong> have prompted other investigators<br />

to derive <strong>and</strong> propose newer risk stratification tools.<br />

The 5-strata Word Health Organization (WHO) classification-based<br />

Prognostic Scoring System (WPSS), proposed<br />

by Malcovati <strong>and</strong> colleagues initially in 2007 <strong>and</strong> since<br />

modified to include marrow fibrosis, attempted to improve<br />

on the IPSS by including transfusion dependence as well as<br />

bringing the prognostic classification in line with the WHO<br />

Classification of Tumors of Haematopoietic <strong>and</strong> Lymphoid<br />

tissues as updated in 2001 <strong>and</strong> again in 2008. In 2008, Kantarjian<br />

<strong>and</strong> colleagues from the MD Anderson Cancer Center<br />

proposed a 4-strata generalized risk model for patients<br />

with MDS that is both sensitive to the degree of cytopenias<br />

<strong>and</strong> emphasizes the importance of high risk karyotype,<br />

which several analyses have suggested is a more powerful<br />

marker of a negative outcome than increased marrow myeloblast<br />

proportion. A review of 1,503 Mayo Clinic patients<br />

comparing the 1997 IPSS, the WPSS, <strong>and</strong> the MD Anderson<br />

Cancer Center general risk model found that while all 3<br />

prognostic systems were useful, the MD Anderson classification<br />

scoring system accurately classified the broadest<br />

range of patients <strong>and</strong> also achieved the greatest separation<br />

between lower risk <strong>and</strong> higher risk groups.<br />

In May 2011, Greenberg discussed a revised/updated version<br />

of the IPSS, IPSS-R, at the 11th MDS Foundation International<br />

Symposium in Edinburgh, Scotl<strong>and</strong>. The IPSS-<br />

R includes a much broader range of karyotypes than the<br />

initial IPSS (based on the German-Austrian consortium data<br />

24<br />

of Haase <strong>and</strong> colleagues), has 5 stratification groups instead<br />

of 4, <strong>and</strong> rebalances the relative risk of cytogenetic abnormalities<br />

comp<strong>are</strong>d to marrow blast proportion, as well as<br />

introducing a graduated system for cytopenias that allows at<br />

least some sensitivity to the degree of cytopenia rather than<br />

just the number of cytopenias. Work on extending <strong>and</strong> validating<br />

the IPSS-R is ongoing.<br />

In addition to inclusion of comorbidities <strong>and</strong> other patient<br />

features, new disease-associated biomarkers that might be<br />

incorporated in future versions of the IPSS include results<br />

of flow cytometric panels, array-based measurement of loss<br />

of heterozygosity <strong>and</strong> gene copy number, <strong>and</strong> assessment<br />

specific molecular abnormalities. The MDS-associated somatic<br />

gene mutation l<strong>and</strong>scape continues to exp<strong>and</strong>, <strong>and</strong><br />

none of the existing risk models incorporates mutational<br />

analysis. For example, Bejar <strong>and</strong> colleagues in the Benjamin<br />

Ebert laboratory in Boston assessed 439 samples from<br />

patients with MDS stratified using the IPSS, <strong>and</strong> found that<br />

mutations in EZH2, ASXL1, TP53, or RUNX1 all conferred<br />

an IPSS-independent adverse prognosis in terms of overall<br />

survival. None of the other mutations tested – ETV6, CBL,<br />

NRAS/KRAS, IDH1/2, TET2, NPM1, or JAK2 – had an<br />

IPSS-independent effect on survival. Since the publication<br />

of Bejar’s paper in the New Engl<strong>and</strong> Journal of Medicine,<br />

the Washington University in St. Louis group have described<br />

DNMT3A mutations, which <strong>are</strong> associated with a strikingly<br />

adverse negative IPSS- <strong>and</strong> karyotype-independent prognostic<br />

effect, both in MDS <strong>and</strong> in acute myeloid leukemia.<br />

Finally, somatic SF3B1 mutations, recently described in<br />

MDS by the Cancer Genome Project of the Wellcome Trust<br />

Sanger Institute – represent a class of mutation that appears<br />

to be particularly strongly associated with MDS subtype refractory<br />

anemia with ring sideroblasts <strong>and</strong> associated with<br />

lower-risk disease. Incorporation of these <strong>and</strong> other novel<br />

prognostic markers will continue to require revision of risk<br />

stratification schemes in the years to come.<br />

OP16 Iron Chelation Therapy in MDS<br />

Luca Malcovati, Department of Hematology Oncology,<br />

S. Matteo Hospital Foundation, University of Pavia<br />

Medical School, Pavia, Italy<br />

Anemia is the most frequent peripheral cytopenia observed<br />

in myelodysplastic syndromes (MDS), <strong>and</strong> is an established<br />

negative prognostic factor in patients with MDS.<br />

Most patients with MDS eventually become dependent on<br />

regular red cell transfusions.The onset of a transfusion-dependency<br />

is associated with a worsening of survival <strong>and</strong> an<br />

increased risk of progression into acute myeloid leukemia. 1<br />

Transfusion-dependent patients invariably develop secondary<br />

iron overload. Transfusion iron is primarily taken up<br />

by the reticuloendothelial cells, but later redistributed to p<strong>are</strong>nchymal<br />

cells. This process is modulated by several factors,<br />

including ineffective erythropoiesis through suppression<br />

of hepcidin production. P<strong>are</strong>nchymal iron overload<br />

from transfusions is responsible for morbidity <strong>and</strong> mortality<br />

in patients with thalassemia major, but clinical consequences<br />

have been reported also in patients with MDS.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

The reference method for determining body iron stores is<br />

the assessment of hepatic iron concentration. Although percutaneous<br />

liver biopsy is difficult to implement in MDS because<br />

of neutropenia <strong>and</strong> thrombocytopenia, data have been<br />

reported in these patients. Schafer <strong>and</strong> coworkers demonstrated<br />

an increased liver iron concentration with signs of<br />

portal fibrosis in a high proportion of transfusion-dependent<br />

adults patients with refractory <strong>and</strong> sideroblastic anemia. 2<br />

In MDS patients, body iron status is routinely assessed by<br />

serum ferritin; although its level can be affected by inflammation<br />

or liver disease, sequential measurements were proven<br />

to be of value in monitoring iron overload. In transfusion-dependent<br />

MDS patients retrospective studies showed<br />

that elevated serum ferritin was associated with worse survival,<br />

3 <strong>and</strong> poor outcome after allogeneic hematopoietic<br />

stem cell transplantation. 4<br />

Magnetic resonance imaging T2* provides a non-invasive<br />

method for detecting <strong>and</strong> quantifying both liver <strong>and</strong><br />

myocardial iron overload. Preliminary studies adopting this<br />

approach showed both hepatic <strong>and</strong> myocardial iron accumulation<br />

can be observed in heavily transfused MDS patients.<br />

5<br />

According to the available evidence- <strong>and</strong> consensus-based<br />

guidelines, patients with refractory anemia with ring<br />

sideroblasts or other forms of refractory anemia, in whom<br />

long-term transfusion therapy is likely, <strong>are</strong> recognized as<br />

the best c<strong>and</strong>idates to receive iron chelation therapy. In addition,<br />

patients who <strong>are</strong> c<strong>and</strong>idates for allogeneic stem cell<br />

transplantation may also benefit from chelation therapy,<br />

since iron overload is associated with increased transplantrelated<br />

mortality. 6,7<br />

In conclusion, the available evidence allows to identify<br />

subsets of MDS patients that at risk of developing p<strong>are</strong>nchymal<br />

iron overload <strong>and</strong> might benefit from iron chelation<br />

therapy. Nevertheless, a more accurate information on the<br />

impact of iron-mediated organ damage on the outcome of<br />

patients with MDS is warranted in order to support clinical<br />

decision with evidence-based criteria.<br />

R E F E R E N C E S<br />

1. Cazzola M, Malcovati L. Myelodysplastic syndromes-coping with ineffective<br />

hematopoiesis. N Engl J Med 2005;352(6):536–8.<br />

2. Schafer AI, Cheron RG, Dluhy R, et al. Clinical consequences of acquired<br />

transfusional iron overload in adults. N Engl J Med 1981;304<br />

(6):319–24.<br />

3. Malcovati L, Della Porta M, Pascutto C, et al. Prognostic factors <strong>and</strong> life<br />

expectancy in myelodysplastic syndromes classified according to WHO<br />

criteria. A basis for clinical decision-making. J Clin Oncol 2005;23:<br />

7594–603.<br />

4. Arm<strong>and</strong> P, Kim HT, Cutler CS, et al. Prognostic impact of elevated pretransplant<br />

serum ferritin in patients undergoing myeloablative stem cell<br />

transplantation. Blood 2007;109(10)4586–8.<br />

5. Di Tucci AA, Matta G, Deplano S, et al. Myocardial iron overload assessment<br />

by T2* magnetic resonance imaging in adult transfusion dependent<br />

patients with acquired anemias. Haematologica 2008;93(9):<br />

1385–8.<br />

6. Aless<strong>and</strong>rino EP, Amadori S, Barosi G, et al. Evidence- <strong>and</strong> consensusbased<br />

practice guidelines for the therapy of primary myelodysplastic<br />

syndromes. A statement from the Italian Society of Hematology. Haematologica<br />

2002;87(12):1286–306.<br />

7. Bowen D, Culligan D, Jowitt S, et al. Guidelines for the diagnosis <strong>and</strong><br />

therapy of adult myelodysplastic syndromes. Br J Haematol 2003;120<br />

(2):187–200.<br />

OP17 Progress with epigenetic therapy<br />

in MDS<br />

Pierre Wijermans, Haga Hospital, The Hague,<br />

The Netherl<strong>and</strong>s<br />

Epigenetic therapy with demethylating agents for intermediate<br />

<strong>and</strong> high risk MDS was developed over the last 10<br />

years. We now know that up to almost 50% of the patients<br />

can reach a response when treated properly. Many problems<br />

in the treatment of these patients however <strong>are</strong> not solved yet<br />

as for instance who should be treated, what is the most efficient<br />

drug, what is the best dosing scheme <strong>and</strong> how long<br />

should we give therapy in case of response.<br />

There <strong>are</strong> some answer to give on the basis of new information<br />

that <strong>are</strong> relevant to the treating physicians.<br />

Who should be treated?<br />

The Italian study with Azacytidine in low risk en intermediate<br />

risk patients confirmed earlier 5-aza-2-deoxycytidine<br />

(Decitabine) experience that treating some of these patients<br />

is helpful. Longer duration of the treatment with probably<br />

also maintenance in case of good response seems appropriate.<br />

Also some of the more high risk patients especially with<br />

adversed cytogenetic abnormalities might be good c<strong>and</strong>idates.<br />

The first results come available that also some of AML<br />

patients may benefit.<br />

Which drugs should we use<br />

Azacytidine <strong>and</strong> Decitabine probably do differ in their effect<br />

on the tumor cell therefore combination therapy seems<br />

an interesting option.<br />

HDAC inhibitors <strong>are</strong> used in multiple phase II trials <strong>and</strong><br />

although potentially very interesting have not showed major<br />

clinical inpact in the treatment of MDS patients yet. However<br />

combination therapy is an option based upon the working<br />

mechanism of the two drugs.<br />

Which dosing scheme<br />

Alternative more convenient dosing schemes with lower<br />

dose both for Azacytidine <strong>and</strong> Decitabine <strong>are</strong> presented.<br />

Some of them <strong>are</strong> using lower doses. How low can we go<br />

with the dose? Prelininary results of studies to intracellular<br />

drug levels revealed that an intra-individual metabolism<br />

might important.<br />

How long should we treat<br />

Complete cure of the MDS is with epigenetic therapy not<br />

possible. We know that often soon after stopping (even in de<br />

cases of a CR) the disease will relapse. Individual experience<br />

of physicians showed us that long term maintenance<br />

treatment might a good option.<br />

The option to treat fit patients with a donor with an allotransplantation<br />

after reaching a CR with epigenetic therapy<br />

has been explored with some success.<br />

25


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

OP18 Hematopoietic cell transplantation<br />

for MDS: New developments<br />

Theo J.M. de Witte, Department of Tumor Immunology,<br />

Radboud University Medical Centre, Nijmegen,<br />

The Netherl<strong>and</strong>s<br />

The spectrum of myelodysplastic syndromes (MDS) varies<br />

from an indolent course over several years to a rapid<br />

progression to acute myeloid leukemia (AML) 1 . MDS classification<br />

systems have been modified recently, leading to<br />

the WHO classifications <strong>and</strong> WHO-based prognostic scoring<br />

system (WPSS) 2, 3 . The new classifications have improved<br />

the accuracy of prognosis by incorporating the impact<br />

of cytogenetic characteristics <strong>and</strong> transfusion dependency.<br />

WHO classifications <strong>and</strong> WPSS showed prognostic<br />

relevance after allo-SCT 4 . According to WPSS criteria, low<br />

risk patients have the highest overall survival rate (80%) <strong>and</strong><br />

the lowest relapse probability (9%) 5 years after transpalntation<br />

5 , but most patients with these low risk criteria do not<br />

require treatment with allo-SCT.<br />

Currently, the most effective curative treatment is allogeneic<br />

stem cell transplantation (alloSCT), with long-term<br />

survival rates between 25% <strong>and</strong> 70%. However, treatmentrelated<br />

mortality continues to be a limiting factor 6 . The most<br />

important factors influencing outcome after alloSCT <strong>are</strong> not<br />

recipient age or intensity of conditioning, but disease stage<br />

at the time of transplantation 7 .<br />

Several large studies in de-novo AML have assessed the<br />

role of allogeneic SCT by a donor versus no donor comparison<br />

8–10 . In these studies the advantage of the presence of<br />

a histocompatible sibling donor was mainly restricted to patients<br />

younger than 40 years. In the MDS Criant study the<br />

average age was considerably higher than 40 years <strong>and</strong> only<br />

28% (38/135) of the patients who reached CR <strong>and</strong> ≤55 years<br />

old, were younger than 40 years 11 . In the Criant study only<br />

patients with unfavorable or intermediate cytogenetic characteristics<br />

appe<strong>are</strong>d to benefit from allogeneic stem cell<br />

transplantation 11 . Whether patients with advanced stages<br />

of MDS should receive remission-induction chemotherapy<br />

prior to the transplant conditioning remains a point for discussion<br />

11 . Retrospective analyses showed conflicting data.<br />

Interpretation of the data is hampered by various selection<br />

biases in the two treatment approaches <strong>and</strong> by lacking details<br />

about the administered chemotherapy prior to the transplant<br />

conditioning 6,12 . The Criant study shows that the great<br />

majority of patients with an identified donor received the<br />

transplant (94%). The 4-year DFS rate of the donor-group<br />

of 46% is encouraging comp<strong>are</strong>d to large registry data. 13<br />

In conclusion: these data suggest that allogeneic SCT<br />

may be the treatment of choice for the young patients (age<br />

≤55 years) with MDS, characterized by poor risk or intermediate<br />

risk cytogenetics, who have a histocompatible donor.<br />

For patients lacking an HLA-compatible sibling donor,<br />

the outcome with autologous SCT or chemotherapy may be<br />

good alternatives for MDS patients with good-risk cytogenetic<br />

characteristics. Until now, no study has proven superiority<br />

of autologous SCT comp<strong>are</strong>d to intensive chemotherapy<br />

11 .<br />

26<br />

Reduced intensity conditioning regimens (RIC) for alloSCT<br />

in MDS has dramatically exp<strong>and</strong>ed the scope of allografting<br />

to many previously ineligible older patients, or<br />

patients with multiple co-morbidities. The heterogeneity<br />

amongst conditioning regimens between centers is considerable,<br />

but usually the procedure appears to be associated<br />

with durable hematopoietic engraftment amongst both sibling<br />

<strong>and</strong> unrelated HLA-matched donor allografts, together<br />

with an acceptable level of transplant-related mortality. The<br />

question whether RIC regimens result in better outcome<br />

when comp<strong>are</strong>d to marrow ablative conditioning has been<br />

addressed in several studies. EBMT performed a retrospective<br />

study of 836 patients with MDS undergoing allogeneic<br />

HSCT with HLA matched siblings, comparing 215 patients<br />

receiving reduced intensity conditioning with 621 patients<br />

receiving st<strong>and</strong>ard myeloablative conditioning (SMC). Patients<br />

receiving RIC were older (age > 50 years in 73% RIC<br />

vs. 28% in SMC, P


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

iron absorption <strong>and</strong> increased iron toxicity. Infections occur<br />

frequently in patients with AML <strong>and</strong> MDS, especially after<br />

during <strong>and</strong> after intensive antileukemic therapy. Infections<br />

<strong>are</strong> associated with increased levels of cytokines, such as<br />

IL6, which also blocks Hepcidin production <strong>and</strong> increased<br />

iron absorption in the gut with its associated iron toxicity.<br />

Finally, every unit of erythrocytes adds 200 mg of iron to<br />

the total iron store of 5 gm in an individual of 75 kg, Several<br />

studies 16 indicate that pre-conditioning ferritin levels<br />

<strong>are</strong> associated with post-transplant complications, such as<br />

mucositis, bacteremia, <strong>and</strong> fever episodes 4, 18 . This association<br />

was app<strong>are</strong>nt in autologous 19 , but more importantly in<br />

the large retrospective analysis performed by the Dana Farber<br />

Institute Group on 543 recipients of allogeneic HSCT 20 .<br />

This study showed a strong association between pretransplantation<br />

serum ferritin level <strong>and</strong> a lower overall survival,<br />

especially in patients with MDS <strong>and</strong> acute leukemia (AL) 20 .<br />

Strikingly, the median ferritin level in this group was 930<br />

mg/l. The percentage of patients with elevated ferritin levels<br />

varied greatly from 25% in chronic myeloid leukemia<br />

(CML) to 70% in lymphoma, 81% in MDS <strong>and</strong> 97% in AL.<br />

The question is what to do to prevent this iron overload<br />

related toxicity? The approach should be multifactorial because<br />

iron overload <strong>and</strong> toxicity cannot be solely contributed<br />

to the number of transfused units of erythrocytes 21 . In<br />

a postmortem study of 10 patients who died early after<br />

HSCT 21 the iron content was in the range of hemachromatosis<br />

patients while the average number of transfusions given<br />

from diagnosis to death was only 48 units. In the pretransplant<br />

period vigorous iron chelation may be important, but<br />

prospective studies should prove a survival benefit. During<br />

the transplant period anti-oxidant therapy, including various<br />

iron chelators, glutamine or the angiotensin-converting enzyme<br />

(ACE)-inhibitor Captopril may be considered, but<br />

clinical experience is limited to anecdotal observations 16 .<br />

During the conditioning <strong>and</strong> the early post-transplant period,<br />

innocuous glutathione-depleting agents like acetaminophen<br />

should be avoided 16 . Finally, in the post-transplant<br />

period phlebotomy, sometimes combined with erythropoietin,<br />

has been successfully applied after HSCT in thalassemic<br />

patients. For those patients who cannot be phlebotomized<br />

iron chelation can be considered as well.<br />

In conclusion: iron toxicity is likely to be an underestimated<br />

cause of HSCT treatment-related mortality. We need<br />

to know more on the pathogenesis, the diagnosis <strong>and</strong> the<br />

monitoring of iron induced organ damage. Prevention of<br />

iron-induced toxicity will require a multidisciplinary approach<br />

of biochemists, pathologists, transfusionists <strong>and</strong> clinicians.<br />

It is clear we do need well-designed prospective<br />

studies both with interventions prior to HSCT <strong>and</strong> after<br />

HSCT.<br />

Several new drugs have now been approved for the treatment<br />

of MDS. The question often arises, especially in older<br />

patients, whether patients should be treated with those (less<br />

toxic) drugs <strong>and</strong> be transplanted only if they do not or no<br />

longer respond or should undergo HSCT at the time of optimum<br />

response. In addition, these new treatment modalities<br />

may be used to reduce more efficiently the MDS clone prior<br />

to the HSCT conditioning. Weighing the pros <strong>and</strong> cons in<br />

view of the potential post-HSCT complications, especially<br />

GVHD, provides a challenging task. GVHD, particularly in<br />

its chronic form, occurs in 50%–60% of patients. Ongoing<br />

trials <strong>are</strong> directed at optimizing pre-HSCT therapy, conditioning<br />

<strong>and</strong> GVHD prophylactic regimens to minimize toxicity<br />

while maintaining a potent cytotoxic effect with low<br />

GVHD incidence. These efforts include various approaches<br />

of pre HSCT therapy, incorporation of ATG, use of antibody-conjugated<br />

radioisotopes <strong>and</strong> post- HSCT administration<br />

of chemotherapy, primarily hypomethylating agents.<br />

R E F E R E N C E S<br />

1. Bennett JM, Catovsky D, Daniel MT et al. Proposals for the classification<br />

of the myelodysplastic syndromes. Br J Haematol 1982;51(2):<br />

189–199.<br />

2. Malcovati L, Porta MG, Pascutto C et al. Prognostic factors <strong>and</strong> life<br />

expectancy in myelodysplastic syndromes classified according to WHO<br />

criteria: a basis for clinical decision making. J Clin Oncol 2005;23<br />

(30):7594–7603.<br />

3. Malcovati L, Germing U, Kuendgen A et al. Time-dependent prognostic<br />

scoring system for predicting survival <strong>and</strong> leukemic evolution in<br />

myelodysplastic syndromes. J Clin Oncol 2007;25(23):3503–3510.<br />

4. Aless<strong>and</strong>rino EP, la Porta MG, Bacigalupo A et al. Prognostic impact<br />

of pre-transplantation transfusion history <strong>and</strong> secondary iron overload<br />

in patients with myelodysplastic syndrome undergoing allogeneic stem<br />

cell transplantation: a GITMO study. Haematologica 2010;95(3):<br />

476–484.<br />

5. Aless<strong>and</strong>rino EP, la Porta MG, Bacigalupo A et al. WHO classification<br />

<strong>and</strong> WPSS predict posttransplantation outcome in patients with myelodysplastic<br />

syndrome: a study from the Gruppo Italiano Trapianto di<br />

Midollo Osseo (GITMO). Blood 2008;112(3):895–902.<br />

6. de Witte T, Hermans J, Vossen J et al. Haematopoietic stem cell transplantation<br />

for patients with myelo-dysplastic syndromes <strong>and</strong> secondary<br />

acute myeloid leukaemias: a report on behalf of the Chronic Leukaemia<br />

Working Party of the European Group for Blood <strong>and</strong> Marrow Transplantation<br />

(EBMT). Br J Haematol 2000;110(3):620–630.<br />

7. Lim Z, Br<strong>and</strong> R, Martino R et al. Allogeneic hematopoietic stem-cell<br />

transplantation for patients 50 years or older with myelodysplastic syndromes<br />

or secondary acute myeloid leukemia. J Clin Oncol 2010;28(3):<br />

405–411.<br />

8. Suciu S, M<strong>and</strong>elli F, de Witte T et al. Allogeneic comp<strong>are</strong>d with autologous<br />

stem cell transplantation in the treatment of patients younger than<br />

46 years with acute myeloid leukemia (AML) in first complete remission<br />

(CR1): an intention-to-treat analysis of the EORTC/GIME-<br />

MAAML-10 trial. Blood 2003;102(4):1232–1240.<br />

9. Cornelissen JJ, van Putten WL, Verdonck LF et al. Results of a HOV-<br />

ON/SAKK donor versus no-donor analysis of myeloablative HLAidentical<br />

sibling stem cell transplantation in first remission acute myeloid<br />

leukemia in young <strong>and</strong> middle-aged adults: benefits for whom?<br />

Blood 2007;109(9):3658–3666.<br />

10. Burnett AK, Wheatley K, Goldstone AH et al. The value of allogeneic<br />

bone marrow transplant in patients with acute myeloid leukaemia at<br />

differing risk of relapse: results of the UK MRC AML 10 trial. Br J<br />

Haematol 2002;118(2):385–400.<br />

11. de Witte T, Hagemeijer A, Suciu S et al. Value of allogeneic versus autologous<br />

stem cell transplantation <strong>and</strong> chemotherapy in patients with<br />

myelodysplastic syndromes <strong>and</strong> secondary acute myeloid leukemia.<br />

Final results of a prospective r<strong>and</strong>omized European Intergroup Trial.<br />

Haematologica 2010;95(10):1754–1761.<br />

12. Nakai K, K<strong>and</strong>a Y, Fukuhara S et al. Value of chemotherapy before allogeneic<br />

hematopoietic stem cell transplantation from an HLA-identical<br />

sibling donor for myelodysplastic syndrome. Leukemia 2005;19(3):<br />

396–401.<br />

13. Sierra J, Perez WS, Rozman C et al. Bone marrow transplantation from<br />

HLA-identical siblings as treatment for myelodysplasia. Blood 2002;<br />

100(6):1997–2004.<br />

14. Martino R, Iacobelli S, Br<strong>and</strong> R et al. Retrospective comparison of reduced-intensity<br />

conditioning <strong>and</strong> conventional high-dose conditioning<br />

for allogeneic hematopoietic stem cell transplantation using HLA-identical<br />

sibling donors in myelodysplastic syndromes. Blood 2006;108<br />

(3):836–846.<br />

27


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

15. Sorror ML, Maris MB, Storb R et al. Hematopoietic cell transplantation<br />

(HCT)-specific comorbidity index: a new tool for risk assessment before<br />

allogeneic HCT. Blood 2005;106(8):2912–2919.<br />

16. Evens AM, Mehta J, Gordon LI. Rust <strong>and</strong> corrosion in hematopoietic<br />

stem cell transplantation: the problem of iron <strong>and</strong> oxidative stress.<br />

Bone Marrow Transplant 2004;34(7):561–571.<br />

17. Tanno T, Bhanu NV, Oneal PA et al. High levels of GDF15 in thalassemia<br />

suppress expression of the iron regulatory protein hepcidin. Nat<br />

Med 2007;13(9):1096–1101.<br />

18. Pullarkat V. Objectives of iron chelation therapy in myelodysplastic syndromes:<br />

more than meets the eye? Blood 2009;114(26):5251–5255.<br />

19. Altes A, Ruiz A, Martinez C et al. The relationship between iron<br />

overload <strong>and</strong> clinical characteristics in a Spanish cohort of 100 C282Y<br />

homozygous hemochromatosis patients. Ann Hematol 2007;86(11):<br />

831–835.<br />

20. Arm<strong>and</strong> P, Kim HT, Cutler CS et al. Prognostic impact of elevated pretransplantation<br />

serum ferritin in patients undergoing myeloablative<br />

stem cell transplantation. Blood 2007;109(10):4586–4588.<br />

21. Strasser SI, Kowdley KV, Sale GE, McDonald GB. Iron overload in<br />

bone marrow transplant recipients. Bone Marrow Transplant 1998;22<br />

(2):167–173.<br />

OP19 Treatment Options for Myelodysplastic<br />

Syndromes<br />

Elias Jabbour, Guillermo Garcia-Manero, Hagop<br />

Kantarjian, Department of Leukemia, The University of<br />

Texas M.D. Anderson Cancer Center, Houston, Texas<br />

Abstract: Myelodysplastic syndromes <strong>are</strong> a group of<br />

heterogeneous hematopoietic stem cell disorders characterized<br />

by peripheral blood cytopenias <strong>and</strong> a risk of transformation<br />

to acute myeloid leukemia. Until recently, treatment<br />

of MDS predominately consisted of supportive c<strong>are</strong> measures.<br />

However, three agents for the treatment of MDS have<br />

recently been approved: lenalidomide, decitabine <strong>and</strong> azacitidine.<br />

These agents have dramatically improved the outcomes<br />

for patients with MDS. To date, azacitidine is the<br />

only agent that has demonstrated a survival advantage when<br />

comp<strong>are</strong>d to conventional c<strong>are</strong>. Novel agents <strong>and</strong> combination<br />

regimens including lenalidomide, decitabine <strong>and</strong> azacitidine<br />

<strong>are</strong> being explored in an effort to further improve<br />

patient outcomes.<br />

Introduction<br />

Myelodysplastic syndromes (MDS) <strong>are</strong> a heterogeneous<br />

group of clonal hematopoietic stem cell disorders that impair<br />

normal hematopoiesis, resulting in a variable number<br />

of cytopenias <strong>and</strong> a potential to evolve into acute myeloid<br />

leukemia (AML). 1 With a median age at diagnosis around<br />

70 years, MDS typically affects the elderly. 1,2 Hence, there<br />

is much morbidity <strong>and</strong> mortality associated with this patient<br />

population, as patients frequently suffer from complications<br />

due to cytopenias as well as other comorbidities. The two<br />

systems used for classifying MDS <strong>are</strong> the French American<br />

<strong>and</strong> British (FAB) criteria <strong>and</strong> the more recently revised<br />

World Health Organization (WHO) classification system. A<br />

third system, the International Prognostic Scoring System<br />

(IPSS) can predict survival based on percentage of bone<br />

marrow blasts, karyotype, <strong>and</strong> number of peripheral blood<br />

cytopenias3 <strong>and</strong> is the most widely used prognostic tool for<br />

assisting with treatment decisions.<br />

For many years, supportive c<strong>are</strong> with blood products (red<br />

blood cell (RBC) <strong>and</strong> platelet transfusions), hematopoietic<br />

28<br />

growth factors, <strong>and</strong> antibiotics remained the only treatment<br />

modality for MDS patients, until the development of three<br />

novel agents that may alter the natural history of this disease.<br />

Within the past decade, the United States (U.S.) Food<br />

<strong>and</strong> Drug Administration (FDA) has approved an immunomodulatory<br />

agent, lenalidomide (Revlimid, Celgene) <strong>and</strong><br />

two hypomethylating agents, decitabine (Dacogen, Eisai,<br />

Inc.) <strong>and</strong> azacitidine (Vidaza, Celgene) for the treatment<br />

of patients with MDS. In simple practice, therapy is tailored<br />

to IPSS score with an emphasis on supportive c<strong>are</strong> therapies<br />

or lenalidomide for lower-risk patients (IPSS low-or intermediate<br />

[int]-1) <strong>and</strong> more intensive therapies such as conventional<br />

chemotherapy, allogeneic hematopoietic stem cell<br />

transplant (HST) <strong>and</strong> clinical trials, as well as hypomethylating<br />

agents, for patients with higher-risk disease (IPSS int-<br />

2 or high) or lower-risk patients with progressive disease. 3<br />

Treatment of Lower-risk MDS<br />

Supportive C<strong>are</strong><br />

Initial clinical management of lower-risk MDS patients<br />

with symptomatic anemia includes the use of erythropoiesis<br />

stimulating agents (ESA), such as epoetin alpha (Epogen,<br />

Amgen; Procrit,Ortho Biotech) or darbepoetin alfa (Aranesp,<br />

Amgen). ESAs have been shown to reduce RBC<br />

transfusion needs in MDS patients <strong>and</strong> when given with<br />

granulocyte colony-stimulating factor (G-CSF), ESAs have<br />

been shown to confer a survival advantage. 4 The likelihood<br />

of response to ESAs has been correlated with RBC transfusion<br />

needs <strong>and</strong> serum erythropoietin levels. Patients with<br />

low transfusion requirements (500 mU/mL <strong>are</strong> least likely to respond to ESAs. 5,6 Demonstrated<br />

in a recent prospective, r<strong>and</strong>omized study by the<br />

<strong>East</strong>ern Cooperative Oncology Group (ECOG), patients<br />

with low-risk MDS <strong>and</strong> low serum EPO levels experienced<br />

higher erythroid response rates when given ESAs with or<br />

without G-CSF comp<strong>are</strong>d to patients with high-risk disease.<br />

7<br />

Immunosuppressive therapy (IST)<br />

A subset of MDS patients with bone marrow failure responds<br />

to IST, which suggests that an immune-mediated<br />

pathogenesis is responsible for the cytopenias. In these patients<br />

with hypocellular bone marrows, IST with antithymocyte<br />

globulin (ATG), cyclosporine or both can be utilized.<br />

In patients with MDS, ATG alone has been shown to decrease<br />

red blood cell transfusion requirements as well as<br />

improve neutropenia <strong>and</strong> thrombocytopenia. 8 Likewise,<br />

long term outcomes in low risk MDS patients have shown<br />

higher response rates with ATG <strong>and</strong> cyclosporine (54%)<br />

than ATG alone (29%)(P=0.004). 9 Factors that have been<br />

shown to favor response to ATG include age, 8–10 HLA-DR15<br />

positivity, 8–10 <strong>and</strong> shorter duration of red blood cell transfusion<br />

requirements. 8,9 In general, patients will respond to IST<br />

within 3 to 4 months <strong>and</strong> will remain clinically stable for<br />

years, but many eventually relapse with return of cytopeni-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

as. Recently, a phase I/II pilot study demonstrated alemtuzumab,<br />

an anti-CD52 monoclonal antibody, to produce durable<br />

responses in patients with intermediate-risk MDS who<br />

fit criteria to respond to IST. Ninety-seven percent of int-1<br />

risk patients <strong>and</strong> 57% of int-2 patients responded to alemtuzumab<br />

by 3 months, which was superior to response rates<br />

reported with ATG alone 9 . Furthermore, at 12 months follow-up,<br />

56% of responding patients had normal blood<br />

counts <strong>and</strong> 78% of patients were transfusion independent. 11<br />

Immunomodulating Therapy (IMID)<br />

Lenalidomide<br />

Lenalidomide was FDA approved in 2005 for the treatment<br />

of patients with transfusion-dependent anemia due to<br />

low- <strong>and</strong> int-1 risk MDS associated with deletion 5q karyotype<br />

(del(5q)). The approval of lenalidomide was based on<br />

two phase II studies demonstrating a high frequency of erythroid<br />

responses in patients with an isolated del(5q) or<br />

del(5q) with other abnormalities. 12,13 The majority of patients<br />

in these studies were IPSS low- or int-1 risk patients<br />

with transfusion-dependent anemia. The majority of patients<br />

achieved a hematologic response (reduced transfusion<br />

needs) <strong>and</strong> became transfusion independent. Of note, responses<br />

occurred despite karyotype complexity <strong>and</strong> patients<br />

with thrombocytopenia at baseline were less likely to respond<br />

to lenalidomide comp<strong>are</strong>d to patients with platelets<br />

>100,000 per cubic milliliter at baseline. Furthermore, a<br />

majority of patients had cytogenetic improvement, <strong>and</strong> 45%<br />

had a complete cytogenetic response. Hence, these responses<br />

illustrate lenalidomide’s effect on the biology of the disease<br />

by reversing the cytogenetic abnormalities associated<br />

with the 5q13 deletion. 13 Following these studies, Raza et al<br />

evaluated the safety <strong>and</strong> efficacy of lenalidomide in 214 patients<br />

with transfusion-dependent anemia due to lower-risk<br />

MDS without del(5q). Hematologic responses were lower<br />

than those observed in patients with del(5q), but the rate of<br />

transfusion independence was 26% <strong>and</strong> the median duration<br />

of transfusion independence was 41 weeks. 14 Lenalidomide<br />

demonstrated a potential benefit for low risk patients without<br />

del(5q) who remain transfusion dependent after supportive<br />

c<strong>are</strong> measures or for patients who <strong>are</strong> not c<strong>and</strong>idates<br />

for ESAs or more intensive therapy. Although lenalidomide<br />

is generally well tolerated, it induces significant cytopenias<br />

resulting in dose reduction or interruption in a majority of<br />

patients.<br />

Treatment of Higher-risk MDS<br />

Hypomethylating Agents<br />

Azacitidine <strong>and</strong> Decitabine<br />

Azacitidine <strong>and</strong> decitabine <strong>are</strong> both hypomethylating<br />

agents that irreversibly inhibit DNA methyltransferase, resulting<br />

in progressive loss of methylation <strong>and</strong> reactivation<br />

of tumor suppressor genes. Azacitidine was the first DNA<br />

methyltransferase inhibitor approved by the U.S. FDA in<br />

2004 for the treatment of all FAB subtypes of MDS. This<br />

approval was based on the results of three CALGB studies<br />

which evaluated the efficacy <strong>and</strong> safety of azacitidine 75<br />

mg/m2 IV or SQ x 7 days every 28 days in MDS patients. A<br />

pooled analysis of the CALGB trials (8421, 8921, <strong>and</strong> 9221)<br />

reported overall response rates (ORR) between 40 <strong>and</strong> 47%<br />

(CR 10–17%, PR 1%, HI 23–36%) for azacitidine <strong>and</strong> 17%<br />

(HI only) for BSC by IWG 2000 criteria 15 . 16<br />

Subsequently, the AZA-001 trial established an improved<br />

overall survival in patients with higher-risk MDS. In this<br />

phase III trial, 358 patients with IPSS int-2 or high-risk<br />

MDS were r<strong>and</strong>omized to receive azacitidine (75 mg/m 2 SQ<br />

daily x 7 days, every 28 days) or conventional c<strong>are</strong> (BSC,<br />

low-dose cytarabine, or intensive chemotherapy). The overall<br />

survival was significantly longer with azacitidine comp<strong>are</strong>d<br />

to conventional c<strong>are</strong> (24.5 vs. 15 months, HR 0.58;<br />

95% CI 0.43–0.77, p=0.0001) <strong>and</strong> was present regardless of<br />

MDS subtype <strong>and</strong> IPSS subgroup. The median time to AML<br />

transformation was reduced with azacitidine comp<strong>are</strong>d to<br />

conventional c<strong>are</strong> (17.8 vs. 11.5 months; HR 0.5; 95% CI<br />

0.35–0.7, p


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

mg/m 2 IV x 5 days every 4 weeks. By modified IWG criteria<br />

21 , the ORR was 51% <strong>and</strong> the median survival was 19.4<br />

months. Patients received a median of five cycles of treatment<br />

<strong>and</strong> 82% of the responses were seen by the second<br />

cycle. 22 Furthermore, the efficacy of low-dose decitabine<br />

<strong>and</strong> intensive chemotherapy was comp<strong>are</strong>d in a historical<br />

comparison of patients with higher-risk MDS at the MD<br />

Anderson Cancer Center. This study showed a similar CR<br />

rate, but significantly lower 6-week <strong>and</strong> 3-month mortality<br />

rates <strong>and</strong> improved survival with decitabine. 23<br />

Patients require several courses of azacitidine <strong>and</strong> decitabine<br />

(3–6 cycles) before demonstrating a response, so<br />

drug- <strong>and</strong> disease-induced myelosuppression can be common<br />

during this time period. Therefore, treatment should<br />

continue with both agents for a minimum of 3–4 cycles before<br />

declaring therapy a failure. Although azacitidine is an<br />

outpatient regimen, it is complicated by a 7- consecutive<br />

day regimen, necessitating weekend administration. More<br />

convenient regimens (i.e. 5 day, 5 days followed by 2 additional<br />

weekdays) have been explored in predominately lower-risk<br />

patients <strong>and</strong> response rates appear similar among the<br />

three regimens. 24,25 However, it is not certain that these regimens<br />

will lead to a survival advantage in higher-risk MDS<br />

patients.<br />

Stem Cell Transplant<br />

The only potential curative option for MDS is allogeneic<br />

stem cell transplant (HSCT), but is restricted to patients<br />

with a donor <strong>and</strong> those free from co-morbidities that may<br />

preclude them from this option. For patients older than 55<br />

years, the mortality rate is approximately 38%. For patients<br />

that <strong>are</strong> eligible for HSCT, the timing of transplantation is<br />

important. Published data identifying the optimal timing of<br />

HSCT is scant. However, a retrospective analysis of patients<br />

younger than 60 years of age who received a myeloablative<br />

conditioning regimen from a sibling donor transplant suggests<br />

that survival is better for low- <strong>and</strong> int-1 risk patients<br />

receiving a transplant at the time of disease progression<br />

rather than at diagnosis. Outcomes for int-2 <strong>and</strong> high-risk<br />

patients <strong>are</strong> better when transplantation is done as early as<br />

possible. 26 Therefore, donor screening for younger patients<br />

should begin as soon as possible regardless of the IPSS<br />

score at diagnosis. This sheds some light on the timing for<br />

younger patients. For elderly patients, reduced intensity<br />

transplants may be an option.<br />

Conclusion<br />

Patients with MDS <strong>are</strong> predominately elderly <strong>and</strong> have<br />

multiple comorbidities that preclude them of curative therapies.<br />

For many years, supportive c<strong>are</strong> measures remained<br />

the only treatment modality for MDS patients, until the development<br />

of three novel agents that may alter the biology<br />

of this disease. The recent development of three FDA-approved<br />

agents, lenalidomide, azacitidine <strong>and</strong> decitabine,<br />

have dramatically changed the MDS l<strong>and</strong>scape. Lenalidomide<br />

is remarkably effective in lower-risk patients, producing<br />

complete transfusion independence in the majority of<br />

del(5q) <strong>and</strong> some non-del(5q) patients. The hypomethylating<br />

agents, azacitidine <strong>and</strong> decitabine, <strong>are</strong> effective in the<br />

30<br />

setting of higher-risk MDS patients, producing responses in<br />

nearly half the treated patients. Comp<strong>are</strong>d to conventional<br />

c<strong>are</strong> regimens, azacitidine has significantly improved the<br />

median survival of higher-risk patients. Despite these therapeutic<br />

advances, the responses in higher-risk patients <strong>are</strong><br />

not durable, making the search for other novel agents necessary.<br />

R E F E R E N C E S<br />

1. Heaney M, Golde D. Myelodysplasia. N Engl J Med 1999;340:<br />

1649–60.<br />

2. Sekeres MA, Schoonen WM, Kantarjian H, et al. Characteristics of US<br />

patients with myelodysplastic syndromes: results of six cross-sectional<br />

physician surveys. J Natl Cancer Inst 2008;100:1542–51.<br />

3. Greenberg P, Cox C, LeBeau MM, et al. International scoring system<br />

for evaluating prognosis in myelodysplastic syndromes. Blood 1997;<br />

89:2079–88.<br />

4. Jadersten M, Malcovati L, Dybedal I, et al. Erythropoietin <strong>and</strong> granulocyte-colony<br />

stimulating factor treatment associated with improved<br />

survival in myelodysplastic syndrome. J Clin Oncol 2008;26:3607–13.<br />

5. Hellstrom-Lindberg E. Efficacy of erythropoietin in the myelodysplastic<br />

syndromes: a meta-analysis of 205 patients from 17 studies. Brit J<br />

Haemat 1995;89:67–71.<br />

6. Hellstrom-Lindberg E, Gulbr<strong>and</strong>sen N, Lindberg G, et al. A validated<br />

decision model for treating the anaemia of myelodysplastic syndromes<br />

with erythropoietin + granulocyte colony-stimulating factor: significant<br />

effects on quality of life. Br J Haematol 2003;120:1037–46.<br />

7. Greenburg PL, Sun Z, Miller KB, et al. Treatment of myelodysplastic<br />

syndrome patients with erythropoietin with or without granulocyte<br />

colony-stimulating factor: results of a prospective r<strong>and</strong>omized phase 3<br />

trial by <strong>East</strong>ern Cooperative Oncology Group (E1996). Blood 2009;<br />

114:2393–2400.<br />

8. Molldrem JJ, Leifer E, Bahceci E, et al. Antithymocyte globulin for<br />

treatment of the bone marrow failure associated with myelodysplastic<br />

syndromes, Ann Intern Med, 2002;137:156–63.<br />

9. Slo<strong>and</strong> EM, Wu CO, Greenberg P, et al. Factors affecting response <strong>and</strong><br />

survival in patients with myelodysplasia treated with immunosuppressive<br />

therapy. J Clinical Oncol 2008;26:2505–11.<br />

10. Saunthararajah Y, Nakamura R, Wesley R, et al. A simple method to<br />

predict response to immunosuppressive therapy in patients with myelodysplastic<br />

syndrome. Blood 2003;102:3025–7.<br />

11. Slo<strong>and</strong> EM, Olnes MJ, Shenoy A, et al. Alemtuzumab treatment of intermediate-1<br />

myelodysplasia patients is associated with sustained improvement<br />

in blood counts <strong>and</strong> cytogenetic remissions. J Clin Oncol<br />

2010;28:5166–73.<br />

12. List A, Kurtin S, Roe DJ, et al. Efficacy of lenalidomide in myelodysplastic<br />

syndromes. N Engl J Med 2005;352: 549–57.<br />

13. List A, Dewald G, Bennett J, et al. Lenalidomide in the myelodysplastic<br />

syndrome with chromosome 5q deletion. New Engl J Med 2006;355:<br />

1456–65.<br />

14. Raza A, Reeves JA, Feldman EJ, et al. Phase 2 study of lenalidomide in<br />

transfusion-dependent, low-risk, <strong>and</strong> intermediate-1- risk myelodysplastic<br />

syndromes with karyotypes other than deletion 5q. Blood 2008;<br />

111:86–93.<br />

15. Cheson BD, Bennett JM, Kantarjian H, et al. Report of an international<br />

working group to st<strong>and</strong>ardize response criteria for myelodysplastic<br />

syndromes. Blood 2000;96:3671–4.<br />

16. Silverman LR, McKenzie DR, Peterson BL, et al. Further analysis of<br />

trials with azacitidine in patients with myelodysplastic syndrome: studies<br />

8421, 8921, <strong>and</strong> 9221 by the Cancer <strong>and</strong> Leukemia Group B. J Clin<br />

Oncol 2006;24:3895–3903.<br />

17. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine<br />

comp<strong>are</strong>d with that of conventional c<strong>are</strong> regimens in the treatment<br />

of higher-risk myelodysplastic syndromes: a r<strong>and</strong>omized, open-label,<br />

phase III study. Lancet Oncol 2009;10:223–32.<br />

18. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient<br />

outcomes in myelodysplastic syndromes: results of a phase III r<strong>and</strong>omized<br />

study. Cancer 2006;106:1794–803.<br />

19. WijerMans P, Suciu S, Liliana Baila L, et al. Low dose decitabine versus<br />

best supportive c<strong>are</strong> in elderly patients with intermediate or high<br />

risk MDS not eligible for intensive chemotherapy: final results of the


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

r<strong>and</strong>omized phase III study (06011) of the EORTC Leukemia <strong>and</strong> German<br />

MDS Study Groups. Blood 2008;112: abstract 226.<br />

20. Kantarjian H, Oki Y, Garcia-Manero G, et al. Results of a r<strong>and</strong>omized<br />

study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic<br />

syndrome <strong>and</strong> chronic myelomonocytic leukemia. Blood 2007;<br />

109:52–7.<br />

21. Cheson BD, Greenberg PL, Bennett JM, et al. Clinical application <strong>and</strong><br />

proposal for modification of the International Working Group (IWG)<br />

response criteria in myelodysplasia. Blood 2006;108:419–25.<br />

22. Steensma DP, Baer MR, Slack JL, et al. Multicenter study of decitabine<br />

administered daily for 5 days every 4 weeks to adults with myelodysplastic<br />

syndromes: the alternative dosing for outpatient treatment<br />

(ADOPT) trial. J Clin Oncol 2009;27: 3842–8.<br />

23. Kantarjian HM, O’Brien S, Huang X, et al. Survival advantage with<br />

decitabine versus intensive chemotherapy in patients with higher risk<br />

myelodysplastic syndrome: comparison with historical experience.<br />

Cancer 2007;109:1133–7.<br />

24. Lyons RM, Cosgriff TM, Modi SS, et al. Hematologic response to three<br />

alternative dosing schedules of azacitidine in patients with myelodysplastic<br />

syndromes. J Clin Oncol 2009; 27:1850–6.<br />

25. Martin MG, Walgren RA, Procknow E, et al. A phase II study of 5-day<br />

intravenous azacitidine in patients with myelodysplastic syndromes.<br />

Am J Hematol 2009;84: 560–4.<br />

26. Cutler CS, Lee SJ, Greenberg P, et al. A decision analysis of allogeneic<br />

bone marrow transplantation for the myelodysplastic syndromes: delayed<br />

transplantation for low-risk myelodysplasia is associated with<br />

improved outcome. Blood 2004;104:579–85.<br />

OP20 Pathophysiology of MPNs <strong>and</strong> role of JAK2<br />

mutations<br />

Richard A Van Etten, Molecular Oncology Research<br />

Institute & Division of Hematology/Oncology, Tufts<br />

Medical Center, Boston, MA, USA<br />

Since the discovery of the somatic JAK2 V617F mutation in<br />

myeloproliferative neoplasms (MPNs) in 2005, there have<br />

been dramatic advances in our underst<strong>and</strong>ing of the molecular<br />

pathogenesis of the non-CML MPNs. We now appreciate<br />

that JAK2 mutations (either JAK2 V617F or mutations in<br />

exon 12 of JAK2) <strong>are</strong> found in virtually all patients with<br />

polycythemia vera, <strong>and</strong> JAK2 mutation testing has become<br />

a central part of the diagnostic evaluation of patients with<br />

suspected MPN. Yet, there <strong>are</strong> still significant gaps in our<br />

knowledge <strong>and</strong> several central questions that remain unanswered.<br />

One unresolved issue is how one mutation<br />

(JAK2 V617F ) can cause distinct MPN phenotypes (PV, ET,<br />

<strong>and</strong> PMF) in different patients. A partial answer to this question<br />

has been provided by transgenic <strong>and</strong> retroviral mouse<br />

models wherein the JAK2 V617F kinase is expressed in mouse<br />

hematopoietic cells. These models lend experimental support<br />

for the concept that expression of JAK2 V617F at levels<br />

similar to or higher than endogenous JAK2 causes erythrocytosis<br />

whereas lower expression favors thrombocytosis,<br />

but several variables, including origin of JAK2 <strong>and</strong> mouse<br />

strain, confound the picture. Contributions from other somatic<br />

mutations <strong>and</strong> possible germline disease-modifying<br />

alleles <strong>are</strong> other potential mechanisms for phenotypic heterogeneity.<br />

Another question is whether there <strong>are</strong> cooperating<br />

mutations that complement or precede the acquisition of<br />

a JAK2 mutation. Several lines of evidence point to the existence<br />

of one or more predisposing mutations in MPNs:<br />

families with occurrence of several different MPNs in multiple<br />

members, patients (mainly ET) with clonal hematopoiesis<br />

whose JAK2 V617F allele burden is less than 50%,<br />

JAK2 V617F patients with other somatic mutations (eg, del20q)<br />

in JAK2 +/+ cells, <strong>and</strong> JAK2 V617F patients who progress to<br />

AML where the blasts lack the JAK2 V617F mutation. C<strong>and</strong>idate<br />

gene <strong>and</strong> genome-wide SNP analyses have identified<br />

correlations with disease phenotype <strong>and</strong> discovered a remarkable<br />

association of an intronic JAK2 allele (rs10974944 or<br />

46/1) that significantly predisposes carriers to develop<br />

JAK2 V617F –positive MPN, where the mutation arises preferentially<br />

on the same JAK2 gene as the SNP. The mechanism<br />

(»hypermutability« vs »fertile ground«) through which this<br />

occurs is unclear, but the same haplotype predisposes to<br />

MPL mutations in PMF. Recent studies have also identified<br />

somatic mutations that can occur in concert with JAK2/<br />

MPL mutations: TET2 loss-of-function mutations in 10%,<br />

ASXL1 mutations in 8–10%, IDH1/2 mutations in 3–5%,<br />

<strong>and</strong> EZH2 mutations in 10–15% of MPN patients. The same<br />

mutations can be found in MDS <strong>and</strong> AML patients, indicating<br />

that these alleles can be associated with other myeloid<br />

neoplasms. Clonal analysis of malignant hematopoietic<br />

colonies demonstrates that these mutations can either precede<br />

or develop after a JAK2 mutation; in some cases<br />

(TET2, IDH1/2) they occur most commonly at time of progression<br />

to AML. Emerging functional data <strong>and</strong> genetic<br />

studies in mice suggest these mutations affect the epigenetic<br />

state of the leukemic cells. Results from ongoing genomic<br />

deep sequencing of MPN patient genomes <strong>are</strong> expected to<br />

shed additional light on the pathogenesis of JAK2 V617F –negative<br />

MPN. Finally, the question remains as to the overall<br />

utility of JAK2 as a target for therapy in the MPNs. Clinical<br />

results with small molecule JAK2 inhibitors (summarized<br />

by Dr. Verstovsek in this Symposium) demonstrate that all<br />

six of the inhibitors in clinical trials <strong>are</strong> effective at reducing<br />

spleen size, reversing elevated proinflammatory cytokines,<br />

<strong>and</strong> improving quality of life <strong>and</strong> performance status in<br />

MPN patients, independent of JAK2 mutation status. However,<br />

the effects of these agents on JAK2 allele burden, myelofibrosis,<br />

cytopenias <strong>and</strong> transfusion requirements, thrombohemorrhagic<br />

complications, <strong>and</strong> risk of progression to<br />

AML in MPN <strong>are</strong> less clear. There appears to be a fundamental<br />

difference between JAK2 <strong>and</strong> ABL1 as therapeutic<br />

targets in MPN <strong>and</strong> CML, respectively. Recent translational<br />

research has begun to identify some of the mechanisms involved,<br />

including Epo-dependent sensitivity of malignant<br />

JAK2 V617F –positive hematopoietic progenitors to JAK2 inhibitors,<br />

<strong>and</strong> a possible compensatory role of other JAK<br />

family members.<br />

OP21 JAK2 inhibitors<br />

Srdjan Verstovsek, MD Anderson Cancer Center, Leukemia<br />

Department, Houston, Texas, USA<br />

Myelofibrosis: Myelofibrosis (MF) is a Philadelphia chromosome-negative<br />

myeloproliferative neoplasm (MPN) of<br />

the bone marrow. This life-threatening disease is characterized<br />

by splenomegaly <strong>and</strong> burdensome symptoms, including<br />

fatigue, night sweats, pruritus, abdominal pain, <strong>and</strong><br />

cachexia. 1 The annual incidence of MF is about 1.0 per<br />

100,000 population, <strong>and</strong> patients have a median life expectancy<br />

of 2 to 11 years due to complications of progressive<br />

31


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

splenomegaly, hepatomegaly with liver failure, portal hypertension,<br />

gastrointestinal bleeding, pulmonary hypertension,<br />

infections, <strong>and</strong> malnutrition. 3 Allogeneic stem cell<br />

transplantation (alloSCT) offers the only potentially cure<br />

for MF, but is associated with a high rate of transplant-related<br />

mortality. 4 Other treatments for MF provide only temporary<br />

<strong>and</strong>/or suboptimal relief of constitutional symptoms,<br />

anemia, <strong>and</strong> splenomegaly, <strong>and</strong> do not generally change the<br />

natural progression of the disease.<br />

JAK2 as a Therapeutic Target in MF: Dysregulation of<br />

the JAK2-STAT pathway is a central pathogenic component<br />

in MF. Some clinical features of MF (anemia, splenomegaly,<br />

risk of transformation to acute myeloid leukemia) have<br />

been related to JAK2 V617F mutation or allele burden; 5–7 however,<br />

the gain-of-function JAK2 V617F mutation is found in<br />

only about 50% of MF patients. Other mutations that result<br />

in activation of JAK-STAT signaling have been detected,<br />

including mutations in exon 12 of the JAK2 gene 8 <strong>and</strong> the<br />

MPL515 mutation in the thrombopoietin receptor. 9<br />

JAK2 Inhibitors in MF: The initial reports of the JAK2 V617F<br />

mutation in MPNs led to the development of ruxolitinib as<br />

the first potent, selective, oral JAK1 <strong>and</strong> JAK2 inhibitor for<br />

clinical use in MF patients. Currently, there <strong>are</strong> several<br />

JAK2 inhibitors in development. Ruxolitinib is currently the<br />

only JAK inhibitor to complete phase III trials in MF.<br />

The safety <strong>and</strong> efficacy of ruxolitinib were evaluated in a<br />

phase I/II study in 153 high- or intermediate-risk patients<br />

with primary MF, post-polycythemia vera (PV) MF, or postessential<br />

thrombocythemia (ET) MF. The median duration<br />

of follow-up was 14.7 months. 11 Phase I results established<br />

25 mg twice daily as the maximum tolerated dose (MTD),<br />

with thrombocytopenia as the dose-limiting toxicity (DLT).<br />

Phase II data established an optimized dose of 15 mg twice<br />

daily (10 mg twice daily if platelets were 100–200 x 10 9 /L)<br />

with monthly dose escalations up to 25 mg twice daily (if no<br />

response <strong>and</strong> no toxicity were observed). Approximately<br />

half of the patients achieved a ≥ 50% reduction in splenomegaly<br />

by palpation. Magnetic resonance imaging (MRI)<br />

confirmed reductions in spleen size. A 50% reduction in<br />

spleen size by palpation was consistent with a 35% reduction<br />

in volume by MRI. In addition, ruxolitinib improved<br />

debilitating MF-related symptoms, including fatigue, night<br />

sweats, <strong>and</strong> pruritus as assessed by the Myelofibrosis Symptom<br />

Assessment Form (MFSAF). 12 Among biomolecular<br />

parameters associated with disease activity, there was a reduction<br />

in plasma levels of proinflammatory cytokines (IL-<br />

6, TNF-α), an increase in plasma levels of leptin <strong>and</strong> erythropoietin,<br />

<strong>and</strong> a dose- <strong>and</strong> time-dependent reduction in constitutively<br />

phosphorylated STAT3 <strong>and</strong>/or STAT5.<br />

COMFORT-I is a r<strong>and</strong>omized, double-blind, placebocontrolled<br />

phase III trial of ruxolitinib in patients with MF. 13<br />

The primary endpoint was the proportion of patients achieving<br />

a ≥ 35% reduction in spleen volume at 24 weeks of<br />

treatment as assessed by MRI. The study met this endpoint:<br />

41.9% of patients had a ≥ 35% reduction in spleen volume<br />

versus 0.7% in the placebo group (P


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

has been confirmed by long term analysis of a subgroup of<br />

the study group who have now been followed up to 91<br />

months (4). Earlier studies had shown safety <strong>and</strong> efficacy of<br />

anagrelide in younger patients (5,6). Meanwhile, long term<br />

data from registries also covering older patients in Austria<br />

(7), Spain (8), Czech Republic (9) or Germany (10) confirm<br />

that anagrelide is well tolerated <strong>and</strong> effective in reducing<br />

platelets to target levels in patients with ET. Addressing the<br />

special issue of cardiovascular adverse events a recent Italian<br />

study has demonstrated a low impact of this concern (11). In<br />

some patients deterioration of renal function is observed under<br />

long term anagrelide treatment: in such patients comedication<br />

or evidence for transformation into myelofibrosis<br />

have to be checked since CMPN can cause glomerulopathy<br />

(12). Progression of bone marrow fibrosis (which is not present<br />

in patients diagnosed according to the WHO classification)<br />

cannot be halted with anagrelide (13).<br />

Taken together, anagrelide is a valuable drug for the treatment<br />

of patients with ET.<br />

R E F E R E N C E S<br />

1. Tefferi A. Polycythemia vera <strong>and</strong> essential thrombocythemia: 2011 update<br />

on diagnosis, risk stratification <strong>and</strong> management. Amer J Hematol<br />

2011;86:293–301.<br />

2. Petrides PE. Anagrelide: What was new in 2005 <strong>and</strong> 2006? Sem Thrombosis<br />

Research 2006;32:399–408.<br />

3. Petrides PE et al. ANAHYDRET: A European multicenter prospective<br />

phase 3 study: Noninferiority of anagrelide comp<strong>are</strong>d to hydroxyurea<br />

in newly WHO diagnosed ET patients.Haematologica 2009;94:440.<br />

4. Gisslinger H et al., in preparation.<br />

5. Storen EC, Tefferi A. Long term use of anagrelide in young patients<br />

with essential thrombocythemia. Blood 2001;97:863–866.<br />

6. Mazzucconi MG et al. A long term study of young patients with essential<br />

thrombocythemia treated with anagrelide. Haematologica 2004;<br />

89:1306–1313.<br />

7. Steurer M et al. Longterm experience with anagrelide (Thromboreductin®)<br />

in the treatment of essential thrombocythemia: data from an international<br />

regsitry. Blood, ASH abstract 2010;32828.<br />

8. Giralt M et al. Retrospective analysis of the efficacy <strong>and</strong> tolerability of<br />

anagrelide in patients with essential thrombocythemia: Spanish registry<br />

of essential thrombocythemia. Med Clin (Barc) 2009;133:86–90.<br />

9. Penka P et al. Essential thrombocythemia <strong>and</strong> other myeloproliferative<br />

disorders treated with thromboreductin: a report from the data base registry.<br />

Vnitr Lek 2010;56:503–512.<br />

10. Petrides PE, Siegel P. Unpublished.<br />

11. Gugliotta L et al. Low impact of cardiovascular adverse events on<br />

anagrelide treatment discontinuation in a cohort of 232 patients with<br />

essential thrombocythemia. Leukemia Research 2011, online.<br />

12. Siad SM et al. Myeloproliferative neoplasms cause glomerulopathy.<br />

Kidney Int 2011, online.<br />

13. Hultdin M et al. Progression of bone marrow fibrosis in patients with<br />

essential thrombocythemia <strong>and</strong> polycythemia vera during anagrelide<br />

treatment. Med Oncol 2007;24:63–70.<br />

OP23 Treatment of Polycythemia Vera<br />

With Interferon<br />

Richard T. Silver, Weill Cornell Medical College, USA<br />

Certain diseases deserve intense medical scrutiny because<br />

the abnormalities encountered stimulate investigation,<br />

which greatly enhance our underst<strong>and</strong>ing of physiologic<br />

mechanisms. The myeloproliferative disorders, including<br />

polycythemia vera (PV), fall into this category. Two recent<br />

major advances have been made in PV: the discovery of the<br />

JAK2 V617F mutation which is useful for diagnosis <strong>and</strong> as a<br />

marker for treatment effect, <strong>and</strong> the use of interferons as a<br />

therapeutic modality.<br />

Although hydroxyurea remains the most commonly used<br />

cytoreductive treatment for PV, limitations of its use have<br />

not received adequate attention. These include its leukemogenic<br />

effect after 10–15 years of use, development of squamous<br />

cell carcinoma of the skin, skin ulcers, <strong>and</strong> other<br />

forms of oral, hepatic <strong>and</strong> renal toxicity. For the younger<br />

patient, hydroxyurea is not appropriate because of its effect<br />

on embryogenesis.<br />

The use of recombinant interferon (rIFN) as a therapeutic<br />

measure has led to significant improvement in the treatment<br />

of all myeloprolifetrative diseases in the past 20 years, <strong>and</strong><br />

especially polycythemia vera. This has been based on appreciation<br />

that a low dose of rIFN used over a prolonged<br />

period may have a significant therapeutic effect. In our presentation,<br />

we will review some of the salient features pertaining<br />

to clinical use of interferon in PV.<br />

Of the two types of interferon, type I (which includes alpha<br />

<strong>and</strong> beta interferon) <strong>and</strong> type II (which includes gamma<br />

interferon), it is alpha interferon which is primarily used in<br />

hematologic practice. Interferon has a wide range of effects<br />

of interest to the hematologist: It augments the functional<br />

activity of T-cells, macrophages, <strong>and</strong> natural killer cells; it<br />

induces dendritic differentiation; it promotes apoptosis of a<br />

variety of tumor cell types; it suppresses the ability of normal<br />

hematopoietic progenitor cells to form colonies in vitro;<br />

it may stimulate dormant hematopoietic cells to cycle, thus<br />

making them more susceptible to the effect of chemotherapeutic<br />

agents. Interferon also inhibits erythroid progenitors<br />

<strong>and</strong> erythroid burst-forming units in vitro <strong>and</strong> produces<br />

morphologic <strong>and</strong> biochemical changes in megakaryocytes<br />

including megakaryocytic proliferation <strong>and</strong> thrombopoeitin-induced<br />

TPO receptor signaling. Interferon alpha antagonizes<br />

platelet derived growth factor. These reasons provide<br />

a physiologic basis for its use in PV.<br />

Two general types of interferons have been clinically<br />

used. In the past, the largest experience has been with interferon<br />

alpha 2b, but more recently with pegylated interferon<br />

alpha 2a (Peg A). More than 350 patients with PV have been<br />

reported with interferon, the largest number (55) treated<br />

with interferon (rIFN) alpha-2b by Silver. 1 Reduction in<br />

phlebotomy rate occurred in 100% of patients <strong>and</strong> freedom<br />

from phlebotomy in 96%. 1 Kiladjian reported on the effects<br />

of Peg A in 37 patients also with a 100% reduction in phlebotomy<br />

rate <strong>and</strong> 97% freedom from phlebotomy. 2 Qualitative<br />

side effects in general have been the same with either<br />

type of interferon – i.e. headache, malaise, fever, chills, fatigue,<br />

myalgia being the most common but less severe with<br />

Peg A than with rIFNα2b. With Peg A, significant reduction<br />

in JAK2 V617F median allele burden at 24 months has been reported<br />

by Kiladjian (45% to 5%) 2 <strong>and</strong> by Quintás-Cardama<br />

(64% to 12%) 3 but not by Silver who reported that hematologic<br />

response, both complete <strong>and</strong> partial, did not correlate<br />

with the molecular response with either Peg A or rIFNα2b<br />

treated patients. 4 However, drop out rates for the three investigations<br />

related to toxicity were 24%, 10%, <strong>and</strong> 6% respectively.<br />

2,3 Thus, it remains to be determined whether or<br />

33


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

not over the long-term there will be significant difference in<br />

the course of disease between a higher discontinuation rate<br />

<strong>and</strong> a lower JAK2 V617F burden comp<strong>are</strong>d to lower discontinuation<br />

rate <strong>and</strong> a higher molecular burden.<br />

In the interferon trials reported by both Silver <strong>and</strong> Kiladjian,<br />

there was a striking decrease in the number of CNS<br />

vascular events comp<strong>are</strong>d to other published data. Whether<br />

this represents a stricter attention to hematocrit levels as<br />

practiced by both the French <strong>and</strong> American groups or some<br />

unique biologic effect of interferon remains to be determined.<br />

Noteworthy <strong>are</strong> two international trials of the Myeloproliferative<br />

Disease Research Consortium with PEG-interferon<br />

alpha 2a which will open soon. One study comp<strong>are</strong>s Peg<br />

A to hydroxyurea in high risk PV <strong>and</strong> ET patients. The other<br />

uses Peg A as a salvage therapy for PV <strong>and</strong> ET in patients<br />

resistant or intolerant to hydroxyurea. Hopefully, these studies<br />

will provide answers to some of the questions raised in<br />

this presentation.<br />

R E F E R E N C E S<br />

1. Silver RT. Long-term effects of the treatment of polycythemia vera with<br />

recombinant interferon-alpha. Cancer 2006;107:451–458.<br />

2. Kiladjian JJ, Cassinat B, Chevret S, Turlure P, Cambier N, Roussel M, et<br />

al. Pegylated interferon-alfa-2a induces complete hematologic <strong>and</strong> molecular<br />

responses with low toxicity in polycythemia vera. Blood 2008;<br />

112:3065–3072.<br />

3. Quintas-Cardama A, Kantarjian H, Manshouri T, Manshouri T, Luthra<br />

R, Estrov Z, et al. Pegylated interferon alfa-2a yields high rates of hematologic<br />

<strong>and</strong> molecular response in patients with advanced essential<br />

thrombocythemia <strong>and</strong> polycythemia vera. J Clin Oncol. 2009;27:5418–<br />

5424.<br />

4. Silver RT, V<strong>and</strong>ris K, Wang YL, Adriano F, Jones AV, Christos PJ, et al.<br />

JAK2(V617F) allele burden in polycythemia vera correlates with grade<br />

of myelofibrosis, but is not substantially affected by therapy. Leuk Res<br />

2011;35:177–182.<br />

OP24 Allogeneic Hematopoietic Stem Cell<br />

Transplantation for Myelofi brosis<br />

Nicolaus Kröger, Department of Stem Cell<br />

Transplantation, University Hospital Hamburg-Eppendorf,<br />

Hamburg, Germany<br />

Hematopoietic stem cell transplantation offers a curative<br />

therapy for patients with myelofibrosis. Due to toxicity, allografting<br />

following myeloablative regimens is mainly applicable<br />

to young patients. Two larger studies reported a 1<br />

year non-relapse mortality of 27% <strong>and</strong> 31%, resulting in a 5<br />

year estimated survival of 47% <strong>and</strong> 58%. (Guardiola et al.<br />

1999 <strong>and</strong> Deeg et al. 2003). The major risk factors in the<br />

myeloablative studies were advanced age, advanced disease<br />

status according to Lille-score, <strong>and</strong> mismatched unrelated<br />

donors.<br />

With the introduction of dose-reduced conditioning using<br />

busulfan or melphalan with fludarabin, transplantation became<br />

tolerable also in older patients. Improvement in management<br />

of transplant-related complications as well as increasing<br />

numbers of volunteer stem cell donors lead to an<br />

increased use of alternative donors. In the so far largest prospective<br />

multicenter study of the Chronic Leukemia Working<br />

Party of the European Group for Blood <strong>and</strong> Marrow<br />

34<br />

Transplantation (EBMT)103 patients were included <strong>and</strong><br />

transplanted after a dose reduced conditioning consisting of<br />

busulfan (10mg/kg) /fludarabin <strong>and</strong> in vivo T-cell depletion<br />

with ATG. All but 2 patients showed leukocyte <strong>and</strong> platelets<br />

engraftment, <strong>and</strong> the cumulative incidence of TRM at 1 year<br />

was 16% which was significantly higher with HLA-mismatched<br />

donors. At 3 years, the cumulative incidence of<br />

relapse was 22% <strong>and</strong> significantly influenced by the risk<br />

status according to Lille. After a median follow up of 33<br />

months (range, 12–76), five-year estimated OS <strong>and</strong> DFS<br />

was 67% <strong>and</strong> 51% respectively. In the multivariate analysis,<br />

age>55 years <strong>and</strong> HLA-mismatched transplantation significantly<br />

predicted inferior overall survival (Kröger et al.<br />

2009).<br />

At the histomorphological level, a complete or near complete<br />

resolution of bone marrow fibrosis resolution was seen<br />

in 59% of patients by day 100, 90% by day 180, <strong>and</strong> 100%<br />

by one year post-AHSCT<br />

Considering the sufficient evidence available that even in<br />

the case of massive splenomegaly donor cells engraftment<br />

<strong>and</strong> full donor chimerism <strong>are</strong> achievable, together with the<br />

significant risks associated with splenectomy pre-transplant<br />

such as surgical risk, delayed immunreconstitution after<br />

AHSCT <strong>and</strong> possibly the reported increased relapse, splenectomy<br />

is not generally recommended in transplant-eligible<br />

patients.<br />

Through the discovery of new disease specific mutations,<br />

such as JAK2V617F or MPL, close monitoring of residual<br />

disease became feasible in many patients <strong>and</strong> can be used a<br />

guide for adoptive immunotherapy such as donor lymphocyte<br />

infusion (Achalby et al 2010) achieving of new transplant-derived<br />

models to estimate risk status <strong>and</strong> possible<br />

outcome in every individual patient <strong>and</strong> help in therapy decision<br />

<strong>and</strong> determine optimal timing of stem cell transplantation.<br />

Such a tool may optimally include not only clinicomorphological<br />

characteristics but also other potentially relevant<br />

factors such as cytogenetics <strong>and</strong> novel molecular<br />

markers.<br />

R E F E R E N C E S<br />

1. Alchalby H, Badbaran A, Zabelina T et al. (2010) Impact of JAK2V617F<br />

mutation status, allele burden, <strong>and</strong> clearance after allogeneic stem cell<br />

transplantation for myelofibrosis. Blood 116:3572–3581.<br />

2. Deeg HJ, Gooley TA, Flowers ME et al. (2003) Allogeneic hematopoietic<br />

stem cell transplantation for myelofibrosis. Blood 102:3912–3918.<br />

3. Guardiola P, Anderson JE, B<strong>and</strong>ini GW et al. (1999) Allogeneic stem<br />

cell transplantation for agnogenic myeloid metaplasia: a European<br />

Group for Blood <strong>and</strong> Marrow Transplantation, Societe Francaise de<br />

Greffe de Moelle, Gruppo Italiano per il Trapianto del Midollo Osseo,<br />

<strong>and</strong> Fred Hutchinson Cancer Research Center Collaborative Study.<br />

Blood 93:2831–2838.<br />

4. Kröger N, Alchalby H, Klyuchnikov E (2009a) et al. JAK2-V617F-triggered<br />

preemptive <strong>and</strong> salvage adoptive immunotherapy with donor-lymphocyte<br />

infusion in patients with myelofibrosis after allogeneic stem<br />

cell transplantation. Blood 113:1866–1868.<br />

5. Kröger N, Holler E, Kobbe G et al. (2009 b) Allogeneic stem cell transplantation<br />

after reduced-intensity conditioning in patients with myelofibrosis:<br />

a prospective, multicenter study of the Chronic Leukemia Working<br />

Party of the European Group for Blood <strong>and</strong> Marrow Transplantation<br />

(EBMT). Blood 114:5264–5270.<br />

6. Kröger N, Badbaran A, Holler E et al. (2007 a) Monitoring of the JAK2-<br />

V617F mutation by highly sensitive quantitative real-time PCR after allogeneic<br />

stem cell transplantation in patients with myelofibrosis. Blood<br />

109:1316–1321.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

OP25 Jumping 1q Translocations <strong>are</strong> Clonal<br />

Markers of Myeloid Malignancies<br />

Associated with Transformation to AML<br />

Vesna Najfeld, J. Tripodi, A. Scalise, L. Silverman, R.T.<br />

Silver, S. Fruchtman, R. Hoffman, Tumor Cytogenetics<br />

Laboratory, Departments of Pathology <strong>and</strong> Medicine, The<br />

Myeloproliferative Disorders Program, The Tisch Cancer<br />

Institute, The Mount Sinai School of Medicine, New York,<br />

NY, Department of Med., Cornell-Weill School of<br />

Medicine, New York, USA<br />

Jumping translocations (JT) <strong>are</strong> r<strong>are</strong> cytogenetic phenomenon<br />

resulting from a part of one chromosome translocating<br />

to several recipient chromosomes, creating multiple<br />

related clones within a single patient. Due to their rarity in<br />

myeloid malignancies it is uncertain whether this abnormality<br />

contributes to disease progression. We retrospectively<br />

investigated 1qJT in 23 patients with myeloproliferative neoplasm<br />

(MPN) <strong>and</strong> myelodysplastic syndrome (MDS) who<br />

were evaluated in the Tumor Cytogenetics Laboratory at the<br />

Mount Sinai School of Medicine between 1984 <strong>and</strong> 2009.<br />

Diagnoses were made according to the World Health Organization<br />

classification criteria. Table1 outlines the characteristics<br />

of the 23 patients with 1qJT. Among 512 studied<br />

patients with MPN (polycythemia vera [PV]=361, primary<br />

myelofibrosis [PMF]=151), 3 (PV=2, PMF=1) were identified<br />

who had 1qJT at diagnosis.<br />

Table 1. Characteristics of 23 patients with Jumping 1q translocations<br />

MPN<br />

No. (%)<br />

MDS<br />

No. (%)<br />

Baseline Analysis<br />

Patients 7 16<br />

Patients with 1qJT 3 5<br />

Normal Karyotype 0 (0) 8 (50)<br />

Abnormal Karyotype 7 (100) 8 (50)<br />

Transformation to AML<br />

Sequential Analysis<br />

2 (29) 4 (25)<br />

Patients 5* 12**<br />

Specimens 24 101<br />

Normal Karyotype 0 (0) 7 (67)<br />

Transformation to AML 4 10<br />

Mean Time from 1qJT to AML in Months (Range) 8 (0–16) 9 (0–42)<br />

Mean Follow up in Months (Range) 65 (2–132) 34 (8–78)<br />

Mean Time (Months) to develop 1qJT (Range) 47 (0–109) 21 (0–58)<br />

* One patient (#3) had 1qJT at diagnosis <strong>and</strong> had one follow-up study<br />

** One patient (#20) had 1qJT at diagnosis <strong>and</strong> 4 sequential studies up to 18<br />

months after the intial study<br />

Additionally four of 165 (PV=96, PMF=69) patients with<br />

MPN-acquired 1qJT over a period of 11 years (the overall<br />

incidence was 1.3% or 4.2% among cytogenetically abnormal).<br />

The mean follow up of the five patients was 65 months<br />

<strong>and</strong> the mean time from diagnosis to the development of<br />

1qJT was 47 months. Four of these five patients transformed<br />

to acute myeloid leukemia (AML) within 8 months of acquiring<br />

1qJT. Patient 6 is particularly informative, he ini-<br />

tially had i(9)(p10) but developed a der(6)t(1;6)(q21;q27)<br />

subclone in two of 18 cells after 6 years. Over time the subclone<br />

involved 100% of cells. The patient was treated solely<br />

with interferon. The most recent analysis revealed 1qJT to<br />

chromosomes 6, 7, Y as well as dup(1)(q21). The initial cytogenetic<br />

analysis of 16 patients with MDS/AML revealed a<br />

normal karyotype in 8 patients <strong>and</strong> an abnormal karyotype<br />

in the other eight patients. Out of 532 studied between 1986<br />

<strong>and</strong> 2009 we identified five patients who had 1qJT at diagnosis.<br />

Four of these five patients presented at the time of<br />

transformation of MDS to AML <strong>and</strong> the fifth patient was<br />

studied after autologous stem cell transplantation. Follow<br />

up studies revealed 11 patients developed 1qJT (the overall<br />

incidence was 3%, or 6% among cytogenetically abnormal).<br />

The mean time to develop 1qJT was 34 months. Among<br />

eight patients with an initial normal karyotype the average<br />

time to acquire 1qJT was 27 months. Once 1qJT were documented<br />

they persisted for an average 9 months in 14 of<br />

(88%) before evolving into AML. Our data do not provide<br />

any common pattern as to the role of the partner chromosomes<br />

in 1qJT. However, 81% of recipient breakpoints<br />

were localized in pericentric regions while the remaining<br />

19% were telomeric fusions. The presence of 1qJT at diagnosis<br />

or the subsequent acquisition of 1qJT appears to be<br />

associated with imminent transformation to AML in patients<br />

with MPN <strong>and</strong> MDS after an average of 8 <strong>and</strong> 9<br />

months, respectively. Once acquired, the prognosis tended<br />

to be dependent on a copy number of 1q indicating that<br />

1qJT was associated with both disease progression <strong>and</strong> poor<br />

prognosis.This work was supported in part by the grant P01<br />

CA1 108671 from the National Institutes of Health.<br />

OP26 Prognostic Tools in CML<br />

Rudiger Hehlmann, Universität Heidelberg, Germany<br />

Prognosis of CML has improved due to the availability of<br />

several new first-line treatment options (optimized imatinib,<br />

2 nd generation TKI first-line). Prediction of prognosis in<br />

CML is currently estimated by criteria at diagnosis <strong>and</strong> by<br />

response to treatment.<br />

At diagnosis, Sokal <strong>and</strong> EUTO scores have been commonly<br />

used for prognostic prediction in the pre-imatinib<br />

era. Since imatinib has changed the natural course of the<br />

disease, a new score (EUTOS score) has been developed<br />

from imatinib treated patients which uses only two variables<br />

(spleen size <strong>and</strong> percentage of basophils in the peripheral<br />

blood) <strong>and</strong> separates a high from a low risk group. The<br />

EUTOS score has a predictive value which is about 1. 4<br />

times more powerful than that of the Sokal score (Hasford<br />

et al, Blood prepublished May 2, 2011).<br />

Under therapy, the gold st<strong>and</strong>ard for prognostic prediction<br />

has been complete cytogenetic remission (CCR). Recently<br />

also major molecular remission (MMR) at 12 months<br />

after start of treatment has been associated with better<br />

survival (Hehlmann et al., JCO 29: 1634–1642, 2011).<br />

MMR is a prerequisite for complete molecular remission<br />

which is considered a necessary step on the path to cure <strong>and</strong><br />

for discontinuation of imatinib. Molecular analysis can be<br />

35


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

performed with peripheral blood <strong>and</strong> avoids bone marrow<br />

puncture.<br />

A further advance in prognostic prediction is early molecular<br />

response at the 10% level (international scale, IS)<br />

after 3 months of treatment (Hanfstein et al., Blood 116,<br />

2010, ASH, abstr. 360). Patients with a BCR-ABL level less<br />

than 10% (IS) after 3 months have a significantly better progression<br />

free survival (96% vs. 83% after 8 years).<br />

It is concluded that molecular analyses performed by harmonized<br />

laboratories with st<strong>and</strong>ardized methods might replace<br />

cytogenetics as the new method of choice for prognostic<br />

prediction at least partially in the future (Müller at al.,<br />

Leukemia 23: 1957–1963, 2009).<br />

OP27 Imatinib, still the best fi rst line therapy?<br />

David Marin, Department of Hematology, Imperial College<br />

London, London, United Kingdom<br />

R<strong>are</strong>ly in the history of medicine has any new drug constituted<br />

such a radical advance in the therapy of a particular<br />

disease as has imatinib for chronic myeloid leukemia<br />

(CML). Imatinib has been the st<strong>and</strong>ard front line therapy for<br />

CML for nearly 10 years <strong>and</strong> although it may still be too<br />

early to appreciate the full extent to which imatinib improves<br />

the survival of CML patients. However at 5 years<br />

more than one third of the patients have ab<strong>and</strong>oned the medication<br />

on account of side effects, lack of efficacy or progression.<br />

Furthermore, imatinib may soon be displaced as<br />

front line therapy by nilotinib, dasatinib or bosutinib. Small<br />

phase II studies using nilotinib or dasatinib as initial therapy<br />

in CML patients have shown that these drugs induce very<br />

deep responses in the majority of patients with virtually nobody<br />

failing to achieve complete cytogenetic response.<br />

Three large phase III studies comparing nilotinib, dasatinib<br />

<strong>and</strong> bosutinib with imatinib <strong>are</strong> ongoing with very promising<br />

early results. It is likely that at least two of these drugs<br />

will prove to be superior to imatinib; if that is the case, it<br />

will be difficult to decide what the initial therapy for CML<br />

patients should be as the there <strong>are</strong> no ongoing studies comparing<br />

the three new drugs between each other.<br />

OP28 Outcome of Treatment of CML with 2 nd<br />

Generation Tyrosine Kinase Inhibitors<br />

After Imatinib Failure<br />

Elias Jabbour, Jorge Cortes, Hagop Kantarjian,<br />

Department of Leukemia, The University of Texas MD<br />

Anderson Cancer Center, Houston, Texas<br />

Abstract: Although imatinib revolutionized the management<br />

of chronic myeloid leukemia (CML), recent data indicate<br />

a transformation in the treatment approach likely in the<br />

near future. For patients who fail with st<strong>and</strong>ard-dose imatinib<br />

therapy, imatinib dose escalation is a second-line option.<br />

However, high-dose imatinib is not an appropriate approach<br />

for patients experiencing drug toxicity, <strong>and</strong> there<br />

remain questions over the durability of responses achieved<br />

with this strategy. Alternative second-line options include<br />

36<br />

the newer tyrosine kinase inhibitors (TKIs) like dasatinib<br />

<strong>and</strong> nilotinib. A substantial amount of long-term data for<br />

these agents is available. Although both <strong>are</strong> potent <strong>and</strong> specific<br />

BCR-ABL TKIs, dasatinib <strong>and</strong> nilotinib exhibit unique<br />

pharmacological profiles <strong>and</strong> response patterns relative to<br />

different patient characteristics, such as disease stage <strong>and</strong><br />

BCR-ABL mutational status. The superiority of second<br />

generation TKIs over imatinib in newly diagnosed disease<br />

has been recognized as well. They induce high <strong>and</strong> rapid<br />

rates of cytogenetic <strong>and</strong> molecular response, with less progression<br />

to advanced forms of disease in comparison with<br />

imatinib. Several investigational agents specific for those<br />

patients with the T315I mutation remain under evaluation.<br />

The future of CML therapy may include early use of these<br />

potent agents to help more patients achieve molecular remission<br />

<strong>and</strong> potentially be a path to a CML cure.<br />

Introduction<br />

Chronic myelogenous leukemia (CML) is a myeloproliferative<br />

neoplasm with an incidence of 1–2 cases per 100,000<br />

adults, with increasing incidence with age <strong>and</strong> a slight male<br />

predominance of 1.3:1. Thirty percent of patients <strong>are</strong> 60<br />

years old or older, with the median age at presentation being<br />

45–50 years old1 . The pathogenesis of CML involves a specific<br />

genetic abnormality involving the fusion of the Abelson<br />

murine leukemia (ABL) gene on chromosome 9 with<br />

the breakpoint cluster region (BCR) gene on chromosome<br />

222 . In 95% of cases, this fusion is the result of a balanced<br />

translocation between chromosomes 9 <strong>and</strong> 22 [t(9;22)(q34;<br />

q11)], termed the Philadelphia chromosome3,4 . The remaining<br />

5% of cases <strong>are</strong> a result of other genetic abnormalities<br />

that result in the fusion of these same genes4 . The resultant<br />

BCR-ABL fusion protein contains the constitutively active<br />

tyrosine kinase region of ABL that produces a proliferative<br />

signal that regulates growth <strong>and</strong> replication through downstream<br />

pathways such as RAS5 , RAF6 , JUN kinase7 , MYC8 ,<br />

<strong>and</strong> STAT 9–11 . This contributes to leukemogenesis by creating<br />

a cytokine-independent cell cycle with aberrant apoptotic<br />

signals in response to cytokine withdrawal2 .<br />

The causal role played by BCR-ABL in leukemogenesis,<br />

specifically as it relates to CML, has led to the development<br />

of tyrosine kinase inhibitors (TKIs) <strong>and</strong> represents the bulk<br />

of pharmaceutical research performed in this <strong>are</strong>na within<br />

the last decade. TKIs mitigate the oncogenic activity of the<br />

BCR-ABL protein through competitive inhibition at the<br />

ATP-binding site, leading to the inhibition of phosphorylation<br />

of proteins involved in BCR-ABL mediated intracellular<br />

signal transduction, <strong>and</strong> thereby causing the arrest of<br />

growth <strong>and</strong> apoptosis in CML cells12–14 .<br />

Imatinib mesylate (Gleevec; Novartis Pharmaceuticals<br />

Corporation, NJ, USA) was the first of these drugs to receive<br />

the approval by the United States Food <strong>and</strong> Drug Administration<br />

(FDA) in 2001 for the treatment of patients<br />

with CML-CP at a dose of 400 mg/day; as well as for advanced<br />

phase (CML-AP) <strong>and</strong> blast phase (CML-BP) diease<br />

at a higher dose of 600 mg/day15–17 . The advent of imatinib<br />

revolutionized the treatment of CML <strong>and</strong> represented improved<br />

outcome over the previously well established gold<br />

st<strong>and</strong>ard of interferon-alpha (IFN-a) based treatments18 . As


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

a result, patients with CML in the chronic phase (CML-CP)<br />

can expect a lifespan of approximately 25 years <strong>and</strong> even<br />

beyond, transforming CML from a disease which was once<br />

fatal, with a 40% 7-year survival to one that is chronic, with<br />

a 90% 7-year survival.<br />

Outcome with imatinib<br />

The l<strong>and</strong>mark phase III IRIS trial (International R<strong>and</strong>omized<br />

Study of Interferon versus STI571) was the first to<br />

clearly demonstrate the superiority of imatinib treatment in<br />

CML-CP as comp<strong>are</strong>d to IFN-a. Complete cytogenetic response<br />

(CCyR) rates were increased in the imatinib group<br />

over the IFN group (68% vs. 7% at 12 months; <strong>and</strong> 76.2%<br />

vs. 14.5% at 18 months) 19,20 . Imatinib produced durable<br />

CCyR with rates of 87% <strong>and</strong> 85% at 5 years <strong>and</strong> 8 years<br />

respectively21,22 . Major molecular response (MMR) rates,<br />

defined as a 3 log reduction in BCR-ABL transcripts19 , or<br />

BCR-ABL:control gene ratio of 0.10 or less23 , were also improved<br />

(39% vs. 2% at 12 months; p


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

of patients who appear to have rising levels of BCR-ABL<br />

transcripts while maintaining a cytogenetic response. This<br />

may indicate an increased risk for development of mutations<br />

or failure to therapy, however, this value should be<br />

considered with caution. It is inherently variable due to the<br />

fact that it is an international normalized ratio, <strong>and</strong> there is<br />

significant variability in values reported from different laboratories.<br />

Thus, a small increase in BCR-ABL transcript<br />

levels does not necessarily indicate treatment failure or loss<br />

of response. This is supported by the 3-year follow up of the<br />

IRIS trial in which CCyR, regardless of MMR, was associated<br />

with improved OS 25 . If a patient who has previously<br />

achieved a MMR is noted to have a 1 log, or 5 fold increase<br />

in BCR-ABL transcripts, the value should be repeated <strong>and</strong><br />

confirmed in 1–3 months to evaluate for loss of cytogenetic<br />

response. Patient compliance should be evaluated in both<br />

cases, <strong>and</strong> mutation analysis should be considered if the patient<br />

has been identified as having a suboptimal response<br />

within the first year of therapy or has had a loss of response<br />

at any time 29 .<br />

Resistance to imatinib<br />

Despite the success of imatinib, a substantial proportion,<br />

33%, of patients have been shown to be resistant to therapy30,31<br />

. This resistance can be classified into two types: primary<br />

<strong>and</strong> secondary. Failure to achieve any of the ELN outlined<br />

criteria for response in accordance with the duration of<br />

therapy is considered primarily resistant; those who have<br />

previously achieved <strong>and</strong> subsequently lost their response in<br />

accordance with those guidelines <strong>are</strong> considered secondarily<br />

resistant18,29,30 . Primary resistance can be further divided<br />

into primary hematologic resistance, which occurs in 2%–<br />

4% of cases who fail to normalize peripheral counts; or primary<br />

cytogenetic resistance, which is more common <strong>and</strong><br />

occurs in approximately 15%–25% of patients who fail to<br />

reduce their Ph+ cells to 35% or fewer of those sampled32 .<br />

Causal factors in imatinib resistance can be classified as<br />

either BCR-ABL dependent, or BCR-ABL independent.<br />

Point mutations <strong>are</strong> the most well-characterized BCR-ABL<br />

dependent factors <strong>and</strong> <strong>are</strong> responsible for 35–75% of cases<br />

of resistance30,33–38 . A key component to imatinib resistance<br />

centers around its ability to bind only to the closed, or inactive,<br />

conformation of the ABL kinase, <strong>and</strong> requires a very<br />

specific structural arrangement for entry <strong>and</strong> binding. Point<br />

mutations can cause steric hindrance as well as elimination<br />

of critical molecules required for hydrogen bonding between<br />

imatinib <strong>and</strong> ABL kinase, thus mitigating imatinib’s<br />

efficacy30,38–40 . Examples of such mutations include T315I,<br />

Y253H, <strong>and</strong> F255K, among others38 .<br />

BCR-ABL independent mechanisms of resistance include<br />

amplification of the ABL kinase oncogene in response<br />

to high plasma imatinib levels30,41–44 ; increased efflux of the<br />

drug by increased expression of P-glycoprotein (Pgp) efflux<br />

pumps30,45–47 ; decreased drug uptake secondary to decreased<br />

expression of the drug uptake transporter, hOCT130,48–51 ;<br />

cytoplasmic sequestration of imatinib by increased serum<br />

protein alfa1-acid glycoprotein (AGP) which binds imatinib<br />

<strong>and</strong> impairs subsequent binding to ABL kinase30,52,53,54 ;<br />

low serum drug concentration30 ; <strong>and</strong> alternative signaling<br />

38<br />

pathway activation, including Ras/Raf/MEK kinase, STAT,<br />

Erk2, <strong>and</strong> SFK phosphorylation of BCR-ABL, all of which<br />

decrease susceptibility to imatinib 30 .<br />

Adherence is another BCR-ABL independent factor that<br />

has been shown to affect outcome. Rates of daily imatinib<br />

adherence have been estimated to range from 75% to<br />

90% 55–57 with higher adherence rates correlated to improved<br />

outcome. In one study of 87 patients with CML-CP treated<br />

with imatinib 400 mg daily, adherence was shown to correlate<br />

with MMR <strong>and</strong> CMR at 6 years. An adherence rate of<br />

90% or less resulted in MMR in only 28.4% as comp<strong>are</strong>d to<br />

94.5% in patients with greater than 90% adherence rates<br />

(P


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

In another study assessing 84 patients over 61 months<br />

with failure to st<strong>and</strong>ard-dose imatinib, doses were escalated<br />

to 600–800 mg daily. In 21 patients with hematologic failure,<br />

48% achieved a CHR, with only 14% achieving a cytogenetic<br />

response. In 63 patients with cytogenetic failure,<br />

75% responded with CCyR. Two <strong>and</strong> 3-year EFS rates were<br />

57% <strong>and</strong> 47%, respectively; with OS rates of 84% <strong>and</strong> 76%,<br />

respectively 59 . Thus, while dose escalation after failure of<br />

st<strong>and</strong>ard-dose imatinib is an important <strong>and</strong> viable treatment<br />

option, it is likely to be effective only in the subset of patients<br />

with previous cytogenetic responses <strong>and</strong> is not indicated<br />

for patients with intolerance to the drug. Thus, clinical<br />

consideration should be given to 2 nd generation TKIs.<br />

Second generation tyrosine kinase inhibitors<br />

[Table2]<br />

Dasatinib<br />

Dasatinib was approved by the US FDA in 2006 for the<br />

2nd line treatment of CML-CP, -AP, -BP, <strong>and</strong> Philadelphiapositive<br />

acute lymphoblastic leukemia [Ph (+) ALL] with<br />

resistance or intolerance to prior therapy with imatinib60,61 .<br />

Dasatinib is 325 times more potent than imatinib38,62 . Unlike<br />

imatinib, dasatinib binds to both the active <strong>and</strong> inactive<br />

forms of BCR-ABL30 .<br />

Several studies have shown that dasatinib is highly effective<br />

in CML-CP In the START-C trial, an international, multicenter,<br />

phase II trial assessing 387 patients with CML-CP<br />

who had demonstrated imatinib intolerance (n= 99) or resistance<br />

(n=288), patients were treated with dasatinib at an<br />

initial dose of 70 mg BID. Twenty four-month follow-up<br />

data showed overall rates of CyR 62%, CCyR 53%, CHR<br />

91%, <strong>and</strong> MMR 47%. Two-year PFS was 80% <strong>and</strong> OS was<br />

94%. Adverse events included grade 3 neutropenia (50%),<br />

grade 3 thromboyctopenia (49%), <strong>and</strong> grade 3 pleural effusions<br />

(9%). Nine percent of all adverse events were considered<br />

»severe62 .« In another study assessed the efficacy of<br />

dasatinib in 445 imatinib-resistant or intolerant patients<br />

with CML-CP (n=186), CML-AP (n=107), CML-myeloid<br />

blast crisis (n=74), CML-lymphoid blast crisis (n=42), or<br />

Ph (+) ALL (n=36). Dasatinib produced MCyR rates of<br />

45% <strong>and</strong> CCyR rates of 33% in patients with CML-CP, <strong>and</strong><br />

major hematologic response rates in advanced stages of disease,<br />

including 59% in CML-AP; 39% in CML myeloid<br />

blast crisis; 31% in lymphoid blast crisis; <strong>and</strong> 42% in Ph (+)<br />

ALL 61 .<br />

The dose optimization study CA180034 r<strong>and</strong>omized 622<br />

patients with imatinib resistant or intolerant CML-CP to 4<br />

dasatinib treatment arms, including 100 mg daily, 50 mg<br />

BID, 140 mg daily, <strong>and</strong> 70 mg BID. Dasatinib dosed at 100<br />

mg daily produced similar CyR <strong>and</strong> PFS rates as other dosing<br />

schedules, with significantly fewer occurrences of grade<br />

3–4 neutropenia, thrombocytopenia, anemia, <strong>and</strong> pleural effusions.<br />

There were also fewer treatment interruptions, reductions,<br />

<strong>and</strong> discontinuations at 100 mg daily as comp<strong>are</strong>d<br />

to the other treatment groups 63 .<br />

Further evaluation of dasatinib therapy in imatinib-resistant<br />

patients with mutations by meta-analysis of 3 dasatinib<br />

studies, including the START-C, START-R, <strong>and</strong> CA180-034<br />

dose optimization study, revealed that rates of response with<br />

dasatinib <strong>are</strong> favorable in a variety of genetic mutations including<br />

E255K/V (CCyR 38%), L248V (CCyR 40%), <strong>and</strong><br />

G250E (CCyR 33%). CCyR rates were lower in F317L(7%),<br />

Q252H (17%), L384M(0%), <strong>and</strong> V299L (0%). Patients with<br />

T315I mutations did not respond to dasatinib 64 .<br />

Dasatinib has also been evaluated in patients with imatinib-resistant<br />

or intolerant CML-AP. One study assessed<br />

174 such patients treated with dasatinib 70 mg BID. Major<br />

hematologic response rates were 64%, CHR rates were<br />

45%; MCyR rates were 39% <strong>and</strong> CCyRrates were 32%.<br />

PFS at 12 months was 66% <strong>and</strong> OS was 82%. Adverse<br />

events included grade 3–4 thrombocytopenia (82%), neutropenia<br />

(76%), <strong>and</strong> diarrhea (52%, grade 3 or 4 in 8%) 65 .<br />

Appropriate dosing in CML-AP was evaluated in another<br />

study, in which 317 patients were r<strong>and</strong>omized to receive dasatinib<br />

at 140 mg daily or 70 mg BID. The results at 2 years<br />

after initiation of treatment demonstrated similar efficancy<br />

in rates of hematologic <strong>and</strong> cytogenetic responses, as well<br />

as in PFS <strong>and</strong> OS rates. Dasatinib 140 mg once daily did<br />

demonstrate an improved safety profile with statistically<br />

Table 2. Response to second generation tyrosine kinase inhibitors (dasatinib, nilotinib <strong>and</strong> bosutinib) in patients who <strong>are</strong> imatinib-resistant or intolerant<br />

in chronic phase, accelerated phase <strong>and</strong> blast phase<br />

Response<br />

Dasatinib<br />

Percent Response<br />

Nilotinib Bosutinib<br />

CP<br />

N=387<br />

AP<br />

n=174<br />

MyBP<br />

n=109<br />

LyBP<br />

n=48<br />

CP<br />

n=321<br />

AP<br />

N=137<br />

MyBP<br />

N=105<br />

LyBP<br />

N=31<br />

CP N=146<br />

AP<br />

N=51<br />

BP<br />

N=38<br />

Median follow-up (mo) 15 14 12+ 12+ 24 9 3 3 7 6 3<br />

% Resistant to imatinib 74 93 91 88 70 80 82 82 69 NR* NR*<br />

% Hematologic Response – 79 50 40 94 56 22 19 85 54 36<br />

CHR 91 45 27 29 76 31 11 13 81 54 36<br />

NEL – 19 7 6 – 12 1 0 – 0 0<br />

% Cytogenetic Response NR 44 36 52 NR NR NR NR – NR NR<br />

Complete 49 32 26 46 46 20 29 32 34 27 35<br />

Partial 11 7 7 6 15 12 10 16 13 20 18<br />

% Survival (at 12 months) 96 (15) 82 (12) 50 (12) 50 (5) 87 (24) 67 (24) 42 (12) 42 (12) 98 (12) 60 (12) 50 (10)<br />

Legend: CP: chronic phase; AP: accelerated phase; MyBP: myeloid blast phase; LyBP: lymphoid blast phase, mo: months; CHR: complete hematologic response;<br />

NEL: no evidence of leukemia<br />

39


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

fewer patients experiencing pleural effusions (20% vs. 39%;<br />

P


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

results show achievement of an 85% CHR rate, 28% CyR<br />

rate, 15% MCyR rate, 15% MMR rate, <strong>and</strong> 57% reduction<br />

in the T315I clone. OS was not met in this group of patients.<br />

In CML-AP, 37.5% showed hematologic response with OS<br />

of 18.8 months. In CML-BP, hematologic response was<br />

demonstrated in 30% with OS of 1.8 months 78 .<br />

An update combining both of the above study populations<br />

was presented at ASH 2010. Thirty six of the 93 chronic<br />

phase patients had been treated previously with 3 or more<br />

TKIs. Over a median followup of 7.5 months, 27/36 (75%)<br />

achieved or maintained a CHR <strong>and</strong> 7 (19%) achieved a<br />

major CyR (4 complete <strong>and</strong> 3 partial) with omacetaxine,<br />

with a median duration of at least 4 months. These findings<br />

support the further investigation of this agent in patients<br />

failing other TKI therapy 79 . Grade 3 to 4 hematologic adverse<br />

events included thrombocytopenia (65%), neutropenia<br />

(48%), <strong>and</strong> anemia (40%), febrile neutropenia (12%),<br />

bone marrow failure (12%), pancytopenia (7%) <strong>and</strong> febrile<br />

bone marrow aplasia (6%). Grade 3 to 4 nonhematologic<br />

adverse events included fatigue (6%). Grade 1–2 adverse<br />

events included diarrhea, fatigue, nausea, vomiting, fever,<br />

headache, <strong>and</strong> asthenia 77,78,79 .<br />

Predictors of outcome<br />

The primary goal of therapy for patients with CML is still<br />

achievement of CCyR. Those who achieve this goal, have a<br />

low probability of eventually progressing. Achieving a major<br />

molecular response is desirable as it further improves the<br />

long-term outcome, but patients who have a CCyR <strong>are</strong> not<br />

considered to have failure to imatinib if they do not have a<br />

major molecular response. This is because the difference in<br />

EFS probability is small, although significant. The timing of<br />

this response is also important. Despite initial suggestions<br />

form the IRIS trial that a MMR at 12 months improved<br />

long-term outcome, comp<strong>are</strong>d to CCyR but no MMR, the<br />

8-year follow-up data has shown no difference in outcome<br />

using this hallmark. By 18 months, patients who have a<br />

CCyR <strong>and</strong> MMR have a better probability of EFS than those<br />

with CCyR but no MMR, but the difference is small (95%<br />

vs. 86%), <strong>and</strong> even smaller if considering only transformation<br />

to AP or BP or death as an event (99% vs 96%) 80 .<br />

The recommendations by the ELN have provided a clear<br />

framework for decision making <strong>and</strong> the significance of the<br />

definitions of failure <strong>and</strong> suboptimal response have been<br />

demonstrated in two independent series27,81 . Patients with<br />

suboptimal response to therapy have an inferior outcome,<br />

although the significance appears to be heterogeneous, with<br />

a more profound adverse prognostic effect for patients with<br />

suboptimal response to therapy at earlier time points (6<br />

months) <strong>and</strong> than at later time points (18 months). It is however<br />

reasonable to consider treatment modifications for patients<br />

who have a suboptimal response to therapy. Treatment<br />

guidelines recommend dose escalation of imatinib to 600 or<br />

800 mg/day in cases of suboptimal response38,70 . However, it<br />

should be acknowledged that there is minimal data available<br />

regarding the effectiveness of this approach58,59 . For patients<br />

with clear failure to imatinib therapy, the current approach<br />

is to change therapy to a second generation TKI, although<br />

ASCT is also an option following treatment failure17,82 .<br />

Clinical trial data have confirmed the efficacy of dasatinib<br />

<strong>and</strong> nilotinib in patients with imatinib resistance or intolerance<br />

<strong>and</strong> the superiority of dasatinib versus imatinib dose<br />

escalation following imatinib resistance 70,81,83 . It is possible<br />

that earlier treatment switch to second-line agents, i.e. after<br />

suboptimal response, could result in more favorable longterm<br />

outcomes than with dose-escalated imatinib, but there<br />

<strong>are</strong> currently no clinical data to support this hypothesis.<br />

In addition to efficacy outcomes, mutational data should<br />

be considered when selecting TKI therapy 84 . The BCR-ABL<br />

genotype can be used to guide treatment decisions since it is<br />

a prognostic factor for disease progression 84–86 . Patients with<br />

T315A/I, F317I/L, <strong>and</strong> V299L mutations do not appear to<br />

respond consistently to therapy with dasatinib 86–89 , whereas<br />

patients with the F359C/V substitution do benefit from dasatinib<br />

86 . In an analysis of 1043 patients who underwent<br />

mutational assessment in phase II/III studies in CP CML, 14<br />

patients had baseline F317L mutations, <strong>and</strong> just one patient<br />

had a baseline V299L mutation 89 . It was found that patients<br />

bearing F317L mutations achieved a high CHR rate (93%),<br />

but cytogenetic response rates (MCyR, 14%; CCyR, 7%)<br />

were lower than in patients without these mutations. Importantly,<br />

in patients treated with dasatinib, high response rates<br />

were obtained with the common imatinib-resistant mutations<br />

in Y253, E255 <strong>and</strong> E359 residues.<br />

Nilotinib resistance is associated with mutations in the<br />

T315, Y253, <strong>and</strong> E255 residues 84–86 . Indeed, recently it was<br />

shown that the presence of E255K/V, Y253H, or F359C/V<br />

mutations at baseline <strong>are</strong> independent predictors of worsened<br />

PFS in patients with CP CML 86 . Therefore, dasatinib<br />

therapy may be more appropriate for patients with these<br />

common mutations, whereas nilotinib may be better suited<br />

for those patients with F317L mutations 88 . Although both<br />

dasatinib <strong>and</strong> nilotinib <strong>are</strong> ineffective against T315I BCR-<br />

ABL 85 , this mutation is more likely to affect patients in<br />

the advanced phases of CML 85 . Patients with T315I may<br />

achieve favorable outcomes with therapies other than the<br />

available second-line TKIs, e.g AP24534, omacetaxine, <strong>and</strong><br />

others 90,91 .<br />

A recent study has explored factors that may predict response<br />

<strong>and</strong> outcome in patients with chronic-phase CML<br />

receiving dasatinib or nilotinib after failing imatinib 92,93 .<br />

The analysis included 123 patients in chronic phase post<br />

imatinib failure: 78 treated with dasatinib <strong>and</strong> 45 treated<br />

with nilotinib. Multivariate analysis for predictive factors<br />

was performed for event-free survival, overall survival, <strong>and</strong><br />

12-month major cytogenetic response (MCyR). Investigators<br />

found that event-free survival in response to a second<br />

TKI in CML depends on achieving a prior cytogenetic response<br />

to imatinib <strong>and</strong> on the patient’s performance status.<br />

In the study, patients with performance status ≥ 1 <strong>and</strong> no<br />

prior cytogenetic response to imatinib had a high likelihood<br />

of responding to a second TKI with poor event-free survival<br />

<strong>and</strong> overall survival. Patients with both risk factors had a<br />

24-month overall survival of only 40% comp<strong>are</strong>d with 95%<br />

for patients with neither risk factor. Therefore, patients with<br />

no previous cytogenetic response to imatinib should be offered<br />

additional treatment approaches including allogeneic<br />

stem cell transplantation <strong>and</strong> clinical trials. However, the<br />

41


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

achievement of MCyR with a second TKI by 12 months<br />

may compensate for the presence unfavorable baseline factors<br />

93 .<br />

Safety <strong>and</strong> tolerability <strong>are</strong> also important considerations<br />

in choosing a TKI. The potential impact of the drug’s AE<br />

profile on any of the patient’s preexisting conditions should<br />

be considered in choosing between second generation BCR-<br />

ABL inhibitors. Since pleural effusion is more common for<br />

patients receiving dasatinib therapy, patients with risk factors<br />

for pleural effusion such as a prior cardiac history,<br />

chronic obstructive pulmonary disease <strong>and</strong> hypertension <strong>are</strong><br />

at greater risk for developing these complications 67 . Risk<br />

factors for developing pleural effusions while taking dasatinib<br />

also include disease stage (BC > AP > CP), previous<br />

lung problems such as smoking or infections <strong>and</strong> those patients<br />

maintained on starting doses of dasatinib 94 .<br />

Blood glucose elevations have been observed in 11% of<br />

CML-CP, <strong>and</strong> 4% of CML-AP patients treated with nilotinib<br />

72,95 . While no CML-CP or CML-AP patients required<br />

dose adjustments, dose interruption or discontinued nilotinib<br />

therapy due to elevated blood glucose, preexisting hyperglycemia<br />

should be c<strong>are</strong>fully monitored to ensure that<br />

the condition is not exacerbated by nilotinib treatment. Prior<br />

severe pancreatitis may also be a concern on nilotinib therapy,<br />

<strong>and</strong> patients with a history should be closely monitored<br />

on nilotinib therapy.<br />

Cardiac events, including congestive heart failure, left<br />

ventricular dysfunction <strong>and</strong> QT prolongation have all been<br />

reported with dasatinib <strong>and</strong> nilotinib. Though they occurred<br />

in


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

also significantly improved the rates of CCyR <strong>and</strong> MMR at<br />

24 months. CCyR at 24 months was 87% with 400 mg twice<br />

daily nilotinib comp<strong>are</strong>d with 77% with imatinib (P =<br />

.0018). Likewise, MMR at 24 months was 59% with 400<br />

mg nilotinib vs 37% with imatinib (P < .0001). There were<br />

also significantly fewer progressions to advanced phase <strong>and</strong><br />

blast crisis with nilotinib. Based on these data, nilotinib has<br />

been approved for the frontline therapy of CML. The gap in<br />

efficacy in favor of nilotinib has persisted over time <strong>and</strong> it<br />

appears that nilotinib may improve both short-term <strong>and</strong><br />

long-term outcomes comp<strong>are</strong>d with imatinib 100 .<br />

In the ENESTnd trial, nilotinib was also shown to be safe<br />

<strong>and</strong> well-tolerated with no increase in side-effects comp<strong>are</strong>d<br />

with imatinib. By contrast, treatment-related gastrointestinal<br />

toxicity <strong>and</strong> fluid retention of all grades were more frequent<br />

with imatinib than they were in either nilotinib<br />

arm 100 .<br />

Dasatinib in the frontline setting [Table 4]<br />

Cortes et al at MD Anderson Cancer Center published a<br />

study in which 50 patients with newly diagnosed CML-CP<br />

were r<strong>and</strong>omized to dasatinib 100 mg once daily or 50 mg<br />

twice daily in the frontline setting. Median follow up was 24<br />

months. Of the 50 patients enrolled in the study, 49 (98%)<br />

attained a CCyR <strong>and</strong> 41 (82%) attained a MMR. Ninety<br />

four percent of patients attained CCyR by 6 months. There<br />

was no difference in response rate by treatment arm. The<br />

projected event-free survival rate at 24 months was 88%.<br />

Grade 3/ 4 hematologic toxicities included neutropenia <strong>and</strong><br />

thrombocytopenia, <strong>and</strong> occurred in 21% <strong>and</strong> 10% of patients,<br />

respectively. Nonhematologic toxicity was usually<br />

grade 1 to 2. There was no significant difference in toxicity<br />

between the two arms, <strong>and</strong> the actual median dose at 12<br />

months was 100 mg (range, 20 to 100 mg). These results<br />

lead to the conclusion that the management of CML-CP in<br />

the frontline setting with dasatinib is an effective approach,<br />

yielding high rates of CCyR <strong>and</strong> MMR101 .<br />

The DASISION trial, an international, multicenter, r<strong>and</strong>omized<br />

phase III trial on frontline dasatinib therapy, r<strong>and</strong>omized<br />

519 patients to receive dasatinib 100 mg once<br />

daily or imatinib 400 mg once daily. Findings from this trial<br />

were recently published102 . The current data presented at<br />

ASH 2010 represented an 18-month follow-up showing that<br />

dasatinib was superior to imatinib with respect to the primary<br />

endpoint, the rate of CCyR. The likelihood of achieving<br />

a CCyR at any time was higher with dasatinib vs imatinib<br />

(85% vs 80%; hazard ratio [HR] = 1.5; P < .0001) 103 .<br />

Table 4. Response rates with frontline dasatinib.<br />

Study<br />

Number of<br />

CCyR (%) MMR (%)<br />

patients<br />

PFS (%) OS (%)<br />

MDACC 50 98 82 – –<br />

DASISION 259 78 57<br />

94.9<br />

(18 months)<br />

96<br />

(18 months)<br />

S0325 123 82 59<br />

99<br />

(12 months)<br />

100<br />

(12 months)<br />

CCyR: complete cytogenetic response, MMR: major molecular response,<br />

PFS: progression free survival, OS: overall survival, MDACC: MD Anderson<br />

Cancer Center<br />

*cCCyR (confirmed complete cytogenetic response) is reported for DASI-<br />

SION study.<br />

In addition, the secondary endpoint, the rate of MMR,<br />

was also significantly improved with dasatinib comp<strong>are</strong>d<br />

with imatinib. The likelihood of achieving MMR at any<br />

time with dasatinib was significantly higher than with imatinib<br />

(57% vs 41%; HR = 1.8; P < .0001). Based on these<br />

data, dasatinib was approved by the US Food <strong>and</strong> Drug Administration<br />

as a st<strong>and</strong>ard of c<strong>are</strong> for CML patients. Dasatinib<br />

was also shown to be well tolerated, with low rates of<br />

grade 3/4 hematologic <strong>and</strong> nonhematologic toxicity, as well<br />

as a low rate of discontinuation due to adverse events although,<br />

pleural effusion occurred only in patients treated<br />

with dasatinib (in 12% of 258 patients, nearly all grade 1<br />

<strong>and</strong> 2) 102,103 .<br />

Another study by the Southwest Oncology Group (S0325)<br />

comp<strong>are</strong>d dasatinib 100 mg to imatinib 400 mg in newly<br />

diagnosed CML in chronic phase. The study showed that<br />

dasatinib was associated with greater MMR comp<strong>are</strong>d with<br />

imatinib at 12 months. However, the 12-month rates of complete<br />

hematologic response (CHR), CCyR, OS, or PFS were<br />

comparable between treatment groups. The lack of statistical<br />

significance in rates of CCyR was likely due to the fact<br />

that patients who were not evaluable were considered nonresponders<br />

104 .<br />

Bosutinib in the frontline setting<br />

The BELA trial, an international, mutlicenter, open-label<br />

phase III trial, comp<strong>are</strong>s bosutinib with imatinib in the<br />

frontline setting using CCyR as a primary endpoint <strong>and</strong><br />

MMR as a secondary endpoint105 . In this study, non-evaluable<br />

patients were considered non-responders. Consequently,<br />

bosutinib was associated with a similar rate of CCyR<br />

comp<strong>are</strong>d with imatinib at 12 months (70% vs 68%; P =<br />

.601), <strong>and</strong> the primary endpoint was not met. In evaluable<br />

patients only, bosutinib did induce a significantly higher<br />

rate of CCyR comp<strong>are</strong>d with imatinib (78% vs 68%; P =<br />

.026). In addition, there was a significantly higher MMR<br />

with bosutinib in both the intent-to-treat <strong>and</strong> evaluable populations<br />

(intent-to-treat patients: 39% vs 26%, P = .002;<br />

evaluable patients: 43% vs 27%, P < .001). More follow-up<br />

on these studies is needed to determine whether these responses<br />

<strong>are</strong> sustained <strong>and</strong> whether bosutinib is truly superior<br />

to imatinib.<br />

Conclusion<br />

The treatment of CML has progressed significantly since<br />

the approval of imatinib in 2001. With the advent of TKIs,<br />

the focus of the scientific community has shifted to the molecular<br />

management of disease, which has had a considerable<br />

impact on the approach to cancer treatment to date.<br />

Imatinib has transformed CML from an immediately lifethreatening<br />

disease to one that is treatable with daily oral<br />

medication that makes it possible to improve both overall<br />

survival <strong>and</strong> maintain quality of life. However, increasing<br />

incidence of imatinib-resistance <strong>and</strong> intolerance necessitates<br />

the development of alternative therapies. There is a<br />

substantial amount of data supporting the improvement in<br />

outcome in long term follow up of the 2nd generation TKIs,<br />

nilotinib <strong>and</strong> dasatinib. Improving outcome in the frontline<br />

setting should be the direction of further scientific research,<br />

43


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

including the implementation of 2 nd generation TKIs as<br />

frontline therapy.<br />

Despite extraordinary progress, a true cure for CML is<br />

not generally achieved by Abl kinase inhibitors. TKIs <strong>are</strong><br />

potent inhibitors of Bcr-Abl kinases (among others), resulting<br />

in rapid reduction of the majority of cells carrying the<br />

Ph chromosomal marker. However, suppression of Abldriven<br />

hematopoiesis may be insufficient to eradicate quiescent<br />

stem cells. Studies assessing the combination of TKIs<br />

with promising agents <strong>are</strong> ongoing. These combinations include<br />

TKI <strong>and</strong> hedgehog inhibitors, omacetaxine, vaccines,<br />

<strong>and</strong> hypomethylatings agents. If successful, this strategy<br />

could lead to a safe <strong>and</strong> permanent discontinuation of therapy<br />

in patients with a good response. The impact of using<br />

more potent agents in the frontline setting on the potential to<br />

discontinue TKI therapy remains to be determined. The future<br />

of CML therapy may include early use of these potent<br />

agents, perhaps in combination with new molecules, to help<br />

more patients achieve CMR, which could lead to therapy<br />

discontinuation <strong>and</strong> cure.<br />

R E F E R E N C E S<br />

1. Faderl S, Kantarjian HM, Talpaz M. Chronic myelogenous leukemia:<br />

update on biology <strong>and</strong> treatment. Oncology (Williston Park) 1999;2:<br />

169–810.<br />

2. Schiffer CA. BCR-ABL tyrosine kinase inhibitors for chronic myelogenous<br />

leukemia. N Engl J Med 2007;357:258–265.<br />

3. Ramch<strong>and</strong>ren R, Schiffer CA. Dasatinib in the treatment of imatinib<br />

refractory chronic myeloid leukemia. Biologics: Targets & Therapy<br />

2009;3:205–214.<br />

4. Sawyers CL. Chronic myeloid leukemia. N Engl J Med 1999;340:<br />

1330–1340.<br />

5. M<strong>and</strong>anas RA, Leibowitz DS, Gh<strong>are</strong>hbaghi K, et al. Role of p21 RAS<br />

in p210 BCR-ABL transformation of murine myeloid cells. Blood<br />

1993;82:1838–47.<br />

6. Okuda K, Matulonis U, Salgia R, Kanakura Y, Druker B, Griffin JD.<br />

Factor independence of human myeloid leukemia cell lines is associated<br />

with increased phosphorylation of the proto-oncogene Raf-1. Exp<br />

Hematol 1994;22:1111–1117.<br />

7. Raitano AB, Halper JR, Hambuch TM, Sawyers CL. The BCR-ABL<br />

leukemia oncogene activates Jun kinase <strong>and</strong> requires Jun for transformation.<br />

Proc Natl Acad Sci USA 1995;92:11746–11750.<br />

8. Sawyers CL, Callahan W, Witte ON. Dominant negative MYC blocks<br />

transformation by ABL oncogenes. Cell 1992;70:901–910.<br />

9. Shuai K, Halpern J, Ten Hoeve J, Rao X, Sawyers CL. Constitutive<br />

activation of STAT5 by the BCR-ABL oncogene in chronic myelogenous<br />

leukemia. Oncogene 1996;13:247–254.<br />

10. Carlesso N, Frank DA, Griffin JD. Tyrosyl phosphorylation <strong>and</strong> DNA<br />

binding activity of signal transducers <strong>and</strong> activators of transcription<br />

(STAT) proteins in hematopoietic cell lines transformed by BCR/ABL.<br />

J Exp Med 1996;183:811–820.<br />

11. Ilaria FL Jr, Van Etten RA. P210 <strong>and</strong> P190 (BCR/ABL) induce the tyrosine<br />

phosphorylation <strong>and</strong> DNA binding activity of multiple specific<br />

STAT family members. J Biol Chem 1996;271:31704–31710.<br />

12. Wong SF. New dosing schedules of dasatinib for CML <strong>and</strong> adverse<br />

event management. J Hematol Oncol 2009;2:10–18.<br />

13. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy <strong>and</strong> safety of a specific<br />

inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.<br />

N Engl J Med 2001;344:1031–1037.<br />

14. Kantarjian H, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant<br />

CML <strong>and</strong> Philadelphia chromosome-positive ALL. N Engl J Med 2006:<br />

354:2542–2551.<br />

15. Johnson JR, Bross P, Cohen M, et al. Approval summary: imatinib mesylate<br />

capsules for treatment of adult patients with newly diagnosed<br />

Philadelphia chromosome-positive chronic myelogenous leukemia in<br />

chronic phase. Clin Cancer Res 2003;9:1972–1979.<br />

16. Pal<strong>and</strong>ri F, Iacobucci I, Castagnetti F, et al. Front-line treatment of Philadelphia<br />

positive chronic myeloid leukemia with imatinib <strong>and</strong> interferon-alpha:<br />

5-year outcome. Haematologica 2008;93:770–774.<br />

44<br />

17. Gleevec {R} [package insert]. <strong>East</strong> Hanover, NJ: Novartis Pharmaceuticals<br />

Corporation;2007.<br />

18. Jabbour E, Cortes J, Kantarjian H. Treatment selection after imatinib<br />

resistance in chronic myeloid leukemia. Targ Oncol 2009;4:3–10.<br />

19. Hughes TP, Kaeda J, Bradford S, et al. Frequency of major molecular<br />

responses to imatinib or interferon alfa plus cytarabine in newly diagnosed<br />

chronic myeloid leukemia. N Engl J Med 2003;349:1423–1432.<br />

20. O’Brien SG, Guilhot F, Larson RA, et al. Imatinib comp<strong>are</strong>d with interferon<br />

<strong>and</strong> low-dose cytarabine for newly diagnosed chronic phase<br />

chronic myeloid leukemia. N Engl J Med 2003;348:994–1004.<br />

21. Druker BJ, Guilhot F, O’Brien S, et al. Five-year follow-up of patients<br />

receiving imatinib for chronic myeloid leukemia. N engl J Med 2006;<br />

355:2408–2417.<br />

22. Deininger M, O’Brien SG, Guilhot F, et al. International r<strong>and</strong>omized<br />

study of interferon vs. STI571 (IRIS) 8-year follow up: sustained survival<br />

<strong>and</strong> low risk for progression or events in patients with newly diagnosed<br />

chronic myeloid leukemia in chronic phase (CML-CP) treated<br />

with imatinib. Blood 2009;114:Abstract 1126.<br />

23. Baccarani M, Saglio G, Goldman J, et al. Evolving concepts in the<br />

management of chronic myeloid leukemia: recommendations from an<br />

expert panel on behalf of the European LeukemiaNet. Blood 2006;<br />

108:1809–1820.<br />

24. Roy L, Guilhot J, Krahnke T, et al. Survival advantage from imatinib<br />

comp<strong>are</strong>d with the combination interferon-a plus cytarabine in chronicphase<br />

chronic myelogenous leukemia: historical comparison between<br />

two phase 3 trials. Blood 2006;108:1478–1484.<br />

25. Kantarjian H, Talpaz M, O’Brien S, et al. Survival benefit with imatinib<br />

mesylate versus interferon-a-based regimens in newly diagnosed chronic-phase<br />

chronic myelogenous leukemia. Blood 2006;108:1835–1840.<br />

26. Marin D, Milojkovic D, Olavarria E, et al. European LeukemiaNet criteria<br />

for failure or suboptimal response reliably identify patients with<br />

CML in early chronic phase treated with imatinib whose eventual outcome<br />

is poor. Blood 2008;112:4437–4444.<br />

27. Alvarado Y, Kantarjian H, O’Brien S, et al. Significance of suboptimal<br />

response to imatinib, as defined by the European LeukemiaNet, in the<br />

long-term outcome of patients with early chronic myeloid leukemia in<br />

chronic phase. Cancer 2009;115:3709–3718.<br />

28. Hughes TP, Kaeda J, Bradford S, et al. Frequency of major molecular<br />

responses to imatinib or interferon alfa plus cytarabine in newly diagnosed<br />

chronic myeloid leukemia. N Engl J Med 2003;349:1423–1432.<br />

29. National Comprehensive Cancer network [Internet]. NCCN Clinical<br />

Practice Guidelines in Oncology: chronic myelogenous leukemia.<br />

2010-[cited 2010 May 23]; Available from: http://www.nccn.org/professionals/physician_gls/PDF/cml.pdf.<br />

30. Bixby D, Talpaz M. Mechanisms of resistance to tyrosine kinase inhibitors<br />

in chronic myeloid leukemia <strong>and</strong> recent therapeutic strategies<br />

to overcome resistance. Hematology Am Soc of Hematology Educ Program<br />

2009:461–476.<br />

31. Hochhaus A, O’Brien SG, Guilhot F, et al. Six-year follow-up of patients<br />

receiving imatinib for the first-line treatment of chronic myeloid<br />

leukemia. Leukemia 23:1054–61,2009.<br />

32. Shah NP: Medial management of CML. Hematology Am Soc Hematol<br />

Educ Program:371–5,2007.<br />

33. Lee F, F<strong>and</strong>i A, Voi M. Overcoming kinase resistance in chronic myeloid<br />

leukemia. Int J Biochem Cell boil 2008;40:334–343.<br />

34. Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, Kuriyan<br />

J. Structural mechanism for STI-571 inhibition of Abelson tyrosine<br />

kinase. Science 2000;289:1938–1942.<br />

35. Branford S, Rudzki Z, Walsh S, et al. Detectino of BCR-ABL mutations<br />

in patients with CML treated with imatinib is virtually always<br />

accompanied by clinical resistance, <strong>and</strong> mutations in the ATP phosphate-binding<br />

loop (P-loop) <strong>are</strong> associated with a poor prognosis.<br />

Blood 2003;102:276–283.<br />

36. Jabbour E, Kantarjian H, Jones D, et al. Frequency <strong>and</strong> clinical significance<br />

of BCR-ABL mutations in patients with chronic myeloid leukemia<br />

treated with imatinib mesylate. Leukemia 2006;20:1767–1773.<br />

37. Hochhaus A, Kreil S, Corbin AS, et al. Molecular <strong>and</strong> chromosomal<br />

mechanisms of resistance to imatinib (STI571) therapy. Leukemia<br />

2002;16:2190–2196.<br />

38. O’H<strong>are</strong> T, Walters DK, Stoffregen EP, et al. In vitro activity of BCR-<br />

ABL inhibitors AMN107 <strong>and</strong> BMS-354825 against clinically relevant<br />

imatinib-resistant ABL kinase domain mutants. Cancer Res 2005;65:<br />

4500–4505.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

39. Shah NP. Loss of response to imatinib: mechanisms <strong>and</strong> management.<br />

Hematology Am Soc Hematol Educ Program 2005:183–187.<br />

40. Deininger M, Buchdunger E, Druker BJ. The development of imatinib<br />

as a therapeutic agent for chronic myeloid leukemia. Blood 2005;105:<br />

2640–2653.<br />

41. Weisberg E, Griffin JD. Mechanism of resistance to the ABL tyrosine<br />

kinase inhibitor STI571 in BCR/ABL-transformed hematopoietic cell<br />

lines. Blood 2000;95:3498–3505.<br />

42. le Coutre P, Tassi E, V<strong>are</strong>lla-Garcia M, et al. Induction of resistance to<br />

the Abelson inhibitor STI571 in human leukemia cells through gene<br />

amplification. Blood 2000;95:1758–1766.<br />

43. Mahon FX, Deininger MW, Schultheis B, et al. Selection <strong>and</strong> characterization<br />

of BCR-ABL positive cell lines with differential sensitivity<br />

to the tyrosine kinase inhibitor STI571: diverse mechanisms of resistance.<br />

Blood 2000;96:1070–1079.<br />

44. Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-<br />

571 cancer therapy caused by BCR-ABL gene mutation or amplification.<br />

Science 2001;293:876–8800.<br />

45. Che XF, Nakajima Y, Sumizawa T, et al. reversal of P-glycoprotein mediated<br />

multidrug resistance by a newly synthesized 1,4-benzothiaxipine<br />

derivative, JTV-519. Cancer Lett 2002;187:111–119.<br />

46. Kotaki M, Motoji T, Takanashi M, Wang YH, Mizoguchi H. Anti-proliferative<br />

effect of the ABL tyrosine kinase inhibitor STI571 on the<br />

P-glycoprotein positive K562/ADM cell line. Cancer Lett 2003;199:<br />

61–68.<br />

47. Rumpold H, Wolf AM, Gruenewald K, Gastl G, Gunsilius E, Wolf D.<br />

RNAi-mediated knockdown of P-glycoprotein using a transposonbased<br />

vector system durably restores imatinib sensitivity in imatinibresistant<br />

CML cell lines. Exp Hematol 2005;33:767–775.<br />

48. Thomas J, Wang L, Clark RE, Pirmohamed M. Active transport of imatinib<br />

into <strong>and</strong> out of cells: implications for drug resistance. Blood<br />

2004;104:3739–3745.<br />

49. White DL, Saunders VA, Dang P, et al. OCT-1-mediated influx is a key<br />

determinant of the intracellular uptake of imatinib but not nilotinib<br />

(AMN107): reduced OCT-1 activity is the cause of low in vitro sensitivity<br />

to imatinib. Blood 2006;108:697–704.<br />

50. White DL, Saunders VA, Dang P, et al. CML patients with low OCT-1<br />

activity achieve better molecular responses on high dose imatinib than<br />

on st<strong>and</strong>ard dose. Those with high OCT-1 activity have excellent responses<br />

on either dose: a TOPS correlative study. Blood 2008;112:<br />

Abstract 3187.<br />

51. Wang L, Giannoudis A, Lane S, Williamson P, Pirmohamed M, Clark<br />

RE. Expression of the uptake drug transporter hOCT1 is an important<br />

clinical determinant of the response to imatinib in chronic myeloid leukemia.<br />

Clin Pharmacol Ther 2008;83:258–264.<br />

52. Gambacorti-Passerini C, Barni R, le Coutre P, et al. Role of alpha1 acid<br />

glycoprotein in the in vivo resistance of human BCR-ABL(+) leukemic<br />

cells to the ABL inhibitor STI571. J Natl Cancer Inst 2000;92:<br />

1641–1650.<br />

53. Gambacorti-Passerini C, Zucchetti M, Russo D, et al. Alpha1 acid glycoprotein<br />

binds to imatinib (STI571) <strong>and</strong> substantially alters its pharmacokinetics<br />

in chronic myeloid leukemia patients. Clin Cancer Res<br />

2003;9:625–632.<br />

54. Widmer N, Decosterd LA, Csajka C, et al. Population pharmacokinetics<br />

of imatinib <strong>and</strong> the role of alpha-acid glycoprotein. Br J Clin Pharmacol<br />

2006;62:97–112.<br />

55. Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical factor<br />

for achieving molecular responses in patients with chronic myeloid leukemia<br />

who achieve complete cytogenetic responses on imatinib. J Clin<br />

Oncol 2010;28:2381–2388.<br />

56. Darkow T, Henk HJ, Thomas SK, et al. Treatment interruptions <strong>and</strong><br />

on-adherence with imatinib <strong>and</strong> associated healthc<strong>are</strong> costs: a retrospective<br />

analysis among managed c<strong>are</strong> patients with chronic myelogenous<br />

leukaemia. Pharmacoeconomics 2007;25:481–496.<br />

57. Noens L, van Lierde MA, De Bock R, et al. Prevalence, determinants,<br />

<strong>and</strong> outcomes of nonadherence to imatinib therapy in patients with<br />

chronic myeloid leukemia: The ADAGIO study. Blood 2009;113:<br />

5401–5411.<br />

58. Kantarjian HM, Larson RA, Guilhot F, O’Brien SG, et al. Efficacy of<br />

imatinib dose escalation in patients with chronic myeloid leukemia in<br />

chronic phase. Cancer 2009;115:551–60.<br />

59. Jabbour E, Kantarjian H, O’Brien S, et al. Imatinib mesylate dose escalation<br />

is associated with durable responses in patients with chronic myeloid<br />

leukemia post cytogenetic failure on st<strong>and</strong>ard-dose imatinib<br />

therapy. Blood 2008;113:2154–2160.<br />

60. Sprycel{R} (dasatinib) [package insert].2007.<br />

61. Brave M, Goodman V, Kaminskas E, et al. Sprycel for chronic myeloid<br />

leukemia <strong>and</strong> Philadelphia chromosome-positive acute lymphoblastic<br />

leukemia resistant to or intolerant of imatinib mesylate. Clin Cancer<br />

Res 2008;14:352–359.<br />

62. Mauro MJ, Baccarani F, Cervantes JH, et al. Dasatinib 2-year efficacy<br />

in patients with chronic-phase chronic myelogenous leukemia (CML_<br />

CP) with resistance or intolerance to imatinib (START-C). J Clin Oncol<br />

2009 ASCO Annual Meeting Abstracts; 26:Abstract 7009.<br />

63. Shah NP, Kim DW, Kantarjian HM, et al. Dasatinib 50 mg or 70 mg<br />

BID comp<strong>are</strong>d to 100 mg or 140 mg QD in patients with CML in<br />

chronic phase (CP) who <strong>are</strong> resistant or intolerant to imatinib: One-year<br />

results of CA180034. J Clin Oncol 2007ASCO Annual Meeting<br />

Abstracts;25(18S):Abstract 7004.<br />

64. Muller MC, Cortes JE, Kim DW, et al. Dasatinib treatment of chronicphase<br />

chronic myeloid leukemia: analysis of responses according to<br />

preexisting BCR-ABL mutations. Blood 2009;114:4944–4953.<br />

65. Apperley JF, Cortes JE, Kim DW, et al. Dasatinib in the treatment of<br />

chronic myeloid leukemia in accelerated phase after imatinib failure:<br />

the START-A trial. J Clin Oncol 2009;27:3472–3479.<br />

66. Kantarjian H, Cortes J, Kim DW, et al. Phase 3 study of dasatinib 140<br />

mg once daily versus 70 mg twice daily in patients with chronic myeloid<br />

leukemia in accelerated phase resistant or intolerant to imatinib:<br />

15-month median follow-up. Blood 2009;113:6322–6329.<br />

67. Quintas-Cardama A, Kantarjian H, O’Brien S, et al. Pleural effusion in<br />

patients with chronic myelogenous leukemia treated with dasatinib after<br />

imatinib failure. J Clin Oncol 2007;25:3908–3914.<br />

68. Quintas-Cardama A, Han X, Kantarjian H, Cortes J. Tyrosine kinase<br />

inhibitor-induced platelet dysfunction in patients with chronic myeloid<br />

leukemia. Blood 2009;114:261–263.<br />

69. Kantarjian H, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant<br />

CML <strong>and</strong> Philadelphia chromosome-positive ALL. N Engl J Med 2006;<br />

354:2542–2551.<br />

70. Deininger MW. Nilotinib. Clin Cancer Res 2008;14:4027–4031.<br />

71. Deininger M, Buchdunger E, Druker BJ. The development of imatinib<br />

as a therapeutic agent for chronic myeloid leukemia. Blood 2005;105:<br />

2640–2653.<br />

72. Hochhaus A, Giles F, Apperley J, et al. Nilotinib in chronic myeloid<br />

leukemia patients in accelerated phase (CML-AP) with imatinib resistance<br />

or intolerance: 24-month follow-up results of a phase 2 study.<br />

Haematologica 2009;94(suppl.2):256. Abstract:0631.<br />

73. Carpiuc KT, Stephens JM, Liou SY, Botteman MF. Incidence of grade<br />

3/4 adverse events in imatinib resistant/intolerant chronic phase CML<br />

(CP-CML): A comparison of nilotinib <strong>and</strong> dasatinib. J Clin Oncol<br />

2007;25:Abstract 17501.<br />

74. Jabbour E, le Coutre P, Baccarani M, et al. Nilotinib is associated with<br />

minimal cross intolerance to imatinib in patients with imatinib-intolerant<br />

chronic myelogenous leukemia (CML) in chronic phase (CP).<br />

Blood 2009;25:Abstract 7039.<br />

75. Cortes J, Kantarjian HM, Kim DW, et al. Efficacy <strong>and</strong> safety of bosutinib<br />

(SKI-606) in patients with chronic phase (CP) Ph+ chronic myelogenous<br />

leukemia (CML) with resistance or intolerance to imatinib.<br />

Blood 2008;112:Abstract 1098.<br />

76. Cortes J, Talpaz M, Deininger M, et al. A phase 1 trial of oral AP24534<br />

in patients with refractory chronic myeloid leukemia <strong>and</strong> other hematologic<br />

malignancies: frist results of safety <strong>and</strong> clinical activity against<br />

T315I <strong>and</strong> resistant mutations. Blood2009;114:Abstract 643.<br />

77. Cortes-Franco J, Raghunadharao D, Parikh P, et al. Safety <strong>and</strong> efficacy<br />

of subcutaneous-administered omacetaxine mepesuccinate in chronic<br />

myeloid leukemia (CML) patients who <strong>are</strong> resistant or intolerant to two<br />

or more tyrosine kinase inhibitors – results of a multicenter phase 2/3<br />

study. Blood 2009;114:Abstract 861.<br />

78. Cortes-Franco J, Khoury HJ, Emmanuel F, et al. Safety <strong>and</strong> efficacy of<br />

subcutaneous-administered omacetaxine mepesuccinate in imatinib-resistant<br />

chronic myeloid leukemia (CML) patients who harbor the BCR-<br />

ABL T315I mutation – results of an ongoing multicenter phase 2/3<br />

study. Blood 2009;114:Abstract 644.<br />

79. Cortes JE, Wetzler M, Lipton J, et al. Subcutaneous omacetaxine (OM)<br />

treatment of chronic phase (cp) chronic myeloid leukemia (CML) patients<br />

following multiple tyrosine kinase inhibitor (TKI) Failure. Program<br />

<strong>and</strong> abstracts of the 52nd American Society of Hematology Annual<br />

Meeting <strong>and</strong> Exposition; December 4–7, 2010; Orl<strong>and</strong>o, Florida.<br />

Abstract 2290.<br />

80. O’Brien SG, Guilhot F, Goldman JM, et al. International r<strong>and</strong>omized<br />

study of interferon versus STI571 (IRIS) 7-Year follow-up: sustained<br />

45


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

survival, low rate of transformation <strong>and</strong> increased rate of major molecular<br />

response (MMR) in patients (pts) with newly diagnosed chronic<br />

myeloid leukemia in chronic phase (CML-CP) treated with imatinib<br />

(IM) [abstract]. Blood. 2008;112(suppl). Abstract 186.<br />

81. Kantarjian H, Pasquinin R, Levy V, et al. Dasatinib or high-dose imatinib<br />

for chronic-phase chronic myeloid leukemia resistant to imatinib<br />

at a dose of 400 to 600 milligrams daily: two-year follow-up of a r<strong>and</strong>omized<br />

phase 2 study (START-R). Cancer 2009 115:4136–4147.<br />

82. Kantarjian H, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant<br />

CML <strong>and</strong> Philadelphia chromosome–positive ALL. N Engl J Med<br />

2006;354:2542–2551.<br />

83. Hochhaus A, Kantarjian HM, Baccarani M, et al. Dasatinib induces<br />

notable hematologic <strong>and</strong> cytogenetic responses in chronic-phase chronic<br />

myeloid leukemia after failure of imatinib therapy. Blood 2007;109:<br />

2303–9.<br />

84. Cortes J, Jabbour E, Kantarjian H, et al. Dynamics of BCR-ABL kinase<br />

domain mutations in chronic myeloid leukemia after sequential treatment<br />

with multiple tyrosine kinase inhibitors. Blood. 2007;110:4005–4011.<br />

85. Jabbour E, Kantarjian H, Jones D, et al. Characteristics <strong>and</strong> outcomes<br />

of patients with chronic myeloid leukemia <strong>and</strong> T315I mutation following<br />

failure of imatinib mesylate therapy. Blood. 2008;112:53–55.<br />

86. Jabbour E, Jones D, Kantarjian HM, et al. Long-term outcome of patients<br />

with chronic myeloid leukemia treated with second-generation<br />

tyrosine kinase inhibitors after imatinib failure is predicted by the in<br />

vitro sensitivity of BCR-ABL kinase domain mutations. Blood. 2009;<br />

114:2037–2043.<br />

87. Soverini S, Martinelli G, Colarossi S, et al. Presence or the emergence<br />

of a F317L BCR-ABL mutation may be associated with resistance to<br />

dasatinib in Philadelphia chromosome–positive leukemia. Blood. 2006<br />

(B);24:e51–e52.<br />

88. Jabbour E, Kantarjian HM, Jones D, et al. Characteristics <strong>and</strong> outcome<br />

of chronic myeloid leukemia patients with F317L BCR-ABL kinase<br />

domain mutation after therapy with tyrosine kinase inhibitors. Blood.<br />

2008(B);112:4839–4842.<br />

89. Müller MC, Cortes JE, Kim D-W, et al. Dasatinib treatment of chronic<br />

phase chronic myeloid leukemia: analysis of responses according to<br />

pre-existing BCR-ABL mutations. Blood 2009;epub ahead of print.<br />

90. Cortes J, Talpaz M, Deininger M, et al. A Phase 1 trial of oral AP24534<br />

in patients with refractory chronic myeloid leukemia <strong>and</strong> other hematologic<br />

malignancies: first results of safety <strong>and</strong> clinical activity against<br />

T315I <strong>and</strong> resistant mutations. Blood 2009;114:Abstract 643.<br />

91. Cortes-Franco J, Khoury HJ, Nicolini FE, et al. Safety <strong>and</strong> efficacy of<br />

subcutaneous-administered omacetaxine mepesuccinate in imatinib-resistant<br />

chronic myeloid leukemia (CML) patients who harbor the Bcr-<br />

Abl T315I mutation-results of an ongoing multicenter phase 2/3 study.<br />

Blood 2009;114:Abstract 644.<br />

92. Kantarjian HM, Jabbour E, Giles FJ, et al. Prognostic factors for progression-free<br />

survival in patients with imatinib-resistant or –intolerant<br />

chronic myeloid leukemia in chronic phase (CML-CP) treated with nilotinib<br />

based on 24 month data. Blood 2009(D);113:3298.<br />

93. Jabbour E, Kantarjian H, O’Brien S, et al. Predictive factors for response<br />

<strong>and</strong> outcome in patients (pts) treated with second generation<br />

tyrosine kinase inhibitors (2-TKI) for chronic myeloid leukemia in<br />

chronic phase (CML-CP) post imatinib failure. Blood. 2009;114:210.<br />

Abstract 509.<br />

94. Lipton JH, Sriharsha L, Bogomilsky S, Casciaro L, Keating A, Messner<br />

H, et al. Pleural effusions in patients treated with dasatinib: Results<br />

from two institutions, risk factors <strong>and</strong> management. Journal of Clinical<br />

Oncology 2007; 25: 680s.<br />

95. Kantarjian H, Giles F, Bhalla K, et al. Update On Imatinib-Resistant<br />

Chronic Myeloid Leukemia Patients in Chronic Phase (CML-CP) On<br />

Nilotinib Therapy at 24 Months: Clinical Response, Safety, <strong>and</strong> Long-<br />

Term Outcomes. Blood (ASH Annual Meeting Abstracts), Nov 2009;<br />

114:1129.<br />

96. Cortes JE, Baccarani M, Guilhot F, et al. Phase III, r<strong>and</strong>omized, openlabel<br />

study of daily imatinib mesylate 400 mg versus 800 mg in patients<br />

with newly diagnosed, previously untreated chronic myeloid leukemia<br />

in chronic phase using molecular end points: tyrosine kinase inhibitor<br />

optimization <strong>and</strong> selectivity study J Clin Oncol. 2010 ;28:424–430.<br />

97. Guilhot. R<strong>and</strong>omized Comparison of Imatinib Versus Imatinib Combination<br />

Therapies in Newly Diagnosed Chronic Myeloid Leukaemia<br />

(CML) Patients in Chronic Phase (CP): First Results of the Phase III<br />

(SPIRIT) Trial from the French CML Group (FI LMC). Blood (ASH<br />

Annual Meeting Abstracts) 2008 112: Abstract 183<br />

46<br />

98. Cortes JE, Jones D, O’Brien et al. Niltonib as front-line therapy for<br />

patients with chronic myeloid lekemia in early chronic phase. J Clin<br />

Oncol 28:392–397. 2010.<br />

99. Rosti G, Castagnetti F, Gugliotta G. Excellent outcomes at 3 years<br />

with Nilotinib 800 mg daily in early chronic phase, Ph(+) chronic<br />

myeloid leukemia (CML): Results of a phase 2 GIMEMA CML WP<br />

clinical trial. Blood (ASH Annual Meeting Abstracts) 2010 116: Abstract<br />

359.<br />

100. Hughes TP, Hochhaus A, Saglio G, et al. ENESTnd update: Continued<br />

superiority of nilotinib versus imatinib in patients with newly diagnosed<br />

chronic myeloid leukemia in chronic phase (CML-CP).<br />

Blood (Annual abstract )Abstract 207<br />

101. Cortes JE, Jones D, O’Brien S, et al. Results of dasatinib therapy in<br />

patients with early chronic-phase chronic myeloid leukemia. J Clin<br />

Oncol 28:398–404.2010.<br />

102. Kantarjian H, Shah NP, Hochhaus A, et al. Dasatinib versus imatinib<br />

in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl<br />

J Med. 2010;362:2260–2270.<br />

103. Shah N, Kantarjian HM, Hochhaus A, et al. Dasatinib versus imatinib<br />

in patients with newly diagnosed chronic myeloid leukemia in chronic<br />

phase (CML-CP) in the DASISION trial: 18-month follow-up. Program<br />

<strong>and</strong> abstracts of the 52nd American Society of Hematology Annual<br />

Meeting <strong>and</strong> Exposition; December 4–7, 2010; Orl<strong>and</strong>o, Florida.<br />

Abstract 206.<br />

104. Jerald P. Radich, Kenneth J. Kopecky, <strong>and</strong> Suzanne Kamel-Reid, et al.<br />

A R<strong>and</strong>omized Phase II Trial of Dasatinib 100 Mg Vs Imatinib 400<br />

Mg In Newly Diagnosed Chronic Myeloid Leukemia In Chronic<br />

Phase (CML-CP): The S0325 Intergroup Trial. Blood (ASH Annual<br />

Meeting Abstracts), Nov 2010; 116: LBA-6.<br />

105. Gambacorti-Passerini C, Kim DW, Kantarjian HM, et al. An ongoing<br />

phase 3 study of bosutinib (SKI-606) versus imatinib in patients with<br />

newly diagnosed chronic phase chronic myeloid leukemia. Program<br />

<strong>and</strong> abstracts of the 52nd American Society of Hematology Annual<br />

Meeting <strong>and</strong> Exposition; December 4–7, 2010; Orl<strong>and</strong>o, Florida. Abstract<br />

208.<br />

OP29 Second line TKI – the best fi rst line therapy?<br />

Gianantonio Rosti, (Italy)<br />

Abstract not submitted.<br />

OP30 How to monitor CML therapy<br />

Elias Jabbour, Department of Leukemia, The University<br />

of Texas M. D. Anderson Cancer Center, Houston, Texas,<br />

USA [see OP28]<br />

OP31 Stem cell transplants for chronic phase<br />

CML<br />

OP32 Pathophysiology of Myeloma<br />

Christoph J. Heuck, University of Arkansas for Medical<br />

Sciences, Myeloma Institute for Research <strong>and</strong> Therapy,<br />

Little Rock, AR, USA<br />

Muchof the recent progress in the treatment of multiple<br />

myeloma has been attributed to the introduction of several<br />

novel agents which, when combined with st<strong>and</strong>ard cytotoxic<br />

agents <strong>and</strong> each other, have imparted a high frequency of<br />

clinical responses. We have previously reported on the superior<br />

survival outcomes in Total Therapy 3A (TT3A), with<br />

added bortezomib (Bz), comp<strong>are</strong>d with Total Therapy 2<br />

(TT2),which r<strong>and</strong>omized patients to a control arm or an ex-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

perimental thalidomide arm of a multi-agent chemotherapy<br />

<strong>and</strong> t<strong>and</strong>em autograft-supported high-dose melphalan program.<br />

The major advance with TT3A was observed in the<br />

85% of patients presenting with gene expression profiling<br />

(GEP)-defined low-risk myeloma <strong>and</strong>. As part of TT3A,<br />

pharmacogenomic investigations of Bz, using GEP analysis<br />

pre- <strong>and</strong> post-48 hr test-dosing, were performed in an attempt<br />

to further delineate low- versus high-risk disease.<br />

Pharmacogenomic investigations after test-dose applications<br />

of Bz have revealed that high expression levels of proteasome<br />

genes <strong>are</strong> linked to inferior prognosis in both of our<br />

TT3 protocols. The GEP80 model discriminated outcomes<br />

whether applied to post-Bz or baseline samples. These findings,<br />

observed in TT3A, were validated in TT3B <strong>and</strong> also<br />

applied to both arms of TT2 (control <strong>and</strong> thalidomide).<br />

Employing post-Bz <strong>and</strong> baseline samples in TT3, the<br />

GEP80 model provided segregation of high-risk subsets of<br />

9% <strong>and</strong> 3%, respectively, in the GEP70 low-risk group <strong>and</strong><br />

low-risk subsets of 41% <strong>and</strong> 55%, respectively, in the<br />

GEP70 high-risk setting. In TT2, the GEP80 model failed to<br />

discern a low-risk subset among the patients with GEP70<br />

high-risk disease, <strong>and</strong> the GEP80 model discriminated only<br />

one patient with high-risk status among those identified to<br />

have GEP70 low-risk disease.<br />

We found a significant upregulation in the expression of<br />

proteasome genes following a 48-hour Bz test dose. PSMD4<br />

was only one of three genes common to both GEP70 <strong>and</strong><br />

GEP80 models. PSMD4 <strong>and</strong> other proteasome genes were<br />

uniquely upregulated by Bz but not by dexamethasone, immunomodulatory<br />

agents, <strong>and</strong> melphalan. Prognostically,<br />

GEP80(BL)-defined high-risk status was the sole genetic<br />

parameter that survived multivariate PFS <strong>and</strong> OS models,<br />

along with low albumin <strong>and</strong> high LDH, although all the<br />

other st<strong>and</strong>ard <strong>and</strong> both cytogenetic <strong>and</strong> molecular genetic<br />

variables also contributed when examined alone.<br />

We have previously reported that both the copy number<br />

<strong>and</strong> the percentage of cells with amp1q21 invariably increase<br />

when comparing samples obtained at diagnosis <strong>and</strong><br />

at relapse. Genes residing on chromosome 1q21 contribute<br />

critically to the high-risk designation in the GEP70 model.<br />

Consistent with this, we have noted that the GEP70 score<br />

inevitably increases at relapse <strong>and</strong> a shift from low to high<br />

risk imparts a significantly shorter post-relapse survival.<br />

OP33 Bortezomib in the Treatment of Multiple<br />

Myeloma<br />

Joan Bladé, Institut of Hematology <strong>and</strong> Oncology,<br />

Department of Hematology Hospital Clinic, Barcelona,<br />

Spain<br />

Bortezomib is a first-in-class selective proteasome inhibitor<br />

approved for the treatment of relapsed <strong>and</strong> refractory<br />

multiple myeloma (MM) as well as in first-line therapy in<br />

combination with melphalan <strong>and</strong> prednisone (MPV) for patients<br />

not eligible for high-dose therapy/autologous stem<br />

cell transplantation (ASCT). In vitro, bortezomib induces<br />

apoptosis of myeloma cells <strong>and</strong> inhibits cell adhesion within<br />

the bone marrow microenvironment. Preclinical <strong>and</strong> clinical<br />

data have shown that bortezomib enhances sensitivity <strong>and</strong><br />

can reverse resistance to st<strong>and</strong>ard therapeutic agents used<br />

in MM.<br />

The first phase II trial of bortezomib (SUMMIT) in patients<br />

with MM who had received at least 3 lines of therapy<br />

showed an overall response rate of 35% including 10% of<br />

CR or nCR. The second pivotal phase II trial (CREST), also<br />

performed in patients with relapsed/refractory myeloma<br />

showed that a reduced dose of 1 mg/m 2 instead of the usual<br />

dose of 1.3 mg/m 2 induced a response rate of 33% with<br />

lower toxicity. A r<strong>and</strong>omized trial comparing bortezomib<br />

versus high-dose dexamethasone (APEX) showed that bortezomib<br />

was superior in RR, PFS <strong>and</strong> OS. A large phase 3<br />

trial of bortezomib plus pegylated liposomal doxorubicin<br />

versus bortezomib alone resulted in a significantly superior<br />

CR plus VGPR rate, PFS <strong>and</strong> OS in favour of the combination.<br />

The subcutaneous administration of bortezomib has<br />

shown similar efficacy with less peripheral neuropathy than<br />

the i.v. formulation. Bortezomib can be effective in patients<br />

with extramedullary disease <strong>and</strong> can partially overcome the<br />

poor prognosis of high-risk cytogenetics. Patients who have<br />

responded to bortezomib with no significant peripheral neuropathy<br />

<strong>and</strong> have been for longer than 6 months off therapy<br />

should be re-treated with the same drug.<br />

In a large phase III (VISTA) trial of newly diagnosed patients<br />

non-eligible for ASCT the association of melphalanprednisone-bortezomib<br />

(MPV) was superior to MP in RR<br />

(71 vs. 36%), CR rate (30 vs. 4%), PFS (median, 24 vs.<br />

16%) <strong>and</strong> OS (82.6 vs. 69.5%). For patients with renal failure<br />

the best initial treatment regimen seems to be the association<br />

of bortezomib/dexamethasone with or without doxorubicin.<br />

In patients eligible for ASCT, the induction with<br />

botezomib/dexamethasone results in pre-transplant RR of<br />

65% including 12% of immunofixation negative CR. The<br />

RR post-transplant is about 90% with 33% CR. The addition<br />

of a third drug, either doxorubicin (PAD, VDD) or thalidomide<br />

(VTD), results in an increased pre-transplant CR<br />

rate from 20 to 35% <strong>and</strong> a post-transplant CR up to 50%. In<br />

consequence, a pre-transplant induction regimen should today<br />

include bortezomib <strong>and</strong> dexamethasone plus a third<br />

agent, preferably an IMiD or a doxorubicin formulation.<br />

It has been shown for the first time outside of the allogeneic<br />

setting, that 18% of patients in at least VGPR after<br />

ASCT achieved molecular remission by qualitative <strong>and</strong><br />

quantitative PCR with intensification therapy with VTD. In<br />

addition, post-transplant intensification with bortezomib<br />

alone significantly improved the CR rate. Finally, encouraging<br />

results with bortezomib maintenance in the ASCT <strong>and</strong><br />

non-transplant setting <strong>are</strong> being reported <strong>and</strong> the possible<br />

efficacy of maintenance with bortezomib is studies in a<br />

number of ongoing clinical trials<br />

OP34 IMIDs in Myeloma<br />

Robert Peter Gale, Haematology Section, Division of<br />

Experimental Medicine, Department of Medicine, Imperial<br />

College, London, UK <strong>and</strong> Celgene Corp, Summit, NJ, USA<br />

The pathophysiology of multiple myeloma is complex.<br />

The initiating event is a mutational event in a B-cell. However,<br />

there <strong>are</strong> important additional mechanisms including:<br />

47


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

(1) additional mutational events within the myeloma clone;<br />

(2) aberrancies of the bone marrow micro-environment; (3)<br />

abnormalities of cytokines <strong>and</strong> hematopoietic growth factors;<br />

<strong>and</strong> (4) immune-deficiency. For several decades myeloma-therapy<br />

focused predominately on control of the myeloma<br />

clone by cytotoxic DNA-alkylating drugs (like melphalan<br />

<strong>and</strong> cyclophosphamide) <strong>and</strong> by drugs directly toxic<br />

to B-cells (like dexamethasone). Recently, new classes of<br />

anti-myeloma drugs were developed including proteosomeinhibitors<br />

(like bortezomib) <strong>and</strong> so-called immune-modulating<br />

drugs (IMiDs) (like thalidomide, lenalidomide <strong>and</strong><br />

pomalidomide). My talk focus on the mechanism(s) of action<br />

of IMiDs in myeloma <strong>and</strong> clinical efficacy.<br />

IMiDs <strong>are</strong> effective anti-myeloma drugs. Their mechanisms<br />

of action <strong>are</strong> diverse <strong>and</strong> complex. They also differ<br />

for different IMiDs. In general, IMiDs act on 4 targets in<br />

myeloma: (1) direct cytotoxicity; (2) immune-modulation;<br />

(3) effects on the bone marrow micro-environment; <strong>and</strong> (4)<br />

modulation of cytokines <strong>and</strong> hematopoietic growth factors.<br />

Exactly how IMiDs mediate these effects <strong>are</strong> complex. For<br />

example, IMiDS cause cell-cycle arrest by increasing P21,<br />

ERG1 <strong>and</strong> P15 expression, altering expression of cancer<br />

suppressor genes CDK2 <strong>and</strong> RB1, decreasing interferon<br />

regulatory factor-4 (IRF4), increasing caspase-3,-8 <strong>and</strong> -9<br />

expression <strong>and</strong> apoptosis <strong>and</strong> by decrease expression of tumour<br />

necrosis factor-alpha (TNFα). There <strong>are</strong> also stimulating<br />

effects on T- <strong>and</strong> NKT-cells. These mechanisms will be<br />

discussed in detail.<br />

Substantial data support the important clinical activity of<br />

IMiDs, alone or with other anti-myeloma drugs (like dexamethasone),<br />

in myeloma-therapy. R<strong>and</strong>omized clinical trials<br />

support the effectiveness of thalidomide <strong>and</strong> lenalidoime<br />

in persons with newly-diagnosed <strong>and</strong> with advanced myeloma.<br />

Interestingly, lenalidomide also prevents myelom<strong>are</strong>lapse<br />

in recipients of high-dose therapy <strong>and</strong> hematopoietic<br />

cell autotransplants. Pomalidomide, the newest IMiD, is active<br />

in persons failing therapy with thalidomide <strong>and</strong> lenalidomide.<br />

These data suggest different proportions of anti-myeloma<br />

mechanisms of action <strong>and</strong> support sequencing of <strong>and</strong><br />

combinations of IMiDs in myeloma-therapy.<br />

OP35 Curing Multiple myeloma – the facts behind<br />

the claim<br />

Bart Barlogie, Alan Mitchell, John D Shaughnessy Jr,<br />

<strong>and</strong> John Crowley, Myeloma Institute for Research <strong>and</strong><br />

Therapy, University of Arkansas for Medical Sciences,<br />

Little Rock, AR; <strong>and</strong> Cancer Research And Biostatistics,<br />

Seattle, WA<br />

Our Total Therapy (TT) approach for newly diagnosed<br />

multiple myeloma (MM) was initiated in 1989 with the<br />

stated goal of attempting MM cure by applying all known<br />

MM-active agents up-front in order to overcome the genomic<br />

chaos present in the majority of patients. We <strong>are</strong> reporting<br />

on updates of TT1 (n=231), TT2 r<strong>and</strong>omizing 668<br />

patients between a control an thalidomide arm, <strong>and</strong> TT3 that<br />

incorporated bortezomib up-front together with thalidomide<br />

<strong>and</strong> applying VTD in maintenance (n=303). With median<br />

follow-up times of 17.8 years in TT1, 9.4 years in TT2 <strong>and</strong><br />

48<br />

6.1 years in TT3, patients alive at last contact include 44,<br />

321, 214, respectively, of whom 24, 208, 194 remain progression-free<br />

including 15, 156,149 in continuous complete<br />

remission (CR). According to an exponential/plateau model,<br />

significant cure fractions (CF) were estimated from overall<br />

survival (OS), progression-free survival (PFS) <strong>and</strong> CR duration<br />

(CRD) as 14.6%, 10.1% <strong>and</strong> 14.1% in TT1; 19.9%,<br />

15.6% <strong>and</strong> 32.2% in TT2’s control arm; 22.5%, 24.7% <strong>and</strong><br />

40.3% in TT2’s thalidomide arm; <strong>and</strong> 61.6%, 58.2% <strong>and</strong><br />

77.2% in TT3. Data will be presented on age- <strong>and</strong> genderadjusted<br />

comparisons of overall survival (OS) with the general<br />

population along with the independent baseline features<br />

linked to 5-yr PFS <strong>and</strong> CRD estimates, also in the context of<br />

gene expression profiling (GEP), available in subsets of<br />

TT2 <strong>and</strong> TT3 trials. Collectively our results support the conclusion<br />

of curability in principle through increasing survival<br />

plateaus especially for GEP-defined low-risk MM reaching<br />

post-CR CF estimates of >80% in TT3 but also 33% in<br />

high-risk MM treated with TT3.<br />

OP36 Adult ALL treatment approach for B-ALL<br />

Dieter Hoelzer, University of Frankfurt, Germany<br />

Therapy of Burkitt leukemia (WHO classification 2008,<br />

also mature B-ALL or L3 ALL) with conventional chemotherapy<br />

resulted in an extremely overall survival of less then<br />

10% in all published studies.<br />

Short intensive sequential therapy<br />

Improvement in outcome of Burkitt lymphoma/ leukemia<br />

was first obtained in paediatric protocols e.g. (CODOX-M,<br />

BFM, LMB). Important treatment elements were high-dose<br />

fractionated alkylating agents, metabolites, high-dose Meth-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

otrexate <strong>and</strong> high-dose Ara-C. When such protocols (e.g.<br />

CODOX-M/IVAC, GMALL-B-NHL, Hyper-CVAD) were<br />

applied in adult B-ALL <strong>and</strong> Burkitt-NHL, the CR rate increased<br />

up to 80%, <strong>and</strong> the overall survival to 50–60%. Further<br />

intensification, e.g. in HDMTX-dose, did not improve<br />

results.<br />

Stem cell transplantation<br />

Autologous stem cell transplantation was considered as<br />

an alternative option in Burkitt lymphoma/ leukemia. Survival<br />

rates in retrospective analysis <strong>and</strong> prospective studies<br />

ranged from 30–60%. A substantial proportion of patients<br />

was not included because of CNS/ BM involvement or rapid<br />

progress. Allogeneic transplant resulted in an overall survival<br />

of 25–30%, most patients had high risk features or far<br />

advanced disease. It was concluded that the allogeneic HCT<br />

may exert a graft-versus-leukemia effect. Overall those SCT<br />

results were not superior to those obtained with short intensive<br />

sequential chemotherapy.<br />

Immunochemotherapy<br />

Burkitt-NHL/ leukemia cells express in >85% CD20.<br />

Therefore some adult studies included the anti-CD20 monoclonal<br />

antibody (MoAb) Rituximab. The response rate increased<br />

<strong>and</strong> overall survival rate improved to 80–90% in<br />

Burkitt lymphoma, in some studies somewhat inferior for<br />

Burkitt leukemia.<br />

Burkitt-NHL in HIV<br />

20–40% of all NHL in HIV+ patients <strong>are</strong> Burkitt NHL.<br />

When the short intensive treatment regimen were applied to<br />

those patients, outcome was clearly superior to CHOP. CR<br />

of approx. 60–70% <strong>and</strong> an overall survival rate of 50% were<br />

achieved when the patients received an additional intensive<br />

antiviral therapy (HAART).<br />

In a recent attempt HIV+ Burkitt NHL patients received<br />

also the MoAb Rituximab in addition to the intensive chemotherapy<br />

<strong>and</strong> – despite the risk of increased immunosuppression<br />

– the survival was improved to >60%.<br />

Prognostic factors<br />

With the improvement in outcome, it is not clear which<br />

adverse prognostic factors for Burkitt-NHL/ BL remain.<br />

Achievement of CR <strong>and</strong> probably stage of disease, but most<br />

likely the double-hit lymphomas with a MYC/8q24 translocation<br />

in combination with another recurrent translocation,<br />

mainly t(14;18) with BCL2-IGH fusion <strong>are</strong> associated with<br />

an unfavourable prognosis. For those patients an early SCT<br />

might be indicated.<br />

Future directions<br />

A new strong adverse prognostic risk factor in Burkitt<br />

leukemia is MRD, but is most unlikely relevant for Burkitt-<br />

NHL.<br />

For the remaining small fraction of non-responding or<br />

relapsed patients it is difficult to find promising approaches,<br />

this could be novel drugs including new antibodies, with the<br />

aim to reduce the tumor load before a SCT.<br />

Realising the r<strong>are</strong>ness of the disease, joined efforts in<br />

ALL study groups to identify risk populations <strong>and</strong> new promising<br />

approaches should be considered.<br />

OP37 T-ALL – Current Treatment<br />

Stefan Faderl, Department of Leukemia, The University of<br />

Texas MD Anderson Cancer Center Texas, USA [see SA01]<br />

Patients with T ALL constitute about 25% of all adult<br />

patients with ALL. The median age at diagnosis is about 30<br />

years <strong>and</strong> it is seen more commonly in males. Not infrequently<br />

do patients with T ALL present with high white<br />

blood cell (WBC) counts <strong>and</strong> an anterior mediastinal mass<br />

which may reach bulky proportions. Immunophenotyping<br />

by flow cytometry is an essential part of the diagnostic work<br />

up to confirm the diagnosis of an acute leukemia, assign the<br />

correct lineage, an define the stage of maturation of the blast<br />

cell population. Based on expression of various CD markers<br />

T cell blasts <strong>are</strong> divided into early, cortical (thymic) <strong>and</strong><br />

meduallary (mature) subtypes. The most T cell lineage specific<br />

marker is expression of surface CD3 (sCD3). This<br />

marker is typically positive in the medullary type, negative<br />

in the early stages <strong>and</strong> ambiguously expressed in the cortical<br />

subtypes. Although less specific but of sufficient sensitivity,<br />

CD1a is expressed in the cortical stages of T ALL but<br />

not in early <strong>and</strong> medullary subtypes.<br />

The prognosis of T ALL was poor in the past but has<br />

improved substantially over recent decades <strong>and</strong> nowadays<br />

surpasses that of patients with the more common precursor<br />

B cell ALL. This evolution is due to changes in treatment<br />

design <strong>and</strong> in particular based on 1) increased intensity; <strong>and</strong><br />

2) addition of other active drugs (eg, cytarabine, cyclophosphamide,<br />

asparaginase). For example, whereas long-term<br />

disease-free survival (DFS) was about 35% with conventional<br />

Non-Hodgkin’s lymphoma (NHL) regimens, it increased<br />

to about 50% when applying NHL regimens for<br />

high-grade lymphomas <strong>and</strong> around 55% when using ALL<br />

protocols. 1<br />

An example for a current high-intensity ALL program is<br />

Hyper-CVAD. 2 Hyper-CVAD consists of 8 intensive induction<br />

/consolidation cycles where hyper-CVAD (hyperfractionated<br />

cyclophosphamide, doxorubicin, vincristine, dexamethasone)<br />

alternates with high-dose methotrexate <strong>and</strong> cytarabine.<br />

This is followed by up to 32 months of the so<br />

called POMP maintenance (vincristine, methotrexate, mercaptopurine,<br />

prednisone). Central nervous system (CNS)<br />

prophylaxis consists of intrathecal therapy with cytarabine<br />

<strong>and</strong> methotrexate for a maximum of 8 injections. The experience<br />

of patients with lymphoblastic lymphoma (T cell lineage<br />

in 79% of the patients) was recently published. Fortynine<br />

patients were included with a median age of 31 years<br />

(range 17 to 59). The complete remission rate was 96% <strong>and</strong><br />

5-year survival 68%. Fourteen of the patients who achieved<br />

CR relapsed at a median of 13 months. The joint study of<br />

the MRC UKALL XII/ECOG E2993 trial included 356 patients<br />

with T ALL with a median age of 29 years. 3 Patients<br />

received 2 induction phases (daunorubicin, vincristine, asparaginase,<br />

prednisone, methotrexate in phase 1 followed<br />

by cyclophosphamide, cytarabine, mercaptopurine, <strong>and</strong> methotrexate<br />

in phase 2). Patients were then either assigned to<br />

an allogeneic stem cell transplant (SCT) arm (if they had an<br />

HLA-compatible donor <strong>and</strong> were younger than 50 years) or<br />

r<strong>and</strong>omized between either an autologous SCT or chemo-<br />

49


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

therapy (if they were at least 50 years old <strong>and</strong> had no donor).<br />

Prior to their assigned or r<strong>and</strong>omized treatment every patient<br />

received high-dose methotrexate <strong>and</strong> asparaginase.<br />

The complete remission rate was 94%; older patients (over<br />

age 50 years) had a significantly lower remission probability<br />

of 79%. The median 5-year survival probability was 48%<br />

with significant differences based on age (38% for patients<br />

over age 35), WBC (41% for patients with a WBC of<br />

>100,000/mcl), immunophenotype (patients who expressed<br />

CD13 <strong>and</strong> lacked CD1a expression did worse), <strong>and</strong> karyotype<br />

(19% with complex karyotypes).<br />

The German ALL Study Group (GMALL) evaluated outcome<br />

of patients with T ALL based on immunophenotype:<br />

early (sCD3 negative, CD1a negative), thymic (CD1a positive),<br />

<strong>and</strong> mature/medullary (sCD3 positive). 4 Three different<br />

studies included a total of 744 patients. Study GMALL<br />

05/93 included 239 patients who all received multiagent<br />

chemotherapy without provisions for SCT in first remission.<br />

Studies GMALL 06/99 <strong>and</strong> 07/03 consisted of 505<br />

patients. In these latter two studies patients received an intensified<br />

treatment (pegaspargase, high-dose methotrexate)<br />

<strong>and</strong> patients with early, mature <strong>and</strong> high-risk thymic T ALL<br />

received SCT in first CR. Outcome was most favorable for<br />

patients with thymic ALL where 10-year survival exceeded<br />

60% with chemotherapy alone. Results were worst for patients<br />

with early <strong>and</strong> mature T-ALL if they received chemotherapy<br />

but were significantly better if these patients received<br />

an allogeneic SCT in first remission.<br />

Novel approaches aim to include additional drugs. In a<br />

study by the MD Anderson group Hyper-CVAD is being<br />

combined with nelarabine. Patients receive Hyper-CVAD as<br />

detailed above. Prior to the POMP maintenance they receive<br />

two cycles of nelarabine followed by one further cycle of<br />

nelarabine later during the maintenance. Interim results<br />

have recently been presented. 5 Of 23 patient with a median<br />

age of 38 years (rate 21 to 76), the CR rate was 96%. With a<br />

median follow up of 14 months, median overall <strong>and</strong> DFS<br />

have not been reached. The survival probability at 3 years<br />

was 65%. These data <strong>are</strong> in keeping with those reported by<br />

other groups before. Longer follow up will be necessary to<br />

determine the benefit if any of the addition of nelarabine<br />

<strong>and</strong> in what sequence.<br />

In summary, modern <strong>and</strong> intensive multiagent chemotherapy<br />

regimens achieve remissions in most patients <strong>and</strong><br />

long-term DFS in about 50 to 60%. New markers of prognosis<br />

(immunophenotype, genotype) <strong>are</strong> valid in determining<br />

whether or not further intensification (SCT) in first remission<br />

can improve outcome. Other markers (eg, mutations of<br />

NOTCH1 or detection of the NUP214-ABL1 fusion gene)<br />

<strong>are</strong> being explored as basis for applying targeted treatment<br />

approaches (gamma secretase inhibitors in the former <strong>and</strong><br />

tyrosine kinase inhibitors in the latter example). New drugs<br />

<strong>and</strong> combinations such as with nelarabine provide further<br />

means to exp<strong>and</strong> therapeutic options in the future.<br />

R E F E R E N C E S<br />

1. Cortelazzo S, Ponozoni M, Ferreri AJM, Hoelzer D. Lymphoblastic<br />

lymphoma. Cri Rev Oncol/Hematol 2011 [epub]<br />

2. Thomas DA, O’Brien S, Cortes J, et al. Outcome with the hyper-CVAD<br />

regimens in lymhoblastic lymphoma. Blood 2004;104:1624–1630.<br />

50<br />

3. Marks DI, Paietta EM, Moorman AV, et al. T-cell acute lymphoblastic<br />

leukemia in adults: clinical features, immunophenotype, cytogenetics,<br />

<strong>and</strong> outcome from the large r<strong>and</strong>omized prospective trial (UKALL XII/<br />

ECOG E2993). Blood 2009;114:5136–5145.<br />

4. Hoelzer D, Thiel E, Arnold R, et al. Successful subtype oriented treatment<br />

strategies in adult T-ALL; Results of 744 patients treated in three<br />

consecutive GMALL Studies. Blood 2009;114:abs 324.<br />

5. Al-Ameri A, Thomas DA, Rav<strong>and</strong>i F, et al. Hyper-CVAD plus nelarabine<br />

in the treatment of newly diagnosed patients with T cell acute lymphoblastic<br />

leukemia/ lymphoblastic lymphoma (T-ALL/LL). J Clin Oncol<br />

2011;29:432S.<br />

OP38 Young adult with ALL – how to treat<br />

Jean-Pierre Marie, Stéphanie Haïat, Anne Vekhoff<br />

& Ollivier Legr<strong>and</strong>, Department of Hematology, Saint-<br />

Antoine Hospital & Université Pierre et Marie Curie,<br />

Paris, EORTC Leukemia Group<br />

Survival for adult with ALL is lower than in children,<br />

mainly due to the relapse rate. A pressing question is whether<br />

the age-related differences in outcome <strong>are</strong> predicated by<br />

the disease per see or whether they result from a different<br />

approach to, or tolerance of, therapy. Boissel et al. (1) comp<strong>are</strong>d<br />

the outcome of adolescents (15–20 years) with ALL<br />

treated in France in either the pediatric FRALLE 93 or the<br />

adult LALA-94 clinical trials. With a median follow up of<br />

about 3.5 years, the CR rate (94% vs. 83%), event free survival<br />

(EFS) at 5 years (67% vs.41%), <strong>and</strong> the DFS for the<br />

CR patients (72% vs. 49%) were far superior in the pediatric<br />

group <strong>and</strong> multivariate analysis confirmed the independent<br />

effect of the treatment trial on outcome. The major differences<br />

between the pediatric or adult approach were the actually<br />

given dosages of asparaginase, corticosteroids <strong>and</strong> vinca-alkaloids.<br />

Another difference was the strict discipline<br />

with which the treatment courses were administered by the<br />

pediatricians comp<strong>are</strong>d to the flexibility of the internists.<br />

Analyses with similar outcome have been reported by comparing<br />

ALL patients aged 15 till 21 years treated in different<br />

countries (2,7). The recent GRAALL-2003 study suggests<br />

that pediatric-inspired therapy markedly improves the outcome<br />

in adult patients with ALL, at least until the age of 45<br />

(8). We recently tested the feasibility of a pediatric regimen<br />

(FRALLE 2000) (9) in 40 adult ALL patients (age 16–57<br />

years), <strong>and</strong> comp<strong>are</strong>d the results to the EORTC ALL4 trial<br />

(10). Median follow-up of alive patients was 1596 days for<br />

patients treated in pediatric protocol <strong>and</strong> 1552 days for patients<br />

treated in ALL4 protocol (p=NS). DFS <strong>and</strong> OS were<br />

63%±9%, <strong>and</strong> 70%±7%, <strong>and</strong> were significantly longer in<br />

patients under 40yo (86%±7% versus 30%±15%, p=0.003<br />

[DFS] <strong>and</strong> 86%±7.8% vs 49%±12%, p=0.01 [OS], respectively)<br />

or cortico/chemo-sensitive (71%±9% versus 36%±<br />

15%, p=0.01 [DFS] <strong>and</strong> 86%±7%, versus 28%±13%, p=<br />

0.0004 [OS]) than in other patients.<br />

Ten patients had undetectable minimal residual disease<br />

(MRD) <strong>and</strong> none of them experienced relapse. Grade 3 – 4<br />

toxicities included a considerable number of severe infections,<br />

liver function abnormalities, unexpected peripheral<br />

neuropathies, osteonecrosis <strong>and</strong> denutrition. These toxicities<br />

increased with age. The adherence discipline with respect<br />

to time-lines of the treatment schedules was successful<br />

in approximately 50% of the limited number of patients


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

studied so far. Similar experience was observed in the HO-<br />

VON-70 study, which included 54 patients aged 18 to 39<br />

years. Overall, it was concluded that results appe<strong>are</strong>d very<br />

encouraging, but that side effects necessitated prolongation<br />

of the induction/consolidation/ intensification phase in a<br />

considerable number of patients <strong>and</strong> that the protocol<br />

seemed too intensive for older patients. The threshold, in<br />

our experience but also in the GRAALL <strong>and</strong> the HOVON is<br />

40 yo.<br />

In conclusion, adolescents but also »young« adults (


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

ond generation ABL TK inhibitors. This provides the rationale<br />

for the administration of TKI after SCT, although the<br />

exact modalities, e.g. type of TKI, dose, starting timepoint<br />

<strong>and</strong> duration of treatment remain to be resolved.<br />

The importance of mutations in clinical resistance to<br />

available TKI has promoted the search for novel inhibitors,<br />

active particularly against the T315I gatekeeper mutation.<br />

Interesting immunologic approaches <strong>are</strong> based on recently<br />

developed bispecific T-cell-engager (BiTE) antibodies<br />

that transiently engage cytotoxic T-cells for lysis of selected<br />

target cells. The bispecific antibody construct called blinatumomab<br />

links T cells with CD19-expressing target cells,<br />

resulting in a non-restricted cytotoxic T-cell response <strong>and</strong><br />

T-cell activation. Preliminary results indicate that treatment<br />

with blinatumomab is able to convert MRD positive ALL<br />

into an MRD negative status, <strong>and</strong> that this is well tolerated.<br />

Current efforts also involve the optimisation of molecular<br />

techniques to quantitate minimal residual disease <strong>and</strong> identify<br />

resistance mechanisms. Optimal treatment results necessitate<br />

the integration of all available therapeutic <strong>and</strong> molecular<br />

diagnostic approaches into predefined, long-term<br />

strategies that take into account both patient <strong>and</strong> disease<br />

characteristics.<br />

OP40 MRD – criteria for postremission therapy<br />

Deborah Thomas, (USA)<br />

52<br />

Abstract not submitted.<br />

OP41 Allogeneic stem cell transplantation for<br />

adult ALL: st<strong>and</strong>ard vs. Reduced-intensity<br />

conditioning<br />

Mohamad Mohty, Hematology Dpt., University-Hopsital<br />

of Nantes, Centre de Recherche en Cancerologie de<br />

Nantes-Angers (CRCNA), INSERM U892, F-44093<br />

Nantes, France<br />

Acute lymphoblastic leukemia (ALL) accounts for approximately<br />

15 to 20% of all adult acute leukemias. Despite<br />

the fact that 80 to 90% of adult patients with ALL succeed<br />

in achieving complete remission (CR), most of them will<br />

relapse <strong>and</strong> die of their disease. Among adults with ALL,<br />

long-term leukemia-free survival (LFS) rates of 30 to 40%<br />

have been obtained with the use of chemotherapy, as comp<strong>are</strong>d<br />

with 45 to 75% with the use of conventional myeloablative<br />

conditioning (MAC) allogeneic stem cell transplantation<br />

(allo-SCT). The latter favorable effect is likely due to a<br />

reduced risk of relapse, especially in those patients in first<br />

CR. However, non-relapse mortality (NRM) may counterbalance<br />

that favorable overall outcome observed after MAC<br />

allo-SCT in those elderly patients <strong>and</strong>/or in patients with<br />

comorbidities. Thus, the use of reduced intensity conditioning<br />

(RIC) prior to allo-SCT may offer hitherto unavailable<br />

opportunities to obtain a graft-vs.-leukemia effect without<br />

the toxicities of intense preparative regimens. However, one<br />

must acknowledge that the role of allo-SCT in adult ALL is<br />

still controversial. In addition, ALL encompasses a group of<br />

chemosensitive diseases, raising concerns that significant<br />

reduction of the intensity of the preparative regimen prior to<br />

allo-SCT may have a negative impact on long-term leukemic<br />

control as it has been already shown in other settings.<br />

Also, very few data is available analyzing RIC allo-SCT for<br />

patients with ALL.<br />

This report will discuss the available research evidence in<br />

this field with a special focus on comparative studies which<br />

assessed the outcomes of adult patients with ALL who underwent<br />

allo-SCT, <strong>and</strong> were analyzed according to the type<br />

of conditioning received prior to allo-SCT (RIC vs. MAC).<br />

OP42 Autologous SCT for ALL – stil a chalange<br />

Sebastian Giebel, Department of Bone Marrow<br />

Transplantation, Maria Sklodowska-Curie Memorial<br />

Cancer Centre <strong>and</strong> Institute of Oncology, Gliwice Branch,<br />

Gliwice, Pol<strong>and</strong><br />

First-line therapy of adults with Ph-negative acute lymphoblastic<br />

leukemia (ALL) consists of remission induction<br />

followed by intensive consolidation. Further treatment usually<br />

depends on the presence of risk factors. Conventional<br />

risk factors include initial tumor burden, age, immunophenotype,<br />

karyotype, <strong>and</strong> time to achieve complete remission<br />

(CR). In recent years, a number of studies demonstrated a<br />

strong prognostic value of the status of minimal residual<br />

disease (MRD) measured after induction <strong>and</strong> consolidation.<br />

1–3 According to some authors MRD together with cytogenetics<br />

may be sufficient for risk stratification. 4,5 Patients<br />

allocated to st<strong>and</strong>ard risk (SR) group used to be treated with<br />

long-term maintenance chemotherapy while those with high<br />

risk (HR) features <strong>are</strong> usually offered allogeneic stem cell<br />

transplantation (alloSCT).<br />

The role of autologous SCT (autoSCT) in adults with<br />

ALL has not been clearly defined. In the 1990ies a series of<br />

studies have been conducted to evaluate whether high-dose<br />

therapy followed by autoHSCT may improve the outcome<br />

of HR patients lacking an HLA-identical sibling comp<strong>are</strong>d<br />

to conventional-dose post-consolidation chemotherapy. 6–9<br />

In the EORTC ALL3 study results of autoHSCT <strong>and</strong> maintenance<br />

treatment appe<strong>are</strong>d superimposable. 6 In the PETH-<br />

EMA ALL93 study no significant difference between the<br />

transplantation <strong>and</strong> chemotherapy arm could be demonstrated,<br />

however, there was some tendency in favor of conventional-dose<br />

treatment (disease-free survival, DFS, 44% vs.<br />

35% at 5 years). 7 In contrast, results of the LALA-94 suggested<br />

some advantage of autoHSCT (DFS 25% vs. 13% at<br />

5 years) although the difference was not statistically significant.<br />

8 A pooled analysis of three subsequent trials by the<br />

French group (LALA-85/87/94) showed that autoHSCT may<br />

contribute to decreased risk of relapse comp<strong>are</strong>d to conventional-dose<br />

chemotherapy (66% vs. 78%, p=0.05) with mno<br />

significant impact on survival. 10 Altogether, the above cited<br />

studies did not provide clear evidence for neither the superiority<br />

nor inferiority of autoHSCT over the maintenance.<br />

In the largest so far MRC UKALLXII/ECOG 2993 study<br />

investigators tested if autoHSCT may be administered instead<br />

of consolidation + maintenance for patients lacking an<br />

HLA-identical sibling, irrespective of the presence of risk<br />

factors. 9 The overall survival was significantly worse in the


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

autoHSCT comp<strong>are</strong>d to chemotherapy arm (46% vs. 37% at<br />

5 years, p=0.03) leading to conclusion that early autoHSCT<br />

cannot substitute consolidation. In addition the donor vs. no<br />

donor analysis revealed advantage of alloHSCT arm in SR<br />

patients. In HR group the non-relapse mortality was 39%<br />

among patients having a donor, overcoming advantage related<br />

to reduced risk of relapse.<br />

Disappointing results of prospective trials evaluating the<br />

role of autoHSCT led to a general tendency to ab<strong>and</strong>on this<br />

treatment option in adults with ALL. It must be stressed,<br />

however, that design of the studies could have been suboptimal.<br />

First of all, the trials have been conducted before the<br />

era of routine MRD assessment. Low tumor burden is prerequisite<br />

of successful autotransplantation as in other conditions<br />

the transplant material may be contaminated by residual<br />

leukemic blasts leading to their re-transplantation <strong>and</strong><br />

early relapse. Evaluation of MRD status in bone marrow<br />

reflecting the tumor load may therefore be crucial for selection<br />

of patients who could benefit from autoHSCT. Indeed,<br />

in a retrospective analysis by the European Working Group<br />

for Adult ALL, the MRD level was demonstrated the most<br />

important prognostic factor with DFS of 58% for patients<br />

with MRD


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

mia (CLL) cells. The availability of these antibodies is making<br />

a great impact in the treatment of CLL. Alemtuzumab, a<br />

chimeric monoclonal antibody that targets the CD52 surface<br />

antigen, was the first antibody approved for the treatment<br />

of patients with CLL. It induces responses in one-third<br />

of relapsed <strong>and</strong> refractory patients, <strong>and</strong> in more than 80% of<br />

the patients when used as initial therapy. Alemtuzumab has<br />

been used as single agent as well as in combination with the<br />

monoclonal antibody rituximab or with chemotherapy in<br />

combination with fludarabine, cyclophosphamide <strong>and</strong> alemtuzumab.<br />

Treatment with alemtuzumab is associated with a<br />

profound lymphocyte depletion <strong>and</strong> acquired immunosuppression;<br />

therefore, opportunistic infections <strong>are</strong> often increased<br />

following this therapy limiting its use. Rituximab,<br />

a chimeric monoclonal antibody against CD20 has been<br />

widely used in lymphoproliferative disorders. In CLL, rituximab<br />

has limited activity when used as single agent, but has<br />

significant synergism when used in combination with other<br />

monoclonal antibodies or chemotherapy. The addition of<br />

rituximab to the combination of fludarabine <strong>and</strong> cyclophosphamide<br />

has been shown to improve response rate <strong>and</strong> prolong<br />

survival in physically fit patients with CLL. Rituximab<br />

has also shown activity in combination with numerous chemotherapeutic<br />

agents such as bendamustine <strong>and</strong> pentostatin.<br />

More recently developed, ofatumumab, is a fully humanized<br />

anti-CD20 antibody with efficacy in patients whose<br />

disease is refractory to both fludarabine <strong>and</strong> alemtuzumab.<br />

Ofatumumab is currently undergoing evaluation in combination<br />

with chemotherapy <strong>and</strong> other biological agents. Other<br />

antibodies or related molecules currently in clinical trials<br />

include GA101, a type II anti-CD20 antibody, lumiliximab,<br />

an antibody targeting CD23, blinatumomab, a bi-specific<br />

single-chain antibody targeting CD19 <strong>and</strong> Tru-016 a small<br />

modular immunopharmaceutical (SMIP) targeting CD37.<br />

These monoclonal antibodies <strong>and</strong> related agents use diverse<br />

mechanisms of action, complement each other <strong>and</strong> when<br />

used in combinations have the potential to enhance the activity<br />

of chemotherapy <strong>and</strong> other biologic agents. Through<br />

the ongoing <strong>and</strong> future development of such combinations,<br />

effective therapeutic options for patients with CLL will continue<br />

to increase.<br />

OP47 New <strong>and</strong> old drugs in CLL – we can do<br />

better<br />

Peter Hillmen, (United Kingdom)<br />

54<br />

Abstract not submitted.<br />

OP48 SCT for CLL – when?<br />

Issa Khouri, Department of Stem Cell Transplantation<br />

<strong>and</strong> Cellular Therapy, The University of Texas MD<br />

Anderson Cancer Center, Houston, Texas, USA<br />

In the past decade, the paradigm for allogeneic stem cell<br />

transplantation (SCT) for chronic lymphocytic leukemia<br />

(CLL) has changed. Confidence in the power of the graftversus-leukemia<br />

effect has encouraged the use of transplan-<br />

tation after low-intensity, nonmyeloablative preparative<br />

regimens. This strategy has resulted in long-term remissions<br />

in a subset of patients with advanced CLL who develop resistance<br />

to conventional treatments. Nonmyeloablative SCT<br />

(NST) can overcome adverse prognostic factors such as the<br />

presence or absence of somatic mutations in the immunoglobulin<br />

heavy chain variable region, Zeta-chain-associated<br />

protein 70 kDa expression, <strong>and</strong> the presence of chromosomal<br />

aberrations, including 17p13.1 deletion. Recent studies<br />

suggested that patients’ hematopoietic SCT-comorbidity index,<br />

tumor bulk, age, <strong>and</strong> disease refractoriness were important<br />

determinants of survival. We review the results of<br />

NST in CLL, discuss the evolution of the new nonmyeloablative<br />

approaches, <strong>and</strong> make recommendations for when<br />

SCT should be considered in patients with CLL.<br />

OP49 Prognostic score for Hematopoietic Stem<br />

Cell Transplantation<br />

Alois Gratwohl, Hematology, Medical Faculty, University<br />

of Basel, Basel, Switzerl<strong>and</strong><br />

Hematopoietic stem cell transplantation (HSCT) has become<br />

the treatment of choice for many patients with a congenital<br />

or acquired severe disorder of the hematopoietic<br />

system. Despite marked improvements over time, HSCT remains<br />

inherently associated with morbidity <strong>and</strong> mortality.<br />

Prior to any decision to proceed with HSCT an assessment<br />

is warranted whether the potential benefits outweigh the<br />

risks of the procedure. Risk assessment is complex, requires<br />

specific skills <strong>and</strong> experience <strong>and</strong> includes risk of the disease<br />

<strong>and</strong> risk of HSCT. Risk of HSCT includes pre-, peri-<br />

<strong>and</strong> post transplant risk elements. Not all of them <strong>are</strong> known<br />

at the time of decision making, pretransplant elements <strong>are</strong>.<br />

The EBMT risk score provides a simple tool to assess instantly<br />

pre-transplant risks for an individual patient. Five<br />

factors, age of the patient (40 years 2 score points), stage of the disease (Early disease<br />

stage 0; intermediate disease stage 1; late stage disease 2<br />

score points), time from diagnosis to HSCT (1<br />

year 1 score point. Note: time interval diagnosis to HSCT<br />

does not apply for patients transplanted in 1 st CR, score is<br />

always 0), donor type (HLA-identical sibling as donor 0;<br />

other donor 1 score point) <strong>and</strong> donor recipient gender combination<br />

(»all other combinations« 0; »female donor for a<br />

male recipient« 1 score point) augment risk for an individual<br />

patient with increasing score from 0 as best to 7 as worst.<br />

Individual risks <strong>are</strong> additive! The score holds for all acquired<br />

hematological disorders, for allogeneic <strong>and</strong> autologous<br />

HSCT (for autologous HSCT consider only age, disease<br />

stage <strong>and</strong> time interval; hence score 0–5). The score is<br />

independent of the HSCT technology; eg, it is valid for st<strong>and</strong>ard<br />

or reduced intensity conditioning, for bone marrow,<br />

peripheral blood <strong>and</strong> cord blood transplants, for grafts with<br />

or without T cell depletion. Survival is uniformly worse for<br />

older patients, transplanted in more advanced disease stage<br />

after a long time interval with a mismatched donor than for<br />

younger patients, transplanted soon in early stage with a<br />

well matched donor. Additional factors such as performance


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

score, comorbidities, CMV serostatus or cytokine polymorphisms<br />

add to the risk <strong>and</strong> improve prediction. There impact<br />

differs for low or high risk patients. Optimal donor recipient<br />

gender combination or CMV serostatus improve outcome in<br />

otherwise low risk patients whereas a low performance<br />

score might be acceptable in low risk but detrimental in otherwise<br />

high risk patients. Such comparative assessment of<br />

disease risk <strong>and</strong> global pre-transplant risk can guide the decisions<br />

for individual patients between HSCT <strong>and</strong> a non<br />

transplant approach. The EBMT score forms the basis for a<br />

risk adapted individualized strategy which should replace<br />

the traditional »donor versus no donor« concept.<br />

OP50 Consideration in Choosing the Optimal<br />

Stem Cell Source<br />

Mary M. Horowitz, Division of Hematology <strong>and</strong> Oncology,<br />

Department of Medicine, Medical College of Wisconsin<br />

<strong>and</strong> Scientific Director, Center for International Blood<br />

<strong>and</strong> Marrow Transplantation (CIBMTR), USA<br />

Considerations in selecting the optimal cell source for allogeneic<br />

transplantation include donor relationship (related<br />

versus unrelated), donor-recipient HLA-matching. <strong>and</strong> cell<br />

source (bone marrow vs peripheral blood versus umbilical<br />

cord blood). HLA-identical siblings <strong>are</strong> generally accepted<br />

as the preferred donor unless donor co-morbidities preclude<br />

donation. Whether to use bone marrow or peripheral blood<br />

grafts from sibling donors depends, in part, on recipient factors.<br />

Although peripheral blood grafts <strong>are</strong> associated with<br />

higher risks of chronic graft-versus-host disease (GVHD),<br />

patients with advanced disease may benefit from the higher<br />

cell doses possible with such grafts. If an HLA-identical<br />

sibling is not available, a search for an unrelated adult volunteer<br />

donor may be done. Best results with unrelated donor<br />

transplantation <strong>are</strong> obtained when donor <strong>and</strong> recipient <strong>are</strong><br />

matched at high-resolution for HLA A, B, C, <strong>and</strong> DRB1. In<br />

the absence of a matched unrelated donor, a donor with a<br />

single mismatch at these loci may be used. In such situations,<br />

matching at low-expression alleles (DQ, DP, DRB3/<br />

4/5) may be beneficial. CIBMTR data suggest that bone<br />

marrow or peripheral blood grafts from unrelated donors<br />

result in similar survival rates but with more GVHD; a large<br />

r<strong>and</strong>omized trial of unrelated donor blood versus bone marrow<br />

grafts is in progress. Some data suggest that peripheral<br />

blood transplantation may be more permissive of HLA mismatching<br />

than bone marrow transplantation. If an adult unrelated<br />

donor is not available in a timely manner, umbilical<br />

cord blood mismatched at one or two loci may be used, with<br />

current data suggesting results similar to results with unrelated<br />

bone marrow or peripheral blood transplantation. The<br />

limiting factor in cord blood transplantation is cell dose,<br />

which may be circumvented by use of multiple units. The<br />

choice of an adult donor versus cord blood is influenced by<br />

graft factors such as HLA-match <strong>and</strong> cell dose but also by<br />

patient factors such as age, weight, type of disease <strong>and</strong> urgency<br />

of the clinical situation. Haploidentical peripheral<br />

blood or bone marrow transplantation may also be effective<br />

graft sources with a recent study suggesting results similar<br />

to cord blood transplantation. With an increasing array of<br />

graft source options, transplantation should be considered<br />

early in the disease course of patients with diseases potentially<br />

benefited by allografting to allow appropriate testing<br />

of potential donors <strong>and</strong> to avoid undue delay when transplantation<br />

becomes necessary. Decisions about when <strong>and</strong><br />

with which graft source to perform transplantation should<br />

be based on a several factors, including donor <strong>and</strong> graft<br />

characteristics but also patient age, performance status <strong>and</strong><br />

disease.<br />

OP51 Source of stem cells in the allogeneic<br />

transplant setting: bone marrow versus<br />

peripheral blood stem cells<br />

Mohamad Mohty, Hematology Dpt., University-Hopsital<br />

of Nantes, Centre de Recherche en Cancerologie de<br />

Nantes-Angers (CRCNA), INSERM U892, F-44093<br />

Nantes, France<br />

While peripheral blood stem cells (PBSC) <strong>are</strong> used almost<br />

exclusively in autologous transplantation, recent surveys<br />

indicate that PBSC <strong>are</strong> used in 50% to 70% of allogeneic<br />

stem-cell transplants. Thus, large variation in practice<br />

<strong>and</strong> considerable uncertainty exists with respect to the relative<br />

effects of allogeneic PBSC transplantation (PBSCT)<br />

versus bone marrow transplantation (BMT) on the outcomes<br />

of patients with hematologic malignancies. Indeed, the use<br />

of PBSCs for allogeneic transplantation is still not universally<br />

accepted. This is in part due to unresolved concerns<br />

about the long-term effects of growth factor treatment in<br />

healthy volunteers <strong>and</strong> data suggesting that this stem cell<br />

source is associated with more chronic graft-versus-host<br />

disease (cGVHD). Comp<strong>are</strong>d with BM, PBSC grafts contain<br />

significantly more total blood cells <strong>and</strong> more CD34+<br />

cells. However, the most striking difference is the 10-fold<br />

higher number of T cells in the PBSC graft. Other differences<br />

include a higher number of monocytes <strong>and</strong> the absence<br />

of mesenchymal stem cells.<br />

In order to address the question of PBSCT versus BMT,<br />

several r<strong>and</strong>omized controlled trials have been conducted.<br />

However, despite these well designed <strong>and</strong> executed clinical<br />

trials, taken individually, most of these trials were relatively<br />

small to draw definitive conclusions, <strong>and</strong> not surprisingly,<br />

substantial controversy still remains regarding the impact<br />

on mortality, disease control, <strong>and</strong> other important clinical<br />

outcomes in relation with the type of donor.<br />

This report will examine the differences in the outcomes<br />

between HLA–matched, related <strong>and</strong> unrelated allogeneic PB-<br />

SCT <strong>and</strong> BMT as therapy for hematologic malignancies.<br />

OP52 Conditioning for SCT – the impact<br />

of disease?!<br />

Dietger Niederwieser, (Germany)<br />

Abstract not submitted.<br />

55


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

OP53 Conditioning for SCT – the impact<br />

of disease?!<br />

John Barrett, Hematology Branch, National Heart,<br />

Lung <strong>and</strong> Blood Institute, National Institutes of Health.<br />

Bethesda, MD, USA<br />

Background<br />

Bone marrow stem cell transplantation for hematological<br />

malignancies was originally perceived as a necessary rescue<br />

from bone marrow failure following the definitive treatment<br />

of the malignant disease by high dose chemotherapy or radiation.<br />

The earliest clinical studies from the mid 1950s involved<br />

autologous bone marrow harvest, a single high dose<br />

infusion of an alkylating agent followed by bone marrow<br />

infusion rescue. Nevertheless, as early as 1956 Barnes <strong>and</strong><br />

Loutit demonstrated that in contradistinction to autologous<br />

marrow grafts an allogeneic bone marrow could confer a<br />

graft versus-leukemia (GVL) effect, albeit with an associated<br />

lethal graft-versus-host reaction (GVH) 1 . Despite this<br />

early recognition that the curative effect of a bone marrow<br />

allograft lay not only in the strength of the conditioning<br />

regimen but also in the alloreactivity of donor against recipient<br />

leukemia it was generally considered that the GVL<br />

effect was modest unless associated with such severe GVH<br />

disease that it would be impracticable to pursue ways to select<br />

<strong>and</strong> maximize GVL. In 1990 two papers changed our<br />

perception by demonstrating that the GVL effect made a<br />

major contribution to the curative effect of stem cell transplantation.<br />

The first – a study by Horowitz, from the International<br />

Bone Marrow Transplantation Registry revealed<br />

the significantly lower relapse risk in survivors who had developed<br />

acute <strong>and</strong> chronic GVHD with lowest relapse in<br />

those who experienced both complications2 . The second,<br />

from Kolb <strong>and</strong> colleagues, revealed the remarkable potential<br />

of infusions of donor lymphocytes to achieve stable<br />

cures of chronic myelogenous leuekemia (CML) relapsing<br />

after bone marrow transplantation3 . The new perception that<br />

GVL was a force to be recognized has underpinned the subsequent<br />

evolution of SCT strategies. Here I will discuss the<br />

interplay between the myelosuppression <strong>and</strong> immunosuppression<br />

of the conditioning regimen, the stem cell graft<br />

type, <strong>and</strong> post graft immunosuppression in the control of<br />

malignant disease. The impact of various transplant approaches<br />

on disease type <strong>and</strong> stage will then be considered<br />

with the aim of identifying the best transplant strategy for<br />

the individual patient.<br />

The multifunctional role of the conditioning regimen<br />

The agents used in conditioning regimens impact both<br />

the hematopoietic <strong>and</strong> immune system as well as causing<br />

off-target damage to other organ systems especially those<br />

showing rapid cell turnover such as the gastrointestinal<br />

tract. Figure 1 shows commonly used conditioning agents<br />

classified by the spectrum of their properties. However it<br />

should be noted that detailed characterization of the immunosuppressive<br />

properties of many agents such as busulfan,<br />

is lacking.<br />

Nevertheless we now have enough underst<strong>and</strong>ing of the<br />

relative actions of conditioning agents that regimens can be<br />

56<br />

Figure 1. Relative properties of common conditioning regimen agents<br />

designed according to their intensity <strong>and</strong> myeolsuppressive<br />

characteristics, as well as their ability to immunoablate.<br />

C<strong>are</strong>fully selected combinations can minimize the unwanted<br />

side effects. Currently used conditioning regimens <strong>are</strong><br />

classified as full intensity or reduced intensity which may or<br />

may not be myeloablative.<br />

Role of myelosuppressive intensity<br />

There <strong>are</strong> now many studies comparing reduced intensity<br />

<strong>and</strong> st<strong>and</strong>ard intensity regimens in specific malignancies.<br />

Myeloablative intensity can be measured by myeloid cell<br />

chimerism using CD15 as a marker. Lower intensity regimens<br />

allow host hematopoietic recovery before their ultimate<br />

rejection <strong>and</strong> replacement by donor myeloid cells<br />

when the donor immunity transcends that of the host. Predictably<br />

many studies show that relapse rates <strong>are</strong> higher in<br />

recipients of reduced intensity transplants while recipients<br />

of full intensity transplants <strong>are</strong> at greater risk of death from<br />

non-relapse complications.<br />

Role of immunosuppressive intensity<br />

In a completely immunoablated recipient with no T cell<br />

or NK cell function the SCT can rapidly engraft. Donor T<br />

cell chimerism, measured in the CD3 fraction of peripheral<br />

blood, reaches 100% by day 14 permitting the early establishment<br />

of GVL effects accompanied by a risk of early<br />

acute GVHD. The type of conditioning regimen affects the<br />

degree to which residual host cells persist <strong>and</strong> thus affects<br />

the pace of engraftment of the donor immune system.<br />

Towards the selection of the best conditioning regimen<br />

for the disease<br />

While it is broadly true that more intensive conditioning<br />

leads to greater antileukemic control as well as conferring<br />

greater immunosuppression <strong>and</strong> permitting early donor immune<br />

function, the relative merits of promoting GVL effects<br />

or relying on direct suppression of malignancy by<br />

chemoradiotherapy depend on the malignant disease in<br />

question. The current challenge is to vary the conditioning<br />

regimen to deliver the optimum conditions for cure for specific<br />

disease subtypes.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

Diseases where GVT effects predominate<br />

The role of conditioning in promoting the GVT effect<br />

transcends any anti-malignancy effect in two situations; In<br />

solid tumors insensitive to chemotherapy such as renal cell<br />

cancer the role of the conditioning is to establish the donor<br />

immune system to initiate GVT activity. Fludarabine <strong>and</strong><br />

cyclophosphamide combinations function well in this regard<br />

achieving rapid donor T cell chimerism without myeolablation.<br />

Some hematological malignancies – CML,<br />

chronic lymphocytic leukemia (CLL), <strong>and</strong> some non-hodgkin<br />

lymphomas (NHL) <strong>are</strong> especially sensitive to GVL effects.<br />

NHL regimens that avoid excessive immunosuppression<br />

from conditioning using Campath (anti CD52) antibody<br />

have a significantly lower relapse rate. In CML prior<br />

to the imatinib era several studies began to explore the use<br />

of non-myeloablative regimens or reduced intensity regimens<br />

to achieve cure cure without complications from intensive<br />

treatment. These studies showed that without the<br />

contribution from some myelosuppression there was an increased<br />

risk of persistent disease. CLL appears to be particularly<br />

susceptible to GVL. As typically older patients<br />

with CLL require transplants there has been much progress<br />

in designing low intensity but effective regimens for such<br />

patients.<br />

Diseases requiring disease control as well as GVL/GVT<br />

Despite a GVL effect being demonstrable most hematological<br />

disorders (including myelodysplastic syndromes,<br />

acute leukemias <strong>and</strong> multiple myeloma) require myeloablation<br />

to maximize the chance of cure. The problem arises in<br />

this group that there is a trade-off between curative potential<br />

<strong>and</strong> risk of mortality from non-relapse causes. This has led<br />

to c<strong>are</strong>ful titration of the dose intensity of the conditioning<br />

especially in older or debilitated patients recognizing that<br />

relapse risk increases with reduction in intensity.<br />

Diseases without GVT<br />

It seems likely that some solid tumors, neuroblastoma,<br />

melanoma <strong>and</strong> breast cancer have minimal benefit from a<br />

GVT effect. While GVT effects have been described in<br />

breast cancer they appear to be in only a minority of patients.<br />

Despite the potential for immunotherapy in this disease<br />

allogeneic stem cell transplants for melanoma have not<br />

been promising. After immunoablative conditioning the disease<br />

appears to rapidly accelerate precluding any delayed<br />

benefit from the GVT effect.<br />

The future – disease control without excessive<br />

conditioning intensity<br />

Improvements in the present status of SCT for malignant<br />

disease await developments in two <strong>are</strong>as; (1) Augmentation<br />

of the GVT /GVL effect. Areas of investigation include<br />

ways to generate tumor-specific or minor antigen-specific<br />

CTL to prevent or treat relapse, use of tumor specific vaccines,<br />

<strong>and</strong> augmentation of NK mediated GVL through NK<br />

infusions <strong>and</strong> cytokines to boost NK function4,5 . Other promising<br />

approaches <strong>are</strong> the use of demethylating agents to<br />

upregulate presentation of cancer-testis antigens on malignant<br />

cells <strong>and</strong> the use of cytokines <strong>and</strong> immunostimulatory<br />

agents such as revlemid (upregulated CTL generation) 5 , <strong>and</strong><br />

bortezomib (upregulated TRAIL mediate killing) 6 .<br />

(2) Delivering focused antimalignant therapy: The introduction<br />

of »small molecules« – tyrosine kinase inhibitors<br />

(TkI), <strong>and</strong> targeted therapy of malignancy while efficacious<br />

as a st<strong>and</strong> alone treatment in CML may best be combined<br />

with SCT. In Ph+ ALL, TkI with SCT may improve the<br />

chances of disease-free survival 7 . Sirolimus appears to have<br />

a unique double therapeutic effect of controlling GVHD<br />

while having a direct anti-lymphoma effect 8 . Lastly the use<br />

of radio-isotopes to target the bone marrow may be a promising<br />

way to deliver high dose treatment to the <strong>are</strong>as where<br />

leukemia resides while avoiding general organ toxicity 9 .<br />

R E F E R E N C E S<br />

1. Barnes DW, Corp MJ, Loutit, JF, Neal FE. Treatment of murine leukaemia<br />

with X rays <strong>and</strong> homologous bone marrow; preliminary communication.<br />

Br Med J 1956;2(4993):626–7.<br />

2. Horowitz MM, Gale RP, Sondel PM, Goldman JM, Kersey J, Kolb HJ,<br />

Rimm AA, Ringdén O, Rozman C, Speck B, et al. Graft-versus-leukemia<br />

reactions after bone marrow transplantation. Blood 1990;75:555–62.<br />

3. Kolb HJ, Mittermüller J, Clemm C, Holler E, Ledderose G, Brehm G,<br />

Heim M, Wilmanns W Donor leukocyte transfusions for treatment of<br />

recurrent chronic myelogenous leukemia in marrow transplant patients.<br />

Blood 1990;76:2462–5.<br />

4. Barrett J, Rezvani K. Immunotherapy: Can we include vaccines with<br />

stem-cell transplantation? Nat Rev Clin Oncol 2009;6:503–5.<br />

5. Parmar S, Fern<strong>and</strong>ez-Vina M, de Lima M.Novel transplant strategies for<br />

generating graft-versus-leukemia effect in acute myeloid leukemia. Curr<br />

Opin Hematol 2011;18:98–104.<br />

6. Yong AS, Keyvanfar K, Hensel N, Eniafe R, Savani BN, Berg M, Lundqvist<br />

A, Adams S, Slo<strong>and</strong> EM, Goldman JM, Childs R, Barrett AJ.<br />

Primitive quiescent CD34+ cells in chronic myeloid leukemia <strong>are</strong> targeted<br />

by in vitro exp<strong>and</strong>ed natural killer cells, which <strong>are</strong> functionally<br />

enhanced by bortezomib. Blood 2009;113:875–82.<br />

7. Lee HJ, Thompson JE, Wang ES, Wetzler M. Philadelphia chromosomepositive<br />

acute lymphoblastic leukemia: current treatment <strong>and</strong> future perspectives.<br />

Cancer 2011;117:1583–94.<br />

8. Arm<strong>and</strong> P, Gannamaneni S, Kim HT, Cutler CS, Ho VT, Koreth J, Alyea<br />

EP, LaCasce AS, Jacobsen ED, Fisher DC, Brown JR, Canellos GP,<br />

Freedman AS, Soiffer RJ, Antin JH. Improved survival in lymphoma<br />

patients receiving sirolimus for graft-versus-host disease prophylaxis<br />

after allogeneic hematopoietic stem-cell transplantation with reducedintensity<br />

conditioning. J Clin Oncol 2008;26:5767–74.<br />

9. Pagel JM, Kenoyer AL, Bäck T, Hamlin DK, Wilbur DS, Fisher DR,<br />

Park SI, Frayo S, Axtman A, Orgun N, Orozco J, Shenoi J, Lin Y, Gopal<br />

AK, Green DJ, Appelbaum FR, Press OW. Anti-CD45 pretargeted radioimmunotherapy<br />

using bismuth-213: high rates of complete remission<br />

<strong>and</strong> long-term survival in a mouse myeloid leukemia xenograft model.<br />

Blood 2011;118:703–1.<br />

OP54 Infectious Complications of Stem Cell<br />

Transplantation<br />

Finn B. Petersen, Intermountain Blood <strong>and</strong> Marrow<br />

Transplant Program, Salt Lake City, Utah, USA<br />

Introduction<br />

Despite major advances in detecting, preventing <strong>and</strong> treating<br />

infections associated with any type of stem cell transplantation<br />

(SCT), non-relapse mortality after SCT r<strong>are</strong>ly<br />

occurs without a direct or indirect contribution from an infectious<br />

episode. Thus, by 2009, infections were reported as<br />

a primary cause of mortality in up to 8% of autologous, <strong>and</strong><br />

up to 20% of allogeneic SCT recipients1 . For this reason,<br />

ongoing focus on cost-effective means to detect, prevent<br />

57


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

<strong>and</strong> treat SCT associated infections need to remain a major<br />

focal point of SCT c<strong>are</strong>.<br />

Methods<br />

5 complementary approaches <strong>are</strong> commonly used by SCT<br />

clinicians to minimize the risk of morbidity <strong>and</strong> mortality<br />

from infectious diseases: 1) prophylactic modalities, 2) empiric<br />

modalities, 3) pre-emptive modalities, 4) therapeutic<br />

modalities, <strong>and</strong> 5) salvage modalities. Which modality will<br />

be most effective for a given situation or patient population<br />

for prophylaxis depends solely on the severity of infection<br />

to be prevented, the cost, <strong>and</strong> expected adverse events, for<br />

empiric therapy on signs or symptoms of infection vs. the<br />

inability of early detection, for pre-emptive therapy on the<br />

ability for early detection vs. available effective therapy, for<br />

therapy on having effective therapeutics available, <strong>and</strong> for<br />

salvage on having effective second line therapies available<br />

in case of primary treatment failure.<br />

Major Advances<br />

Since the beginning of clinical SCT, <strong>and</strong> the recognition<br />

of the high morbidity <strong>and</strong> mortality from infections in this<br />

patient population, infection prevention <strong>and</strong> control have<br />

been a main <strong>are</strong>a of clinical SCT research, already in the<br />

1970’s documenting the benefit of protective environments,<br />

prophylactic antimicrobials <strong>and</strong> rapid diagnosis of the most<br />

serious infections.<br />

The most significant advances over the past 25 years in<br />

reducing the impact <strong>and</strong> threat of infectious diseases in SCT<br />

<strong>are</strong> shown in table 1. However, these improvements have<br />

most often been used to exp<strong>and</strong> SCT treatments to older <strong>and</strong><br />

higher risk patients, or for permitting us of more potent immunesuppression<br />

with the SCT, for a net stable treatment<br />

related morbidity <strong>and</strong> mortality from infectious diseases<br />

over time.<br />

Future Directions<br />

Aside from continuing to refine <strong>and</strong> adapt the modalities<br />

listed in table 1 to the ever changing transplant <strong>and</strong> microbiological<br />

environments, future improvements in reducing<br />

the impact of infectious diseases associated with SCT may<br />

Table 1. Modalities to reduce the impact of infectious diseases in stem<br />

cell transplantation<br />

Modality<br />

Year<br />

Introduced<br />

Use<br />

Broad Spectrum Systemic Antibiotics 1986 P, E, PE, T, S<br />

Acyclovir 1983 P, E, PE, T<br />

Post-transplant Filgrastim 1989 P, E, PE<br />

Peripheral Blood Stem Cells 1990 P<br />

1st generation triazoles (fluconazol) 1990 P, E, PE, T<br />

Quinolone Prophylaxis during neutropenia 1991 P, T<br />

CMV Early Diagnosis / Treatment 1992 P, E, PE, T<br />

Lipid Compound Polyenes 1992 E, T, S<br />

Echinoc<strong>and</strong>ens / 2nd generation triazoles 2000 P, E, PE, T, S<br />

Pre-/post-transplant Palifermin 2005 P<br />

P=Prophylactic, E=Empiric, PE=pre-emptive, T=therapeutic, S=Salvage.<br />

Bolded letters designate primary use.<br />

58<br />

come from clinical adaptation of single nucleotide polymorphisms<br />

(SNPs) of toll-like receptor (TLR) or similar inflammatory<br />

response associated genes, potentially permitting an<br />

infectious disease risk allocations of patients based on specific<br />

infection response genotypes.<br />

Also, a better ability to promote earlier <strong>and</strong> more complete<br />

post-transplant immune reconstitution, <strong>and</strong> to be able<br />

to control antigen-discrepancies between donor <strong>and</strong> recipient<br />

without having to use infection facilitating immunosuppressive<br />

agents such as corticosteroids <strong>and</strong> tumor-necrosisfactor<br />

blockers, would promote infection control in SCT<br />

patients. Lastly, shifting all anti-infectious activities from<br />

empiric <strong>and</strong> therapeutic to prophylactic <strong>and</strong> pre-emptive use<br />

also would represent a significant step forward, but the realization<br />

of such a goal will depend on further development of<br />

sensitive <strong>and</strong> microbe specific early detection tests, of which<br />

only a few <strong>are</strong> available for clinical use today. Finally, in<br />

today’s strained budgetary environment, an ongoing weighing<br />

of the use of often expensive, but effective, infection<br />

prophylaxis modalities against the cost, both monetary <strong>and</strong><br />

human, of devastating <strong>and</strong> potentially fatal infections, is an<br />

absolute necessity.<br />

Of help to the transplant clinicians <strong>are</strong> an increasing body<br />

of evidence based guidelines published by specialty groups<br />

<strong>and</strong> societies, such as what the major national <strong>and</strong> international<br />

societies involved in SCT did in their most recent update<br />

of »Guidelines for preventing infectious complications<br />

among hematopoietic cell transplant recipients: a global<br />

perspective« in a special issue of Bone Marrow Transplantation<br />

published in 2009 1<br />

Conclusion<br />

While major improvements in detection, prevention <strong>and</strong><br />

therapy of SCT related infectious diseases have been significant<br />

over time, <strong>and</strong> permitted SCT of higher risk patients<br />

<strong>and</strong> using higher risk transplants, the microbiological, transplant,<br />

<strong>and</strong> regulatory fields <strong>are</strong> constantly evolving in unpredictable<br />

ways, presenting clinical stem cell transplant<br />

clinicians with ever new infectious disease challenges.<br />

R E F E R E N C E S<br />

1. M Tomblyn et al: »Guidelines for preventing infectious complications<br />

among hematopoietic cell transplant recipients: a global perspective«.<br />

Bone Marrow Transplantation 2009;44:453–526<br />

OP55 Old drug, new tricks in prevention of graftversus-host<br />

disease (GVHD): Development<br />

<strong>and</strong> clinical applicability of high-dose<br />

cyclophosphamide<br />

Leo Luznik, Division of Hematologic Malignancies;<br />

Department of Oncology, Sidney Kimmel Comprehensive<br />

Cancer Center at Johns Hopkins, Baltimore, MD. USA<br />

GVHD is a major complication of allogeneic hematopoietic<br />

stem cell transplantation (alloHSCT). The successful<br />

prevention of acute GVHD with CNIs <strong>and</strong> methotrexate<br />

(MTX) was developed in the 1980s <strong>and</strong> has revolutionized<br />

the field of alloHSCT; however, at least 50% of patients still


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

develop this complication.(1, 2) There is currently no successful<br />

strategy for the prevention of chronic GVHD, the<br />

most common cause of late morbidity <strong>and</strong> mortality. It affects<br />

over 50% of patients who would otherwise have been<br />

cured of their primary malignancy.(3) Mechanistically, immunosuppressive<br />

drugs given to control GVHD suppress<br />

alloimmunity by the nonspecific inhibition of alloreactive T<br />

cell activation, proliferation, <strong>and</strong> differentiation. By blocking<br />

T-cell activation, currently used agents in the GVHD<br />

prophylaxis regimens interfere with the apoptosis of the<br />

alloreactive T cells, causing global immunosuppression<br />

<strong>and</strong> a delay in the time for induction of transplantation<br />

tolerance. Of all the commonly used immunosuppressants,<br />

only MTX <strong>and</strong> cyclophosphamide (Cy) can induce the<br />

apoptosis of alloantigen-activated human T cells.(4) MTX,<br />

although active as a single agent for GVHD prevention, due<br />

to its intrinsic toxicity profile, it cannot be administered in<br />

higher doses critical for inducing the en masse alloreactive<br />

T-cell apoptosis. On the other h<strong>and</strong>, Cy can safely be administered<br />

in high-doses, which <strong>are</strong> required to eliminate alloreactive<br />

clones after alloHSCT because of its favorable safety<br />

profile, including lack of toxicity to the primitive hematopoietic<br />

stem cells.(5) Our recently published results suggest<br />

that high-dose Cy is sufficient as a single agent in the<br />

prevention of both acute <strong>and</strong> chronic GVHD after myeloablative<br />

conditioning <strong>and</strong> allografting using HLA-matched<br />

related <strong>and</strong> unrelated donors.(6) Patients transplanted using<br />

this strategy have a low incidence of infections <strong>and</strong>, despite<br />

the advanced disease stage, their survival is favorable, suggesting<br />

that this approach retains the graft-versus-leukemia<br />

(GVL) effect. Our group has been also exploring high dose,<br />

post-transplantation Cy as prophylaxis of GVHD after nonmyeloablative,<br />

HLA-haploidentical bone marrow transplantation,<br />

or mini-haploBMT.(7) Among 210 recipients of<br />

mini-haploBMT, 87% of patients have experienced sustained<br />

donor cell engraftment. The cumulative incidences of<br />

grades II-IV acute GVHD <strong>and</strong> chronic GVHD <strong>are</strong> 27% <strong>and</strong><br />

13%, respectively. Five-year cumulative incidence of nonrelapse<br />

mortality is 18% <strong>and</strong> actuarial overall survival <strong>and</strong><br />

event-free survivals <strong>are</strong> 35% <strong>and</strong> 27%, respectively. Recent<br />

multi-center phase II trial conducted by Blood <strong>and</strong> Marrow<br />

Transplant Clinical Trials Network confirmed the validity<br />

of posttransplant Cy in GVHD prevention across HLA-barriers.(8)<br />

Survival rates after posttransplant Cy-based minihaploBMT<br />

were comparable to survival rates in patients<br />

with high-risk hematologic malignancies who were transplanted<br />

with blood or marrow from HLA- matched unrelated<br />

donors after reduced conditioning. In conclusion, by<br />

taking advantage of the differential susceptibility of proliferating,<br />

alloreactive T cells over non-proliferating, non-alloreactive<br />

T cells to high dose Cy, this novel strategy provides<br />

a unique opportunity to optimize GvHD prophylaxis<br />

after HLA-matched alloBMT <strong>and</strong> increase the use of HLAmismatched<br />

donors. (9) (10)<br />

R E F E R E N C E S<br />

1. Appelbaum FR. Hematopoietic-cell transplantation at 50. N Engl J<br />

Med 2007;357:1472–1475.<br />

2. Storb R, Deeg HJ, Whitehead J, Appelbaum F, Beatty P, Bensinger W,<br />

Buckner CD, Clift R, Doney K, F<strong>are</strong>well V, et al. Methotrexate <strong>and</strong><br />

cyclosporine comp<strong>are</strong>d with cyclosporine alone for prophylaxis of<br />

acute graft versus host disease after marrow transplantation for leukemia.<br />

N Engl J Med 1986;314:729–735.<br />

3. Lee SJ. Have we made progress in the management of chronic graft-vshost<br />

disease? Best Pract Res Clin Haematol 23:529–535.<br />

4. Strauss G, Osen W, Debatin KM. Induction of apoptosis <strong>and</strong> modulation<br />

of activation <strong>and</strong> effector function in T cells by immunosuppressive<br />

drugs. Clin Exp Immunol 2002;128:255–266.<br />

5. Jones RJ, Barber JP, Vala MS, Collector MI, Kaufmann SH, Ludeman<br />

SM, Colvin OM, Hilton J. Assessment of aldehyde dehydrogenase in<br />

viable cells. Blood 1995;85:2742–2746.<br />

6. Luznik L, Bolanos-Meade J, Zahurak M, Chen AR, Smith BD, Brodsky<br />

R, Huff CA, Borrello I, Matsui W, Powell JD, Kasamon Y, Goodman<br />

SN, Hess A, Levitsky HI, Ambinder RF, Jones RJ, Fuchs EJ. High-dose<br />

cyclophosphamide as single-agent, short-course prophylaxis of graftversus-host<br />

disease. Blood 115:3224–3230.<br />

7. Luznik L, O’Donnell PV, Symons HJ, Chen AR, Leffell MS, Zahurak<br />

M, Gooley TA, Piantadosi S, Kaup M, Ambinder RF, Huff CA, Matsui<br />

W, Bolanos-Meade J, Borrello I, Powell JD, Harrington E, Warnock S,<br />

Flowers M, Brodsky RA, S<strong>and</strong>maier BM, Storb RF, Jones RJ, Fuchs<br />

EJ. HLA-haploidentical bone marrow transplantation for hematologic<br />

malignancies using nonmyeloablative conditioning <strong>and</strong> high-dose,<br />

posttransplantation cyclophosphamide. Biol Blood Marrow Transplant<br />

2008;14:641–650.<br />

8. Brunstein CG, Fuchs EJ, Carter SL, Karanes C, Costa LJ, Wu J, Devine<br />

SM, Wingard JR, Aljitawi OS, Cutler CS, Jagasia MH, Ballen KK,<br />

Eapen M, O’Donnell PV. Alternative donor transplantation after reduced<br />

intensity conditioning: results of parallel phase 2 trials using<br />

partially HLA-mismatched related bone marrow or unrelated double<br />

umbilical cord blood grafts. Blood 118:282–288.<br />

9. Luznik L, Fuchs EJ. High-dose, post-transplantation cyclophosphamide<br />

to promote graft-host tolerance after allogeneic hematopoietic<br />

stem cell transplantation. Immunol Res 47:65–77.<br />

10. Luznik L, Jones RJ, Fuchs EJ. High-dose cyclophosphamide for graftversus-host<br />

disease prevention. Curr Opin Hematol 17:493–499.<br />

OP56 Chronic Graft-versus-Host Disease<br />

– Anything New in Prevention <strong>and</strong><br />

Treatment?<br />

Steven Zivko Pavletic, National Cancer Institute,<br />

Bethesda, Maryl<strong>and</strong>, USA<br />

Chronic graft-versus-host disease (cGVHD) is the leading<br />

cause of non-relapse morbidity <strong>and</strong> mortality in survivors<br />

after allogeneic hematopoietic stem cell transplantation<br />

(HSCT), but is also associated with lower malignancy<br />

relapse rates. In 2005 an NIH Consensus Conference galvanized<br />

the cGVHD clinical research field by providing st<strong>and</strong>ardized<br />

definitions about diagnosis, staging, response criteria,<br />

data collection <strong>and</strong> conduct of clinical trials. These<br />

consensus documents set the stage for a number of prospective<br />

cohort studies in the United States <strong>and</strong> internationally<br />

which <strong>are</strong> currently in progress.<br />

In spite of effective measures available for preventing<br />

acute GVHD, until recently, there were no signs that similar<br />

preventive strategies could be developed for cGVHD. However,<br />

emerging data from cohort studies <strong>and</strong> clinical trials<br />

indicate <strong>are</strong>as where progress could be made. The key challenges<br />

for prevention in cGVHD <strong>are</strong> the lack of specificity<br />

of the current cGVHD therapies <strong>and</strong> our poor underst<strong>and</strong>ing<br />

of the graft-versus-malignancy effect (GVM) targets. There<br />

is a hope that ongoing studies which integrate in depth investigation<br />

of the cGVHD biology in conjunction with well<br />

annotated patient tissue samples will generate necessary information<br />

<strong>and</strong> lead towards the development of early tar-<br />

59


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

geted therapeutic interventions to prevent severe cGVHD.<br />

Parallel research is needed to identify precisely the GVM<br />

targets on the malignant cells or to integrate some of the<br />

newly emerging anti-cancer drugs in the cGVHD prevention<br />

<strong>and</strong> treatment algorithms. One of the strategies to develop<br />

better timing of cGVHD prevention is to identify<br />

markers of cGVHD activity that differentiate active disease<br />

manifestations from those that <strong>are</strong> caused by the irreversible<br />

damage.<br />

In the <strong>are</strong>a of front line therapy there have been several<br />

attempts to bring promising agents as first line in addition to<br />

steroids. None of these combinations have succeeded in improving<br />

symptoms of cGVHD <strong>and</strong> commonly have been associated<br />

with prohibitive side effects including increased<br />

relapse. Therefore 0.5–1.0 mg/kg/day of prednisone remains<br />

the st<strong>and</strong>ard front line systemic treatment for cGVHD.<br />

Unfortunately, about 50% of cGVHD patients fail steroids<br />

<strong>and</strong> need second line treatments. Currently there is no st<strong>and</strong>ard<br />

recommended second line therapy for cGVHD <strong>and</strong> the<br />

choice of agent is based on the patient <strong>and</strong> clinician preferences,<br />

prior treatment history <strong>and</strong> expected side effects profile.<br />

Agents which <strong>are</strong> most commonly used in steroid refractory<br />

cGVHD include extracorporeal photopheresis, mycophenolate,<br />

sirolimus, rituximab, pentostatin <strong>and</strong> imatinib.<br />

Currently st<strong>and</strong>ard second line therapy is a clinical trial if<br />

available. The treatment of the bronchiolitis obliterans syndrome<br />

(BOS) which is the lung manifestation of cGVHD<br />

poses a specific challenge to clinicians due to the high mortality<br />

<strong>and</strong> lack of effective treatments for this most devastating<br />

presentation of cGVHD. BOS is increasingly recognized<br />

as an <strong>are</strong>a for clinical research. In summary, although little<br />

progress in improving our ability to prevent or treat cGVHD<br />

can yet be decl<strong>are</strong>d, there <strong>are</strong> a number of promising basic<br />

research studies <strong>and</strong> clinical trials which suggest significant<br />

breakthroughs in the near future.<br />

OP57 Relapse after Allogeneic Hematopoietic<br />

Stem Cell Transplantation – The Necessity<br />

for New Strategies <strong>and</strong> Treatments<br />

Michael R. Bishop, Medical College of Wisconsin,<br />

Milwaukee, Wisconsin, USA<br />

Recurrence of disease is the leading cause of mortality<br />

after allogeneic hematopoietic stem cell transplantation (alloHSCT),<br />

whose efficacy is largely attributed to graft-versus-tumor<br />

(GVT) effects. There <strong>are</strong> several factors that <strong>are</strong><br />

responsible for the biologic resistance of malignancies to<br />

beneficial effects of alloHSCT including clonal evolution of<br />

cancer, drug <strong>and</strong> radiation resistance, cancer epigenetics,<br />

<strong>and</strong> possibly cancer stem cells. However, several other factors<br />

affect the occurrence <strong>and</strong> outcome of relapse, including<br />

conditioning regimen, type of allograft, <strong>and</strong> disease histology,<br />

status, <strong>and</strong> sensitivity to chemotherapy. Optimally strategies<br />

<strong>are</strong> needed to precisely balance immune responses to<br />

favor GVT without harmful graft-versus-host disease to protect<br />

against relapse <strong>and</strong> treat recurrent disease after HSCT.<br />

The outcomes of patients relapsing after an alloHSCT generally<br />

remain poor even with the use of donor lymphocytes<br />

infusions (DLI). Developing strategies to promptly identify<br />

60<br />

patients at risk for relapse after alloHSCT, while malignancy<br />

is in a more treatable stage, could decrease relapse rates<br />

after alloHSCT. Monitoring of minimal residual disease<br />

(MRD) post-transplant, through methods such as molecular<br />

genetics, tumor-specific molecular primers, fluorescein in<br />

situ hybridization, <strong>and</strong> multi-parameter flow cytometry,<br />

could lead to novel preemptive treatments of relapse. Studies<br />

<strong>are</strong> also needed to provide data with detail on disease<br />

status, prior treatments, biologic markers, <strong>and</strong> post-transplant<br />

events to identify patients at high risk for relapse. The<br />

development of strategies to intervene prior to florid relapse<br />

may include non-immunologic therapies, such as targeted<br />

preparative regimens <strong>and</strong> post-transplant drug therapy, as<br />

well as immunologic interventions, including graft <strong>and</strong> Tcell<br />

engineering, vaccination, <strong>and</strong> dendritic cell-based approaches.<br />

There is no st<strong>and</strong>ard approach to treating relapse<br />

after alloHSCT. Withdrawal of immune suppression <strong>and</strong><br />

DLI <strong>are</strong> commonly used for all diseases; however, the efficacy<br />

of DLI is quite variable depending on disease histology<br />

<strong>and</strong> state <strong>and</strong> has limited efficacy in the majority of hematologic<br />

malignancies. As such, there is a significant need for<br />

novel therapies, strategies, <strong>and</strong> well-designed, disease-specific<br />

trials for relapse following alloHSCT, particularly in<br />

patients for whom DLI is not an option. The 2009 National<br />

Cancer Institute International Workshop on the Biology,<br />

Prevention, <strong>and</strong> Treatment of Relapse after Allogeneic Hematopoietic<br />

Stem Cell Transplantation was convened to<br />

identify, prioritize, <strong>and</strong> coordinate future research activities<br />

related to relapse after alloHSCT. The Workshop suggested<br />

3 major initiatives for a coordinated research effort to address<br />

the problem of relapse after alloHSCT: (1) to establish<br />

multicenter correlative <strong>and</strong> clinical trial networks for basic/<br />

translational, epidemiologic, <strong>and</strong> clinical research; (2) to establish<br />

a network of biorepositories for the collection of<br />

samples before <strong>and</strong> after alloHSCT to aid in laboratory <strong>and</strong><br />

clinical studies; <strong>and</strong> (3) to further refine, implement, <strong>and</strong><br />

study the Workshop-proposed definitions for disease-specific<br />

response <strong>and</strong> relapse <strong>and</strong> recommendations for monitoring<br />

of MRD. These recommendations, in coordination<br />

with ongoing research initiatives <strong>and</strong> transplantation organizations,<br />

provide a research framework to rapidly <strong>and</strong> efficiently<br />

address the significant problem of relapse after alloHSCT.<br />

OP58 Design, conduct <strong>and</strong> analysis of clinical<br />

trials in acute leukemia: the keys of success<br />

Stefan Suciu, EORTC Headquaters, Brussels, Belgium<br />

Clinical trials <strong>are</strong> experiments performed in medical settings<br />

in order to determine maximum tolerated dose of a<br />

drug/regimen (phase I), its activity (phase II) or efficacy<br />

(phase III) in a given patient population. In order to shorten<br />

the drug development, phase I/II, or phase II/III, or even<br />

phase I/III trials may be set-up as well. Phase I <strong>and</strong> II trials<br />

require a limited number of patients, as their endpoints <strong>are</strong><br />

toxicity <strong>and</strong> activity (e.g. CR rate) respectively. In order to<br />

detect moderate treatment differences in term of efficacy,<br />

r<strong>and</strong>omized phase III trials require large numbers of patients<br />

<strong>and</strong> long follow-up. Such trials <strong>are</strong> sized according to a pri-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

mary endpoint, generally overall survival, event-free survival,<br />

progression-free survival or disease-free survival (for<br />

patients who reached CR). Secondary endpoints contain remaining<br />

efficacy endpoints, CR rate, highest toxicity/adverse<br />

events observed during treatment courses, time to recovery<br />

<strong>and</strong>, if possible, quality-of-life evaluations. Longterm<br />

outcomes <strong>and</strong> toxicities, incidences of secondary tumors<br />

<strong>are</strong> evaluated as well, in case of potentially toxic or<br />

long lasting treatments (drugs, drug combinations, irradiations).<br />

A phase III trial may contain several r<strong>and</strong>omized questions.<br />

If no interactions between them <strong>are</strong> foreseen, such<br />

factorial designs represent an efficient way to use a patient<br />

population. To overcome possible interactions, 10–20% more<br />

patients should be added as comp<strong>are</strong>d to a classical one<br />

question trial.<br />

Acute leukemias, lymphoblastic (ALL) or myeloblastic<br />

(AML), <strong>are</strong> heterogeneous diseases. In AML distinct entities<br />

have been determined (e.g. APL, t(8;21)/inv(16), FLT3,<br />

etc). Age <strong>and</strong> performance status <strong>are</strong> important patient-factors<br />

to adjust treatment intensity. In the personalised treatment<br />

era, patient population available to enter into a phase<br />

III trial becomes quite limited, even within a large cooperative<br />

group. Intergroup trials should be set-up in order to<br />

reach adequate patient accrual. The leading group, in cooperation<br />

with a Company, should set-up the protocol, with<br />

the input of other interested groups. Agreements regarding<br />

submissions (e.g. protocol, SAE) to local regulatory authorities,<br />

data collection process, publication rules <strong>and</strong> authorship,<br />

etc should be set-up <strong>and</strong> applied.<br />

Phase III trial analyses should be done when mature efficacy<br />

data <strong>are</strong> available: final analysis, when the required<br />

number of events is reached, <strong>and</strong> interim looks, if specified<br />

in the protocol. Early stopping rules for efficacy <strong>and</strong>/or futility<br />

should be based on well defined statistical rules. In<br />

order to avoid any conflict of interest, only an Independent<br />

Data Monitoring committee (IDMC) should evaluate the interim<br />

results <strong>and</strong> provide adequate recommendations to the<br />

trialists. If there is sufficient evidence that the experimental<br />

treatment is superior to the st<strong>and</strong>ard one, or the treatment<br />

difference is already too low or goes in the wrong direction,<br />

one may draw early conclusions, <strong>and</strong>, if necessary, switch<br />

patients already entered in the trial, from one arm to another.<br />

Statistical analyses should be done in accordance to the trial<br />

design, using efficient statistical tests. If r<strong>and</strong>omization was<br />

stratified by some prognostic factors (e.g. age <strong>and</strong> WBC),<br />

statistical test (e.g. logrank test or Cox model) should be<br />

stratified/adjusted by these factors, in case a block-size/<br />

minimization method was used for r<strong>and</strong>omization. Such test<br />

will provide unbiased treatment difference estimate, having<br />

an increased efficiency as comp<strong>are</strong>d to the unstratified/unadjusted<br />

test. Adjustments by other important factors could<br />

be done. Interactions between some factors <strong>and</strong> treatment<br />

could be investigated as well, for detection of potential predictive<br />

factors for treatment efficacy.<br />

A successful trial requires a good idea, e.g. the assessment<br />

of a targeted drug to be used alone or in an already<br />

existing treatment scheme, in a »marker«-positive patient<br />

population, in whom the experimental treatment may be applied.<br />

The trial should be properly designed, preferentially<br />

be r<strong>and</strong>omized in order to avoid any systematic bias. Disease<br />

assessments should mirror as much as possible the<br />

usual clinical practice, <strong>and</strong> be scheduled <strong>and</strong> made evenly in<br />

the treatment groups. Efficient data collection regarding the<br />

main <strong>and</strong> secondary endpoints should be done. A good cooperation<br />

of clinical investigators should provide a fast recruitment.<br />

Interim <strong>and</strong> final analyses using proper statistical<br />

technique should be done on (relatively) mature data in order<br />

to be able drawing valid conclusions. In case of any<br />

safety concerns, in some trials (e.g. phase III, r<strong>and</strong>omized<br />

phase I/III), safety data should be monitored by an Independent<br />

Data Safety Monitoring Board (IDSMB), with no conflict<br />

of interest. This should meet regularly in order to provide<br />

adequate recommendations regarding the study conduct.<br />

OP59 Front line therapy for indolent B-cell<br />

lymphoma<br />

Nathan Fowler, Department of Lymphoma <strong>and</strong> Myeloma,<br />

MD Anderson Cancer Center, Houston, Texas, USA<br />

A number of treatment options exist for newly diagnosed<br />

patients with indolent non-Hodgkins lymphoma (iNHL).<br />

Several r<strong>and</strong>omized studies have demonstrated an improvement<br />

in both progression free survival (PFS) <strong>and</strong> overall<br />

survival (OS) with the addition of anti-CD20 directed monoclonal<br />

antibody (MAB) therapy into frontline combinations.<br />

Despite these developments, the majority of advanced<br />

stage patients still relapse with iNHL or undergo aggressive<br />

transformation, <strong>and</strong> improvements in frontline treatment <strong>are</strong><br />

needed. Although maintenance treatment with MABs <strong>and</strong><br />

consolidation with anti-CD20 radioconjugates results in<br />

prolongation of PFS, their effect on OS is unknown. Bendamustine<br />

has significant activity in iNHL, <strong>and</strong> recent r<strong>and</strong>omized<br />

studies suggest a potential role in untreated patients.<br />

Newer agents, such as the immumomodulatory agent<br />

lenalidomide, the proteasome inhibitor bortezomib, <strong>and</strong><br />

drugs targeting essential steps in the B-cell receptor pathway<br />

such as Bruton’s tyrosine kinase (BTK) <strong>and</strong> PI-3 kinase<br />

show promise. Studies <strong>are</strong> currently underway exploring the<br />

potential to incorporate these <strong>and</strong> other novel biologic<br />

agents into frontline treatment for iNHL.<br />

OP60 Rituximab maintenance treatment<br />

in indolent lymphoma<br />

Ofer Sphilberg, Inst. of Hematology, Davidoff Center,<br />

Beilinson Hospital & Tel-Aviv University, Israel<br />

Follicular lymphoma, which represents 15%–30% of newly<br />

diagnosed lymphomas, is an indolent lymphoma that is<br />

characterized by slow growth <strong>and</strong> a high initial response rate<br />

but relapsing <strong>and</strong> progressive disease. Most patients <strong>are</strong> diagnosed<br />

with advanced disease, that is, stage III or IV, <strong>and</strong> cannot<br />

be cured with currently available conventional therapies.<br />

New treatment modalities <strong>are</strong> therefore urgently needed.<br />

The chimeric monoclonal antibody rituximab targeted<br />

against CD20, a protein that is expressed on the surface of<br />

61


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

all mature B cells, is active in many B-cell lymphomas that<br />

express this molecule. Rituximab administered intravenously<br />

in combination with chemotherapy improves overall survival<br />

in patients with newly diagnosed <strong>and</strong> relapsed indolent<br />

lymphoma comp<strong>are</strong>d with chemotherapy alone.<br />

Rituximab maintenance therapy following successful induction<br />

has recently emerged as a highly effective treatment<br />

for follicular lymphoma. R<strong>and</strong>omized clinical trials as well<br />

as systematic review <strong>and</strong> meta-analysis, analyzing the impact<br />

of rituximab maintenance comp<strong>are</strong>d to observation<br />

alone have demonstrated a significantly better outcome in<br />

terms of progression-free survival in patients who received<br />

as first-line treatment single-agent rituximab, st<strong>and</strong>ard chemotherapy<br />

<strong>and</strong> recently also immuno-chemotherapy. In the<br />

setting of relapsed disease, rituximab maintenance has shown<br />

a significant benefit even in terms of overall survival.<br />

These data suggest that maintenance therapy with rituximab<br />

as four weekly infusions every 6 months or as a single<br />

infusion every 2–3 months, should be added to st<strong>and</strong>ard<br />

therapy for patients with follicular lymphoma after successful<br />

induction therapy.<br />

OP61 Risk adapted treatment for patients<br />

with B-DLBCL<br />

M<strong>are</strong>k Trneny, (Czech Republic)<br />

62<br />

Abstract not submitted.<br />

OP62 Treatment of peripheral T-Cell lymphomas<br />

Pier Luigi Zinzani, Institute of Hematology »Seràgnoli«,<br />

University of Bologna, Italy<br />

Up till recent years, no major therapeutic advances have<br />

been observed in peripheral T/NK-cell lymphomas (PTCL),<br />

with overall survival (OS) values for most of the systemic<br />

entities in the order of 25–35% at five years. Furthermore,<br />

there has been a rather consistent lack of PTCL-specific<br />

clinical trials, partly due to the rarity of this condition, <strong>and</strong><br />

partly to diagnostic difficulties <strong>and</strong> controversial views on<br />

whether immunophenotype-specific treatment strategies<br />

should be adopted. As a consequence of the REAL <strong>and</strong> the<br />

subsequent WHO classification of malignant lymphomas,<br />

present day diagnostic approaches identify PTCL entities<br />

based upon immunophenotype in association with clinical,<br />

morphological, <strong>and</strong> molecular genetic data. These advances<br />

have recently led to PTCL-specific clinical trials.<br />

Analogue to the treatment strategy applied in diffuse<br />

large B-cell lymphoma (DLBCL), combination chemotherapy<br />

with CHOP (cyclophosphamide, adriamycin, vincristine,<br />

prednisone) or CHOP-like variants has up till now been<br />

the most frequently used first line therapy also in PTCL.<br />

Given as st<strong>and</strong>ard schedule every three weeks, this approach<br />

does not seem to benefit more than 25–30% of the patients,<br />

while schedule intensification, e.g. shortening of the timeinterval<br />

between courses <strong>and</strong>/or addition of etoposide, has<br />

by some authors been reported to improve outcome. However,<br />

reported OS values have been varying, often depend-<br />

ing on the included number of prognostically more favourable<br />

cases belonging to the histological subtype anaplastic<br />

large cell lymphoma (ALCL), alk-positive.<br />

Based on small-scale observations on single-agent efficacy,<br />

gemcitabine has also been investigated in some PTCLspecific<br />

trials. Response rates were encouraging, but of relatively<br />

short duration.<br />

High-dose therapy with autologous stem cell transplant<br />

(HDT/ASCT) proved feasible in both relapsed/refractory<br />

<strong>and</strong> treatment-naïve PTCL. Overall results suggest a more<br />

favorable impact of this approach in first line as comp<strong>are</strong>d<br />

to salvage treatment. However, most data <strong>are</strong> of selected <strong>and</strong><br />

retrospective nature. At present, only a few PTCL-restricted<br />

phase II trials investigating HDT/ASCT as part of a firstline<br />

therapeutic approach have been reported. Of these only<br />

very few have a sufficient cohort-size to compensate for the<br />

heterogeneity of this disorder.<br />

Combinations of chemotherapy with T-cell specific monoclonal<br />

antibodies (MoAb) have shown high overall response<br />

rates (ORR) <strong>and</strong> <strong>are</strong> currently being investigated in<br />

ongoing clinical trials.<br />

Due to the rarity of T/NK-cell neoplasms, no PTCL-restricted<br />

phase III trials have yet been completed. However,<br />

the first international efforts have recently been launched.<br />

OP63 Therapy of Mantle cell Lymphoma<br />

Christian Geisler, Rigshospitalet, Copenhagen,<br />

Denmark<br />

The treatment of choice in younger, fit MCL patients is<br />

induction immunochemotherapy followed by high-dose<br />

therapy with autologous stem cell support. Phase-II data<br />

have strongly suggested that Ara-C containing immunochemotherapy<br />

is superior to R-CHOP, <strong>and</strong> this has now been<br />

confirmed by a r<strong>and</strong>omized trial by the European MCL Network.<br />

In elderly/frail patients, however, R-CHOP was superior<br />

to R – fludarabine + cyclophosphamide, possibly as the<br />

combined net effect of efficacy <strong>and</strong> tolerability. The combination<br />

of R-CHOP with rituximab maintenance has furher<br />

prolonged progression-free survival <strong>and</strong> may be the regimen<br />

of choice in elderly frail patients, but needs testing<br />

against R-AraC. The treatment goal in both settings is complete<br />

clinical <strong>and</strong> molecula remission.Targeted therapy with<br />

proteasome inhibitors, m-Tor inhibitors <strong>and</strong> other new compounds<br />

represents promising <strong>are</strong>as of development in firstline<br />

<strong>and</strong> salvage treatment.<br />

OP64 Is there a role of radioimmunotherapy<br />

in NHL<br />

OP65 Therapy for relapsed/refractory aggressive<br />

NHL<br />

Anas Younes, (USA)<br />

Abstract not submitted.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

OP66 Alternative monoclonal antibodies<br />

for the treatment of NHL<br />

Luis Fayad, (USA)<br />

Abstract not submitted.<br />

OP67 Early favorable <strong>and</strong> unfavorable Hodgkin<br />

lymphoma in adults, st<strong>and</strong>ards <strong>and</strong><br />

directions<br />

Patrice Carde, Institut Gustave Roussy, Villejuif, France<br />

Few malignancies, if any, benefited a step-by-step improvement<br />

in tumor control <strong>and</strong> survival rates as early favorable<br />

<strong>and</strong> unfavorable Hodgkin lymphoma. Competition<br />

between Cooperative groups through to rigorous prospective<br />

r<strong>and</strong>omized phase III trials result in the current proposal<br />

of an optimal treatment balancing relapse <strong>and</strong> longterm<br />

risks. St<strong>and</strong>ard treatment consists in 2 to 3 cycles of<br />

ABVD followed by 20–30 Gy involved field radiation therapy<br />

(IFRT) in early favorable <strong>and</strong> in a minimum of 4 cycles<br />

<strong>and</strong> of 30 Gy in early unfavorable stages (E. Lugtenburg &<br />

A. Hagenbeek Treatment of Early Favorable HL chapt. 10,<br />

pp. 179 in Hodgkin Lymphoma -A. Engert <strong>and</strong> S.J. Horning<br />

(eds.) DOI: 10.1007/978-3-642-12780-9_11, © Springer-<br />

Verlag Berlin Heidelberg 2011; Raemaekers JJJ & Engert A<br />

Treatment of Early Unfavorable HL, chapt. 11, pp. 185–187<br />

ibid); These st<strong>and</strong>ards provide EFS/PFS/FFTF <strong>and</strong> OS rates<br />

of 93% <strong>and</strong> 97%, respectively for early favorable stages<br />

(GHSG HD-10) <strong>and</strong> of 91 an >97%, respectively for early<br />

unfavorable stages (GHSG HD-14); see Table 1.<br />

Taking into account the already very high level of the current<br />

results, the relatively short follow-up of 3–5 years for<br />

most of these studies <strong>and</strong>, unhappily, the rarity of late reports,<br />

due to the hard work <strong>and</strong> low reward from long-term<br />

updates, only dismal differences can be expected between<br />

treatment arms, even adequately powered, as in the HD-13<br />

GHSG study looking at the value of each of the individual<br />

ABVD components. For instance the maximal Progression<br />

free survival (PFS) differential seen among recent trials has<br />

been the 91 to 97% 3-year yield (p


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

Time-dependent covariates (markers, time-to-response,<br />

»quality« or »speed« of response) <strong>are</strong> particularly subject to<br />

flaws for 2 reasons: (1) they <strong>are</strong> highly treatment-dependent<br />

(2) as occasional brilliant predictors of response, they <strong>are</strong> -<br />

wrongly- expected to direct the treatment strategy, <strong>and</strong><br />

therefore improve global survival. Actually, even identification<br />

of a poor-faring group in no way means that a more<br />

adequate treatment is available for this precise group. The<br />

EORTC paved the way for such a strategy in advanced HL<br />

(Carde P. Early response to chemotherapy (CT) is a faithful<br />

surrogate independent predictive factor that can influence<br />

treatment strategy in Hodgkin’s Disease (HD). Proc. Fifth<br />

International Symposium on Hodgkin’s Lymphoma 22–25<br />

September 2001, Köln, Germany. Leukemia & Lymphoma<br />

42,suppl.2 :35,2001 (oral presentation, abst. # I-40)).<br />

FDG-PET scans better assess tumor burden than CT scans<br />

<strong>and</strong> functional imaging has been shown to predict outcome<br />

after combined modality therapy in early stage HL with a<br />

very high negative predictive value (Hutchings M, Loft A,<br />

Hansen M, et al: FDG PET after two cycles of chemotherapy<br />

predicts treatment failure <strong>and</strong> progression-free survival in<br />

Hodgkin lymphoma. Blood 107:52–59, 2006). FDG-PET,<br />

already known important in staging <strong>and</strong> treatment evaluation<br />

recently became fashionable to adapt the treatment, for less,<br />

or more...<br />

Calling for less treatment recently proved disappointing,<br />

according to the results of the H10 International/EORTC<br />

study, where deleting involved node irradiation (IN RT) in<br />

PET 2 negative patients (early favorable <strong>and</strong> unfavorable<br />

stages) resulted in lower PFS (XXXXXX). Other cooperative<br />

groups attempt not to duplicate these results.<br />

Calling for more treatment in early positive PET 2 patients<br />

<strong>and</strong> being successful is tricky, apart from the false positives,<br />

that in part depend upon the treatment; indeed, most patients<br />

with positive interim PET <strong>are</strong> cured, so that the positive predictive<br />

value in early stage has been exceeded 15% in a recent<br />

study (Sher DH, Mauch PM, Van Deen AAet al. Prognostic<br />

significance of mid- <strong>and</strong> post ABVD PET imaging in<br />

Hodgkin’s lymphoma: the importance of involved – field<br />

radiotherapy. Ann Oncol. 2009;20(11):1848–53). In this respect<br />

the result of the International/EORTC H10 trial is eagerly<br />

awaited for.<br />

TARGETED TREATMENT TO HODGKIN <strong>LYMPHOMA</strong><br />

TUMOR CELLS<br />

Targeted therapy to receptors <strong>and</strong> antigens expressed by<br />

the Hodgkin <strong>and</strong> Reed Sternberg cells (HRS), proves efficient,<br />

whether or not these <strong>are</strong> the ultimate tumor stem cells<br />

(A. Younes <strong>and</strong> A. Engert. New agents for patients with<br />

Hodgkin lymphoma. chapt. 20, pp. 283–294 in Hodgkin<br />

Lymphoma -A. Engert <strong>and</strong> S.J. Horning (eds.) DOI:<br />

10.1007/978-3-642-12780-9_11, © Springer-Verlag Berlin<br />

Heidelberg 2011). Since two decades, among the many c<strong>and</strong>idate-new<br />

drugs for HL, very few reached clinical practice,<br />

<strong>and</strong> none to first-line advanced HL. However, m-Tor inhibitors,<br />

histone deacetylases (HDAC) inhibitors, immunomodulators<br />

(lenalidomide), <strong>and</strong> anti-CD30 antibody drug conjugate<br />

brentuximab vedotin (SGN-35) demonstrate a significant<br />

activity in phase II studies.<br />

64<br />

Table 2. References of main large phase 2 <strong>and</strong> r<strong>and</strong>omized trials reported<br />

in localized HL<br />

4-drug regimens : ABVD (CMT)<br />

Favourable patients<br />

ABVD x2 + IF RT HD-7 Engert JCO 2007. (Engert et al. 3495-502)<br />

Unfavourable patients<br />

ABVD x4 + IF RT Milan Bonadonna JCO 2004.<br />

(Bonadonna et al. 2835-41)<br />

ABVD x6 + M RT H6 Carde JCO 1993. (Carde et al. 2258-72)<br />

ABVD x6 + IF RT H9U Fermé Blood ASH 2005. (Fermé et al. 813)<br />

Noordijk JCO 2005. (Noordijk et al. 6505)<br />

ABVD x4 + IF RT H9U Fermé Blood ASH 2005.(Fermé et al. 813)<br />

Noordijk JCO 2005. (Noordijk et al. 6505)<br />

ABVD x4 + IF RT H9U Fermé Blood ASH 2005.(Fermé et al. 813)<br />

Noordijk JCO 2005. (Noordijk et al. 6505)<br />

ABVD x 4 + IF RT HD-10 Engert NEJM 2010. (Engert et al.)<br />

ABVD x 2 + IF RT HD-10 Engert NEJM 2010. (Engert et al.)<br />

ABVD x4 + IF RT 20 or 30 Gy HD-11 Eich 2010 submitted. (Eich et al.)<br />

ABVD x4 + IF RT 30 Gy HD-14 Borchmann Blood ASH 2008<br />

ABVD x4 + IN RT & FDG-PET adapted) H10 EORTC André Blood<br />

ASH 2009<br />

response adapted strategies – based on FDG-PET imaging<br />

Favourable patients<br />

ABVD x2 & PET2 <strong>and</strong> adaptation H10F cf André ASH 2009.<br />

(Andre et al. 97)<br />

ABVD x2 & PET2 <strong>and</strong> adaptation HD-16 GHSG ongoing 2010<br />

Unfavourable patients<br />

ABVD x2 & PET2 <strong>and</strong> adaptation H10U André ASH 2009 (Andre et al. 97)<br />

escalated BEACOPP x2 + ABVD x 2 +IF/IN RT HD-17 GHSG project 2010<br />

SGN-35 demonstrates an activity/toxicity profile that<br />

may allow rapid upgrading to phase III trials, even in early<br />

stage disease (Younes A, Bartlett NL, Leonard JP et al.<br />

Brentuximab Vedotin (SGN-35) for Relapsed CD30-Positive<br />

Lymphomas. N Engl J Med 2010; 363:1812–1821November<br />

4, 2010). The Italian, French <strong>and</strong> EORTC groups<br />

may seize a unique opportunity launch a phase III trial<br />

(H12) to assess the benefit of this new drugs in a large number<br />

of early stage unfavorable patients. The purpose is to<br />

comp<strong>are</strong> SGN-35 + AVD (=EXP) with ABVD alone (=STD),<br />

both followed by 30 Gy RT. Population is early stage unfavorable<br />

Hodgkin disease. There would be a phase 2 trial<br />

followed by a phase 3 trial.<br />

TREATMENT STRATEGY FOR HL TENDS TO BE<br />

DIRECTED TO ITHE INDIVIDUAL PATIENT<br />

Unlike »targeted« treatments for certain types of solid<br />

tumors such as colon (KRAS) or lung (EGFR) carcinomas,<br />

or hematological conditions (chronic myeloid leukemia),<br />

where treatment is based on individual patient’s tumor characteristics,<br />

HL strategy remains individually blind. However<br />

biological approaches develop <strong>and</strong> may comfort the huge<br />

knowledge that a 40 year experience could accumulate in<br />

terms of conventional treatment <strong>and</strong> late effects, a unique<br />

situation in onco-hematology.<br />

OP68 21st Century Radiation treatments<br />

in Hodgkin lymphoma<br />

Theodore Girinsky on behalf of the EORTC Lymphoma<br />

Group, Institut Gustave Roussy, Villejuif, France<br />

Background. The occurrence of late complications due<br />

to old concepts <strong>and</strong> techniques forced the radiation oncolo-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

gy community to evolve. Nowadays modern radiotherapy<br />

allows radiation to be delivered almost »surgically.«<br />

Radiation therapy is now always used in combination<br />

with chemotherapy. This combination allows the implementation<br />

of »mini radiotherapy« in which radiation doses <strong>and</strong><br />

fields <strong>are</strong> reduced to a minimum <strong>and</strong> precisely implemented<br />

using sophisticated radiation delivery techniques.<br />

Radiation doses can now be reduced to 20–30 Gy, as<br />

demonstrated by the German Hodgkin Study Group. Radiation<br />

fields have become smaller <strong>and</strong> more precise as a result<br />

of the involved node radiotherapy concept developed a decade<br />

ago by the EORTC group. Tremendous progress in<br />

imaging technology such as CT/PET allow an increasingly<br />

accurate identification of initially involved lymph nodes. In<br />

our experience with 141 patients, FDG-PET depicted undetected<br />

LNs in 70% of the patients. Sophisticated radiation<br />

delivery such as IMRT or the deep inspiration breath hold<br />

technique allow precise implementation of these »surgical<br />

fields« with good results. In our study of 50 patients with<br />

localized Hodgkin lymphoma, 5-year progression-free survival<br />

<strong>and</strong> overall survival were 92% <strong>and</strong> 94% respectively.<br />

Conclusions. Sophisticated <strong>and</strong> precise radiation treatments<br />

combined with chemotherapy should allow excellent<br />

cure <strong>and</strong> quality of life in patients with Hodgkin lymphoma.<br />

OP69 Improving Outcome in High Risk Hodgkin’s<br />

lymphoma<br />

Andreas Engert, University Hospital of Cologne, Köln,<br />

Germany<br />

Hodgkin lymphoma has become one of the best-curable<br />

malignancies with tumor-free survival rates in excess of<br />

80%. However, the prognosis for high-risk advanced stage<br />

patients or those with relapsed or refractory disease still<br />

needs improving. To this end, the German Hodgkin Study<br />

Group (GHSG) has developed a more dose intense regimen,<br />

BEACOPP, which demonstrated superiority of our old st<strong>and</strong>ard<br />

of c<strong>are</strong> in a prospectively r<strong>and</strong>omized trial (HD9). The<br />

ten-years update of this study indicates significant differences<br />

in terms of tumor control (18%) <strong>and</strong> overall survival<br />

(11%) between the old st<strong>and</strong>ard (8xCOPP/ABVD) <strong>and</strong> 8<br />

cycles of BEAOPP escalated (Engert et al, JCO, 2009). The<br />

follow-up study, HD12 suggested that radiotherapy (RT)<br />

might not be needed in all patients. This was proven by our<br />

HD15 study in which only PET-positive patients having residual<br />

disease of at least 2,5 cm after chemotherapy received<br />

additional RT.<br />

In order to further reduce toxicity of treatment <strong>and</strong> increase<br />

efficacy, numerous new drugs <strong>are</strong> currently being<br />

developed for HL. The most promising is SGN-35 (Brentuximab<br />

Vedotin) which has shown 75% responses in the<br />

pivotal phase II study. This <strong>and</strong> other new approaches will<br />

be discussed.<br />

OP70 What is the role of PET in Hodgkin’s<br />

lymphoma<br />

Martin Hutchings, Departments of Hematology <strong>and</strong><br />

Oncology, Rigshospitalet, Copenhagen University<br />

Hospital, Denmark<br />

Combined computed tomography <strong>and</strong> positron emission<br />

tomography with 18F-fluorodeoxyglucose (FDG-PET/CT)<br />

has become a cornerstone imaging method in the management<br />

of malignant lymphoma, particularly in the management<br />

of Hodgkin lymphoma. FDG-PET/CT provides an<br />

improved staging accuracy, with higher sensitivity than conventional<br />

imaging methods including CT. The FDG uptake<br />

in Hodgkin lymphoma masses decreases rapidly after initiation<br />

of therapy in well responding patients, <strong>and</strong> this makes<br />

early interim FDG-PET/CT the strongest available predictor<br />

of treatment response <strong>and</strong> prognosis. A negative posttreatment<br />

FDG-PET/CT is highly predictive of long-term<br />

disease-free survival, <strong>and</strong> this has led to the incorporation of<br />

FDG-PET/CT into the revised response criteria for malignant<br />

lymphoma. This presentation will critically address the<br />

value of FDG-PET in Hodgkin lymphoma, including shortcomings<br />

of the available scientific evidence <strong>and</strong> common<br />

problems <strong>and</strong> pitfalls when FDG-PET/CT is used in the<br />

daily clinic <strong>and</strong> in clinical trials. The potential value of<br />

FDG-PET/CT in the follow-up setting, <strong>and</strong> in the management<br />

of relapsed Hodgkin lymphoma will be discussed.<br />

OP71 Treatment of Relapsed/Refractory<br />

Hodgkin’s lymphoma<br />

Igor Aurer, Division of Hematology, Department of<br />

Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

Hodgkin’s lymphoma (HL) is one of the most curable<br />

forms of cancer. Depending on stage <strong>and</strong> prognostic factors<br />

between 60% <strong>and</strong> 95% of patients <strong>are</strong> cured with st<strong>and</strong>ard<br />

front-line therapy. Therefore, about 25% of patients will, at<br />

some point in the course of their disease, need salvage treatment.<br />

Best results in patients below 60–65 (70) years of age in<br />

1 st relapse without significant comorbidities <strong>are</strong> achieved<br />

with intensive salvage regimens followed by autologous<br />

stem cell transplantation (ASCT) with or without consolidative<br />

radiotherapy. Long-term outcome of this approach is<br />

quite favorable, with disease-free survival rates of about<br />

60%. Although a substantial proportion of patients respond<br />

to alternative front-line chemotherapy regimens, such as<br />

COPP, the advantage of ASCT to chemotherapy alone has<br />

been proven in r<strong>and</strong>omized trials. DHAP is the most frequently<br />

used <strong>and</strong> studied second-line chemotherapy regimen.<br />

In a recently published r<strong>and</strong>omized trial, 2 cycles of<br />

DHAP followed by BEAM conditioning <strong>and</strong> ASCT were<br />

less toxic <strong>and</strong> at least as effective as 2 cycles of DHAP,<br />

high-dose single agent chemotherapy, BEAM <strong>and</strong> ASCT.<br />

Based on these data, 2 cycles of chemotherapy prior to<br />

ASCT <strong>are</strong> considered st<strong>and</strong>ard. There <strong>are</strong> no r<strong>and</strong>omized<br />

trials comparing DHAP with other regimens such as ICE,<br />

65


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

HDIM or IGEV, <strong>and</strong> the results of published phase II trials<br />

<strong>are</strong> similar. Therefore, no recommendations regarding the<br />

choice of intensive salvage regimens can be made at present.<br />

The outlook of refractory patients (i.e. those that progress<br />

during or relapse within 3 months after the end of frontline<br />

treatment) is substantially worse. Only about 30% of<br />

them <strong>are</strong> cured with this approach. For both, refractory or<br />

relapsing patients, response to second line treatment is the<br />

most important prognostic factor. ASCT is ineffective in<br />

progressing patients <strong>and</strong> only about 20% of patients with<br />

stable disease benefit from the procedure.<br />

There <strong>are</strong> no r<strong>and</strong>omized trials on the role of radiotherapy<br />

(RT) in this setting but most centers use it either before or<br />

after ASCT. The radiation fields vary, some irradiate only<br />

<strong>are</strong>as that were not in CR (by PET or CT) after ASCT, some<br />

those that were not in remission after salvage chemotherapy<br />

<strong>and</strong> some all <strong>are</strong>as involved at relapse.<br />

Patients who <strong>are</strong> not c<strong>and</strong>idates for ASCT with a localized<br />

relapse in a previously unirradiated <strong>are</strong>a can be effectively<br />

treated with salvage radiotherapy.<br />

About 40–50% of patients fail second-line treatment. For<br />

them the st<strong>and</strong>ard approach used to be chemotherapy, either<br />

with different drug combinations or single-agents, such as<br />

vinblastine or gemcitabine, <strong>and</strong> the judicious use of radiotherapy.<br />

Average survival of such patients is more than 1<br />

year, bearing witness to the chemo- <strong>and</strong> radiosensitivity of<br />

HL. This approach is probably going to change very soon,<br />

because very promising results have been reported with<br />

brentuximab-vedotin (also known as SGN-35) an anti-CD30<br />

monoclonal antibody linked to a cytotoxic agent with antitubullary<br />

activity. Other new agents that <strong>are</strong> being tested in<br />

this setting include histone-deacetylase inhibitors, lenalidomide<br />

<strong>and</strong> m-TOR inhibitors. Finally, the role of allogeneic<br />

stem-cell transplantation remains unclear. While long-term<br />

disease free survival can be achieved with reduced intensity<br />

conditioning in about 40% of patients responding to salvage<br />

chemotherapy, treatment-related mortality <strong>and</strong> morbidity<br />

remains a significant problem <strong>and</strong> comparative trials still<br />

have to show which population of patients will benefit from<br />

this approach.<br />

OP72 Allogeneic SCT for Hodkin’s lymphoma<br />

Ana Sureda, Haematology Department, Addenbrooke’s<br />

Hospital, Cambridge, UK<br />

Myeloablative conditioning <strong>and</strong> allo-SCT<br />

in Hodgkin’s lymphoma<br />

The first reports on allogeneic stem cell transplantation<br />

(allo-SCT) in patients with HL appe<strong>are</strong>d in the mid eighties<br />

[1, 2]. Two larger registry-based studies published in 1996<br />

gave disappointing results. Gajewski et al analyzed 100 HL<br />

patients allografted from HLA-identical siblings <strong>and</strong> reported<br />

to the International Bone Marrow Transplant Registry<br />

(IBMTR) [3]. The 3-year-rates for OS, DFS, <strong>and</strong> the probability<br />

of relapse were 21%, 15%, <strong>and</strong> 65%, respectively.<br />

The major problems after transplantation were persistent or<br />

recurrent disease or respiratory complications, which accounted<br />

for 35% to 51% of deaths. A case-matched analysis<br />

66<br />

including 45 allografts <strong>and</strong> 45 autografts reported to the<br />

EBMT was performed by Milpied et al [4]. They did not<br />

find significant differences in actuarial probabilities of OS,<br />

PFS, <strong>and</strong> relapse rates between allo-SCT <strong>and</strong> ASCT (25%,<br />

15%, 61% vs. 37%, 24%, 61%, respectively). The actuarial<br />

TRM at 4 years was significantly higher for allografts than<br />

for autografts (48% vs 27%, p=0.04). Acute GvHD ≥ grade<br />

II was associated with a significantly lower risk of relapse,<br />

but also with a lower survival rate.<br />

A number of reports confirmed the registry data: allo-<br />

SCT resulted in lower relapse rates but significantly higher<br />

toxicity with no improvement over ASCT when PFS or OS<br />

were considered [5–7]. Although the poor results after myeloablative<br />

conditioning could at least partly be explained<br />

by the very poor-risk features of many individuals included<br />

in these early studies, the high procedure-related morbidity<br />

<strong>and</strong> mortality prevented the widespread use of allo-SCT.<br />

Reduced intensity conditioning <strong>and</strong> allo-SCT<br />

in Hodgkin’s lymphoma<br />

The largest cohort of patients treated with RIC/allo-SCT<br />

in HL was recently reported by the LWP of the EBMT [8]<br />

(n=285). Median time from diagnosis to allo-SCT was 41 (4<br />

– 332) months. Patients had received an average of four<br />

lines of prior therapy (1 – 8) <strong>and</strong> 288 patients (77%) had<br />

failed one or two ASCT. At the time of allo-SCT, 47 patients<br />

(17%) were in CR, 123 patients (43%) had chemosensitive<br />

disease <strong>and</strong> 115 patients (40%) had chemoresistant disease<br />

or untested relapse. One hundred <strong>and</strong> seventy two patients<br />

(63%) were allografted from a matched sibling donor<br />

(MRD), 94 (33%) from a matched unrelated donor (MUD),<br />

<strong>and</strong> 19 from a mismatched donor (7%). Grade II-IV acute<br />

GvHD (aGVHD) was reported in 27% of patients, chronic<br />

GvHD (cGVHD) in 40% of patients at risk. The 100-day<br />

TRM was 12% but increased to 20% at 12 months, <strong>and</strong> to<br />

22% at three years. The development of chronic GVHD was<br />

associated with a higher TRM <strong>and</strong> a trend to a lower relapse<br />

rate. In a l<strong>and</strong>mark analysis the development of either acute<br />

or chronic GVHD by 9 months post transplant was associated<br />

with a significantly lower relapse rate.<br />

The MD Anderson Cancer Center updated their experience<br />

[9] in 58 patients with relapsed or refractory HL who<br />

underwent a RIC/allo-SCT from a MRD (n=25) or a MUD<br />

(n=33). Forty-eight (83%) patients had received a prior<br />

ASCT. Disease status at RIC/allo-SCT was sensitive relapse<br />

(n=30) or refractory relapse (n=28). The conditioning regimen<br />

employed was fludarabine (125–130 mg/m2 over 4–5<br />

days), melphalan (140 mg/m2 IV over 2 days) (FM) <strong>and</strong> antithymocyte<br />

globulin (thymoglobulin 6 mg/kg over 3 days)<br />

was added for the 14 most recent MUD transplants. Cumulative<br />

100-day <strong>and</strong> 2-year TRM were 7% <strong>and</strong> 15%, respectively.<br />

The cumulative incidence (CI) of grade II-IV acute<br />

GVHD was 28%. The CI of chronic GVHD at any time was<br />

74%. Fourteen patients (24%) received a total of 25 (range<br />

1–5) donor leukocyte infusions (DLIs) for disease progression/relapse.<br />

Five of them (35%) received CT as well, <strong>and</strong><br />

nine (64%) developed acute GVHD after the DLI. Projected<br />

2-year OS <strong>and</strong> PFS were 64% <strong>and</strong> 32%, with 2-year projected<br />

disease progression at 55%. There was no statistically


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

significant difference between MRD <strong>and</strong> MUD transplants<br />

with regard to OS, PFS <strong>and</strong> disease progression. There was<br />

a trend for the response status prior to allo-SCT to favorably<br />

impact PFS (p=0.07) <strong>and</strong> disease progression (p=0.049),<br />

but not OS (p=0.4). Partial responders <strong>and</strong> patients with<br />

stable/ refractory disease f<strong>are</strong>d similarly with regards to OS<br />

<strong>and</strong> PFS.<br />

Forty patients with relapsed or refractory HL treated with<br />

the combination of fludarabine (150 mg/m 2 ) <strong>and</strong> melphalan<br />

(140 mg/m 2 ) have recently been presented by the Spanish<br />

group [10]. GvHD prophylaxis consisted of cyclosporine A<br />

(CsA) <strong>and</strong> methotrexate (MTX). Twenty-one patients (53%)<br />

had received >2 lines of CT, 23 patients (58%) had been irradiated,<br />

<strong>and</strong> 29 patients (73%) had failed a previous ASCT.<br />

Twenty patients were allografted in resistant relapse <strong>and</strong> 38<br />

patients received hematopoietic cells from a MRD. Oneyear<br />

TRM was 25%. Acute GvHD developed in 18 patients<br />

(45%) <strong>and</strong> cGvHD in 17 (45%) of the 31 evaluable patients.<br />

Extensive cGvHD was associated with a trend to a lower<br />

relapse rate (71% vs 44% at 24 months, p=0.07). The response<br />

rate three months after RIC/allo-SCT was 67%.<br />

Eleven patients received donor lymphocyte infusions (DLIs)<br />

for relapse or persistent disease. Six patients (54%) responded.<br />

OS <strong>and</strong> PFS were 48% <strong>and</strong> 32% at 2 years, respectively.<br />

Refractoriness to CT was the only adverse prognostic<br />

factor for both OS <strong>and</strong> PFS.<br />

Investigators from Seattle reported their results in relapsed<br />

/ refractory HL patients [11]. Thirty-eight patients<br />

had a MRD, 24 a MUD <strong>and</strong> 28 a HLA-haploidentical related<br />

donor. The patients received 2 Gy total body irradiation<br />

(TBI) alone (n=17) or in combination with fludarabine<br />

(90 mg/m 2 ) <strong>and</strong> immunosuppression consisted of micophenolate<br />

mofetil (MMF) <strong>and</strong> CsA. All patients were heavily<br />

pre-treated with a median of five prior regimens administered.<br />

92% of the patients had failed a previous ASCT. Prior<br />

to RIC/allo-SCT 22 patients were in CR, 30 in PR, 9 had<br />

relapsed disease, <strong>and</strong> 29 had refractory disease. The overall<br />

incidence of grade II-IV aGvHD was 50%. The incidence of<br />

extensive cGvHD was 55% at 1 year. TRM was significantly<br />

lower in those patients being allografted from a HLAhaploidentical<br />

donor. Relapse risk was also lower in haploidentical<br />

recipients.<br />

Peggs et al explored the effects of in vivo T-cell depletion<br />

with alemtuzumab followed by fludarabine (150 mg/m 2 )<br />

<strong>and</strong> melphalan (140 mg/m 2 ) in multiply relapsed patients;<br />

90% of them had failed a previous autograft [12]. At transplant,<br />

8 patients were in CR, 25 patients were in PR, one<br />

patient was in untested relapse, <strong>and</strong> 15 patients had refractory<br />

disease. Thirty-one patients were allografted from a<br />

MRD <strong>and</strong> 18 from MUDs. All patients engrafted, grade II-<br />

IV aGvHD occurred in 16% of patients, 14% developed<br />

cGvHD before DLIs. Nineteen patients received DLIs for<br />

progression (n=16) or mixed chimerism (n=3). Nine patients<br />

(56%) showed a response, which was significantly associated<br />

with, acute <strong>and</strong>/or extensive chronic GvHD. Nonrelapse<br />

mortality was 16% at 730 days. Projected 4-year OS<br />

<strong>and</strong> PFS were 56% <strong>and</strong> 39%, respectively.<br />

Finally, the final results of a multicenter phase II prospective<br />

study on the role of RIC/Allo-SCT were presented at<br />

ASH2010 [13]. 92 HL patients with an HLA identical sib-<br />

ling or a MUD were treated with 2 courses of salvage chemotherapy.<br />

Seventy-eight patients (85%) who showed at<br />

least stable disease were eligible to receive a RIC/Allo-SCT;<br />

all 14 patients with chemorefractory disease died from progressive<br />

lymphoma. Most allografted patients had failed a<br />

prior autologous transplantation (86%); 50 patients were allografted<br />

with chemosensitive <strong>and</strong> 28 with resistant disease;<br />

MUDs were used in 23 patients. Fludarabine (150 mg/m 2<br />

iv) <strong>and</strong> melphalan (140 mg/m 2 iv) were used as conditioning<br />

regimen <strong>and</strong> cyclosporine A plus methotrexate as graft-versus-host<br />

disease prophylaxis. Non-relapse mortality was<br />

8% at 100-days <strong>and</strong> 15% at 1-year. Relapse was the major<br />

cause of failure. PFS was 48% at 1-year <strong>and</strong> 24% at 4-years.<br />

Chronic GVHD was associated with a lower relapse incidence<br />

<strong>and</strong> a better PFS (figure 3). Patients allografted in CR<br />

had a significantly better outcome. OS was 71% at 1-year<br />

<strong>and</strong> 43% at 4-years.<br />

No definitive information is available with respect to the<br />

best conditioning protocol or the impact of T-cell depletion<br />

in this setting. If one accepts that attempting an effective<br />

graft-versus-HL reaction may require several months, preventing<br />

early progression by administering a vigorous conditioning<br />

regimen remains an essential goal still to accomplish.<br />

In this sense, the combination of a more intensive<br />

preparative regimen, the BEAM protocol together with a<br />

profound T-cell depletion with alemtuzumab as aGVHD<br />

prophylaxis has demonstrated to be associated with sustained<br />

donor engraftment, a high response rate, minimal<br />

toxicity (NRM 7.6%) <strong>and</strong> a low incidence of GVHD [14].<br />

The two analyses presented by the Lymphoma WP of the<br />

EBMT also strengthen this argument.<br />

Comparison of myeloablative <strong>and</strong> reduced-intensity<br />

conditioning prior to allo-SCT in relapsed <strong>and</strong><br />

refractory Hodgkin’s lymphoma<br />

The LWP has performed the only analysis reported so far<br />

which comp<strong>are</strong>s outcomes after reduced-intensity or myeloablative<br />

conditioning in patients with HL [15]. Ninetyseven<br />

patients with HL were allografted after RIC <strong>and</strong> 93<br />

patients were allografted after a conventional regimen. A<br />

previous ASCT was more frequent in the RIC/allo-SCT<br />

group (59% vs 41%, p=0.03) as was the use of peripheral<br />

blood stem cells (82% vs 56% p


Oral presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

HL effect comes from two main sources: the demonstration<br />

that the development of acute or chronic GVHD after allo-<br />

SCT is associated to a lower relapse rate <strong>and</strong> the clinical<br />

information coming from DLIs. Relapse rate is significantly<br />

lower in those patients developing GVHD after transplantation.<br />

This fact was already indicated by single center analysis<br />

including low number of patients [9,140, 12, 13] but also<br />

by larger retrospective analyses such as those performed by<br />

the Lymphoma WP of the EBMT. The development of either<br />

acute or chronic GVHD by 9 months after transplant<br />

was associated with a lower relapse rate in Robinson’s analysis<br />

[8]. The development of acute GVHD alone did not<br />

reduce the relapse rate but was associated with a significantly<br />

higher TRM <strong>and</strong> a lower PFS <strong>and</strong> OS. In contrast,<br />

the development of chronic GVHD alone was associated<br />

with a trend to lower relapse rate, a significantly higher<br />

TRM but had no impact on OS or PFS. In the comparative<br />

analysis between conventional allo-SCT <strong>and</strong> RIC/allo-SCT<br />

the development of chronic GVHD after transplantation<br />

significantly reduced relapse rate after transplant <strong>and</strong> improved<br />

PFS in those patients presenting with this complication<br />

[15].<br />

The most direct evidence for a graft versus malignancy<br />

effect comes from observations relating to disease responses<br />

to DLIs. Peggs et al have previously reported clinical responses<br />

in 9 of 16 patients receiving DLIs for measurable<br />

disease post RIC/allo-SCT [12]. Other small series have reported<br />

response rates of 44–54% following DLI administration<br />

[9, 10]. In Robinson’s analysis [8] many DLIs were<br />

administered following adjunctive therapy but disease responses<br />

were reported in 30% of patients receiving DLI<br />

alone. Patients responding to DLIs had a higher incidence<br />

of GVHD post DLI than those not responding. These data<br />

confirm the presence of a clinically effective graft versus<br />

Hodgkin’s effect. However, the efficacy of DLIs is likely to<br />

depend upon the bulk of disease at the time of administration<br />

<strong>and</strong> the optimal use of DLI requires further refinement.<br />

Preemptive, dose escalating or PET scan guided strategies<br />

may improve the overall efficacy of DLIs [16, 17].<br />

How to improve the results of allo-SCT?<br />

Relapse rate is the major cause of failure for those patients<br />

with relapsed / refractory HL being considered c<strong>and</strong>idates<br />

for such an approach. There <strong>are</strong> several possible ways<br />

to address this issue: better patient selection <strong>and</strong> chemosensitivity<br />

of the tumour. Disease status at the time of allo-SCT<br />

is the most important prognostic factor for the long-term<br />

outcome of this procedure. Only those patients in CR or<br />

very good PR should be considered adequate c<strong>and</strong>idates for<br />

an allo-SCT at least with the current protocols. In this sense,<br />

new salvage strategies to try to put patients into a better response<br />

should be sought. Pre-transplant PET does not seem<br />

to have a prognostic impact on either OS or PFS after allo-<br />

SCT but PET was able to diagnose relapse after allo-SCT<br />

earlier than conventional computed tomography [16, 18]. In<br />

this sense, the role of PET should be further explored in this<br />

setting.<br />

Modulation of the intensity of the conditioning regimen<br />

can also result in lower relapse rate after allo-SCT. Low-<br />

68<br />

dose TBI containing regimens seem to be associated to a<br />

high relapse rate in the RIC-allo setting [8, 15]. New drugs<br />

with potential antitumoral activity in front of HL but with a<br />

safe profile <strong>are</strong> being currently tested by different groups.<br />

The intensity of conditioning regimen can be eventually<br />

augmented without significantly modifying Non-Relapse<br />

Mortality (NRM) taking into consideration the population<br />

of patients that <strong>are</strong> considered c<strong>and</strong>idates for an allo-SCT<br />

<strong>and</strong> the fact that GVL effect is not as potent as in low-grade<br />

lymphoproliferative disorders.<br />

Finally, the »so called« maintenance strategy currently<br />

being explored in the ASCT setting can also be analyzed in<br />

the allogeneic one.<br />

R E F E R E N C E S<br />

1. Appelbaum FR, Sullivan KM, Thomas ED, et al. Allogeneic marrow<br />

transplantation in the treatment of MOPP-resistant Hodgkin’s disease.<br />

J Clin Oncol 1985;3:1490–1494.<br />

2. Phillips GL, Reece DE, Barnett MJ, et al. Allogeneic marrow transplantation<br />

for re fractory Hodgkin’ disease. J Clin Oncol 1989;7:1039–<br />

1045.<br />

3. Gajewski JL, Phillips GL, Sobocinski KA, et al. Bone marrow transplants<br />

from HLA-identical siblings in advanced Hodgkin’s disease. J<br />

Clin Oncol 1996;14:572–578.<br />

4. Milpied N, Fielding AK, Pearce RM, Ernst P, Goldstone AH. Allogeneic<br />

bone marrow transplant is not better than autologous transplant for<br />

patients with relapsed Hodgkin’s disease. J Clin Oncol. 1996;14:1291–<br />

1296.<br />

5. Anderson JE, Litzow MR, Appelbaum FR, et al. Allogeneic, syngeneic,<br />

<strong>and</strong> autolo gous marrow transplantation for Hodgkin’s disease: The 21year<br />

Seattle Experience. J Clin Oncol 1993;11:2342–2350.<br />

6. Jones RJ, Ambinder RF, Piantadosi S, Santos GW. Evidence of a graftversus-lymphoma<br />

effect associated with allogeneic bone marrow transplantation.<br />

Blood 1991;77:649–653.<br />

7. Akpek G, Ambinder RF, Piantadosi S, Abrams RA, Brodsky RA, Vogelsang<br />

GB, et al. Long-term results of blood <strong>and</strong> marrow transplantation<br />

for Hodgkin’s disease. J Clin Oncol 2001;19:4314–4321.<br />

8. Robinson S, Sureda A, Canals C, et al. Reduced intensity allogeneic<br />

stem cell transplantation for Hodgkin’s lymphoma: Identification of<br />

prognostic factors predicting outcome. Haematol 2008;94:230–238.<br />

9. Anderlini P, Saliba R, Acholonu S, Okoroji GJ, Donato M, Giralt S, et<br />

al. Reduced-intensity allogeneic stem cell transplantation in relapsed<br />

<strong>and</strong> refractory Hodgkin’s disease: low transplant-related mortality <strong>and</strong><br />

impact of intensity of conditioning regimen. Bone Marrow Transplant<br />

2005;35:943–951.<br />

10. Alv<strong>are</strong>z I, Sureda A, Caballero MD, et al. Non-myeloablative stem cell<br />

transplantation is an effective therapy for refractory or relapsed Hodgkin’s<br />

lymphoma: Results of a Spanish prospective cooperative protocol.<br />

Biol Blood Marrow Transplant 2006;12:172–183.<br />

11. Burroughs LM, O’Donnell PV, S<strong>and</strong>maier BM, et al. Comparison of<br />

outcomes of HLA-matched related, unrelated, or HLA-haploidentical<br />

related hematopoietic cell transplantation following nonmyeloablative<br />

conditioning for relapsed or refractory Hodgkin lymphoma. Biol Blood<br />

Marrow Transplantation 2008;14:1279–1287.<br />

12. Peggs KS, Hunter A, Chopra R, et al. Clinical evidence of a graft-versus-Hodgkin’s-lymphoma<br />

effect after reduced-intensity allogeneic<br />

transplantation. Lancet 2005;365:1934–1941.<br />

13. Sureda A, Canals C, Arranz R, et al. Allogeneic stem cell transplantation<br />

after reduced-intensity conditioning in patients with relapsed or<br />

refractory Hodgkin’s lymphoma: results of the HDR-Allo Study, a prospective<br />

clinical trial by the Grupo Español de Linfomas/Trasplante de<br />

Médula Ósea (GEL/TAMO) <strong>and</strong> the Lymphoma Working Party of the<br />

European Group for Blood <strong>and</strong> Marrow Transplantation. Leukemia<br />

(submitted).<br />

14. Faulkner RD, Craddock C, Byrne JL, et al. BEAM-alemtuzumab reduced-intensity<br />

allogeneic stem cell transplantation for lymphoproliferative<br />

diseases: GVHD, toxicity, <strong>and</strong> survival in 65 patients. Blood<br />

2004;103:428–434.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Oral presentations<br />

15. Sureda A, Robinson S, Çanals C, et al Reduced-intensity conditioning<br />

comp<strong>are</strong>d with conventional allogeneic stem-cell transplantation in relapsed<br />

or refractory Hodgkin’s lymphoma: an analysis from the Lymphoma<br />

Working Party of the European Group for Blood <strong>and</strong> Marrow<br />

Transplantation. J Clin Oncol 2008;26:455–462.<br />

16. Peggs KS, Thomson KJ, Hart DP et al. Dose-escalated donor lymphocyte<br />

infusions following reduced intensity transplantation: toxicity,<br />

chimerism, <strong>and</strong> disease responses. Blood 2004;103:1548–1556.<br />

17. Hart DP, Avivi I, Thomson KJ, et al. Use of 18 F-FDG positron emission<br />

tomography following allogeneic transplantation to guide adoptive immunotherapy<br />

with donor lymphocyte infusions. Br J Haematol 2005;<br />

128:824–829.<br />

18. Lambert JR, Bomanju JB, Pegas KS, et al. Prognostic role of PET scanning<br />

before <strong>and</strong> alter reduced-intensity allogeneic stem cell transplantation<br />

for lymphoma. Blood 2010;115:2763–2768.<br />

OP73 New Drugs in the treatment of Hodkin’s<br />

lymphoma<br />

Anas Younes, (USA)<br />

Abstract not submitted.<br />

69


Meet the Professor<br />

MP01 Chronic myeloid leukemia<br />

Martin Mistrik, Dept. of Hematology <strong>and</strong> Transfusiology,<br />

University Hospital Bratislava, Slovakia<br />

Introduction<br />

Chronic myeloid leukemia (CML) was first described in<br />

the nineteenth century, but treatment with arsenicals <strong>and</strong> radiotherapy<br />

was unsatisfactory. In the 1950s busulfan <strong>and</strong><br />

hydroxycarbamid were introduced, <strong>and</strong> the disease control<br />

has improved. Significant progress brought discovery of the<br />

Philadelphia (Ph) chromosome (4), that results from a t(9;22)<br />

translocation <strong>and</strong> that leukemia originates from a single hematopoietic<br />

»stem cell« with BCR-ABL fusion gene (2; 3).<br />

In the 1980s interferon-alpha replaced chemotherapeuticals<br />

<strong>and</strong> induced in some patients Ph-chromosome negativity.<br />

Between 1980 <strong>and</strong> 2000, allogeneic stem cell transplantation<br />

(allo-SCT) was initial treatment for younger patients<br />

with HLA-matched donors. In the last decade, the introduction<br />

of imatinib (imatinib mesylate, IM), the Abl tyrosine<br />

kinase inhibitor (TKI), has dramatically changed CML therapeutic<br />

strategy (1). The annual number of SCT for CML<br />

has fallen markedly <strong>and</strong> according to the latest ELN recommendations<br />

from 2009 most hematologists today recommend<br />

initial treatment with IM. IM has transformed CML<br />

from an invariably fatal disease in its natural evolution to a<br />

chronic illness, like hypertension, diabetes, or AIDS if patients<br />

comply with daily oral therapy. Although CML is a<br />

relatively r<strong>are</strong> malignancy, effective treatment has significantly<br />

enlarged population of patients, increased its prevalence<br />

<strong>and</strong> raised new problems of management.<br />

What is important at diagnosis?<br />

At first spleen size (by palpation cm bellow costal margin<br />

at maximal inspirium) has to be recorded, <strong>and</strong> complete <strong>and</strong><br />

differential blood count, bone marrow aspiration <strong>and</strong> conventional<br />

cytogenetics performed. In case of Ph-negative<br />

karyotype FISH <strong>and</strong> PCR for BCR-ABL is m<strong>and</strong>atory. HLA<br />

typing of siblings should be done in every patient fit for<br />

allo-SCT.<br />

What <strong>are</strong> the next steps at diagnosis?<br />

1. disease phase assessment according to<br />

blasts (chronic phase: < 10% in peripheral blood<br />

<strong>and</strong>/ or nucleated bone marrow cells; accelerated<br />

phase: 10 – 19%; blastic phase: ≥ 20%)<br />

basophils (chronic phase: < 20% in peripheral blood;<br />

accelerated phase: ≥ 20%)<br />

platelet count (accelerated phase: persistent thrombocytopenia<br />

< 100 x 109 /l)<br />

spleen size (accelerated phase: increasing spleen<br />

size)<br />

cytogenetics (accelerated phase: evidence of clonal<br />

evolution)<br />

2. Risk score assessment (5)(www.roc.se/sokal.asp) –<br />

Sokal risk: low < 0,8; intermediate 0,8 – 1,2 <strong>and</strong> high<br />

> 1,2:<br />

Age (years)<br />

Myeloblasts in blood (%)<br />

Platelets (x 10 9 /l)<br />

Spleen size (cm)<br />

What is the first line treatment?<br />

Leukocytosis has to be controlled with hydroxyurea, addition<br />

of allopurinol is important as well. Low risk patients<br />

should start treatment with IM, intermediate <strong>and</strong> high risk<br />

patients could profit from upfront second generation TKI<br />

therapy. The treatment goal is achievement of major molecular<br />

response (it means 3 log bcr/abl reduction, or bcr/abl<br />


Meet the Professor Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

In summary, there is a lot one can get right or wrong at<br />

diagnosis. The majority or all patients who present with de<br />

novo CML should if possible receive initial treatment with<br />

TKI (imatinib, nilotinib or dasatinib). Monitoring blood<br />

counts, cytogenetics <strong>and</strong> quantitative PCR <strong>are</strong> used according<br />

to the level of response. Allo-SCT can be considered<br />

according to the TKI effectivity. For non chronic phase<br />

CML patients TKI ± chemotherapy followed by allo-SCT is<br />

the treatment of first choice.<br />

R E F E R E N C E S<br />

1. Druker BJ, Talpaz M, Resta DJ, et al. Efficacy <strong>and</strong> safety of a specific<br />

inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.<br />

New Engl J Med 2001;344:1031–1037<br />

2. Geary CG. The story of chronic myeloid leukaemia. Br J Haematol<br />

2000;110:2–11<br />

3. Goldman JM, Daley GQ. Chronic myeloid leukemia: a brief history. In:<br />

Melo JV, Goldman JM, editors. Myeloproliferative Disorders. New<br />

York, NY: Springer; 2007. p. 10–13<br />

4. Nowell PC, Hungerford DA. A minute chromosome in human granulocytic<br />

leukemia. Science 1960;132:1497<br />

5. Sokal JE, Baccarani M, Russo D, et al. Staging <strong>and</strong> prognosis in chronic<br />

myelogenous leukemia. Semin Hematol 1988;25:49–61<br />

MP02 Essential (primary) Thrombocythemia<br />

Petro E. Petrides, Hematology Oncology Center <strong>and</strong><br />

Ludwig-Maximilians-University Medical School Munich,<br />

Germany<br />

This seminar will cover all relevant aspects of essential<br />

(primary) thrombocythemia (ET). Starting with the precise<br />

histological diagnosis of ET, which is based on the current<br />

WHO-classification <strong>and</strong> the discussion of differential diagnosis<br />

from reactive thrombocythemia, it continues with the<br />

novel molecular markers JAK2 V617F <strong>and</strong> MPL which <strong>are</strong><br />

present in 50 or 5% resp.of all ET patients.<br />

Next, platelet production <strong>and</strong> function under normal <strong>and</strong><br />

pathological (ET) circumstances <strong>are</strong> outlined.<br />

From the limited data on epidemiology (since ET is an<br />

orphan disease), the question of life expectancy is discussed.<br />

Then signs <strong>and</strong> symptoms of ET patients <strong>are</strong> outlined.<br />

which <strong>are</strong> caused by microcirculatory, thrombotic or hemorrhagic<br />

events.<br />

When ET has been diagnosed by histological <strong>and</strong> molecular<br />

markers, a staging procedure should be performed:<br />

this includes a differential blood smear, serum electrolytes<br />

(with the occurrence of pseudohyperkalemia), uric acid, renal<br />

<strong>and</strong> hepatic values as well as cholesterol (HDL <strong>and</strong><br />

LDL) <strong>and</strong> triglycerides.<br />

Clinical investigation should include a physical examination,<br />

an ultrasound of the spleen (in three dimensions with<br />

volume determination), Doppler ultrasound of the carotid<br />

arteries (plaque formation, intima media thickness) <strong>and</strong> an<br />

exercise electrocardiogram.<br />

Goal of therapeutic interventions is primary <strong>and</strong> secondary<br />

prevention of disease associated complications. At the<br />

moment therapeutical recommendations <strong>are</strong> based upon<br />

age, thrombosis <strong>and</strong> platelet counts. A more comprehensive<br />

approach includes life style (physical activities, smoking,<br />

72<br />

diet, biological age), additional thrombophilic risk factors<br />

(through appropriate testing) <strong>and</strong> how well the patient is informed<br />

about the disease <strong>and</strong> its potential complications.<br />

Basic therapy includes »wait <strong>and</strong> see« or platelet antiaggregatory<br />

treatment with aspirin although only retrospective<br />

data confirm the validity of the latter approach. For cytoreduction<br />

three substances <strong>are</strong> nowadays in use: anagrelide,<br />

hydroxyurea <strong>and</strong> pegylated interferon-alpha with different<br />

mechanisms of action. Only three r<strong>and</strong>omized clinical trials<br />

testing hydroxyurea <strong>and</strong>/or anagrelide (Bergamo/Vicenza,<br />

PT1 <strong>and</strong> Anahydret) have been carried out. Long term efficacy<br />

<strong>and</strong> adverse effect data <strong>are</strong> available only for anagrelide<br />

from post marketing registries from different countries. Pros<br />

<strong>and</strong> cons of the three therapeutical regimens will be discussed.<br />

In addition, novel approaches such as telomerase<br />

inhibitors will be outlined.<br />

Treatment of aged people (up to 96 years), children or<br />

during pregnancy pose additional chalenges.<br />

Case examples for the questions addressed will be given<br />

from daily practise.<br />

MP03 Acute myeloid leukemia<br />

Boris Labar, Division of Hematology, Department of<br />

Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

Classification of acute myeloid leukemia<br />

The current WHO classification is using morphology, immunophenotyping,<br />

cytogenetic <strong>and</strong> molecular genetic abnormalities<br />

to delineate entities of AML with a different<br />

prognostic significance (1). The WHO classification of AML<br />

is given in Table 1. A number of recurrent genetic abnormalities<br />

<strong>are</strong> sufficiently defined to be recognized as entities<br />

of AML.<br />

Moreover some of new genetic abnormalities, mutation<br />

of NPM1 gene <strong>and</strong> CEBPA gene <strong>are</strong> also added to classification<br />

(2, 3). These mutations <strong>are</strong> associated with good prognosis.<br />

FLT3 mutations (4) <strong>are</strong> not defined as a separate entity<br />

but there <strong>are</strong> sufficient data that these mutations have<br />

the prognostic impact. The entity of AML with myelodysplasia<br />

related changes is practically based on the morphological<br />

specific changes, i.e. morphological dysplastic changes<br />

in ≥50% of ≥2 cell lineages in bone marrow. In this<br />

group <strong>are</strong> patients with previous myelodysplastic syndrome<br />

(MDS) or myelodysplastic/myeloproliferative neoplasm<br />

(MDS/MPN) who evolve to AML with marrow or blood<br />

blast counts ≥20%, or if they have myelodysplasia related<br />

cytogenetic abnormality (see Table 1.) (5). Therapy related<br />

myeloid neoplasm (6) is high risk AML <strong>and</strong> should be treated<br />

with intensive chemotherapy <strong>and</strong> allogeneic SCT.<br />

A previous FAB classification is recognized in WHO<br />

classification ss the entity acute myeloid leukemia, not otherwise<br />

specified. In this subgroup one can find the morphologic<br />

separation of AML according to the immaturity of<br />

leukemic cells as well as according to the hematopoietic lineage<br />

involved. This subtype of AML is reserved for the patients<br />

without the known cytogenetic or molecular genetic


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Meet the Professor<br />

Table 1. Acute myeloid leukemia <strong>and</strong> related precursor neoplasm, <strong>and</strong><br />

acute leukemia of ambiguous lineage (WHO 2008) (1)<br />

Acute myeloid leukemia with recurrent genetic abnormalities<br />

– AML with t(8;21)(q22;q22); RUNX1-RUNX1T1<br />

– AML with inv(16)(p13.1;q22) or t(16;16)(p13.1;q22); CBFB-MYH11<br />

– APL with t(15;17)(q22;q12); PML-RARA*<br />

– AML with t(9;11)(p22;q23); MLLT3-MLL**<br />

– AML with t(6;9)(p23:q34); DEK-NUP214<br />

– AML with inv(3)(q21;q26.2) or t(3;3)(q21;q25.2); RPN1-EVI1<br />

– AML (megakaryoblastic) with t(1;22)(p13;q13); RBM15-MKL1<br />

– Provisional entity: AML with mutated NPM1<br />

– Provisional entity: AML with mutated CEBPA<br />

Acute myeloid leukemia with myelodysplasia related changes †<br />

Therapy related myeloid neoplasm ‡<br />

Acute myeloid leukemia, not otherwise specified (NOS)<br />

– Acute myeloid leukemia with minimal differentiation<br />

– Acute myeloid leukemia without maturation<br />

– Acute myeloid leukemia with maturation<br />

– Acute myelomonocytic leukemia<br />

– Acute monoblastic/monocytic leukemia<br />

– Acute erythroid leukemia<br />

Pure erythroid leukemia<br />

Erythroleukemia, erythroid/myeloid<br />

– Acute megakaryoblastic leukemia<br />

– Acute basophilic leukemia<br />

– Acute panmyelosis with myelofibrosis (syn.: acute myelofibrosis; acute<br />

myelosclerosis)<br />

Myeloid sarcoma (syn.: extramedullary myeloid tumor; granulocytic<br />

sarcoma, chloroma)<br />

Myeloid proliferation related to Down syndrome<br />

– Transient abnormal myelopoiesis (syn.: transient myeloproliferative<br />

disorder)<br />

– Myeloid leukemia associated with Down syndrome<br />

Blastic plasmacytoid dendritic cell neoplasm<br />

Acute leukemias of ambiguous lineage<br />

– Acute undifferentiated leukemia<br />

– Mixed phenotype acute leukemia with t(9;22)(q34;q11.2); BCR-ABL1 §<br />

– Mixed phenotype acute leukemia with t(v;11q23); MLL rearranged<br />

– Mixed phenotype acute leukemia, B/myeloid, NOS<br />

– Mixed phenotype acute leukemia, T/myeloid, NOS<br />

– Provisional entity: Natural killer (NK) cell lymphoblastic leukemia/<br />

lymphoma<br />

* Other recurring translocations involving RARA should be reported accordingly<br />

e.g. AML with t(1;17)(q23;q12)/ZBTB16-RARA; AML with t(11;17)<br />

(q13;q12); NUMA1-RARA; AML with T(5;17)(q35;q12); NPM1-RARA; or<br />

AML with STAT5B-RARA (the later have normal chromosome 17 on conventional<br />

cytogenetics)<br />

** Other translocations involving MLL should be reported accordingly: e.g.<br />

AML with t(6;11)(q27;q23); MLLT4-MLL; AML with t(11;19)(q23;p13.3);<br />

MLL-MLLT1; AML with t(11;19)(q23;p13.1); MLL-ELL; AML with t(10;11)<br />

(p12;q23); MLLT10-MLL.<br />

† >20% blood or marrow blasts <strong>and</strong> any of the following previous history of<br />

myelodysplastic syndrome (MDS) or myelodysplastic/myeloproliferative<br />

neoplasm (MDS/MPN): myelodysplasia-related cytogenetic abnormality (see<br />

below); multilineage dysplasia; <strong>and</strong> absence of both prior cytotoxic therapies<br />

for unrelated disease <strong>and</strong> aforementioned recurring genetic abnormalities;<br />

cytogenetic abnormalities sufficient to diagnose AML with myelodysplasi<strong>are</strong>lated<br />

changes <strong>are</strong>:<br />

– Complex karyotype (defined as 3 or more chromosomal abnormalities)<br />

– Unbalanced changes: -7 or del(7q); -5 or del(5q); i(17q) or t(17p); -13 or<br />

del(13q); del(11q); del(12p) or t(12p); del(9q); idic(X)(q13)<br />

– Balanced changes: t(11;16)(q23;p13.3); t(3;21)(q26;q22.1);<br />

t(1;3)(p36.3;q21.1); t(2;11)(p21;q23); t(5;12)(q33;p12); t(5;7)(q33;p13);<br />

t(5;10)(q33;q21); t(3;5)(q25;q34).<br />

‡ Cytotoxic agents implicated in therapy related hematologic neoplasm: alkylating<br />

agents; ionizing radiation therapy; topoisomerase II inhibitors; others.<br />

§ BCR-ABL1 positive leukemia may present as mixed phenotype leukemia,<br />

but should be treated as BCR-ABL1 positive acute lymphoblastic leukemia.<br />

abnormalities. In some of them, there <strong>are</strong> markers associated<br />

with prognostic significance.<br />

The r<strong>are</strong> forms of AML <strong>and</strong> acute leukemia of ambiguous<br />

lineage recognized in WHO classification <strong>are</strong> also associated<br />

with poor prognosis. The other WHO AML entities <strong>are</strong><br />

rather r<strong>are</strong> disorders <strong>and</strong> will not be discussed in detail.<br />

Prognosis of AML<br />

Patient’s related prognostic factors<br />

Age, co-morbidities, performance status <strong>and</strong> genetic variation<br />

in drug metabolism <strong>are</strong> the main prognostic factors<br />

related to patients with AML. Increasing age is important<br />

independent adverse prognostic factor (7). Currently all patients<br />

under the age of 60 <strong>are</strong> c<strong>and</strong>idate to receive st<strong>and</strong>ard<br />

intensive chemotherapy <strong>and</strong> according to prognostic factors<br />

stem cell transplantation or intensive chemotherapy as postremission<br />

therapy. It has to be stressed that age as a factor<br />

is not only dependent on so-called »calendar age«. Recently<br />

many older patients in a good clinical status have been successfully<br />

treated with intensive chemotherapy. Even more,<br />

stem cell transplantation with reduced intensity conditioning<br />

could be offered to older patients with reasonable toxicity<br />

<strong>and</strong> treatment related mortality (TRM) (8). So to define<br />

the treatment approach it is important to evaluate <strong>and</strong> to determine<br />

co-morbidities before any therapy. Many different<br />

comorbidity scores have been proposed to outline patients<br />

who <strong>are</strong> the c<strong>and</strong>idate for intensive therapy (9). It is very<br />

popular especially in older group of patients to differentiate<br />

»fit« from »unfit« or »frail« patients.<br />

Co-morbidity scoring is currently still under the investigation<br />

in many Cooperative groups. In younger patients comorbidity<br />

score is »defined« as inclusion/exclusion criteria.<br />

That means patients with e.g. »unacceptable« renal, cardial<br />

or hepatic abnormalities <strong>are</strong> not included into the study. By<br />

such approach at least 20–30% of younger patients <strong>are</strong> excluded<br />

from the treatment in the clinical trials. On the other<br />

h<strong>and</strong>s in older group more than 50% of patients <strong>are</strong> also<br />

excluded from the clinical trials. So, these patients have not<br />

been reported in any results?! Because of that it would be<br />

important to propose comorbidity score for all AML patients<br />

<strong>and</strong> to evaluate how many of the patients <strong>are</strong> able to<br />

receive st<strong>and</strong>ard therapy <strong>and</strong> stem cell transplantation, how<br />

many of them <strong>are</strong> c<strong>and</strong>idate for low-intensity treatment <strong>and</strong><br />

supportive c<strong>are</strong>. It is important to have the prospective data<br />

for the patients treated in the Centers of CELG. This will<br />

give us the real situation concerning the treatment applicability<br />

in the Central <strong>and</strong> <strong>East</strong>ern Europe.<br />

Many trials showed a better outcome for patients less than<br />

45 years of age comp<strong>are</strong>d to the group of patients from 45 to<br />

60 years of age. Nonetheless most of the cooperative groups<br />

for adult AML <strong>are</strong> including patients from 18 to 60 years of<br />

age as younger ones. They received intensive chemotherapy<br />

<strong>and</strong> stem cell transplantation with st<strong>and</strong>ard conditioning. Reduced<br />

intensive conditioning has been predominantly used in<br />

patients older than 50 years of age because of lower toxicity.<br />

Older group of patients <strong>are</strong> those with age >60. The comorbidity<br />

scoring treatment approaches mentioned before has<br />

been used practically only for older patients.<br />

Disease related prognostic factors<br />

According to AML working party of European Leukemia<br />

Net several important <strong>and</strong> independent prognostic factors<br />

have been recognized as white blood cell counts, existence<br />

of prior MDS or AML with MDS feature, previous cytotoxic<br />

therapy for another malignancy, <strong>and</strong> cytogenetic <strong>and</strong><br />

molecular abnormalities in leukemic cells (10). The signifi-<br />

73


Meet the Professor Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

cance of prognostic factors is dependent on the therapy<br />

given to a patient. The genetic abnormalities according to<br />

prognostic impact <strong>are</strong> given in Table 2.<br />

Therapy of AML<br />

In acute leukemia still the first goal of the treatment is<br />

to induce good control of the leukemia (i.e. complete remission)<br />

<strong>and</strong> after that to maintain the control of leukemia<br />

or to cure it. The first part of therapy is induction therapy<br />

by which we induce remission of leukemia. In the second<br />

one so-called postremission therapy our goal is to maintain<br />

the remission state or by intensive treatment approach<br />

to cure the leukemia.<br />

74<br />

Table 2. Prognostic impact of genetic abnormalities (10)<br />

Genetic group Subsets<br />

Favorable t(8;21)(q22;q22); RUNX1-RUNX1T1<br />

inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11<br />

Mutated NPM1 without FLT3-ITD (normal karyotype)<br />

Mutated CEBPA (normal karyotype)<br />

Intermediate I Mutated NPM1 <strong>and</strong> FLT3-ITD (normal karyotype)<br />

Wild-type NPM1 <strong>and</strong> FLT3-ITD (normal karyotype)<br />

Wild-type NPM1 without FLT3-ITD (normal karyotype)<br />

Intermediate II t(9;11)(p22;q23); MLLT3-MLL<br />

Cytogenetic abnormalities not classified as favorable<br />

or adverse<br />

Adverse inv(3)(q21q26.2) or t(3;3)(q21;q26.2); RPN1-EVI1<br />

t(6;9)(p23;q34); DEK-NUP214<br />

t(v;11)(v;q23); MLL rearranged<br />

-5 or del(5q); -7; abnl(17p); complex karyotype<br />

Remission-Induction Therapy<br />

Combination chemotherapy is the primary treatment<br />

modality for patients with AML. Options for induction<br />

chemotherapy include st<strong>and</strong>ard or high dose cytarabine in<br />

combination with an anthracycline. Approximately half of<br />

patients will require a second induction cycle to obtain a<br />

complete remission. A combination of an anthracycline<br />

such as daunorubicin for three days <strong>and</strong> »st<strong>and</strong>ard« dose<br />

cytarabine for 7 days should be recommended (11). High<br />

dose of cytarabine may increase the complete remission<br />

rate in patients less than 60 years of age. It does so at the<br />

cost of increased toxicity. Treatment-related mortality, the<br />

rate of early relapse, <strong>and</strong> overall survival <strong>are</strong> not clearly<br />

improved (12).<br />

At present, there is no conclusive evidence to recommend<br />

one »7+3« regimen over another. Depending on<br />

dose, schedule, <strong>and</strong> patient selection criteria, they have<br />

similar response rates <strong>and</strong> sh<strong>are</strong> common toxicities such<br />

as severe degrees of myelosuppression with moderate mucositis,<br />

<strong>and</strong> diarrhea. Idarubicin or mitoxantrone could be<br />

used instead of daunorubicine with a similar outcome.<br />

The best induction chemotherapy for older patients with<br />

AML remains to be identified. Intensive chemotherapy may<br />

be appropriate for selected patients with low or intermediate<br />

risk disease in whom the complete remission (CR) rate<br />

can be as high as 70 to 80 percent. With this approach, median<br />

survival is approximately eight months, but 9 to 12<br />

percent of patients will be alive at five years. Although pilot<br />

studies have used more intensive initial chemotherapy, a<br />

reasonable st<strong>and</strong>ard regimen for many older patients who<br />

<strong>are</strong> medically fit is seven days of continuous infusion cytarabine<br />

plus three days of daunorubicin (13).<br />

In general, the choice of anthracycline (eg, daunorubicin,<br />

mitoxantrone, or idarubicin) does not appear to affect<br />

overall outcome. However, higher doses of anthracyclines<br />

may result in superior rates of complete remission without<br />

an app<strong>are</strong>nt increase in toxicity.<br />

So, for most older adults with favorable or intermediate<br />

risk AML <strong>and</strong> an ECOG performance status of two or less<br />

<strong>and</strong> few comorbidities, we recommend remission induction<br />

treatment with a combination of daunorubicin for<br />

three days <strong>and</strong> »st<strong>and</strong>ard« dose cytarabine for seven days<br />

rather than other chemotherapy regimens or supportive<br />

c<strong>are</strong> alone.<br />

While not curative, many Centers consider low-dose<br />

cytarabine to be the st<strong>and</strong>ard against which other treatments<br />

for AML in the older patient should be evaluated. A<br />

number of trials have investigated the use of low-dose cytarabine<br />

in older subjects with AML, both for induction<br />

<strong>and</strong> later for maintenance of remission. Although the number<br />

of complete remissions was greater with intensive<br />

chemotherapy, the early death rate was also higher. As a<br />

result, there were no differences in survival or remission<br />

duration between the patients receiving more intensive<br />

therapy <strong>and</strong> patients on low-dose cytarabine (14).<br />

Postremission therapy<br />

Post-remission therapy in acute myeloid leukemia remains<br />

problematic <strong>and</strong> controversial. It has been demonstrated<br />

that younger patients can maintain longer complete<br />

remissions (CR) with aggressive post-remission therapies<br />

after induction treatment: allogeneic autologous stem cell<br />

transplantation, or intensive chemotherapy. Still there is<br />

no evidence that auto-SCT is superior in terms of overall<br />

survival to intensive chemotherapy (15). Data clearly<br />

show that patients with unfavorable risk disease <strong>are</strong> more<br />

often addressed to allo-SCT <strong>and</strong> patients with low-risk<br />

disease receive more often intensive consolidation chemotherapy<br />

(16). At the moment most of AML patients generally<br />

receive allo-SCT on the basis of donor availability<br />

(the so called »genetic r<strong>and</strong>omization«). In the recent year<br />

allo-SCT with reduced intensity conditioning have been<br />

successfully performed for the treatment of older AML<br />

patients (8).<br />

R E F E R E N C E S<br />

1. Vardiman JW et al. Blood 2009;114:937–951.<br />

2. Döhner K et al. Blood 2005;106:3740–3746.<br />

3. Schlenk RF et al. N Engl J Med 2008;358(18):1909–1918.<br />

4. Gale RE et al. Blood 2008;111:2776–2784.<br />

5. Mufti GJ et al. Haematologica 2008;93:1712–7.<br />

6. Godley LA, Larson RA. Semin Oncol 2008;35:418–429.<br />

7. Juliusson G et al. Blood 2009;113:4179–4187.<br />

8. Blaise D et al. Exp Hematol 2010;38:1241–50.<br />

9. Sorror ML et al. Blood 2005;106:2912:2919<br />

10. Döhner H et al. Blood 2010;115:453–474<br />

11. Kolitz JE. Br J Haematol 2006;134:555.<br />

12. Weick JK et al. Blood 1996;88:2841.<br />

13. Martin MG, Abboud CN. Blood Rev 2008;22:311–20.<br />

14. Zwierzina H et al. Leukemia 2005;19:1929–33.<br />

15. Cassileth PA et al. Blood 1992;79:1924.<br />

16. Cornelissen JJ et al. Blood 2007;109:3658.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Meet the Professor<br />

MP04 Allogeneic Stem cell transplantation<br />

for hematological malignancies – current<br />

approach <strong>and</strong> future trends<br />

Boris Labar, Division of Hematology, Department of<br />

Medicine University Hospital Center Zagreb <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

At the beginning of the second decade of 21 st century, 40<br />

years after first clinical use of allogeneic stem cell transplantation<br />

(SCT) for hematological malignancies I searched<br />

through the PUBMED to find out what is the current scientific<br />

interest in the field of stem cell transplantation. For this<br />

purpose I analyzed 300 recent publications (mostly 2011).<br />

In these publications 10% of them <strong>are</strong> committed to SCT in<br />

hematological malignancies. Most of them dealing with the<br />

reports of unusual sources of stem cells predominantly cord<br />

blood mesenchimal stem cells. Some reports <strong>are</strong> showing<br />

the results of transplants with reduced intensity conditioning.<br />

Late complications <strong>and</strong> never ending story how to induce<br />

GvL effect without GvHD <strong>are</strong> also reported. Contrary<br />

to that the majority of publication describe current results of<br />

stem cell treatment in regenerative medicine, chronic disease<br />

<strong>and</strong> autoimmune diseases, as well as in many non-hematological<br />

tumors. Logical conclusion after this very preliminary<br />

survey could be: The era of allogeneic SCT for<br />

hematological malignancies from the scientific point of<br />

view is finishing?! We solved(?) the most questions as how<br />

<strong>and</strong> when to perform SCT. We know the efficacy <strong>and</strong> limitation<br />

of SCT for hematological malignancies(?!). It seems<br />

much more interesting to study the trans-differentiation <strong>and</strong><br />

immunomodulatory capacity of stem cells comp<strong>are</strong>d to the<br />

investigation how to improve the outcome of allografting in<br />

hematological malignancies. I am not going to say that there<br />

is no interest for these approaches but it may be that scientific<br />

interest is really decreasing. Moreover targeting treatment<br />

approach as in CML with TKI, seems to be more attractive<br />

for the future investigation.<br />

On the other h<strong>and</strong> in every day clinical medicine transplant<br />

is very efficient therapy for leukemia <strong>and</strong> lymphoma.<br />

For high risk patients transplant is until now the only curable<br />

therapy. The outcome is improving every decade. Still<br />

this could be a st<strong>and</strong>ard therapy for many years from now.<br />

In many transitional countries SCT is not yet a routine treatment<br />

approach. To support this let me sh<strong>are</strong> with you some<br />

recent data from the Center(s) in transitional country(ies).<br />

Incidence of stem cell transplant is increasing every year<br />

especially for acute leukemia. The reasons for that could be:<br />

SCT is the most efficient treatment options for acute leukemia<br />

<strong>and</strong> MDS (1,2);<br />

By using more efficient supportive therapy transplant related<br />

toxicity <strong>and</strong> mortality is decreasing. This make SCT<br />

more popular (3).<br />

Less intensive conditioning is associated with lower early<br />

mortality. By this approach transplant could be offer an<br />

older patient group, i.e. 50 to 60 years of age. In well developed<br />

countries many centers treated patients older than 60<br />

years of age (4, 5) (Fig. 1.).<br />

The results of transplants with match unrelated donor<br />

(MUD) according to HLA-typing with high resolution (10/10<br />

or 9/10) is similar to the results of transplants with sibling<br />

Figure 1. Incidence of SCT according to st<strong>and</strong>ard <strong>and</strong> RIC conditioning<br />

at Zagreb Center<br />

Figure 2. The incidence of SCT according to sibling <strong>and</strong> MUD donor in<br />

Zagreb Center<br />

compatible donor; this clearly increase the number of SCT<br />

with MUD donor (6, 7) (Fig. 2.)<br />

In the recent year use of cord blood (two cord blood) <strong>are</strong><br />

feasible approach for adult patients (8).<br />

Transplant should be performed in almost all hematological<br />

malignancies. But the approach when to perform transplants<br />

depends on many factors. The high toxicity <strong>and</strong> mortality<br />

<strong>are</strong> the major determinants to perform allotransplant<br />

in early phase of the disease when the antitumor efficacy is<br />

the best. Transplant related mortality with st<strong>and</strong>ard conditioning<br />

depending on patient’s age range from 20% to 50%<br />

(9, 10).<br />

The main causes of death <strong>are</strong> given In Fig. 3.<br />

Figure 3. Causes of death in 1 st 100 days following allogeneic transplant<br />

in Zagreb Center.<br />

75


Meet the Professor Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

For the diseases with the high prediction of progression<br />

or relapse (acute leukemia <strong>and</strong> myelodysplastic syndrome)<br />

after st<strong>and</strong>ard therapy allotransplant should be perform immediately.<br />

Indolent hematological malignancies or tumors<br />

with effective <strong>and</strong> less toxic therapy (lymphoma, CLL, multiple<br />

myeloma <strong>and</strong> CML) should be treated in more advanced<br />

phase of disease, when other treatment options<br />

failed. In such situation it is crucial to define evidence based<br />

medicine criteria i.e. criteria that allografting have a benefit<br />

comp<strong>are</strong>d with the other treatment options.<br />

The transplant outcome is also influenced by the availability<br />

of compatible donor, the source of stem cells, the<br />

intensity of conditioning, the incidence <strong>and</strong> severioty of<br />

early <strong>and</strong> late complications after allografting as GvHD <strong>and</strong><br />

infections. This will be also presenting during the session.<br />

R E F E R E N C E S<br />

1. Serventi-Seiwerth R et al. Acta Med Croatica 2009;63:205–8.<br />

2. Oosterveld M et al. Leukemia 2003;17:859–68.<br />

3. Vicente D et al. Bone Marrow Transplant 2007;40:349–54.<br />

4. Luger SM et al. Bone Marrow Transplant 2011; in press.<br />

5. Servais S et al. Transfus Apher Sci 2011;44:205–10.<br />

6. Ciurea SO et al. Biol Blood Marrow Transplant 2011;17:923–9<br />

7. Schlenk RF et al. J Clin Oncol 2010;28:4492–9.<br />

8. Brunstein CG et al. Blood 2010;116:4693–9.<br />

9. Remberger M et al. Biol Blood Marrow Transplant 2011; in press.<br />

10. Gupta T et al. Hematol Oncol Stem Cell Ther 2011;4:17–29.<br />

MP05 Acute lymphoblastic leukemia<br />

Deborah Thomas, (USA)<br />

76<br />

Abstract not submitted.<br />

MP06 Myelodysplastic Syndromes (MDS)<br />

Petra Muus, Theo de Witte, Dept Hematology, Radboud<br />

University Nijmegen, Medical Centre, The Netherl<strong>and</strong>s<br />

General characteristics of MDS<br />

Myelodysplastic syndromes (MDS) <strong>are</strong> a heterogeneous<br />

group of disorders affecting the normal differentiation of<br />

hematopoietic cells. Clinical manifestations of MDS overlap<br />

with aplastic anemia (AA), paroxysmal nocturnal hemoglobinuria<br />

(PNH), acute myeloid leukemia (AML) <strong>and</strong> myeloproliferative<br />

syndromes (MPS). Differential diagnosis<br />

can be challenging. Most experts do agree that the majority<br />

of MDS clones arise at the level of a pluripotent hematopoietic<br />

stem cell.<br />

All types of MDS sh<strong>are</strong> dysplastic features in one or all<br />

of the myeloid, megakaryocytic, <strong>and</strong>/or erythroid lineages,<br />

frequently resulting in cytopenias of the affected lineages<br />

(thresholds: Hb 10g/dL, absolute neutrophil count


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Meet the Professor<br />

The differential diagnosis of pancytopenia <strong>and</strong> a hypocellular<br />

bone marrow with or without relative erythroid hyperplasia<br />

is particularly challenging. The patient may have hypoplastic<br />

MDS. But it is important to know that PNH may<br />

have a similar presentation, particularly with a concomitant<br />

viral disease. Alternatively in the presence or absence of a<br />

small PNH clone the diagnosis may be aplastic anemia<br />

(AA). Hypoplastic MDS is relatively r<strong>are</strong>. Hemolytic PNH<br />

may develop from AA (AA-PNH) or r<strong>are</strong>ly out of what first<br />

appe<strong>are</strong>d to be MDS (MDS-PNH) 9 . In all cases it is important<br />

to follow the patient c<strong>are</strong>fully <strong>and</strong> repeat diagnostics in<br />

time.<br />

Treatment of MDS<br />

Treatment of MDS is as diverse as its clinical manifestations.<br />

For elderly <strong>and</strong> low risk patients supportive treatment<br />

is most commonly consisting of transfusions with erythrocytes<br />

<strong>and</strong>/or platelets. Patients with ineffective hematopoiesis<br />

generally have low intracellular levels of folic acid <strong>and</strong><br />

may profit from folic acid suppletion. Early iron chelation<br />

therapy seems important to prevent iron overload <strong>and</strong> toxicity.<br />

Growth factors (ESA’s with <strong>and</strong> without G-CSF) influencing<br />

cytopenias have a place in lower risk MDS. International<br />

guidelines suggest that patients who <strong>are</strong> either erythrocyte<br />

transfusion dependent or have low epo levels <strong>are</strong><br />

c<strong>and</strong>idates for ESA’s. G-CSF may be synergistic to ESA’s in<br />

MDS10 . Fase I studies for Romiplostim in patients with<br />

MDS <strong>and</strong> low platelet counts were promising11 <strong>and</strong> a r<strong>and</strong>omized<br />

placebo controlled phase II study is presently being<br />

analyzed. Differentiation inducing agents <strong>and</strong> low-dose<br />

chemotherapy <strong>are</strong> currently not often used anymore.<br />

An important development in recent years is Lenalidomide<br />

in lower risk MDS with 5q-. A r<strong>and</strong>omized placebo<br />

controlled study demonstrated the effectivity of the agent12 .<br />

Registration of Lenalidomide in Europe is waiting safety<br />

reports, with regards to a potentially increased incidence of<br />

AML. Studies investigating the role of lenalidomide in low<br />

risk MDS without 5q abnormalities <strong>are</strong> ongoing.<br />

Hypomethylating agents <strong>are</strong> effective in intermediate <strong>and</strong><br />

high risk (IPSS) MDS. Patients on Vidaza had a better survival<br />

than those receiving st<strong>and</strong>ard c<strong>are</strong>13 . In an almost comparable<br />

group of patients decitabine demonstrated activity<br />

as well14 . In Europe only Vidaza is approved. Currently the<br />

drug needs to be injected sc for 7 consecutive days every 28<br />

days which is quite a burden for the patient. Of note: in<br />

cases were stable disease is achieved vidaza cannot be stopped.<br />

The only accepted curative treatment for MDS is an allogeneic<br />

stem cell transplantation (alloSCT) alone or after<br />

remission induction with high-dose chemotherapy. Various<br />

studies showed a 4-year survival between 20 <strong>and</strong> 30% of<br />

treated high-risk patients. This treatment is available to only<br />

a limited number of patients because of the lack of suitable<br />

donors <strong>and</strong> the intensity of the procedure, which is in many<br />

cases incompatible with the age of the patient. Development<br />

of other transplant modalities (matched unrelated donors,<br />

non-myeloablative conditioning) have rendered alloSCT<br />

accessible to more MDS patients. Hypomethylating agents<br />

may prove sufficient <strong>and</strong> relative non-toxic cytoreductive<br />

therapy before alloSCT.<br />

As an alternative intensive remission induction, followed<br />

by intensive consolidation chemotherapy alone can be considered.<br />

Some patients responded well to intensive chemotherapy<br />

followed by autologous stem cell transplantation 15 .<br />

MDS patients may have prolonged hypoplasia following intensive<br />

chemotherapy.<br />

MDS classification, prognostic characterization plus well<br />

defined response criteria <strong>are</strong> helping advance treatment options<br />

for patients with MDS 16 .<br />

R E F E R E N C E S<br />

1. Harris NL, Jaffe ES, Diebold J, Fl<strong>and</strong>rin G, Muller-Hermelink HK,<br />

Vardiman J, Lister TA, Bloomfield CD. The World Health Organization<br />

classification of neoplastic diseases of the hematopoietic <strong>and</strong> lymphoid<br />

tissues. Report of the Clinical Advisory Committee meeting,<br />

Airlie House, Virginia, November, 1997. Ann Oncol 1999;10:1419–32.<br />

2. Bennett JM, Catovsky D, Daniel MT, Fl<strong>and</strong>rin G, Galton DA, Gralnick<br />

HR, Sultan C. Proposals for the classification of the myelodysplastic<br />

syndromes. Br J Haematol 1982;51:189–99.<br />

3. Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M,<br />

Vallespi T, Hamblin T, Oscier D, Ohyashiki K, Toyama K, Aul C, Mufti<br />

G, Bennett J. International scoring system for evaluating prognosis in<br />

myelodysplastic syndromes. Blood 1997;89:2079–88.<br />

4. Greenberg PL, Sanz GF, Sanz MA. Prognostic scoring systems for risk<br />

assessment in myelodysplastic syndromes. Forum (Genova) 1999;9:<br />

17–31.<br />

5. Malcovati L, Germing U, Kuendgen A et al. Time-dependent Prognostic<br />

Scoring System for Predicting Survival <strong>and</strong> Leukemic Evolution in<br />

MDS. J CLin Oncol 2007;25:3503–3510.<br />

6. Aul C, Giagounidis A, Germing U. Epidemiological features of myelodysplastic<br />

syndromes: results from regional cancer surveys <strong>and</strong> hospital-based<br />

statistics. Int J Hematol 2001;73:405–10.<br />

7. Cogle R, Craig BJ, Rollison DE <strong>and</strong> List AF. Incidence of MDS using<br />

a novel claims-based algorithm: high number of uncaptured cases by<br />

cancer registries. Blood 2011;117:7121–7125.<br />

8. Madkaikar M, Gupta M, Jijina F, Ghosh K.Paroxysmal nocturnal haemoglobinuria:<br />

diagnostic tests, advantages, & limitations. Eur J Haematol<br />

2009;83(6):503–11.<br />

9. Li Y, Li X, Ge M, Shi J, Qian L, Zheng Y, Wang J. Long-term follow-up<br />

of clonal evolutions in 802 aplastic anemia patients: a single-center experience.<br />

Ann Hematol 2011;90:529–37.<br />

10. Jadersten M, Montgomery SM, Dybedal I et al. Longterm outcome of<br />

treatment of anemia in MDS with Erythropoietin <strong>and</strong> G-CSF. Blood<br />

2005;106:803–811.<br />

11. Kantarjian H, Fenaux P, Sekeres MA, Becker PS, Boruchov A, Bowen<br />

D, Hellstrom-Lindberg E, Larson RA, Lyons RM, Muus P, Shammo J,<br />

Siegel R, Hu K, Franklin J, Berger DP Safety <strong>and</strong> efficacy of romiplostim<br />

in patients with lower-risk myelodysplastic syndrome <strong>and</strong> thrombocytopenia.<br />

J Clin Oncol 2010;20;28(3):437–44.<br />

12. Fenaux P, Giagounidis A, Selleslag D, Beyne-Rauzy O, Mufti G, Mittelman<br />

M, Muus P, Te Boekhorst P, Sanz G, Del Cañizo C, Guerci-Bresler<br />

A, Nilsson L, Platzbecker U, Lübbert M, Quesnel B, Cazzola M,<br />

Ganser A, Bowen D Dr, Schlegelberger B, Aul C, Knight R, Francis J,<br />

Fu T, Hellström-Lindberg E. A r<strong>and</strong>omized phase 3 study of lenalidomide<br />

vs placebo in RBC transfusion-dependent patients with Low-/Intermediate-1-risk<br />

MDS with del5q. Blood. 2011 Jul 13. [Epub ahead of<br />

print].<br />

13. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis<br />

A, Schoch R, Gattermann N, Sanz G, List A, Gore SD, Seymour<br />

JF, Bennett JM, Byrd J, Backstrom J, Zimmerman L, McKenzie<br />

D, Beach C, Silverman LR; International Vidaza High-Risk MDS Survival<br />

Study Group. Efficacy of azacitidine comp<strong>are</strong>d with that of conventional<br />

c<strong>are</strong> regimens in the treatment of higher-risk myelodysplastic<br />

syndromes: a r<strong>and</strong>omised, open-label, phase III study. Lancet Oncol<br />

2009;10:223–32.<br />

14. Lübbert M, Suciu S, Baila L, Rüter BH, Platzbecker U, Giagounidis A,<br />

Selleslag D, Labar B, Germing U, Salih HR, Beeldens F, Muus P,<br />

Pflüger KH, Coens C, Hagemeijer A, Eckart Schaefer H, Ganser A, Aul<br />

77


Meet the Professor Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

C, de Witte T, Wijermans PW. Low-dose decitabine versus best supportive<br />

c<strong>are</strong> in elderly patients with intermediate- or high-risk myelodysplastic<br />

syndrome (MDS) ineligible for intensive chemotherapy: final<br />

results of the r<strong>and</strong>omized phase III study of the European Organisation<br />

for Research <strong>and</strong> Treatment of Cancer Leukemia Group <strong>and</strong> the<br />

German MDS Study Group. J Clin Oncol 2011;29(15):1987–96.<br />

15. de Witte T, Suciu S, Verhoef G, Labar B, Archimbaud E, Aul C, Selleslag<br />

D, Ferrant A, Wijermans P, M<strong>and</strong>elli F, Amadori S, Jehn U, Muus<br />

P, Boogaerts M, Zittoun R, Gratwohl A, Zwierzina H, Hagemeijer A,<br />

Willemze R: Intensive chemotherapy followed by allogeneic or autologous<br />

stem cell transplantation for patients with myelodysplastic syndromes<br />

(MDSs) <strong>and</strong> acute myeloid leukemia following MDS. Blood<br />

2001;98:2326–31.<br />

16. Greenberg P. Current therapeutic approaches for patients with MDS.<br />

Brit J of Haematology 2010;150:131–143.<br />

MP07 Diagnostic <strong>and</strong> therapeutic challanges<br />

in B – Chronic lymphocytic leukemia<br />

(B-CLL)<br />

Branimir Jaksic, Department of Medicine, University<br />

Hospital Merkur <strong>and</strong> School of Medicine, University of<br />

Zagreb, Croatia.<br />

In this session we will address the frequent challenges in<br />

the management of B-CLL. Some of the recent major clinical<br />

research advancements in the diagnostics <strong>and</strong> clinical<br />

therapeutic trials have put a new perspective on the actual<br />

nature of the disorder <strong>and</strong> helped to modify conceptual <strong>and</strong><br />

practical clinical approaches. Therefore we will address<br />

challenges referring to the diagnostics first, followed by the<br />

therapy, <strong>and</strong> finally discuss some of the typical scenarios<br />

from the practical clinical point of view.<br />

Diagnostics. Diagnostic criteria have recently been both<br />

simplified <strong>and</strong> made more specific. Thus, only a peripheral<br />

blood sample is required for the diagnosis, but at the same<br />

time the morphology supplemented by flow cytometry defined<br />

immune phenotype has become m<strong>and</strong>atory. In clinical<br />

practice this opened the problem in a definition of required<br />

panel of monoclonal antibodies <strong>and</strong> required level of their<br />

expression to distinguish between »typical« B-CLL <strong>and</strong><br />

»atypical or B-CLL variant(s)« as far as immune phenotype<br />

is concerned. Moreover, the concept of variants may encompass<br />

more clinical variability, such as distinct patterns<br />

of the tumor mass distribution <strong>and</strong> similar.<br />

The simple clinical <strong>and</strong> hematological parameters <strong>are</strong><br />

also commonly used for the evolution monitoring. We have<br />

been using for many years the Total Tumor Mass Scoring<br />

System (TTM) that is exclusively disease related. It proved<br />

to be very useful for both monitoring of the disease evolution<br />

<strong>and</strong> its response to the treatment independently of toxicities.<br />

Perhaps, the addition of more cellular <strong>and</strong> humoral<br />

immunological new parameters <strong>and</strong>/or infection related<br />

triggers may improve prediction of disease evolution.<br />

The comorbidity evaluation is important prerequisite for<br />

sound clinical decisions. As a rule, more effective treatments<br />

<strong>are</strong> at the same time associated with more toxicities<br />

as shown in the recently published trials. The overall clinical<br />

benefit should be c<strong>are</strong>fully balanced taking into account<br />

expected effect <strong>and</strong> expected tolerance. Thus, this part of<br />

the diagnostic process aimed to predict tolerance should<br />

never be neglected. The cumulative illness rating scales <strong>are</strong><br />

78<br />

helpful (CIRS-G is widely used for CLL) both in clinical<br />

trials <strong>and</strong> routine practice.<br />

The prognostic factors <strong>are</strong> individual variables that <strong>are</strong><br />

associated with prognosis. The prognosis is usually measured<br />

by the length of survival. The clinical usefulness of a<br />

given prognostic factor is related to the strength of its independent<br />

prognostic power. In addition to help prognostication,<br />

the prognostic analyses represent a powerful research<br />

tool to evaluate both biological <strong>and</strong> clinical relevance of a<br />

given variable. It is important to note that prognostic factors<br />

may be classified into (1) disease related, (2) patient related<br />

(B-CLL unrelated) <strong>and</strong> (3) intermediate group that may be<br />

in variable extent both disease <strong>and</strong> patient related. The distinction<br />

is important since disease related <strong>and</strong> patient related<br />

factors have different clinical meanings. The c<strong>are</strong>ful clinical<br />

evaluation is particularly important in the intermediate<br />

group, to classify a prognostic factor according to underlying<br />

cause. For example the presence of anemia or elevation<br />

of serum beta -2-microglobulin may be present due to disease<br />

itself or to disease unrelated condition, <strong>and</strong> consequently<br />

may have different impact on clinical decision on<br />

starting therapy<br />

The prognostic indices <strong>are</strong> composite scales that comprise<br />

several individual variables. The variables <strong>are</strong> chosen<br />

by their mutually independent prognostic power(s), <strong>and</strong> <strong>are</strong><br />

therefore excellent predictors of prognosis. However, if<br />

combining prognostic factors related to disease with those<br />

unrelated to the disease (which is a very common situation)<br />

they may have limited clinical impact. While a bad prognosis<br />

due to presence of the factors related to the disease warrants<br />

more aggressive treatment, patient related prognostic<br />

factors as for example advanced age may have clinically opposite<br />

indication. Failure to recognize this concept may<br />

have deleterious clinical repercussions.<br />

Therapy. There <strong>are</strong> two important basic questions for<br />

clinical practice: (1) When to treat? <strong>and</strong> (2) How to treat?<br />

Both questions <strong>are</strong> interrelated <strong>and</strong> depend mostly on the<br />

achievable aims of therapy definition. Based on the clinical<br />

trials results performed in 1980s that showed no advantage<br />

of early institution of therapy, it is today recommended in<br />

most of guidelines to refrain therapy until indication of active<br />

disease present. Although this is the common clinical<br />

practice, it is somehow problematic conceptually. Namely,<br />

this policy has resulted in a watch <strong>and</strong> wait practice for variable<br />

period of time, but once the indication to start therapy<br />

is adopted, there is a shift in goal, with a tendency to apply<br />

the most effective treatment aiming molecular response, or<br />

in other words, the eradication of the disease. This is in contrast<br />

to general paradigm in oncology that effective treatment<br />

should be applied with no delay. Thus, in current B-<br />

CLL clinical practice we often wait until the full blown<br />

disease to try to eradicate it. If we really believe in eradication<br />

potential of the given antineoplastic therapy (as may be<br />

the case according to the latest reports), this therapy should<br />

definitely be tried earlier in the course of the disease.<br />

As far as to define achievable aims of therapy, there is<br />

little doubt that the best would be to achieve eradication of<br />

the disease resulting in cure. The eradication will also mean<br />

to document molecular remission up to the sensitivity of<br />

available techniques. Also, this approach may be effective


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Meet the Professor<br />

in preventing emergence of therapy resistant, usually more<br />

malignant clones of the disease in contrast to the low intensity<br />

treatment that may favor emergence of resistance. However,<br />

this conceptually sound approach may not be feasible,<br />

because more effective treatment is usually more toxic. For<br />

this reason the aim should be balanced with expected tolerance,<br />

<strong>and</strong> if appropriate, by defining less ambitious aims<br />

such as major clinical remission or minor remission or stable<br />

disease. To achieve targeted aim the least toxic therapies<br />

should be chosen. This approach is a variant of »toxicity<br />

tailored therapy« taking into account comorbidity <strong>and</strong> other<br />

patient related factors, which all requires high level of clinical<br />

expertise.<br />

These general principles should be applied with caution<br />

on individual patients, particularly because an individual<br />

response <strong>and</strong> tolerance may vary considerably <strong>and</strong> they <strong>are</strong><br />

difficult to anticipate with certainty. Therefore therapy should<br />

be interactive <strong>and</strong> should be modified according to the<br />

achieved results. If necessary the goals should be reset. It is<br />

logical to start with the goal set as high as possible in accordance<br />

with the available data <strong>and</strong> clinical judgment, <strong>and</strong><br />

later proceed pending on the documented response <strong>and</strong> tolerance.<br />

Note that supportive c<strong>are</strong> is of the utmost importance,<br />

<strong>and</strong> therefore should be always delivered at the highest possible<br />

level.<br />

Clinical practice typical scenarios. Although the diagnostics<br />

<strong>and</strong> particularly therapy should be individualized in<br />

order to conform to the above principles <strong>and</strong> concepts, several<br />

clinical examples of the frequent clinical situations will<br />

be discussed, to illustrate the important challenges that <strong>are</strong><br />

present in the B-CLL management.<br />

MP08 Myelofi brosis<br />

Srdjan Verstovsek, MD Anderson Cancer Center, Leukemia<br />

Department, Houston, Texas, USA<br />

Myelofibrosis (MF) is a myeloproliferative neoplasm<br />

(MPN) characterized by abnormal proliferation of megakaryocytes,<br />

fi brosis of the bone marrow (BM), osteosclerosis,<br />

extramedullary hematopoiesis (EMH), <strong>and</strong> progressive<br />

BM failure with an increased risk of progression to acute<br />

myeloid leukemia (AML). MF was known by other names<br />

in the past, including chronic idiopathic MF, agnogenic<br />

myeloid metaplasia, <strong>and</strong> MF with myeloid metaplasia. MF<br />

is part of the classic Philadelphia chromosome–negative<br />

MPNs alongside polycythemia vera (PV) <strong>and</strong> essential<br />

thrombocythemia (ET). MF can occur as a primary disorder<br />

(PMF) or secondary to other MPNs, mainly PV (post-PV<br />

MF) <strong>and</strong> ET (post-ET MF). MF is a r<strong>are</strong> disease, with an<br />

incidence rate estimated at 1.46 cases/100,000 persons/year.<br />

Median age at diagnosis is 67 years, <strong>and</strong> both sexes <strong>are</strong><br />

equally affected. The prognosis of patients with MF is quite<br />

variable, as some patients can have a very indolent disorder<br />

with prolonged survival, whereas in other cases, the disease<br />

follows an aggressive course. Median survival of patients<br />

with MF is in the range of 69 months, <strong>and</strong> survival at 5 <strong>and</strong><br />

10 years is decreased 40% <strong>and</strong> 60%, respectively, comp<strong>are</strong>d<br />

with age <strong>and</strong> sex-matched controls. Major causes of death<br />

include transformation to acute leukemia, progressive BM<br />

failure due to MF, thrombotic <strong>and</strong> cardiovascular complications,<br />

<strong>and</strong> portal hypertension. The pathogenesis of MF is<br />

still unknown. Great advances in our underst<strong>and</strong>ing of the<br />

pathogenesis of this disorder were made with the discovery<br />

of mutations involved in cytokine receptor signaling pathways<br />

(JAK2V617F <strong>and</strong> MPLW515K/L). First-degree relatives<br />

of patients with MF <strong>and</strong> PV/ET have a 5- to 7-fold increased<br />

risk of developing MPNs, suggesting a strong genetic<br />

component in the susceptibility to this disease. Indeed,<br />

a specific single nucleotide polymorphism of the JAK2 gene<br />

(rs10974944) has been associated with an increased risk of<br />

developing JAK2V617F positive MPNs. Treatment of MF<br />

is suboptimal <strong>and</strong> palliative in nature, mainly directed toward<br />

alleviation of symptoms due to cytopenias or splenomegaly.<br />

In the past decade, several new drugs have been<br />

introduced for the treatment of patients with MF, <strong>and</strong> it is<br />

expected that the outcomes for these patients will soon improve<br />

as new compounds become available.<br />

MP09 Non-Hodgkin’s lymphoma<br />

Igor Aurer, Department of Medicine, Clinical Hospital<br />

Center <strong>and</strong> School of Medicine, University of Zagreb,<br />

Croatia<br />

New treatment modalities <strong>and</strong> new lymphoma classifications<br />

have substantially increased the number of different<br />

scenarios a physician could be facing when seeing patients<br />

with non-Hodgkin lymphomas (NHL). While good evidence<br />

exists for early treatment phases in patients with the<br />

more frequent NHL types, the same is not true for advanced<br />

disease, elderly <strong>and</strong> patients with significant comorbidities<br />

<strong>and</strong> r<strong>are</strong> NHL types.<br />

During this session I will review treatment st<strong>and</strong>ards <strong>and</strong><br />

recommendations for different types of NHL in different<br />

patient populations. Attendees will be urged to present problems<br />

they <strong>are</strong> or have been facing <strong>and</strong> discuss treatment options<br />

with the audience.<br />

Specific problems we will address include:<br />

1) Diffuse large-B cell lymphoma in elderly <strong>and</strong> patients<br />

with limited cardiac function<br />

2) Burkitt’s lymphoma<br />

3) Relapsing / refractory mantle-cell lymphoma<br />

4) Nodal non-follicular indolent lymphoma<br />

5) Extranodal marginal zone lymphoma<br />

6) Localized nodal indolent lymphomas<br />

7) Peripheral T-cell lymphomas<br />

MP10 Multiple Myeloma<br />

Damir Nemet, Division of Hematology, Department of<br />

Medicine, Clinical Hospital Centre Zagreb <strong>and</strong> School of<br />

Medicine, University of Zagreb, Zagreb, Croatia<br />

Multiple myeloma (MM) is a malignant hematological<br />

disease characterized by the accumulation of clonal plasma<br />

cells in bone marrow, presence of monoclonal protein in the<br />

blood <strong>and</strong>/or urine, destructive bone disease, hypercalce-<br />

79


Meet the Professor Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

mia, renal failure, <strong>and</strong> hematologic dysfunction. MM accounts<br />

for about 13% of all hematological neoplastic disease<br />

<strong>and</strong> 1% of all cancers. The annual age-adjusted incidence<br />

is 4,0–5,6 cases per 100.000 persons. The median age<br />

of patients at the time of diagnosis is about 65–70 years.<br />

In the marrow, MM cells interact with stromal components<br />

<strong>and</strong> other cells in the microenvironment to receive<br />

growth <strong>and</strong> survival factors <strong>and</strong> to modulate their environment<br />

inducing pathological phenomena, such as neovascularisation<br />

(angiogenesis <strong>and</strong> vasculogenesis) <strong>and</strong> bone disruption<br />

(osteoclastogenesis). Almost all patients with MM<br />

evolve from a premalignant condition known as monoclonal<br />

gammapathy of undetermined significance (MGUS). Smoldering<br />

myeloma (SMM) is an intermediate asymptomatic<br />

advanced stage which much more probability of progression<br />

to symptomatic disease. There is no need for treatment<br />

of smoldering MM until progression to active, symptomatic<br />

disease. The most important is to assess risk of progression<br />

in individual patient.<br />

Major clinical manifestations of MM <strong>are</strong> osteolytic bone<br />

lesions, anemia, hypercalcemia, renal failure, <strong>and</strong> an increased<br />

risk of infections. Extramedullary plasmacytomas<br />

<strong>and</strong> extramedullary expansion of lesions originated in the<br />

bone can be observed.<br />

The diagnosis of MM requires ≥10% of clonal BM plasma<br />

cells or a biopsy proven plasmacytoma, presence of serum<br />

<strong>and</strong>/or urinary monoclonal protein (true non-secretory<br />

myeloma is r<strong>are</strong>), <strong>and</strong> evidence of end organ damage known<br />

as CRAB: hypercalcemia, renal impairment, anemia, <strong>and</strong><br />

bone lesions. Several staging systems have been built to<br />

separate patients into various risk groups with different outcomes.<br />

The International Staging System (ISS) is now<br />

widely used; it is highly prognostic but presents some limitations.<br />

ISS separates patients into three groups based on<br />

beta-2-microglobulin <strong>and</strong> serum albumin concentrations<br />

with significant difference in survival. However, chromosomal<br />

abnormalities like del 13 on karyotyping, t(4;14),<br />

t(14;16), del 17p <strong>and</strong> 1q gains significantly impact outcome<br />

<strong>and</strong> some prognostic models <strong>are</strong> based solely on the presence<br />

of chromosomal changes.<br />

Treatment options have traditionally included corticosteroids,<br />

alkylating or other cytotoxic agents, <strong>and</strong> the use of<br />

high-dose chemotherapy followed by autologous stem cell<br />

transplantation (ASCT). Recent advances include use of<br />

novel immunomodulatory drugs thalidomide <strong>and</strong> lenalidomide,<br />

<strong>and</strong> proteasome inhibitor bortzomib. Despite these<br />

advances, most of the patients <strong>are</strong> not cured <strong>and</strong> ultimately<br />

develop chemotherapy-refractory disease.<br />

With the use of ASCT <strong>and</strong> after emergence of novel<br />

agents overall survival in MM has improved significantly in<br />

the last two decades. ASCT is a st<strong>and</strong>ard treatment approach<br />

80<br />

for younger (≥65 or even 70 years old) patients, therefore at<br />

initial treatment considerations MM patients <strong>are</strong> generally<br />

divided in »transplant eligible« <strong>and</strong> »transplant ineligible«<br />

group. Clinical trials so far have shown that high-dose therapy<br />

<strong>and</strong> ASCT improves response rates, disease-free survival<br />

(DSF), <strong>and</strong> overall survival (OS) in symptomatic patients<br />

with MM. More than half of MM patients <strong>are</strong> not eligible<br />

for high-dose therapy including elderly patients <strong>and</strong><br />

those with comorbidities. Recent results indicate significant<br />

improvement by adding novel drugs to st<strong>and</strong>ard therapy. AlloSCT<br />

with myeloablative or reduced intensity conditioning<br />

(RIC) is still investigational in myeloma <strong>and</strong> not widely accepted<br />

primarily due to still high transplant related mortality<br />

<strong>and</strong> high GvHD rates.<br />

Post-transplant strategies could be defined as »consolidation«<br />

(short term drug therapy, usually 3–6 months of duration)<br />

or »maintenance« (prolonged low dose drug therapy).<br />

At present it is unclear what strategies <strong>are</strong> effective, do these<br />

strategies prolong OS, do all patients should receive consolidation/maintenance,<br />

<strong>and</strong>, if not, who should receive<br />

such therapy.<br />

Almost all patients with MM eventually relapse. Options<br />

for further therapy include alkylating agents, corticosteroids,<br />

thalidomide, lenalidomide, bortezomib or liposomal<br />

doxorubicin in various combinations. The choice of treatment<br />

depends on several factors including drugs used as<br />

initial therapy, the degree <strong>and</strong> duration of response to previous<br />

therapy, type of relapse/progression (aggressive relapse<br />

or more indolent relapse), <strong>and</strong> previous toxicity. Pomalidomide,<br />

a new immunomodulatory drug <strong>and</strong> carfilzomib, a<br />

novel proteasome inhibitor, have significant activity in relapsed<br />

<strong>and</strong> refractory MM. Beside this, promising agents<br />

<strong>are</strong> histone deacetylase inhibitors vorinostat <strong>and</strong> panobiostat<br />

<strong>and</strong> potent anti-CS-1 antibody elotuzumab. Results of<br />

phase II studies with bendamustin <strong>are</strong> encouraging.<br />

Supportive therapy is an important part of whole strategy<br />

of myeloma treatment. Bisphosphonates, especially nitrogen-containing<br />

compounds (pamidronate, zoledronic acid)<br />

<strong>are</strong> potent inhibitors of bone resorption. They reduce skeletal<br />

complications in MM patients. RANKL inhibition by<br />

monoclonal antibody denosumab show promising results in<br />

clinical studies. Patients with MM <strong>are</strong> at high risk of developing<br />

infections. Appropriate prophylaxis is therefore m<strong>and</strong>atory<br />

during myeloma treatment. Renal failure is an important<br />

complication of myeloma. Symptomatic c<strong>are</strong> (appropriate<br />

hydration, urine alkalization, avoidance of nephrotoxic<br />

therapy <strong>and</strong> bisphosphonates) <strong>are</strong> important as well<br />

as immediate anti-myeloma treatment. Bortezomib is probably<br />

the best choice in patients with renal impairment due to<br />

his fast anti-myeloma activity, influence on inflammatory<br />

response, <strong>and</strong> no need for dose reduction.


State of the Art II.<br />

SA02 Diagnostic <strong>and</strong> Treatment Approach for<br />

Acute Myeloblastic Leukemia – Report from<br />

Central <strong>and</strong> <strong>East</strong>ern European Leukemia<br />

Group (CELG)<br />

Boris Labar 1 , Agnieszka Wierzbovska 2 , Georgi Mihaylov 3 ,<br />

Andreaa Moicean 4 , Dragica Tomin 5 , Zita Borbenyi 6 ,<br />

K<strong>are</strong>l Indrak 7 , Lidija Cevreska 8 , Martin Mistrik 9 ,<br />

W<strong>and</strong>a Knopinska-Posluszny 10 , Vildan Bijedic 11 .<br />

1 Division of Hematology, Department of Medicine,<br />

University Hospital Center <strong>and</strong> School of Medicine<br />

University of Zagreb, Croatia; 2 Department of<br />

Hematology, Medical University of Lodz, Copernicus<br />

Memorial Hospital, Lodz, Pol<strong>and</strong>; 3 Queen Joanna<br />

University Hospital, Sofia, Bulgaria; 4 Fundeni Clinical<br />

Institute, Second Department of Hematology, Buch<strong>are</strong>st,<br />

Romania; 5 Clinic of Hematology, Clinical Center Serbia,<br />

Belgrade, Serbia; 6 Second Department of Medicine,<br />

University Medical School of Szeged,<br />

Hungary; 7 Department of Hemato-Oncology, University<br />

Hospital Olomouc <strong>and</strong> Faculty of Medicine <strong>and</strong> Dentistry,<br />

Palacky University Olomouc, Czech Republic;<br />

8 Department of Hematology, Medical Faculty, Skopje,<br />

Republic of Macedonia; 9 Department of Hematology <strong>and</strong><br />

Transfusiology, University Hospital, Bratislava, Slovakia;<br />

10 Department of Clinical Biochemistry, University Medical<br />

School, Gdańsk, Pol<strong>and</strong>; 11 Department of Hematology,<br />

KCU, Sarajevo, Bosnia <strong>and</strong> Herzegovina.<br />

Abstract: Central <strong>and</strong> <strong>East</strong>ern Leukemia Cooperative<br />

Group (CELG) perform retrospective analysis of diagnostic<br />

<strong>and</strong> treatment possibilities for acute myeloblastic leukemia<br />

(AML). The results of the questionnaire for acute myeloblastic<br />

leukemia (AML) <strong>are</strong> presented.<br />

In a retrospective manner using the Questionnaire we analyzed<br />

in 26 centers of 10 countries the diagnostic <strong>and</strong> treatment<br />

possibilities for AML.<br />

In 10 countries more than 880 patients with AML have<br />

been treated per year. Most of the countries have the st<strong>and</strong>ard<br />

diagnostic tools. In some countries there <strong>are</strong> no facilities<br />

for cytogenetic <strong>and</strong> cryopreservation. Induction <strong>and</strong><br />

consolidation chemotherapy is cytarabine based in combination<br />

with anthracycline. The number of transplants is low<br />

due to insufficient number of transplant centers. In four<br />

countries patients <strong>are</strong> treated in rooms with two or more<br />

beds.<br />

The number of patients is sufficient for the future prospective<br />

clinical trials. Concerning the diagnostic tools,<br />

st<strong>and</strong>ard approach has been routinely performing in all centers.<br />

Therapy approach is cytarabine based in combination<br />

with anthracycline. Stem cell transplantation is performing<br />

in majority of Centers but still the number of transplants is<br />

low. The level of supportive c<strong>are</strong> differs from country to<br />

country <strong>and</strong> one of the priorities of CELG group is to st<strong>and</strong>ardize<br />

the diagnostic <strong>and</strong> treatment approaches. This is a<br />

prerequisite for the future clinical trials.<br />

Introduction<br />

In 2009 the 13 representatives of Central <strong>and</strong> <strong>East</strong>ern European<br />

countries founded the new group named Central <strong>and</strong><br />

<strong>East</strong>ern European Cooperative Leukemia Group (CELG).<br />

During the several meetings two main goals of the group<br />

activity were defined: 1. to design the prospective clinical<br />

trials for treatment of hematological malignancies <strong>and</strong> 2. To<br />

propose the guidelines <strong>and</strong> st<strong>and</strong>ards of diagnosis <strong>and</strong> treatment.<br />

One of the first activities of CELG group was to perform<br />

retrospective analysis dealing with the diagnostic <strong>and</strong> treatment<br />

possibilities for acute myeloblastic leukemia (AML).<br />

The results of the questionnaire for AML <strong>are</strong> presented.<br />

Methods<br />

Participating countries <strong>and</strong> centers:<br />

Twenty-six Centers from 10 countries respond the Questionnaire<br />

(Fig.1).<br />

Figure 1. Number of Centers from participating countries<br />

Questionnaire:<br />

The Questionnaire was create to get the answer about the<br />

AML diagnostic <strong>and</strong> treatment approach in Central <strong>and</strong><br />

<strong>East</strong>ern European countries. The questionnaire consists of<br />

three parts:<br />

Part one give the answers about the participating countries,<br />

number of Centers in each country, <strong>and</strong> number of patients<br />

treated per year.<br />

Part two is dealing with diagnostic possibilities for AML:<br />

morphology/cytochemistry, immunophenotyping, cytogenetic/molecular<br />

genetics, biobanking, HLA typing, microbiology.<br />

Part three is treatment part for AML: Induction therapy,<br />

postremission therapy (intensive chemotherapy, maintenance<br />

chemotherapy, autologous <strong>and</strong> allogeneic stem cell transplantation),<br />

supportive c<strong>are</strong> (infection prevention, growth<br />

factors, transfusion support).<br />

81


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

Statistical analysis<br />

Descriptive methods 1 have been used to present the obtained<br />

data from Questionnaire.<br />

Results<br />

Number of patients treated in CELG Centers.<br />

In 26 Centers from 10 countries (Fig 2.) 881 patients with<br />

AML have been treated during a period of 1 year.<br />

Figure 2. Number of patients with AML treated per year according to<br />

participating countries<br />

Most Centers were treated more than 20 patients per year;<br />

but in 4 of them less than 10 patients received therapy for<br />

AML. More than 60% of treated patients were younger than<br />

60 years of age (Fig. 3).<br />

Figure 3. Age of treated patients with AML according to participating<br />

countries<br />

Diagnosis of AML<br />

The majority of Centers <strong>are</strong> able to perform the st<strong>and</strong>ard<br />

diagnostic tools for acute leukemia as cytomorphology, cytochemistry<br />

<strong>and</strong> multiparameter (≥ 3 color) flow cytometry,<br />

as well as qualitative PCR (Fig. 4.).<br />

In one country there is no cytogenetic laboratory. Centers<br />

in 4 countries have no facilities for cryopreservation (Fig.<br />

5.) <strong>and</strong> they <strong>are</strong> not able to store the samples of peripheral<br />

blood <strong>and</strong> marrow. In 6 countries galactommanan test <strong>are</strong><br />

routinely performing for diagnosis of fungal infection (Fig<br />

5.). Comorbidity prognostic index2 has been used as a treatment<br />

predictor in 8 countries, especially in older patients.<br />

82<br />

Legends: BMA – bone marrow aspiration, Biopsy – bone marrow biopsy;<br />

Cytochem – cytochmistry; FC/+3 – fl ow cytometry using 3color;<br />

Genome – Genome molecular assays<br />

Figure 4. St<strong>and</strong>ard diagnostic facilities in centers of participating countries.<br />

Legend: Cryopres. – Cryopreservation<br />

Figure 5. Biobanking, cryopreservation, HLA-typing <strong>and</strong> infection diagnostic<br />

possibilities in centers of participating countries.<br />

Treatment of AML<br />

For patient younger than 60 years of age cytarabine based<br />

chemotherapy was the main regimen (Fig. 6.). For induction<br />

therapy »3+7« regimen 3 is the most frequently used treatment.<br />

For consolidation combination of intermediate dose<br />

of cytarabine in combination with anthracycline 4 is the most<br />

popular treatment option. Some centers treated AML according<br />

to EORTC protocol 5 . They added etopozide to anthracycline<br />

<strong>and</strong> cytarabine for induction therapy. Polish<br />

Centers have been using their own protocol, combination of<br />

cytarabine, cladribine <strong>and</strong> anthracycline 6,7 .<br />

Legend: EORTC – European Organization for Research <strong>and</strong> Treatment of<br />

Cancer; PALG – Polish Acute Leukemia Group; IntAraC – Intermediate<br />

dose of cytarabine<br />

Figure 6. Induction <strong>and</strong> postremission chemotherapy for patients 18 to<br />

60 years of age


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

For older fit patients most of the CELG centers have been<br />

using 3+7 protocol (Fig 7.) For frail patients low dose cytarabine<br />

or symptomatic therapy has been the most frequent<br />

treatment approach (Fig. 7.).<br />

Legend: 3+7 – (daunoblastin 3 days + cytarabine 7 days); 2+5 (daunoblastin 2<br />

days + cytarabine 5 days); MICE – mitoxantrone, cytarabine, etoposide;<br />

Ara-C based – cytarabine based; LD-Ara-C – Low dose cytarabine;<br />

ASCT – Autologous stem cell transplantation; Hydrea – hydroxyurea<br />

Figure 7. Induction <strong>and</strong> postremission chemotherapy for patients older<br />

than 60 years of age<br />

Out of 667 patients eligible for transplantation, 194 of<br />

them (28%) received stem cells in post-consolidation period;<br />

15% were treated with allogeneic stem cell transplantation<br />

while 13% of patients received autologous stem cell<br />

(Fig. 8.).<br />

Legend: Allo-SCT – allogeneic Stem cell transplantation;<br />

Auto-SCT – autologous stem cell transplantation<br />

Figure 8. Number of patients treated with allogeneic <strong>and</strong> autologous<br />

stem cell transplantation<br />

Concerning the source of stem cells in autologous group<br />

most of the patients received stem cells from peripheral<br />

blood (97%). In allogeneic group in ¾ of patients stem cells<br />

were from peripheral blood, while in ¼ of them marrow was<br />

the source of stem cells. Two patients received stem cells<br />

from cord blood (Fig 9.).<br />

Allogeneic stem cell transplantation is not a st<strong>and</strong>ard<br />

treatment options in two countries. Therapy with match unrelated<br />

donor as well as conditioning with reduced intensity<br />

is still not available in two countries (Fig 10.).<br />

Legend: Allo-SCT – allogeneic Stem cell transplantation;<br />

Auto-SCT – autologous stem cell transplantation<br />

Figure 9. Source for stem cells<br />

Legend: Allo-SCT – allogeneic stem cell transplantation; MUD – Match<br />

unrelated donor; RIC – reduced intensity conditioning<br />

Figure 10. Availability of allogeneic stem cell transplantation, transplantation<br />

with unrelated donor <strong>and</strong> transplantation with reduced intensity<br />

conditioning<br />

Concerning the supportive c<strong>are</strong> 6 countries (four of them<br />

with laminar air-flow) <strong>are</strong> treated AML patients in 1-bed<br />

room, according to the principles of reverse isolation, while<br />

in 4 countries patients <strong>are</strong> caring in two or more than 2-bed<br />

rooms (Fig. 11.) Laminar air-flow unites <strong>are</strong> used exclusively<br />

for allogeneic transplantation.<br />

Legend: LAF – laminar air-fl ow<br />

Figure 11. St<strong>and</strong>ard of c<strong>are</strong> of the neutropenic patients<br />

Quinolones <strong>and</strong> fluconazole <strong>are</strong> the principal antimicrobial<br />

<strong>and</strong> antifungal drugs for bacterial <strong>and</strong> fungal infection<br />

prophylaxis respectively. All centers have the possibilities<br />

for platelet <strong>and</strong> red blood cell transfusion support although<br />

the criteria for transfusion differ from center to center.<br />

83


State of the Art Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

Discussion<br />

Most of the Centers in the Central <strong>and</strong> <strong>East</strong>ern European<br />

countries <strong>are</strong> well experienced centers for the diagnosis <strong>and</strong><br />

treatment of AML. In most of them the number of patient is<br />

sufficient to get all the necessary skills for AML diagnosis<br />

<strong>and</strong> treatment. But still some centers <strong>are</strong> treating small number<br />

of patients. In view of the current diagnostic <strong>and</strong> treatment<br />

approach especially the need for new molecular genome<br />

technique <strong>and</strong> transplantation together with the targeted<br />

therapy it is not rational to develop the center with a<br />

small number of patients. The cost of proper c<strong>are</strong> <strong>and</strong> treatment<br />

is too high for such a small number of patients. The<br />

recommendation is to treat not less than 30 patients per<br />

year.<br />

The total number of patients treated in 10 countries is<br />

quite sufficient to design <strong>and</strong> conduct prospective clinical<br />

trial. Another very important information coming from the<br />

data is age of the patients treated. Most of the patients (60–<br />

70%) <strong>are</strong> younger than 60 years of age. The median age of<br />

AML patients is 658 . This means that many patients did not<br />

reach these centers, <strong>and</strong> they <strong>are</strong> treated in the local hospitals.<br />

St<strong>and</strong>ard diagnostic facilities <strong>are</strong> sufficient in most centers<br />

for AML. Cytomorphology, immunophenotyping, cytogenetic<br />

<strong>and</strong> molecular technique is m<strong>and</strong>atory according to<br />

ELN criteria9 . The questionnaire delineated that most of the<br />

centers <strong>are</strong> able to perform these st<strong>and</strong>ard diagnostic tools.<br />

In one country cytogenetic is not performing routinely. According<br />

to WHO classification for myeloid neoplasm10 cytogenetic<br />

is important prognostic factor for AML <strong>and</strong> should<br />

be the part of st<strong>and</strong>ard diagnostic test. In the recent years we<br />

<strong>are</strong> faced with the detection of many new genetic alterations<br />

with prognostic impact11–15 . This represents a new diagnostic<br />

challenge for many centers, especially for four countries<br />

without the cryopreservation facilities. The centers in these<br />

countries have no possibilities to keep the samples for the<br />

more dem<strong>and</strong>ing test in AML. The recommendation is to<br />

organize referent laboratory for cryopreservation in a reasonable<br />

period of time.<br />

Induction <strong>and</strong> consolidation therapy <strong>are</strong> cytarabine-based<br />

in combination with anthracycline. The well known regime<br />

»3+7« is the most popular one. Still there is no data showing<br />

better treatment protocol for induction therapy3,16–18 . In elderly,<br />

treatment depends on co-morbidities. Fit elderly patients<br />

received less intensive chemotherapy comp<strong>are</strong>d to<br />

young patients, who predominantly received »3+7 regimen«.<br />

Frail patients have been treated with low-dose cytarabine<br />

or supportive c<strong>are</strong>19 .<br />

Only the minority of younger patients underwent stem<br />

cell transplantation. About 15% of them have been allografted,<br />

<strong>and</strong> 13% received their own marrow as postremission<br />

therapy. The reasons that might explain the small number<br />

of transplanted patients <strong>are</strong> insufficient facilities for<br />

transplants or low number of patients eligible for transplants.<br />

The questionnaire data shows that in some countries<br />

there <strong>are</strong> no sufficient facilities for allografting. At the moment<br />

the centers in two countries <strong>are</strong> not able to perform<br />

allogeneic transplants. In four countries allografting from<br />

matched unrelated donor (MUD) (20) was not performing.<br />

84<br />

In addition total body irradiation (TBI) for conditioning <strong>and</strong><br />

reduced intensity conditioning 21,22 have not been performing<br />

in 3 <strong>and</strong> 4 countries respectively. .<br />

Patient c<strong>are</strong> is not on the expected level for immunocompromised<br />

<strong>and</strong> neutropenic patients. In centers of 4 countries<br />

acute leukemia <strong>are</strong> treating in the room of two or more beds.<br />

It is m<strong>and</strong>atory to state that intensive chemotherapy <strong>and</strong><br />

stem cell transplantation could be performed only in the<br />

centers with 1-bed room.<br />

Conclusions<br />

The questionnaire revealed important data concerning the<br />

diagnosis <strong>and</strong> treatment of AML in centers of Central <strong>and</strong><br />

<strong>East</strong>ern Europe. A large number of patients have been currently<br />

treated in these centers. The number of patients is<br />

quite enough for the future prospective clinical trials. Concerning<br />

the diagnostic tools, st<strong>and</strong>ard approach has been<br />

performing routinely in most centers. Therapy approach is<br />

cytarabine based in combination with anthracycline. Stem<br />

cell transplantation is performing in the majority of Centers<br />

but still the number of transplants is low. The level of st<strong>and</strong>ard<br />

<strong>and</strong> supportive c<strong>are</strong> differs from country to country <strong>and</strong><br />

one of the priorities of CELG group is to st<strong>and</strong>ardize the<br />

diagnostic <strong>and</strong> treatment approaches. This is a prerequisite<br />

for the future clinical trials.<br />

R E F E R E N C E S<br />

1. Dawson B, Trapp RG, ur. Basic & Clinical biostatistics. LangeMedical<br />

Books/ McGraw-Hill 2001; 211–221.<br />

2. Charlson ME, Pompei P, Ales KL, MacKenzie CR. A new method of<br />

classifying prognostic comorbidity in longitudinal studies: development<br />

<strong>and</strong> validation. J Chronic Dis 1987;40:373–383.<br />

3. Löwenberg B, Pabst T, Vellenga E, van Putten W, Schouten HC, Graux<br />

C, et al. Dutch-Belgian Cooperative Trial Group for Hemato-Oncology<br />

(HOVON) <strong>and</strong> Swiss Group for Clinical Cancer Research (SAKK)<br />

Collaborative Group. Cytarabine dose for acute myeloid leukemia. N<br />

Engl J Med 2011; 364:1027–36.<br />

4. Ofran Y, Rowe JM. Induction <strong>and</strong> postremission strategies in acute myeloid<br />

leukemia: what is new? Curr Opin Hematol 2011; 18:83–8.<br />

5. M<strong>and</strong>elli F, Vignetti M, Suciu S, Stasi R, Petti MC, Meloni G, et al.<br />

Daunorubicin versus mitoxantrone versus idarubicin as induction <strong>and</strong><br />

consolidation chemotherapy for adults with acute myeloid leukemia:<br />

the EORTC <strong>and</strong> GIMEMA Groups Study AML-10. J Clin Oncol 2009;<br />

27:5397–403.<br />

6. Holowiecki J, Grosicki S, Robak T, Kyrcz-Krzemien S, Giebel S, Hellmann<br />

A, et al. Polish Adult Leukemia Group (PALG). Addition of<br />

cladribine to daunorubicin <strong>and</strong> cytarabine increases complete remission<br />

rate after a single course of induction treatment in acute myeloid<br />

leukemia. Multicenter, phase III study. Leukemia 2004; 18:989–97.<br />

7. Robak T, Wierzbowska A. Current <strong>and</strong> emerging therapies for acute<br />

myeloid leukemia. Clin Ther 2009; 2:2349–70.<br />

8. Yamamoto JF, Goodman MT. Patterns of leukemia incidence in the<br />

United States by subtype <strong>and</strong> demographic characteristics, 1997–2002.<br />

Cancer Causes Control 2008; 19:379–90.<br />

9. Döhner H, Estey EH, Amadori S, Appelbaum FR, Büchner T, Burnett<br />

AK, et al. Diagnosis <strong>and</strong> management of acute myeloid leukemia in<br />

adults: recommendations from an international expert panel, on behalf<br />

of the European LeukemiaNet. Blood 2010;115:453–474.<br />

10. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit<br />

A, et al. The 2008 revision of the WHO Classification of Myeloid<br />

Neoplasms <strong>and</strong> Acute Leukemia: rationale <strong>and</strong> important changes.<br />

Blood 2009;114(5):937–951.<br />

11. Grimwade D. The clinical significance of cytogenetic abnormalities in<br />

acute myeloid leukaemia. Best Pract Res Clin Haematol 2001;14:<br />

497–529.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) State of the Art<br />

12. Gillil<strong>and</strong> DG, Griffin JD. Role of FLT3 in leukemia. Curr Opin Hematol<br />

2002;9(4):274–81.<br />

13. Schlenk RF, Döhner K, Krauter J, Fröhling S, Corbacioglu A, Bullinger<br />

L, et al. Mutations <strong>and</strong> treatment outcome in cytogenetically normal<br />

acute myeloid leukemia. N Engl J Med 2008;358(18):1909–1918.<br />

14. Green CL, Koo KK, Hills RK, Burnett AK, Linch DC, Gale RE. Prognostic<br />

significance of CEBPA mutations in a large cohort of younger<br />

adult patients with acute myeloid leukemia: impact of double CEBPA<br />

mutations <strong>and</strong> the interaction with FLT3 <strong>and</strong> NPM1 mutations. J Clin<br />

Oncol 2010 ; 28:2739–47.<br />

15. Marchesi F, Annibali O, Cerchiara E, Tirindelli MC, Avvisati G. Cytogenetic<br />

abnormalities in adult non-promyelocytic acute myeloid leukemia:<br />

A concise review. Crit Rev Oncol Hematol. 2010 Epub ahead of<br />

print.<br />

16. Estey E, Döhner H. Acute myeloid leukaemia. Lancet. 2006;368(9550):<br />

1894–1907.<br />

17. Kolitz, JE. Current therapeutic strategies for acute myeloid leukaemia.<br />

Br J Haematol 2006; 134:555.<br />

18. Fern<strong>and</strong>ez HF, Rowe JM. Induction therapy in acute myeloid leukemia:<br />

intensifying <strong>and</strong> targeting the approach. Curr Opin Hematol 2010;17:<br />

79–84.<br />

19. Estey E. Acute myeloid leukemia <strong>and</strong> myelodysplastic syndromes in<br />

older patients. J Clin Oncol 2007; 25:1908–1915.<br />

20. Schlenk RF, Döhner K, Mack S, Stoppel M, Király F, Götze K, et al.<br />

Prospective evaluation of allogeneic hematopoietic stem-cell transplantation<br />

from matched related <strong>and</strong> matched unrelated donors in younger<br />

adults with high-risk acute myeloid leukemia: German-Austrian trial<br />

AMLHD98A. J Clin Oncol 2010; 28:4642–8.<br />

21. Blaise D, Farnault L, Faucher C, Marchetti N, Fürst S, El Cheikh J et al.<br />

Reduced-intensity conditioning with Fludarabin, oral Busulfan, <strong>and</strong><br />

thymoglobulin allows long-term disease control <strong>and</strong> low transplant-related<br />

mortality in patients with hematological malignancies. Exp Hematol<br />

2010; 38:1241–50.<br />

22. Luger SM, Ringdén O, Zhang MJ, Pérez WS, Bishop MR,et al. Similar<br />

outcomes using myeloablative vs reduced-intensity allogeneic transplant<br />

preparative regimens for AML or MDS. Bone Marrow Transplant.<br />

2011 in press.<br />

85


Poster Presentations<br />

PP01 Rapamycin enhances dimethyl sulfoxidemediated<br />

growth arrest in human<br />

myelogenous leukemia cells<br />

Lalic H, Lukinovic-Skudar V, Banfic H, Visnjic D.<br />

Department pt of Physiology & CIBR, School of Medicine,<br />

University of Zagreb<br />

Aim: The pharmacological inhibitors of phosphoinositide<br />

3-kinase (PI3K)/Akt/mammalian target of rapamycin<br />

(mTOR) pathway have been proposed in the treatment of<br />

leukemia based on their antiproliferative effects, but their<br />

possible role in differentiation therapy is less explored. Rapamycin,<br />

an mTOR-inhibitor, has been recently reported to<br />

potentiate all-trans retinoic acid-mediated differentiation of<br />

HL-60 <strong>and</strong> NB4 cell lines along granulocytic pathway, <strong>and</strong><br />

monocytic differentiation of U937 cells induced by vitamin<br />

D3. Dimethyl sulfoxide (DMSO) is a potent inducer of<br />

granulocytic differentiation of myeloid cell lines <strong>and</strong> erythroid<br />

differentiation of K562 cells. The aim of the present<br />

study was to test for the possible synergistic effects of rapamycin<br />

<strong>and</strong> DMSO on growth arrest <strong>and</strong> differentiaton of<br />

human myelogenous leukemia cells.<br />

Materials <strong>and</strong> methods: Myeloblastic (AML-M2) HL-<br />

-60, promyelocytic (AML-M3) NB4, monocytic (AML-<br />

-M5) U937, immature (AML-M6) KG-1, <strong>and</strong> erythro-megakaryocytic<br />

K562 cell lines were maintained in exponential<br />

growth <strong>and</strong> differentiated in the presence of DMSO (0.6 or<br />

1.25%). The number of viable cells was quantified using a<br />

hemocytometer <strong>and</strong> trypan blue exclusion. The expression<br />

of differentiation markers <strong>and</strong> the cell cycle distribution of<br />

propidium iodide-labeled cells were determined by FACS<br />

analyses. Cell morphology was evaluated on May-Grunwald-Giemsa-stained<br />

cytospin preparations. The level of<br />

phosphorylated (Thr389) <strong>and</strong> total p70 S6 Kinase (p70<br />

S6K) in total cell lysates was determined by <strong>West</strong>ern blot<br />

analysis.<br />

Results: <strong>West</strong>ern blot analysis demonstrated that the incubation<br />

of leukemia cells in the presence of 20 nM rapamycin<br />

for 20 min completely reduced the level of Thr389phosphorylated<br />

p70 S6K, which is one of the principle<br />

downstream targets of activated mTOR. When applied at<br />

the same dose for 96 h, rapamycin alone exerted minimal<br />

antiproliferative effects; significant decrease in the number<br />

of viable cells was observed in rapamycin-treated HL-60<br />

<strong>and</strong> KG-1 cells. The presence of 1.25% DMSO for 96 h<br />

significantly inhibited the growth of all cell lines tested, <strong>and</strong><br />

the combination of rapamycin <strong>and</strong> DMSO inhibited the<br />

number of viable cells significantly more than either agent<br />

alone. FACS analysis of propidium iodide-labeled cells revealed<br />

a synergistic effect of rapamycin <strong>and</strong> DMSO on cell<br />

cycle arrest in G0/G1 phase.<br />

Conclusion: Rapamycin potentiates growth-inhibitory<br />

effects of DMSO in the established acute myeloid leukemia<br />

lines. These results suggest that mTOR inhibitors may have<br />

beneficial effects in combination with differentiation agents<br />

in therapy of AML<br />

PP02 Ikaros transcription factors expression<br />

in cell differentiation<br />

Antica M 1 , Paradzik M 1 , Matulic M 2 , Batinic D 3 , Labar B 3 .<br />

1 Division of Molecular Biology, Ru|er Bo{kovi} Institute,<br />

Zagreb, 2 Department of Molecular Biology, Faculty of<br />

Science, Zagreb, 3 University Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb Croatia.<br />

Ikaros family transcription factors <strong>are</strong> involved in lymphocyte<br />

differentiation <strong>and</strong> have a critical role at specific<br />

check points of the haemopoietic pathway. However, how<br />

developmentaly regulated changes <strong>are</strong> reflected in gene expression<br />

programs of lymphocyte differentiation is not well<br />

understood. We study the mechanisms <strong>and</strong> predisposition<br />

for leukemia development by analysing genes involved in<br />

hematopoietic differentiation. Gene targeting studies show<br />

that major players in the commitment of hematopoietic progenitors<br />

<strong>are</strong> Ikaros family transcription factors, <strong>and</strong> inappropriate<br />

expression of these genes might be involved in<br />

leukemogenesis. Accordingly, we analyzed the differential<br />

expression of Ikaros transcription factors <strong>and</strong> their splicing<br />

variants in the most frequent leukemia types, CLL, ALL <strong>and</strong><br />

AML <strong>and</strong> in hematological cell lines. We provide evidence<br />

that splicing variants of these transcription factors occur but<br />

they <strong>are</strong> not restricted to leukemic cells only. Further, the<br />

analysis of Aiolos, showed a clear difference among groups<br />

due to its lower expression in all types of acute leukemia<br />

investigated.<br />

Aims: In this work we analyzed the state of Ikaros family<br />

members in different leukemic cells with the aim to explore<br />

the transcriptional control of human hematopoietic lineages<br />

<strong>and</strong> shed some new light on our underst<strong>and</strong>ing of transcription<br />

factor significance in human leukemia.<br />

Methods: By means of qRT-PCR we investigated the<br />

mRNA expression of Ikaros transcription factors <strong>and</strong> their<br />

splicing variants in leukemia cell lines <strong>and</strong> in 30 samples of<br />

leukemia patients, with AML, ALL or CLL <strong>and</strong> healthy controls.<br />

Results: Ikaros family members <strong>are</strong> expressed as multiple<br />

splice variants <strong>and</strong> proteins arising from alternatively<br />

spliced transcripts may act as dominant negatives. In all of<br />

the cell lines examined Ikaros was present as dominant Ik1<br />

to Ik4 isoforms <strong>and</strong> the small Ik6 isoform was absent. Aiolos<br />

was expressed in the majority of the cell lines, of both, B<br />

<strong>and</strong> T origin, in the form of the full length Aio1. Helios was<br />

87


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

also present only in two long isoforms Hel1 <strong>and</strong> Hel2, <strong>and</strong><br />

was absent in one third of the samples. Although expression<br />

of these short isoforms has been linked to lymphoid <strong>and</strong><br />

myeloid leukemia the quantitative distribution of Ikaros,<br />

Aiolos <strong>and</strong> Helios mRNA expression level in hemopoietic<br />

cells of patients with lymphocytic leukemia, represented by<br />

the most frequent B-CLL <strong>and</strong> ALL (T cell ALL <strong>and</strong> common<br />

ALL), <strong>and</strong> comp<strong>are</strong>d to acute leukemia of myeloid lineage,<br />

AML has not been reported. We found that the relative<br />

quantity of Ikaros <strong>and</strong> Helios mRNA was similar in all subgroups.<br />

Though, analysis of Aiolos, showed a clear difference<br />

among groups due to its lower expression in all types<br />

of acute leukemia investigated.<br />

Conclusion: Our results, associating a lower Aiolos expression<br />

with acute leukemia point to Aiolos role in human<br />

lymphocyte development as it has been previously shown in<br />

mice.<br />

PP03 Multicenter performance evaluation<br />

of the multidrugquant assay kit<br />

Márki-Zay J 1 , Tauber Jakab K 1 , Sipka S 2 , Nagy G 2 ,<br />

Baráth S 2 , Gyimesi E 2 , Hevessy Zs 3 , Sziráki Kiss V 3 ,<br />

Szabó P 5 , Tõkés-Füzesi M 5 , Nagy É 7 , Trucza É 7 ,<br />

Udvardy M 4 , Borbényi Z 8 , Dávid M 6 , Kappelmayer J 3 .<br />

1 Solvo Biotechnology, Szeged, Hungary; 2 Regional<br />

Immunological Laboratory, 3 Department of Clinical<br />

Biochemistry <strong>and</strong> Molecular Pathology, 4 2nd Department<br />

of Internal Medicine, University of Debrecen-Medical <strong>and</strong><br />

Health Science Center, Debrecen, Hungary; 5 Department<br />

of Laboratory Medicine, University of Pécs-Medical<br />

School, 6 1st Department of Internal Medicine, Pécs,<br />

Hungary; 7 Department of Laboratory Medicine, 8 2nd<br />

Department of Medicine <strong>and</strong> Cardiology, University of<br />

Szeged – Albert Szent-Györgyi Clinical Center, Szeged,<br />

Hungary<br />

In the last decade it has become widely accepted that<br />

clinical resistance to chemotherapy correlates with the overexpression<br />

of ATP-binding cassette (ABC) transporters, such<br />

as ABCB1 (MDR1 or P-gp), ABCC1 (MRP1), <strong>and</strong> ABCG2<br />

(MXR or BCRP). There <strong>are</strong> numerous technical approaches<br />

to study multidrug resistance (MDR), which came into the<br />

centre of interest, but the correlating results derived from<br />

different methods. Although the functional methods separately<br />

gave promising results, st<strong>and</strong>ardization <strong>and</strong> reproducibility<br />

of these tests failed to conform to the values required<br />

from routine diagnostic methods. The MultiDrugQuant kit<br />

(MDQ kit) is a flow cytometric method to measure the functional<br />

activity of the three, clinically most relevant efflux<br />

transporters, such as MDR1, MRP1 <strong>and</strong> BCRP in viable<br />

cells. The kit applies fluorescent dyes (substrates) <strong>and</strong> inhibitors<br />

of the all three transporters implicated. After short<br />

incubation of the cell suspension, the MDR activities (MAF<br />

values) of the multidrug transporter were calculated from<br />

the difference between the geo-mean fluorescent intensity<br />

of cells w/o the specific inhibitors, respectively.<br />

Aim: The purpose of the study was the performance evaluation<br />

of the MDQ-kit as routine clinical laboratory flow<br />

cytometric assay.<br />

88<br />

Patients <strong>and</strong> methods: Performance evaluation of the<br />

MDQ kit was carried out to fulfil the requirements of the<br />

EU IVD directive (98/79/EC) <strong>and</strong> the harmonised European<br />

norms. Laboratory validation of the kit was performed in<br />

three university centres in Hungary according to the st<strong>and</strong>ards<br />

of the Clinical Laboratory St<strong>and</strong>ards Institute. Inaccuracy<br />

<strong>and</strong> comparative measurements were carried out using<br />

cell lines with low <strong>and</strong> high activity of transporters.<br />

Mononuclear cells from healthy donors were separated using<br />

Ficoll gradient <strong>and</strong> results on different flow cytometers<br />

were comp<strong>are</strong>d using CD45 or CD19 or CD3 monoclonal<br />

antibodies for gating the population of interest using a harmonized<br />

procedure. The reference interval of MDR activities<br />

in lymphocytes has been determined in a population of<br />

120 healthy volunteers.<br />

Results: The assay proved to be specific <strong>and</strong> robust at<br />

various concentrations of the fluorescent dyes (10–100% of<br />

the original) or inhibitors (50–150% of the original). Both<br />

intra-assay <strong>and</strong> inter-assay reproducibilities were < 24.2;<br />

MAF-MRP < 13.2; MAF-BCRP < 23.2.<br />

Conclusion: This functional assay is reliable <strong>and</strong> provides<br />

transporter specific quantitative results on the function<br />

of the MDR1, MRP1 <strong>and</strong> BCRP transporters in the<br />

target cells. Recently, clinical trials have been started in haematological<br />

diseases to evaluate the predictive <strong>and</strong> prognostic<br />

values of these biomarkers.<br />

PP04 NPM1 mutations in AML detected by<br />

fragment analysis <strong>and</strong> Sanger sequencing<br />

Crncec I, Musani V, Marusic Vrsalovic M, Livun A,<br />

Pejsa V, Jaksic O, Haris V, Ajdukovic R, Stoos Veic T,<br />

Kusec R. Departments of Hematology, Divison of<br />

molecular diagnostics <strong>and</strong> genetics <strong>and</strong> Cytology,<br />

Universtiy hospital Dubrava <strong>and</strong> Zagreb School of<br />

Medicine, Division of molecular medicine, Institute Rudjer<br />

Boskovic, Zagreb, Croatia<br />

NPM1gene mutations can be detected in approximately<br />

1/3 of all de novo AMLs particularly in cases with normal<br />

karyotype <strong>and</strong> predicts good prognosis. However, associated<br />

with FLT3-ITD is of adverse prognosis<br />

Aim: We have developed a two-step molecular assay for<br />

the detection of NPM1 mutations.<br />

Patients <strong>and</strong> methods: The first part, prescreening for<br />

mutation, consists of RT-PCR amplification of exon 12 of<br />

the gene with the subsequent gene scan (fragment analysis)<br />

by capillary electrophoresis in AB310 Sequencer analyzer.<br />

What follows is re-amplification of mutation-positive cases<br />

<strong>and</strong> direct sequencing for the determination of the precise<br />

type of mutation. We tested 60 patients with de novo (46)<br />

<strong>and</strong> secondary (14) AMLs from our institution. The karyotype<br />

was available for 32% of them <strong>and</strong> FLT3-ITD analysis<br />

was done in all.<br />

Results: NMP1 mutations were found in 12 patients with<br />

de novo AML (26% of de novo AML; 20% for all AMLs).<br />

None was from the sAML group. Sequencing revealed 8<br />

cases of A-type (TCTG), 2 of D-type (CCTG) <strong>and</strong> 1 each of<br />

H-type (CTTG) <strong>and</strong> Nm-type (CCAG) mutation. Expected<br />

frequencies <strong>are</strong> given in the table.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

Mutation type N % (95% CI) Expected %<br />

A 8 67 (35-90) 70-80<br />

D 2 17 (2-48) approx. 6, < 10<br />

H 1 8 (0-27) approx. 2<br />

Nm 1 8 (0-27) approx. 0,4, < 1<br />

Σ 12 - -<br />

There was also a significant association of NPM1 <strong>and</strong><br />

FLT3-ITD mutation (H 2 =6,322; DF=1; P=0,012).<br />

Conclusion: In conclusion, the two-step molecular assay<br />

for NPM1 mutation detection is rational <strong>and</strong> acceptable<br />

method for the routine diagnostic application. Frequencies<br />

of NPM1 mutations in our patients <strong>are</strong> comparable to the<br />

ones reported previously, with type A being the most frequent.<br />

However, although in the small number of patients,<br />

we have observed increased occurrence of relatively r<strong>are</strong><br />

molecular types. We can also confirm that there is an increased<br />

association of FLT3-ITD with NPM1 mutation.<br />

PP05 NPM1 mutation A <strong>and</strong> FLT3/ITD in AML<br />

with myelodysplasia-related changes<br />

Radic Antolic M, Ries S, Zadro R, Davidovic S, Labar B.<br />

Clinical Hospital Center <strong>and</strong> School of Medicine,<br />

University of Zagreb, Croatia<br />

According to WHO criteria, acute myeloid leukemia<br />

(AML) with myelodysplasia-related changes requires the<br />

presence of at least 20% blasts in the bone marrow <strong>and</strong>/or<br />

blood <strong>and</strong> morphologic features of dysplasia in two or more<br />

mature bone marrow cell lines. Chromosomal abnormalities<br />

may be identical to those in myelodysplasia, with either<br />

complex or normal karyotypes. In patients with normal<br />

karyotype, it is important to detect mutations in NPM1 <strong>and</strong><br />

FLT3 gene as a predictor of clinical outcome.<br />

The aim of this study was to determine the prevalence of<br />

NPM1 mutation A <strong>and</strong> FLT3/ITD <strong>and</strong> to relate them to cytogenetic<br />

abnormalities in patients with AML with myelodysplasia-related<br />

changes.<br />

The study included 52 patients (23 female <strong>and</strong> 29 male)<br />

with de novo AML with myelodysplasia-related changes.<br />

Bone marrow aspirate at diagnosis was used for cytogenetic<br />

<strong>and</strong> molecular analysis. GTG-b<strong>and</strong>ing was used for karyotype<br />

analysis <strong>and</strong> FISH was used for detection of del(5q),<br />

del(5), del(7q), del(7) <strong>and</strong> rearrangement of the MLL gene.<br />

RQ-PCR was used to determine NPM1 mutation A (Ipsogen,<br />

France) <strong>and</strong> FLT3/ITD was determined by RT-PCR<br />

analysis (Nakao et al, Leukemia 1996).<br />

Among 52 patients included in the study, 29 (56%) had<br />

normal karyotype, 9 (17%) had intermediate, <strong>and</strong> 12 (23%)<br />

adverse cytogenetic risk, while cytogenetic analysis was not<br />

performed for two patients (4%). FLT3/ITD was positive in<br />

7 (13%) patients: 6 of them were with normal karyotype <strong>and</strong><br />

one had adverse cytogenetic risk. NPM1 mutation A was<br />

positive in 9 (17%) patients; 6 of them had normal karyotype,<br />

2 had intermediate <strong>and</strong> one patient adverse cytogenetic<br />

risk. Three patients (6%) with NPM1 mutation A <strong>and</strong><br />

FLT3/ITD had normal karyotype. Although patients with<br />

normal karyotype had higher incidence of NPM1 mutation<br />

A <strong>and</strong> FLT3/ITD than patients with intermediate <strong>and</strong> adverse<br />

cytogenetic risk, the difference was not statistically<br />

significant. There was no difference in patient distribution<br />

in cytogenetic risk groups according to gender. In normal<br />

karyotype patients group, higher incidence of NPM1 mutation<br />

A <strong>and</strong> FLT3/ITD was observed in female patients (4/14)<br />

than in male patients (2/15) but without statistical significance.<br />

AML with myelodysplasia-related changes is a distinctive<br />

myeloid disorder which requires multidisciplinary approach<br />

in diagnostics. Cytogenetic abnormalities have the<br />

most important role in predicting outcome, but in patients<br />

with normal karyotype only small gene mutations (NPM1,<br />

FLT3) can help in risk categorization. In future, those mutations<br />

could be real predicting factors correlating with therapy<br />

response <strong>and</strong> clinical outcome.<br />

PP06 Prognostic Signifi cance of CD56 Antigen<br />

Expression in Patients with Acute Myeloid<br />

Leukemia<br />

Djunic I, Virijevic M, Djurasinovic V, Novkovic A,<br />

Colovic N, Kraguljac-Kurtovic N, Vidovic A,<br />

Suvajdzic-Vukovic N, Tomin D. Clinic of hematology,<br />

Clinical Center Serbia, Belgrade, Serbia<br />

Aim: CD56 antigen, identified as an isoform of the neural<br />

adhesion molecules, has been found to be associated<br />

with poor prognosis in several hematologic malignancies,<br />

including acute myeloid leukemias (AML). The aims of this<br />

study were to investigate frequency <strong>and</strong> prognostic relevance<br />

of CD56 expression in patients with AML, as well as<br />

to comp<strong>are</strong> the importance of CD56 expression with st<strong>and</strong>ard<br />

prognostic factor such <strong>are</strong> age, cytogenetic abnormalities<br />

<strong>and</strong> performance status.<br />

Patients <strong>and</strong> methods: In this single-center study we<br />

analyzed the data of 184 newly diagnosed patients with<br />

nonpromyelocytic AML with follow-up of 36 months. As<br />

the risk factors for overall survival (OS), rate of achievement<br />

CR <strong>and</strong> duration of CR, were estimated: age, ECOG<br />

PS, FAB subtype AML, ELN cytogenetic risk group <strong>and</strong><br />

CD56 expression on blasts cells. Performance status (PS)<br />

was evaluated by <strong>East</strong>ern Cooperative Oncology Group<br />

(ECOG), ranged 0–4 (=2). AML subtypes were classified<br />

by French-American-British (FAB) Cooperative Group.<br />

Cytogenetic risk groups were assessed by recommendation<br />

of European LeukemiaNet (ELN): favorable, intermediate-<br />

I, intermediate-II <strong>and</strong> adverse group. For CD56 antigen expression,<br />

detected by flow-cytometry, a cut off >=20% was<br />

used as positive (CD56+). Patients were treated with Medical<br />

Research Council (MRC) 12 regimen. Risk factors were<br />

identified using the univariate <strong>and</strong> multivariate analysis.<br />

Results: The median patients’ age was 58 years, range<br />

18–79. CD56+ antigen was recorded in 40 patients (21.7%).<br />

Significant risk factors for poor OS in univariate analysis<br />

were: age >=55 years (p=2 (p=0.001), adverse cytogenetic<br />

(p=0.019) <strong>and</strong> CD56+ (p=0.003). The multivariate analysis<br />

indicated that CD56+ was the most significant risk factor<br />

89


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

for OS: p=0.05, relative risk (RR)=1.598 (95% CI 0.984-<br />

2.595). The most significant factor for poor rate of CR was<br />

age >=55 years: p=0.001, RR=0,274 (95% CI 0.125-0.597).<br />

CD56 positivity had not significant influence on CR rate,<br />

but it was the most significant risk factor for shorter duration<br />

of CR: p=0.005, RR=0.443 (95% CI 0.253-0.779). According<br />

to FAB subtypes of AML, CD56 positivity had the<br />

most significant impact on patients’ outcome in M5 subtype.<br />

In those patients CD56+ was the most significant risk<br />

factor for poor OS (p=0.021, RR=2.981, 95% CI 1.174-<br />

7.122) <strong>and</strong> shorter duration of CR (p=0.03, RR=3.563, 95%<br />

CI 1.132-11.215.<br />

Conclusion: Our data underline the independent negative<br />

prognostic role of CD56 antigen expression in AML,<br />

particularly in M5 subtype. Its presence should be regularly<br />

estimate for a better prognostic assessment of AML patients.<br />

PP07 FLT3 internal t<strong>and</strong>em duplication is a poor<br />

risk factor in patients with AML with<br />

diverse cytogenetic features<br />

M. Mikulic, R. Zadro, S. Davidovic, R. Serventi Seiwerth,<br />

D. Sertic, D. Nemet, J. Batinic, D. Batinic, K. Gjadrov,<br />

S. Ries, B. Labar. Department of Medicine <strong>and</strong><br />

Department of Clinical Laboratory, Clinical Hospital<br />

Center <strong>and</strong> School of Medicine, University of Zagreb,<br />

Croatia<br />

Aim: Mutations of Fms-like tyrosine kinase 3 (FLT3) <strong>are</strong><br />

present in about 30% of acute myelogenous leukemia<br />

(AML) patients that have been shown to have poor prognosis.<br />

The aim of this study was to analyze the prognostic impact<br />

of FLT3 internal t<strong>and</strong>em duplication (ITD) on the outcome<br />

of patients diagnosed with AML in comparison with<br />

patients with wild-type (WT) FLT3.<br />

Patients <strong>and</strong> methods: We retrospectively analyzed the<br />

laboratory <strong>and</strong> clinical data in 78 newly diagnosed patients<br />

with AML who have been treated in our Center from<br />

02/2000-03/2010. Median patients’ age was 45 years (range,<br />

22–72 yrs.). AML diagnosis was based on morphological,<br />

immunophenotypical <strong>and</strong> cytogenetical features. The presence<br />

of FLT3 ITD was detected by RT-PCR analysis. All<br />

patients received intensive induction chemotherapy followed<br />

by consolidation chemotherapy; 36 patients were<br />

treated with autologous hematopoietic stem cell transplantation<br />

(SCT) in 1 st CR, while 8 <strong>and</strong> 3 patients were allografted<br />

in 1 st <strong>and</strong> >1 st CR, respectively.<br />

Results: FLT3-ITD was detected in 20 patients (25%).<br />

Eight FLT3-ITD patients had normal karyotypes, two had<br />

good, <strong>and</strong> two had intermediate cytogenetic features. Thirteen<br />

FLT3-WT patients had normal karyotypes, five had<br />

good, nine had poor, <strong>and</strong> nine had intermediate cytogenetic<br />

features. In 30 patients from both groups cytogenetic studies<br />

had failed. CR rate was 85% in FLT3-ITD patients <strong>and</strong><br />

71% in FLT3-WT patients (p=0.249). 2-year DFS was 33.8<br />

<strong>and</strong> 52.5% in the FLT3-ITD <strong>and</strong> FLT3-WT group, respectively<br />

(p=0.029). 2-year OS was 47.3 <strong>and</strong> 63% in the FLT3-<br />

ITD <strong>and</strong> FLT3-WT group, respectively (p=0.665).<br />

90<br />

Conclusion: A higher remission rate after induction, although<br />

not statistically significant, was found in the FLT3-<br />

ITD group. Contrary to that, the DFS was significantly better<br />

in patients with FLT3-WT <strong>and</strong> a trend for better OS was<br />

observed in patients with FLT3-WT. This data support the<br />

evidence for FLT3-ITD being a poor risk factor, not only in<br />

patients with normal karyotypes, but also in patients with<br />

different cytogenetic risk factors.<br />

PP08 A Philadelphia-negative Chronic Myeloid<br />

Leukemia with a BCR/ABL Fusion Gene<br />

Lasan Trcic R 1 , Kardum I 2 , Jaksic B 2 , Jaksic O 3 , Kusec R 3 ,<br />

Pejsa V 3 , Zadro R 4 , Batinic D 4 , Labar B 5 , Begovic D 1 .<br />

1 Division of Medical Genetics, Department of Paediatrics<br />

Univesity Hospital Center Zagreb, Zagreb, Croatia.<br />

2 Division of Hematology, Department of Internal Medicine,<br />

Clinical Hospital Merkur, Zagreb, Croatia. 3 Division of<br />

Hematology Departmrnt of Internal Medicine, Clinical<br />

Hospital Dubrava, Zagreb, Croatia. 4 Clinical Laboratory<br />

Diagnostics, University Hospital Center Zagreb, Zagreb,<br />

Croatia. 5 Division of Hematology, Department of Internal<br />

Medicine Univesity Hospital Center Zagreb, Zagreb,<br />

Croatia<br />

Aim: Objective. The causing factor in chronic myeloid<br />

leukemia (CML) is the BCR/ABL chimeric gene. In most<br />

cases, it is cytogeneticlly visualized as a translocation between<br />

chromosomes 9 <strong>and</strong> 22 as the Philadelphia (Ph) chromosome.<br />

Five to ten percent of patients lack cytogenetics<br />

evidence of the Ph chromosome (CML-Ph) but show BCR/<br />

ABL fusion by fluorescence in situ hybridization (FISH) or<br />

reverse transcriptase-polymerase chain reaction (RT-PCR).<br />

Patients <strong>and</strong> methods: Methods. Diagnostic bone marrow<br />

samples were diagnosed by morphological, immunophenotypical<br />

criteria, <strong>and</strong> reverse transcriptase-polymerase<br />

chain reaction. Chromosome studies were performed on<br />

GTG-b<strong>and</strong>ed chromosomes obtained from bone marrow after<br />

direct preparation or short-time cultures. FISH studies<br />

were done using BCR/ABL1 dual-color/dual_fusion probes<br />

(Kreatech, Vysys)<br />

Results: Among 231 adult patients with CML, 10%<br />

(n=23) had variant translocations (9;22;v), involving one or<br />

two additional translocation partner chromosome. Two cases<br />

(0.8% in total) had an app<strong>are</strong>ntly normal karyotype. In<br />

first case, 56-year-old female diagnosed with CML, conventional<br />

cytogenetic analysis shoved a normal karyotype.<br />

Methaphase FISH showed an insertion of BCR sequences<br />

onto a chromosome 9 within ABL, the karyotype was<br />

46,XX. ish ins(9;22)(q34;q11.2). Second case, 58-year-old<br />

female with clinical findings consistent with CML, by conventionl<br />

cytogenetic analysis showed app<strong>are</strong>ntly normal<br />

karyotype. Findings with methapase FISH were interpreted<br />

as an insertion of the ABL gene onto chromosome 22 with<br />

BCR, the karyotype was 46,XX. ish ins(22;9)(q11.2;q34).<br />

The RT-PCR provided molecular evidence that a typical<br />

CML chimeric product resulting from a fusion of BCR exon<br />

with ABL exon is present in both cases.<br />

Conclusion: The clinical significance Ph-CML with app<strong>are</strong>ntly<br />

normal karyotype, with 5’BCR/3’ABL fusion gene


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

located on 9q34 <strong>and</strong> 22q11.2 is unclear. Some studies have<br />

suggested that these cryptic translocationes have no impact<br />

on prognosis, while others have suggested an adverse prognosis,<br />

it is more likely that the prognosis in Ph- is linked to<br />

the presence or absence of associated deletion on the der(9)<br />

rather than positioning of the fusion gene alone.<br />

PP09 Croatian CML Registry based on European<br />

LeukemiaNet CML Registry<br />

Sertic D 1 , Peric Z 1 , Sincic-Petricevic J 2 , Lozic D 3 ,<br />

M<strong>and</strong>ac-Rogulj I 4 , Duletic Nacinovic A 5 ,<br />

Gveric-Krecak V 6 , Corovic-Arneri E 7 , Pejsa V 8 ,<br />

Suvic-Krizanic V 9 , Skunca Z 10 , Babok-Flegaric R 11 ,<br />

Krmek-Zupanic D 12 , Carzavec D 13 , Lazic-Prodan V 14 ,<br />

Coha B 15 , Ajdukovic R 8 , Romic I 7 , Zadro R 1 , Davidovic S 1 ,<br />

Lasan-Trcic R 1 , Labar B 1 . Croatian Cooperative Group for<br />

Hematologic Diseases-CML Group: 1 University Hospital<br />

Center Zagreb, 2 University Hospital Center Osijek,<br />

3 University Hospital Center Split, 4 Clinical Hospital<br />

Merkur Zagreb, 5 University Hospital Center Rijeka,<br />

6 General Hospital [ibenik, 7 General Hospital Dubrovnik,<br />

8 Clinical Hospital Dubrava-Zagreb, 9 General Hospital<br />

Sisak, 10 General Hospital Zadar, 11 General Hospital<br />

Vara`din, 12 General Hospital Sv.Duh-Zagreb, 13 Clinical<br />

Hospital Center Sestre milosrdnice-Zagreb, 14 General<br />

Hospital Pula, 15 General Hospital Slavonski Brod<br />

Background: Based on European Leukemianet (ELN)<br />

Registry for Philadelphia positive chronic myeloid leukemia<br />

(CML) we collected data on CML epidemiology, the<br />

disease characteristics <strong>and</strong> treatment, as well as the data<br />

concerning the monitoring <strong>and</strong> clinical outcome of CML in<br />

Croatia. Here we present the preliminary data of CML Registry<br />

in Croatia.<br />

Methods: Data were prospectively collected from November<br />

15, 2009 until June 30, 2011. Data was sent to the<br />

national coordinator in the University Hospital Center Zagreb<br />

from all the hematologists in Croatia. All patients<br />

signed informed consent prior to the data reporting in the<br />

Registry.<br />

Results: From November 15, 2009 until June 30, 2011,<br />

69 patients with CML were registered. For 66 patients data<br />

were collected in CML Registry. Median age was 54.6 years<br />

(range 17–87). There were more male than female patients<br />

(63.6% versus, 36.4%). At time of diagnosis all were in<br />

chronic phase of CML. The Euro Score risk group distribution<br />

was: 36.4% low, 51.5% intermediate <strong>and</strong> 12.1% high<br />

risk; the Sokal risk distribution showed: 36.4% low, 30.3%<br />

intermediate <strong>and</strong> 30.3% high risk The majority of the patients<br />

showed a WHO performance status score of 0 (54.5%)<br />

or 1 (37.9%). FISH for bcr-abl was positive in 100% of<br />

cases <strong>and</strong> 51 (77.3%) of them were diagnosed by conventional<br />

cytogenetic (G-b<strong>and</strong>ing). Molecular Diagnostics (RT-<br />

PCR, quantitative PCR or both) was performed in 48<br />

(72.8%) patients Out of 27 patients who were committed<br />

quantitative-PCR at diagnosis, in 19 of them it is expressed<br />

by the international score. In relation to the population<br />

based approach preliminary data showed a higher frequency<br />

of newly diagnosed CML in two regions (Sisak <strong>and</strong> Sibenik).<br />

Summary: According to data collected so far the incidence<br />

of CML in Croatia is 0.99 / 100 000 inhabitants.<br />

There is uneven incidence distribution of CML by region,<br />

but due to the small number of patients we need a longer<br />

follow-up. Participation in ELN Registry study clearly improved<br />

the diagnostic possibilities (use of quantitative PCR<br />

for monitoring of CML). We also implemented for most patients<br />

ELN recommendations for treatment response with<br />

TKI.<br />

PP10 Use of Imatinib mesylate in Chronic Phase<br />

CML in Clinical Practice – Our Experience<br />

Miljkovic E, Markovic D, Cojbasic I, Nikolic V,<br />

Marjanovic G, Govedarovic N, Macukanovic-Golubovic L,<br />

Vucic M, Pavlovic M, Vukicevic T, Tijanic I, Simonovic O,<br />

Clinic of Hematology, Clinical Center Nis, Serbia<br />

Chronic myelogenous leukemia (CML) is a pluripotential<br />

stem cell disease characterized by anemia, extreme<br />

blood granulocytosis <strong>and</strong> granulocytic immaturity, basophilia,<br />

often thrombocytosis, <strong>and</strong> splenomegaly.<br />

Hallmark of chronic myeloid leukemia is Philadelphia<br />

chromosome that results from a reciprocal translocation between<br />

chromosomes 9 <strong>and</strong> 22.<br />

T (9;22)(q34;11) creates an aberrant fusion gene derived<br />

from the ABL gene encoded on chromosome 9 <strong>and</strong> the BCR<br />

gene encoded on chromosome 22.The product of this fusion<br />

BCR-ABL gene is a constitutively active protein-tyrosine<br />

kinase that promotes cellular proliferation <strong>and</strong> suppresses<br />

apoptosis.<br />

The incidence of CML increases with age, with a peak<br />

incidence of 53 years. Men <strong>are</strong> affected more often than<br />

women (3:2).<br />

CML has a tri-phasic clinical course: chronic phase, accelerated<br />

phase <strong>and</strong> blastic phase (myeloid or lymphatic)<br />

defines the transformation of CML in an acute leukemia <strong>and</strong><br />

is difficult to treat. Myeloid transformation of CML is more<br />

common (70%) than lymphoid blastic crisis (30%).<br />

Inevitable progression to blast crisis used to occur after a<br />

median of 3.5 years prior to the development of Bcr Abltargeted<br />

therapie.<br />

The Abl tyrosine kinase inhibitor imatinib (IM) induces<br />

durable responses in a high proportion of patients with<br />

chronic phase chronic myeloid leukemia <strong>and</strong> therefore represents<br />

the current st<strong>and</strong>ard first line treatment.<br />

Aim: Aim was to evaluate hematological <strong>and</strong> cytogenetic<br />

response in patients treating with IM longer then 6 months.<br />

Second aim was to evaluate molecular response in patients<br />

treating with IM longer then 24 months.<br />

Methods: Patients were monitored for hematologic <strong>and</strong><br />

cytogenetc reponse at 6 months intervals. Complete hematologic<br />

response is defined by WBC count


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

ter short-term culture (24 or 48 hours or both) with st<strong>and</strong>ard<br />

G or Q b<strong>and</strong>ing techniques.<br />

Achievement of a complete molecular response (CCyR)<br />

was assessed with the use of a quantitative reverse-transcriptase–polymerase-chain-reaction<br />

(RT-PCR) assay.<br />

Results: 38 patients, 21 men <strong>and</strong> 17 women were<br />

analised. Median age of patients was 46.6 years (18–73).<br />

After 32.6 months of median follow up (6–80), complete<br />

hematologic <strong>and</strong> cytogenetic response (mitosis with no<br />

Ph+) was achieved in 79.0 % patients (30),complete hematologic<br />

<strong>and</strong> no cytogenetic response (>95% mitosis Ph+) in<br />

2.6% (1). 15.8% of patients (6) had resistance to imatinib<br />

mesylate <strong>and</strong> were switched to other therapeutical options,<br />

two of them with additional chromosomal abnormalities.<br />

(one with 2 Ph+,<strong>and</strong> one with trisomy 8 <strong>and</strong> trisomy 22<br />

chromosomes). 1 patient (2.6%) had a severe adverse effect<br />

after 6 months with IM therapy-pancytopenia, mostly<br />

thrombocytopenia <strong>and</strong> the drug was interrupted.<br />

Achievement of CCyR was assessed with (RT-PCR) assay<br />

in patients treating with IM longer then 24 months. 15<br />

patients were tested for molecular response. A major molecular<br />

response was defined as a BCR-ABL transcript level<br />

of 0.1% or lower on the International Scale (conversion factor<br />

of 0.81), corresponding to a reduction in the BCR-ABL<br />

transcript level by at least 3 log from the st<strong>and</strong>ardized baseline<br />

level. Complete molecular response achieved 86.7% of<br />

patients (13) <strong>and</strong> 13.3% (2) had no molecular response.<br />

Conclusions: Chronic myeloid leukemia has substantially<br />

improved survival with the application of selective<br />

ABL tyrosine kinase inhibitor imatinib mesylate as st<strong>and</strong>rad<br />

treatment of chronic phase CML, with very good quality of<br />

life <strong>and</strong> few adverse effects.<br />

PP11 Imatinib for CML as a frontline therapy<br />

– Zagreb Experience<br />

Sertic D 1 , Zadro R 2 , Nemet D 1 , Davidovic S 3 ,<br />

Lasan-Trcic R 3 , Radic Antolic M 2 , Horvat I 2 ,<br />

Frani} Simic I 3 , Roncevic P 1 , Radman I 1 , Basic-Kinda S 1 ,<br />

Aurer I 1 , Labar B 1 . 1 Division of Hematolofy Department of<br />

Medicine, 2 Department of Clinical Laboratory,<br />

3 Department of Pediatric, Clinical Hospitral Center <strong>and</strong><br />

School od Medicine, University of Zagreb, Croatia<br />

Background: Imatinib is frontline therapy for chronic<br />

phase Philadelphia positive chronic myeloid leukemia (Ph+<br />

CML). In about 70% of patients optimal treatment response<br />

could be achieved <strong>and</strong> sustained for more than 7 years of<br />

therapy.<br />

We present the experience of the imatinib frontline treatment<br />

for CML in University Hospital Center Zagreb, Department<br />

of Hematology.<br />

Patients <strong>and</strong> Methods: A total of sixty seven patients<br />

with Ph+CML were treated with first line imatinib, from<br />

February 2003. Here we evaluated fifty eight of them, who<br />

were eligible for twelve months follow up. Median followup<br />

time was fifty two months (range 12–103, months).<br />

There were males <strong>and</strong> females, with a median age of forty<br />

nine years (range 14–74, years). Patients were treated with<br />

92<br />

400 mg/day. They were regularly checked for treatment response<br />

every 3 months with cytogenetic methods (G-b<strong>and</strong>ing<br />

<strong>and</strong> FISH) <strong>and</strong> quantitative RT-PCR.<br />

Results: In forty one patients (70%) complete cytogenetic<br />

response (CCR) was documented. Out of those forty<br />

one patients twelve patients (20%) achieved a complete molecular<br />

response (CMR). Twenty three patients (40%)<br />

achieved a major molecular response (MMR), while six patients<br />

(10%) had less than MMR. Two patients (3.4%) were<br />

resistant to first line treatment <strong>and</strong> were subsequently treated<br />

with nilotinib. Another two patients (3.4%) progressed to<br />

acute lymphoblastic leukemia, both after 2.5 months of therapy<br />

with imatinib. One of them had a T315I mutation, <strong>and</strong><br />

was subsequently treated with MUD allogeneic stem cell<br />

transplantation. One patient progressed to acute myeloid<br />

leukemia. Another patient had a progression of the disease<br />

during the chronic phase, with a verified T315I mutation.<br />

He was treated with allogeneic stem cell transplantation.<br />

Eight patients (13%) died during the course of the treatment,<br />

four of which related to CML therapy or progression<br />

of the disease. In four patients imatinib dose was increased<br />

to 600 mg or 800 mg. Subsequent optimal response was<br />

documented in all four patients. Two patients (3%) had serious<br />

side effects to the treatment <strong>and</strong> were therefore switched<br />

to nilotinib.<br />

Conclusion: Optimal response with imatinib is reported<br />

in 41 patients. Progression of the disease to accelerated/<br />

blastic phase was found in 3 patients. Eight patients died,<br />

four because of CML. Our experience has proved also that<br />

imatinib is an effective frontline therapy for Ph+CML.<br />

PP12 Frequency of 4 bcr-abl1 kinase domain<br />

mutations in chronic myeloid leukemia<br />

patients resistant to imatinib mesylate<br />

therapy<br />

Horvat I 1 , Radic Antolic M 1 , Zadro R 1 , Sertic D 2 , Labar B 2 .<br />

1 Department of Clinical Laboratory, 2 Department of<br />

Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

Introduction of imatinib mesylate (IM) for treatment of<br />

chronic myeloid leukemia (CML) has brought revolution in<br />

patient survival. Most patients in chronic phase of disease<br />

have high rates of responses to IM with the optimum response<br />

understood as major molecular response <strong>and</strong> bcrabl1<br />

transcript level ≤ 0.1% according to International Scale<br />

(IS). A proportion of patients become resistant to the drug<br />

after some time. Mutations in bcr-abl1 kinase domain <strong>are</strong><br />

found in 50% of resistant patients <strong>and</strong> IM cannot bind to its<br />

target.<br />

The aim of this study was to determine the frequency of<br />

4 bcr-abl1 kinase domain mutations (T315I, E255K, E255V<br />

<strong>and</strong> Y253H) in a group of CML patients who became resistant<br />

to IM therapy, <strong>and</strong> to show the percentage of bcr-abl1<br />

transcript levels in patients with <strong>and</strong> without mutations. The<br />

study included 28 CML patients, 27/28 were monitored during<br />

the course of IM therapy by real time quantitative PCR<br />

(Ipsogen, France) <strong>and</strong> their bcr-abl1 transcript values were


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

expressed according to IS. ASO-PCR for detection of T315I,<br />

E255K, E255V <strong>and</strong> Y253H mutations was performed according<br />

to Kang et al. (Haematologica, 2006).<br />

Among 28 patients tested, T315I was detected in 9 patients<br />

(32.1%), E255K in 3 patients (10.7%), E255V in 2<br />

patients (7.1%) <strong>and</strong> Y253H in 6 patients (21.4%). Overall,<br />

14/28 patients (50.0%) had one or more mutations. In T315I,<br />

E255K, E255V <strong>and</strong> Y253H positive patients, bcr-abl1 transcript<br />

levels were 7.7–39.1% IS (median 23.2%), 7.7–12.7%<br />

(median 12.3%), 1.7–12.7% (median 7.2%) <strong>and</strong> 12.7–39.1%<br />

(median 18.0%), respectively. In 14 patients tested negative<br />

for analyzed mutations, bcr-abl1 transcript levels were<br />

0.55–46.3% IS (median 12.3%). In 4 T315I positive patients<br />

one or more additional mutations were detected; one patient<br />

accumulated all four mutations over time (T315I, E255K,<br />

E255V, Y253H), 2 patients gained Y253H, <strong>and</strong> E255K mutation<br />

was detected in 1 patient. Among 4 patients who died,<br />

3 were T315I positive <strong>and</strong> 2 of them had additional mutations.<br />

Mutations in bcr-abl1 kinase domain <strong>are</strong> to a large extent<br />

responsible for resistance to IM therapy. We have shown<br />

that the monitoring of bcr-abl1 transcript level is important<br />

for detecting resistance to IM <strong>and</strong> that mutations <strong>are</strong> found<br />

in a high proportion of resistant patients. Searching for mutations<br />

in patient who lost response to IM therapy is significant<br />

part of CML patient management as the result of mutation<br />

analysis directly affects therapy approach, particularly<br />

in T315I positive patients for whom the treatment of choice<br />

is stem-cell transplantation.<br />

PP13 Imatinib mesylate in Patients with CML<br />

Relapse after Allogeneic Transplantation<br />

Serventi Seiwerth R 1 , Sertic D 1 , Radic Antolic M 2 ,<br />

Mrsic M 1 , Zadro R 2 , Davidovic S 3 , Grubic Z 2 , Mikulic M 1 ,<br />

Labar B 1 . 1 Department of Medicine, 2 Department of<br />

Clinical Laboratory, 3 Department of Pediatric, Clinical<br />

Hospital Center <strong>and</strong> School of Medicine, University of<br />

Zagreb, Croatia<br />

Intronduction: Allogenic stem cell transplantation is a<br />

curable therapy for substantial number of patients with Ph+<br />

CML. Because of high treatment related toxicity <strong>and</strong> mortality<br />

in the current era of TKI terapy allografting is not any<br />

more 1st line treatment. CML relapse after allogeneic transplantation<br />

is still a problem in 20% of patients. For these<br />

patient therapy with tyrosine kinase inhibitors (TKI) should<br />

be an treatment option. Here we present the outcome of<br />

three patients in realpse after allografting <strong>and</strong> treated with<br />

TKI.<br />

Patients <strong>and</strong> methods: Three patients, one male <strong>and</strong> two<br />

females with Ph+ CML in chronic phase recived bone marrow<br />

transplant from their HLA identical siblings after myeloablative<br />

conditioning with busulfan <strong>and</strong> cyclophosphamide.<br />

For GvHD prophylaxis cyclosporin <strong>and</strong> short methotrexate<br />

was given. In all patients sustained three lineage<br />

engraftmnet occured. Two of them experienced acute GvHD<br />

of gastrointestinal tract gr. II <strong>and</strong> skin gr. II respectively,<br />

which responded to corticosteroides. The relapse of CML<br />

was documented on 132, 168 <strong>and</strong> 30 months after allotransplantation<br />

Two of them experienced molecular relapse; one<br />

recieved donor lymphocyte infusion in escalating doses of<br />

CD3+ cells, but without any effect. Third patient had hematologycal<br />

relapse <strong>and</strong> was treated with hydroxyurea for two<br />

months.All patients received imatinib mesylate 400 mg<br />

daily dose within the 3 months of relapse. Two out of three<br />

patients achieved complete molecular response 10 <strong>and</strong> 12<br />

months after imatinib was started, while in third patient<br />

maior molecular response was documented 9 months after<br />

initiation of imatinib mesylate. All three patients <strong>are</strong> 100%<br />

donor chimeras according to the unseparated peripheral<br />

blood cells.<br />

Conclusion: imatinib mesylate is efficient in treating patients<br />

with Ph positive chronic myeloic leukemia in relapse<br />

after allogeneic transplantation. Major or complete molecular<br />

response could be achieved after 9–12 months of therapy<br />

with imatinib mesylate 400 mg/day. Longer follow up time<br />

is m<strong>and</strong>atory to asses if the treatment efficacy is sustained.<br />

PP14 Long-term survival <strong>and</strong> late-onset<br />

complications after reduced-intensity<br />

conditioning (RIC) allogeneic stem cell<br />

transplantation (allo-SCT)<br />

Peric Z, Clavert A, Chevallier P, Brissot E, Malard F,<br />

Guillaume T, Delaunay J, Ayari S, Dubruille V,<br />

Le Gouill S, Mahe B, Gastinne T, Blin N, Harousseau JL,<br />

Moreau P, Milpied N, Mohty M, Service d’Hématologie<br />

Clinique, Centre Hospitalier et Universitaire (CHU) de<br />

Nantes, Nantes, France<br />

RIC allo-SCT is increasingly used in elderly or frail patients<br />

not eligible for st<strong>and</strong>ard myeloablative conditioning.<br />

While the features <strong>and</strong> natural history of long-term complications<br />

(LTC) <strong>are</strong> rather well described in the st<strong>and</strong>ard allo-<br />

SCT setting, data <strong>are</strong> still sparse in the RIC setting.<br />

This report analyzed the outcome <strong>and</strong> features of LTC in<br />

110 RIC allo-SCT patients who survived for a minimum of<br />

2 years after transplantation. The median age of recipients<br />

was 55 (range, 19–68) years. In all, 74 patients (67%) had a<br />

lymphoid malignancy, whereas 34 (31%) patients were diagnosed<br />

with myeloid malignancies. Two patients (2%) were<br />

treated for severe aplastic anemia. In total, 94 patients (86%)<br />

received G-CSF-mobilized PBSCs whereas 9 patients (8%)<br />

received unmanipulated bone marrow, <strong>and</strong> 7 patients (6%)<br />

received cord blood. 67 grafts (61%) were obtained from<br />

HLA identical sibling donors, 35 (32%) from HLA matched<br />

unrelated donors <strong>and</strong> 8 (7%) from 1 Ag HLA mismatched<br />

unrelated donors. The majority of patients received a fludarabine,<br />

busulfan, <strong>and</strong> ATG-based RIC regimen (n=80; 73%),<br />

11 patients (10%) received fludarabine <strong>and</strong> low-dose TBI,<br />

while the remaining 17 patients (15%) received different<br />

chemotherapy-based RIC regimens.<br />

Recurrence of the primary malignancy was found in 4<br />

(4%) cases <strong>and</strong> was the primary cause of late death. A total<br />

of 6 (6%) patients died of non-relapse causes at a median of<br />

3.2 (range, 2.4–6.1) years post allo-SCT. The non-relapse<br />

causes of death were secondary malignancy (n=3), chronic<br />

93


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

GVHD (n=2), <strong>and</strong> infection (n=1). The Kaplan-Meier estimate<br />

of OS was 81% (95%CI, 71–94) at 10 years. With a<br />

median follow-up of 4.6 years (range, 2–12.1), chronic<br />

GVHD was the most prevalent late complication with a cumulative<br />

incidence of 74% (95%CI, 62–83) at 10 years.<br />

Cardiovascular complications were diagnosed in 47 (43%)<br />

patients, with arterial hypertension ocuring in 32 (29%) patients<br />

<strong>and</strong> cardial insufficiency in 15 (14%) patients. Renal<br />

impairment occured in 37 (34%) patients. Pulmonary complications<br />

were found in 28 (25 %) patients <strong>and</strong> were mostly<br />

related to chronic GVHD. Endocrine disorders were diagnosed<br />

in 18 (16%) involving thyroid dysfunction in 6 (5%)<br />

patients. Psychological disorders (mostly depression) were<br />

observed in 9 (8%) survivors. A secondary malignancy<br />

occured in 9 (8%) patients; 3 patients were diagnosed with<br />

two malignancies. The main organs involved were skin<br />

(n=4), colon (n=3), prostate (n=2), lungs (n=1), urinary bladder<br />

(n=1) <strong>and</strong> neuroendocrine tissue. (n=1)<br />

We conclude that although RIC allo-SCT has the potential<br />

to cure a significant proportion of patients with otherwise<br />

fatal diseases, <strong>and</strong> though many patients can survive the<br />

acute complications of the procedure, many other patients<br />

may not enjoy full restoration of health with onset of a significant<br />

rate of LTC, waranting lifelong surveillance through<br />

a close partnership between the transplant center, <strong>and</strong><br />

organ-specific specialties.<br />

PP15 Factors predicting overall survival in<br />

patients with advanced chronic graft versus<br />

host disease diagnosed by NIH consensus<br />

criteria.<br />

Grkovic L 1,2 , Steinberg SM 1 , Baird K 1 , Williams KM 1 ,<br />

Cowen EW 1 , Mitchell SA 1 , Pulanic D 1,2 , Avila DN 1 ,<br />

Taylor TN 1 , Wroblewski SG 1 , Serventi Seiwerth R 1,2 ,<br />

Gress RE 1 , Pavletic SZ 1 . 1 National Cancer Institute,<br />

National Institutes of Health, Bethesda, USA, 2 Department<br />

of Medicine, Clinical Hospital Center Zagreb, Croatia<br />

Aim: To determine factors predictive for survival in patients<br />

with advanced cGVHD (chronic graft-versus-host<br />

disease) defined by NIH (National Institutes of Health) criteria.<br />

Patients <strong>and</strong> methods: 189 adults were enrolled onto the<br />

cross-sectional prospective cGVHD natural history study<br />

between 2004 <strong>and</strong> 2010. Median patient age was 48 years<br />

[18–70], median time from transplant to enrollment was 3<br />

years [0.3–22], <strong>and</strong> median time from cGVHD diagnosis to<br />

enrollment was 2 years [0–19]. 66% of the patients had severe<br />

<strong>and</strong> 33% had moderate cGVHD according to the NIH<br />

global scoring with a median of 4 organs involved [1–8].<br />

80% of the patients were receiving systemic immunosuppression<br />

<strong>and</strong> have failed a median of 4 [0–9] prior systemic<br />

therapies for their cGVHD. Median follow-up of surviving<br />

patients was 2.5 years [0.1–5.8]. 28% of the patients had<br />

Karnofsky performance status less than 80% <strong>and</strong> 7% had<br />

thrombocytopenia (


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

plant, received HLA-matched related (n=79) or unrelated<br />

(n=60) BM after myeloablative BuCy conditioning. HiCy<br />

(50 mg/kg/day) was given on days +3, <strong>and</strong> +4 as only<br />

planned GVHD prophylaxis. Patients with acute grade II-IV<br />

GVHD were treated with methylprednisolone 1–2.5 mg/kg/<br />

day IV as a first line therapy, <strong>and</strong> those with visceral GVHD<br />

also received calcineurin inhibitors (CNI) or other non-CNI<br />

immunosuppressants.<br />

Results: In the competing risk model (death <strong>and</strong> graft<br />

failure as competing risks), the incidence of acute grade II-<br />

IV <strong>and</strong> III-IV GVHD by day 200 was 50% <strong>and</strong> 16%, respectively.<br />

Thirteen percent of patients who developed acute<br />

grade II-IV GVHD were not treated 15% were treated with<br />

steroids alone, 63% with steroids plus a CNI <strong>and</strong> 9% with<br />

steroids plus non CNI-based agents. Overall cumulative incidence<br />

of SSI use was 45%. Median time to initiation of<br />

SSI was 42 (19–142) days <strong>and</strong> the median duration of SSI<br />

use was 152 (13–981) days. The cumulative incidence of<br />

glucocorticoid use was 38% in related <strong>and</strong> 52% in unrelated<br />

group. Steroids were tapered rather quickly, 9 months after<br />

BMT all patients in related group were off glucocorticoid<br />

treatment, while in unrelated group only 14% of patients<br />

were on therapy at that time. Cumulative incidence of nonglucocorticoid<br />

therapy initiation was 29% in related <strong>and</strong><br />

44% in unrelated group At 6 months <strong>and</strong> 1 year, 63% <strong>and</strong><br />

83% of patients were off all immunosuppressive therapy,<br />

respectively. With a median follow-up of 26 months, cumulative<br />

incidence of chronic GVHD was 10%. Only 3 patients<br />

have died with refractory GVHD.<br />

Conclusion: These results extend our previous observations<br />

that post-transplantation Cy is effective single agent<br />

strategy for prophylaxis of acute GVHD with both a low<br />

rate of grade III-IV <strong>and</strong> more than half of the patients never<br />

requiring additional SSI. The limited use of SSI may be responsible<br />

for low infectious rate <strong>and</strong> excellent immune reconstitution<br />

seen in these patients. This approach also provides<br />

novel platform to facilitate the use of post-transplant<br />

immunotherapy aimed at reducing relapse.<br />

PP17 Allogeneic transplantation from HLA<br />

matched unrelated donor – single Center<br />

experience<br />

Serventi Seiwerth R 1 , Mikulic M 1 , Mrsic M 1 , Grubic Z 2 ,<br />

Bojanic I 3 , Durakovic N 1 , Stingl K 2 , Labar B 1 . 1 Division of<br />

Hematology, Department of Medicine, 2 Department of<br />

Clinical Laboratory, 3 Department of Transfusiology,<br />

Clinical Hospital Center <strong>and</strong> School of Medicine,<br />

University of Zagreb, Croatia<br />

Introduction: Allogeneic transplantation is a st<strong>and</strong>ard<br />

therapeutic approach for malignant <strong>and</strong> some nonmalignant<br />

congenital <strong>and</strong> acquired hematopetic diseases. Only one<br />

third of patients who would benefit from allogeneic transplantation<br />

have an HLA identical family donor. Search for<br />

an HLA matched unrelated donor from the International<br />

Bone Marrow Donor Registry gives more patients the opportunity<br />

to get optimal treatment. Acute leukemias <strong>and</strong><br />

myelodysplastic syndromes followed by bone marrow failure<br />

syndromes <strong>are</strong> major indications for allogeneic stem<br />

cell transplantation from matched unrelated donor. With<br />

high resolution HLA typing techniques on the allele level,<br />

results of alogeneic transplantation from matched unrelated<br />

donors have reached results in family transplants.<br />

We present patients transplanted from HLA matched unrelated<br />

donor in University Hospital Center Zagreb in the<br />

period from 1991. – 2010.<br />

Patients <strong>and</strong> treatment: From 1991 till December 2010.<br />

71 patients, median age 28 years (range 1 month – 61 year)<br />

received HLA matched unrelated donor transplant; 30 female<br />

<strong>and</strong> 41 male. HLA matches were 10/10 for 23 patients,<br />

9/10 for 14 patients, 8/10 in two patients, 13 patients were<br />

transplanted from 8/8 HLA matched donors; one patient<br />

was transplanted with two umbilical cord blood 6/6 HLA<br />

match. Eight patients transplanted before 2000 received<br />

stem cells from unrelated donor 6 out of 6. Most of our patients<br />

were transplanted for acute leukemia (34 out of 71<br />

patient): 18 for acute myeloid <strong>and</strong> 16 for acute lymphoblastic<br />

leukemia; 19 out of 71 patients were transplanted because<br />

of chronic myeloid leukemia <strong>and</strong> myelodisplastic<br />

syndrome while 9 of them received stem cells because of<br />

lymphoproliferative neoplasm. Seven patients were allografted<br />

for other diseases. Only 9 out of 34 acute leukemia<br />

patients (26,5%) were transplanted in first complete remission;<br />

7 out of 16 CML patients received stem cells in first<br />

chronic phase; while patients with non-Hodgkin lymphoma<br />

were allografted in refractory disease; 16 out of 71 patients<br />

who were previously autografted received stem cells from<br />

matched unrelated donor in relapse; one patient received<br />

MUD transplant after rejection of first unrelated transplant<br />

from different donor.<br />

St<strong>and</strong>ard myeloablative conditioning regimen consisting<br />

of busulfan <strong>and</strong> cyclophosphamide or cyclophosphamide<br />

<strong>and</strong> total body irradiation with antitymocytic globulin were<br />

given to 46 patients;;. 25 (35, 2%) patients were transplanted<br />

after reduced intensity conditioning regimens, most of<br />

them being fludarabine based (fludarabine + cyclophosphamide;<br />

fludarabine + busulfan) in combination with antithymocytic<br />

globulin. Cyclosporine <strong>and</strong> short courses of metothrexate<br />

(for st<strong>and</strong>ard conditioning) <strong>and</strong> cyclosporine <strong>and</strong><br />

mofetil mycophenolate (for nonmyeloablative conditioning)<br />

were used in GvHD prophylaxis. As a stem cell source<br />

marrow, peripheral blood <strong>and</strong> umbilical cord blood were<br />

used in 31 patients, 36 patients <strong>and</strong> 4 patients respectively.<br />

Results: In this high risk group of patients 55% (39/71)<br />

of them were alive on day +100. Infections, acute Graft versus<br />

Host Disease grade III-IV, multiorgan failure <strong>and</strong> underlying<br />

disease were the principal cause of death in 17, 14, 4<br />

<strong>and</strong> 8 patients respectively. Data <strong>are</strong> missing for 9 patients.<br />

Most of the deaths occurred during first 12 months; Overall<br />

survival at 5 years is 24% (17/ 71); Platou was reached after<br />

18 months. Although not significant there is a trend for better<br />

survival in patients with unrelated donor (10/10) comp<strong>are</strong>d<br />

to patients with a donor 9/10 or 8/8.<br />

Conclusion: In this high risk group of patients 55% of<br />

them <strong>are</strong> alive on 100 days after allografting, with 5-year<br />

overall survival of 24%. Main causes of death <strong>are</strong> serious<br />

infections, GvHD, multiorgan failure <strong>and</strong> underlying disease.<br />

Better patient selection <strong>and</strong> transplantation in earlier<br />

phase of the disease would give better antitumor effect with<br />

less toxicity <strong>and</strong> superior long-term survival.<br />

95


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

PP18 Extracorporeal Photochemotherapy in<br />

Treatment of Chronic Graft-versus-Host<br />

Disease<br />

Bojanic I 1 , Serventi Seiwerth R 2 , Golubic Cepulic B 1 ,<br />

Mazic S 1 , Lukic M 1 , Raos M 1 , Plenkovic F 1 ,<br />

Golemovic M 1 , Dubravcic K 3 , Perkovic S 3 , Batinic D 3 ,<br />

Labar B 2 . 1 Department of Transfusiology, 2 Division of<br />

Hematology, Department of Medicine, 3 Department of<br />

Clinical Laboratory, Clinical Hospital Center <strong>and</strong> School<br />

of Medicine University of Zagreb, Croatia.<br />

Extracorporeal photochemotherapy (ECP) is an immunomodulatory<br />

therapy which has been used in treatment of<br />

chronic GVHD (cGVHD). ECP involves separation of the<br />

mononuclear cells (MNC) with leukapheresis, followed by<br />

ex vivo administration of photosensitizer 8-methoxypsoralen<br />

(8-MOP) <strong>and</strong> ultraviolet A (UV-A) radiation, before<br />

reinfusion to the patient.<br />

Aim: Aim of the study was to evaluate clinical <strong>and</strong> immunomodulatory<br />

effect of ECP procedures performed in<br />

patients with cGVHD. The frequency of adverse reactions<br />

associated with ECP was also evaluated.<br />

Patients <strong>and</strong> methods: We analyzed 351 ECP procedures<br />

performed in 6 patients; median ECP per patient was 52<br />

(range 13–127). Patients’ median age was 29 years (range,<br />

12–76 yrs). The patients suffered from generalized sclerodermatous<br />

skin changes, impaired join mobility <strong>and</strong> one<br />

patient had symptoms of oral disease. In all patients concomitant<br />

immunosuppressive treatments for cGVHD were<br />

used as necessary. ECP procedures were performed for two<br />

consecutive days: in initial phase weekly, followed every<br />

two weeks <strong>and</strong> than monthly according to clinical response.<br />

ECP was performed using the »off line« technique. MNCs<br />

were collected using COBE Spectra cell separator (Caridian<br />

BCT). In all leukapheresis 2 patient’s total blood volumes<br />

were processed. Collected MNC concentrate was transferred<br />

to Extracorporeal UV-A Bag Set (Cell Max GmbH)<br />

<strong>and</strong> diluted with saline solution. 8-MOP (Gerot) was injected<br />

into the UV-A Bag Set <strong>and</strong> bag was irradiated by PUVA<br />

Combi-Light UVA Illuminator at wave length of 350 nm<br />

with the irradiation dose of 2 J/cm 2 . Irradiated cells were<br />

reinfused back to the patient. During apheresis <strong>and</strong> reinfusion<br />

of irradiated cells patients were monitored for adverse<br />

reactions. Number of T-lymphocyte subsets (CD3+, CD3+4+,<br />

CD3+8+, CD4+ CD8+ ratio) <strong>and</strong> B-lymphocytes (CD 19+),<br />

in patient’s peripheral blood were tested monthly.<br />

Results: The effect of ECP in patients with cGVHD with<br />

skin <strong>and</strong> joint involvement was mostly beneficial. Five patients<br />

experienced either improvement or stabilization in<br />

sclerodermatous skin changes <strong>and</strong> joint mobility. The overall<br />

cutaneous response rate was 83% (complete response<br />

rate 50% 3/6 pts), <strong>and</strong> partial response rate 33% (2/6 pts). In<br />

patient who suffered from oral disease, the total recovery<br />

was observed. Clinical response was typically delayed until<br />

2 to 3 months. In patients who responded to ECP efficiently,<br />

the influence of ECP on T-cell subsets leads to the suggestion<br />

that interactions between T-cell subsets may participate<br />

in the process of ECP.<br />

96<br />

In general ECP was well tolerated. No increased incidence<br />

of infections <strong>and</strong> no serious adverse reactions have<br />

been observed. After reinfusion of MNCs, one patient experienced<br />

increase in body temperature, likely due to the release<br />

of cytokines from photomodified cells.<br />

Conclusion: ECP proved to be efficient <strong>and</strong> safe procedure<br />

that may be recommended for patients with cGVHD<br />

who do not respond to conventional therapy.<br />

PP19 Wernicke’s encephalopathy in an<br />

adolescent after allogeneic hematopoietic<br />

stem cell transplantation: a case report<br />

Rajic Lj 1 , Pavlovic M 1 , Femenic R 1 , Bilic E 1 , Krnic N 1 ,<br />

Barisic N 1 , Ozretic D 1 , Tesovic G 4 , Dusek D 4 , Zadro R 2 ,<br />

Serventi Seiwerth R 3 , Mikulic M 3 , Konja J 1 , Labar B 3 .<br />

1 Department of Pediatric, 2 Department of Clinical<br />

Laboratory, 3 Department of Medicine, Clinical Hospital<br />

Center Zagreb, 4 Clinical Hospital for Infectious Diseases,<br />

1,2,3,4 School of Medicine University of Zagreb, Croatia<br />

Introduction: Wernicke’s encephalopathy (WE) is a neurological<br />

emergency caused by a thiamine (vitamin B1) deficiency.<br />

It was first described in 1881 by German neurologist<br />

Carl Wernicke as a triad comprising of global confusion,<br />

ataxia <strong>and</strong> ophtalmoplegia. The proposed mechanism<br />

of brain lesions is impaired myelination due to decreased<br />

NADPH production. Although most commonly associated<br />

with chronic alcoholism in adults, non-alcoholic WE has<br />

been described in conditions predisposing to poor nutritional<br />

status. Reports of WE in infants <strong>and</strong> children <strong>are</strong> r<strong>are</strong>, <strong>and</strong><br />

the majority of paediatric cases <strong>are</strong> associated with malignancy<br />

<strong>and</strong> gastrointestinal disease. Patients undergoing hematopoietic<br />

stem cell transplantation (HSCT) <strong>are</strong> at high<br />

risk of this potentially lethal condition due to chemotherapy-induced<br />

nausea <strong>and</strong> vomiting, <strong>and</strong> depletion of thiamine<br />

stores by long-term total p<strong>are</strong>nteral nutrition (TPN) <strong>and</strong> glucose-containing<br />

i.v.solutions.<br />

Aim: To present the case report of an 18 yr old male adolescent<br />

presenting with Wernicke’s encephalopathy following<br />

unrelated allogeneic HSCT for underlying acute myelogenous<br />

leukemia.<br />

Case report: On day + 42 after HSCT, the patient presented<br />

with vertical nystagmus <strong>and</strong> hallucinations. He complained<br />

of generalized weakness, gait disturbance, hyperacusia,<br />

difficult swallowing <strong>and</strong> insomnia. Neurogical examination<br />

revealed mild confusion, lower limb <strong>and</strong> truncal<br />

ataxia, bilateral abducens nerve palsy, both vertical <strong>and</strong> rotatory<br />

nystagmus, soft palate palsy <strong>and</strong> <strong>are</strong>flexia. Cerebrospinal<br />

fluid analysis <strong>and</strong> cranial CT scans imaging revealed<br />

no abnormality. Magnetic resonance imaging (MRI) of the<br />

brain showed symmetric high-signal intensities in dorsomedial<br />

nuclei of the thalamus, mamillary bodies, inferior colliculi,<br />

periaqueductal gray <strong>and</strong> pontine tegmentu, which<br />

were consistent with diagnosis of WE. The patient was<br />

treated with thiamine 500 mg/day i.v. for one month. His<br />

general condition <strong>and</strong> neurological signs were improved but<br />

not completely.<br />

Discussion: Neurological complications occur in 30–<br />

40% of patients undergoing HSCT. They may be of infec-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

tious, cerebrovascular, toxic, immune-mediated, or metabolic<br />

origin. The diagnosis of WE is clinical but only 16%<br />

of affected patients develop pathognomonic clinical triad.<br />

Measurements of blood or erythrocyte thiamine concentrations<br />

<strong>are</strong> limited by a lack of specificity <strong>and</strong> <strong>are</strong> not routinely<br />

available in hospital laboratories. Due to selective<br />

vulnerability of the midline gray matter <strong>are</strong>as, MRI plays an<br />

important role in the diagnosis of this condition. It is crucial<br />

to consider WE in the differential diagnosis for all patients<br />

after HSCT presenting with confussion, ataxia <strong>and</strong> ophtalmoplegia.<br />

To prevent this devastating <strong>and</strong> often fatal neurologic<br />

complication, all patients underwent HSTC should be<br />

empirically treated with thiamine.<br />

PP20 Need + Information + Satisfaction<br />

Vieira S, Monroe D, Librojo M. London, United Kingdom<br />

Over the last decade, information for cancer patients has<br />

become increasingly more importance.<br />

The very nature of the cancer requires patients to learn<br />

about their disease in order to cope with the consequences<br />

of treatment <strong>and</strong> in inform the decision making process.<br />

It is therefore vital to provide enough information for patients<br />

<strong>and</strong> relevant others.<br />

People/patients vary in the amount of information they<br />

require. Evidence suggests that some patients do not want<br />

very much information about their diseases or any treatments.<br />

Stage Information provided on the following topics<br />

On admission<br />

N %<br />

1. Side effects of therapy or chemotherapy 20 (40%)<br />

2. Procedures<br />

(Bone Marrow Aspiration, Hickman Line insertion etc)<br />

19 (38%)<br />

3. Sexuality 5 (10%)<br />

4. Fertility 9 (18%)<br />

5. Results of diagnostic test 27 (54%)<br />

6. Follow up, appointments 25 (50%)<br />

7. Diet 14 (28%)<br />

other 0 (0%)<br />

During their stay 1. Side effects of therapy or chemotherapy 18 (36%)<br />

2. Procedures<br />

(Bone Marrow Aspiration, Hickman Line insertion etc)<br />

20 (40%)<br />

3. Infection Control 21 (42%)<br />

4. Sexuality 4 (8%)<br />

5. Fertility 6 (12%)<br />

6. Results of diagnostic test 29 (58%)<br />

7. others<br />

blood exams<br />

3 (6%)<br />

On discharge 1. Outcome of your treatment 30 (60%)<br />

2. Follow up, appointment 47 (94%)<br />

3. Medication 49 (98%)<br />

4. What happens next 38 (76%)<br />

5. When to call the medical team (Hospital) 41 (82%)<br />

6. Work life 26 (52%)<br />

7. others:<br />

alternative treatment models<br />

1 (2%)<br />

Total 50 (100%)<br />

Aim: This study aims to explore how much information<br />

our patient group wants to know <strong>and</strong> how much information<br />

our team provides to the patients<br />

Patients <strong>and</strong> methods: A questionnaire was prep<strong>are</strong>d to<br />

collect data on how much information was given to patients<br />

on three different stages of their treatment plans. These<br />

stages were: on admission, during their inpatient stay <strong>and</strong><br />

on discharge. A fourth section was added, to investigate<br />

other information need patient felt required.<br />

The questionnaire was used between May 2010 till October<br />

2010, total of 50 questionnaire. Outcome of these data<br />

collection is shown in table I.<br />

Results: 50 malignant haematology patients were admitted<br />

to our unit for various reasons (See Table I).<br />

Our audit was evaluated in three different stages. In admission<br />

stage most of the information provided were about<br />

side effects of treatment, test results. (See Table I).<br />

During in patient period was the llowest stages where information<br />

provided. It should be b<strong>are</strong> in mind that haematology<br />

patient group has long hospitalisation, the data collection<br />

tool was given on discharge. Therefore, data might<br />

be lost due to poor recalling the information required.<br />

Results also showed our patient were happy about the information<br />

they were given especially on discharge as most<br />

aspect of information covered (See Table I)<br />

PP21 CD43 expression is associated with inferior<br />

survival within activated B-cell group of<br />

diffuse large B-cell lymphoma<br />

Mitrovic Z 1,4 *, Iqbal J 1 , Fu K 1 , Smith LM 2 ,<br />

Weisenburger DD 1 , Greiner T 1 , Auon P 1 , Bast M 3 ,<br />

Armitage JO 3 , Vose JM 3 , Chan WC 1 . Departments of<br />

1 Pathology/Microbiology <strong>and</strong> 3 Hematology/Oncology,<br />

2 College of Public Health, University of Nebraska Medical<br />

Center, Omaha, NE, USA, 4 School of Medicine University<br />

of Zagreb, Croatia<br />

CD43 expression is associated with inferior survival within<br />

activated B-cell group of diffuse large B-cell lymphoma.<br />

Aim: The expression of CD43, an abundant membrane<br />

glycoprotein, was associated with an inferior survival of patients<br />

with diffuse large B-cell lymphoma (DLBCL) in a<br />

single-center study. The aim of the present study is to evaluate<br />

the prognostic significance of CD43 in our cohort of<br />

patients <strong>and</strong> correlate protein expression data with gene expression<br />

profiling (GEP).<br />

Patients <strong>and</strong> methods: A total of 129 patients treated<br />

with R-CHOP from Nebraska Lymphoma Study Group<br />

were included. Tumor samples on tissue microarray (TMA)<br />

were stained for CD43, CD10, BCL6, <strong>and</strong> MUM1 to correlate<br />

CD43 expression <strong>and</strong> cell of origin according to the<br />

Hans algorithm. GEP data were available for 31 patients.<br />

Results: CD43 was expressed in 25 of 129 DLBCLs<br />

(19.4%). There was no difference in the pre-treatment clinical<br />

characteristics between CD43+ <strong>and</strong> CD43-group of patients.<br />

CD43 expression was associated with inferior failure-free<br />

survival (FFS) <strong>and</strong> overall survival (OS), p=0.032<br />

<strong>and</strong> p=0.018, respectively. Furthermore, CD43 was signifi-<br />

97


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

cantly related to activated B-cell (ABC) group defined by<br />

GEP <strong>and</strong> by immunohistochemistry, p=0.006 <strong>and</strong> p=0.0485,<br />

respectively. Interestingly, patients with CD43+ DLBCL<br />

had very poor OS (p < 0.001) within the ABC group <strong>and</strong> 4.3<br />

times the risk of death by multivariate analysis comp<strong>are</strong>d to<br />

CD43-. When patients with GEP data were comp<strong>are</strong>d within<br />

the ABC group, a significant enrichment in stromal-1 signature<br />

<strong>and</strong> immune-response-1 signature was observed in<br />

CD43- group.<br />

Conclusion: CD43 is an adverse prognostic marker in<br />

DLBCL <strong>and</strong> is preferentially expressed in the ABC group. It<br />

can identify patients with extremely poor outcome within<br />

the ABC group. A possible explanation is the association<br />

with more favorable microenvironment signatures for CD43-<br />

cases within the ABC group.<br />

* This work was financed in part by a grant from the National<br />

Foundation for Science, Higher Education <strong>and</strong> Technological<br />

Development of the Republic of Croatia<br />

PP22 Association of Interleukin-10,TNF-Alpha<br />

<strong>and</strong> TGF-Beta Gene Polymorphisms on The<br />

Outcome of Diffuse Large B-cell Lymphoma<br />

Tarabar O, Tukic Lj, Cikota B, Milanovic N, Aleksic A,<br />

Magic Z. Military Medical Academy, Belgrade, Serbia<br />

Abstract: Diffuse large B-cell lymphomas (DLBCL)<br />

represent a heterogeneous group of lymphoproliferative disorders<br />

with highly variable clinical course <strong>and</strong> outcome.<br />

Published date indicated that common genetic variants in<br />

immune/ inflammatory response genes can affect outcome<br />

of diffuse large B-cell lymphomas (DLBCL).<br />

Aim: To analyze the association of IL-10 –3575, –1082,<br />

TNF-alpha –308 <strong>and</strong> TGF-beta Leu10Pro polymorphic<br />

variants with clinical characteristics <strong>and</strong> outcome of DLB-<br />

CL patients (pts) treated with rituximab (R)-CHOP therapy.<br />

Patients <strong>and</strong> methods:Between January 2004 <strong>and</strong> December<br />

2007, a total of 64 pts (M/F 33/31; aged 22–76, median<br />

48 yrs) with newly diagnosed DLBCL were entered<br />

into this study. Factors examined for prognostic significance<br />

included sex, age (60 yrs or younger vs. older), Ann Arbor<br />

(CS) stage (I-II vs. III-IV), serum lactate dehydrogenase<br />

level (normal vs. elevated), number of extranodal sites of<br />

disease (0 or 1 vs. 2 or more), performance status (0 or 1 vs.<br />

2 or more), <strong>and</strong> IPI score (0–2 vs. 3–5). The therapy for all<br />

pts was R-CHOP (6–8 cycles). Genotypes were determined<br />

with PCR-based methodology.<br />

Results:TGF-beta Pro10 was associated with an unfavorable<br />

features of DLBCL. A significantly more frequent<br />

stage III-IV (OR 4.65, 95% CI 1.33-16.12; p=0.016) <strong>and</strong> IPI<br />

score 3–5 (OR 5.37, 95% CI 1.45-20.0; p=0.012) were<br />

found in pts with the Pro allele variant (LeuPro/ProPro)<br />

than in carriers of LeuLeu genotype. IL-10 <strong>and</strong> TNF-alpha<br />

gene polymorphisms were not associated with established<br />

prognostic factors. Considering survival, only TNF-alpha –<br />

308 influenced overall survival (OS) in R-CHOP treated<br />

pts. With a median follow-up of 23 months, there was significantly<br />

difference between GG <strong>and</strong> GA/AA carriers (log<br />

98<br />

rank test, p=0.048). Overall survival at 3 years was 61.5%<br />

(95% CI 28.8-46.9) for GA/AA genotypes <strong>and</strong> 83.2% (95%<br />

CI 50.8-98.9) for GG genotype of TNF-alpha –308.<br />

Conclusion:Our results indicate that TNF-alpha –308<br />

<strong>and</strong> TGF-beta Leu10Pro gene variations could be associated<br />

to the clinical course <strong>and</strong> outcome of R-CHOP treated DL-<br />

BCL patients.<br />

PP23 Dose Adjusted EPOCH – Rituximab as First<br />

Line Treatment for High Risk Diffuse Large<br />

B-Cell Lymphoma: A Single Center<br />

Experience<br />

Pejsa V, Prka Z, Lucijanic M, Jaksic O, Pirsic M,<br />

Ajdukovic R, Kusec R. Division of Hematolofy,<br />

Department of Medicine, Universiti Hospital Dubrava<br />

<strong>and</strong> School of Medicine, University of Zagreb, Croatia<br />

Abstract: Dose-adjusted EPOCH- rituximab (DA-EP-<br />

OCH-R) is an infusional protocol which is based on pharmacodinamical<br />

adjustment of drug dosage depending on<br />

laboratory values of absolute neutrophil count <strong>and</strong> platelets<br />

(Wilson WH et al. Blood 2002;99:2685–2693). Rationale<br />

for this regimen is well known fact that tumor cells <strong>are</strong> less<br />

resistant on prolonged exposition to low dose of chemotherapy<br />

than to short exposition to high dose of drugs.<br />

Aim: To assess clinical outcome in patients with diffuse<br />

large B-cell lymphoma (DLBCL) with poor prognostic factors<br />

(IPI ≥2 <strong>and</strong>/or high proliferation index; PI>80%, measured<br />

as percentage of Ki67+ cells) treated with DA-EP-<br />

OCH-R as first line treatment.<br />

Patients <strong>and</strong> methods: From May 2005. to July 2011.,<br />

22 patients, median age 48.5 years (range 17–75) with diffuse<br />

large B-cell lymphoma were included. Elevated LDH<br />

had 16 out of 22 patients (73%) <strong>and</strong> 17 out of 22 patients<br />

(77%) were in Ann Arbor stages 3 <strong>and</strong> 4. IPI ≥2 had 18 out<br />

of 22 patients (82%). DA-EPOCH-R was administred according<br />

to original schedule. Six of these patients proceed<br />

to autoHSCT as consolidation therapy due to very aggressive<br />

histology <strong>and</strong> biology of disease.<br />

Results: Overall response rate (ORR) was 86% (19/22),<br />

including 13 (59%) complete responses (CR) <strong>and</strong> 6 (27%)<br />

partial responses (PR). Overall survival (OS) was 64% at 5<br />

years (median not reached) with median follow up of 14<br />

months (range 3–68). Progression free survival (PFS) was<br />

70% at 5 years. Of patients who achieved CR, 12 <strong>are</strong> still in<br />

complete remission.<br />

Conclusion: In this report we presented 22 patients with<br />

high risk diffuse large B-cell lymphoma. Our results showed<br />

that DA-EPOCH-R is highly effective regimen in this group<br />

of patients. These results <strong>are</strong> conclusive with results of Garcia-Su<strong>are</strong>z<br />

<strong>and</strong> colleagues (Garcia-Su<strong>are</strong>z J et al. British<br />

journal of Haematology 2007;136:276–285) who also find<br />

this regimen effective as first line treatment in DLBCL patients.<br />

Some of our patients were consolidated with autoHSCT<br />

due to high proliferation index or IPI≥3. Only 1<br />

patient relapsed during follow up period. Of 6 patients who<br />

were transplated, all but one <strong>are</strong> still in CR. We conclude<br />

that DA-EPOCH-R is highly effective regimen as first line<br />

treatment in high risk diffuse large B-cell lymphoma.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

PP24 Immunoblastic Morphology as a Possible<br />

Prognostic Indicator for the Outcome of the<br />

Patients with Diffuse Large B cell<br />

Lymphoma in Era of the Rituximab Based<br />

Treatment: Single Centre Experience<br />

Trajkova S, Panovska-Stavridis I, Stojanovik A,<br />

Petrusevska G, Dukovski D, Cevreska L. Department of<br />

Hematology, Medical Faculty, Skopje, Republic of<br />

Macedonia<br />

Immunoblastic morphology as a possible prognostic indicator<br />

for the outcome of the patients with diffuse large B<br />

cell lymphoma in era of the Rituximab based treatment:<br />

single centre experience<br />

Aim: In order to investigate the prediction value of the<br />

immunoblastic morphology(IB) as a possible prognostic indicator<br />

for the outcome of our DLBL patient treated with<br />

the Rituximab (R)-CHOP regimen we conducted a retrospective<br />

study.<br />

Patients <strong>and</strong> methods: Our study enrolled 192 DLBL<br />

patients diagnosed <strong>and</strong> treated at the University Clinic of<br />

Hematology in the period between February 2002 <strong>and</strong> December<br />

2007. They were all treated with R-CHOP regimen<br />

<strong>and</strong> the median follow-up of the patient was 36 months. We<br />

analyzed the biopsy samples immunohistochemically for<br />

markers of germinal center (Bcl6), post-germinal center<br />

(MUM1) <strong>and</strong> apoptosis (Bcl-2).<br />

Results: The patients were categorized as DLBL(132;<br />

68,7%), IB(60;31,2). The median overall survival time (OS)<br />

were 59,3 months in DLBL group <strong>and</strong>,<strong>and</strong> 42,2months in<br />

IB group, <strong>and</strong> time to treatment (TT) were 56,8, <strong>and</strong><br />

30,6months respectively for the IB group. The DLBL <strong>and</strong><br />

IB groups were comparable regarding the age, gender distributions<br />

<strong>and</strong> all others already established prognostic parameters<br />

as performance status, advanced IPI, albumin level<br />

except for the low IPI 0–2 which was statistically associated<br />

with the DLBL group (p=.024).<br />

Conclusion: Our results did not show any statistical survival<br />

advantage <strong>and</strong> better outcome for the patient classified<br />

as DLBL when treated with R-CHOP <strong>and</strong> indicate that immunohistohemical<br />

markers do not really reflect the molecular<br />

diversity of the tumor. Our work shows that IB morphology<br />

is a major risk factor in DLBL patients treated with R-<br />

CHOP. Therefore this morphology appears to capture some<br />

adverse molecular events that a currently hard to detect with<br />

routine diagnostic procedures.<br />

PP25 Primary Mediastinal Large B-Cell<br />

Lymphoma – Results of our Center<br />

Miljkovic E, Marjanovic G, Cojbasic I, Tijanic I,<br />

Markovic D, Nikolic V, Macukanovic-Golubovic L,<br />

Govedarovic N, Vucic M. Clinic of Hematology, Clinical<br />

Center Nis, Serbia<br />

Abstract: Primary mediastinal large B-cell lymphoma<br />

(PMBCL) is a distinct clinico-pathological subtype of diffuse<br />

large B-cell lymphoma (DLBCL) <strong>and</strong> represents less<br />

than 3% of all non-Hodgkin lymphoma cases.The tumor ap-<br />

pears to be derived from medullary B cells within the thymus<br />

gl<strong>and</strong>.Phenotypically tumor cells <strong>are</strong> positive for CD45,<br />

B cell markers(Pax-5, CD19, CD20, CD22, CD79a) <strong>and</strong><br />

bcl-2, often positive for CD30, MUM1 <strong>and</strong> bcl-6. <strong>and</strong> negative<br />

for CD10 <strong>and</strong> CD21. The optimal treatment is unknown,<br />

with some studies suggesting a superior outcome with doseintensive<br />

chemotherapy regimens. The role of mediastinal<br />

radiotherapy upon completion of chemotherapy remains unclear<br />

Aim: Aim was to evaluate response in patients treating<br />

with R-MACOP-B regimen.<br />

Patients <strong>and</strong> methods:We performed a retrospective review<br />

of 5 patients with PMBCL, who were treated at our<br />

clinic during the period between Januar 2009 <strong>and</strong> December<br />

2010.<br />

Results: Among 5 patients there were 4 women <strong>and</strong> 1<br />

man.Median age was 31 years (24–33). Most patients were<br />

in stage II–80% (4) <strong>and</strong> one in stage III–20%, all of them<br />

with B simptoms. Bulky mediastinal mass (>5cm in diameter)<br />

was present in all of our patients. Superior vena cava<br />

syndrom with facial edema, neck vein distention <strong>and</strong> upper<br />

extemity swelling was seen in 60% of the patients (3). After<br />

treating with R-MACOP-B regimen (rituximab, methotrexate,<br />

doxorubicin, cyclophoshamide, vincristine, prednisone,<br />

beomycin) complete response was accomplished in 40%<br />

(2),partial response in 20% (1).Two patients (40%) died,<br />

one due to progression of disease <strong>and</strong> one due to the toxicity<br />

of the treatment. Two patients who complited R-MA-<br />

COP-B had involved-field radiation therapy (IFRT) following<br />

chemotherapy <strong>and</strong> complete remission was confirmed<br />

by performing PET scan.<br />

Conclusion: PMBCL, an uncommon disease affects<br />

young people with a female prevalence <strong>and</strong> frequent bulky<br />

mediastinal mass.Studies have demonstrated advantage of<br />

R-MACOP B comp<strong>are</strong>d to CHOP,so we were treated our<br />

patients with this regimen. Two patients who complited R-<br />

MACOP B <strong>and</strong> had IFRT <strong>are</strong> still in complete remission<br />

after two years of follow up.<br />

PP26 Dose-intense BACOP for Treatment<br />

of High-risk Peripheral T-cell NHL<br />

Aurer I 1 , Dujmovic D 1 , Basic-Kinda S 1 , Nemet D 1 ,<br />

Radman I 1 , Ilic I 2 , Stern-Padovan R 3 , Santek F 4 , Sertic D 1 ,<br />

Cigrovski N 3 , Labar B 1 . 1 Department of Medicine,<br />

2 Pathology, 3 Radiology <strong>and</strong> 4 Oncology, University Hospital<br />

Center <strong>and</strong> School of Medicine, University of Zagreb,<br />

Croatia<br />

PTCLs, except ALK+ ALCL, have a bad prognosis with<br />

overall 5-year survival rates below 30%. Because of their<br />

relative rarity, no large r<strong>and</strong>omized trials have been performed<br />

<strong>and</strong> the importance of individual cytotoxic agents,<br />

their dose density <strong>and</strong> intensity as well as that of autografting<br />

remains unproven. At our center we have been using<br />

dose-intense BACOP, a regimen modeled after the French<br />

ACVBP, for treatment of patients with high risk PTCL. The<br />

regimen consisted of cyclophosphamide 1500 mg/m 2 day 1,<br />

doxorubicin 75 mg/m 2 day 1, bleomycin 15 mg days 1 <strong>and</strong><br />

99


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

5, vincristine 2 mg days 1 <strong>and</strong> 5 <strong>and</strong> prednisone 50 mg/m 2<br />

days 1–5 every 3 weeks for 4–6 cycles. Responding patients<br />

underwent stem-cell collection <strong>and</strong> autografting after<br />

BEAM conditioning. Areas of initially bulky disease <strong>and</strong><br />

those not in CR prior to transplantation were irradiated posttransplant<br />

with 30–40 Gy. Patients with high risk for development<br />

of CNS disease received intrathecal methotrexate<br />

prophylaxis. Response evaluations were performed after<br />

2–3 cycles <strong>and</strong> at the end of chemotherapy. Patients were<br />

considered high risk if they had any PTCL type with IPI 3<br />

or more, bulky disease or failed to respond to 2–3 cycles of<br />

st<strong>and</strong>ard CHOP-21. We treated 28 patients, 20 men <strong>and</strong> 8<br />

women, 17–67 years old (median 46). Thirteen had PTCL-<br />

NOS, 8 ALCL, 3 AILD, 2 nasal type, 1 panniculitis-like <strong>and</strong><br />

1 hepatosplenic PTCL. Two had stage IE bulky, 1 stage II<br />

bulky, 2 stage III <strong>and</strong> 23 stage IV disease. The toxicity of<br />

treatment was substantial, all patients experienced serious<br />

hematological toxicity <strong>and</strong> infections or neutropenic fever,<br />

3 died of infection during chemotherapy <strong>and</strong> 1 during stemcell<br />

transplantation, 2 had DVT/PE, 1 died of complications<br />

related to the necrosis of an extremely bulky tumor <strong>and</strong> one<br />

underwent emergency splenectomy after tumor necrosis<br />

caused splenic rupture. Three patients were unavailable for<br />

efficacy analysis due to early death, 4 progressed <strong>and</strong> 21<br />

initially responded (75%); 14 achieved CR <strong>and</strong> 7 PR. None<br />

of the 3 patients with nasal type or hepatosplenic PTCL responded.<br />

Of the responders, 11 underwent autografting as<br />

100<br />

intended, 3 relapsed prior to transplantation, treatment was<br />

modified in 3 due to toxicity, 2 were not transplanted due to<br />

patient refusal or physician decision, 1 underwent allogeneic<br />

transplantation <strong>and</strong> 1 failed to mobilize.<br />

After a median follow-up of survivors of 20 months, 9<br />

patients <strong>are</strong> still alive, including 8 who were autografted as<br />

intended. Median response duration was 12 months, FFS 6<br />

months <strong>and</strong> OS 10 months (Fig 1 <strong>and</strong> 2.). In conclusion, a<br />

significant proportion of high-risk PTCL-NOS, ALCL <strong>and</strong><br />

AILD patients respond to dose-intense BACOP followed by<br />

autologous stem-cell transplantation but the toxicity of<br />

treatment is substantial.<br />

PP27 Oral lomustine, chlorambucil, etoposide<br />

<strong>and</strong> prednisolone (CCEP) for treatment of<br />

patients with advanced lymphoma<br />

Radman I, Vodanovic M, Mitrovic Z, Aurer I, Basic-Kinda<br />

S, Labar B. Division of Hematology, Department of<br />

Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

Abstract: Oral palliative chemotherapy is reserved for<br />

the refractory <strong>and</strong> relapsed lymphoma patients who <strong>are</strong> not<br />

c<strong>and</strong>idates for further aggressive treatment. Herein we report<br />

single institution experience.<br />

Aim: CCEP, all – oral chemotherapy containing CCNU,<br />

chlorambucil, etoposide <strong>and</strong> prednisone was tested in refractory<br />

<strong>and</strong> relapsed lymphoma patients in advanced stages.<br />

Patients <strong>and</strong> methods: Twenty patients with refractory/<br />

relapsed lymphoma observed between 2000 <strong>and</strong> 2010 were<br />

treated with CCEP. Eight patients were diagnosed with<br />

Hodgkin lymphoma, 9 with aggressive non-Hodgkin lymphoma<br />

<strong>and</strong> 3 with indolent lymphoma. All patients were in<br />

advanced stage disease (Ann Arbor III/IV) <strong>and</strong> previously<br />

treated > 2 lines of therapy.<br />

Results: The median age was 51 (range 20–76 years).<br />

Thirteen patients (65%) had been extensively pretreated (≥3<br />

treatment), including autologous or allogeneic stem cell<br />

transplantation. All patients were in advanced stage disease<br />

(Ann Arbor III/IV), with extramedullary spread; lung 10 patients<br />

(50%), liver 7 patients (35%), spleen 5 patients (25%)<br />

<strong>and</strong> bone marrow 4 patients (20%). Overall response rate<br />

was 50%; 6 patients (30%) obtained stable state <strong>and</strong> 4 (20%)<br />

progressive disease. Median follow-up was 11 months with<br />

30% of 2-year survival rate. Patients receiving ≥ 3 cycles of<br />

CCEP achieved better response rate <strong>and</strong> longer survival.<br />

The progressive disease <strong>and</strong> toxicity were the main reasons<br />

for discontinuation of treatment.<br />

Conclusion: CCEP is low cost <strong>and</strong> effective chemotherapy<br />

with acceptable toxicity <strong>and</strong> good tolerability. In half of<br />

the patients with very adverse prognosis one can expect<br />

temporary treatment success.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

PP28 Effi cacy <strong>and</strong> Safety of Ruxolitinib, a JAK1<br />

<strong>and</strong> JAK2 Inhibitor, in Patients With<br />

Myelofi brosis: Results From a R<strong>and</strong>omized,<br />

Double-Blind, Placebo-Controlled, Phase III<br />

Trial (COMFORT-I)<br />

Verstovsek S 1 , Mesa R 2 , Gotlib J 3 , Levy R 4 , Gupta V 5 ,<br />

DiPersio J 6 , Catalano J 7 , Deininger M 8 , Miller C 9 ,<br />

Silver R 10 , Talpaz M 11 , Winton E 12 , Harvey J 13 ,<br />

Arcasoy M 14 , Hexner E 15 , Lyons R 16 , Paquette R 17 ,<br />

Raza A 18 , Vaddi K 4 , Erickson-Viitanen S 4 , Koumenis I 4 ,<br />

Sun W 4 , S<strong>and</strong>or V 4 , Kantarjian H 1 . 1 The University of Texas<br />

MD Anderson Cancer Center, Houston, TX, USA; 2 Mayo<br />

Clinic, Scottsdale, AZ, USA; 3 Stanford Cancer Center,<br />

Stanford, CA, USA; 4 Incyte Corporation, Wilmington, DE,<br />

USA; 5 Princess Marg<strong>are</strong>t Hospital, Toronto, Canada;<br />

6 Washington University School of Medicine, St. Louis, MO,<br />

USA; 7 Frankston Hospital, Frankston, Australia;<br />

8 University of Utah Huntsman Cancer Institute, Salt Lake<br />

City, UT, USA; 9 Saint Agnes Cancer Institute, Baltimore,<br />

MD, USA; 10 Weill Cornell Medical College, New York, NY,<br />

USA; 11 University of Michigan, Ann Arbor, MI, USA;<br />

12 Emory University School of Medicine, Atlanta, GA, USA;<br />

13 Birmingham Hematology & Oncology, Birmingham, AL,<br />

USA; 14 Duke University Health System, Durham, NC,<br />

USA; 15 University of Pennsylvania Health System,<br />

Philadelphia, PA, USA; 16 Cancer C<strong>are</strong> Center of South<br />

Texas, San Antonio, TX, USA; 17 UCLA Medical Hematology<br />

& Oncology, Los Angeles, CA, USA; 18 Columbia<br />

Presbyterian Medical Center, New York, NY, USA<br />

Background: Myelofibrosis (MF) is characterized by<br />

splenomegaly, debilitating symptoms, cytopenias, <strong>and</strong> shortened<br />

survival. No effective drug therapies <strong>are</strong> available;<br />

however, dysregulated JAK-STAT signaling, a key feature<br />

of MF, is a promising therapeutic target. Ruxolitinib (Rux),<br />

a selective JAK1 <strong>and</strong> JAK2 inhibitor, showed clinical activity<br />

in MF.<br />

Aim: COMFORT-I was designed to comp<strong>are</strong> the efficacy<br />

<strong>and</strong> safety of Rux with that of placebo (PB) in MF patients<br />

(pts).<br />

Patients <strong>and</strong> methods: Pts with intermediate-2 or highrisk<br />

MF (per IWG-MRT) were r<strong>and</strong>omized to Rux or PB.<br />

Rux starting dose was 15 mg or 20 mg PO BID for pts with<br />

baseline platelet count of 100–200 x 10 9 /L or >200 x 10 9 /L,<br />

respectively; the dose could be adjusted during the study to<br />

optimize efficacy <strong>and</strong> safety. The primary endpoint was the<br />

proportion of pts with >=35% reduction in spleen volume at<br />

week (wk) 24, as assessed by magnetic resonance imaging<br />

or computed tomography (blinded review). Secondary endpoints<br />

included duration of spleen response, changes in<br />

daily symptom burden (Total Symptom Score [TSS] using<br />

the modified Myelofibrosis Symptom Assessment Form<br />

[MFSAF] v2.0), <strong>and</strong> survival. Changes in quality of life<br />

(QOL) <strong>and</strong> fatigue were exploratory measures, as were molecular<br />

<strong>and</strong> serum biomarkers <strong>and</strong> transfusion dependence.<br />

Results: 155 pts were r<strong>and</strong>omized to Rux <strong>and</strong> 154 to PB<br />

(median follow-up: 32.2 wks). At 24 wks, the proportion of<br />

pts achieving a >=35% reduction in spleen volume was<br />

41.9% for Rux vs 0.7% for PB (p=50% improvement in<br />

TSS was 45.9% for Rux vs 5.3% for PB (p< 0.0001). Ruxtreated<br />

pts had a mean 46.1% improvement in TSS; PBtreated<br />

pts experienced a 41.8% worsening (p20% of pts)<br />

non-hematologic adverse events in pts on Rux vs PB were<br />

abdominal pain (10.3% vs 41.1%), fatigue (25.2% vs<br />

33.8%), diarrhea (23.2% vs 21.2%), <strong>and</strong> peripheral edema<br />

(18.7% vs 22.5%). The most common grade 3 or 4 hematologic<br />

laboratory abnormalities at any time during the study<br />

(Rux vs PB) were low hemoglobin (45.2% vs 19.2%) <strong>and</strong><br />

low platelets (12.9% vs 1.3%). Anemia <strong>and</strong> thrombocytopenia<br />

were manageable through dose modification/ temporary<br />

hold <strong>and</strong> r<strong>are</strong>ly led to study discontinuation (1 pt in each<br />

arm for each event).<br />

Conclusion: Rux demonstrated rapid <strong>and</strong> sustained clinical<br />

benefits in spleen size <strong>and</strong> debilitating symptoms, <strong>and</strong><br />

improved QOL in pts with MF; conversely, pts on PB had<br />

progressive worsening of disease. Rux was generally well<br />

tolerated. These results suggest that Rux may become an<br />

important new treatment option for the management of this<br />

chronic, advancing neoplasm.<br />

Support: Incyte Corp<br />

PP29 Prevalence of the JAK2 V617F Mutation in<br />

Croatian Patients with Classic Ph-Negative<br />

Myeloproliferative neoplasm<br />

Duletic Nacinovic A, Dekanic A, Hadzisejdic I, Seili I,<br />

Grahovac B, Grohovac D, Valkovic T, Host I, Petranovic<br />

D, Jonjic N. Department of Medicine <strong>and</strong> Deppartment of<br />

Pathology, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Rijeka, Croatia.<br />

The classic Ph-negative myeloproliferative neoplasms<br />

(MPNs) <strong>are</strong> clonal disorders of multipotent haematopoietic<br />

progenitors. The Janus-associated Kinase-2 mutation JAK2<br />

V617F in such neoplasms has been described as a frequent<br />

genetic event in majority of these patients.<br />

Aim: To investigate the status of the JAK2-V617F mutation<br />

in patients with classic Ph-negative MPNs treated in<br />

Clinical Hospital Center Rijeka <strong>and</strong> to comp<strong>are</strong> it with hemoglobin<br />

<strong>and</strong> hema-tocrit level, white blood cells <strong>and</strong> platelet<br />

count, splenomegaly, leukocyte alkaline phosphatase<br />

score <strong>and</strong> clinical features.<br />

Patients <strong>and</strong> methods: DNA was isolated from peripheral<br />

blood granulocytes in 115 patients: 41 with polycythemia<br />

vera (PV), 43 with essential thrombocythemia (ET), 9<br />

with primary myelofibrosis (PMF), 10 with myeloproliferative<br />

neoplasm-unclassifiable (MPN-u), 4 with secondary<br />

erithrocytosis <strong>and</strong> 5 patients with secondary thrombocytosis.<br />

The JAK2-V617 mutation was determined using allele<br />

specific PCR.<br />

Results: The JAK2-V617F mutation was found in 71/106<br />

(66.98%) patients with MPNs or in 36/41 with PV (87.80%),<br />

25/43 with ET (58.14%), 5/9 with PMF (55.56%) <strong>and</strong> 5/13<br />

MPNs-unclassified (38.46%) disorders. The JAK2-V617<br />

mutation was absent in patients with secondary erythrocytosis<br />

<strong>and</strong> seconrary thrombocytosis. There were significant<br />

differences in hemoglobin <strong>and</strong> hematocrit level, leukocyte<br />

alkaline phosphatase score <strong>and</strong> white blood cells at diagno-<br />

101


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

sis in JAK2-V617F non mutated versus mutated patients<br />

with MPNs. Vascular events were present in 23/106 (21.7%)<br />

patients with MPNs or more specifically in 13/41 (31.7%)<br />

with PV, 8/43 (18.6%) with ET, 1/9 (11.1%) with PMF <strong>and</strong><br />

1/13 (7.7%) with MPN-u. The majority of those patients<br />

with one or more vascular events were JAK2-V617F positive.<br />

When analyzed within each entity than it could be emphasized<br />

that one or more vascular events, or recurrent<br />

thrombosis was detected in 13/41 (31.7%), 7/41 (17.1%)<br />

cases, respectively with PV, or in 10/13 with JAK2-V617F<br />

positive status. In patients with ET, comp<strong>are</strong> to PV, vascular<br />

events (8/43) were less present (18.6%). Though, as noticed<br />

for PV, most of those patients (6/8) were associated with<br />

JAK2-V617F mutation. In patients with PMF only 1vascular<br />

event was confirmed (patient was JAK2V617F negative),<br />

as well as in patients with MPN-u (patient was JAK2 positive).<br />

Conclusion: The JAK2-V617F mutation was frequently<br />

detected in our patients with MPNs in accordance with literature<br />

data, <strong>and</strong> therefore should be incorporated in the<br />

diagnostic evaluation of patients with suspected MPNs.<br />

Further analysis should focus on contribution of the JAK2-<br />

V617F mutation in the clinical phenotype of patients with<br />

distinct subgroups of MPNs. These correlations imply that<br />

detection of this mutation will not only have a diagnostic<br />

value, but also a role in treatment given the development of<br />

STAT/JAK patway inhibiting drugs.<br />

PP30 Frequency <strong>and</strong> Clinical Correlates of JAK2<br />

46/1 Haplotype in Essential<br />

Thrombocythemia: Single Center<br />

Experience<br />

Panovska-Stavridis I, Matevska N, Ivanovski M,<br />

Trajkova S, Dukovski D, Pivkova Veljanovska A,<br />

Cevreska L, Dimovski A. Department of Hematology,<br />

Medical Faculty, Skopje, Republic of Macedonia<br />

Background: It is predicted that the inherited genetic<br />

background in the individual patients with myeloproliferative<br />

neoplasm (MPN) influences the disease susceptibility<br />

<strong>and</strong> the phenotype expression of the MPN. Several groups<br />

discovered that the germline JAK2 haplotype, tagged by the<br />

»C« allele of single nucleotide polymorphism (SNP)<br />

rs12343867 (C/T) is associated with the JAK2V617F positive<br />

MPN. Also, some recent studies showed equal distribution<br />

of this SNP among JAK2V617F negative MPN <strong>and</strong><br />

questioned the role of this hyplotype in MPN patients.<br />

Aim: In order to extend further those observations we<br />

conduct a retrospective study. First, we assess the frequency<br />

of JAK2 46/1 haplotype in Essential thrombocyhemia (ET)<br />

patients in comparison with population controls. As second<br />

we evaluate the association of 46/1 with the JAK2V617F<br />

mutational status <strong>and</strong> the clinical characteristics in the series<br />

of patients with ET that were diagnosed <strong>and</strong> treated at<br />

the University Clinic of hematology-Skopje, Republic of<br />

Macedonia.<br />

Patients <strong>and</strong> methods: Ninthly five consecutive patients<br />

diagnosed with ET according to proposed criteria for diag-<br />

102<br />

nosis in 2008 by the World Health Organization were included<br />

in our study. The 46/1 tag SNP rs12343867 (C/T)<br />

was genotyped on a MxPro 3005P real-time PCR system<br />

(Stratagene, La Jolla, CA,USA) using the TaqMan SNP genotyping<br />

assay (Applied Biosystems, Foster City, CA,<br />

USA) according to the manufacturer’s instructions.<br />

Results: The incidence of 46/1-linked C allele was significantly<br />

higher in ET (genotype: CC 13%, CT 60%, TT<br />

27%; C-allele frequency: 43,7) than in population control<br />

(C-allele frequency 29%), P


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

resection.Three patients treated with CT underwent haematopoietic<br />

stem cell transplantation (HSCT), autologous in<br />

one, <strong>and</strong> allogeneic from related donor in other two patients,<br />

Tottaly, five patients died. Median group overall survival<br />

was 28 months. Mean survival time in the group of patients<br />

treated with chemotherapy was 22.1 months. Within the<br />

group of patients initially treated with surgical resection<br />

who were followed later on, mean survival time was 25.2<br />

months. No significant difference in survival was observed<br />

between these two groups of patients (p>0.05). Mean survival<br />

time to systemic leukemia development was 21.9<br />

months.<br />

Conclusion: According to the results in our series of patients,<br />

it is clear that in cases of suspected MS, employment<br />

of wide spectrum of antibodies during immunohistochemical<br />

work-up of tumor tissue is m<strong>and</strong>atory. Also, optimal<br />

treatment is not defined, in part because of different clinical<br />

presentation. Chemotherapy, HSCT, surgical resection, or<br />

combination of these approaches, is used depending on actual<br />

case.<br />

PP32 The Risk of Hematopoetic Malignancy<br />

in Children Born after in vitro Fertilization<br />

Bilic E, Stemberger L, Slipac J, Stojsavljevic S, Bilic E,<br />

Konjevoda P, Konja J, Femenic R, Rajic Lj. Department<br />

of Pediatrics <strong>and</strong> Departmen of Neurology, Clinical<br />

Hospital Center <strong>and</strong> School of Medicine, University<br />

of Zagreb, Croatia<br />

Infertility is one of the major health <strong>and</strong> social problems<br />

where assisted reproductive technology such as in vitro fertilization<br />

(IVF) can offer an efficient solution. Evaluation of<br />

children conceived by IVF is neccessary to determine the<br />

safety of the method. Hematopoetic tumors (leukemia <strong>and</strong><br />

lymphoma) <strong>are</strong> the most common childhood cancers. Some<br />

study found an association with IVF <strong>and</strong> increased risk for<br />

acute lymphoblastic leukemia (ALL) <strong>and</strong> non-Hodgkin<br />

lymphoma (NHL) in children.<br />

Aim: The aim of present study was to determine whether<br />

there is an increased risk for hematopoetic tumors in children<br />

conceived by IVF in comparison to children born after<br />

natural conception.<br />

Patients <strong>and</strong> methods: The study included 550 children<br />

treated for leukemia or lymphoma (345 ALL, 67 acute myeloblastic<br />

leukemia (AML), 63 Hodgkin lymphoma <strong>and</strong> 75<br />

NHL) in the Department of Pediatric, University of Zagreb,<br />

School of Medicine Zagreb, Croatia, between 1994. <strong>and</strong><br />

2009.y.<br />

Results: Three children of 550 included in the study were<br />

born after IVF. Two suffered from ALL <strong>and</strong> one from AML<br />

Conclusion: Results of our study showed no increased<br />

risk for childhood leukemia <strong>and</strong> lymphoma in children conceived<br />

by IVF in comparison to children conceived naturally.<br />

The main limitation of our, <strong>and</strong> most of other similar<br />

studies was the sample size not large enough to decline the<br />

discrepancy of literature data <strong>and</strong> to provide an unambiguous<br />

answer to the question of increased risk of hematopoetic<br />

tumors in children concieved by IVF<br />

PP33 H1N1 Infl uenza pneumonia in patient with<br />

chronic lymphocytic leukemia – case report<br />

Vince A 1 , Barsic B 1 , Dusek D 1 , Vrhovac R 2 , Kutlesa M 1 ,<br />

Santini M 1 . 1 University Hospital for Infectious Diseases<br />

»Dr. Fran Mihaljevic«, Zagreb, Croatia, 2 Department of<br />

Medicine, Division of Hematology, University Hospital<br />

»Merkur« Zagreb, Croatia<br />

Initial experience with the 2009 H1N1-Influenza A<br />

(H1N1) suggests that this virus can cause severe disease in<br />

immunosuppressed patients such as influenza pneumonia<br />

<strong>and</strong> respiratory failure.<br />

We present a case of 41-year old man with chronic lymphocytic<br />

leukemia (CLL) who was treated at the University<br />

Hospital for Infectious Diseases in February 2011 because<br />

of influenza pneumonia. The patient was first diagnosed in<br />

2008, with small tumor mass (TTM 5.7) CLL, Rai stage I.<br />

His phenotype was typical for B-CLL, <strong>and</strong> due to low CD38<br />

expression <strong>and</strong> trisomy 12 he was not regarded as a patient<br />

with adverse prognosis. Although his tumor mass gradually<br />

increased over time, he never required specific treatment.<br />

Patient was not vaccinated against influenza although recommended<br />

by his hematologist. Present illness started on<br />

2/11/2011 with dry cough <strong>and</strong> low-grade temperature. He<br />

was initially prescribed clarithromycin. On 2/16/2011 patient<br />

became febrile up to 40 C with shaking chills <strong>and</strong> productive<br />

cough. He was admitted to small local hospital on<br />

2/17/2011 because of bilateral pneumonia. Upon admission,<br />

the patient was febrile (40 C), with decreased breath sounds<br />

on the left side with fine crackles bilaterally. The remainder<br />

of physical examination was unremarkable. Therapy with<br />

oseltamivir, ceftriaxone <strong>and</strong> doxycycline was commenced.<br />

On the fifth day of hospital stay patient’s condition started<br />

to deteriorate with development of respiratory insufficiency<br />

<strong>and</strong> he was transferred to Intensive C<strong>are</strong> Unit (ICU) where<br />

mechanical ventilation was started. Ceftriaxone was changed<br />

to meropenem <strong>and</strong> vancomycin; doxycycline <strong>and</strong> oseltamivir<br />

were continued. Unfortunately, respiratory insufficiency<br />

worsened <strong>and</strong> extracorporeal membrane oxygenation was<br />

considered as only life-saving procedure. He was transferred<br />

to ICU of University Hospital for Infectious Diseases<br />

on the twelfth day of illness. Upon admission patient was<br />

mechanically ventilated (fiO2 100%) <strong>and</strong> hypoxemic (satO2<br />

80%). Pulmonary X-ray showed bilateral confluent, mixed<br />

alveolar <strong>and</strong> interstitial infiltrates, consistent with acute respiratory<br />

distress syndrome (ARDS). ECMO was started<br />

upon admission. On the seventh day of hospitalization there<br />

was further deterioration of patient’s condition, probably<br />

because of nosocomial sepsis with development of septic<br />

shock <strong>and</strong> multiple organ dysfunction syndrome. Patient<br />

died on the tenth day of hospitalization, i.e. twenty-first day<br />

of influenza illness. Diagnosis of influenza A-H1N1 was<br />

confirmed by RT-PCR from tracheal aspirate.<br />

CLL patients <strong>are</strong> particularly at risk for influenza complications<br />

because of hypogammaglobulinemia <strong>and</strong> deficient<br />

T-cell compartment. Therefore physicians should strongly<br />

encourage their patients to get influenza vaccine. Otherwise,<br />

during influenza season empirical therapy with oseltamivir<br />

should be started at the first symptoms of influenza.<br />

103


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

PP34 Treatment of paroxismal nocturnal<br />

hemoglobinuria – a case report<br />

Boban A 1 , Serventi Seiwerth R 1 , Basic-Kinda S 1 , Peric Z 1 ,<br />

Dubravcic K 2 , Batinic D 2 , Labar B 1 . 1 Division of<br />

Hematology, Department of Medicine, 2 Department of<br />

Clinical Laboratory, Clinical Hospital Center <strong>and</strong> School<br />

of Medicine, University of Zagreb, Croatia<br />

Introduction: Paroxismal nocturnal hemoglobinuria<br />

(PNH) is clonal acquired disorder characterized by hemolytic<br />

anemia, thrombosis, fatigue <strong>and</strong> bone marrow failure.<br />

It is caused by acquired mutation of PIG-A gene in mutipotent<br />

hematopoietic stem cell. As a consequence, cells <strong>are</strong><br />

lacking glycosil phosphatidylinosiol (GPI) – anchored proteins,<br />

namely CD55 <strong>and</strong> CG 59, which <strong>are</strong> complement<br />

regulatory proteins. The lack of complement regulatory proteins<br />

makes erythrocytes susceptible to both intravascular<br />

<strong>and</strong> extravascular hemolysis. Intravascular hemolysis is responsible<br />

for majority of morbidity form the disease. Eculizumab<br />

is a monoclonal antibody that inhibits terminal complement<br />

activation, which reduces intravascular hemolysis<br />

in PNH patients.<br />

Aim: Here we present the first PNH patient in our Center<br />

treated with eculizumab.<br />

Case presentation: The patient was diagnosed having<br />

aplastic anemia in 2009, at the age of 21. The treatment<br />

started with corticosteroids, <strong>and</strong> later altered into cyclosporine.<br />

In 2005 the patient presented with severe headache <strong>and</strong><br />

transitory ischemic attack. At that point PNH was suspected,<br />

<strong>and</strong> diagnostic procedure was carried out. Diagnose of<br />

PNH was made based on PNH clone on flow cytometry. In<br />

2007, the progression of intravascular hemolysis was noted,<br />

with significant increase in bilirubin <strong>and</strong> lactate dehidrogenase<br />

levels, followed by the signs of bone marrow failure.<br />

The need for blood transfusions <strong>and</strong> fatigue increased, while<br />

quality of life decreased. Allogenic stem cell transplantation<br />

was indicated, but no related donor was found. The patient<br />

was eligible for eculizumab treatment.<br />

The treatment with eculizumab started in January 2010.<br />

Patient was vaccinated for Naisseria meningitides, <strong>and</strong> developed<br />

severe cellulitis with skin necrosis on the place of<br />

the puncture, hence the reconstructive surgery of the left upper<br />

arm was needed. At the time when eculizumab was<br />

started, patient had severe hemolysis, thrombocytopenia,<br />

<strong>and</strong> anemia with need of blood transfusions every 2–3 weeks<br />

<strong>and</strong> secondary hemochromatosis. Subjectively, she complained<br />

mostly on severe fatigue. The patient received induction<br />

treatment without side effects <strong>and</strong> continued with<br />

maintenance treatment for 19 months. A significant decrease<br />

of hemolysis was noted, <strong>and</strong> consequently reduced<br />

need for transfusions <strong>and</strong> disappearance of symptoms,<br />

mainly fatigue with dramatic improvement in her quality of<br />

life.<br />

Conclusion: Eculizumab treatment significantly improved<br />

quality of life of the PNH patient form our center. Patients<br />

main symptom, fatigue, disappe<strong>are</strong>d almost completely,<br />

<strong>and</strong> rate of blood transfusion was reduced to once in<br />

5–6 weeks. We found eculizumab effective in treatment of<br />

PNH patients.<br />

104<br />

PP35 Anti-proliferative <strong>and</strong> pro-apoptotic action<br />

of the combined cytostatic/<br />

immunosuppressive treatment of myeloma<br />

cell lines in vitro<br />

Zelic A 1 , Ivcevic S 1 , Kovacic N 1 , Kusec R 2 , Grcevi} D 1 .<br />

1 Zagreb University School of Medicine; 2 Dubrava<br />

University Hospital, Zagreb, Croatia<br />

Multiple myeloma (MM) is B-lymphocyte neoplasia,<br />

characterized by the slow proliferation of malignant plasma<br />

cells in the bone-marrow (BM). Multiple anti-apoptotic signaling<br />

mechanisms contribute to the accumulation of myeloma<br />

cells within the BM <strong>and</strong> account for their resistance<br />

to chemotherapy. Therefore, investigation of the effects of<br />

different agents on the balance between pro- <strong>and</strong> anti-apoptotic<br />

factors in the malignant clone would contribute to the<br />

development of novel therapeutic strategies in MM.<br />

Aim: We determined the effects of several currently used<br />

anti-myeloma agents <strong>and</strong> their possible additive action on<br />

the inhibition of proliferation <strong>and</strong> enhancement of apoptosis<br />

of different human myeloma cell lines.<br />

Material <strong>and</strong> methods: Human myeloma cell lines NCI-<br />

H929, RPMI 8226 <strong>and</strong> JJN3 (ADCC-LGC <strong>and</strong> DSMZ cell<br />

line collection) <strong>and</strong> Theil (a gift from Dr. K. Pulford, University<br />

of Oxford, UK) were treated with bortezomib (Millenium<br />

Pharmaceuticals), dexamethasone, cyclosporine <strong>and</strong><br />

thalidomide (Sigma-Aldrich) for 48 hrs. Cell line proliferation<br />

was assessed by the colorimetric MTT assay, whereas<br />

viability <strong>and</strong> apoptosis were evaluated by annexin V/propidium<br />

iodide staining. Gene expression of pro-apoptotic<br />

(Bax <strong>and</strong> p21) <strong>and</strong> pro-survival molecules (Bcl-2 <strong>and</strong> c-<br />

Myc) was assessed by real-time AB7500 instrument (Applied<br />

Biosystems). Additionally, we assessed the expression<br />

of SPARC (secreted protein acidic <strong>and</strong> rich in cysteine), involved<br />

in the cell de-adhesion from BM matrix <strong>and</strong> tumor<br />

pathogenesis.<br />

Results: Anti-myeloma agents (dexamethasone, bortezomib<br />

<strong>and</strong> thalidomide) achieve their effects by different cellular<br />

mechanisms. In addition, cyclosporine was included<br />

for its ability to increase cell sensitivity to other cytostatics.<br />

Each cell line, representing different biological variant of<br />

MM, has shown unique pattern of the expression of pro- <strong>and</strong><br />

anti-apoptotic genes <strong>and</strong>, in addition, responded differently<br />

to anti-myeloma treatment. The most potent additive effect<br />

of drug combinations, including thalidomide/bortezomib,<br />

thalidomide/cyclosporine <strong>and</strong> bortezomib/cyclosporine,<br />

was noticed on the cellular <strong>and</strong> molecular level, as suppressed<br />

proliferation <strong>and</strong> shift in the balance of pro- <strong>and</strong><br />

anti-apoptotic genes.<br />

Conclusion: Resistance to apoptosis, common in myeloma<br />

cells, depends on the constitutive expression of molecules<br />

acting at cell-cycle checkpoints <strong>and</strong> activated intracellular<br />

pathways in the particular circumstances. We observed<br />

that the susceptibility of myeloma cells to specific<br />

treatment is determined by their intrinsic balance between<br />

pro-survival <strong>and</strong> pro-apoptotic factors. Certain drug combinations,<br />

specifically thalidomide or bortezomib with cyclosporine,<br />

decreased Bcl-2/Bax ratio, a key indicator of enhanced<br />

apoptosis. Our findings suggest that the insight into


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

the molecular events in the affected clone could enable administration<br />

of a specific individualized therapy which<br />

would be more effective in eradicating the malignant clone.<br />

PP36 T<strong>and</strong>em Transplantation in Multiple<br />

Myeloma – Single Center Experience<br />

Batinic J 1 , Durakovic N 1 , Sertic D 1 , Bojanic I 2 , Batinic D 3 ,<br />

Golubic-Cepulic B 2 , Mazic S 2 , Radman Livaja I 1 ,<br />

Basic-Kinda S 1 , Aurer I 1 , Labar B 1 , Nemet D 1 . 1 Division of<br />

Hematology, Department of Medicine, 2 Department of<br />

Transfusiology, 3 Deaprtment of Clinical Laboratory,<br />

Clinical Hospital Center <strong>and</strong> School of Medicine,<br />

University of Zagreb, Croatia<br />

High-dose chemotherapy followed by autologous hematopoetic<br />

stem cell transplantation is well established first<br />

line therapy for multiple myeloma. It has been previously<br />

shown that t<strong>and</strong>em transplantation carries survival benefit<br />

when comp<strong>are</strong>d to a single transplantation. We have conducted<br />

a trial of two successive high-dose chemotherapy<br />

followed by peripheral blood stem cell rescue for patients<br />

with advanced stage multiple myeloma.<br />

Aim: Aim was to evaluate feasibility, efficacy <strong>and</strong> toxicity<br />

of t<strong>and</strong>em transplantation.<br />

Patients <strong>and</strong> Methods: We included 107 patients, median<br />

age 54 years (32–66), 49 were female, <strong>and</strong> 58 male<br />

patients. Most of the patients had IgG monoclonal protein<br />

(54.2%), 17.7% patients had IgA monoclonal protein,<br />

17.7% had light chain myeloma, while 9.3% had non-secretory<br />

myeloma, <strong>and</strong> one patient had IgD monoclonal protein.<br />

Patients were evaluated on a intention to treat basis, <strong>and</strong><br />

from 107 included patients, 83 (77.5%) received t<strong>and</strong>em<br />

transplantation; 4 patients did not collect adequate number<br />

of cells for two transplantations, 10 patients had low performance<br />

score, 7 were not transplanted due to physician decision,<br />

1 patient refused second transplantation, 1 patient relapsed<br />

<strong>and</strong> for 1 patient the reason is unknown.<br />

Results: Progression free survival (PFS) <strong>and</strong> overall survival<br />

(OS) were evaluated. Median time of progression free<br />

survival for the entire group was 77 months, while median<br />

time of overall survival was 138 months. Prior to first transplantation<br />

8.6% of patients were in complete remission or<br />

very good partial remission (CR/VGPR), 78% were in partial<br />

remission (PR) <strong>and</strong> 11.4% had minimal response, while<br />

1.9% had progressive disease. After the first transplantation<br />

42.3% achieved CR/VGPR <strong>and</strong> 50% of patients were in PR,<br />

while after the second transplantation 71.8% of patients<br />

were in CR/VGPR <strong>and</strong> 26.2% were in PR. We conducted<br />

analysis of PFS <strong>and</strong> OS according to disease status prior to<br />

second transplantion <strong>and</strong> found no significant difference between<br />

patients that achieved CR or VGPR <strong>and</strong> the patients<br />

that did not do so. However, when the same analysis was<br />

done according to disease status 6 months after the second<br />

transplantation, a significant difference in both PFS <strong>and</strong> OS<br />

was found between patients that achieved CR/VGPR <strong>and</strong><br />

the patients that did not achieve CR/VGPR (p=0.0179 <strong>and</strong><br />

p=0.0056 respectively).<br />

Conclusion: T<strong>and</strong>em transplantation as first line treatment<br />

for multiple myeloma is an effective therapy offering<br />

patients long term progression free survival <strong>and</strong> overall survival.<br />

We found no significant difference in long term PFS<br />

<strong>and</strong> OS between patients that achieved CR/VGPR prior to<br />

second transplantation <strong>and</strong> those that did not, indicating important<br />

therapy efficacy of second transplantation. Also, patients<br />

achieving CR/VGPR after second transplantation<br />

have statistically better PFS <strong>and</strong> OS than patients failing to<br />

do so, which emphasizes importance of achieving CR as a<br />

final outcome.<br />

PP37 The Role of Thalidomide as Maintenance<br />

Therapy in Multiple Myeloma<br />

Bila J 1,2 , Bodrozic J 2 , Todorovic M 1,2 ,<br />

Sefer D 2 , Kraguljac N 2 , Antic D 2 , Andjelic B 2 , Elezovic I 1,2 ,<br />

Tomin D 1,2 , Gotic M 1,2 , Mihaljevic B 1,2 . 1 Medical Faculty,<br />

University of Belgrade, 2 Clinic of Hematology, CCS,<br />

Belgrade, Serbia<br />

The aim of study was to analyse results of thalidomide<br />

maintenance in patients with multiple myeloma.<br />

Patients <strong>and</strong> methods: The study included 130 newly<br />

diagnosed patients: 1) 50pts treated with high-dose treatment<br />

(HDT) <strong>and</strong> autologous stem cell transplant (ASCT, 29<br />

male/21 female, mean age 53 yrs, range 41–65); <strong>and</strong> 2) 80<br />

elderly pts (45 male/35 female, mean age 68 yrs, range 66–<br />

83yrs); with IgG myeloma in 75pts; IgA 27pts; light chains<br />

25pt; non-secretory 3pts. Distribution according to the clinical<br />

stage <strong>and</strong> ISS score was as follows: 1) II 56pts; III<br />

74pts; ISS3 in <strong>and</strong> 59pts with ISS3 score. Induction treatment<br />

in the first group was: a) VAD (average 4 cycles, range<br />

3–6 cycles in 35/50pts); b) CTD (average 4 cycles, range<br />

3–6 cycles in 15/50pts), followed by Melphalan 200mg/m²<br />

<strong>and</strong> ASCT. The maintenance therapy was: Alfa-Interferon<br />

(aIFN) in 10/35pts after VAD; Thalidomide (100mg/day) in<br />

35/50pts after VAD/CTD. Median duration of maintenance<br />

was 19m (range 6–36 m). Initial treatment for the elderly<br />

pts: a) MP (30/80pts); b) MPT (35/80pts); c) Thal-Dex<br />

(15/80pts). The maintenance therapy was: Alfa-Interferon<br />

(aIFN) after MP (30/30pts); Thalidomide (100mg/day) after<br />

MPT/Thal-Dex (50/80pts). Median duration of maintenance<br />

was 16m (range 4–28 m).<br />

Results: In the first pts group, after VAD, VGPR was<br />

achieved in 4/35pts; PR in 25/35pts. Following HDT,<br />

7/35pts achieved CR; VGPR 9/35pts; PR 14/35pts. After<br />

CTD induction, CR existed in 3/15pts; VGPR/PR in 8/15pts.<br />

Median follow-up was 26m (range 18–60 m). Thalidomide<br />

maintenance after CTD significantly improved 3-yrs probability<br />

of relapse-free survival (VAD+HDT+aIFN: RFS<br />

25% vs. VAD+HDT+Thal: RFS 37% vs. CTD+HDT+Thal:<br />

RFS 51%, P<br />

Conclusion: Thalidomide as maintenance therapy significantly<br />

improves treatment results in myeloma patients<br />

with limitations due to toxic effects.<br />

105


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

PP38 Modifi cated dosing of the VMP /Velcade +<br />

melphalan + prednisone/ regime in multiple<br />

myeloma /MM/ patients<br />

Masarova K, Stefanikova Z, Mistrik M. Department of<br />

Hematology <strong>and</strong> Transfusiology, University Hospital,<br />

Bratislava, Slovakia<br />

Aim: To establish efficacy <strong>and</strong> sefety modificated dosing<br />

of VMP regime once weekly in MM patients not eligible for<br />

autologous stem cell transplantation /ASCT/.<br />

Abstract: In september 2008, J. San Miguel et al. published<br />

the results of the study Phase III VISTA comparing<br />

treatment regime VMP /Velcade+melphalan + prednisone/<br />

vs MP in newly diagnosed patients with MM not eligible for<br />

ASCT. In the VISTA study bortezomib administration was<br />

1,3 mg/m 2 in 9 six weeks cycles, in the first 4 cycles the<br />

administration was more intense /day 1,4,8,11,22,25,29,32/<br />

<strong>and</strong> the following 5 cycles the administration of bortezomib<br />

in days 1,8,22,29. This study confirmed significantly longer<br />

medium OS /medium overal survival/ of patients in the<br />

VMP arm. ORR in this arm was 71% /CR: 31%, PR: 40%/,<br />

existence of negative adverse events of grade 3 /4 was: neutrophenia<br />

40%, trombocytophenia 37%, anaemia 19%,<br />

polyneuropathy /PN/ 13%. In 2008 A.Palumbo at al. published<br />

a study which comp<strong>are</strong>d effectiveness <strong>and</strong> safety of<br />

the VMP regime vs. VMPT in newly diagnosed patients not<br />

eligible for ASCT. In this study, at the beginning the bortezomib<br />

administration was as in the VISTA study, but in<br />

march 2007 the protocol was changed <strong>and</strong> the administration<br />

of bortezomib was changed to 9 five weeks cycles /1,3<br />

mg/m 2 in days 1,8,15,22/. The effectiveness of bortezomib<br />

remained unchanged /ORR: 81%, CR: 21%/ but the adverse<br />

events, especially PN of grade 3/4 /2%/ decreased <strong>and</strong> also<br />

decreased the amount of the treatment discontinuations<br />

/10%/.<br />

Published results led us to administration VMP regime in<br />

treatment of MM in patients not eligible for ASCT in five<br />

weeks cycles with administration of bortezomib 1x weekly<br />

/1,3 mg/m 2 in days 1,8,15,22/, melphalan 9mg/m 2 <strong>and</strong> prednisone<br />

60 mg/m 2 in days 1–4. Since April 2009 we have<br />

collected group of 33 patients with median line of previous<br />

MM treatment 3. Currently is possible to evauluate 33 patients<br />

who absolved min. 4 cycles of previous treatments. In<br />

this patients we achieved CR in 33%, PR in 42%, ORR in<br />

75% with no PN of grade 3 /4. No patient had to discontinuate<br />

or terminate treatment due to adverse events. Median of<br />

administered cycles was 6,9 cycles. Median TTP /time to<br />

progression/ was 25,7 months /VISTA 24 months/. Median<br />

OS was not yet reached /VISTA not yet reached/.<br />

Conclusion: On the base of our experience we can confirm<br />

that the effectiveness of modificated VMP regime in<br />

MM patients not eligible for ASCT remained stable <strong>and</strong> we<br />

did not observed any adverse events of grade 3/4 or discontinuations<br />

of the treatment due to adverse events. Decreased<br />

frequency of bortezomib administration was well tolerated<br />

also by the patients who were comming for the administration<br />

once weekly.<br />

106<br />

PP39 Tevagrastim <strong>and</strong> Plerixafor as fi rst line<br />

mobilizing regimen in patients with<br />

lymphoma <strong>and</strong> Multiple Myeloma<br />

Andreola G., Babic A., Negri M., Drera M., Martinelli G.,<br />

Laszlo D. Department of Haematology, European Institute<br />

of Oncology, Milan, Italy<br />

Patients affected by hematologic malignancies might benefit<br />

from high dose chemotherapy followed by peripheral<br />

stem cells (PBSC) transplant. Chemotherapy in combination<br />

with G-CSF is effective in mobilizing stem cells but<br />

often toxic, might require prolonged hospitalization <strong>and</strong> extensive<br />

supportive c<strong>are</strong>. Moreover a high proportion of patients,<br />

ranging from 11 to 53%, fail to collect an adequate<br />

number of stem cells with this approach. Plerixafor, a<br />

CXCR4 chemokine antagonist, has shown to increase the<br />

number of circulating CD34+ cells in cancer patients when<br />

used alone or with G-CSF <strong>and</strong> to be able to rescue patients<br />

unable to mobilize with traditional regimens. Recently several<br />

forms of biosimilar nonglycosylated recombinant human<br />

G-CSF have been clinically developed <strong>and</strong> approved<br />

by the European Medicines Agency for the same indications<br />

as the reference filgrastim product on the basis of comparable<br />

quality, efficacy <strong>and</strong> safety. Biosimilars also represent<br />

a more cost-effective strategy <strong>and</strong> their use in clinical setting<br />

may provide cost savings in their indicated uses. In order<br />

to test the efficacy of the combination of biosimilar version<br />

of G-CSF (Tevagrastim) <strong>and</strong> plerixafor in mobilizing<br />

PBSC, from 12/10 to 5/11, 11 patients, affected by Non-<br />

Hodgkin Lymphoma (3), Hodgkin Disease (2) <strong>and</strong> Multiple<br />

Myeloma (MM) (6), were treated as follows: Tevagrastim<br />

(10µg/kg/die) was self administered for 3 days; on day 4<br />

patients were admitted to the hospital, circulating CD34+<br />

cells counted <strong>and</strong> if 500 of 12 (9–13) <strong>and</strong> of PLT>20.000 of<br />

13 (9–19) days.<br />

The combination of tevagrastim <strong>and</strong> plerixafor is safe <strong>and</strong><br />

effective in mobilizing PBSC <strong>and</strong> allows a collection of a<br />

more than adequate number of cells in most of the patients<br />

in a maximum of 2 apheresis procedures, even in patients<br />

with MM who need to collect a double amount of cells.<br />

PP40 Does Prelixafor do the trick in mobilising<br />

Stem Cells?<br />

Vieira S, Monroe D, Librojo M. London, United Kingdom<br />

Abstract: Plerixafor (MOZOBIL) is an antagonist of alpha-chemokine<br />

receptor CXCR <strong>and</strong> one of the new additions<br />

to Haematology discipline <strong>and</strong> it is used as a stem cell<br />

mobiliser. CXCR4 alpha-chemokine receptors <strong>are</strong> important<br />

in a hematopoietic stem cell homing to the bone marrow<br />

<strong>and</strong> in hematopoietic stem cell quiescence. It is indicated<br />

in combination with Granulocyte Colony Stimulating<br />

Factor (GCSF) for Peripheral Blood Stem Cell (PBSC) collections<br />

in the Multiple Myeloma (MM) <strong>and</strong> Lymphoma<br />

patient group with a poor mobilisation history<br />

Patients <strong>and</strong> methods: In our organisation, data was collected<br />

prospectively. 14 patients received Plerixafor for<br />

PBSC collection between May 2009 <strong>and</strong> September 2010.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

All patients except one met the criteria prior to treatment<br />

(White Blood Count> 2.5 x 109/L, Absolute Neutrophil<br />

Count > 1.5 x 109/L, Platelet> 85 x 109/L, Serum Creatinine<br />

< 1.5 mg/dL, Aspartate transaminase (AST), Alanine<br />

Transaminase (ALT), Bilirubin < 2 x ULN with no evidence<br />

of Hepatitis B <strong>and</strong> C. One patient had low platelet count.<br />

Patient demographic data is shown in Table 1. Three patients<br />

had an underlying documented medical problem (cortochondritis<br />

<strong>and</strong> glaucoma).<br />

Results: Nine of the patients were diagnosed with MM<br />

<strong>and</strong> three with Non-Hodgkin’s Lymphoma (NHL), two patients<br />

with Hodgkin’s Disease <strong>and</strong> one with Neuroblastoma.<br />

All patients received four consecutive days of GCSF 10 ľg/<br />

kg prior to Plerixafor. Plerixafor was given 10 hours before<br />

the PBSC collection <strong>and</strong> GCSF was repeated one hour prior<br />

to PBSC collection time in the morning. All patients received<br />

0.24mg/kg/day of Plerixafor. There were no side effects<br />

observed. Only one patient required a second dose of<br />

Plerixafor.<br />

For all cases, the target CD34+ count was 4 x 106 cells/<br />

kg of recipient body weight. Two patients responded extremely<br />

well with a CD34+ count of 21.79 x 106 cells/kg<br />

<strong>and</strong> 15.16 x 106 cells/kg achieved <strong>and</strong> six patient had<br />

achieved the target of 4 X 106 cells/kg or above with positive<br />

outcome. However seven patients failed to achieve to<br />

reach target CD+ 34 4 X109cells/kg of CD34 count was<br />

collected. Six of these patients had adequate amount of<br />

CD34 count for an autologus transplant. Only one patient<br />

failed to mobilise any CD34 at all.<br />

Conclusion: Plerixafor was used at The London Clinic<br />

recently with mostly favourable outcomes. According to<br />

this study Plerixafor has improved PBSC outcomes with<br />

one exception case. Prelixafor definitely made a positive<br />

difference 54% of our patient <strong>and</strong> 40% of our patient had<br />

enough cells for an aoutologus transplant. However, results<br />

<strong>are</strong> inconclusive due to small patient numbers. Ongoing<br />

studies with new patient experiences <strong>are</strong> needed. More than<br />

one dose of Plerixafor maybe required for heavily pre-treated<br />

patients who <strong>are</strong> historically poor mobiliser of PBSC<br />

collections.<br />

PP41 Multidrug Resistance: From Research<br />

to Clinics<br />

Tauber Jakab K 1 , Márki-Zay J 1 , Kis E 1 , Udvardy M 2 ,<br />

Borbényi Z 3 , Dávid M 4 , Krajcsi P 1 . 1 Solvo Biotechnology,<br />

Szeged, Hungary; 2 2nd Department of Internal Medicine,<br />

University of Debrecen-Medical <strong>and</strong> Health Science<br />

Center, Debrecen, Hungary; 3 2nd Department of Medicine<br />

<strong>and</strong> Cardiology, University of Szeged – Albert Szent-<br />

Györgyi Clinical Center, Szeged, Hungary; 4 1st<br />

Department of Internal Medicine, Pécs, Hungary<br />

Since the beginning of the 90’s several studies have confirmed<br />

the role of multidrug transporter proteins as major<br />

mechanism of chemoresistance in a variety of haematological<br />

malignancies. This phenomenon usually results from the<br />

overexpression of ATP-binding cassette (ABC) transporters,<br />

such as ABCB1 (MDR1 or P-gp), ABCC1 (MRP1), <strong>and</strong><br />

ABCG2 (MXR or BCRP), which <strong>are</strong> known to function as<br />

drug efflux pumps causing drug resistance by actively extruding<br />

multiple anticancer drugs with expenses of ATP. According<br />

to a number of multicentric trials, P-glycoprotein or<br />

ABCG2 expression <strong>are</strong> bad prognostic markers in acute myeloid<br />

leukemia (AML) regarding remission rate <strong>and</strong> overall<br />

survival.<br />

Aim: The primary goal of our study was to evaluate the<br />

frequency of the three clinically relevant ABC transporters<br />

in AML <strong>and</strong> chronic lymphoid leukemia (CLL) using a<br />

functional routine clinical laboratory flow cytometric assay.<br />

In addition, we have characterized several anticancer drugs<br />

for their interactions with efflux transporters using cellular<br />

<strong>and</strong> membrane-based assays.<br />

Patients <strong>and</strong> methods: In the frame of a multicentric<br />

trial, flow cytometric assays were performed using the MultiDrugQuant<br />

kit (MDQ kit) on CD45dim blast cells of 26<br />

AML patients <strong>and</strong> CD19+ 28 CLL patients prior to the initiation<br />

of chemotherapy. The MDQ kit applies the calcein-<br />

<strong>and</strong> mitoxantrone-assays for the measurement of the function<br />

of MDR1, MRP1 <strong>and</strong> BCRP transporters. Activities of<br />

the multidrug transporters <strong>are</strong> calculated from the difference<br />

between the geomean cellular fluorescent intensities<br />

determined w/o transporter specific inhibitors <strong>and</strong> <strong>are</strong> expressed<br />

as multidrug resistance activity factor (MAF) values.<br />

Substrate or inhibitory profiles of 20 anticancer drugs<br />

have been characterized using membrane-based ATPase <strong>and</strong><br />

vesicular transport assays <strong>and</strong> cellular dye efflux assays<br />

such as the Calcein- <strong>and</strong> Hoechst-assays. The results of<br />

these experiments have been correlated to the cytotoxicity<br />

of the compounds on control (HL60, PLB) <strong>and</strong> transporter<br />

overexpressing (HL60-MDR1, HL60 MRP1 <strong>and</strong> PLB-<br />

BCRP) cell lines.<br />

Results: The median age of AML <strong>and</strong> CLL pts was 55<br />

<strong>and</strong> 67 years, respectively. MAF values were above the cutoff<br />

value in 9 of the 26 AML <strong>and</strong> in 12 of the 28 CLL patients<br />

at enrollment. Results of the membrane-based <strong>and</strong><br />

cellular assays to test the drug-transporter interactions<br />

agreed well.<br />

Conclusion: Based on these preliminary data the MDQ<br />

kit is reliable <strong>and</strong> provides transporter specific quantitative<br />

results on the function of the MDR1, MRP1 <strong>and</strong> BCRP<br />

transporters in the target cells. Besides other prognostic factors<br />

measurement of transporter activity allows risk stratification<br />

of the AML or CLL patients. The high-throughput in<br />

vitro assays presented here enable to characterize the interactions<br />

of antineoplastic drugs with multidrug transporters.<br />

PP42 P-glycoprotein activity <strong>and</strong> Flt3 internal<br />

t<strong>and</strong>em duplication in 39 patients with<br />

acute myeloid leukemia<br />

Batinic D 1 , Perkovic S 1 , Zadro R 1 , Labar B 2 , 1 Department<br />

of Clinical Laboratory, 2 Division of Hematology, Clinical<br />

Hospital Center <strong>and</strong> School of Medicine, University of<br />

Zagreb, Croatia<br />

Aim: Flt3 internal t<strong>and</strong>em duplication (Flt3/ITD) <strong>and</strong><br />

high P-glycoprotein (Pgp) activity <strong>are</strong> adverse prognostic<br />

factors in patients with acute myeloid leukemia (AML). The<br />

107


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

aim of this study was to determine the association of Flt3/<br />

ITD, Pgp activity <strong>and</strong> CD34 antigen expression in AML.<br />

Patients <strong>and</strong> methods: Data on Flt3/ITD status <strong>and</strong> Pgp<br />

activity were retrospectively analyzed in a group of 39 patients<br />

with AML diagnosed <strong>and</strong> treated at the University<br />

Hospital Centre Zagreb between January 2010 <strong>and</strong> May<br />

2011. Patients with acute promyelocytic leukemia were excluded<br />

from the study. CD34 antigen expression was determined<br />

by routine flow cytometric immunophenotyping of<br />

AML blasts. Pgp functional activity was assessed using<br />

Rhodamine 123 (Rho123) as a fluorescent probe <strong>and</strong> Pgp<br />

substrate, <strong>and</strong> verapamil (Vpm) as an inhibitor of Pgp activity.<br />

Pgp was considered to be active if ratio of fluorescence<br />

intensity (RFI) (Vpm+/Vpm-) was ≥ 2,0. FLT3/ITD was detected<br />

by RT-PCR analysis (Nakao et al, Leukemia 1996).<br />

Results: Flt3/ITD was found in 7/39 (18%), Pgp activity<br />

in 22/39 (56%) <strong>and</strong> CD34 expression in 26/39 (67%) of all<br />

our patients. Flt3/ITD- Pgp- group accounted for 13/39<br />

(33%); Flt3/ITD+ Pgp- 4/39 (10%); Flt3/ITD- Pgp+ 19/39<br />

(49%); Flt3/ITD+ Pgp+ 3/39 (8%). Four out of seven (57%)<br />

Flt3/ITD+ patients did not express active Pgp, <strong>and</strong> only 3<br />

out of 22 (14%) Pgp+ patients were Flt3/ITD+, demonstrating<br />

no significant association between these two parameters<br />

(Fisher’s exact test; p=0.6765). In addition, Flt3/ITD status<br />

was not associated with CD34 expression (p=0.1938), in<br />

contrast to Pgp+ cases that were CD34+ positive in 19/22<br />

(86%) AMLs (p=0.0054).<br />

Conclusion: Our results confirm the published data on<br />

the association between Pgp activity <strong>and</strong> CD34 marker expression,<br />

<strong>and</strong> the lack of association between the Pgp activity<br />

<strong>and</strong> Flt3/ITD in patients with AML. One possible explanation<br />

for this mutual exclusion might be the origin of leukemic<br />

cells, i.e. more frequent finding of Flt3/ITD in CD34negative<br />

blasts of monocytic origin. Further analysis of<br />

Flt3/ITD <strong>and</strong> Pgp in well-defined subgroups of AML in a<br />

larger cohort of our patients should clarify the role of combined<br />

Pgp <strong>and</strong> Flt3/ITD determination in risk assessment<br />

<strong>and</strong> prognostic classification of patients with AML.<br />

PP43 The pretreatment risk factors <strong>and</strong><br />

importance of comorbidity index for overall<br />

survival, complete remission <strong>and</strong> early<br />

death in patients with acute myeloid<br />

leukemia<br />

Djunic I, Virijevic M, Novkovic A, Djurasinovic V,<br />

Colovic N, Vidovic A, Suvajdzic-Vukovic N, Tomin D,<br />

Clinic of hematology, Clinical Center Serbia, Belgrade,<br />

Serbia<br />

Aim: The aims of this study were to determine as the<br />

pretreatment risk factors for overall survival (OS), rate of<br />

complete remission (CR) <strong>and</strong> early death in patients with<br />

acute myeloid leukemia (AML), as to estimate the influence<br />

of comorbidity on patients’ outcome.<br />

Patients <strong>and</strong> methods: This single-center study involved<br />

184 patients with nonpromyelocytic AML with follow-up<br />

of 58 months. The following parameters were estimated as<br />

the risk factors for OS, CR <strong>and</strong> early death: age, WBC<br />

(=30x10 6 /L), level of serum lactate dehydrogenase (LDH)<br />

108<br />

more than 1.5 x upper limit of normal <strong>and</strong> expression of<br />

CD34 antigen on leukemic blasts (=10%). Performance status<br />

(PS) was evaluated by <strong>East</strong>ern Cooperative Oncology<br />

Group (ECOG), ranged 0–4 (=2). Cytogenetic risk group<br />

was assessed by recommendation of European LeukemiaNet<br />

(ELN). Comorbidities evaluated by using the hematopoietic<br />

cell transplantation-specific comorbidity index<br />

(HCT-CI). Patients treated by Medical Research Council<br />

(MRC) 12 regimen. Risk factors were identified using the<br />

univariate <strong>and</strong> multivariate analysis.<br />

Results: The mean patients age was 56 years, range 18–<br />

79. The most significant risk factor for poor OS was the<br />

adverse cytogenetics: p=0.014, relative risk (RR)=1.314<br />

(95% CI 1.057-1.633). The age >=55 years indicated as the<br />

most significant risk factor for poor rate of CR: p=0.001,<br />

RR=0.274 (95% CI 0.125-0.597). The most significant factor<br />

for early death was the HCT-CI>=3: p=0.027, RR=4.310<br />

(95% CI 1.186-15.665). The cut-off of the age >=55 years<br />

was significant for poor OS <strong>and</strong> CR. The incidence of HCT-<br />

CI>=3 was significantly increased in patient with age >=55<br />

years than in patients younger than 55 years (p=3 had half<br />

of patient aged >=55 years, while 75% of patients younger<br />

than 55 years have HCT-CI=0. Mostly patients (87.9%)<br />

with age=1) had impact of infection on HCT-CI score (according<br />

HCT-CI definition). So, we estimated risk factors<br />

for both of these groups. The most significant risk factor for<br />

OS in group aged=55 years it was HCT-CI>=3: p=0.005,<br />

RR=1.874 (95% CI 1.208-2.907). In the group aged=55<br />

years, elevated serum LDH level was the most significant<br />

factor for poor rate of CR: p=0.015, RR=0.291, (95% CI<br />

0.107-0.790) <strong>and</strong> ECOG PS>=2: p=0.005, RR=0.730 (95%<br />

CI 0.586-0.910) for early death.<br />

Conclusion: This study identified the pretreatment risk<br />

factors for OS, CR <strong>and</strong> early death in patients with AML.<br />

We point out the importance of comorbidity for OS <strong>and</strong><br />

early death, as well as the impact of infection on outcome in<br />

patients with AML.<br />

PP44 Treatment of Adolescents with Acute<br />

Lymphoblastic Leukemia<br />

Konja J, Rajic Lj, Bilic E, Femenic R, Anicic M.<br />

Department of Pediatric, Clinical Hospital Center <strong>and</strong><br />

School of Medicine, University of Zagreb, Croatia<br />

Adolescents with acute lymphoblastic leukemia (ALL)<br />

have languished in the shadow of successful therapeutic<br />

outcome in childhood ALL. While 80% of children aged<br />

1–15 years <strong>are</strong> long-term survivors, less than 40% of adults<br />

<strong>are</strong> cured with current therapies. Adolescents <strong>and</strong> young<br />

adults who may be eligible for both adult <strong>and</strong> pediatric protocols<br />

have continued to have an intermediate outcome,<br />

which has remained inferior to that in children.<br />

Aim: This article has summarized the recent <strong>and</strong> updated<br />

retrospective comparative analysis of adolescents treated<br />

with pediatric <strong>and</strong> adult trials.<br />

Patients <strong>and</strong> methods: Data on ALL adolescents aged<br />

16–18 treated with pediatric trial ALL IC-BFM 2002 were<br />

comp<strong>are</strong>d with literature reports on the treatment of ALL<br />

adolescents according to adult protocols.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

Results: During the 2002–2006 period, 143 ALL patients<br />

aged 16–18 were treated with pediatric trial ALL IC-BFM<br />

2002. The 3-year event free survival (EFS) was 71%. During<br />

the 1997–2002 period, 67 ALL patients aged 15–17<br />

were treated with adult trial UKALLXII/E2993, <strong>and</strong> 61<br />

ALL age-matched patients were treated with pediatric trial<br />

MRC ALL97, with 5-year EFS of 49% <strong>and</strong> 65%, respectively.<br />

A similar 6-year EFS has been reported for the American<br />

pediatric regimen (CCG) that was by 26% higher in<br />

comparison with their adult regimen (Cancer <strong>and</strong> Leukemia<br />

Group B,CALCB).<br />

Conclusion: Therapeutic outcome in adolescents with<br />

ALL treated according to pediatric protocols is better (16 –<br />

26%) in comparison with adult regimen.<br />

PP45 FLAG-IDA as the Salvage Regime for<br />

Relapsed <strong>and</strong> Refractory Acute Leukemias<br />

Virijevic M, Suvajdzic N, Djunic I, Novkovic A,<br />

Djurasinovic V, Colovic N, Vidovic A, Tomin D. Clinic<br />

for hematology, Clinical Center Serbia, Belgrade, Serbia<br />

Aim: Relapsed <strong>and</strong> refractory acute leukemias (AL) have<br />

poor prognosis. There is no st<strong>and</strong>ard chemotherapy regimen<br />

that provides a durable second complete remission (CR).<br />

One of the most effective salvage regimen is FLAG-Ida.<br />

This study evaluated the efficiency <strong>and</strong> the toxicity of<br />

FLAG-Ida regimen in a relapsed <strong>and</strong> primary refractory AL.<br />

Patients <strong>and</strong> methods: This single-center study involved<br />

42 patients (pts) with AL with follow-up of 5 years. Twentyfour<br />

(52.4%) pts with acute myeloid leukemia (AML) <strong>and</strong><br />

18 (42.9%) with acute lymphoblastic leukemia (ALL) were<br />

included. All patients were treated with FLAG-Ida regimen<br />

(Fludarabin 30mg/m 2 days 1–5; Cytarabin 2000mg/m 2 days<br />

1–5; Idarubicin 8mg/m 2 days 1–3; G-CSF 300mg/m 2 was<br />

given subcutaneously from day 1 until neutrophil recovery).<br />

The following parameters were estimated as the risk factors<br />

for CR achievement <strong>and</strong> overall survival (OS): age, WBC<br />

(=30x10 9 /l), cytogenetics at diagnosis, refractory vs relapsed<br />

AL, early (in first 6–12 months after CR) vs late relapse<br />

(after 1 year) of disease. Cytogenetic risk in AML<br />

patients was according to European LeukemiaNet (ELN).<br />

Patients with ALL with unfavorable cytogenetics (Philadelphia<br />

chromosome or bcr-abl positivity) were considered as<br />

a high risk group. Event-free survival (EFS) <strong>and</strong> OS were<br />

calculated by Kaplan-Meier <strong>and</strong> differences log rank method.<br />

Toxicity was evaluated according to the World Health<br />

Organization (WHO) classification.<br />

Results: The mean pts’ age was 35 years, range 18–55;<br />

twenty-two (52.4%) had relapsed <strong>and</strong> 20 (47.6%) primary<br />

refractory AL. Sixteen of 42 (38.1%) patients achieved CR,<br />

10 (41.7%) pts with AML <strong>and</strong> 6 (33%) pts with ALL<br />

(p=0.03). Hematological toxicity (grade III-IV) occurred in<br />

all AL cases. The median time to neuthrophil recovery<br />

(ANC >500/µl) <strong>and</strong> platelet recovery (>20x10 9 /l) were 17<br />

<strong>and</strong> 20 days from the start of therapy. Nonhematological<br />

toxicity was mild (grade I-II). Febrile neutropenia was registered<br />

in 39 (90.5%) pts. Infection of unknown cause was<br />

registered in 21 (54.7%) <strong>and</strong> of known cause in 18 (46.3%)<br />

pts, respectively. The significant risk factor for poor CR rate<br />

in relapsed AL was WBC>=30x10 9 /l (p=0.043). The most<br />

significant risk factor for OS was early relapse vs late relapse<br />

(10 vs 24 months, p<br />

Conclusion: FLAG-Ida proved to be highly effective <strong>and</strong><br />

low toxic regimen, very useful in our group of relapsed <strong>and</strong><br />

primary refractory AL pts, enabling them to achieve CR <strong>and</strong><br />

to undergo allogeneic stem cell transplantation.<br />

PP46 Clinical outcome <strong>and</strong> prognosis related<br />

factor in acute promyelocytic leukemia<br />

patients treating with PETHEMA LPA 099<br />

protocol.<br />

Mitrovic M, Suvajdzic N, Bogdanovic A, Novkovic A,<br />

Kraguljac N, Sretenovic A, Antic D, Djunic I, Vidovic A,<br />

Colovic N, Virijevic M, Djurasinovic V, Elezovic I,<br />

Tomin D. Clinic of hematology, Clinical Center Serbia,<br />

Belgrade, Serbia<br />

In acute promyelocytic leukemia (APL) a cure rate of<br />

75–80% can be anticipated with a combination of all-trans<br />

retinoic acid (ATRA) <strong>and</strong> anthracyclines. But, underst<strong>and</strong>ing<br />

of the prognostic factors associated with the induction<br />

mortality has remained controversial <strong>and</strong> limited.<br />

Aim: To analyze the clinical outcome <strong>and</strong> the prognostic<br />

factor in APL patients (pts) treating with PETHEMA LPA<br />

099 protocol.<br />

Patients <strong>and</strong> methods: From 2004 to 2010, 42 consecutive<br />

APL pts confirmed either by t(15; 17) or PML/RARA<br />

were treated with PETHEMA LPA 99. The median followup<br />

was 32 months (range: 1–78).<br />

Results: Median time from the first symptoms to diagnosis<br />

was 23 days (range: 5–90). Pretreatment pts characteristic<br />

were as follows: median age 42 years (range: 21–69),<br />

22/42 male; mean WBC 25.6 x10 9 /L (range: 0.6–183); mean<br />

platelet count 34x10 9 /L (range: 4–101); hypergranular form<br />

in 39/42 (93%) pts; PETHEMA risk stratification: high<br />

17/42 (40%), intermediate 17/42 (40%), low 8/42 (20%);<br />

mean D-dimer 3606 ľg/L (range: 996–11340). DIC was<br />

confirmed in 32/42 (86%) pts, mean DIC score 6 (range:<br />

3–7). Additional cytogenetics abnormalities were detected<br />

in 8/42 (19%) pts.<br />

Therapy results: 33/42 (76%) pts achieved complete remission<br />

(CR). Induction death occurred in 9/42 (21%) pts,<br />

due to: differentiation syndrome (DS)- 4/42 (9.5%), central<br />

nervous system hemorrhage- 4/42 (9.5%) <strong>and</strong> infection-<br />

1/42 (2.3%). Early died pts had higher WBC (55.9x10 9 /L<br />

vs. 14.1x10 9 /L, p=0.014), higher percentage of peripheral<br />

blood (PB) promyelocytes (15.0% vs. 9.59%, p=0.03) <strong>and</strong><br />

higher DIC score (6.32 vs. 5.85, p=0.014). All died patients<br />

were CD15- with DIC score >5. Predictors of induction<br />

death in univariate analysis were: WBC >30x10 9 /l (p=0.03),<br />

DIC score >5 (p=0.01), CD15- (p=0.04). In multivariate<br />

analysis only WBC >30x10 9 /l was an independent predictor<br />

factor. DS occurred in 11 pts (26%). Pts with DS had significantly<br />

higher percentage of PB promyelocytes (48% vs.<br />

28.68%, p=0.04). Microgranular subtype (p=0.048) <strong>and</strong><br />

CD15- (p=0.04) were predictive for DS. There <strong>are</strong> no predictive<br />

parameters for bleeding. Three relapses occurred<br />

after the third consolidation, during maintenance <strong>and</strong> 6<br />

109


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

months after the maintenance termination, respectively.<br />

Mean time to relapse was14 months (range: 2–31). The<br />

26/42 (62%) pts <strong>are</strong> alive in first continuous CR. Three-year<br />

overall survival (OS) is 76% <strong>and</strong> disease free survival (DFS)<br />

82%.<br />

Conclusion: PETHEMA LPA-99 proved effective in our<br />

APL patients concerning OS, DFS <strong>and</strong> relapse rate. High<br />

rate of early death could be attributed to the numerous highrisk<br />

pts <strong>and</strong> increased rate of DIC. WBC>10x10 9 /L, DIC<br />

score>5 <strong>and</strong> CD15 negativity were poor prognostic factors<br />

for early death. Reduction of the incidence of early deaths is<br />

m<strong>and</strong>atory <strong>and</strong> requires a timely diagnosis <strong>and</strong> better management<br />

of both DS <strong>and</strong> bleeding.<br />

PP47 Acute myeloid leukemia with<br />

t(8;21)(q22;q22); RUNX1-RUNX1T1<br />

– case report<br />

Radic Antolic M, Rajic Lj, Femenic R, Bilic E,<br />

Pavlovic M, Perkovic S, Lasan R, Markovic-Glamocak M,<br />

Zadro R. Department of Clinical Laboratory <strong>and</strong><br />

Department of Pediatrics, Clinical Hospital Center <strong>and</strong><br />

School of Medicine, University of Zagreb, Croatia<br />

A 17-year-old male patient was diagnosed with AML in<br />

December 2009; hepatosplenomegaly was present along<br />

with increased white blood cell count (12.1x10 9 /L), anemia<br />

<strong>and</strong> thrombocytopenia. Cytomorphological diagnosis was<br />

AML M2 with eosinophils according to FAB classification.<br />

Immature aberrant myleoid cell immunophenotype (MPO+<br />

CD13+/–CD33+CD34+CD117+CD133+HLADR+) with<br />

coexpression of TdT but without other lymphoid, erythroid,<br />

monocytic or megakaryocytic markers was confirmed. Presence<br />

of t(8;21)(q22;q22) <strong>and</strong> RUNX1-RUNX1T1 fusion<br />

gene was detected in the bone marrow with negative FLT3/<br />

ITD. The patient was treated according to AML-BFM 2004<br />

(combination of intensive chemotherapy <strong>and</strong> hematopoietic<br />

stem cell transplantation, HSCT). Although the patient<br />

achieved cytogenetic response in March 2010, one month<br />

later 3% of bone marrow cells showed t(8;21)(22q;22q).<br />

Because of his age, gender <strong>and</strong> poor response after 15th day<br />

of intensive chemotherapy, the patient was scheduled for allogeneic<br />

HSCT. After allogeneic HSCT, the patient developed<br />

Wernicke encephalopathy with bacterial infections<br />

that were successfully treated. However, he never achieved<br />

molecular response during chemotherapy as well as in posttransplantation<br />

period. This prompted us to quantify<br />

RUNX1-RUNX1T1 fusion gene <strong>and</strong> WT1 gene expression.<br />

Expression of WT1 gene showed decreased tendency prior<br />

to allogeneic HSCT <strong>and</strong> was lower than that of RUNX1-<br />

RUNX1T1 fusion gene. In posttransplatation period WT1<br />

gene expression increased 3.5 times <strong>and</strong> was higher than<br />

RUNX1-RUNX1T1 fusion gene expression. Three months<br />

later allogeneic HSCT bone marrow was negative for t(8;21)<br />

but del(20q) was present in 10% of nuclei with the lowest<br />

value of RUNX1-RUNX1T1 fusion gene expression during<br />

the entire follow-up period. NPM1 mutation A <strong>and</strong> FLT3/<br />

ITD were negative at that time as well. Two months later<br />

del(20q) was present in 2% of nuclei. Nine months after<br />

110<br />

SCT the patient is still in clinical, cytologic <strong>and</strong> cytogenetic<br />

remission, but with positive molecular finding of RUNX1-<br />

RUNX1T1 fusion gene expression. This case report shows<br />

that patients, despite favorable clinical, cytogenetic <strong>and</strong><br />

molecular prognostic markers, do not always have a good<br />

outcome. It also confirms that molecular monitoring of a<br />

patient with leukemia represents the most sensitive method<br />

for estimating response to therapy <strong>and</strong> assessing disease<br />

course.<br />

PP48 Successful treatment of B-cell<br />

prolymphocytic leukemia (B-PLL)<br />

with FCR-Lite protocol<br />

Batar P, Telek B, Udvardy M. Department of Hematology,<br />

University of Debrecen Health Sciences Center, Debrecen,<br />

Hungary<br />

B-cell prolymphocytic leukemia is a r<strong>are</strong> disorder accounting<br />

approximately for 2% of all lymphoid malignancies.<br />

It is chracterized by marked lymphocytosis in the peripheral<br />

blood, matured lymphocytic infiltration in the bone<br />

marrow <strong>and</strong> prominent splenomegaly. It has a distinct immunophenotype<br />

pattern (CD19, CD20, CD22, FMC7, intensive<br />

surface immunoglobulin positivity) which helps to<br />

differentiate from other lymphoproliferative malignancies.<br />

The disease has poor prognosis <strong>and</strong> its treatment is unsettled.<br />

Here we present a case of a 71 year old male patient<br />

who was admitted to the outpatient facility of the University<br />

of Debrecen Health Sciences Center Department of Hematology<br />

on December 2008. He was presented with 71.0 G/L<br />

WBC of witch 65% were prolymphocyte. Prominent splenomegaly<br />

was also present. The lower border of the spleen<br />

exceeded the left costal margin by 14 cm. No anaemia,<br />

thrombocytopenia or lymphadenopathy were present. Bone<br />

marrow aspiration revealed hypercellularity <strong>and</strong> 80% matured<br />

lymphocytic infiltration. Flow cytometry identified CD5-<br />

, CD19+, CD20+, CD22+ <strong>and</strong> FMC+ phenotype. Surface<br />

kappa light chain positivity was also present. Cytogenetics<br />

<strong>and</strong> FISH were also performed. Chromosomal abnormalities<br />

were not detected. The patient had an excellent clinical<br />

performance status (ECOG: 0). FCR-Lite protocol (fludarabine,<br />

cyclophosphamide, rituximab)l was initiated. Lymphocytosis<br />

<strong>and</strong> splenomegaly have disappe<strong>are</strong>d after 4 cycles.<br />

Six weeks after 6 cycles of chemotherapy flow cytometry<br />

showed complete remission. No major hematological<br />

complications were observed during treatment. However G-<br />

CSF support was needed due to grade III-IV neutropenia<br />

but no fever or infection has developed. After completion of<br />

6 cycles of FCR-Lite treatment maintenance rituximab<br />

monotherapy was started administering 500 mg/m 2 rituximab<br />

every three monts. After a follow up of 2.5 years the<br />

patient is still in complete remission. According to our experience<br />

the FCR-Lite protocol can not only be used in patients<br />

with B-CLL but it also can be effective in patients<br />

with B-PLL. No clinical experience has been reported yet in<br />

the literature with this protocol.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

PP49 Combination of rituximab <strong>and</strong> high-dose<br />

chlorambucil in therapy of small<br />

lymphocyte lymphoma / chronic<br />

lymphocytic leukemia<br />

Aurer I 1 , Hude I 1 , Basic-Kinda S 1 , Dujmovic D 1 ,<br />

Nemet D 1 , Radman I 1 , Ilic I 2 , Stern-Padovan R 3 , Santek F 4 ,<br />

Sertic D 1 , Labar B 1 . 1 Department of Medicine, 2 Parthology,<br />

3 Radiology, 4 Oncology, University Hospital Center <strong>and</strong><br />

School of Medicine, University of Zagreb, Croatia.<br />

Introduction: Chronic lymphocytic leukemia / small lymphocyte<br />

lymphoma (CLL) is the most common leukemia<br />

among adult Caucasians. It is typically a disease of the elderly,<br />

with a median age of onset of 69 years. Clinical course<br />

of the disease is variable <strong>and</strong> the prognosis depends on the<br />

clinical stage at the time of diagnosis <strong>and</strong> other established<br />

prognostic factors. Despite the intensive promotion of fludarabin-based<br />

protocols as the fundamental treatment of CLL,<br />

high dose chlorambucil remains the most efficient monochemotherapy<br />

for these patients. Low toxicity, short treatment<br />

duration <strong>and</strong> favorable cost make this therapy a convenient<br />

platform for addition of non-toxic targeted drugs, like rituximab.<br />

Rituximab is a monoclonal antibody aimed at the<br />

CD20-antigen on the surface of B-lymphocytes.<br />

Aim: Combination of rituximab <strong>and</strong> chlorambucil has<br />

been in routine clinical use for some time. The aim of this<br />

research is to assess its effectiveness.<br />

Patients <strong>and</strong> methods: Twenty-five patients treated at<br />

KBC-Zagreb were included in this study. Median age was<br />

67 years. Patients had received high-dose chlorambucil (10–<br />

20 mg, median 18 mg) continuously <strong>and</strong> 700–800 mg of<br />

rituximab. Eight cycels were planned. Treatment was conducted<br />

with or without addition of glucocorticoids. In analysis,<br />

patients were divided into groups depending on Binet<br />

<strong>and</strong> Rai stages <strong>and</strong> total tumor mass score as well as other<br />

prognostic factors. Adverse events (AE) were evaluated. Assessed<br />

outcomes were response rates <strong>and</strong> survival.<br />

Results: Complete remission (CR) was achieved in 12<br />

patients (48%), partial remission (PR) in 7 (28%), 2 patients<br />

were refractory to therapy (8%) <strong>and</strong> in 4 treatment is still<br />

ongoing. After median follow-up of 15 months of survivors,<br />

progression-free survival (PFS) is 72% <strong>and</strong> overall survival<br />

(OS) 92%. Patients with Binet stage C <strong>and</strong> Rai stages 3 <strong>and</strong><br />

4 have a statistically significant lower PFS than patients<br />

with favorable stages (26% vs, 86%, p=0, 047). The effect<br />

of total tumor mass (TTM) score as well as other prognostic<br />

factors on PFS was not statistically significant (p > 0,05).<br />

Likewise, correlation between assessed prognostic factors<br />

hasn’t been proven. Serious toxicity occurred in 7 patients,<br />

6 of whom were ≥ 65 years. However, correlation between<br />

age <strong>and</strong> toxicity wasn’t statistically significant. The most<br />

common serious AE was leukopenia.<br />

Conclusion: Combination of rituximab <strong>and</strong> continuous<br />

high-dose chlorambucil is a very effective front-line treatment<br />

of CLL, leading to high overall response. Therapy<br />

does not cause unexpected toxicity <strong>and</strong> is well tolerated. To<br />

obtain more relevant results for comparison with larger<br />

studies, further follow-up <strong>and</strong> inclusion of new patients is<br />

needed.<br />

PP50 Atypical Blastoid Variant of Hairy Cell<br />

Leukemia: a Case Report<br />

Hadji-Pecova L, Petrusevska G, Panovska I, Stojanovski Z,<br />

Stojanovic A. Department of Hematology, Medical Faculty,<br />

Skopje, Republic of Macedonia<br />

Hairy Cell Leukemia (HCL) is a r<strong>are</strong> B-cell indolent lymphoproliferative<br />

disorder, also known as one of the B-cell<br />

chronic lymphatic leukemia (B-CLL). HCL-blastoid variant<br />

describes an entity of HCL that is important from the point<br />

of view of requiring differential diagnosis from classic<br />

HCL, HCL-variant <strong>and</strong> others Non-Hodgkin’s Lymphoma.<br />

Differential diagnosis includes HCL, HCL-v, B-CLL,<br />

Mantle cell lymphoma, Folicular lymphoma (FL). <strong>and</strong><br />

Splenic lymphoma vith vilous lymphocyte.<br />

HCL-blastoid variant differs from the classic form of<br />

HCL with respect to the lack of monocytopenia, leucocyosis<br />

<strong>and</strong> unique morphology <strong>and</strong> immunophenotype.<br />

We report a 30 years male patient who presented at our<br />

department in May 2005 with leucocytosis, mild anemia,<br />

skin bleeds <strong>and</strong> splenimegaly. The blood smear showed<br />

more than 50% of heterogenous lymphoid population with<br />

balstoid hairy cells prdominance. In the bone marrow biopsy<br />

diffuse infiltration of lymphoid cells were seen positive<br />

for CD20, CD79a <strong>and</strong> HLA-DR, <strong>and</strong> negative for CD103,<br />

CD11, DBA44, CD5, <strong>and</strong> CD23. The splenic biopsy showed<br />

diffuse lymphoid infiltration of the white <strong>and</strong> red pulp immunohoistochemically<br />

positive for CD20, CD79a, BCL-2,<br />

HLA-DR, <strong>and</strong> CD10 slightly positive in a subset of cells,<br />

<strong>and</strong> negative for CD103, CD11 <strong>and</strong> DBA44.<br />

Based on bone marrow <strong>and</strong> spleen morphology <strong>and</strong> immunohistochemistry<br />

the patient was unfortunately misdiagnosed<br />

as having diffuse FL.<br />

He was initially treated with splenectomy followed by<br />

seven cycles of CHOP chemotherapy but with minimal response.<br />

In the meantime the second opinion of the bone marrow<br />

<strong>and</strong> spleen histopathology <strong>and</strong> immunohistochemistry were<br />

received, <strong>and</strong> the patient was rediagnosed as a case with r<strong>are</strong><br />

atypical blastoid variant of HCL exhibiting aberrant phenotype.<br />

The therapy with cladribine was immediately started<br />

which resulted with complete remission (CR). The patient<br />

was in CR for two years (April 2006 – April 2008).<br />

By the end of April 2008 relapse was confirmed. Peripheral<br />

blood <strong>and</strong> bone marrow showed massive, almost 100%<br />

infiltration with bizarre blastoid cells.<br />

The patient was treated as acute leukemia. He received 3<br />

cycles of Hyper C-VAD chemotherapy (he was in CR after<br />

the first cycle) followed by consolidation with cladribine.<br />

Recently, positive mutation of BRAF was confirmed.<br />

Since November 2008 the patient is in CR.<br />

This case suggests that we should be extremely c<strong>are</strong>ful<br />

during the period of diagnosis having in account the existence<br />

of aberrant phenotypes, <strong>and</strong> that the morphology still<br />

remains important tool. Literature research reveals that aberrant<br />

phenotype CD20+, CD10+ in particular the conjunction<br />

with CD103 negative has been known for several years,<br />

comprising roughly 2–5% of HCL cases.<br />

111


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

PP51 Intravascular T-cell lymphoma presented as<br />

a subcutaneous panniculitis-like lymphoma:<br />

A case report<br />

Ilic I 1 , Basic-Kinda S 2 , Bosnic D 2 , Aurer I 2 , Dotlic S 2 ,<br />

Gasparovic V 1 . 1 Department of Pathology, 2 Department of<br />

Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, University of Zagreb, Croatia<br />

Introduction: Intravascular lymphoma (IVL) is a r<strong>are</strong><br />

type of non-Hodgkin lymphoma characterized by proliferation<br />

of lymphoma cells only within the vessel lumina involving<br />

in most cases the skin <strong>and</strong>/or the brain. Majority of<br />

IVL <strong>are</strong> of B-cell origin but r<strong>are</strong> cases of T-cell <strong>and</strong> NK cell<br />

IVL have been reported.<br />

Case presentation: A 45-year old female was admitted<br />

to the hospital because of the fever of four month duration,<br />

elevated erythrocyte sedimentation rate, elevated level of<br />

lactate dehydrogenase, thrombocytopenia <strong>and</strong> multiple erythematous<br />

patches <strong>and</strong> nodules on her legs <strong>and</strong> lower back,<br />

suggesting a subcutaneous panniculitis-like T-cell non Hodgkin<br />

lymphoma. During the observation she developed severe<br />

dyspnea <strong>and</strong> impairment of consciousness. Biopsy of<br />

skin lesion was performed <strong>and</strong> it showed dermal vessels<br />

filled with large atypical lymphatic cells. Immunophenotypic<br />

analysis showed that the tumor cells were CD2-,<br />

CD3+, CD4+, CD5−, CD7+, CD8−, CD30-, EMA-, CD56−,<br />

ALK−, TIA1-, Granzyme B-, Perforin-, CD20-, Cytokeratin<br />

-. There was no tumor cell infiltrate outside the vessels or<br />

in the subcutaneous fat tissue. Treatment with Methotrexate<br />

combined with Asparginase <strong>and</strong> Dexamethasone was initiated,<br />

but the patient died of multiple organ failure about a<br />

month after admission to the hospital.<br />

Discussion: We found about 20 cases of intravascular Tcell<br />

lymphoma reported so far in the literature, <strong>and</strong> therefore<br />

T-cell IVL is still considered a very r<strong>are</strong> entity. The<br />

patients may show a variety of symptoms, usually nonspecific,<br />

such as abdominal pain, altered mental status or fever,<br />

making this diagnosis difficult to recognize <strong>and</strong> delaying<br />

the treatment. Some of the patients with IVL present with<br />

skin lesions reminiscent of panniculitis or subcutaneous<br />

panniculitis-like T-cell lymphoma. The only way to obtain a<br />

correct diagnosis is to perform a biopsy of such lesions.<br />

Histopathological differential diagnosis includes intravascular<br />

lymphomatosis seen in hepatosplenic T-cell lymphoma<br />

or in angioimmunoblastic peripheral T-cell lymphoma,<br />

making an IVL a diagnosis of exclusion. Thus, without<br />

proper clinical information the diagnosis cannot be established.<br />

Conclusion: Intravascular lymphoma is a r<strong>are</strong> disease,<br />

however in patients with fever of unknown origin, pancytopenia<br />

or nonspecific neurological symptoms, IVL should be<br />

considered. The biopsy, as well as thorough examination of<br />

the patient is crucial for reaching the exact diagnosis.<br />

112<br />

PP52 Primarily Localized non Hodgkin<br />

Lymphoma of Testes with Relapse<br />

Localized in Buccal Area – Case Report<br />

Hotic-Laz<strong>are</strong>vic S, Kezic Lj, M<strong>and</strong>ic D, Stojcic B,<br />

Malinovic J, Mrdja J. Division of Hematology, Department<br />

of Internal Medicine, Clinical Center Banja Luka, Banja<br />

Luka, Bosnia <strong>and</strong> Herzegovina<br />

Introduction: Primarily localized lymphoma of testes is<br />

r<strong>are</strong> <strong>and</strong> aggressive extranodal non Hodgkin lymphoma. It<br />

is the most frequent tumor of testes in men aged 60–80<br />

years.<br />

The Aim of the study: to present the case of a patient<br />

with primarily localized lymphoma of testes, in whom there<br />

has been performed orchiectomy within two different periods,<br />

performed chemotherapy <strong>and</strong> radio therapy, <strong>and</strong> the<br />

relapse of the disease in both cheeks <strong>and</strong> subm<strong>and</strong>ibular<br />

gl<strong>and</strong> on the left.<br />

Case presentation: a patient of 56 years of age, with<br />

painless swelling of the left scrotal <strong>are</strong>a <strong>and</strong> non-homogenhypoechogne<br />

mass (48x32 mm) of the left testis with microcalcifications<br />

verified by ultrasound. After the left orchiectomy<br />

(pathohystological diagnosis was Lymphoma non<br />

Hodgkin of the left testis- diffused large cell B-phenotype,<br />

CD-20+, of high proliferation activity) there was implemented<br />

the chemotherapy R-CVP on the 21st day in the<br />

planned therapy doses, as well, which the patient underwent<br />

well. During the therapy of maintaining by rituximab, after<br />

the second treatment, swelling of the right scrotal <strong>are</strong>a.<br />

There was indicated the right orchiectomy, <strong>and</strong> the obtained<br />

pathohystological finding was almost identical to the first<br />

one. The implemented irradiation of both testes boxes, abdomen<br />

<strong>and</strong> pelvis with 36 Gy/20 fractions by the method of<br />

reversed Y. Seven moths upon the completed irradiation,<br />

<strong>and</strong> after the first dose of substance of Testosterone depot<br />

(due to exceptionally low values of testosterone <strong>and</strong> difficulties<br />

related to it), occurrence of tm forming of both buccal<br />

<strong>are</strong>as <strong>and</strong> subm<strong>and</strong>ibular gl<strong>and</strong> on the left. After biopsy,<br />

by pathohystological finding, there was verified the Hodgkin<br />

lymphoma understood as the relapse of the disease <strong>and</strong><br />

there started the therapy according to MINE protocol, which<br />

is still being realized.<br />

Conclusion: Primarily localized lymphoma of testes <strong>and</strong><br />

buccal <strong>are</strong>a <strong>are</strong> r<strong>are</strong> extranodal localizations of non Hodgkin<br />

lymphoma.<br />

PP53 A 48 Year Old Woman with a R<strong>are</strong> Case<br />

of a Midline Destructive Disease Presenting<br />

as Having both Midline <strong>and</strong> Nonmidline<br />

Lesions – a Case Report<br />

Fern<strong>and</strong>ez S, Lapus F, B<strong>are</strong>z MY. Davao Medical Center,<br />

Davao City, Philippines<br />

Objectives: To describe a r<strong>are</strong> case of lethal midline granuloma<br />

presenting in a r<strong>are</strong> location. This type of lymphoma<br />

which needs a tissue biopsy for diagnosis <strong>and</strong> immunohistochemical<br />

staining for confirmation is a challenge in a developing<br />

nation to diagnose, treat <strong>and</strong> follow up. This paper is


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

presented to be able to show how a r<strong>are</strong> case of lymphoma<br />

was appropriately diagnosed in sans the modern methods of<br />

immunohistochemical staining with promising results of<br />

chemotherapy.<br />

Midline destructive lesions <strong>are</strong> a heterogeneous group of<br />

r<strong>are</strong> lesions where the classification angiocentric lymphoma,<br />

or peripheral Natural Killer/T cell lymphoma, nasal<br />

type belongs. It usually presents with destructive mass lesions<br />

<strong>and</strong> extensive destruction in the midfacial <strong>are</strong>as. We<br />

describe a case of an angiocentric lymphoma, or Natural<br />

Killer/T cell lymphoma who presented with both a nasal<br />

mass <strong>and</strong> a skin lesion in the medial aspect of her left leg. A<br />

tissue biopsy was done to make the diagnosis. She underwent<br />

six cycles of chemotherapy <strong>and</strong> radiotherapy with very<br />

promising results<br />

Conclusions: We report this case to document a r<strong>are</strong> type<br />

of lymphoma. Our patient was unique in that she presented<br />

with a r<strong>are</strong> type of lymphoma that is not usually documented<br />

in the Philippines <strong>and</strong> is r<strong>are</strong> even in international reports.<br />

Moreover, she presented also unusually having both a<br />

nasal mass <strong>and</strong> a skin lesion in the medial aspect of her left<br />

leg.<br />

PP54 Successful mobilisation with Plerixafor-<br />

clinical report<br />

Babic A, Istituto Europeo di Oncologia, Divisione di<br />

Ematologia, Milan, Italy<br />

Background: Our Clinical Transplantation Program sometimes<br />

sees a young people treated in abroad interrupting<br />

or changing their clinical pathway.<br />

Clinical Case: Here is the case of a young russian girl,<br />

16 year old girl, affected by Hodgkin Lymphoma, nodular<br />

sclerosis, stage IIIB. Diagnosis was performed in Russia in<br />

October 2009 on a biopsy of a supraclavicolar lymphnode.<br />

She then underwent 6 cycles of chemotherapy including antracycline,<br />

cyclophosphamide, vincristine, procarbazine <strong>and</strong><br />

prednisone which was followed by mantle <strong>and</strong> inverted Yfield<br />

radiotherapy until June 2010. All these treatments has<br />

been performed by using peripheral veins. During the treatment<br />

she developed Herpes Zoster <strong>and</strong> Parvovirus infection<br />

treated respectively with antiviral drugs <strong>and</strong> Ig ev. In August<br />

2010, a CT scan showed relapse of disease. She was inserted<br />

the Gronshong central catheter <strong>and</strong> on October 25th, she<br />

underwent the first cycle of chemotherapy with ifosfamide,<br />

etoposide <strong>and</strong> prednisone <strong>and</strong> on November 2nd she was<br />

admitted in our inpatient department<br />

Methods: We wanted to use this chemotherapy cycle as<br />

»mobilization« adding GCSF if necessary to see if we <strong>are</strong><br />

able to collect her stem cells for autologous transplant. We<br />

followed her blood cell counts recovery, monitoring circulating<br />

CD34+ as soon as white blood cells reached 500 /ul,<br />

but counts showed very low numbers, only 2 cells/ul at 11th<br />

November. We therefore decided to ad plerixafor <strong>and</strong> the<br />

very next day she had 10 cells/ul of CD34+ <strong>and</strong> we positioned<br />

a central venous cathether in the femoral vein deciding<br />

to perform the firsth apheresis procedure <strong>and</strong> permitting<br />

her to achieve 0.7 x 106 cells/kg at the very day (12 November).<br />

We therefore administered a second dose of plerixafor<br />

<strong>and</strong> repeated the procedure the day after with 19 cells/ul <strong>and</strong><br />

a collection of 1.5 x 106 cells/kg, for a total of 2.2 x10E6<br />

CD34+/kg.<br />

Results: Therefore she underwent additional two cycles<br />

with ifosfamide, carboplatin <strong>and</strong> etoposide therapy <strong>and</strong><br />

achieved a complete response PET <strong>and</strong> CT scan documented<br />

in Jaunary 2011.<br />

Conclusions: She then was able to undergo the autologous<br />

stem cell transplantation after a conditioning regimen<br />

according to BEAM schedule <strong>and</strong> in February 2011 PET<br />

<strong>and</strong> CT scans confirmed the complete response.<br />

PP55 Isolation <strong>and</strong> in vitro cell culture of<br />

mesenchymal stem cells from human<br />

umbilical cord blood<br />

Mazic S 1 , Golemovic M 1 , Skific M 1 , Bojanic I 1 , Humar I 2 ,<br />

Golubic Cepulic B 1 . 1 Department of Transfusiology,<br />

2 Deaprtment of Clinical Laboratory, Cliniacal Hospital<br />

Center, Zagreb, Croatia<br />

Mesenchymal stem cells (MSCs) <strong>are</strong> multipotent bone<br />

marrow (BM) cells that give stromal support for hematopoiesis<br />

<strong>and</strong> express immunomodulatory properties <strong>and</strong> regenerative<br />

potential. Although cord blood (CB) is easily accessible<br />

source of MSCs <strong>and</strong> could serve as alternative to BM,<br />

isolation success of MSCs is very low. Therefore attempts<br />

<strong>are</strong> made to enhace isolation <strong>and</strong> expansion rates <strong>and</strong> in this<br />

regard different culture conditions in vitro <strong>are</strong> being examined.<br />

Lately, human platelet lysate (PL) has been proposed<br />

as fetal bovine serum substitute.<br />

Aim: The aim of this study was to evaluate success rate<br />

of MSCs isolation from CB by imunomagnetic depletion of<br />

CD34+ hematopoietic fraction <strong>and</strong> to investigate biological<br />

properties of MSCs exp<strong>and</strong>ed in presence of 10%PL.<br />

Samples <strong>and</strong> methods: 15 CB units were selected according<br />

to predefined criteria; volume >40ml, total nucleated<br />

cells (TNC) >6x10 8 , mononucleated cells (MNC) >1x10 8<br />

<strong>and</strong> processing within 15 hr from collection. Each CB was<br />

divided in two cell fractions; CD34- <strong>and</strong> unmanipulated<br />

fraction. MNCs of each fraction were plated at 1x10 6 /cm 2 .<br />

Adherent cells were cultured in presence of 10% PL until<br />

reaching confluence <strong>and</strong> then harvested by trypsinization<br />

<strong>and</strong> replated at 5000 MSCs/cm 2 . At each passage (P) cells<br />

were counted <strong>and</strong> population doublings (PD) were calculated.<br />

In order to evaluate clonogenic potential of cultured<br />

cells, colony-forming unit-fibroblast (CFU-F) assays were<br />

performed starting at P2. Flow cytometry analysis of surface<br />

phenotype was done in P3. Cells harvested in P5 were<br />

added to mixed lymphocyte cultures (MLR) in order to test<br />

their immunomodulatory properties.<br />

Results: Expansion of MSCs in presence of PL resulted<br />

in 27% (4/15) overall success rate. Within successfully cultured<br />

MSCs 3/4 (75%) originated from CD34- fraction.<br />

Cells cultivated from two fractions were morfologicaly different.<br />

Cultured MSCs showed slow growth kinetics; time<br />

needed for one MSCs PD in 4 samples was 8.5, 18, 16 <strong>and</strong><br />

7.4 days respectively. Initialy high clonogenic potential rapidly<br />

decreased by P5. Analysis of surface phenotype showed<br />

113


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

expected lack of hematopoietic markers <strong>and</strong> variable expression<br />

of typical MSCs markers. All cultivated MSCs expressed<br />

CD73 while CD90 was expressed in variable intensity.<br />

Expression of CD105 was not detected. Addition of<br />

cultivated MSCs in different concentrations to MLR resulted<br />

in modest immunomodulatory effect.<br />

Conclusion: None of the predefined selection criteria<br />

correlated with MSCs isolation success. Depletion of<br />

CD34+ cells favoured growth of MSCs. Whether unexpected<br />

immunophenotype of cultured MSCs correlates with<br />

specific culture conditions or with some additonal parameter<br />

needs to be evaluated in larger group of samples. Despite<br />

availability of CB as potential source of MSCs, their low<br />

frequency <strong>and</strong> unpredictable expansion success along with<br />

time consuming <strong>and</strong> laborious procedure, make CB unreliable<br />

source of MSCs for clinical use.<br />

PP56 Biological properties of human<br />

mesenchymal stem cells exp<strong>and</strong>ed in vitro<br />

in media supplemented with human<br />

platelet lysate<br />

Skific M 1 , Golemovic M 1 , Mazic S 1 , Bojanic I 1 , Humar I 2 ,<br />

Davidovic S 3 , Crkvenac Gornik K 3 , Ilic I 4 , Durakovic N 5 ,<br />

Mikulic M 5 , Serventi Seiwerth R 5 , Labar B 5 ,<br />

Golubic Cepulic B 1 . 1 Department of Transfusiology,<br />

2 Department of Clinical Laboratory, 3 Department of<br />

Pediatric, 4 Department of Pathology, 5 Department of<br />

Medicine, Clinical Hospital Center <strong>and</strong> School of<br />

Medicine, Universiuty of Zagreb, Croatia<br />

Human mesenchymal stem cells (MSCs) <strong>are</strong> nonhematopoietic<br />

multipotent cells that express regenerative <strong>and</strong> immunomodulatory<br />

properties in vitro <strong>and</strong> appear immunoprivileged<br />

what gives them considerable therapeutic potential.<br />

Because of very low incidence in the bone marrow<br />

(BM) aspirates MSCs need to be exp<strong>and</strong>ed in vitro. Recently<br />

human platelet lysate (PL) has been evaluated as fetal<br />

bovine serum (FBS) substitute in MSCs in vitro expansion.<br />

Aim: The aim of this study was to evaluate biological properties<br />

of MSCs exp<strong>and</strong>ed in vitro in the presence of PL <strong>and</strong><br />

create basis for the development of clinical expansion protocol.<br />

Patients <strong>and</strong> methods: Mononuclear cells (MNC)<br />

were isolated from the BM of 8 healthy individuals by Ficoll<br />

density gradient centrifugation <strong>and</strong> plated at 160000<br />

MNC/cm 2 in media suplemented with 10%FBS, 10%PL or<br />

5%PL. When the cells reached 80% confluence, they were<br />

harvested by trypsinization <strong>and</strong> replated at 1000 MSCs/cm 2<br />

until reaching passage 5 (P5). To evaluate growth kinetics in<br />

different culture conditions, number of cells was determined<br />

by light microscopy techniques <strong>and</strong> to evaluate MSCs clonogenic<br />

potential, colony forming unit-fibroblast (CFU-F)<br />

assays were performed at each P. Characteristic immunophenotype<br />

was assesed by flow cytometry whenever sufficient<br />

number of cells was obtained. To assess their multipotency,<br />

MSCs were induced into adipogenesis <strong>and</strong> osteogenesis<br />

in vitro (n=6). Immunomodulatory properties were<br />

evaluated by adding 10-40000 MSCs to mixed lymphocyte<br />

reaction (MLR) (n=1). Karyotype analysis was performed<br />

to validate chromosomal stability of exp<strong>and</strong>ed cells (n=5).<br />

114<br />

Results: Dominant growth of MSCs was observed in the<br />

presence of 10%PL <strong>and</strong> confirmed by CFU-F tests. Cell<br />

growth analysis showed that, by the P5, median time needed<br />

for one MSC population doubling (PD) in the presence of<br />

10%FBS was 154hr comp<strong>are</strong>d to 50hr <strong>and</strong> 40hr in 5%PL<br />

<strong>and</strong> 10%PL, respectively. More than 96% of cells cultured<br />

in the presence of 10%PL expressed CD90 <strong>and</strong> CD73 already<br />

in P1 <strong>and</strong> P2, while CD105 expression was either<br />

very low or not detected. Cultured cells were devoid of hematopoietic<br />

markers already in P1. Cells cultured in presence<br />

of 10%PL differentiated into adipocytes <strong>and</strong> osteoblasts.<br />

In MLR experiment, MSCs cultured with 10%PL<br />

displayed stronger immunomodulatory effect comp<strong>are</strong>d to<br />

those cultured in 5%PL. Karyotype was analyzed in 5 samples<br />

of different donors mostly obtained in P3 or P4. Analysed<br />

metaphases displayed normal karyotype with the exception<br />

of one sample where trisomy 8 was detected in P4,<br />

but not in subsequent P5.<br />

Conclusion: MSCs cultured in the presence of 10%PL<br />

display almost all expected biological <strong>and</strong> immunomodulatory<br />

properties. Whether absence of CD105 expression correlates<br />

with specific culture conditions, needs to be confirmed<br />

in the bigger cohort of samples. This study provides<br />

a basis for further efforts in evaluating PL as a valuable FBS<br />

substitute in clinical grade MSCs expansion.<br />

PP57 Croatian Cord Blood Bank: the fi rst 4 years<br />

Mazic S 1 , Bojanic I 1 , Zlopasa G 2 , Mrsic M 3 , Plenkovic F 1 ,<br />

Lukic M 1 , Raos M 1 , Gojceta K 1 , Golemovic M 1 , Skific M 1 ,<br />

Tomac G 1 , Burnac IL 1 , Novoselac J 1 , Vidovic I 1 ,<br />

Biskup M 1 , Labar B 3 , Golubic Cepulic B 1 . 1 Department of<br />

Transfusiology, 2 Department of Gynecology <strong>and</strong><br />

Obstetrics, 3 Department of Medicine, Clinical Hospital<br />

Center <strong>and</strong> School of Medicine, Universiuty of Zagreb,<br />

Croatia<br />

Croatian cord blood bank (CBB) was established in<br />

March 2007 with the aim to provide patients with transplants<br />

according to international st<strong>and</strong>ards <strong>and</strong> increase<br />

their chance in finding a match. Furthermore, our goal was<br />

to promote cord blood (CB) donation <strong>and</strong> by educating all<br />

croatian maternities give every mother an opportunity for<br />

donation.<br />

In that regard a team of doctors from CBB, maternity<br />

hospital <strong>and</strong> national bone marrow donor registry, starting<br />

with Zagreb <strong>and</strong> gradually spreading out throughout the<br />

country, visited every maternity ward <strong>and</strong> educated complete<br />

staff about donor information <strong>and</strong> selection, CB collection,<br />

processing, storage <strong>and</strong> therapeutic use. Our CBB<br />

collaborates with 22 maternities for which we give full logistic<br />

support. We give them feedback through quaterly reports<br />

of collected <strong>and</strong> discarded donations, point out problems<br />

<strong>and</strong> if necessary reeducate the staff.<br />

Samples <strong>and</strong> methods: CB is collected in utero, in term<br />

deliveries with mother´s informed consent. Transport is organized<br />

within 24 h through courier service. The unit is<br />

checked for volume <strong>and</strong> total nucleated cell number (TNC)<br />

<strong>and</strong> accepted if TNC is > 10x10 8 . CB is processed using


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

Sepax cell separator, ISBT code labelled <strong>and</strong> stored in Bioarchive.<br />

Results: In a period from 2007–2011 our CBB received<br />

2879 donations out of which 1411 (49%) were stored. The<br />

reasons for rejection of donated units at initial control were<br />

low TNC (97,7%), period from collection to storage exceeding<br />

48 h (0,3%), administrative error (0,1%), twin pregnancy<br />

(0,1%), validation study (1,6%). Additional 240 doses<br />

were rejected at final control due to quality of CB unit<br />

(76,7%), problems during transport or processing (14,7%),<br />

medical information about newborn (0,8%), mother (3,3%)<br />

or father (2,9%), or incomplete documentation (0,4%).<br />

Among reasons for inadequate quality of CB unit, low TNC<br />

after processing accounted for most rejections (44%), microbiological<br />

contamination for 31,1% while no detectable<br />

growth in clonogenic assay accounted 1,6% of cases.<br />

For this period we had 1171 doses enter the registry of<br />

World Marrow Donor Association (WMDA) which makes<br />

40,6% of recieved donations. Despite numerous preliminary<br />

search requests for our units up till now we issued only one<br />

CB to treat a patient in Croatia.<br />

In the past year we had a noticable drop in the percentage<br />

of stored CB units; 39% versus average of 56,6% in first 3<br />

years. Reason for this is increased number of donations<br />

which allowed us to set higher initial TNC criteria. Units<br />

that entered the WMDA registry in 2010. had mean TNC<br />

count of 9,64±2,7 <strong>and</strong> mean CD34+ count of 5,58±3,85.<br />

Conclusion: To increase our chances for issuing units for<br />

transplantation we plan to continue promoting CB donation,<br />

setting higher initial TNC criteria <strong>and</strong> working on receiving<br />

FACT accreditation <strong>and</strong> entering Netcord.<br />

PP58 Italian nurses group in mobilisation <strong>and</strong><br />

apheresis (GIIMA): ideation <strong>and</strong> activity<br />

Babic A, Laszlo D, Galgano L, De Marchi E, Orl<strong>and</strong>o L,<br />

Martinelli G. Istituto Europeo di Oncologia, Divisione di<br />

Ematologia, Milan, Italy<br />

Background: The Clinical Transplantation Program consists<br />

in a process performed by a multidisciplinary team<br />

working with common staff training, programs, protocols,<br />

<strong>and</strong> quality management systems. Peripheral blood stem cells<br />

represent the most used source of hematopoietic cells <strong>and</strong><br />

apheresis is a routine technique applied to collect them.<br />

The collection is generally performed by National Transfusion<br />

Services while the mobilization <strong>and</strong> reinfusion of<br />

stem cells is carried out by Clinical Units. For these reason,<br />

there is difficult to assure a continuity of patient assistance,<br />

especially from a nurse’s point of view.<br />

Methods: We therefore created in April 2010 a dedicated<br />

group (Gruppo Italiano Infermieristico in Mobilizzazione<br />

ed Aferesi – GIIMA), with a committee linked to the GIT-<br />

MO <strong>and</strong> EBMT nurses groups. The group includes medical<br />

doctors <strong>and</strong> nurses working in National Transfusion Services<br />

<strong>and</strong> Transplant Centers with the aim to:<br />

1) create the stabile link between the apheresis nurses<br />

who works in transfusion centers <strong>and</strong> those who operate<br />

in transplant centers, following both the patients in<br />

mobilization <strong>and</strong> apheresis.<br />

2) create the material for nurse education in apheresis<br />

<strong>and</strong> for patient information<br />

3) develop the program for nurses education in apheresis<br />

for new students based by Jacie’s regulation <strong>and</strong> coherent<br />

with Italian low.<br />

Results: The group translated the EBMT h<strong>and</strong>book »The<br />

Practical Guide for Nurses <strong>and</strong> Other Health C<strong>are</strong> Professionals«<br />

in Italian language <strong>and</strong> have distributed it to each<br />

Italian transplant <strong>and</strong> transfusion center. Currently the GI-<br />

IMA is distributing the DVD already available on dem<strong>and</strong><br />

on: Giima.italy@gmail.com after been distributed, again to<br />

all transplant <strong>and</strong> transfusion centers in Italy <strong>and</strong> Switzerl<strong>and</strong><br />

(Italian Canton). This DVD will be an additional tool<br />

for nurse education. It contains interactive presentation of<br />

autoPBSC process, from mobilization, apheresis, autoPBSC<br />

to follow-up.<br />

The next step is to collaborate in developing the specialized<br />

university program for apheresis nurse education to be<br />

inserted in regional university program REL – SIDEM<br />

Conclusions: GIIMA is a group who created a link between<br />

two realities who performs similar protocols in parallel<br />

ways. The aim of the group is to promote the unique<br />

pathway in stem cell mobilization <strong>and</strong> apheresis. The next<br />

step is to collaborate in developing the specialized university<br />

program for apheresis nurse education hopefully to be<br />

inserted in regional university program REL-SIDEM.<br />

PP59 Unrelated Donor Search Experience<br />

in the Croatian Hematopoietic Stem Cell<br />

Transplantation Program<br />

Grubic Z 1 , Stingl K 1 , Serventi Seiwerth R 2 , Labar B 2 ,<br />

Zunec R 1 . 1 Tissue Typing Centre, 2 Department of<br />

hematology, Clinical Hospital Centre Zagreb, Croatia<br />

The donor search protocols in the unrelated hematopoietic<br />

stem cell transplantation (HSCT) program vary between<br />

transplantation centers due to many different factors, such<br />

as preferences in the source of stem cells (unrelated donor<br />

or umbilical cord unit – CBU), characteristics of the donor,<br />

resolution of the required HLA typing etc. However, the differences<br />

among populations arising from the HLA polymorphism<br />

also play a role <strong>and</strong> should be considered in establishing<br />

the donor search protocol. The aim of this study<br />

was to address this question with respect to the Croatian<br />

population.<br />

In the period 2009–2010, a successful search for an unrelated<br />

donor/CBU in the Bone Marrrow Donors Worldwide<br />

(BMDW) was performed at our centre for 51 patients for<br />

whom a matched donor could not be previously found<br />

among family members. The donor was an unrelated individual<br />

in 48 cases while for the remaining two patients CBU<br />

was chosen as a source of HSCT, double CBU transplantation<br />

in one case. The gender mismatched donor was chosen<br />

for 43% of male patients, while this percentage was slightly<br />

higher for female patients (54%). The median donor age<br />

was 36.2 (range 21 to 57). The HLA resolution typing criteria<br />

in our transplantation centre is high resolution typing for<br />

HLA-A, -B, -C, -DRB1 <strong>and</strong> -DQB1 loci. However, the<br />

115


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

10/10 matching level was not achieved for 24 patients.<br />

Twenty-one patient received a 9/10 matched transplant,<br />

while three had an 8/10 matched donor. The highest number<br />

of mismatches was found at the HLA-C locus (9 patients).<br />

However, the most interesting finding regarding the matching<br />

at the HLA loci was observed among our patients positive<br />

for the HLA-DRB1*11 specificity. Namely, patients<br />

with HLA-DRB1*11:04 allele were more difficult to match<br />

than those with HLA-DRB1*11:01 allele. This was the case<br />

for four patients in the group of patients mismatched for one<br />

DRB1 allele (N=6), <strong>and</strong> one patient who received an 8/10<br />

matched transplant <strong>and</strong> in addition to a r<strong>are</strong> DRB1 allele<br />

also had the HLA-DRB1*11:04 allele. All those patients ultimately<br />

received a DRB1*11:01 positive transplant. The<br />

explanation for this finding lies in the almost equal frequency<br />

of DRB1*11:01 <strong>and</strong> DRB1*11:04 alleles in the Croatian<br />

population as opposed to the predominance of DRB1*11:01<br />

elsewhere. The mismatches in other cases were mainly due<br />

to the either a r<strong>are</strong> allele or an unusual haplotype of the patient.<br />

In conclusion, results of the study revealed some specific<br />

characteristics of the Croatian population which should be<br />

taken into account while performing the donor search. The<br />

finding regarding the HLA-DRB1*11 alleles is an important<br />

example of how the differences in allele distribution<br />

between Croatian population <strong>and</strong> populations with potential<br />

donors can influence the search outcome.<br />

PP60 Following the Chimerism Status of Patients<br />

after Hematopoietic Stem Cell<br />

Transplantation – Ten Years Experience<br />

Stingl K 1 , Grubic Z 1 , Serventi Seiwerth R 2 , Labar B 2 ,<br />

Rajic Lj 3 , Vrhovac R 4 , Zunec R 1 . 1 Tissue Typing Centre,<br />

2 Department of Medicine, 3 Clinic of pediatrics, Clinical<br />

Hospital Centre, 4 Department of Medicine, Clinical<br />

Hospital Merkur, Zagreb, Croatia<br />

The analysis of the patient’s chimerism status following<br />

the hematopoietic stem cell transplantation (HSCT) has<br />

been accepted in the last decade as a valuable tool in the<br />

post-HSCT treatment <strong>and</strong> prediction of the transplantation<br />

outcome. The aim of this retrospective study was to evaluate<br />

the current protocol used for this purpose in our centre.<br />

In the period from 2001–2011, the peripheral blood or<br />

bone marrow samples of 273 patients have been received<br />

for the chimerism analysis. The protocol consisted out of<br />

DNA isolation using the commercial kit, PCR amplification<br />

of the chosen microsatellite loci <strong>and</strong> electrophoresis on a<br />

6% polyacrilamide gel. Microsatellite loci tested were<br />

HUMTH01, HUMVWFA31, HUMFES/FPS, HUMF13A01,<br />

SE33, D1S80 <strong>and</strong> D22S683. In cases when two or less tested<br />

loci were informative (both recipient <strong>and</strong> donor specific<br />

alleles could be identified), additional microsatellites were<br />

analysed (D1S549, D1S1656, D12S391, D18S535).<br />

The number of informative loci ranged from 2 (7.3%) to<br />

7 (2.6%) with the highest percentage of patients having 4<br />

informative loci (32.2%). The most informative locus was<br />

SE33 (61.1%). Additional loci had to be tested for 68 pa-<br />

116<br />

tients. In this period, the analysis could not be performed<br />

only once, in a case when patient <strong>and</strong> donor were identical<br />

twins. The number of times the patient underwent the chimerism<br />

analysis varied, depending on the conditioning regimen,<br />

diagnosis <strong>and</strong> the clinical status of the patient. This<br />

number ranged from one to 25. The majority of the patients<br />

had a full donor chimerism (FDC) status for the entire time<br />

of the follow-up (69.2%). However, various other situations<br />

have been encountered: patients who reached FDC after a<br />

period of decreasing mixed chimerism (MC, 10.3%), patients<br />

with a transient MC (5.9%), patients who had a stable<br />

or decreasing MC (2.6%), patients with an increasing proportion<br />

of recipient cells either after a period of FDC status<br />

or without ever reaching FDC (5.1%), patients who, after a<br />

FDC status, showed MC in the last performed analysis<br />

(3.7%), <strong>and</strong> patients who never achieved engraftment with<br />

only recipient cells detectable (4.0%). A good correlation<br />

between the clinical status <strong>and</strong> the results of the chimerism<br />

analysis has been observed, especially in the cases of increasing<br />

MC.<br />

In conclusion, the protocol for chimerism analysis established<br />

in our centre has proven to be a reliable <strong>and</strong> useful<br />

tool in the patient treatment after transplantation. Regular,<br />

consecutive analyses enable the determination of the chimerism<br />

dynamics <strong>and</strong> thus provide necessary data for timely<br />

interventions <strong>and</strong> improvement of the transplantation outcome.<br />

PP61 Venous access for Apheresis <strong>and</strong><br />

Photopheresis procedures<br />

Babic A. Istituto Europeo di Oncologia, Divisione di<br />

Ematologia, Milan, Italy<br />

Background: Our Clinical Transplantation Program consists<br />

in a process performed by a multidisciplinary team<br />

working with common staff training, programs, protocols,<br />

<strong>and</strong> quality management systems, according to JACIE <strong>and</strong><br />

ISO guidelines. Peripheral blood stem cells represent the<br />

most used source of hematopoietic cells <strong>and</strong> apheresis is a<br />

routinary technique applied to collect them. In our Collection<br />

Unit we use Cobe Spectra System for apheresis procedures<br />

<strong>and</strong> also perform photopheresis procedures with<br />

UVA-PIT off line system for GvHD treatment.<br />

Aims: To perform the apheresis pocedures, single or<br />

multiple, we need the presence of good vascular access,<br />

able to provide constant blood flow pressure of 40–90 ml/h<br />

during the whole procedure time (3 hours approximately)<br />

with minor complication risks, due to the procedure itself or<br />

to a long time presence of central venous catheter.<br />

Methods: For apheresis procedures to collect CD34+<br />

cells or lymphocytes, where possible, usually with healthy<br />

donors, we prefer to use the peripheral veins (cephalic or<br />

basilical) for the procedure. On the contrary, in patients the<br />

peripheral veins <strong>are</strong> often insufficient or severely compromised<br />

by multiple chemotherapy cycles, therefore we need<br />

to use central veins to perform the procedure. Frequently<br />

our patients <strong>are</strong> provided with a Porth-a-Cath Catheter<br />

which can be used as a return way during the procedure so<br />

it becomes easier to find only one peripheral access rather


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Poster presentations<br />

than two. In case of complete absence of peripheral access<br />

we put another central venous catheter preferably in femoral<br />

vein. On the other h<strong>and</strong>, when we perform the extracorporeal<br />

photopheresis procedure with off line system we first<br />

need to execute the lympho+monocytes collection by Cobe<br />

Spectra; therefore we need two good venous access available<br />

during the whole program of cure. There <strong>are</strong> a variety<br />

of schedules depending on disease but usually the treatment<br />

takes 5–6 months to be completed. The choice of the central<br />

venous catheter to be used also depends on the psychophysical<br />

conditions of each patients <strong>and</strong> it needs to be evaluated<br />

c<strong>are</strong>fully <strong>and</strong> on a one-to-one basis.<br />

Results: Depending on the situation, the choice of the<br />

catheters to be used for procedures is personalised. Venous<br />

access, risks of infections <strong>and</strong> thrombosis <strong>are</strong> evaluated by<br />

the apheresis team while the nurse is dedicated to perform<br />

the procedure <strong>and</strong> to schedule catheter’s medications. Complications<br />

due to catheters used for apheresis procedure <strong>are</strong><br />

usually r<strong>are</strong> , mostly those linked to thrombotic or infection<br />

complications. (Bern MM e coll.,1990)<br />

Conclusions: Choosing the right venous access we can<br />

guarantee the optimum blood flow able to collect in less<br />

time the selected cell types with low contamination of unwanted<br />

cells, minor risks due to the platelet loose, minor<br />

stress to the patient <strong>and</strong> major security to patient <strong>and</strong> to operator.<br />

PP62 Pure red cell aplasia preceding a DLBCL<br />

with 6 years<br />

Barca G, Marin S, Nada S, Dragan C, Barbu D,<br />

Angelescu S, Tevet M, Saguna C, Bizu I, Lupu AR,<br />

Coltea Clinical Hospital, Hematology Clinic, Buch<strong>are</strong>st,<br />

Romania<br />

Background: Pure red cell aplasia (PRCA) is one of the<br />

autoimmune diseases seen during the course of lymphomas.<br />

However, the relation of PRCA with the underlying lymphomas<br />

remains unclear, <strong>and</strong> cases were reported in which<br />

PRCA occurred concomitantly, preceded, or followed the<br />

diagnosis of lymphoma.<br />

Aims <strong>and</strong> case description: we report the case of a 49<br />

years-old women with pure red cell aplasia diagnosed in<br />

2003 <strong>and</strong> confirmed by bone marrow biopsy with immunohistochemical<br />

analysis. After 6 years of treatment with red<br />

blood cell mass transfusion, erythropoietin <strong>and</strong> iron chelation<br />

therapy she developed a left supraclavicular tumor. The<br />

biopsy followed by imunohistochemical analysis showed a<br />

diffuse large B cell lymphoma <strong>and</strong> the initial workup established<br />

stage IIIB disease. She received 8 R-CHOP cycles of<br />

chemotherapy, with PET/CT confirming complete remission<br />

at the end. During chemotherapy the hemoglobin concentration<br />

started rising, despite the grim expectations. At<br />

the time this report was written, she does not need transfusions<br />

any more.<br />

Discussion <strong>and</strong> conclusions: Immunotherapy with<br />

Rituximab <strong>and</strong> immunosuppressive medication, including<br />

ciclofosfamide <strong>are</strong> mentioned in the literature as viable<br />

treatments for PRCA. Both Rituximab <strong>and</strong> the immunosuppressive<br />

effects of the CHOP regimen could be responsible<br />

for the favorable response seen in this case. The peculiarity<br />

of the case consists in the the occurence of lymphoma at a<br />

considerable interval from the diagnosis of PRCA <strong>and</strong> the<br />

prompt response of PRCA to the R-CHOP therapy for lymphoma.<br />

PP63 Can Platelet be Unnecessary<br />

Vieira S, Monroe D, Librojo M. London, United Kingdom<br />

Blood transfusion continues to be an essential part of<br />

modern practise but not without risks. During the last decade<br />

there has been an increase interest in national level<br />

across the United Kingdom (UK) <strong>and</strong> Europe for collecting<br />

data on the hazards of transfusion of blood components.<br />

Therefore avoiding unnecessary transfusion is accepted as<br />

one way of reducing the risk which is associated with blood<br />

transfusion. In our stem cell transplant unit we have audited<br />

platelet transfusion (PT) frequency <strong>and</strong> reason for it.<br />

Patients <strong>and</strong> methods: The audit was carried out prospectively.<br />

A log in book was kept in unit for recording the<br />

red cell transfusion orders. The data was validated with<br />

cross check retrospectively all inpatient episodes record on<br />

monthly basis. The audit was carried over 6 months of period<br />

form May 2010 till end of October 2010.<br />

Results: Unit policy is to keep platelet count above 10<br />

with the exception of 20 if the patient is febrile. It is also<br />

aimed to keep plt above 50 if pat is receiving anticoagulant<br />

or bleeding for various reasons.<br />

In six months period, total of 105 episodes platelet transfusion<br />

occurred. Only 10% of these were routine transfusion<br />

<strong>and</strong> they were below 10 X109/L. <strong>and</strong> 26% of plt was<br />

given due to bleeding. However 10% of routine transfusion<br />

was given when platelet count was higher than 31 X 109/L.<br />

In overall 36% of the total platelet transfusion was given<br />

when platelet count was higher than 31 x109/L. 26% of the<br />

PT was given due to bleeding <strong>and</strong> 19% was given as unit<br />

policy i.e. anticoagulant treatment.<br />

Conclusion: Our organisation recognises the importance<br />

of risk associated in unnecessary blood product transfusion.<br />

Our 6 moths audit has shown interesting figures.<br />

In six months period total of 105 platelet transfusion carried<br />

out, only 36.5% of these transfusion was fitting the unit<br />

policy. 9.5% of these were given when platelet count was<br />

higher than 31 x109/L. There were various reason to transfuse<br />

platelet even though count was reasonable for haematology<br />

patient group such as bleeding <strong>and</strong> anticoagulant<br />

use.<br />

PP64 Transfuse or Not to Transfuse?<br />

Vieira S, Monroe D, Librojo M. London, United Kingdom<br />

Blood transfusion continues to be an essential part of<br />

modern practise but not without risks. During the last decade<br />

there has been an increase INTEREST in national<br />

level across the United Kingdom (UK) <strong>and</strong> Europe for collecting<br />

data on the hazards of transfusion of blood components.<br />

Therefore avoiding unnecessary transfusion is ac-<br />

117


Poster presentations Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

cepted as one way of reducing the risk which is associated<br />

with blood transfusion. In our stem cell transplant unit we<br />

have audited red cell transfusion frequency <strong>and</strong> reason for<br />

it. Unit policy is to keep Haemoglobin level above 8gr/dl,<br />

this may change time to time to 8.5 mg/dl depends on patient<br />

symptoms or if post chemo.<br />

Patients <strong>and</strong> methods: The audit was carried out prospectively.<br />

A log in book was kept in unit for recording the<br />

red cell transfusion orders. The data was validated with<br />

cross check retrospectively all inpatient record on monthly<br />

basis. The audit was carried over 6 months of period form<br />

May 2010 till end of October 2010.<br />

Results: During this 6 months period total of 90 episodes<br />

RCT performed in our unit. Haemoglobin level was 8.5 gr/L<br />

or below in 75.5% episodes, where as only 11% of the trans-<br />

118<br />

fusion performed when haemoglobin level was 9.1 gr/L or<br />

above.<br />

The reason for transfusion was categorised as routine as<br />

unit protocol, bleeding, pre procedure <strong>and</strong> on doctor’s instruction<br />

due to patient symptoms or keeping high trash<br />

hold haemoglobin level. Routine RCT was 87% of total<br />

episodes <strong>and</strong> 76% of this was 8.5 gr/L or below. However<br />

10% of the RCT transfusion was done when haemoglobin<br />

level is 9.1 gr/L or higher.<br />

Conclusion: Our organisation recognises the importance<br />

of risk associated in unnecessary blood product transfusion.<br />

Our 6 moths audit has shown interesting figures. 10% of<br />

total episodes RCT was given as routine were above 9.1 gr/<br />

L where as 70% of routine RCT was performed due to low<br />

haemoglobin level according to our unit policy.


Satellite Symposia<br />

Satellite Symposium:<br />

Therapy of fungal infections<br />

in immunocompromised patients<br />

SP01 Hematologic patients dying of IFI? Trends<br />

in IFI epidemiology<br />

Lubos Drgona, Slovakia<br />

Invasive fungal infections/diseases (IFD) <strong>are</strong> life threatening<br />

infectious complications in the population of immunocompromised<br />

patients. Patients with acute acute leukemia<br />

<strong>and</strong> patients undergoing hematopoietic stem cell transplantation<br />

(HSCT) <strong>are</strong> considered as the high risk group for<br />

development of IFD. During the last years of previous millenium<br />

an increase of incidence of invasive aspergillosis<br />

(IA) <strong>and</strong> decrease of invasive c<strong>and</strong>ida infection were observed<br />

at hematological departments. Today, invasive c<strong>and</strong>idaemia<br />

(IC) in high-risk haematological patient is quite r<strong>are</strong><br />

fungal infection when comp<strong>are</strong>d with IA but IC is still associated<br />

with high mortality (40–50%). Incidence of IC<br />

started to decline after the introduction of fluconazole to<br />

profylaxis of high risk patients during the 90-ties. But, results<br />

from our consecutive, prospective studies documented<br />

that the incidence of c<strong>and</strong>ideamia in Slovakia during the recent<br />

5 years is increasing. The population at risk is situated<br />

at ICUs <strong>and</strong> hematology-oncology departments <strong>and</strong> the<br />

most common pathogen is C.albicans in 38–50% of all episodes<br />

followed by C. parapsilosis in 26–29%. Infections<br />

caused by non-albicans species dominated at our hematooncology<br />

wards. The resistance of some non-albicans strains<br />

to fluconazole <strong>and</strong> voriconazole is slightly increasing. The<br />

local epidemiological data <strong>are</strong> important <strong>and</strong> they should<br />

help to guide the therapy for the patients with c<strong>and</strong>idaemia.<br />

The incidence of IA is 5–15% among the patients after<br />

allogeneic HSCT; the incidence in AML patients after induction<br />

therapy can reach 5–10%. Autopsy surveys identifyied<br />

the high prevalence of IA (30%) in patients with AML<br />

or after allogeneic HSCT but only about 1/3 of them were<br />

diagnosed before the death. Unfortunately, autopsy rate is<br />

very low in some countries. Several factors <strong>are</strong> associated<br />

with the increased risk for invasive fungal disease <strong>and</strong> higher<br />

mortality. Many studies have confirmed that good performance<br />

status, controlled malignancy, rapid neutropenia recovery,<br />

other than allogeneic transplantation <strong>and</strong> localized<br />

infection <strong>are</strong> factors associated with the higher probability<br />

of successfull outcome. The ascent of novel therapies for<br />

lymphoid malignancies (e.g. fludarabine, alemtuzumab) is<br />

associated with the higher incidence of opportunistic infections,<br />

including the IA. Despite the better knowledge of risk<br />

factors, useful diagnostic tools (e.g.GM, HRCT) <strong>and</strong> appropriate<br />

use of antifungal drugs – which <strong>are</strong> able to decrease<br />

mortality – the overall <strong>and</strong> infection-related mortality is still<br />

a problem. Mortality of high-risk haematological patients<br />

with IA is still high (40–80%) but there is a trend to decrease<br />

of mortality of IA confirmed by recently published<br />

studies. Our data from Czech <strong>and</strong> Slovak Fungal Infection<br />

Database (FIND) showed the overall mortality of 59% at 1<br />

year after the diagnosis of IA. The long-term overall survival<br />

of patients patients with IA who failed on the 1.line<br />

therapy was only 30% according to our observation in another<br />

survey. Genetic <strong>and</strong> prognostic risk factors <strong>are</strong> more<br />

widely studied now to better define the high-risk population<br />

<strong>and</strong> to improve the selection of appropriate antifungal strategy.<br />

A better knowledge about the epidemiology <strong>and</strong> risk factors<br />

is one of the key issues in the complex management of<br />

haematologic patients with IFD <strong>and</strong> may help to decrease<br />

mortality.<br />

SP02 Invasive aspergillosis in hematological<br />

patients – from empirical to early<br />

pre-emptive therapy<br />

Michael Girschikofsky, Austria<br />

The management of invasive fungal infection (IFI) is still<br />

a major challenge in patients with hematological malignancies<br />

or undergoing allogeneic stem cell transplantation. The<br />

incidence depends on the severity <strong>and</strong> duration of neutropenia,<br />

co-morbidity <strong>and</strong> co-medication, as well as local hospital<br />

factors like reconstruction works. Despite development<br />

of novel agents the mortality of proven invasive aspergillosis<br />

remains about 50%, which is mainly attributed to a delayed<br />

onset of antifungal therapy.<br />

Several guidelines support the decision finding for a prophylactic<br />

use of antimycotics in this high risk population.<br />

Nevertheless about 30% of patients develop neutropenic fever<br />

<strong>and</strong> 72–96 hours after failure of broad spectrum antibiotics<br />

the use of empirical antifungal therapy is considered as<br />

st<strong>and</strong>ard of c<strong>are</strong>. Overtreatment und drug related toxicity is<br />

a well known problem of this approach. Targeting the time<br />

frame between the first evidence of pathogens <strong>and</strong> manifest<br />

clinically signs of IFI, the pre-emptive strategy which is<br />

based on regularly screening with non-culture microbiological<br />

techniques (aspergillus galactomannan <strong>and</strong> PCR)<br />

<strong>and</strong> early CT-scanning may lead to a more effective use of<br />

antimycotic therapy.<br />

The detection of fungal associated signs in computed tomography<br />

scan (CT) is helpful but never pathognomonic.<br />

Thus, if clinically justifiable invasive diagnostic procedures,<br />

like CT guided biopsy <strong>are</strong> recommended to exclude other<br />

diseases or to determine the fungal species.<br />

119


Satellite Symposia Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

SP03 Management of Invasive aspergillosis:<br />

guidelines at the bedside<br />

Ben De Pauw (The Netherl<strong>and</strong>s)<br />

The relative incidence of the various fungal infections depends<br />

on geography as well as on local medical practices in<br />

a given hospital. In general, the number of patients at risk of<br />

invasive fungal infections has increased over the last decennium<br />

with the introduction of modern immunosuppressive<br />

<strong>and</strong> monoclonal antibodies for the treatment of both malignant<br />

<strong>and</strong> immune-mediated diseases. The problems in the<br />

management of invasive fungal disease hinge around three<br />

pivotal factors: 1) Difficulty to establish the diagnosis in an<br />

early stage of development; 2) Availability of safe, efficacious<br />

<strong>and</strong> affordable antifungal agents; 3) Impact of the<br />

evolvement of the underlying immune deficiency on the<br />

outcome of the infection.<br />

Diagnostic tools, notably serological tests <strong>and</strong> imaging<br />

techniques have improved over the last decade but their still<br />

doubts about their potency to detect an invasive fungal infection<br />

in an early stage of disease, partly because the required<br />

knowledge <strong>and</strong> facilities <strong>are</strong> only available at specialized<br />

centers where only a minority of the patients is being<br />

treated. As to avoid the confrontation with a stage of<br />

infection beyond a chance of cure, strategies based on early<br />

intervention became very popular. However, after having<br />

r<strong>and</strong>omized more than 7000 patients in clinical trials there<br />

is still no convincing scientific evidence to support a theoretically<br />

attractive prophylactic strategy. Using fluconazole<br />

as prophylaxis against c<strong>and</strong>idiasis in reasonably well defined<br />

risk groups appe<strong>are</strong>d effective but the story is different<br />

for mould infections. Even the most recent trials on posaconazole<br />

in the prevention of aspergillosis failed to provide a<br />

definite answer. Moreover, the risk of inducing resistance of<br />

selection of resistant stains is the price to be paid for intensive<br />

use of antifungals as has been witnessed after the introduction<br />

of fluconazole. Empirical treatment, i.e. institution<br />

of systemically active antifungals for fever not responding<br />

to adequate antibacterial therapy, was originally designed to<br />

eliminate life-threatening c<strong>and</strong>idosis but gained general recognition<br />

as an anti-aspergillosis strategy as well. As there<br />

<strong>are</strong> myriad causes of fever, it is inevitable that, following an<br />

empirical strategy, many patients will receive antifungals<br />

without actually requiring them. Improved diagnostics<br />

might help to reduce the number of c<strong>and</strong>idates for antifungals<br />

substantially; it is expected that such an pre-emptive<br />

strategy will become more popular.<br />

Systemically active antifungal compounds from four different<br />

classes of drugs <strong>are</strong> regularly used to treat invasive<br />

fungal infections. Amphotericin B was the drug of choice<br />

for all invasive fungal infections for more than 30 years.<br />

Presently, fluconazole is prescribed mainly to treat new infections<br />

by C<strong>and</strong>ida species <strong>and</strong> Cryptococcus neoformans.<br />

Recommendations on the management of aspergillosis for<br />

high-risk patients is now confusing since posaconazole<br />

would appear the drug of choice for prophylaxis, while voriconazole<br />

remains the drug of choice for the treatment of<br />

proven <strong>and</strong> probable aspergillosis leaving caspofungin <strong>and</strong><br />

liposomal amphotericin B the preferred drugs for empirical<br />

120<br />

therapy. This is the unfortunate consequence of mixing efficacy<br />

studies that ought to be designed to explore the options<br />

<strong>and</strong> limitations of a given drug with strategy trials.<br />

It has become customary to issue guidelines for the management<br />

of life-threatening diseases such as invasive fungal<br />

infections. These guidelines <strong>are</strong> supposed to offer the clinician<br />

easy access to the overwhelming amount of sometimes<br />

confusing data assuming that the assorted information will<br />

help to select the most suitable treatment for the seriously ill<br />

patient. Moreover, patients <strong>and</strong> authorities alike dem<strong>and</strong><br />

clear regulations to provide insight in the medical procedures<br />

<strong>and</strong> an objective benchmark for quality control, repectively.<br />

It is obvious that administrative people <strong>and</strong> clinician<br />

appreciate treatment guidelines in a different way. The individual<br />

patient occupies the central position in the vision of<br />

the clinician, while administrators see a group of patients<br />

with a similar disease who require a particular therapy.<br />

Guidelines for its treatment have been published in English<br />

by working parties from various countries <strong>and</strong> as all<br />

committees had the same data set at their disposal, great<br />

congruency amongst the various documents could be expected.<br />

The differences, however, <strong>are</strong> considerable. Most<br />

discrepancies <strong>are</strong> related to a different interpretation of the<br />

clinical trials. Empirical trials <strong>and</strong> salvage studies, which<br />

<strong>are</strong> rather strategic than drug-efficacy studies <strong>are</strong> of no value<br />

at all. The value of clinical trials for this purpose is largely<br />

overestimated, especially by unexperienced practitioners<br />

<strong>and</strong> regulatory authories. Therefore the published guidelines<br />

for the treatment of invasive fungal disease have to be<br />

taken into consideration but applied in clinical practice with<br />

prudence. They <strong>are</strong> provided for groups but there <strong>are</strong> frequently<br />

good reasons to deviate from the recommendations<br />

in individual cases.<br />

Satellite Symposium:<br />

Iron chelation therapy for MDS<br />

SP04 The Role of Iron Chelation Therapy in MDS<br />

Heather A. Leitch (Canada)<br />

Most patients with MDS develop RBC transfusion dependence<br />

<strong>and</strong> resulting iron overload (IOL) impacts negatively<br />

on survival. 1 Mechanisms of toxicity may include<br />

non-transferrin-bound iron (NTBI) resulting in oxidative<br />

stress, damaging lipids, proteins, <strong>and</strong> nucleic acids, <strong>and</strong> potentially<br />

leading to apoptosis or mutagenesis <strong>and</strong> malignant<br />

progression, both features of MDS. Guidelines recommend<br />

iron chelation therapy in lower-risk MDS with reasonable<br />

life expectancy, where retrospective studies show a survival<br />

benefit; these data will be reviewed. 2–4<br />

Chelation is also considered for patients eligible for stem<br />

cell transplantation (SCT), where decreased survival <strong>and</strong><br />

increased treatment-related mortality with IOL is seen, a<br />

sharp increase in NTBI occurs, <strong>and</strong> lower infection risk, delayed<br />

leukaemic transformation <strong>and</strong> improved outcome<br />

with chelation <strong>are</strong> suggested. 5,6<br />

While awaiting results of phase 3 trials of chelation in<br />

MDS, treatment should be individualized <strong>and</strong> monitored ac-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Satellite Symposia<br />

cording to guidelines. If observations that chelation could<br />

improve outcome in higher-risk MDS <strong>are</strong> confirmed, future<br />

expansion in the role for iron-lowering therapy may be<br />

seen.<br />

R E F E R E N C E S<br />

1. Sanz G, Nomdedeu B, Such E, Bernal T, Belkaid M, Ardanaz MT, et al.<br />

Independent impact of iron overload <strong>and</strong> transfusion dependency on survival<br />

<strong>and</strong> leukemic evolution in patients with myelodysplastic syndrome.<br />

Blood 2008;112:[abstract 640].<br />

2. Leitch HA, Leger CS, Goodman TA, Wong KK, Wong DHC, Ramadan<br />

KM, et al. Improved survival in patients with myelodysplastic syndrome<br />

receiving iron chelation therapy. Clin Leuk 2008;2:205–11.<br />

3. Fox F, Kündgen A, Nachtkamp K, Strupp C, Haas R, Germing U, Gattermann<br />

N. Matched-pair analysis of 186 MDS patients receiving iron<br />

chelation therapy or transfusion therapy only. Blood 2009;114:[abstract<br />

1747].<br />

4. Rose C, Brechignac S, Vassilief D, Pascal L, Stamatoullas A, Guerci A,<br />

et al. Does iron chelation therapy improve survival in regularly transfused<br />

lower risk MDS patients? A multicenter study by the GFM. Leuk<br />

Res. [Epub ahead of print 2010 Feb 1].<br />

5. Pullarkat V, Blanchard S, Tegtmeier B, Dagis A, Patane K, Ito J, Forman<br />

SJ. Iron overload adversely affects outcome of allogeneic hematopoietic<br />

cell transplantation. Bone Marrow Transplant 2008;42:799–805.<br />

6. Sorror ML, Storer BE, Schoch G, S<strong>and</strong>maier BM, Martin PJ, Scott BL,<br />

et al. Low albumin, high ferritin, <strong>and</strong> thrombocytopenia before transplant<br />

predict non-relapse mortality (NRM) independent of the hematopoietic<br />

cell transplantation comorbidity index (HCT-CI). Blood 2009;<br />

114:[abstract 651].<br />

SP05 Which MDS patients <strong>are</strong> eligible for iron<br />

chelation therapy?<br />

Aristoteles Giagounidis (Germany)<br />

SP06 Deferasirox in iron-overloaded patients<br />

with transfusion-dependent MDS<br />

de Witte T, de Swart L, Swinkels D, MacKenzie M<br />

(The Netherl<strong>and</strong>s)<br />

Myelodysplastic syndromes (MDS) <strong>are</strong> a heterogeneous<br />

group of clonal hematopoietic stem cell disorders. They <strong>are</strong><br />

characterized by abnormal differentiation <strong>and</strong> maturation of<br />

myeloid cells, bone marrow failure <strong>and</strong> genetic instability<br />

with an enhanced risk of progression to secondary leukaemia.<br />

The International Prognostic Scoring System (IPSS)<br />

for MDS is based on the percentage of marrow blasts, the<br />

number of cytopenias <strong>and</strong> cytogenetic characteristics <strong>and</strong><br />

has shown to be effective in predicting outcome mainly for<br />

MDS patients treated with supportive c<strong>are</strong> 1 . A new scoring<br />

system, the WHO Prognostic Scoring System (WPSS), is<br />

based on the WHO classification, transfusion dependency<br />

<strong>and</strong> cytogenetic abnormalities.<br />

Diagnosis <strong>and</strong> treatment of iron overload <strong>and</strong> iron toxicity<br />

is of great importance in the clinical management of<br />

MDS patients, because iron overload is likely an important<br />

predictor of morbidity <strong>and</strong> mortality. In a retrospective<br />

study 2 , heart failure was the most important non-leukemic<br />

cause of death (50%), followed by infection (31%) <strong>and</strong> liver<br />

cirrhosis (8%). These causes can be the result of iron overload.<br />

Iron overload in MDS is mainly caused by red blood cell<br />

transfusions 3 . Transfused iron probably accumulates initially<br />

in the liver before it is loaded into the heart as is described<br />

in β-thalassemia patients 4 . In a study with transfusion-dependent<br />

MDS patients, iron accumulation of the heart occurred<br />

after 75–100 blood transfusions when patients did<br />

not receive iron chelation therapy 6 .<br />

Ineffective erythropoiesis may also play an important<br />

role in iron accumulation in MDS patients in addition to red<br />

blood cell transfusions associated iron overload. Similar to<br />

β-thalassemia patients, ineffective erythropoiesis may lead<br />

to an increased growth differentiation factor (GDF)15 level,<br />

which subsequently lowers the hepcidin levels <strong>and</strong> results in<br />

an increased iron absorption from the intestinal tract 7 . Until<br />

now, only small studies have been performed in MDS patients<br />

to evaluate the hepcidin level 8–10 , but GDF15 has<br />

shown to be increased in RARS patients 11 . It is not clear if<br />

ineffective erythropoiesis itself leads to significant symptomatic<br />

iron overload. However, serum ferritin levels can be<br />

elevated before blood transfusions <strong>are</strong> given, especially in<br />

patients with RARS 12 , suggesting that iron overload is already<br />

present.<br />

Besides iron accumulation, iron toxicity probably plays<br />

an important role in MDS. Free iron in the plasma becomes<br />

available when the capacity of transferrin to carry iron is<br />

exceeded. This non-transferrin-bound iron (NTBI) is even<br />

elevated in MDS patients who did not yet receive any blood<br />

cell transfusions <strong>and</strong> is associated with a higher level of<br />

apoptosis 13 . In a recent study with thalassemia patients,<br />

NTBI was correlated with transferrin saturation but not with<br />

serum ferritin level. Patients with heart diseases had significantly<br />

higher NTBI levels than those without heart disease,<br />

suggesting that NTBI will be responsible for functional organ<br />

damage 14 . The presence of NTBI will be reflected in a<br />

rise of labile plasma iron (LPI) <strong>and</strong> reactive oxygen species<br />

(ROS) that causes cell <strong>and</strong> tissue damage 15,16 . LPI is the<br />

fraction of NTBI that is redox active <strong>and</strong> eliminated by iron<br />

chelators 17 . The determination of NTBI remains difficult<br />

<strong>and</strong> new methods <strong>are</strong> under investigation to improve the reliability<br />

of this important iron toxicity parameter. In the near<br />

future a more reliable assessment of NTBI in combination<br />

with LPI will be most valuable to determine iron toxicity in<br />

MDS patients.<br />

Recently new oral iron chelation therapy has become<br />

available (deferasirox), which makes iron chelation therapy<br />

more feasible <strong>and</strong> effective despite possible side effects.<br />

Several prospective studies have demonstrated that oral<br />

iron chelation therapy is effective in the reduction of the<br />

iron accumulation, but it is not yet clear which patients benefit<br />

most of this treatment 18–21 . Iron chelation therapy will<br />

probably reduce iron accumulation in end target organs as<br />

well as decrease iron toxicity by reducing potentially toxic<br />

iron 19,22 . Some studies have shown benefit of iron chelation<br />

therapy for survival in MDS patients 23,24 .<br />

Several guidelines for iron chelation therapy in MDS patients<br />

have been published the last years, without clear evidence<br />

for the optimum treatment of iron overload 25 . By<br />

gaining more insight into the pathophysiology <strong>and</strong> toxicity<br />

of iron overload in MDS patients, the guidelines for iron<br />

121


Satellite Symposia Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

chelation therapy can be optimized to reduce iron related<br />

morbidity <strong>and</strong> mortality.<br />

An ongoing registry study organized by the MDS Working<br />

Package of the European LeukemiaNet has collected<br />

prospectively demographic <strong>and</strong> treatment data on more than<br />

1,000 patients with lower risk MDS from 15 European<br />

countries. An interim analysis presented at the most recent<br />

EHA meeting showed that transfusions dependent patients<br />

had a 4.1 times higher chance of dying during a short observation<br />

period of 18 months only. This registry consortium<br />

will continue to collect data, including data on the impact of<br />

iron chelation on outcome.<br />

R E F E R E N C E S<br />

1. Greenberg P, Cox C, LeBeau MM et al. International scoring system<br />

for evaluating prognosis in myelodysplastic syndromes. Blood 1997;89<br />

(6):2079–2088.<br />

2. Malcovati L, Porta MG, Pascutto C et al. Prognostic factors <strong>and</strong> life<br />

expectancy in myelodysplastic syndromes classified according to WHO<br />

criteria: a basis for clinical decision making. J Clin Oncol 2005;23<br />

(30):7594–7603.<br />

3. Porter JB. Practical management of iron overload. Br J Haematol<br />

2001;115(2):239–252.<br />

4. Chacko J, Pennell DJ, Tanner MA et al. Myocardial iron loading by<br />

magnetic resonance imaging T2* in good prognostic myelodysplastic<br />

syndrome patients on long-term blood transfusions. Br J Haematol<br />

2007;138(5):587–593.<br />

5. Jensen PD, Jensen FT, Christensen T, Eiskjaer H, Ba<strong>and</strong>rup U, Nielsen<br />

JL. Evaluation of myocardial iron by magnetic resonance imaging during<br />

iron chelation therapy with deferrioxamine: indication of close relation<br />

between myocardial iron content <strong>and</strong> chelatable iron pool. Blood<br />

2003;101(11):4632–4639.<br />

7. Tanno T, Bhanu NV, Oneal PA et al. High levels of GDF15 in thalassemia<br />

suppress expression of the iron regulatory protein hepcidin. Nat<br />

Med 2007;13(9):1096–1101.<br />

8. Murphy PT, Mitra S, Gleeson M, Desmond R, Swinkels DW. Urinary<br />

hepcidin excretion in patients with low grade myelodysplastic syndrome.<br />

Br J Haematol 2009;144(3):451–452.<br />

9. Tanno T, Bhanu NV, Oneal PA et al. High levels of GDF15 in thalassemia<br />

suppress expression of the iron regulatory protein hepcidin. Nat<br />

Med 2007;13(9):1096–1101.<br />

10. Winder A, Lefkowitz R, Ghoti H et al. Urinary hepcidin excretion in<br />

patients with myelodysplastic syndrome <strong>and</strong> myelofibrosis. Br J Haematol<br />

2008;142(4):669–671.<br />

11. Ramirez JM, Schaad O, Durual S et al. Growth differentiation factor 15<br />

production is necessary for normal erythroid differentiation <strong>and</strong> is increased<br />

in refractory anaemia with ring-sideroblasts. Br J Haematol<br />

2009;144(2):251–262.<br />

12. Gattermann N. Clinical consequences of iron overload in myelodysplastic<br />

syndromes <strong>and</strong> treatment with chelators. Hematology/Oncology<br />

Clinics 2005;19:13–17.<br />

13. Cortelezzi A, Cattaneo C, Cristiani S et al. Non-transferrin-bound iron<br />

in myelodysplastic syndromes: a marker of ineffective erythropoiesis?<br />

Hematol J 2000;1(3):153–158.<br />

14. Piga A, Longo F, Duca L et al. High nontransferrin bound iron levels <strong>and</strong><br />

heart disease in thalassemia major. Am J Hematol 2009;84(1):29–33.<br />

15. Esposito BP, Breuer W, Sirankapracha P, Pootrakul P, Hershko C, Cabantchik<br />

ZI. Labile plasma iron in iron overload: redox activity <strong>and</strong><br />

susceptibility to chelation. Blood 2003;102(7):2670–2677.<br />

16. Ghoti H, Amer J, Winder A, Rachmilewitz E, Fibach E. Oxidative<br />

stress in red blood cells, platelets <strong>and</strong> polymorphonuclear leukocytes<br />

from patients with myelodysplastic syndrome. Eur J Haematol 2007;<br />

79(6):463–467.<br />

17. Pootrakul P, Breuer W, Sametb<strong>and</strong> M, Sirankapracha P, Hershko C,<br />

Cabantchik ZI. Labile plasma iron (LPI) as an indicator of chelatable<br />

plasma redox activity in iron-overloaded beta-thalassemia/HbE patients<br />

treated with an oral chelator. Blood 2004;104(5):1504–1510.<br />

18. Gattermann N, Schmid M, Della PM, et al. Efficacy <strong>and</strong> safety of deferasirox<br />

during 1 year of treatment in transfusion-dependent patients<br />

122<br />

with myelodysplastic syndromes: results from EPIC trial. ASH Annual<br />

Meeting Abstracts 633. 2008. Ref Type: Abstract<br />

19. List AF, Baer MR, Steensma DP, et al. Iron chelation with Deferasirox<br />

improves iron burden in patients with myelodysplastic syndromes<br />

(MDS). ASH Annual Meeting Abstracts 634. 2008. Ref Type: Abstract<br />

20. Porter J, Galanello R, Saglio G et al. Relative response of patients with<br />

myelodysplastic syndromes <strong>and</strong> other transfusion-dependent anaemias<br />

to deferasirox (ICL670): a 1-yr prospective study. Eur J Haematol<br />

2008;80(2):168–176.<br />

21. Wood JC, Kang BP, Thompson A et al. The effect of deferasirox on<br />

cardiac iron in thalassemia major: impact of total body iron stores.<br />

Blood 2010;116(4):537–543.<br />

22. Ghoti H, Amer J, Winder A, Rachmilewitz E, Fibach E. Oxidative<br />

stress in red blood cells, platelets <strong>and</strong> polymorphonuclear leukocytes<br />

from patients with myelodysplastic syndrome. Eur J Haematol 2007;<br />

79(6):463–467.<br />

23. Rose C, Brechignac S, Vassilief D et al. Does iron chelation therapy<br />

improve survival in regularly transfused lower risk MDS patients? A<br />

multicenter study by the GFM (Groupe Francophone des Myelodysplasies).<br />

Leuk Res 2010;34(7):864–870.<br />

24. Leitch HA, Leger CS, Goodman TA et al. Improved survival in patients<br />

with myelodysplastic syndrome receiving iron chelation therapy. Clinical<br />

Leukemia 2008;2(3):205–211.<br />

25. Messa E, Cilloni D, Saglio G. Iron chelation therapy in myelodysplastic<br />

syndromes. Adv Hematol 2010;2010:756289.<br />

Satellite Symposium<br />

The Optimal Management of CML<br />

SP07 Nilotinib versus Imatinib for Newly<br />

Diagnosed CML patients: ENESTnd study<br />

results<br />

Giuseppe Saglio (Italy)<br />

Patients with newly diagnosed chronic phase (CP) chronic<br />

myeloid leukemia (CML) have a high probability of longterm<br />

survival, which is mainly due to the efficacy of imatinib,<br />

the first targeted inhibitor of the oncogenic BCR-<br />

ABL1 kinase to be registered. In a recent report from IRIS<br />

study, patients in the imatinib arm had a 8-year rate of overall<br />

survival (OS) of 85% <strong>and</strong> freedom from progression to<br />

accelerated- (AP) or blast-phase (BP) disease of 93%. Cumulative<br />

rates of complete cytogenetic response (CCyR)<br />

were 69% at 1 year, 87% at 5 years, <strong>and</strong> 89% at 8 years.<br />

These data highlight that for at least one-third of patients,<br />

the potential exists to improve on what can be achieved with<br />

st<strong>and</strong>ard imatinib 400 mg/d treatment. After imatinib failure,<br />

current treatment recommendations include dose escalation<br />

to 600–800 mg/d, or switch to an approved next-generation<br />

tyrosine kinase inhibitor (TKI. Although stem cell<br />

transplant (SCT) remains an important <strong>and</strong> potentially curative<br />

option, favorable response rates <strong>and</strong> tolerability achieved<br />

with TKIs have resulted in SCT being increasingly reserved<br />

for patients who have failed on TKIs <strong>and</strong> for those who have<br />

progressed to advanced phases. The results from ENESTnd<br />

trial testing nilotinib at two different dosages, 300mg BID<br />

<strong>and</strong> 400 BID, clearly indicate a superior efficacy of the more<br />

potent second-generation TKI nilotinib with respect to imatinib,<br />

particularly in terms of higher rates of major molecular<br />

responses (MMR), of complete cytogenetic responses<br />

(CCyR) that, more importantly, lead to a decreased number<br />

of progressions to more advances phases of the disease.


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Satellite Symposia<br />

These results will probably modify imatinib-based strategies<br />

in the first-line therapy of CML <strong>and</strong> <strong>are</strong> becoming a key<br />

<strong>are</strong>a of clinical research during the next few years.<br />

REFERENCES<br />

1. Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib versus Imatinib for<br />

Newly Diagnosed Chronic Myeloid Leukemia. N Engl J Med 2010;362:<br />

2251–2259.<br />

2. Kantarjian H, Shah NP, Hochhaus A, et al. Dasatinib versus Imatinib in<br />

Newly Diagnosed Chronic-Phase Chronic Myeloid Leukemia. N Engl J<br />

Med 2010;362:2260–2270.<br />

SP08 Optimal treatment for newly diagnosed<br />

CML patients<br />

Francis Giles (Irel<strong>and</strong>)<br />

Nilotinib (Tasigna ® ) is a much more potent <strong>and</strong> selective<br />

BCR-ABL inhibitor than imatinib <strong>and</strong> was designed to<br />

overcome imatinib’s deficiencies. Nilotinib has very significant<br />

efficacy in patients with CML in chronic (CP) <strong>and</strong><br />

accelerated (AP) following imatinib failure. Based on the<br />

results of the Evaluating Nilotinib Efficacy <strong>and</strong> Safety in<br />

Clinical Trials-Newly Diagnosed Patients (ENESTnd) study,<br />

the Food <strong>and</strong> Drug Administration (FDA) on June 17 th 2010<br />

granted approval of nilotinib for the treatment of adult patients<br />

with newly diagnosed Philadelphia-chromosome positive<br />

CML in CP. Imatinib changed our perceptions of the<br />

therapeutic power of targeted inhibition of a pathologically<br />

active kinase. Nilotinib, a designer agent built on the imatinib<br />

scaffold, has proven superior to its template agent by<br />

every significant surrogate marker used in monitoring CML.<br />

The largest CML-related threat to life on imatinib therapy is<br />

development of AP/BP within the first 3 years of imatinib<br />

therapy. In terms of both the clinically relevant surrogate<br />

markers, CCyR <strong>and</strong> MMR, <strong>and</strong> of the clinically vital endpoints<br />

of early transformation events <strong>and</strong> overall survival,<br />

nilotinib has proved superior to imatinib as first line therapy<br />

in CML. Nilotinib has a low incidence of grade 3/4 extramedullary<br />

adverse events, predominantly transient biochemical<br />

abnormalities, which tend to occur early in therapy.<br />

Nilotinib in the frontline setting is associated with less<br />

myelosuppression than imatinib. The ENEST1st study, a<br />

pan-European study, will further examine the efficacy of nilotinib<br />

300mg BID in newly diagnosed patients with CML<br />

using CMR as the primary study endpoint. The ENEST1st<br />

study has a range of integral international collaborative biology<br />

studies which include a focus on CML stem cells,<br />

indices of genomic instability, very sensitive mutated bcrabl<br />

phenotype clone detection <strong>and</strong> quantitation, <strong>and</strong> genomic<br />

analyses. Nilotinib PK <strong>and</strong> patient adherence to therapy<br />

will also be focused on in this study.<br />

R E F E R E N C E S<br />

1. Hochhaus A, Saglio G, le Coutre PD, et al. Superior efficacy of nilotinib<br />

comp<strong>are</strong>d with imatinib in newly-diagnosed patients with chronic myeloid<br />

leukemia in chronic phase (CML-CP): ENESTnd minimum 24month<br />

follow-up. Paper presented at: 16th Annual Congress of the European<br />

Hematology Association; June 9–12, 2011; London, UK.<br />

2. Saglio G, Hochhaus A, Guilhot F, et al. Nilotinib is associated with<br />

fewer treatment failures <strong>and</strong> suboptimal responses vs imatinib in pa-<br />

tients with newly diagnosed chronic myeloid leukemia in chronic phase<br />

(CML-CP): results from ENESTnd. Paper presented at: 16th Annual<br />

Congress of the European Hematology Association; June 9–12, 2011;<br />

London, UK.<br />

3. Weisberg E, Manley PW, Cowan-Jacob SW, Hochhaus A, Griffin JD.<br />

Second generation inhibitors of BCR-ABL for the treatment of imatinib-resistant<br />

chronic myeloid leukaemia. Nat Rev Cancer. 2007;7(5):<br />

345–356.<br />

4. Hughes TP, Hochhaus A, Saglio G, et al; for the ENESTnd investigators.<br />

ENESTnd 24-month update: continued superiority of nilotinib versus<br />

imatinib in patients with newly diagnosed chronic myeloid leukemia<br />

in chronic phase (CML-CP). Slides presented at: 52nd Annual Meeting<br />

of the American Society of Hematology; December 4–7, 2010; Orl<strong>and</strong>o,<br />

FL. Abstract 207.<br />

5. Saglio G, Kim D-W, Issaragrisil S, et al; for the ENESTnd investigators.<br />

Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia.<br />

N Engl J Med. 2010;362(24):2251–2259<br />

6. Pinilla-Ibarz J, Cortes J, Mauro MJ. Intolerance to tyrosine kinase inhibitors<br />

in chronic myeloid leukemia: definitions <strong>and</strong> clinical implications.<br />

Cancer. 2011;117(4):688–697.<br />

7. Kantarjian HM, Giles FJ, Bhalla KN, et al. Nilotinib is effective in patients<br />

with chronic myeloid leukemia in chronic phase following imatinib<br />

resistance or intolerance: 24-month follow-up results. Blood. 2010;<br />

117(4):1141–1145.<br />

SP09 The importance of molecular monitoring<br />

in CML patients<br />

Martin M. Mueller (Germany)<br />

Topic: The importance of molecular monitoring in CML<br />

patients<br />

Abstract: Molecular characterization of diseases has<br />

been implemented in routine work-up of hematologic patients<br />

in many countries. Molecular monitoring of CML patients<br />

represents one of the most advanced fields applying<br />

qualitative <strong>and</strong> quantitative RT-PCR using peripheral blood<br />

samples. Besides mentioning the advantages <strong>and</strong> disadvantages<br />

of the techniques, the differences comp<strong>are</strong>d to cytogenetic<br />

assessment as the current gold-st<strong>and</strong>ard will be highlighted.<br />

Quantification of BCR-ABL mRNA transcripts<br />

represents the most sensitive tool of monitoring CML patients<br />

allowing to predict for future response at early timepoints<br />

<strong>and</strong> to find suboptimal treatment outcomes triggering<br />

mutation analysis. Further, current European efforts of harmonizing<br />

laboratories for BCR-ABL quantification will be<br />

discussed.<br />

Satellite Symposium:<br />

Optimal Management of PNH<br />

SP10 PNH – Morbidities & Mortality<br />

Boris Labar (Croatia)<br />

Paroxysmal nocturnal hemoglobinuria (PNH) is a r<strong>are</strong>,<br />

chronic, life-threatening disorder caused by the somatic<br />

mutation of the PIG A gene in hematopoetic stem cells (1).<br />

Because of this mutation PNH cells <strong>are</strong> missing glycosilphosphatidylinositol<br />

(GPI) So GPI anchored proteins <strong>are</strong><br />

not bind <strong>and</strong> present on the cell surface. For PNH it is important<br />

that CD59 (1)(forms a defensive shield for erythrocytes<br />

from complement-mediated lysis) <strong>and</strong> CD55 (2) (pre-<br />

123


Satellite Symposia Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

vents formation <strong>and</strong> augments the instability of the C3 convertases,<br />

attenuating the complement cascades) <strong>are</strong> not<br />

present at the surface of red cells. Such cells underwent hemolysis<br />

because of uncontrolled complement activation (3).<br />

As a consequence of chronic hemolysis, progressive morbidities<br />

could develop such as thrombotic events (TEs),<br />

chronic kidney disease (CKD), <strong>and</strong> pulmonary hypertension.<br />

These <strong>and</strong> other PNH-associated symptoms (ie, abdominal<br />

pain, dyspnea, dysphagia, fatigue, hemoglobinuria,<br />

<strong>and</strong> erectile dysfunction) have a significant effect on patient<br />

quality of life (QoL). The 5-year mortality rate for patients<br />

with PNH is ∼35%,(4) <strong>and</strong> the leading cause of death is<br />

TE.(5) As many as 29% to 44% of patients with PNH suffer<br />

from at least one TE during their disease,(6) <strong>and</strong> TE may<br />

occur regardless of transfusion history. Many patients with<br />

PNH have impaired renal function, with up to 64% having<br />

CKD; renal failure is a common (8% to 18%) cause of death<br />

among patients with PNH.(7, 8) Nearly 50% of patients<br />

have some evidence of pulmonary hypertension.(9) Although<br />

not life-threatening, fatigue <strong>and</strong> pain <strong>are</strong> common in<br />

patients with PNH <strong>and</strong> can substantially impair QoL.<br />

Historically, PNH has been managed with transfusions,<br />

red blood cell supplements, anticoagulants, <strong>and</strong> steroids,(5,<br />

10) none of which have reduced the risks of disease progression<br />

or severe morbidity. Hematopoeitic stem cell transplant<br />

is associated with a high risk of morbidity <strong>and</strong> mortality;<br />

thus, for most patients, the associated risks preclude this option.<br />

As such, recent studies in PNH have focused on inhibiting<br />

the complement system to reduce the chronic hemolysis<br />

underlying the progressive morbidities <strong>and</strong> mortality.<br />

Treatment with eculizumab, a fully humanized monoclonal<br />

antibody that inhibits terminal complement activity, can<br />

normalize the survival of patients with PNH(11).<br />

R E F E R E N C E S<br />

1. Johnson JR et al. J Clin Pathol Mol Pathol. 2002;55:145–52<br />

2. Brodsky RA. In Hematology, Basic Principles <strong>and</strong> Practices, 4th ed. R<br />

Hoffman, EJ et al. Eds Philadelphia, Elsevier Churchil Livingstone,<br />

2005, pp 419–27.<br />

3. Rother RP et al. Nat Biotechnol. 2007;25:1256–1264.<br />

4. Hillmen P et al. N Engl J Med. 1995;25:1253–1258<br />

5. Parker C et al. Blood. 2005;106:3699–3709<br />

6. Hillmen P et al. Blood. 2007;110:4123–4128.<br />

7. Hillmen P et al. Am J Hematol. 2010;85:553–559.<br />

8. Nishimura J et al. Medicine (Baltimore). 2004;83:193–207.<br />

9. Hill A. et al. Br J Haematol. 2007;137:181–192<br />

10. Rachidi S et al. Eur J Intern Med. 2010;21:260–267.<br />

11. Kelly RJ et al. Blood. 2011;117:6786–6792<br />

SP11 Shifting the Paradigm in PNH Management<br />

Peter Hillmen (United Kingdom)<br />

Paroxysmal nocturnal hemoglobinuria (PNH) is a r<strong>are</strong>,<br />

acquired, chronic <strong>and</strong> life-threatening hematopoietic stem<br />

cell disorder characterized by chronic uncontrolled activation<br />

of the complement system. 1–3<br />

The primary clinical manifestation of PNH is chronic hemolysis,<br />

which is associated with severe morbidities, including<br />

thromboembolism (TE), chronic kidney disease<br />

(CKD), anemia, <strong>and</strong> fatigue. 4 For patients with PNH, treatment<br />

with eculizumab, a fully humanized monoclonal anti-<br />

124<br />

body that inhibits terminal complement activity, has represented<br />

a major paradigm shift.<br />

The clinical program of eculizumab in patients with PNH<br />

consists of 195 patients in total, <strong>and</strong> 4 trials [Pilot study<br />

(N=11), TRIUMPH (phase III, placebo-controlled trial)<br />

(N=87), SHEPHERD (broader patient population) (N=97)]<br />

<strong>and</strong> a subsequent long-term extension trial evaluating longterm<br />

efficacy <strong>and</strong> safety. 5 In the long-term trial, following<br />

eculizumab treatment, the reduction in lactate dehydrogenase<br />

(LDH) was sustained after 36 months follow-up (P <<br />

.0001), TE events were significantly reduced from 52 pretreatment<br />

to 10 trial events by matched time analysis (P <<br />

.0005), <strong>and</strong> CKD stage (stages 1–5) was reduced from 69%<br />

of patients at baseline to 31% at 36 months. 5 Hemoglobin<br />

increased significantly <strong>and</strong> was sustained over 36 months<br />

(mean increase over baseline 9.5 g/L; range, –31 to 68; P <<br />

.0001). Among patients receiving ≥ 36 months of eculizumab<br />

(n=87), 29% became transfusion independent <strong>and</strong> remained<br />

so throughout treatment. Eculizumab was well tolerated;<br />

most adverse events (AEs) were mild-to-moderate.<br />

Of the 10% of patients who discontinued (n=20), 9 were the<br />

result of an AE. In 16-weeks of follow-up, 3 of the 20 discontinuing<br />

patients had TE, including 1 death. Two cases of<br />

meningococcal sepsis were successfully treated without sequelae.<br />

Four patients died during study; 3 were considered<br />

unrelated <strong>and</strong> 1 possibly related to eculizumab. Per Kaplan<br />

Meier estimation, overall survival was 97.6% at 3 years <strong>and</strong><br />

maintained through 5.5 years of treatment; this comp<strong>are</strong>s<br />

favorably to a survival rate previously reported in historical<br />

controls (65% at 5 years).<br />

In another analysis of survival (N=79), the efficacy of<br />

eculizumab was sustained over 8 years of therapy <strong>and</strong> was<br />

not different to that in an age-, sex-matched normal control<br />

population 6 .<br />

In conclusion, eculizumab significantly reduces chronic<br />

hemolysis <strong>and</strong> TE, improves CKD <strong>and</strong> PNH-associated<br />

symptoms, <strong>and</strong> improves probability of survival. These effects<br />

<strong>are</strong> sustained during long-term treatment. Thus, eculizumab<br />

inhibition of terminal complement activity is an effective<br />

treatment approach for patients with PNH.<br />

Alexion Pharmaceuticals, Inc: Consultancy, Honoraria,<br />

Membership on an entity’s Board of Directors or advisory<br />

committees, Research Funding.<br />

R E F E R E N C E S<br />

1. Brodsky R, Schrezenmeier H, Muus P et al. Blood [ASH]. 2009;114:<br />

Abstract 3007.<br />

2. Brodsky RA. Blood. 2009;113(26):6522–6527.<br />

3. Hillmen P, Elebute M, Kelly R et al. Am J Hematol. 2010;85(8):553–<br />

559.<br />

4. Parker C, Omine M, Richards S et al. Blood. 2005;106(12):3699–3709.<br />

5. Hillmen P, Risitano A, Schrezenmeier H et al. Haematologica [EHA]<br />

2011;96(s2):105. Abstract 0254.<br />

6. Kelly RJ, Hill A, Arnold LM et al. Blood. 2011;117(25):6786–6792.<br />

SP12 Acute renal failure <strong>and</strong> PNH – Case report<br />

Jaroslav Cermak (Czech Republic)<br />

Diagnosis <strong>and</strong> management of patients with PNH can be<br />

challenging. This case-based presentation provides a real-


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Satellite Symposia<br />

world context for PNH treatment <strong>and</strong> clinical trial data <strong>and</strong><br />

provides a forum for discussion of their application to evidence-based<br />

medicine in day-to-day practice.<br />

Patients with PNH often experience increased rates of infection,<br />

fatigue, insomnia, pain in the joints <strong>and</strong> abdomen,<br />

dyspnea, <strong>and</strong> dysuria, each of which can undermine quality<br />

of life <strong>and</strong> can also lead to an increased risk of thrombotic<br />

events <strong>and</strong> mortality. 1,2 In patients who present with these<br />

symptoms, a differential diagnosis based on histologic <strong>and</strong><br />

flow cytometry analyses is essential. 3 With only supportive<br />

c<strong>are</strong>, PNH is invariably progressive. 4 In later stages, renal<br />

failure, infections, <strong>and</strong> cardiopulmonary <strong>and</strong> hematologic<br />

complications can become life-limiting. 1,3<br />

Historical therapies for PNH have included supportive<br />

c<strong>are</strong> with red blood cell transfusions <strong>and</strong> iron chelation as<br />

needed, steroids, antithrombotic prophylaxis, <strong>and</strong> dialysis. 1,4<br />

However, the introduction of eculizumab, a terminal complement<br />

inhibitor, has provided an option to treat the underlying<br />

disease rather than palliating disease symptoms <strong>and</strong><br />

end-organ damage. 4,5 Nonetheless, several questions remain:<br />

What is the role for stem cell transplant? Will antithrombosis<br />

prophylaxis be needed during eculizumab treatment?<br />

What is the role of eculizumab in patients with aplastic<br />

anemia/PNH syndrome <strong>and</strong> other PNH-related conditions?<br />

Generation of an international PHN registry may<br />

provide insight into these questions.<br />

R E F E R E N C E S<br />

1. Parker et al. Blood. 2005;106:3699–3709.<br />

2. Lee et al. Haematologica. 2010;95: Abstracts 505 <strong>and</strong> 506.<br />

2. Borowitz et al. Cytometry B Clin Cytom. 2010;78:211–230.<br />

3. Rachidi et al. Eur J Intern Med. 2010;21:260–267.<br />

4. Hill A et al. Haematologica. 2010;95:567–573.<br />

SP13 Treatment of Paroxismal nocturnal<br />

hemoglobinuria – a case report<br />

Ana Boban (Croatia)<br />

Paroxismal nocturnal hemoglobinuria (PNH) is clonal<br />

acquired disorder characterized by hemolytic anemia, thrombosis,<br />

fatigue <strong>and</strong> bone marrow failure. It is caused by acquired<br />

mutation of PIG-A gene in mutipotent hematopoietic<br />

stem cell. As a consequence, cells <strong>are</strong> lacking glycosil phos-<br />

phatidylinosiol (GPI) – anchored proteins, namely CD55<br />

<strong>and</strong> CG 59, which <strong>are</strong> complement regulatory proteins. The<br />

lack of complement regulatory proteins makes erythrocytes<br />

susceptible to both intravascular <strong>and</strong> extravascular hemolysis.<br />

Intravascular hemolysis is responsible for majority of<br />

morbidity form the disease. Eculizumab is a monoclonal<br />

antibody that inhibits terminal complement activation,<br />

which reduces intravascular hemolysis in PNH patients.<br />

Here we present the first PNH patient in our Center treated<br />

with eculizumab.<br />

The patient was diagnosed having aplastic anemia in<br />

2009, at the age of 21. The treatment started with corticosteroids,<br />

<strong>and</strong> later altered into cyclosporine. In 2005 the patient<br />

presented with severe headache <strong>and</strong> transitory ischemic attack.<br />

At that point PNH was suspected, <strong>and</strong> diagnostic procedure<br />

was carried out. Diagnose of PNH was made based<br />

on PNH clone on flow cytometry. In 2007, the progression<br />

of intravascular hemolysis was noted, with significant increase<br />

in bilirubin <strong>and</strong> lactate dehidrogenase levels, followed<br />

by the signs of bone marrow failure. The need for<br />

blood transfusions <strong>and</strong> fatigue increased, while quality of<br />

life decreased. Allogenic stem cell transplantation was indicated,<br />

but no related donor was found. The patient was eligible<br />

for eculizumab treatment.<br />

The treatment with eculizumab started in January 2010.<br />

Patient was vaccinated for Naisseria meningitides, <strong>and</strong> developed<br />

severe cellulitis with skin necrosis on the place of<br />

the puncture, hence the reconstructive surgery of the left upper<br />

arm was needed. At the time when eculizumab was<br />

started, patient had severe hemolysis, thrombocytopenia,<br />

<strong>and</strong> anemia with need of blood transfusions every 2–3 weeks<br />

<strong>and</strong> secondary hemochromatosis. Subjectively, she complained<br />

mostly on severe fatigue. The patient received induction<br />

treatment without side effects <strong>and</strong> continued with<br />

maintenance treatment for 19 months. A significant decrease<br />

of hemolysis was noted, <strong>and</strong> consequently reduced<br />

need for transfusions <strong>and</strong> disappearance of symptoms,<br />

mainly fatigue with dramatic improvement in her quality of<br />

life.<br />

Eculizumab treatment significantly improved quality of<br />

life of the PNH patient. Patient’s main symptom, fatigue,<br />

disappe<strong>are</strong>d almost completely, <strong>and</strong> rate of blood transfusion<br />

was reduced to once in 5–6 weeks. We found eculizumab<br />

effective in treatment of PNH patients.<br />

125


Author Index<br />

Abdel-Wahab, O., 23<br />

Ajdukovic, R., 88, 91, 98<br />

Aleksic, A., 98<br />

Amadori, S., 17, 22<br />

Andjelic, B., 105<br />

Andreola, G., 106<br />

Angelescu, S., 117<br />

Anicic, M., 108<br />

Antic, D., 102, 105, 109<br />

Antica, M., 87<br />

Arcasoy, M., 101<br />

Armitage, JO., 97<br />

Auon, P., 97<br />

Aurer, I., 65, 79, 92, 99, 100, 105,<br />

111, 112<br />

Avila, DN., 94<br />

Ayari, S., 93<br />

Babic, A., 106, 113, 115, 116<br />

Babok-Flegaric, R., 91<br />

Baird, K., 94<br />

Banfic, H., 87<br />

Baráth, S., 88<br />

Barbu, D., 117<br />

Barca, G., 117<br />

B<strong>are</strong>z, MY, 112<br />

Barisic, N., 96<br />

Barlogie, B., 48<br />

Barrett, J., 56<br />

Barsic, B., 103<br />

Basic-Kinda, S., 92, 99, 100, 104,<br />

105, 111, 112<br />

Bast, M., 97<br />

Batar, P., 110<br />

Batinic, D., 87, 90, 96, 104, 105, 107<br />

Batinic, J., 90, 105<br />

Begovic, D., 90<br />

Bijedic, V., 81<br />

Bila, J., 105<br />

Bilic, E., 96, 103, 108, 110<br />

Bishop, M. R., 60<br />

Biskup, M., 114<br />

Bizu, I., 117<br />

Bladé, J., 47<br />

Blaise, D., 23<br />

Blin, N., 93<br />

Boban, A., 104, 125<br />

Bodrozic, J., 105<br />

Bogdanovic, A., 109<br />

Bojanic, I., 95, 96, 105, 113, 114<br />

Bolaños Meade, J., 94<br />

Borbenyi, Z., 81, 88, 107<br />

Borckhardt, A., 19<br />

Bosnic, D., 112<br />

Brissot, E., 93<br />

Burnac, IL., 114<br />

Burnett, A., 21<br />

Carde, P., 63<br />

Carzavec, D., 91<br />

Catalano, J., 101<br />

Cermak, J., 124<br />

Cevreska, L., 81, 99, 102<br />

Chan, WC., 97<br />

Chevallier, P., 93<br />

Cigrovski, N., 99<br />

Cikota, B., 98<br />

Clavert, A., 93<br />

Coha, B., 91<br />

Cojbasic, I., 91, 99<br />

Colovic, N., 89, 108, 109<br />

Corovic-Arneri, E., 91<br />

Cortes, J., 3, 36<br />

Cowen, EW., 94<br />

Crkvenac Gornik, K., 114<br />

Crncec, I., 88<br />

Crowley, J., 48<br />

Dávid, M., 88, 107<br />

Davidovic, S., 89, 90, 91, 92,<br />

93, 114<br />

Deininger, M., 101<br />

Dekanic, A., 101<br />

Delaunay, J., 93<br />

De Marchi, E., 115<br />

De Pauw, B., 120<br />

de Swart, L., 121<br />

de Witte, T., 17, 26, 76, 121<br />

Dimovski, A., 102<br />

DiPersio, J., 101<br />

Djunic, I., 89, 102, 108, 109<br />

Djurasinovic, V., 89, 108, 109<br />

Dotlic, S., 112<br />

Dragan, C., 117<br />

Drera, M., 106<br />

Drgona, L., 119<br />

Dubravcic, K., 96, 104<br />

Dubruille, V., 93<br />

Dujmovic, D., 99, 111<br />

Dukovski, D., 99, 102<br />

Duletic Nacinovic, A., 91, 101<br />

Durakovic, N., 94, 95, 105,<br />

114<br />

Dusek, D., 96, 103<br />

Elezovic, I., 102, 105, 109<br />

Engert, A., 65<br />

Erickson-Viitanen, S., 101<br />

Faderl, S., 3, 49<br />

Fayad, L., 63<br />

Femenic, R., 96, 103, 108, 110<br />

Fern<strong>and</strong>ez, S., 112<br />

Ferrajoli, A., 53<br />

Fowler, N., 61<br />

Frani} Simic, I., 92<br />

Freireich, E., 17<br />

Fruchtman, S., 35<br />

Fu, K., 97<br />

Fuchs, EJ., 94<br />

Gale, R. P., 47<br />

Galgano, L., 115<br />

Garcia-Manero, G., 28<br />

Gasparovic, V., 112<br />

Gastinne, T., 93<br />

Geisler, C., 62<br />

Giagounidis, A., 121<br />

Giebel, S., 52<br />

Giles, F., 123<br />

Girinsky, T., 64<br />

Girschikofsky, M., 119<br />

Gjadrov, K., 90<br />

Gojceta, K., 114<br />

Golemovic, M., 96, 113, 114<br />

Golubic Cepulic, B., 96, 105, 113,<br />

114<br />

Gotic, M., 105<br />

Gotlib, J., 101<br />

Govedarovic, N., 91, 99<br />

Grahovac, B., 101<br />

Gratwohl, A., 54<br />

Grcevi}, D., 104<br />

Greiner, T., 97<br />

Gress, RE., 94<br />

Grkovic, L., 94<br />

Grohovac, D., 101<br />

Grubic, Z., 93, 95, 115, 116<br />

Guillaume, T., 93<br />

Gupta, V., 101<br />

Gveric-Krecak, V., 91<br />

Gyimesi, E., 88<br />

Hadji-Pecova, L., 111<br />

Hadzisejdic, I., 101<br />

Haferlach, T., 20<br />

Haïat, S., 50<br />

127


Author Index Lije~ Vjesn 2011; godi{te 133; (Supl. 4)<br />

Halkes, S., 17<br />

Haris, V., 88<br />

Harousseau, JL., 93<br />

Harvey, J., 101<br />

Hehlmann, R., 35<br />

Heuck, C. J., 46<br />

Hevessy, Zs., 88<br />

Hexner, E., 101<br />

Hillmen, P., 54, 124<br />

Hoelzer, D., 48<br />

Hoffman, R., 35<br />

Horowitz, M. M., 55<br />

Horvat, I., 92<br />

Host, I., 101<br />

Hotic-Laz<strong>are</strong>vic, S., 112<br />

Hude, I., 111<br />

Humar, I., 113, 114<br />

Hutchings, M., 65<br />

Ilic, I., 99, 111, 112, 114<br />

Indrak, K., 81<br />

Iqbal, J., 97<br />

Ivanovski, M., 102<br />

Ivcevic, S., 104<br />

Jabbour, E., 3, 28, 36, 46<br />

Jaksic, B., 78, 90<br />

Jaksic, O., 88, 90, 98<br />

Jansen, J., 21<br />

Jones, RJ., 94<br />

Jonjic, N., 101<br />

Kantarjian, H., 3, 23, 28, 36,<br />

101<br />

Kappelmayer, J., 88<br />

Kardum, I., 90<br />

Keating, M., 18<br />

Kezic, Lj., 112<br />

Khouri, I., 54<br />

Kis, E., 107<br />

Knopinska-Posluszny, W.,<br />

81<br />

Kolb, H.-J., 19<br />

Konja, J., 96, 103, 108<br />

Konjevoda, P., 103<br />

Koumenis, I., 101<br />

Kovacic, N., 104<br />

Kowalski, J., 94<br />

Kraguljac, N., 105, 109<br />

Kraguljac-Kurtovic, N., 89<br />

Krajcsi, P., 107<br />

Krmek-Zupanic, D., 91<br />

Krnic, N., 96<br />

Kröger, N., 34<br />

Kusec, R., 88, 90, 98, 104<br />

Kutlesa, M., 103<br />

Labar, B., 17, 72, 75, 81, 87, 89, 90,<br />

91, 92, 93, 95, 96, 99, 100, 104,<br />

105, 107, 111, 114, 115, 116, 123<br />

128<br />

Lalic, H., 87<br />

Lapus, F., 112<br />

Lasan-Trcic, R., 90, 91, 92, 110<br />

Laszlo, D., 106, 115<br />

Lazic-Prodan, V., 91<br />

Le Gouill, S., 93<br />

Legr<strong>and</strong>, O., 50<br />

Leitch, H. A., 120<br />

Levine, R. L., 23<br />

Levy, R., 101<br />

Librojo, M., 97, 106, 117<br />

Livun, A., 88<br />

Lozic, D., 91<br />

Lucijanic, M., 98<br />

Lukic, M., 96, 114<br />

Lukinovic-Skudar, V., 87<br />

Lupu, AR., 117<br />

Luznik, L., 58, 94<br />

Lyons, R., 101<br />

MacKenzie, M., 121<br />

Macukanovic-Golubovic, L., 91, 99<br />

Magic, Z., 98<br />

Mahe, B., 93<br />

Malard, F., 93<br />

Malcovati, L., 24<br />

Malinovic, J., 112<br />

M<strong>and</strong>ac-Rogulj, I., 91<br />

M<strong>and</strong>ic, D., 112<br />

Marie, J.-P., 17, 50<br />

Marin, D., 36<br />

Marin, S., 117<br />

Marjanovic, G., 91, 99<br />

Márki-Zay, J., 88, 107<br />

Markovic, D., 91, 99<br />

Markovic-Glamocak, M., 110<br />

Martinelli, G., 106, 115<br />

Marusic Vrsalovic, M., 88<br />

Masarova, K., 106<br />

Matevska, N., 102<br />

Matulic, M., 87<br />

Mazic, S., 96, 105, 113, 114<br />

Meloni, G., 17<br />

Mesa, R., 101<br />

Mihaljevic, B., 105<br />

Mihaylov, G., 81<br />

Mikulic, M., 90, 93, 95, 96, 114<br />

Milanovic, N., 98<br />

Miljkovic, E., 91, 99<br />

Miller, C., 101<br />

Milpied, N., 93<br />

Mistrik, M., 71, 81, 106<br />

Mitchell, A., 48<br />

Mitchell, SA., 94<br />

Mitrovic, M., 102, 109<br />

Mitrovic, Z., 97, 100<br />

Mohty, M., 52, 55, 93<br />

Moicean, A., 81<br />

Monroe, D., 97, 106, 117<br />

Moreau, P., 93<br />

Mrdja, J., 112<br />

Mrsic, M., 93, 95, 114<br />

Mueller, M. M., 123<br />

Musani, V., 88<br />

Muus, P., 17, 76<br />

Nada, S., 117<br />

Nagy, É., 88<br />

Nagy, G., 88<br />

Najfeld, V., 35<br />

Negri, M., 106<br />

Nemet, D., 79, 90, 92, 99, 105,<br />

111<br />

Niederwieser, D., 55<br />

Nikolic, V., 91, 99<br />

Novkovic, A., 89, 108, 109<br />

Novoselac, J., 114<br />

O’Brien, S., 3, 53<br />

Orl<strong>and</strong>o, L., 115<br />

Ottman, O., 51<br />

Ozretic, D., 96<br />

Panovska, I., 111<br />

Panovska-Stavridis, I., 99, 102<br />

Paquette, R., 101<br />

Paradzik, M., 87<br />

Pavletic, SZ., 59, 94<br />

Pavlovic, M., 91<br />

Pavlovic, Maja 96, 110<br />

Pejsa, V., 88, 90, 91, 98<br />

Peric, Z., 91, 93, 104<br />

Perkovic, S., 96, 107, 110<br />

Perunicic Jovanovic, M., 102<br />

Petersen, F. B., 57<br />

Petranovic, D., 101<br />

Petrides, P. E., 32, 72<br />

Petrusevska, G., 99, 111<br />

Pirsic, M., 98<br />

Pivkova Veljanovska, A., 102<br />

Plenkovic, F., 96, 114<br />

Prka, Z., 98<br />

Pulanic, D., 94<br />

Radic Antolic, M., 89, 92, 93, 110<br />

Radman, I., 92, 99, 100, 105, 111<br />

Rajic, Lj., 96, 103, 108, 110, 116<br />

Raos, M., 96, 114<br />

Rav<strong>and</strong>i, F., 3, 22<br />

Raza, A., 101<br />

Ries, S., 89, 90<br />

Romic, I., 91<br />

Roncevic, P., 92<br />

Rosti, G., 46<br />

Saglio, G., 122<br />

Saguna, C., 117<br />

S<strong>and</strong>or, V., 101<br />

Santek, F., 99, 111<br />

Santini, M., 103


Lije~ Vjesn 2011; godi{te 133; (Supl. 4) Author Index<br />

Sanz, M. A., 22<br />

Scalise, A., 35<br />

Schmetzer, H., 19<br />

Sefer, D., 105<br />

Seili, I., 101<br />

Selleslag D, D., 17<br />

Sertic, D., 90, 91, 92, 93, 99,<br />

105, 111<br />

Serventi Seiwerth, R., 90, 93, 94, 95,<br />

96, 104, 114, 115, 116<br />

Shaughnessy, J. D., Jr, 48<br />

Shih, A., 23<br />

Silver, R. T., 33, 35, 101<br />

Silverman, L., 35<br />

Simonovic, O., 91<br />

Sincic-Petricevic, J., 91<br />

Sipka, S., 88<br />

Skific, M., 113, 114<br />

Skunca, Z., 91<br />

Slipac, J., 103<br />

Smith, LM., 97<br />

Sphilberg, O., 61<br />

Sretenovic, A., 109<br />

Steensma, D., 24<br />

Stefanikova, Z., 106<br />

Steinberg, SM., 94<br />

Stemberger, L., 103<br />

Stern-Padovan, R., 99, 111<br />

Stingl, K., 95, 115, 116<br />

Stojanovic, A., 99, 111<br />

Stojanovski, Z., 111<br />

Stojcic, B., 112<br />

Stojsavljevic, S., 103<br />

Stoos Veic, T., 88<br />

Suciu, S., 17, 60<br />

Sun, W., 101<br />

Sureda, A., 66<br />

Suvajdzic, N., 102, 109<br />

Suvajdzic-Vukovic, N., 89, 108<br />

Suvic-Krizanic, V., 91<br />

Swinkels, D., 121<br />

Szabó, P., 88<br />

Sziráki Kiss, V., 88<br />

Talpaz, M., 101<br />

Tarabar, O., 98<br />

Tauber Jakab, K., 88, 107<br />

Taylor, TN., 94<br />

Telek, B., 110<br />

Tesovic, G., 96<br />

Tevet, M., 117<br />

Thomas, D., 52, 76<br />

Tijanic, I., 91, 99<br />

Todorovic, M., 105<br />

Tõkés-Füzesi, M., 88<br />

Tomac, G., 114<br />

Tomin, D., 81, 89, 102, 105, 108,<br />

109<br />

Trajkova, S., 99, 102<br />

Tripodi, J., 35<br />

Trneny, M., 62<br />

Trucza, É., 88<br />

Tukic, Lj., 98<br />

Udvardy, M., 88, 107, 110<br />

Vaddi, K., 101<br />

Valkovic, T., 101<br />

Van Etten, R. A., 31<br />

Vekhoff, A., 50<br />

Verstovsek, S., 31, 79, 101<br />

Vidovic, A., 89, 108, 109<br />

Vidovic, I., 114<br />

Vieira, S., 97, 106, 117<br />

Vignetti, M., 17<br />

Vince, A., 103<br />

Virijevic, M., 89, 108, 109<br />

Visnjic, D., 87<br />

Vodanovic, M., 100<br />

Vose, JM., 97<br />

Vrhovac, R., 103, 116<br />

Vucic, M., 91, 99<br />

Vukicevic, T., 91<br />

Weisenburger, DD., 97<br />

Wierda, W., 53<br />

Wierzbovska, A., 81<br />

Wijermans, P., 25<br />

Willemze, R., 17<br />

Williams, KM., 94<br />

Winton, E., 101<br />

Wroblewski, SG., 94<br />

Younes, A., 62, 69<br />

Zadro, R., 89, 90, 91, 92, 93, 96,<br />

107, 110<br />

Zahurak, M., 94<br />

Zelic, A., 104<br />

Zinzani, P. L., 62<br />

Zlopasa, G., 114<br />

Zunec, R., 115, 116<br />

129

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!