10.08.2013 Views

Report in English with a Dutch summary (KCE reports 75A)

Report in English with a Dutch summary (KCE reports 75A)

Report in English with a Dutch summary (KCE reports 75A)

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.

Wetenschappelijke ondersteun<strong>in</strong>g van het<br />

College voor Oncologie:<br />

een nationale praktijkrichtlijn voor de aanpak<br />

van slokdarm- en maagkanker<br />

<strong>KCE</strong> <strong>reports</strong> <strong>75A</strong><br />

Federaal Kenniscentrum voor de Gezondheidszorg<br />

Centre fédéral d’expertise des so<strong>in</strong>s de santé<br />

2008


Het Federaal Kenniscentrum voor de Gezondheidszorg<br />

Voorstell<strong>in</strong>g : Het Federaal Kenniscentrum voor de Gezondheidszorg is een<br />

parastatale, opgericht door de programma-wet van 24 december 2002<br />

(artikelen 262 tot 266) die onder de bevoegdheid valt van de M<strong>in</strong>ister<br />

van Volksgezondheid en Sociale Zaken. Het Centrum is belast met het<br />

realiseren van beleidsondersteunende studies b<strong>in</strong>nen de sector van de<br />

gezondheidszorg en de ziekteverzeker<strong>in</strong>g.<br />

Raad van Bestuur<br />

Effectieve leden : Gillet Pierre (Voorzitter), Cuypers Dirk (Ondervoorzitter),<br />

Avontroodt Yolande, De Cock Jo (Ondervoorzitter), Demeyere Frank,<br />

De Ridder Henri, Gillet Jean-Bernard, God<strong>in</strong> Jean-Noël, Goyens Floris,<br />

Maes Jef, Mertens Pascal, Mertens Raf, Moens Marc, Perl François, Van<br />

Massenhove Frank, Vandermeeren Philippe, Verertbruggen Patrick,<br />

Vermeyen Karel.<br />

Plaatsvervangers : Annemans Lieven, Coll<strong>in</strong> Benoît, Cuypers Rita, Dercq Jean-Paul, Désir<br />

Daniel, Lemye Roland, Palsterman Paul, Ponce Annick, Pirlot Viviane,<br />

Remacle Anne, Schoonjans Chris, Schrooten Renaat, Vanderstappen<br />

Anne.<br />

Reger<strong>in</strong>gscommissaris : Roger Yves<br />

Directie<br />

Algemeen Directeur : Dirk Ramaekers<br />

Adjunct-Algemeen Directeur : Jean-Pierre Closon<br />

Contact<br />

Federaal Kenniscentrum voor de Gezondheidszorg (<strong>KCE</strong>)<br />

Wetstraat 62<br />

B-1040 Brussel<br />

Belgium<br />

Tel: +32 [0]2 287 33 88<br />

Fax: +32 [0]2 287 33 85<br />

Email : <strong>in</strong>fo@kce.fgov.be<br />

Web : http://www.kce.fgov.be


Wetenschappelijke ondersteun<strong>in</strong>g<br />

van het College voor Oncologie:<br />

een nationale praktijkrichtlijn voor<br />

de aanpak van slokdarm- en<br />

maagkanker<br />

<strong>KCE</strong> <strong>reports</strong> <strong>75A</strong><br />

M. PEETERS, T. LERUT, J. VLAYEN, F. MAMBOURG,<br />

N. ECTORS, P. DEPREZ, T. BOTERBERG, J. DE MEY,<br />

P. FLAMEN, J.-L. VAN LAETHEM, B. NEYNS, P. PATTYN<br />

Federaal Kenniscentrum voor de Gezondheidszorg<br />

Centre fédéral d’expertise des so<strong>in</strong>s de santé<br />

2008


<strong>KCE</strong> REPORTS <strong>75A</strong><br />

Titel : Wetenschappelijke ondersteun<strong>in</strong>g van het College voor Oncologie: een<br />

nationale praktijkrichtlijn voor de aanpak van slokdarm- en maagkanker<br />

Auteurs : M. Peeters (UZ Gent; College Oncologie), T. Lerut (UZ Leuven), J. Vlayen<br />

(<strong>KCE</strong>), F. Mambourg (<strong>KCE</strong>), N. Ectors (UZ Leuven), P. Deprez (UCL), T.<br />

Boterberg (UZ Gent), J. De Mey (UZ Brussel), P. Flamen (Jules Bordet<br />

Instituut), J.-L. Van Laethem (ULB), B. Neyns (UZ Brussel), P. Pattyn (UZ<br />

Gent)<br />

Externe experten : Michel Buset (Belgian Society of Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy), Wim<br />

Ceelen (Belgian Society of Surgical Oncology), Paul Cheyns (Upper GI<br />

sectie van het Kon<strong>in</strong>klijk Belgisch Genootschap Heelkunde), Jean-Marie<br />

Collard (Belgian Society of Surgical Oncology), Jochen Decaestecker<br />

(Vlaamse Verenig<strong>in</strong>g voor Gastro-enterologie), Jacques De Grève<br />

(Voorzitter Work<strong>in</strong>g Party Manual and Guidel<strong>in</strong>es, College Oncologie),<br />

Pierre Demetter (Belgian Digestive Pathology Club), Louis Ferrant<br />

(Domus Medica), Roland Hust<strong>in</strong>x (Belgische Genootschap voor Nucleaire<br />

Geneeskunde), Anne Jouret-Mour<strong>in</strong> (Belgian Digestive Pathology Club),<br />

Cathy Mah<strong>in</strong> (Belgische Verenig<strong>in</strong>g voor Radiotherapie-Oncologie), Max<br />

Mano (Belgische Verenig<strong>in</strong>g voor Medische Oncologie), Hubert Piessevaux<br />

(Société Royale Belge de Gastro-enterologie), Daniel Urba<strong>in</strong> (Belgian<br />

Society of Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy), Eric Van Cutsem (Vlaamse<br />

Verenig<strong>in</strong>g voor Gastro-enterologie), Bart Van den Eynden (Domus<br />

Medica), Didier Verhoeven (Belgische Verenig<strong>in</strong>g voor Medische<br />

Oncologie), Joseph Weerts (Upper GI sectie van het Kon<strong>in</strong>klijk Belgisch<br />

Genootschap Heelkunde)<br />

Acknowledgements Liesbet Van Eycken (Sticht<strong>in</strong>g Kankerregister), Kris Henau (Sticht<strong>in</strong>g<br />

Kankerregister)<br />

Externe validatoren : Harry Bleiberg (Jules Bordet Instituut), Marc De Man (OLV Ziekenhuis<br />

Aalst), Hugo W. Tilanus (Erasmus MC Rotterdam)<br />

Conflict of <strong>in</strong>terest : De meeste auteurs, externe experten en validatoren werken <strong>in</strong> een<br />

centrum gespecialiseerd <strong>in</strong> de behandel<strong>in</strong>g van slokdarm- en maagkanker.<br />

Joseph Weerts ontv<strong>in</strong>g betal<strong>in</strong>g voor deelname aan een postgraduaat<br />

cursus (IRCAD Straatsburg). Andere belangenconflicten werden niet<br />

meegedeeld.<br />

Disclaimer: De externe experten hebben aan het wetenschappelijke rapport<br />

meegewerkt dat daarna aan de validatoren werd voorgelegd. De validatie<br />

van het rapport volgt uit een consensus of een meerderheidsstem tussen<br />

de validatoren. Alleen het <strong>KCE</strong> is verantwoordelijk voor de eventuele<br />

resterende vergiss<strong>in</strong>gen of onvolledigheden alsook voor de aanbevel<strong>in</strong>gen<br />

aan de overheid.<br />

Layout :<br />

Brussel, 21 maart 2008<br />

Studie nr 2007-28-01<br />

Ine Verhulst<br />

Dome<strong>in</strong> : Good Cl<strong>in</strong>ical Practice (GCP)<br />

MeSH : Esophageal Neoplasms ; Stomach Neoplasms ; Gastro<strong>in</strong>test<strong>in</strong>al Stromal Tumors ; Lymphoma,<br />

B-Cell, Marg<strong>in</strong>al Zone<br />

NLM classification : WI 149<br />

Taal : Nederlands, Engels<br />

Format : Adobe® PDF (A4)<br />

Wettelijk depot : D/2008/10.273/16<br />

Elke gedeeltelijke reproductie van dit document is toegestaan mits bronvermeld<strong>in</strong>g.<br />

Dit document is beschikbaar van op de website van het Federaal Kenniscentrum voor de<br />

gezondheidszorg.


Hoe refereren naar dit document?<br />

Peeters M, Lerut T, Vlayen J, Mambourg F, Ectors N, Deprez P, et al. Wetenschappelijke<br />

ondersteun<strong>in</strong>g van het College voor Oncologie: een nationale praktijkrichtlijn voor de aanpak van<br />

slokdarm- en maagkanker. Good Cl<strong>in</strong>ical Practice (GCP). Brussel: Federaal Kenniscentrum voor de<br />

Gezondheidszorg (<strong>KCE</strong>); 2008. <strong>KCE</strong> <strong>Report</strong>s <strong>75A</strong> (D/2008/10.273/16)


<strong>KCE</strong> <strong>Report</strong>s <strong>75A</strong> Slokdarm- en maagkanker i<br />

VOORWOORD<br />

Jaarlijks worden ongeveer 2000 Belgen met de diagnose van slokdarm- of maagkanker<br />

geconfronteerd. Gezien de slechte prognose vormen ze samen de vijfde belangrijkste<br />

oncologische doodsoorzaak <strong>in</strong> België, na long-, dikdarm-, borst- en prostaatkanker. Een<br />

belangrijk deel van de patiënten kan niet genezen worden en valt terug op een<br />

palliatieve behandel<strong>in</strong>g.<br />

In dit rapport wordt een evidence-based praktijkrichtlijn voorgesteld die zorgverleners<br />

en patiënten met slokdarm- of maagkanker moet helpen bij het nemen van kl<strong>in</strong>ische<br />

besliss<strong>in</strong>gen. Voor het opstellen van de aanbevel<strong>in</strong>gen werd opnieuw vertrokken van<br />

bestaande <strong>in</strong>ternationale richtlijnen van goede kwaliteit, aangevuld met de meest<br />

recente wetenschappelijke literatuur. Bij potentieel behandelbare tumoren staat<br />

chirurgie centraal, waarbij een goede opleid<strong>in</strong>g en voldoende ervar<strong>in</strong>g van de chirurg<br />

cruciaal is. In de palliatieve situatie primeert de levenskwaliteit. Eens te meer wordt <strong>in</strong><br />

deze richtlijn het belang van multidiscipl<strong>in</strong>air overleg benadrukt.<br />

Voorliggende richtlijn is de vierde die tot stand kwam dankzij een ondertussen<br />

vertrouwde samenwerk<strong>in</strong>g tussen het College voor Oncologie en het <strong>KCE</strong>. Een grote<br />

groep experten, waaronder ook vertegenwoordigers van de Vlaamse wetenschappelijke<br />

huisartsenverenig<strong>in</strong>g, werkte aan dit rapport mee. Hun enthousiasme en <strong>in</strong>zet waren<br />

wederom groot en bepaalden <strong>in</strong> belangrijke mate de hoge kwaliteit van dit document.<br />

Onze welgemeende dank hiervoor.<br />

De ontwikkel<strong>in</strong>g van deze richtlijn vormt de eerste stap <strong>in</strong> een lang en cont<strong>in</strong>u proces.<br />

Met het onl<strong>in</strong>e ter beschikk<strong>in</strong>g stellen via de website van het College voor Oncologie<br />

wordt de verspreid<strong>in</strong>g en implementatie van deze richtlijn bevorderd. In de richtlijn<br />

wordt bovendien een prille stap gezet naar de ontwikkel<strong>in</strong>g van kwaliteits<strong>in</strong>dicatoren,<br />

die moeten toelaten de kwaliteit van de kankerzorg op te volgen. Enkele lopende<br />

kwaliteits<strong>in</strong>itiatieven, zoals het PROCARE project, zullen ongetwijfeld duidelijkheid<br />

brengen over de te volgen strategie hieromtrent.<br />

De aanpak van kanker krijgt momenteel behoorlijk wat aandacht met de ontwikkel<strong>in</strong>g<br />

van een nationaal kankerplan. We hopen dat voorliggende richtlijn nuttig kan bijdragen<br />

aan de verdere uitwerk<strong>in</strong>g van dit plan.<br />

Closon Jean Pierre Ramaekers Dirk<br />

Adjunct algemeen directeur Algemeen directeur


ii Slokdarm- en maagkanker <strong>KCE</strong> <strong>reports</strong> <strong>75A</strong><br />

INLEIDING<br />

Samenvatt<strong>in</strong>g<br />

B<strong>in</strong>nen de samenwerk<strong>in</strong>g tussen het College voor Oncologie en het <strong>KCE</strong> werden<br />

aanbevel<strong>in</strong>gen ontwikkeld voor de aanpak van slokdarm- en maagkanker (<strong>in</strong>clusief het<br />

primaire maaglymfoom en gastro<strong>in</strong>test<strong>in</strong>ale stromale tumoren [GIST]). Deze<br />

aanbevel<strong>in</strong>gen beslaan het ganse traject van deze patiënten, gaande van diagnose tot<br />

follow-up. Ze zijn bedoeld voor alle zorgverleners die betrokken zijn <strong>in</strong> de zorg voor<br />

deze patiënten.<br />

METHODOLOGIE<br />

DEFINITIES<br />

Voor de ontwikkel<strong>in</strong>g van deze richtlijn werd de ADAPTE methodologie gebruikt. In<br />

eerste <strong>in</strong>stantie werden met behulp van kl<strong>in</strong>ische experten de belangrijkste kl<strong>in</strong>ische<br />

zoekvragen geformuleerd. Bestaande (<strong>in</strong>ter)nationale richtlijnen werden gezocht <strong>in</strong><br />

Medl<strong>in</strong>e, Embase, National Guidel<strong>in</strong>e Clear<strong>in</strong>ghouse en websites van richtlijnorganisaties<br />

en oncologische organisaties. De 17 gevonden richtlijnen werden door middel van het<br />

AGREE <strong>in</strong>strument beoordeeld op hun kwaliteit door twee onafhankelijke reviewers en<br />

al dan niet geselecteerd op basis van een algemeen kwaliteitsoordeel. Vervolgens<br />

werden de 7 geselecteerde richtlijnen geupdated voor elke kl<strong>in</strong>ische vraag, door<br />

bijkomende evidentie te zoeken <strong>in</strong> Medl<strong>in</strong>e en de Cochrane Database of Systematic<br />

Reviews. Een level of evidence werd toegekend aan elke orig<strong>in</strong>ele aanbevel<strong>in</strong>g en<br />

bijkomende studie door gebruik te maken van het GRADE systeem.<br />

Gebaseerd op de gevonden evidentie werden aanbevel<strong>in</strong>gen geformuleerd door een<br />

multidiscipl<strong>in</strong>aire richtlijnontwikkel<strong>in</strong>gsgroep. Deze aanbevel<strong>in</strong>gen werden op een<br />

formele manier beoordeeld door vertegenwoordigers van de Professionele en<br />

Wetenschappelijke Verenig<strong>in</strong>gen. Belangenconflicten werden genoteerd.<br />

TOPOGRAFISCHE DEFINITIES<br />

VROEGE LETSELS<br />

Er bestaat veel discussie over de classificatie van tumoren van de gastro-oesofagale<br />

overgang. Voor dit rapport werden de volgende def<strong>in</strong>ities gehanteerd:<br />

• Indien meer dan 50% van de tumor zich <strong>in</strong> de maagcardia bev<strong>in</strong>dt,<br />

wordt hij geklasseerd als maagtumor.<br />

• Indien de tumormassa zich vooral <strong>in</strong> de slokdarm bev<strong>in</strong>dt, wordt hij als<br />

slokdarmtumor geklasseerd.<br />

• Tumoren van de gastro-oesofagale overgang dienen beschouwd te<br />

worden als slokdarmtumoren, en dienen als dusdanig te worden<br />

behandeld.<br />

Over de def<strong>in</strong>itie van een Barrett letsel bestaat er geen consensus. De meest gangbare<br />

def<strong>in</strong>itie is: een verander<strong>in</strong>g van het slokdarmepitheel die visueel kan herkend worden<br />

tijdens endoscopie en waarbij op biopsie een <strong>in</strong>test<strong>in</strong>ale metaplasie bevestigd wordt.<br />

Meerdere classificaties van dysplasie zijn beschikbaar. Kl<strong>in</strong>ische relevantie is een<br />

belangrijk criterium bij de keuze tussen deze classificaties.


<strong>KCE</strong> <strong>Report</strong>s <strong>75A</strong> Slokdarm- en maagkanker iii<br />

FINALE AANBEVELINGEN<br />

1. SLOKDARMKANKER<br />

MDT: Multidiscipl<strong>in</strong>air Team<br />

PET: Positronen Emissie Tomografie


iv Slokdarm- en maagkanker <strong>KCE</strong> <strong>reports</strong> <strong>75A</strong><br />

Diagnose<br />

De diagnose van slokdarmkanker is, naast een anamnese en kl<strong>in</strong>isch onderzoek,<br />

gebaseerd op een oesofagogastroscopie met biopsies. Patiënten met de volgende<br />

alarmsymptomen zouden vroegtijdig verwezen moeten worden voor endoscopie:<br />

dysfagie, aanhoudend braken, anorexie, gewichtsverlies en gastro<strong>in</strong>test<strong>in</strong>aal bloedverlies.<br />

Hoge-resolutie endoscopie en chromo-endoscopie zijn niet rout<strong>in</strong>ematig aangewezen,<br />

maar kunnen nuttig zijn bij de opvolg<strong>in</strong>g van patiënten met een hoog risico op<br />

slokdarmkanker, zoals patiënten met een Barrett-slokdarm of hooggradige dysplasie.<br />

Aanpak van dysplastische letsels<br />

Stag<strong>in</strong>g<br />

Bij patiënten met een Barrett letsel moet een gestructureerd biopsieprotocol<br />

gehanteerd worden, met biopsies elke 2 centimeter uit 4 kwadranten en biopsies van<br />

elk zichtbaar letsel. Bij een vermoeden van hooggradige dysplasie dient er reken<strong>in</strong>g<br />

gehouden te worden met de kl<strong>in</strong>iek en endoscopische bev<strong>in</strong>d<strong>in</strong>gen. Indien de<br />

hooggradige dysplasie bevestigd wordt en een therapeutische <strong>in</strong>terventie aangewezen is,<br />

is een grondige endoscopische (hoge-resolutie endoscopie +/- chromo-endoscopie,<br />

biopsies elke centimeter uit 4 kwadranten) en pathologische evaluatie (bevestig<strong>in</strong>g van<br />

de diagnose door tweede patholoog, eventueel bijkomende biopsies of diagnostische<br />

endoscopische mucosale resectie [EMR]) aangewezen. Behandel<strong>in</strong>g dient te worden<br />

besproken tijdens een multidiscipl<strong>in</strong>air overleg, en gebeurt best <strong>in</strong> centra met de nodige<br />

endoscopische en chirurgische expertise en voorzien<strong>in</strong>gen.<br />

Bij patiënten met slokdarmkanker is een CT van de thorax (<strong>in</strong>clusief lage halsregio) en<br />

het abdomen aangewezen ter uitsluit<strong>in</strong>g van metastasen. Indien curatieve behandel<strong>in</strong>g<br />

mogelijk geacht wordt, is verdere stag<strong>in</strong>g met echo-endoscopie (EUS) met of zonder<br />

fijne naald aspiratie cytologie (FNAC) aangewezen. Aanvullend kan PET(/CT) scan<br />

overwogen worden voor de evaluatie van lymfeklier- en andere metastasen. Bijkomende<br />

onderzoeken, zoals kernsp<strong>in</strong>tomografie, bronchoscopie (+/- bronchiale echografie +/-<br />

biopsie), thoracoscopie of laparoscopie, kunnen gebeuren op <strong>in</strong>dicatie.<br />

Behandel<strong>in</strong>g van mucosale kanker<br />

Bij patiënten met een T1a letsel dient de diagnose bevestigd te worden door een<br />

tweede patholoog. Bijkomende biopsies of eventuele diagnostische EMR kunnen<br />

aangewezen zijn bij twijfel. Behandel<strong>in</strong>g dient besproken te worden tijdens een<br />

multidiscipl<strong>in</strong>air overleg, waarbij – reken<strong>in</strong>g houdende met het stadium, grootte, lengte<br />

van het Barrett letsel, histologische type, differentiatiegraad en lymfovasculaire <strong>in</strong>vasie –<br />

EMR de voorkeur draagt boven chirurgie.<br />

Behandel<strong>in</strong>g van kanker voorbij de mucosa<br />

Neoadjuvante behandel<strong>in</strong>g<br />

Neoadjuvante behandel<strong>in</strong>g is niet rout<strong>in</strong>ematig aangewezen bij patiënten met<br />

slokdarmkanker, en moet besproken worden tijdens een multidiscipl<strong>in</strong>air overleg.<br />

Neoadjuvante radiotherapie is niet aangewezen.<br />

Kl<strong>in</strong>ische uitkomsten en nevenwerk<strong>in</strong>gen van gecomb<strong>in</strong>eerde behandel<strong>in</strong>g (neoadjuvante<br />

behandel<strong>in</strong>g en chirurgie) dienen prospectief geregistreerd te worden.<br />

Chirurgie<br />

Chirurgie is de standaardbehandel<strong>in</strong>g bij patiënten met resecabele slokdarmkanker, en<br />

gebeurt bij voorkeur via een transthoracale ‘en bloc’ resectie met uitgebreide twee-veld<br />

lymfadenectomie. Het doel van chirurgie is volledige verwijder<strong>in</strong>g van de tumor (R0<br />

resectie). Chirurgie voor slokdarmkanker gebeurt bij voorkeur <strong>in</strong> gespecialiseerde<br />

diensten met een hoog volume door chirurgen met de nodige ervar<strong>in</strong>g en/of opleid<strong>in</strong>g


<strong>KCE</strong> <strong>Report</strong>s <strong>75A</strong> Slokdarm- en maagkanker v<br />

Adjuvante behandel<strong>in</strong>g<br />

Adjuvante behandel<strong>in</strong>g is niet aangewezen bij patiënten met slokdarmkanker.<br />

Niet-chirurgische behandel<strong>in</strong>g met curatief opzet<br />

Uitsluitend radiochemotherapeutische behandel<strong>in</strong>g is te overwegen bij patiënten met<br />

een lokaal uitgebreide slokdarmkanker die niet-resecabel is, of bij patiënten met een<br />

lokaal uitgebreide slokdarmkanker die om medische redenen <strong>in</strong>operabel zijn of chirurgie<br />

weigeren.<br />

Palliatieve behandel<strong>in</strong>g en metastatische ziekte<br />

Follow-up<br />

Bij obstructie door een slokdarmtumor is stent<strong>in</strong>g, laserbehandel<strong>in</strong>g of argon plasma<br />

coagulatie (APC) aangewezen, afhankelijk van de plaatselijke beschikbaarheid en<br />

expertise. De behandel<strong>in</strong>g van dysfagie t.g.v. een slokdarmtumor gebeurt best met zelfontplooibare<br />

metalen stents of plastic ontplooibare stents. Bij tumor <strong>in</strong>- of overgroei <strong>in</strong><br />

patiënten met een stent is laserbehandel<strong>in</strong>g, APC of re-stent<strong>in</strong>g te overwegen.<br />

Radiotherapie (extern of endolum<strong>in</strong>eel) is te overwegen bij patiënten met dysfagie en<br />

een relatief langere overlev<strong>in</strong>gsverwacht<strong>in</strong>g.<br />

Chemotherapie met of zonder radiotherapie is een behandel<strong>in</strong>gsoptie voor patiënten<br />

met lokaal uitgebreide of metastatische slokdarmkanker, die moet besproken worden<br />

tijdens een multidiscipl<strong>in</strong>air overleg. Palliatieve chirurgie, zoals oesofagectomie of<br />

substernale bypass, is niet aangewezen.<br />

Patiënten met slokdarmkanker moeten toegang hebben tot een gespecialiseerd palliatief<br />

team, dat specifieke aandacht dient te geven aan symptoomcontrole, voed<strong>in</strong>g en<br />

levenskwaliteit. De huisarts moet een coörd<strong>in</strong>erende rol hebben <strong>in</strong> de organisatie van<br />

de palliatieve thuiszorg.<br />

Bij patiënten die behandeld werden voor slokdarmkanker is een kl<strong>in</strong>isch onderzoek om<br />

de 3 maanden aangewezen. Een CT scan is aangewezen om de 6 maanden gedurende<br />

het eerste jaar, en nadien jaarlijks. Patiënten die behandeld werden met EMR moeten<br />

een controle endoscopie krijgen na 3 maanden, nadien elke 6 maanden gedurende 2<br />

jaar, en nadien jaarlijks.<br />

Behandel<strong>in</strong>g van recidief<br />

Bij patiënten met een recidief slokdarmkanker dienen de therapeutische opties<br />

besproken te worden tijdens een multidiscipl<strong>in</strong>air overleg. Bij patiënten met een locaal<br />

recidief of nieuwe tumor na EMR behoort nieuwe lokale behandel<strong>in</strong>g tot de<br />

behandelopties.


vi Slokdarm- en maagkanker <strong>KCE</strong> <strong>reports</strong> <strong>75A</strong><br />

2. MAAGKANKER


<strong>KCE</strong> <strong>Report</strong>s <strong>75A</strong> Slokdarm- en maagkanker vii<br />

Diagnose<br />

De diagnose van maagkanker is, naast een anamnese en kl<strong>in</strong>isch onderzoek, gebaseerd<br />

op een oesofagogastroscopie met biopsies. Patiënten met de volgende alarmsymptomen<br />

zouden vroegtijdig verwezen moeten worden voor endoscopie: dysfagie, aanhoudend<br />

braken, anorexie, gewichtsverlies en gastro<strong>in</strong>test<strong>in</strong>aal bloedverlies. Een test voor H.<br />

pylori positiviteit moet systematisch op weefsel gebeuren, en bij voorkeur bevestigd<br />

worden met een tweede test.<br />

Hoge-resolutie endoscopie en chromo-endoscopie zijn niet rout<strong>in</strong>ematig aangewezen,<br />

maar kunnen nuttig zijn bij de opvolg<strong>in</strong>g van patiënten met een hoog risico op<br />

maagkanker, zoals patiënten met een dysplasie.<br />

Aanpak van dysplastische letsels<br />

Stag<strong>in</strong>g<br />

Bij patiënten met een hooggradige dysplasie waarvoor therapeutische <strong>in</strong>terventie<br />

aangewezen is, is een grondige endoscopische en pathologische evaluatie (bevestig<strong>in</strong>g<br />

van de diagnose door tweede patholoog, eventueel bijkomende biopsies) aangewezen.<br />

Behandel<strong>in</strong>g dient te worden besproken tijdens een multidiscipl<strong>in</strong>air overleg, en gebeurt<br />

best <strong>in</strong> centra met de nodige endoscopische en chirurgische expertise en voorzien<strong>in</strong>gen.<br />

Bij patiënten met maagkanker is een CT van de thorax en het abdomen aangewezen ter<br />

uitsluit<strong>in</strong>g van metastasen. Indien curatieve behandel<strong>in</strong>g mogelijk geacht wordt, is<br />

verdere stag<strong>in</strong>g met EUS met of zonder FNAC aangewezen. Bijkomende onderzoeken,<br />

zoals PET scan, kernsp<strong>in</strong>tomografie of laparoscopie, kunnen gebeuren op <strong>in</strong>dicatie.<br />

Behandel<strong>in</strong>g van mucosale kanker<br />

Biopsies uit een mucosale maagkanker dienen beoordeeld te worden door een<br />

patholoog met relevante ervar<strong>in</strong>g. Behandel<strong>in</strong>g dient besproken te worden tijdens een<br />

multidiscipl<strong>in</strong>air overleg, waarbij – reken<strong>in</strong>g houdende met het stadium, grootte,<br />

histologische type, differentiatiegraad – EMR de voorkeur draagt boven chirurgie.<br />

Behandel<strong>in</strong>g van kanker voorbij de mucosa<br />

Neoadjuvante behandel<strong>in</strong>g<br />

Neoadjuvante behandel<strong>in</strong>g is niet rout<strong>in</strong>ematig aangewezen bij patiënten met<br />

maagkanker, maar is een optie die moet besproken worden tijdens een multidiscipl<strong>in</strong>air<br />

overleg.<br />

Kl<strong>in</strong>ische uitkomsten en nevenwerk<strong>in</strong>gen van gecomb<strong>in</strong>eerde behandel<strong>in</strong>g (neoadjuvante<br />

behandel<strong>in</strong>g en chirurgie) dienen prospectief geregistreerd te worden.<br />

Chirurgie<br />

Adjuvante behandel<strong>in</strong>g<br />

Chirurgie is de standaardbehandel<strong>in</strong>g bij patiënten met resecabele maagkanker. Distale<br />

tumoren worden behandeld met een partiële gastrectomie, proximale tumoren met een<br />

totale gastrectomie. D2 lymfadenectomie dient standaard te gebeuren. Het doel van<br />

chirurgie is volledige verwijder<strong>in</strong>g van de tumor (R0 resectie). Chirurgie voor<br />

maagkanker gebeurt bij voorkeur <strong>in</strong> gespecialiseerde diensten met een hoog volume<br />

door chirurgen met de nodige ervar<strong>in</strong>g en/of opleid<strong>in</strong>g<br />

Adjuvante chemo- of radiotherapie is niet aangewezen bij patiënten met maagkanker.<br />

Adjuvante chemoradiotherapie wordt niet rout<strong>in</strong>ematig aanbevolen, maar kan<br />

overwogen worden na besprek<strong>in</strong>g tijdens een multidiscipl<strong>in</strong>air overleg.


viii Slokdarm- en maagkanker <strong>KCE</strong> <strong>reports</strong> <strong>75A</strong><br />

Palliatieve behandel<strong>in</strong>g en metastatische ziekte<br />

Follow-up<br />

Palliatieve chirurgie is beperkt tot patiënten met symptomatische stenosen, bloedende<br />

tumoren en perforatie. Bij patiënten met een maligne maagobstructie bestaat de keuze<br />

tussen endoscopische stent<strong>in</strong>g of chirurgische gastro-enterostomie.<br />

Bij patiënten met lokaal uitgebreide of metastatische maagkanker en een goede<br />

algemene toestand is comb<strong>in</strong>atie chemotherapie te overwegen.<br />

Patiënten met maagkanker moeten toegang hebben tot een gespecialiseerd palliatief<br />

team, dat specifieke aandacht dient te geven aan symptoomcontrole, voed<strong>in</strong>g en<br />

levenskwaliteit. De huisarts moet een coörd<strong>in</strong>erende rol hebben <strong>in</strong> de organisatie van<br />

de palliatieve thuiszorg.<br />

Bij patiënten die behandeld werden voor maagkanker is een kl<strong>in</strong>isch onderzoek en<br />

bloedanalyse om de 3 maanden aangewezen. Een CT scan is aangewezen om de 6<br />

maanden gedurende het eerste jaar, en nadien jaarlijks. Patiënten die behandeld werden<br />

met EMR moeten een controle endoscopie krijgen na 3 maanden, nadien elke 6<br />

maanden gedurende 2 jaar, en nadien jaarlijks.<br />

Behandel<strong>in</strong>g van recidief<br />

Bij patiënten met een recidief maagkanker dienen de therapeutische opties besproken te<br />

worden tijdens een multidiscipl<strong>in</strong>air overleg. Bij patiënten met een locaal recidief of<br />

nieuwe tumor na EMR behoort nieuwe lokale behandel<strong>in</strong>g tot de behandelopties.<br />

3. PRIMAIR MAAGLYMFOOM<br />

Diagnose en stag<strong>in</strong>g<br />

Behandel<strong>in</strong>g<br />

Bij een vermoeden van een primair maaglymfoom zijn m<strong>in</strong>stens 8-12 biopsies<br />

aangewezen. Biopsies dienen op die manier bewaard te worden dat moleculaire<br />

diagnostiek mogelijk is. Bij patiënten waar een primair maaglymfoom histologisch<br />

bevestigd wordt, is een EUS (zonder FNAC) aangewezen. Verdere stag<strong>in</strong>g is niet nodig<br />

bij patiënten met een laaggradig ‘mucosa-associated lymphoid tissue’ (MALT) lymfoom,<br />

tenzij noodzakelijk voor differentieeldiagnostische redenen.<br />

H. pylori eradicatie is de eerste keuze behandel<strong>in</strong>g voor patiënten met een stadium I<br />

laaggradig MALT lymfoom die geïnfecteerd zijn met H. pylori. Nadien is strikte<br />

opvolg<strong>in</strong>g met endoscopie en biopsies noodzakelijk. Evaluatie van de H. pylori eradicatie<br />

is nodig b<strong>in</strong>nen de 3 maanden. Patiënten met een succesvolle H. pylori eradicatie, maar<br />

zonder tumorregressie na 1 jaar of met tumorprogressie, dienen doorverwezen te<br />

worden naar een gespecialiseerd hematologisch centrum.


<strong>KCE</strong> <strong>Report</strong>s <strong>75A</strong> Slokdarm- en maagkanker ix<br />

4. GASTROINTESTINALE STROMALE MAAGTUMOREN<br />

Diagnose en stag<strong>in</strong>g<br />

Behandel<strong>in</strong>g<br />

Bij patiënten met een kl<strong>in</strong>isch vermoeden van een gastro<strong>in</strong>test<strong>in</strong>ale stromale tumor<br />

(GIST) van de maag is een EUS met FNAC en een immunohistochemische test van<br />

CD117 aangewezen. Indien therapie overwogen wordt, wordt een CT van het abdomen<br />

aanbevolen.<br />

Niet-metastatische resecabele GIST<br />

Bij patiënten <strong>in</strong> goede algemene toestand met een histologisch bevestigde nietmetastatische<br />

GIST, is chirurgische resectie aangewezen. Ook bij patiënten <strong>in</strong> goede<br />

algemene toestand met een maagtumor van > 5 cm die suggestief is voor een GIST en<br />

zonder aanwijz<strong>in</strong>gen voor metastasen, is chirurgische resectie aangewezen.<br />

Bij patiënten <strong>in</strong> goede algemene toestand met een maagtumor van 2-5 cm die suggestief<br />

is voor een GIST en zonder aanwijz<strong>in</strong>gen voor metastasen, dient de keuze tussen<br />

chirurgische resectie en een afwachtende houd<strong>in</strong>g besproken te worden tijdens een<br />

multidiscipl<strong>in</strong>air overleg. Bij patiënten met een maagtumor van < 2 cm die suggestief is<br />

voor een GIST en zonder aanwijz<strong>in</strong>gen voor metastasen, is een afwachtende houd<strong>in</strong>g<br />

aangewezen.<br />

Metastatische en/of niet-resecabele GIST<br />

Bij patiënten met een metastatische en/of niet-resecabele GIST wordt behandel<strong>in</strong>g met<br />

imat<strong>in</strong>ib aanbevolen. PET/CT wordt aanbevolen om de respons op imat<strong>in</strong>ib te<br />

beoordelen. Bij patiënten met imat<strong>in</strong>ib resistentie of <strong>in</strong>tolerantie kan sunit<strong>in</strong>ib<br />

overwogen worden als tweedelijns behandel<strong>in</strong>g.<br />

IMPLEMENTATIE EN HERZIENING VAN DE<br />

RICHTLIJN<br />

De implementatie van deze richtlijn dient bevorderd te worden door middel van een<br />

onl<strong>in</strong>e implementatie <strong>in</strong>strument dat gebaseerd is op de algemene algoritmes van de<br />

richtlijn. Dit <strong>in</strong>strument dient beschikbaar gesteld te worden op de website van het<br />

College voor Oncologie.<br />

Er dienen geschikte kwaliteits<strong>in</strong>dicatoren ontwikkeld te worden op basis van de<br />

belangrijkste aanbevel<strong>in</strong>gen van deze richtlijn.<br />

Gezien de evidence evolueert, zal een update van deze richtlijn – na een pre-assessment<br />

van de literatuur – vermoedelijk noodzakelijk zijn over 5 jaar.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 1<br />

Table of contents<br />

Scientific <strong>summary</strong><br />

1 INTRODUCTION ................................................................................................... 5<br />

1.1 SCOPE.................................................................................................................................................. 5<br />

1.2 EPIDEMIOLOGY .................................................................................................................................. 5<br />

1.2.1 Oesophageal cancer............................................................................................................ 5<br />

1.2.2 Gastric cancer ...................................................................................................................... 6<br />

2 METHODOLOGY................................................................................................... 8<br />

2.1 GENERAL APPROACH........................................................................................................................ 8<br />

2.2 CLINICAL QUESTIONS....................................................................................................................... 8<br />

2.3 SEARCH FOR EVIDENCE..................................................................................................................... 9<br />

2.3.1 Cl<strong>in</strong>ical practice guidel<strong>in</strong>es................................................................................................. 9<br />

2.3.2 Additional evidence...........................................................................................................10<br />

2.4 QUALITY APPRAISAL........................................................................................................................10<br />

2.4.1 Cl<strong>in</strong>ical practice guidel<strong>in</strong>es...............................................................................................10<br />

2.4.2 Additional evidence...........................................................................................................10<br />

2.5 DATA EXTRACTION AND SUMMARY ............................................................................................10<br />

2.6 FORMULATION OF RECOMMENDATIONS.....................................................................................10<br />

2.7 EXTERNAL REVIEW ..........................................................................................................................10<br />

3 DEFINITIONS...................................................................................................... 12<br />

3.1 TOPOGRAPHIC DEFINITIONS .........................................................................................................12<br />

3.2 EARLY LESIONS.................................................................................................................................13<br />

3.2.1 Histology of the normal oesophagus ............................................................................13<br />

3.2.2 Barrett’s oesophagus ........................................................................................................13<br />

3.2.3 Dysplasia <strong>in</strong> squamous epithelium .................................................................................15<br />

3.2.4 Dysplasia <strong>in</strong> columnar epithelium ..................................................................................15<br />

4 FINAL RECOMMENDATIONS ON OESOPHAGEAL CANCER ................................. 18<br />

4.1 FLOWCHART....................................................................................................................................18<br />

4.2 DIAGNOSIS .......................................................................................................................................19<br />

4.3 WORK-UP OF DYSPLASTIC LESIONS..............................................................................................20<br />

4.4 STAGING...........................................................................................................................................22<br />

4.5 TREATMENT OF MUCOSAL CANCER .............................................................................................23<br />

4.6 TREATMENT OF CANCER BEYOND THE MUCOSA .......................................................................24<br />

4.6.1 Neoadjuvant treatment....................................................................................................24<br />

4.6.2 Surgical treatment .............................................................................................................25<br />

4.6.3 Adjuvant treatment ...........................................................................................................27


2 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

4.6.4 Non-surgical treatment <strong>with</strong> curative <strong>in</strong>tent ..............................................................27<br />

4.7 PALLIATIVE TREATMENT & METASTATIC DISEASE ........................................................................28<br />

4.8 FOLLOW-UP .....................................................................................................................................30<br />

4.9 RECURRENT DISEASE .......................................................................................................................30<br />

5 FINAL RECOMMENDATIONS ON GASTRIC CANCER............................................ 31<br />

5.1 FLOWCHART....................................................................................................................................31<br />

5.2 DIAGNOSIS .......................................................................................................................................32<br />

5.3 WORK-UP OF DYSPLASTIC LESIONS..............................................................................................33<br />

5.4 STAGING...........................................................................................................................................34<br />

5.5 TREATMENT OF MUCOSAL CANCER .............................................................................................35<br />

5.6 TREATMENT OF CANCER BEYOND THE MUCOSA .......................................................................36<br />

5.6.1 Neoadjuvant treatment....................................................................................................36<br />

5.6.2 Surgical treatment .............................................................................................................37<br />

5.6.3 Adjuvant treatment ...........................................................................................................38<br />

5.7 PALLIATIVE TREATMENT & METASTATIC DISEASE ........................................................................39<br />

5.8 FOLLOW-UP .....................................................................................................................................40<br />

5.9 RECURRENT DISEASE .......................................................................................................................41<br />

5.10 TREATMENT OF GASTRIC LYMPHOMA ..........................................................................................41<br />

5.10.1 Introduction ........................................................................................................................41<br />

5.10.2 Diagnosis and stag<strong>in</strong>g ........................................................................................................41<br />

5.10.3 Treatment............................................................................................................................42<br />

5.11 TREATMENT OF GASTROINTESTINAL STROMAL TUMOURS........................................................43<br />

5.11.1 Introduction ........................................................................................................................43<br />

5.11.2 Diagnosis and stag<strong>in</strong>g ........................................................................................................43<br />

5.11.3 Treatment............................................................................................................................44<br />

6 IMPLEMENTATION AND UPDATING OF THE UPPER GI CANCER GUIDELINE...... 46<br />

6.1 IMPLEMENTATION............................................................................................................................46<br />

6.2 QUALITY CONTROL........................................................................................................................46<br />

6.3 GUIDELINE UPDATE.........................................................................................................................46<br />

7 REFERENCES ...................................................................................................... 47<br />

8 APPENDICES ...................................................................................................... 65


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 3<br />

ABBREVIATIONS<br />

95% CI 95 percent confidence <strong>in</strong>terval<br />

5-FU 5-fluorouracil<br />

AFE Autofluorescence endoscopy<br />

AGA American Gastroenterological Association<br />

APC Argon plasma coagulation<br />

ASCO American Society of Cl<strong>in</strong>ical Oncology<br />

BUS Bronchoscopic ultrasound<br />

CBO <strong>Dutch</strong> Institute for Healthcare Improvement<br />

CCO Cancer Care Ontario<br />

CODG Conventional open distal gastrectomy<br />

CPG Cl<strong>in</strong>ical Practice Guidel<strong>in</strong>e<br />

CRT Chemoradiotherapy<br />

CT Computed tomography<br />

EMR Endoscopic mucosal resection<br />

ESD Endoscopic submucosal dissection<br />

EUS Endoscopic ultrasound<br />

FNA(C F<strong>in</strong>e needle aspiration (cytology)<br />

FNCLCC Fédération Nationale des Centres de Lutte Contre le Cancer<br />

GI Gastro<strong>in</strong>test<strong>in</strong>al<br />

GIST Gastro<strong>in</strong>test<strong>in</strong>al stromal tumours<br />

GOJ Gastro-oesophageal junction<br />

GRADE Grad<strong>in</strong>g of Recommendations Assessment, Development and<br />

HR<br />

Evaluation<br />

Hazard ratio<br />

HRE High-resolution endoscopy<br />

IARC International Agency for Research on Cancer<br />

IBD Inflammatory bowel disease<br />

IUAC International Union Aga<strong>in</strong>st Cancer<br />

LN Lymph node<br />

MALT Mucosa-associated lymphoid tissue<br />

MDCT Multidetector row CT<br />

MDT Multidiscipl<strong>in</strong>ary team<br />

MeSH Medical Subject Head<strong>in</strong>gs<br />

MRI Magnetic resonance imag<strong>in</strong>g<br />

NBI Narrow band imag<strong>in</strong>g<br />

NICE National Institute for Health and Cl<strong>in</strong>ical Excellence<br />

OR Odds ratio<br />

PDT Photodynamic therapy<br />

PET Positron-emission tomography


4 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

RCT Randomised Controlled Trial<br />

RR Risk ratio<br />

SCJ Squamo-columnar junction<br />

SEMS Self-expand<strong>in</strong>g metal stents<br />

SIGN Scottish Intercollegiate Guidel<strong>in</strong>es Network<br />

SSBE Short-segment Barrett’s oesophagus<br />

UK United K<strong>in</strong>gdom<br />

US United States of America<br />

WHO World Health Organisation


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 5<br />

1 INTRODUCTION<br />

1.1 SCOPE<br />

In the present report, a cl<strong>in</strong>ical practice guidel<strong>in</strong>e (CPG) on upper gastro<strong>in</strong>test<strong>in</strong>al<br />

cancer is presented, which is the result of a collaboration of the College of Physicians<br />

for Oncology and the <strong>KCE</strong>. This cl<strong>in</strong>ical practice guidel<strong>in</strong>e will cover a broad range of<br />

topics: diagnosis, stag<strong>in</strong>g, treatment, supportive therapy, and follow-up. The guidel<strong>in</strong>e<br />

primarily concerns <strong>in</strong>dividuals <strong>with</strong> oesophageal or gastric cancer, and is <strong>in</strong>tended to be<br />

used by all care providers <strong>in</strong>volved <strong>in</strong> the care for these patients.<br />

1.2 EPIDEMIOLOGY<br />

1.2.1 Oesophageal cancer<br />

Oesophageal cancer is the eighth most common cancer <strong>in</strong> the world and one of the<br />

most lethal [1]. Incidence rates of oesophageal cancer show well-known regional<br />

disparities. Overall, <strong>in</strong>cidence rates for all types of oesophageal cancer range from four<br />

to n<strong>in</strong>e cases per 100.000 males per year (1975 – 1997) <strong>in</strong> Western countries [2].<br />

Lower <strong>in</strong>cidence rates are found <strong>in</strong> Northern Europe (F<strong>in</strong>land, Norway, and Sweden),<br />

whereas the French regions of Burgundy and Calvados have <strong>in</strong>cidence rates of > 14 per<br />

100.000 males per year.<br />

In the Netherlands, Crane et al. found an <strong>in</strong>crease <strong>in</strong> age standardised <strong>in</strong>cidence by 3.4%<br />

and 1.9% per year (1989 – 2003) for males and females respectively [3]. This <strong>in</strong>crease<br />

was almost exclusively caused by oesophageal adenocarc<strong>in</strong>omas. In 14 years, age<br />

standardised mortality <strong>in</strong>creased 2.5% per year among males and 1.7% per year among<br />

females. Similar trends were found <strong>in</strong> the UK and the US, but not <strong>in</strong> France [4]. Relative<br />

survival <strong>in</strong> the Netherlands improved significantly from 8.1% <strong>in</strong> 1989-1993 to 12.6% <strong>in</strong><br />

1999-2003 (p


6 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

The differences <strong>in</strong> <strong>in</strong>cidence trends of oesophageal squamous cell carc<strong>in</strong>oma and<br />

adenocarc<strong>in</strong>oma are less pronounced <strong>in</strong> Belgium (Table 2 and Figure 1). This is probably<br />

due to the fact that the Belgian cancer registration is of recent years (only go<strong>in</strong>g back<br />

until 1997). Dur<strong>in</strong>g the first registration years a high percentage of tumours <strong>with</strong><br />

unspecified histology were registered. However, recently the quality of the cancer<br />

registration markedly improved. This lead to an <strong>in</strong>crease <strong>in</strong> <strong>in</strong>cidence of both squamous<br />

cell carc<strong>in</strong>oma and adenocarc<strong>in</strong>oma and a decrease <strong>in</strong> the <strong>in</strong>cidence of tumours <strong>with</strong><br />

unspecified histology (Table 2 and Figure 1) (Belgian Cancer Registry, personal<br />

communication).<br />

Table 2. Age standardised <strong>in</strong>cidence $ of oesophageal cancer <strong>in</strong> Belgium<br />

accord<strong>in</strong>g to histological type, 1997 – 2003 (n/100.000 person years) (Belgian<br />

Cancer Registry, personal communication).<br />

1997 1998 1999 2000 2001 2002 2003<br />

Males<br />

• SCC # 4.0 3.9 4.2 4.4 4.9 5.2 4.7<br />

• Adenocarc<strong>in</strong>oma 2.7 2.8 2.9 3.6 4.5 4.0 4.3<br />

• Unspecified tumour<br />

Females<br />

1.0 0.6 0.5 0.4 0.2 0.1 0.2<br />

• SCC # 0.9 1.0 1.3 1.4 1.6 1.5 1.5<br />

• Adenocarc<strong>in</strong>oma 0.4 0.4 0.4 0.7 0.6 0.5 0.8<br />

• Unspecified tumour 0.1 0.1 0.2 0.0 0.1 0.1 0.1<br />

$us<strong>in</strong>g a European standard population. #SCC: squamous cell carc<strong>in</strong>oma.<br />

Figure 1. Age standardised <strong>in</strong>cidence $ of oesophageal cancer <strong>in</strong> Belgium for<br />

males accord<strong>in</strong>g to histological type, 1997 – 2003 (n/100.000 person years)<br />

(Belgian Cancer Registry, personal communication).<br />

$us<strong>in</strong>g a European standard population.<br />

1.2.2 Gastric cancer<br />

With an estimated 934.000 new cases per year <strong>in</strong> 2002 worldwide (8.6% of all new<br />

cancer cases), gastric cancer is <strong>in</strong> fourth place beh<strong>in</strong>d cancers of the lung, breast, and<br />

colon and rectum, <strong>with</strong> almost two-third of the cases occurr<strong>in</strong>g <strong>in</strong> develop<strong>in</strong>g countries<br />

[1]. It is the second most common cause of death from cancer.<br />

Gastric cancer <strong>in</strong>cidence rates vary by up to ten-fold throughout the world. Japan and<br />

Korea have the highest gastric cancer <strong>in</strong>cidence rates <strong>in</strong> the world.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 7<br />

High-<strong>in</strong>cidence areas for non-cardia gastric adenocarc<strong>in</strong>oma <strong>in</strong>clude East Asia, Eastern<br />

Europe, and Central and South America. Low <strong>in</strong>cidence rates are found <strong>in</strong> South Asia,<br />

North and East Africa, North America, Australia, and New Zealand [6]. Survival is<br />

moderately good only <strong>in</strong> Japan (52%), where mass screen<strong>in</strong>g by photofluoroscopy has<br />

been practiced s<strong>in</strong>ce the 1960s. Survival is also relatively high <strong>in</strong> North America<br />

(approximatively 21%), possibly due to early diagnosis follow<strong>in</strong>g a higher number of<br />

endoscopic exam<strong>in</strong>ations performed for gastric disorders. Estimated survival is 27% <strong>in</strong><br />

Western Europe [1].<br />

In the Netherlands, age standardised <strong>in</strong>cidence of gastric cancer decl<strong>in</strong>ed from 21.6 to<br />

15.9 per 100.000 person years <strong>in</strong> males and from 10.4 to 6.2 per 100.000 person years<br />

<strong>in</strong> females s<strong>in</strong>ce 1978 [7]. The age standardised mortality rates decreased from 20.7 to<br />

12.8 per 100.000 person years <strong>in</strong> males and from 8.2 to 4.2 per 100.000 person years <strong>in</strong><br />

females.<br />

In Belgium, the crude <strong>in</strong>cidence rate of gastric cancer rose from 12.9 per 100.000 males<br />

<strong>in</strong> 1997 to 14.9 per 100.000 males <strong>in</strong> 2003, and from 8.0 per 100.000 females <strong>in</strong> 1997 to<br />

8.4 per 100.000 females <strong>in</strong> 2003 (Belgian Cancer Registry, personal communication).<br />

Age standardised <strong>in</strong>cidence <strong>in</strong>creased by 2.6% and 0.8% per year (1997 – 2003) for<br />

males and females respectively (Table 3). However, <strong>in</strong> these rates tumours of the<br />

gastro-oesophageal junction (GOJ) are also <strong>in</strong>cluded.<br />

While the <strong>in</strong>cidence rates of these GOJ tumours recently <strong>in</strong>creased, the <strong>in</strong>cidence rates<br />

of ‘real’ gastric tumours (see chapter 3.1) decl<strong>in</strong>ed [8].<br />

Table 3. Age standardised <strong>in</strong>cidence $ of gastric cancer <strong>in</strong> Belgium, 1997 –<br />

2003 (n/100.000 person years) (Belgian Cancer Registry, personal<br />

communication).<br />

Year<br />

Males Females<br />

Belgium Flanders Belgium Flanders<br />

1997 7.4 9.2 3.1 4.3<br />

1998 7.3 8.3 3.4 4.1<br />

1999 7.9 9.1 3.5 4.4<br />

2000 7.5 9.0 3.9 4.5<br />

2001 8.2 9.8 3.5 4.2<br />

2002 8.5 9.8 3.9 4.5<br />

2003 8.2<br />

$us<strong>in</strong>g a world standard population.<br />

9.1 3.2 3.6


8 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

2 METHODOLOGY<br />

2.1 GENERAL APPROACH<br />

As for the previous CPGs developed <strong>with</strong><strong>in</strong> the collaboration between the College and<br />

the <strong>KCE</strong>, the present CPG was developed by adapt<strong>in</strong>g (<strong>in</strong>ter)national CPGs to the<br />

Belgian context [9]. This approach is currently be<strong>in</strong>g structured <strong>in</strong> a formal<br />

methodology by the ADAPTE group, an <strong>in</strong>ternational group of guidel<strong>in</strong>e developers and<br />

researchers [10]. The ADAPTE methodology generally consists of three major phases:<br />

1. Set-up Phase: Outl<strong>in</strong>es the necessary tasks to be completed prior to<br />

beg<strong>in</strong>n<strong>in</strong>g the adaptation process (e.g., identify<strong>in</strong>g necessary skills and<br />

resources).<br />

2. Adaptation Phase: Assists guidel<strong>in</strong>e developers <strong>in</strong> mov<strong>in</strong>g from selection of<br />

a topic to identification of specific cl<strong>in</strong>ical questions; search<strong>in</strong>g for and<br />

retriev<strong>in</strong>g guidel<strong>in</strong>es; assess<strong>in</strong>g the consistency of the evidence there<strong>in</strong>, their<br />

quality, currency, content and applicability; decision mak<strong>in</strong>g around<br />

adaptation; and prepar<strong>in</strong>g the draft adapted guidel<strong>in</strong>e.<br />

3. F<strong>in</strong>alization Phase: Guides guidel<strong>in</strong>e developers through gett<strong>in</strong>g feedback<br />

on the document from stakeholders who will be impacted by the guidel<strong>in</strong>e,<br />

consult<strong>in</strong>g <strong>with</strong> the source developers of guidel<strong>in</strong>es used <strong>in</strong> the adaptation<br />

process, establish<strong>in</strong>g a process for review and updat<strong>in</strong>g of the adapted<br />

guidel<strong>in</strong>e and the process of creat<strong>in</strong>g a f<strong>in</strong>al document.<br />

2.2 CLINICAL QUESTIONS<br />

The cl<strong>in</strong>ical practice guidel<strong>in</strong>e addresses the follow<strong>in</strong>g cl<strong>in</strong>ical questions:<br />

1. Which diagnostic techniques should be used for the diagnosis of oesophageal<br />

and gastric cancer?<br />

2. What are the best treatment options for dysplastic lesions of the oesophagus<br />

and stomach?<br />

3. What stag<strong>in</strong>g techniques should be used for oesophageal and gastric cancer?<br />

4. What are the best treatment options for mucosal oesophageal and gastric<br />

cancer?<br />

5. What are the best treatment options for oesophageal and gastric cancer<br />

beyond the mucosa?<br />

a. neoadjuvant treatment<br />

b. surgical treatment<br />

c. adjuvant treatment<br />

d. non-surgical treatment <strong>with</strong> curative <strong>in</strong>tent<br />

6. What are the best palliative treatment options for extensive disease?<br />

7. What are the best follow-up strategies for oesophageal and gastric cancer?<br />

8. What are the best treatment options for gastric lymphoma?<br />

9. What are the best treatment options for gastro<strong>in</strong>test<strong>in</strong>al stromal tumours?


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 9<br />

2.3 SEARCH FOR EVIDENCE<br />

2.3.1 Cl<strong>in</strong>ical practice guidel<strong>in</strong>es<br />

2.3.1.1 Sources<br />

A broad search of electronic databases (Medl<strong>in</strong>e, EMBASE), specific guidel<strong>in</strong>e websites<br />

and websites of oncologic organisations (Table 4) was conducted <strong>in</strong> July 2007.<br />

Table 4: Searched guidel<strong>in</strong>e websites and websites of oncologic<br />

organisations.<br />

Alberta Heritage Foundation For Medical http://www.ahfmr.ab.ca/<br />

Research (AHFMR)<br />

American Society of Cl<strong>in</strong>ical Oncology (ASCO) http://www.asco.org/<br />

American College of Surgeons (ACS) http://www.facs.org/cancer/coc/<br />

CMA Infobase http://mdm.ca/cpgsnew/cpgs/<strong>in</strong>dex.asp<br />

Guidel<strong>in</strong>es International Network (GIN) http://www.g-i-n.net/<br />

National Comprehensive Cancer Network http://www.nccn.org/<br />

(NCCN)<br />

National Guidel<strong>in</strong>e Clear<strong>in</strong>ghouse http://www.guidel<strong>in</strong>e.gov/<br />

National Cancer Institute http://www.cancer.gov/<br />

Haute Autorité de Santé (HAS) http://bfes.hassante.fr/HTML/<strong>in</strong>dexBFES_HAS.html<br />

BC Cancer Agency http://www.bccancer.bc.ca/default.htm<br />

Institute for Cl<strong>in</strong>ical Systems Improvement (ICSI) http://www.icsi.org/<strong>in</strong>dex.asp<br />

National Health and Medical Research Council<br />

(NHMRC)<br />

http://www.nhmrc.gov.au/<br />

Scottish Intercollegiate Guidel<strong>in</strong>es Network<br />

(SIGN)<br />

http://www.sign.ac.uk/<br />

New Zealand Guidel<strong>in</strong>es Group (NZGG) http://www.nzgg.org.nz/<br />

Fédération Nationale des Centres de Lutte http://www.fnclcc.fr/sor/structure/<strong>in</strong>dex-<br />

Contre le Cancer (FNCLCC)<br />

National Institute for Health and Cl<strong>in</strong>ical<br />

Excellence (NICE)<br />

2.3.1.2 Search terms<br />

sorspecialistes.html<br />

http://www.nice.org.uk/<br />

For Medl<strong>in</strong>e the follow<strong>in</strong>g MeSH terms were used <strong>in</strong> comb<strong>in</strong>ation: stomach neoplasms,<br />

esophageal neoplasms. For EMBASE the follow<strong>in</strong>g Emtree terms were used <strong>in</strong><br />

comb<strong>in</strong>ation: stomach tumor, esophagus tumor. These MeSH and Emtree terms were<br />

comb<strong>in</strong>ed <strong>with</strong> a standardised search strategy to identify CPGs (Table 5).<br />

Table 5: Standardised search strategy for CPGs.<br />

Database Search strategy<br />

Medl<strong>in</strong>e guidel<strong>in</strong>e [pt] OR practice guidel<strong>in</strong>e [pt] OR<br />

recommendation* [ti] OR standard* [ti] OR<br />

guidel<strong>in</strong>e* [ti]<br />

EMBASE 'practice guidel<strong>in</strong>e'/exp<br />

2.3.1.3 In- and exclusion criteria<br />

Both national and <strong>in</strong>ternational CPGs on oesophageal and gastric cancer were searched.<br />

A language (<strong>English</strong>, <strong>Dutch</strong>, French) and date restriction (2001 – 2007) were used.<br />

CPGs <strong>with</strong>out references were excluded, as were CPGs <strong>with</strong>out clear<br />

recommendations.


10 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

2.3.2 Additional evidence<br />

For each cl<strong>in</strong>ical question, the evidence – identified through the <strong>in</strong>cluded CPGs – was<br />

updated by search<strong>in</strong>g Medl<strong>in</strong>e and the Cochrane Database of Systematic Reviews from<br />

the search date of the CPG on (search date August – September 2007). A comb<strong>in</strong>ation<br />

of appropriate MeSH terms and free text words was used (see appendix).<br />

For therapeutic <strong>in</strong>terventions, only systematic reviews and randomized controlled trials<br />

(RCT) were <strong>in</strong>cluded. However, for diagnostic <strong>in</strong>terventions we also searched for<br />

observational studies <strong>in</strong> case no systematic review or RCT was found. The identified<br />

studies were selected based on title and abstract. For all eligible studies, the full-text<br />

was retrieved. In case no full-text was available, the study was not taken <strong>in</strong>to account<br />

for the f<strong>in</strong>al recommendations.<br />

2.4 QUALITY APPRAISAL<br />

2.4.1 Cl<strong>in</strong>ical practice guidel<strong>in</strong>es<br />

In total, 17 CPGs were identified. All were quality appraised by two <strong>in</strong>dependent<br />

reviewers (JV, FM) us<strong>in</strong>g the AGREE <strong>in</strong>strument. Disagreement was discussed face-toface.<br />

At the end, agreement was reached for all CPGs, and 7 CPGs were <strong>in</strong>cluded (see<br />

appendix).<br />

2.4.2 Additional evidence<br />

The quality of the retrieved systematic reviews and RCTs was assessed us<strong>in</strong>g the<br />

checklists of the <strong>Dutch</strong> Cochrane Centre (www.cochrane.nl).<br />

2.5 DATA EXTRACTION AND SUMMARY<br />

For each <strong>in</strong>cluded CPG the follow<strong>in</strong>g data were extracted: search date & publication<br />

year, searched databases, availability of evidence tables, recommendations and<br />

referenced evidence.<br />

For each systematic review, the search date, publication year, <strong>in</strong>cluded studies and ma<strong>in</strong><br />

results were extracted. For RCTs, the follow<strong>in</strong>g data were extracted: publication year,<br />

study population, study <strong>in</strong>tervention, and outcomes.<br />

For each cl<strong>in</strong>ical question, the recommendations from the identified CPGs and the<br />

additional evidence were summarized <strong>in</strong> evidence tables. A level of evidence was<br />

assigned to each recommendation and additional study us<strong>in</strong>g the GRADE system (see<br />

appendix).<br />

2.6 FORMULATION OF RECOMMENDATIONS<br />

Based on the retrieved evidence, a first draft of recommendations was prepared by a<br />

small work<strong>in</strong>g group (JV, FM). This first draft together <strong>with</strong> the evidence tables was<br />

circulated to the guidel<strong>in</strong>e development group 2 weeks prior to the first face-to-face<br />

meet<strong>in</strong>g. The guidel<strong>in</strong>e development group met on several occasions (October 1 st 2007,<br />

November 5 th 2007, December 10 th 2007 and January 9 th 2008) to discuss the first draft.<br />

Recommendations were changed if important evidence supported this change. Based on<br />

the discussion meet<strong>in</strong>gs a second draft of recommendations was prepared. A grade of<br />

recommendation was assigned to each recommendation us<strong>in</strong>g the GRADE system (see<br />

appendix). The second draft was once more circulated to the guidel<strong>in</strong>e development<br />

group for f<strong>in</strong>al approval.<br />

2.7 EXTERNAL REVIEW<br />

The recommendations prepared by the guidel<strong>in</strong>e development group were circulated to<br />

the Professional Associations (Table 6). Each association was asked to assign 2 key<br />

persons to discuss the recommendations dur<strong>in</strong>g an open meet<strong>in</strong>g.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 11<br />

These panellists received the recommendations one week prior to this open meet<strong>in</strong>g.<br />

As a preparation of the meet<strong>in</strong>g all <strong>in</strong>vited panellists were asked to score each<br />

recommendation on a 5-po<strong>in</strong>t Likert-scale to <strong>in</strong>dicate their agreement <strong>with</strong> the<br />

recommendation, <strong>with</strong> a score of ‘1’ <strong>in</strong>dicat<strong>in</strong>g ‘completely disagree’, ‘2’ <strong>in</strong>dicat<strong>in</strong>g<br />

‘somewhat disagree’, ‘3’ <strong>in</strong>dicat<strong>in</strong>g ‘unsure’, ‘4’ <strong>in</strong>dicat<strong>in</strong>g ‘somewhat agree’, and ‘5’<br />

<strong>in</strong>dicat<strong>in</strong>g ‘completely agree’ (the panellists were also able to answer ‘not applicable’ <strong>in</strong><br />

case they were not familiar <strong>with</strong> the underly<strong>in</strong>g evidence). In case a panellist disagreed<br />

<strong>with</strong> the recommendation (score ‘1’ or ‘2’), (s)he was asked to provide appropriate<br />

evidence. All scores (n = 15) were than anonymized and summarized <strong>in</strong>to a mean score,<br />

standard deviation and % of ‘agree’-scores (score ‘4’ and ‘5’) to allow a targeted<br />

discussion (see appendix). The recommendations were then discussed dur<strong>in</strong>g a face-toface<br />

meet<strong>in</strong>g on January 23 rd 2008. Based on this discussion a f<strong>in</strong>al draft of the<br />

recommendations was prepared, and discussed by the guidel<strong>in</strong>e development group by<br />

email. In appendix, an overview is provided of how the comments of the experts were<br />

taken <strong>in</strong>to account.<br />

Table 6: List of Professional Associations to which the recommendations<br />

were communicated.<br />

Belgian Society of Medical Oncology (BSMO)<br />

Belgian Society of Radiotherapy (BVRO – ABRO)<br />

Belgian Society of Nuclear Medic<strong>in</strong>e<br />

Belgian Society of Surgical Oncology (BSSO)<br />

Upper GI section of the Royal Belgian Society of Surgery<br />

Flemish Society of Gastroenterology (VVGE)<br />

Belgian Group of Digestive Oncology (BGDO)<br />

Royal Belgian Society of Gastroenterology (SRBGE)<br />

Domus Medica (Scientific association of Flemish general practitioners)<br />

Belgian Society of Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy (BSGIE)<br />

Belgian Digestive Pathology Club<br />

Belgian Society of Pathology<br />

Royal Belgian Radiological Society<br />

Scientific Society of General Medic<strong>in</strong>e (SSMG)<br />

Belgian Group for Endoscopic Surgery (BGES)


12 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

3 DEFINITIONS<br />

3.1 TOPOGRAPHIC DEFINITIONS<br />

Traditionally, when discuss<strong>in</strong>g cancer of the oesophagus, cancer of the gastrooesophageal<br />

junction (GOJ) is also <strong>in</strong>cluded. Cl<strong>in</strong>icians are very often confronted <strong>with</strong><br />

adenocarc<strong>in</strong>omas that straddle the GOJ. Various criteria have been used to categorize<br />

tumours situated at the GOJ. In most classification systems, the anatomic location of the<br />

epicentre or predom<strong>in</strong>ant mass of the tumour is used to determ<strong>in</strong>e whether the<br />

neoplasm is oesophageal or gastric (cardia) <strong>in</strong> orig<strong>in</strong>.<br />

Siewert and Ste<strong>in</strong> proposed a topographic classification for cardia carc<strong>in</strong>omas [11].<br />

Accord<strong>in</strong>g to these authors, epidemiologic, cl<strong>in</strong>ical, and pathologic data support a<br />

subclassification of adenocarc<strong>in</strong>omas aris<strong>in</strong>g <strong>in</strong>to the vic<strong>in</strong>ity (i.e. that have their centre<br />

<strong>with</strong><strong>in</strong> 5 cm proximal and distal of the anatomical cardia) of the GOJ <strong>in</strong>to:<br />

1. adenocarc<strong>in</strong>oma of the distal oesophagus, which usually arises from an area <strong>with</strong><br />

specialized <strong>in</strong>test<strong>in</strong>al metaplasia (i.e. Barrett oesophagus) and may <strong>in</strong>filtrate<br />

the GOJ from above (type I);<br />

2. true carc<strong>in</strong>oma of the cardia aris<strong>in</strong>g immediately at the GOJ (type II);<br />

3. subcardial carc<strong>in</strong>oma that <strong>in</strong>filtrates the GOJ and distal oesophagus from below<br />

(type III).<br />

In contrast to previously described classification systems, Siewert and Ste<strong>in</strong> attempted<br />

to solve the problem of splitt<strong>in</strong>g up GOJ tumours <strong>in</strong>to oesophageal and gastric tumours<br />

by creat<strong>in</strong>g a third entity [11]. This third entity, the so-called cardiacarc<strong>in</strong>oma, is<br />

lump<strong>in</strong>g a large group of tumours and ‘squeezes’ the true GOJ tumours between the<br />

type I and type II tumours. Their effort seems rather add<strong>in</strong>g to the confusion than<br />

help<strong>in</strong>g to solve the true problem. This classification is entirely based on identify<strong>in</strong>g the<br />

“anatomical” cardia and measur<strong>in</strong>g the centre of the tumour <strong>in</strong> relation to this<br />

anatomical cardia on the resected specimen (i.e. pathological stag<strong>in</strong>g). However,<br />

measur<strong>in</strong>g the centre of the tumour is impractical if not impossible for cl<strong>in</strong>ical stag<strong>in</strong>g<br />

purposes, which need to be as accurate as possible for mak<strong>in</strong>g appropriate therapeutic<br />

decisions.<br />

In 2000, the World Health Organization Classification of Tumours published Pathology<br />

and Genetics of Tumours of the Digestive System [12]. The authors formulated<br />

diagnostic criteria based on the follow<strong>in</strong>g def<strong>in</strong>ition of the GOJ: “the GOJ is the<br />

anatomical region at which the tubular oesophagus jo<strong>in</strong>s the stomach”. Accord<strong>in</strong>g to<br />

these authors, adenocarc<strong>in</strong>omas that cross the GOJ are called adenocarc<strong>in</strong>omas of the<br />

GOJ, regardless of where the bulk of the tumour lies. Adenocarc<strong>in</strong>omas located entirely<br />

above the GOJ, as def<strong>in</strong>ed above, are considered oesophageal carc<strong>in</strong>omas.<br />

Adenocarc<strong>in</strong>omas located entirely below the GOJ are considered gastric <strong>in</strong> orig<strong>in</strong>. The<br />

use of the ambiguous and often mislead<strong>in</strong>g term ‘carc<strong>in</strong>oma of the gastric cardia’ is<br />

discouraged. Depend<strong>in</strong>g on their size, these tumours should <strong>in</strong>stead be referred to as<br />

carc<strong>in</strong>oma of the body of the stomach [12].<br />

In the most recent recommendations of the International Union Aga<strong>in</strong>st Cancer (IUAC)<br />

[13], accord<strong>in</strong>g to the advice formulated <strong>in</strong> the supplement on the TNM classification,<br />

adenocarc<strong>in</strong>omas situated at the GOJ are to be classified <strong>in</strong>to oesophageal, GOJ or<br />

cardiac adenocarc<strong>in</strong>omas us<strong>in</strong>g a s<strong>in</strong>gle major criterion, i.e. the localization of the bulk of<br />

the tumour. If more than 50% of the mass of the tumour is situated <strong>in</strong> the cardia, the<br />

tumour should be considered to be of cardiac orig<strong>in</strong> and classified as a gastric tumour. If<br />

the mass of the tumour is predom<strong>in</strong>antly found <strong>in</strong> the oesophagus, it is to be classified<br />

as an oesophageal tumour. Furthermore, it is specified that a tumour situated on the<br />

GOJ is likely to be of oesophageal orig<strong>in</strong> when the neoplastic lesion was associated <strong>with</strong><br />

a Barrett oesophagus of the specialized or <strong>in</strong>test<strong>in</strong>al type. Unfortunately, the<br />

recommendations <strong>in</strong> the most recent Cancer Stag<strong>in</strong>g Manual on how to handle these<br />

tumours may not always be compatible <strong>with</strong> this classification, aga<strong>in</strong> creat<strong>in</strong>g confusion.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 13<br />

The chapter on stomach refers to the 50% rule, whereas the chapter on oesophagus<br />

<strong>in</strong>dicates that “tumours aris<strong>in</strong>g <strong>with</strong><strong>in</strong> the GOJ and gastric cardia that have m<strong>in</strong>imal<br />

<strong>in</strong>volvement (2 cm or less) of the oesophagus are considered primary gastric cancers”.<br />

A cont<strong>in</strong>u<strong>in</strong>g <strong>in</strong>crease <strong>in</strong> the <strong>in</strong>cidence of cardia cancer has been reported s<strong>in</strong>ce the mid<br />

1970s [14]. The output of scientific publications on cardia and cardiac cancer has<br />

evolved <strong>in</strong> parallel <strong>in</strong>ternationally. Today, the vast majority of data available on cardia<br />

and cardiac cancer are not comparable because of lack/variability of diagnostic criteria.<br />

This may result <strong>in</strong> therapeutic approaches based on loose (and non-scientific) grounds.<br />

Uniformity <strong>in</strong> classification, term<strong>in</strong>ology and diagnostic criteria is urgently needed. At<br />

present, at least <strong>in</strong> our experience, it would appear that the similarities between<br />

adenocarc<strong>in</strong>oma of the GOJ or cardia and Barrett adenocarc<strong>in</strong>oma outnumber the<br />

dissimilarities and are to be differentiated from gastric (<strong>in</strong>clud<strong>in</strong>g “subcardia”) cancer<br />

[15, 16].<br />

S<strong>in</strong>ce the ‘50% rule’ as proposed by the IUAC [13] is practical from a cl<strong>in</strong>ical stag<strong>in</strong>g<br />

po<strong>in</strong>t of view, easily be<strong>in</strong>g applicable on endoscopic ultrasonography (EUS) (the top of<br />

the longitud<strong>in</strong>al gastric mucosal folds be<strong>in</strong>g the endoscopic landmark between<br />

oesophagus and stomach), this rule will be used <strong>in</strong> the present CPG. “True” GOJ<br />

tumours should be classified and treated as oesophageal tumours.<br />

Key po<strong>in</strong>ts<br />

• If more than 50% of the mass of the tumour is situated <strong>in</strong> the cardia, the<br />

tumour should be considered to be of cardiac orig<strong>in</strong> and classified as a<br />

gastric tumour.<br />

• If the mass of the tumour is predom<strong>in</strong>antly found <strong>in</strong> the oesophagus, it<br />

should be classified as an oesophageal tumour.<br />

• Tumours of the gastro-oesophageal junction should be classified and have<br />

the same concept of treatment as oesophageal tumours.<br />

3.2 EARLY LESIONS<br />

3.2.1 Histology of the normal oesophagus<br />

The lum<strong>in</strong>al side of the normal oesophagus is l<strong>in</strong>ed by mucosa composed of epithelium,<br />

lam<strong>in</strong>a propria and the muscularis mucosae. Except for a short segment of columnar<br />

epithelium <strong>in</strong> the distal oesophagus at the gastro-oesophageal junction the normal<br />

oesophageal epithelium is a tough non-kerat<strong>in</strong>iz<strong>in</strong>g stratified squamous epithelium. This<br />

epithelium consists of a dynamic cell population which is renewed cont<strong>in</strong>uously. The<br />

different cell layers <strong>in</strong> the squamous epithelium - basal, <strong>in</strong>termediate or prickle cell<br />

layers and superficial layers (functional and surface) - are the morphological expression<br />

of processes of proliferation, differentiation or maturation and dy<strong>in</strong>g cells. A variety of<br />

cell types such as neuroendocr<strong>in</strong>e cells (Merkel cells), rare melanocytes, lymphocytes<br />

and Langerhans cells are normally present <strong>with</strong><strong>in</strong> the squamous epithelium of the<br />

oesophagus. The lam<strong>in</strong>a propria rests on a muscularis mucosae. The lam<strong>in</strong>a propria<br />

conta<strong>in</strong>s lymphatics, blood vessels, nerve fibres and occasional <strong>in</strong>flammatory cells. The<br />

three rema<strong>in</strong><strong>in</strong>g layers of the oesophageal wall are the submucosa, an area of loose<br />

connective tissue conta<strong>in</strong><strong>in</strong>g mucus-secret<strong>in</strong>g glands that open <strong>in</strong>to the lumen via ducts,<br />

the muscularis propria <strong>with</strong> an <strong>in</strong>ner circular and an outer longitud<strong>in</strong>al layer, and the<br />

adventitia. The oesophagus lacks a def<strong>in</strong><strong>in</strong>g layer of mesothelial cells.<br />

3.2.2 Barrett’s oesophagus<br />

3.2.2.1 Anatomy<br />

The muscular GOJ is the site at which the most distal portion of the oesophagus (the<br />

most distal segment of the lower oesophageal sph<strong>in</strong>cter [LOS]) meets the proximal<br />

stomach. Endoscopically, one can closely approximate the muscular GOJ by identify<strong>in</strong>g<br />

the proximal marg<strong>in</strong> of the gastric folds.


14 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

3.2.2.2 Histology<br />

The mucosal GOJ, also known as the mucosal squamo-columnar junction (SCJ) or Zl<strong>in</strong>e,<br />

is the site at which the squamous mucosa of the oesophagus meets columnar- l<strong>in</strong>ed<br />

mucosa. It is important to understand, however, that the SCJ may be at the same level<br />

as the muscular GOJ or may lie 1-2 cm above the muscular GOJ <strong>in</strong> ‘normal’ <strong>in</strong>dividuals.<br />

In order to avoid confusion between Barrett’s mucosa and normal – gastric – junctional<br />

columnar mucosa, especially <strong>in</strong> cases further complicated by the presence of hiatus<br />

hernia, an arbitrary m<strong>in</strong>imal length of 3 cm of Barrett’s mucosa from the GOJ was<br />

required before the diagnosis of Barrett’s mucosa could be made [17]. Short-segment<br />

Barrett’s oesophagus (SSBE) was later def<strong>in</strong>ed as Barrett’s mucosa


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 15<br />

The ‘double’ muscularis mucosae can only be visualized <strong>in</strong> (good quality) endoscopic<br />

mucosal resection (EMR) and operation specimens. Only <strong>in</strong>vasion through the orig<strong>in</strong>al,<br />

deeper muscularis mucosae is def<strong>in</strong>ed as “submucosal” <strong>in</strong>vasion [28, 29].<br />

3.2.3 Dysplasia <strong>in</strong> squamous epithelium<br />

In squamous epithelium dysplasia is classified as mild, moderate, or severe. With<br />

<strong>in</strong>creas<strong>in</strong>g grades of dysplasia there is a progressive <strong>in</strong>crease <strong>in</strong> dysplastic cells from the<br />

basal layer onwards untill the entire thickness of the epithelium is replaced. The latter<br />

state is described as carc<strong>in</strong>oma <strong>in</strong> situ [30]. When dysplastic cells reach the lum<strong>in</strong>al<br />

aspect of the epithelium <strong>with</strong>out cytoplasmic maturation the term carc<strong>in</strong>oma <strong>in</strong> situ can<br />

be used.<br />

Some authors use a simplified classification for non-<strong>in</strong>vasive squamous neoplastic<br />

lesions: low-grade <strong>in</strong>traepithelial neoplasia, which <strong>in</strong>cludes mild and moderate dysplasia,<br />

and high-grade <strong>in</strong>traepithelial neoplasia, which <strong>in</strong>cludes severe dysplasia and carc<strong>in</strong>oma<br />

<strong>in</strong> situ.<br />

By def<strong>in</strong>ition, the basement membrane is <strong>in</strong>tact <strong>in</strong> dysplasia. However, the junction of<br />

the epithelium <strong>with</strong> the underly<strong>in</strong>g stroma may become irregular [31].<br />

3.2.4 Dysplasia <strong>in</strong> columnar epithelium<br />

From a biological po<strong>in</strong>t of view, the progression of precursors and precursor lesions<br />

<strong>in</strong>to carc<strong>in</strong>omas is driven by the evolution and proliferation of clones of cells <strong>with</strong><br />

accumulated genetic errors related to control of cell proliferation, <strong>in</strong>tercellular<br />

adhesion, tumour suppression, etc., result<strong>in</strong>g <strong>in</strong> genomic <strong>in</strong>stability.<br />

Dysplasia is def<strong>in</strong>ed as ‘an unequivocal neoplastic epithelium conf<strong>in</strong>ed <strong>with</strong><strong>in</strong> its<br />

basement membrane [32]. Furthermore, dysplasia has the potential to progress <strong>in</strong>to<br />

<strong>in</strong>vasive malignancy. Although morphological detection <strong>in</strong> mucosal biopsy specimens is<br />

still the best method of detect<strong>in</strong>g patients at risk of develop<strong>in</strong>g cancer, it has its<br />

limitations:<br />

INTRA-OBSERVER AND INTER-OBSERVER VARIABILITY<br />

Numerous articles have been published on this topic demonstrat<strong>in</strong>g specific areas of<br />

discordance at the lower and higher end of the spectrum of early lesions. There is a<br />

significant degree of <strong>in</strong>tra-observer and <strong>in</strong>ter-observer variability <strong>in</strong> the diagnosis of<br />

dysplasia (unequivocal neoplastic atypia) versus reactive atypia even among experienced<br />

gastro<strong>in</strong>test<strong>in</strong>al pathologists [33-35]. Similarly, it has become apparent, especially from<br />

comparative studies between Western and Japanese pathologists, that the differential<br />

diagnosis between high-grade dysplasia, carc<strong>in</strong>oma <strong>in</strong> situ and <strong>in</strong>tramucosal carc<strong>in</strong>oma is<br />

prone to <strong>in</strong>tra-observer and <strong>in</strong>ter-observer variability [36-38].<br />

CLASSIFICATIONS<br />

Diagnosis of dysplasia is based on the detection of morphological changes. Accord<strong>in</strong>g to<br />

the severity of histological changes, dysplasia has been graded us<strong>in</strong>g either a three tier<br />

system or a two tier system (Table 7). The changes were <strong>in</strong>itially described as mild,<br />

moderate and severe dysplasia (morphological classification, 3-tier). In 1983, the<br />

Inflammatory Bowel Disease (IBD) study group classified dysplasia as negative, <strong>in</strong>def<strong>in</strong>ite<br />

or positive, i.e. low and high-grade (cl<strong>in</strong>ical classification, 2-tier: low-grade = mild and<br />

moderate dysplasia; high-grade = severe dysplasia) (Table 7)) [32].<br />

Grad<strong>in</strong>g <strong>in</strong> a two tier system would seem to be easier and more reproducible, and<br />

moreover to correlate <strong>with</strong> cl<strong>in</strong>ical implications. In the US and Western Europe, there<br />

was a general agreement that this cl<strong>in</strong>ically based classification was applicable to<br />

neoplastic changes <strong>in</strong> Barrett’s mucosa too [35, 39, 40]. In 2000, three new<br />

classifications for gastro<strong>in</strong>test<strong>in</strong>al dysplasia have been proposed: the Padova classification<br />

(gastric) [41], the Vienna classification [42] and the revised Vienna classification [43]<br />

(Table 7).


16 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

These new classifications aimed at : 1) chang<strong>in</strong>g the term<strong>in</strong>ology used, i.e. replac<strong>in</strong>g<br />

dysplasia by (<strong>in</strong>tra)epithelial neoplasia; 2) <strong>in</strong>clud<strong>in</strong>g not only dysplasia but also (<strong>in</strong>vasive)<br />

carc<strong>in</strong>omas; and 3) dist<strong>in</strong>guish<strong>in</strong>g ‘mucosal high-grade neoplasia’ <strong>in</strong>to high-grade<br />

dysplasia, suspicion for non-<strong>in</strong>vasive or <strong>in</strong>vasive carc<strong>in</strong>oma, non-<strong>in</strong>vasive carc<strong>in</strong>oma,<br />

<strong>in</strong>tramucosal carc<strong>in</strong>oma and carc<strong>in</strong>oma <strong>in</strong>vad<strong>in</strong>g submucosa or beyond. The major<br />

differences amongst these classifications reside <strong>in</strong> the subcategories <strong>in</strong>cluded/grouped<br />

and the figures attributed. Comparative studies us<strong>in</strong>g the new classifications have shown<br />

an improvement of the <strong>in</strong>ter-observer variability especially between the Western and<br />

Japanese pathologists [36-38, 44]. F<strong>in</strong>ally, a revision of the WHO classification of<br />

tumours of the digestive system was published at the end of 2000 [45]. The latter<br />

<strong>in</strong>troduced ‘high-grade <strong>in</strong>traepithelial neoplasia’ (<strong>in</strong>clud<strong>in</strong>g severe dysplasia and<br />

carc<strong>in</strong>oma <strong>in</strong> situ), but did not recommend one or another of the previously mentioned<br />

classifications. Up till now, these new classifications have not yet ga<strong>in</strong>ed widespread<br />

acceptance [25, 46]. Moreover, the authors of the Vienna classification stressed that the<br />

subdivisions related to ‘mucosal high-grade neoplasia’ (high-grade dysplasia, suspicion for<br />

<strong>in</strong>vasive carc<strong>in</strong>oma, non-<strong>in</strong>vasive carc<strong>in</strong>oma and <strong>in</strong>tramucosal carc<strong>in</strong>oma) may be<br />

important for research purposes and may not be needed for cl<strong>in</strong>ical purposes [42].<br />

Moreover, for resection specimens, only specific histological diagnoses should be given.<br />

Group classifications such as ‘mucosal high-grade neoplasia’ should not be used [44].<br />

In conclusion, especially concern<strong>in</strong>g the higher end of the spectrum of early lesions and<br />

<strong>in</strong> view of the importance of a multidiscipl<strong>in</strong>ary approach, it is important for a<br />

pathologist to have a clear understand<strong>in</strong>g of the particular treatment regimens available<br />

and to be applied under particular circumstances. Classification is a chosen arrangement<br />

of elements <strong>in</strong> relation to a purpose. For the physician, a classification should be<br />

cl<strong>in</strong>ically relevant. Currently, the ma<strong>in</strong> cl<strong>in</strong>ical options are no follow-up, follow-up, local<br />

treatment by endoscopy, m<strong>in</strong>imally <strong>in</strong>vasive (laparoscopic) surgery and extensive<br />

surgery <strong>in</strong>clud<strong>in</strong>g lymph node dissection (see below). Many studies have evaluated the<br />

potential utility of immunohistochemical or molecular markers as additional techniques<br />

<strong>in</strong> detect<strong>in</strong>g dysplasia, however <strong>with</strong> limited success. Additional confirmation by an<br />

expert pathologist (second op<strong>in</strong>ion) is advocated, especially when therapeutic<br />

<strong>in</strong>tervention is considered [25]. Today, the new classifications should be taken for what<br />

they are, i.e. attempts at an <strong>in</strong>ternational level to reach consensus on histological<br />

diagnosis of dysplasia <strong>in</strong> ‘chosen arrangements of elements’ which may eventually<br />

generate guidel<strong>in</strong>es for the development of diagnostic and management strategies.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 17<br />

Table 7. Overview of classifications for dysplasia.<br />

IBD Padova Vienna Revised Vienna TNM<br />

Negative for 1. Negative for dysplasia 1. No neoplasia 1. No neoplasia<br />

dysplasia 1.0. Normal<br />

1.1. Reactive foveolar<br />

hyperplasia<br />

1.2. Intest<strong>in</strong>al metaplasia (IM)<br />

1.2.1. IM complete type<br />

1.2.1. IM <strong>in</strong>complete type<br />

Indef<strong>in</strong>ite for 2. Indef<strong>in</strong>ite for dysplasia 2. Indef<strong>in</strong>ite for dysplasia 2. Indef<strong>in</strong>ite for dysplasia<br />

dysplasia 2.1. Foveolar hyperplasia<br />

2.2. Hyperproliferative IM<br />

3. Non-<strong>in</strong>vasive neoplasia (flat<br />

or elevated)<br />

Low-grade 3.1. Low-grade 3. Low-grade<br />

3. Low-grade<br />

dysplasia<br />

adenoma/dysplasia adenoma/dysplasia<br />

High-grade<br />

dysplasia<br />

3.2. High-grade 4. Non-<strong>in</strong>vasive highgrade<br />

neoplasia<br />

4.1. High-grade<br />

adenoma/dysplasia<br />

3.2.1. High-grade <strong>in</strong>clud<strong>in</strong>g<br />

suspicious carc<strong>in</strong>oma <strong>with</strong>out<br />

<strong>in</strong>vasion (<strong>in</strong>traglandular)<br />

3.2.2. High-grade <strong>in</strong>clud<strong>in</strong>g 4.2. Non-<strong>in</strong>vasive<br />

carc<strong>in</strong>oma <strong>with</strong>out <strong>in</strong>vasion carc<strong>in</strong>oma (carc<strong>in</strong>oma <strong>in</strong><br />

(<strong>in</strong>traglandular)<br />

situ)<br />

4. Suspicious for <strong>in</strong>vasive 4.3. Suspicious for<br />

carc<strong>in</strong>oma<br />

<strong>in</strong>vasive carc<strong>in</strong>oma<br />

5. Invasive adenocarc<strong>in</strong>oma 5. Invasive neoplasia<br />

5.1. Intramucosal<br />

carc<strong>in</strong>oma<br />

5.2. Submucosal<br />

carc<strong>in</strong>oma (or deeper<br />

<strong>in</strong>filtration)<br />

Key po<strong>in</strong>ts<br />

4. High-grade neoplasia<br />

4.1. High-grade<br />

adenoma/dysplasia<br />

4.2. Non-<strong>in</strong>vasive<br />

carc<strong>in</strong>oma (carc<strong>in</strong>oma <strong>in</strong><br />

situ)<br />

4.3. Suspicious for<br />

<strong>in</strong>vasive carc<strong>in</strong>oma<br />

4.4. Intramucosal<br />

carc<strong>in</strong>oma<br />

• There is no consensus about the def<strong>in</strong>ition of Barrett’s oesophagus.<br />

• Several classifications are available for dysplasia. For the physician, the used<br />

classification should be cl<strong>in</strong>ically relevant.<br />

Tis<br />

T1a<br />

T1b


18 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

4 FINAL RECOMMENDATIONS ON<br />

OESOPHAGEAL CANCER<br />

4.1 FLOWCHART


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 19<br />

4.2 DIAGNOSIS<br />

The diagnosis of oesophageal cancer should <strong>in</strong>clude a history and cl<strong>in</strong>ical exam<strong>in</strong>ation.<br />

However, a diagnosis purely based on cl<strong>in</strong>ical grounds is unreliable [47]. Two diagnostic<br />

procedures, i.e. flexible upper gastro<strong>in</strong>test<strong>in</strong>al (GI) endoscopy and barium contrast<br />

radiology, are available. However, barium studies are less sensitive than endoscopy for<br />

the diagnosis of early malignancy and they cannot reliably diagnose premalignant lesions<br />

<strong>in</strong>clud<strong>in</strong>g dysplasia. Therefore, flexible upper GI endoscopy <strong>with</strong> at least biopsies of all<br />

suspicious lesions is recommended as the diagnostic procedure of choice <strong>in</strong> patients<br />

<strong>with</strong> suspected oesophageal cancer [47]. Nevertheless, barium studies can be useful to<br />

determ<strong>in</strong>e the oesophageal or gastric extent of a tumour, to confirm the presence of a<br />

tracheo-oesophageal fistula or to document complete lum<strong>in</strong>al obstruction [48].<br />

The follow<strong>in</strong>g alarm symptoms are associated <strong>with</strong> oesophageal (and gastric) cancer and<br />

should lead to a referral for early endoscopy and biopsies: dysphagia, recurrent<br />

vomit<strong>in</strong>g, anorexia, weight loss and gastro<strong>in</strong>test<strong>in</strong>al blood loss [47, 49]. The general<br />

practitioner has an important role <strong>in</strong> recogniz<strong>in</strong>g these symptoms. In a recent<br />

prospective study, Bowrey et al. found a sensitivity of 85% us<strong>in</strong>g alarm symptoms for<br />

the referral for endoscopy [49]. This means that 15% of patients <strong>with</strong> oesophageal<br />

cancer would not have been referred for <strong>in</strong>itial endoscopic assessment because of the<br />

absence of alarm symptoms. Importantly, patients <strong>with</strong> alarm symptoms had a<br />

significantly more advanced tumour stage (metastatic disease <strong>in</strong> 47% vs. 11%; p < 0.001),<br />

were less likely to undergo surgical resection (50% vs. 95%; p


20 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Table 8. Features associated <strong>with</strong> high risk for develop<strong>in</strong>g oesophageal<br />

cancer.<br />

Cl<strong>in</strong>ical features:<br />

• Alcohol and tobacco abuse<br />

• Longstand<strong>in</strong>g gastro-oesophageal reflux disease<br />

• Neck, mouth and head cancer<br />

• Previous oesophageal cancer<br />

• Barrett’s oesophagus<br />

• Caustic <strong>in</strong>jury<br />

• Papilloma<br />

• Genetic diseases<br />

• Various: Achalasia, Plummer-V<strong>in</strong>son, Celiac disease<br />

Endoscopic features:<br />

• stricture<br />

• ulceration<br />

• nodularity<br />

Histological features:<br />

Recommendations<br />

• high-grade dysplasia<br />

• multifocal dysplastic lesions, persist<strong>in</strong>g after adequate conservative<br />

treatment for 6-8 weeks<br />

1. Patients present<strong>in</strong>g <strong>with</strong> any of the follow<strong>in</strong>g alarm symptoms <strong>with</strong><strong>in</strong><br />

the cl<strong>in</strong>ical context of potential oesophageal pathology should be<br />

referred for early endoscopy and biopsies: dysphagia, recurrent<br />

vomit<strong>in</strong>g, anorexia, weight loss, gastro<strong>in</strong>test<strong>in</strong>al blood loss (1C<br />

recommendation).<br />

2. Flexible upper gastro<strong>in</strong>test<strong>in</strong>al endoscopy <strong>with</strong> at least biopsies of all<br />

suspicious lesions is recommended as the diagnostic procedure of<br />

choice <strong>in</strong> patients <strong>with</strong> suspected oesophageal cancer (1C<br />

recommendation).<br />

3. High-resolution endoscopy (HRE) and chromoendoscopy is not<br />

rout<strong>in</strong>ely recommended, but may be of value <strong>in</strong> screen<strong>in</strong>g and<br />

follow-up of high-risk patients (2C recommendation).<br />

4.3 WORK-UP OF DYSPLASTIC LESIONS<br />

Evidence from a few RCTs [47] and one low-quality systematic review [55] suggests<br />

that anti-reflux surgery is not <strong>in</strong>dicated <strong>in</strong> patients <strong>with</strong> Barrett’s oesophagus to reduce<br />

the risk of progression to adenocarc<strong>in</strong>oma.<br />

Patients diagnosed <strong>with</strong> high-grade dysplasia should have careful endoscopic assessment<br />

[47]. However, dysplastic lesions <strong>in</strong> patients <strong>with</strong> Barrett’s oesophagus are difficult to<br />

detect us<strong>in</strong>g conventional endoscopy. Recent studies suggest that high-resolution<br />

endoscopy (HRE) – <strong>with</strong> or <strong>with</strong>out chromoendoscopy –, narrow band imag<strong>in</strong>g (NBI)<br />

or autofluorescence may improve the detection of dysplastic lesions (see above) [56].


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 21<br />

In patients <strong>with</strong> Barrett's oesophagus, a structured biopsy protocol <strong>with</strong> quadrantic<br />

biopsies every two centimetres and biopsy of any visible lesion is recommended. In the<br />

presence of dysplasia, quadrantic biopsies should be taken every 1 cm dur<strong>in</strong>g<br />

surveillance and control endoscopies [57, 58]. In addition to the endoscopic assessment,<br />

patients <strong>with</strong> high-grade dysplasia should have a careful pathological assessment [47].<br />

Evaluation of suspected high-grade dysplasia <strong>in</strong> Barrett's oesophagus biopsies should be<br />

undertaken <strong>with</strong> knowledge of the cl<strong>in</strong>ical and endoscopic background. Pathologists<br />

should follow a classification for report<strong>in</strong>g dysplasia that the multidiscipl<strong>in</strong>ary team is<br />

familiar <strong>with</strong>. An overview of classifications is already provided <strong>in</strong> chapter 3.2.4. Where<br />

therapeutic <strong>in</strong>tervention is contemplated on the basis of high-grade dysplasia, the<br />

diagnosis should be validated by a second pathologist experienced <strong>in</strong> this area because<br />

of the significant <strong>in</strong>ter- and <strong>in</strong>tra-observer variation [47, 56]. Further biopsies or<br />

diagnostic endoscopic mucosal resection (EMR) should be done if there is uncerta<strong>in</strong>ty.<br />

Biopsies should also be reviewed at a multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong> access to the<br />

cl<strong>in</strong>ical <strong>in</strong>formation.<br />

F<strong>in</strong>ally, as the diagnosis, treatment and follow-up of patients <strong>with</strong> oesophageal highgrade<br />

dysplastic lesions is very specific and new techniques rapidly become available,<br />

these patients should be referred to centres or network reference centres <strong>with</strong> the<br />

appropriate endoscopic and surgical expertise and facilities [47]<br />

Recommendations<br />

4. Reduction of risk of progression to adenocarc<strong>in</strong>oma is not an<br />

<strong>in</strong>dication for anti-reflux surgery <strong>in</strong> patients <strong>with</strong> Barrett's<br />

oesophagus (2A recommendation).<br />

5. In patients <strong>with</strong> Barrett's oesophagus there should be a structured<br />

biopsy protocol <strong>with</strong> quadrantic biopsies every two centimetres and<br />

biopsy of any visible lesion (1C recommendation).<br />

6. Pathologists should follow a classification for report<strong>in</strong>g dysplasia that<br />

the multidiscipl<strong>in</strong>ary team is familiar <strong>with</strong> (1C recommendation).<br />

7. Evaluation of suspected high-grade dysplasia <strong>in</strong> Barrett's oesophagus<br />

biopsies should be undertaken <strong>with</strong> knowledge of the cl<strong>in</strong>ical and<br />

endoscopic background (1C recommendation).<br />

8. Patients confirmed <strong>with</strong> high-grade dysplasia should have careful<br />

endoscopic and pathological assessment (1C recommendation).<br />

9. High-resolution endoscopy +/- chromoendoscopy as well as every 1<br />

cm qaudrantic biopsies is recommended <strong>in</strong> patients <strong>with</strong> a dysplastic<br />

or neoplastic lesion <strong>in</strong> a Barrett's oesophagus (1C recommendation).<br />

10. Where therapeutic <strong>in</strong>tervention is contemplated on the basis of<br />

high-grade dysplasia, the diagnosis should be validated by a second<br />

pathologist experienced <strong>in</strong> this area and further biopsies or<br />

eventually diagnostic endoscopic mucosal resection (EMR) should be<br />

done if there is uncerta<strong>in</strong>ty (1C recommendation).<br />

11. Treatment options for patients <strong>with</strong> high-grade dysplasia should be<br />

discussed at a multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong> access to the cl<strong>in</strong>ical<br />

and pathological <strong>in</strong>formation (expert op<strong>in</strong>ion).<br />

12. Patients <strong>with</strong> high-grade dysplasia should be referred to centres or<br />

network reference centres <strong>with</strong> the appropriate endoscopic and<br />

surgical expertise and facilities (1C recommendation).


22 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

4.4 STAGING<br />

In patients <strong>with</strong> oesophageal cancer, CT scan of the chest and abdomen <strong>with</strong><br />

<strong>in</strong>travenous contrast and gastric distension <strong>with</strong> oral contrast or water should be<br />

performed rout<strong>in</strong>ely. The liver should at least be imaged <strong>in</strong> the arterial and portal<br />

venous phase [47]. Slice thickness should be 5 mm or less [56]. The ma<strong>in</strong> contribution<br />

of CT scan to the stag<strong>in</strong>g of oesophageal cancer is the detection of distant metastases<br />

and gross <strong>in</strong>vasion of adjacent structures/organs [47, 56, 59]. If metastatic disease is<br />

detected <strong>with</strong> CT scan, curative treatment is excluded and additional stag<strong>in</strong>g <strong>with</strong> EUS<br />

and/or PET scan is unnecessary. CT scan has also an acceptable diagnostic accuracy for<br />

locoregional stag<strong>in</strong>g (T and N), although EUS is clearly more sensitive (see below) [47,<br />

56]. Given the high <strong>in</strong>cidence of cervical lymph node metastasis [60], CT scan should<br />

also <strong>in</strong>clude the cervical region.<br />

EUS has emerged as the imag<strong>in</strong>g technique of choice for locoregional stag<strong>in</strong>g [47, 56],<br />

<strong>with</strong> a diagnostic accuracy rang<strong>in</strong>g from 53-94% (median 83%) for T-stag<strong>in</strong>g and from<br />

54-94% for N-stag<strong>in</strong>g (median 76%) [61]. In stenotic tumours, the accuracy of EUS is<br />

further improved <strong>with</strong> the use of dilation, which <strong>in</strong> most cases permits passage of the<br />

endoscope [62, 63]. However, this is associated <strong>with</strong> a risk of perforation [64].<br />

Importantly, most studies deal<strong>in</strong>g <strong>with</strong> the accuracy of N-stag<strong>in</strong>g only assess the yes-no<br />

possibility of positive lymph nodes, <strong>with</strong>out correlat<strong>in</strong>g the EUS f<strong>in</strong>d<strong>in</strong>gs of a positive<br />

lymph node to the histopathological f<strong>in</strong>d<strong>in</strong>gs of that particular node. As a result,<br />

although a f<strong>in</strong>al pathology report may conclude N1 disease to be present, a node <strong>with</strong><br />

positive EUS f<strong>in</strong>d<strong>in</strong>gs may be histopathologically negative and vice versa. Therefore, the<br />

diagnostic accuracy of EUS for N-stag<strong>in</strong>g may be overestimated.<br />

The accuracy of N stag<strong>in</strong>g <strong>with</strong> EUS is further improved <strong>with</strong> f<strong>in</strong>e needle aspiration<br />

cytology (FNAC) [47, 56]. FNAC needs to be <strong>in</strong>terpreted by an experienced<br />

pathologist.<br />

Neck imag<strong>in</strong>g <strong>with</strong> ultrasonography or CT enables the detection of metastatic lymph<br />

nodes that are cl<strong>in</strong>ically not palpable [47] or can be used to guide f<strong>in</strong>e needle aspiration<br />

of suspicious lymph nodes <strong>with</strong> very high accuracy and specificity [56, 65].<br />

In the absence of clear metastatic disease on conventional CT/EUS/cervical US,<br />

PET(/CT) may provide valuable additional <strong>in</strong>formation, <strong>in</strong> particular when consider<strong>in</strong>g<br />

multimodality treatment <strong>with</strong> curative option. In such cases, basel<strong>in</strong>e PET(/CT) allows<br />

better response assessment as compared to conventional CT/EUS/cervical US.<br />

FNCLCC made specific recommendations on the use of PET scan <strong>in</strong> the diagnosis and<br />

stag<strong>in</strong>g of oesophageal cancer [66]. In addition, the <strong>KCE</strong> recently published an HTA<br />

report on the use of PET scan [67]. S<strong>in</strong>ce this HTA report, numerous observational<br />

studies of variable quality have been published [68-75]. For <strong>in</strong>itial stag<strong>in</strong>g of patients<br />

<strong>with</strong>out apparent metastases on CT scan, especially <strong>in</strong> locally advanced disease (cT>2 or<br />

cN1 or cN doubtful), PET(/CT) is useful for detect<strong>in</strong>g positive lymph nodes and distant<br />

metastatic disease [67]. However, <strong>in</strong> early-stage disease (cT1-2N0) the probability that<br />

PET(/CT) will significantly upstage disease is low.<br />

Magnetic resonance imag<strong>in</strong>g (MRI) was not found to be superior to conventional<br />

imag<strong>in</strong>g techniques such as CT scan ([47, 56]. Therefore, MRI should be reserved for<br />

those patients who cannot undergo CT or used for additional <strong>in</strong>vestigation follow<strong>in</strong>g CT<br />

and/or EUS.<br />

In patients <strong>with</strong> cl<strong>in</strong>ical or imag<strong>in</strong>g features suspicious of tracheobronchial <strong>in</strong>vasion (e.g.<br />

cough aggravated by swallow<strong>in</strong>g <strong>in</strong> case of a fistula) or extr<strong>in</strong>sic compression,<br />

bronchoscopy comb<strong>in</strong>ed <strong>with</strong> bronchoscopic ultrasound (BUS)<br />

and/or biopsy can be useful [47, 76, 77]. The underly<strong>in</strong>g evidence, however, is weak to<br />

very weak. Observational data also provide weak evidence for the occurrence of<br />

synchronous neoplasms of the bronchial tree <strong>in</strong> patients <strong>with</strong> squamous cell carc<strong>in</strong>oma<br />

of the oropharynx and the oesophagus, which is an <strong>in</strong>dication for bronchoscopy [78].<br />

Few studies are available to support the use of <strong>in</strong>vasive stag<strong>in</strong>g <strong>with</strong> thoracoscopy and<br />

laparoscopy (+/- peritoneal cytology and/or laparoscopic ultrasound) [47, 56, 79].


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 23<br />

Thoracoscopy can be useful for the detection of positive mediast<strong>in</strong>al lymph nodes, while<br />

laparoscopy can be considered <strong>in</strong> patients <strong>with</strong> oesophageal tumours <strong>with</strong> a gastric<br />

component [47]. Laparoscopy has a higher specificity for M stag<strong>in</strong>g <strong>in</strong> comparison <strong>with</strong><br />

CT scan, but obviously carries a higher risk of morbidity [56].<br />

Recommendations<br />

13. In patients <strong>with</strong> oesophageal cancer, CT scan of the chest (<strong>in</strong>clud<strong>in</strong>g<br />

lower neck region) and abdomen <strong>with</strong> <strong>in</strong>travenous contrast and<br />

gastric distension <strong>with</strong> oral contrast or water should be performed<br />

rout<strong>in</strong>ely. The liver should at least be imaged <strong>in</strong> the arterial and<br />

portal venous phase (1C recommendation).<br />

14. Patients <strong>with</strong> oesophageal or gastro-oesophageal junction cancers<br />

who are candidates for any curative therapy should have their<br />

tumours staged <strong>with</strong> endoscopic ultrasonography +/- f<strong>in</strong>e needle<br />

aspiration cytology (FNAC) and ultrasonography of the neck (1B<br />

recommendation).<br />

15. F<strong>in</strong>e needle aspiration cytology (FNAC) needs to be <strong>in</strong>terpreted by<br />

an experienced pathologist (expert op<strong>in</strong>ion).<br />

16. In patients <strong>with</strong> oesophageal cancer and an option for curative<br />

treatment after conventional stag<strong>in</strong>g (CT/endoscopic<br />

ultrasonography), PET(/CT) scan may be considered for the stag<strong>in</strong>g<br />

of lymph nodes (loco-regional, distal or all lymph nodes) and distant<br />

sites other than lymph nodes (1C recommendation).<br />

17. The follow<strong>in</strong>g exam<strong>in</strong>ations can be considered for specific<br />

<strong>in</strong>dications: MRI, bronchoscopy +/- bronchial ultrasonography (BUS)<br />

+/- biopsy, thoracoscopy, or laparoscopy (1C recommendation).<br />

4.5 TREATMENT OF MUCOSAL CANCER<br />

The pr<strong>in</strong>ciples of anatomopathological evaluation of mucosal cancer are similar to those<br />

of high-grade dysplasia, <strong>in</strong>clud<strong>in</strong>g validation of the diagnosis by a second pathologist,<br />

additional biopsies or diagnostic EMR <strong>in</strong> case of uncerta<strong>in</strong>ty, and discussion of treatment<br />

options at the MDT [47].<br />

Only observational studies are available compar<strong>in</strong>g endoscopic mucosal resection (EMR)<br />

to surgery for the treatment of superficial oesophageal cancer [56]. Both treatments<br />

were found to be equally effective, but EMR was associated <strong>with</strong> less complications.<br />

Therefore, superficial oesophageal cancer limited to the mucosa should be treated <strong>with</strong><br />

EMR, tak<strong>in</strong>g <strong>in</strong>to account the stage, size, length of Barrett, histological type,<br />

differentiation grade, and lymphovascular <strong>in</strong>vasion [47]. For example, <strong>in</strong> case of mucosal<br />

cancer <strong>in</strong> a long Barrett’s segment, complete resection of the Barrett’s area <strong>with</strong> EMR is<br />

unlikely. In these cases, surgery (e.g. gastric pull-up or vagal spar<strong>in</strong>g oesophagectomy<br />

<strong>with</strong> colon <strong>in</strong>terposition) may rema<strong>in</strong> the treatment of choice.<br />

En-bloc resection – allow<strong>in</strong>g better pathology stag<strong>in</strong>g of deep and lateral marg<strong>in</strong>s –<br />

should be aimed at <strong>with</strong> the appropriate technique (EMR for lesions less than 12 mm)<br />

[80-82].<br />

Mucosal ablative techniques, such as argon plasma coagulation (APC), photodynamic<br />

therapy (PDT) or laser, are <strong>in</strong>vestigational [47, 56]. Only small observational studies<br />

showed the benefits of these techniques, and they should therefore be limited to units<br />

<strong>with</strong> appropriate expertise.<br />

Moreover, APC was associated <strong>with</strong> buried neoplastic glands and should not be<br />

considered for curative treatment. Importantly, PDT is associated <strong>with</strong> a risk of<br />

stricture formation <strong>in</strong> up to 30% of patients [83]. Above this, mucosal ablative<br />

techniques make an anatomopathological control of the stag<strong>in</strong>g results impossible.


24 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Recommendations<br />

18. Where therapeutic <strong>in</strong>tervention is considered for a supposedly T1a<br />

mucosal cancer, the diagnosis should be validated by a second<br />

pathologist experienced <strong>in</strong> this area. Further biopsies or eventually<br />

diagnostic endoscopic mucosal resection (EMR) should be done if<br />

there is uncerta<strong>in</strong>ty (1C recommendation).<br />

19. Treatment options for patients <strong>with</strong> mucosal cancer should be<br />

discussed at a multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong> access to the cl<strong>in</strong>ical<br />

and pathological <strong>in</strong>formation (expert op<strong>in</strong>ion).<br />

20. Superficial oesophageal cancer limited to the mucosa should be<br />

treated <strong>with</strong> endoscopic mucosal resection (EMR), tak<strong>in</strong>g <strong>in</strong>to<br />

account well-def<strong>in</strong>ed criteria relat<strong>in</strong>g to stage, size, length of Barrett,<br />

histological type, differentiation grade, and lymphovascular <strong>in</strong>vasion<br />

(1C recommendation).<br />

21. Mucosal ablative techniques, such as argon plasma coagulation<br />

(APC), photodynamic therapy (PDT) or laser, are <strong>in</strong>vestigational and<br />

should be limited to units <strong>with</strong> appropriate expertise (1C<br />

recommendation).<br />

4.6 TREATMENT OF CANCER BEYOND THE MUCOSA<br />

4.6.1 Neoadjuvant treatment<br />

In 2006, Cancer Care Ontario (CCO) published a high-quality CPG (<strong>in</strong>clud<strong>in</strong>g metaanalyses)<br />

on the use of neoadjuvant treatment for oesophageal cancer [84]. Three<br />

additional meta-analyses [85-87] and one systematic review [88] have been published<br />

s<strong>in</strong>ce then. Only Gebski et al. found new evidence <strong>in</strong> addition to that presented by<br />

CCO [86].<br />

CCO identified 6 RCTs compar<strong>in</strong>g preoperative radiotherapy and surgery vs. surgery<br />

alone [84]. No significant difference was detected <strong>in</strong> the risk of death at one year (RR<br />

1.01; 95%CI 0.88 – 1.16; p=0.90). These results are <strong>in</strong> l<strong>in</strong>e <strong>with</strong> those of Arnott et al.,<br />

who found a hazard ratio (HR) of 0.98 (95%CI 0.78 – 1.01), suggest<strong>in</strong>g a small but nonsignificant<br />

absolute survival benefit of 3% at 2 years and 4% at 5 years <strong>in</strong> favour of<br />

preoperative radiotherapy [85].<br />

N<strong>in</strong>e different RCTs were identified by CCO compar<strong>in</strong>g preoperative chemotherapy<br />

and surgery vs. surgery alone [84]. In addition, two RCTs were identified compar<strong>in</strong>g<br />

preoperative and postoperative chemotherapy and surgery vs. surgery alone. All eleven<br />

trials were also <strong>in</strong>cluded <strong>in</strong> a recent Cochrane review [87]. A meta-analysis of the first 9<br />

RCTs found a pooled RR of 1.00 (95%CI 0.83 – 1.19; p=0.97), detect<strong>in</strong>g no difference <strong>in</strong><br />

survival at one year [84]. These results are <strong>in</strong> l<strong>in</strong>e <strong>with</strong> the Cochrane review (HR 0.88;<br />

95%CI 0.75 – 1.04). Trials also reported risks of toxicity <strong>with</strong> chemotherapy that ranged<br />

from 11% to 90% [87].<br />

Gebski et al. identified 10 RCTs (of which one was an unpublished thesis, and two were<br />

published as an abstract) compar<strong>in</strong>g preoperative CRT and surgery vs. surgery alone<br />

[86]. A HR for all-cause mortality of 0.81 (95%CI 0.70 – 0.93; p=0.002) <strong>in</strong> favour of<br />

preoperative CRT was found, correspond<strong>in</strong>g to a 13% absolute difference <strong>in</strong> survival at<br />

2 years. Patients <strong>with</strong> adenocarc<strong>in</strong>oma seemed to obta<strong>in</strong> the highest benefit, although<br />

conflict<strong>in</strong>g evidence is available.<br />

These results are not <strong>in</strong> l<strong>in</strong>e <strong>with</strong> those presented by CCO, who found no difference <strong>in</strong><br />

one-year survival between the 2 treatment arms (RR 0.91; 95%CI 0.79 – 1.06; p=0.21)<br />

[84]. CCO also identified 1 RCT compar<strong>in</strong>g preoperative CRT (bleomyc<strong>in</strong> +<br />

radiotherapy) to preoperative radiotherapy alone. No significant difference <strong>in</strong> survival<br />

between the two treatment groups was found [84].


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 25<br />

In a systematic review of 6 RCTs by Graham et al., a RR of 0.81 (95%CI 0.64 – 1.02)<br />

compared to surgery alone was found. This corresponded to an additional 40 days of<br />

quality-adjusted life expectancy [88].<br />

S<strong>in</strong>ce the literature search of CCO and Gebski et al., results of two additional small<br />

RCTs became available, compar<strong>in</strong>g preoperative CRT (cisplat<strong>in</strong> + 5-FU) to surgery<br />

alone [89, 90]. No significant difference <strong>in</strong> survival was found.<br />

Based on the evidence presented, the advantage of neoadjuvant treatment is small and<br />

although neoadjuvant treatment is frequently used, it cannot be considered as a rout<strong>in</strong>e<br />

practice <strong>in</strong> patients <strong>with</strong> oesophageal cancer. It is clear that for each <strong>in</strong>dividual patient<br />

the need for neoadjuvant treatment should be discussed dur<strong>in</strong>g a multidiscipl<strong>in</strong>ary<br />

meet<strong>in</strong>g. If neoadjuvant treatment is part of the treatment plan, it should be<br />

adm<strong>in</strong>istered accord<strong>in</strong>g to a predef<strong>in</strong>ed protocol. This protocol should also specify the<br />

role of PET(/CT) scan <strong>in</strong> the evaluation of treatment response to neoadjuvant<br />

treatment. In a previous <strong>KCE</strong> report, some evidence was identified support<strong>in</strong>g the role<br />

of PET scan <strong>in</strong> the assessment of treatment response to neoadjuvant treatment, us<strong>in</strong>g<br />

the <strong>in</strong>itial stag<strong>in</strong>g PET results as a comparison [67]. These conclusions are confirmed by<br />

subsequent prospective trials [74, 91]. F<strong>in</strong>ally, <strong>in</strong> all trials, a complete pathological<br />

response was associated <strong>with</strong> the most favourable prognosis.<br />

S<strong>in</strong>ce the use of neoadjuvant treatment for patients <strong>with</strong> oesophageal cancer still is<br />

controversial, it is recommended to prospectively register cl<strong>in</strong>ical outcomes and<br />

adverse events of the comb<strong>in</strong>ed treatment. In fact, besides the mandatory cancer<br />

registration, prospective registration of cl<strong>in</strong>ical outcomes and adverse events is<br />

mandatory for all oncologic patients accord<strong>in</strong>g to the Royal Decree of March 21 st 2003<br />

settl<strong>in</strong>g the norms for the healthcare programme <strong>in</strong> oncology [92].<br />

Recommendations<br />

4.6.2 Surgical treatment<br />

22. Preoperative radiotherapy alone is not recommended for patients<br />

<strong>with</strong> oesophageal cancer (2A recommendation).<br />

23. Neoadjuvant treatment is not rout<strong>in</strong>ely <strong>in</strong>dicated for patients <strong>with</strong><br />

oesophageal cancer (2A recommendation).<br />

24. The need for neoadjuvant treatment should be discussed dur<strong>in</strong>g a<br />

multidiscipl<strong>in</strong>ary meet<strong>in</strong>g (expert op<strong>in</strong>ion).<br />

25. Prospective registration of cl<strong>in</strong>ical outcomes and adverse events of<br />

comb<strong>in</strong>ed treatment is recommended (expert op<strong>in</strong>ion).<br />

Surgical resection rema<strong>in</strong>s the standard treatment for patients <strong>with</strong> resectable<br />

oesophageal cancer [47, 56, 84, 88]. Patients are considered to have unresectable<br />

disease <strong>in</strong> case of metastatic disease, positive lymph nodes outside the region of surgery<br />

and/or radiotherapy (i.e. the cervical, thoracic and upper abdom<strong>in</strong>al compartment),<br />

advanced locoregional disease (e.g. tracheo-oesophageal fistula), or severe comorbidity.<br />

Until now, only few studies are available compar<strong>in</strong>g surgery to other treatment<br />

modalities [56, 93, 94]. One moderate-quality eastern RCT compared primary<br />

chemoradiotherapy (CRT) and salvage surgery to primary ‘standard’ oesophagectomy<br />

[94]. No significant difference <strong>in</strong> 2-year survival and disease-free survival was found<br />

between the 2 treatment groups. Six out of 36 patients (17%) assigned to primary CRT<br />

were treated <strong>with</strong> salvage oesophagectomy [94]. A French study compared CRT<br />

followed by surgery to CRT alone <strong>in</strong> patients <strong>with</strong> operable oesophageal cancer that<br />

responded to CRT [93]. Two-year survival and 2-year local recurrence rate did not<br />

differ significantly.<br />

The results of these two RCTs need to be <strong>in</strong>terpreted <strong>with</strong> caution, s<strong>in</strong>ce they had<br />

important methodological limitations (no accurate <strong>in</strong>formation on randomisation<br />

procedure, absence of bl<strong>in</strong>d<strong>in</strong>g). Above this, as <strong>in</strong> many other trials, surgery was not<br />

standardized [93, 94].


26 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Surgery for oesophageal cancer should be aimed at achiev<strong>in</strong>g an R0 resection, and<br />

should be considered preferentially through an en bloc resection (i.e. <strong>with</strong> systematic<br />

abdom<strong>in</strong>al and mediast<strong>in</strong>al lymph node dissection, the so-called two field<br />

lymphadenectomy) [47, 56]. This recommendation is ma<strong>in</strong>ly based on the results of 1<br />

RCT that compared extended transthoracic resection <strong>with</strong> two-field lymphadenectomy<br />

to limited transhiatal resection <strong>with</strong>out two-field lymphadenectomy [95]. A trend<br />

towards improved 5-year survival was found <strong>in</strong> favour of the extended thoracic group.<br />

The goal of lymphadenectomy is to optimize stag<strong>in</strong>g and to improve long-term<br />

outcomes by decreas<strong>in</strong>g the risk for local recurrence and <strong>in</strong>creas<strong>in</strong>g the probability of<br />

an R0 resection [47, 56]. Two-field lymphadenectomy <strong>in</strong>volves the clearance of<br />

mediast<strong>in</strong>al (1 st field) and abdom<strong>in</strong>al (2 nd field) lymph nodes, while three-field<br />

lymphadenectomy also <strong>in</strong>volves cervical lymph node clearance (3 rd field). The extent of<br />

two-field lymphadenectomy is specified <strong>in</strong> relation to the extent of the mediast<strong>in</strong>al part<br />

of the lymphadenectomy. Standard two-field lymphadenectomy <strong>in</strong>volves the clearance of<br />

lymph nodes from the car<strong>in</strong>a down to the diaphragm. In extended two-field<br />

lymphadenectomy, clearance is expanded to the right paratracheal and recurrent nerve,<br />

while <strong>in</strong> total two-field lymphadenectomy clearance is further extended to the left<br />

recurrent nerve [96]. Extensive two-field lymphadenectomy should be standard dur<strong>in</strong>g<br />

oesophagectomy [47]. Three-field lymphadenectomy is considered strictly<br />

<strong>in</strong>vestigational, especially <strong>in</strong> distal third adenocarc<strong>in</strong>oma [60]. Indeed, data from<br />

Western studies <strong>with</strong> three-field lymphadenectomy are scarce and are based on<br />

relatively small sample sizes [60, 97].<br />

Many studies have shown a relationship between patient outcomes (e.g. 30-day<br />

mortality) and surgeon or hospital volume [47, 56, 98, 99]. Recently, a meta-analysis of<br />

13 studies calculated that the experience of 20 oesophagectomies per year is needed to<br />

significantly reduce postoperative mortality [100]. Therefore, oesophageal cancer<br />

surgery should be carried out <strong>in</strong> high volume specialist surgical units by surgeons <strong>with</strong><br />

experience <strong>in</strong> oesophageal and GOJ cancer [47]. Recently, it was shown that specialty<br />

tra<strong>in</strong><strong>in</strong>g <strong>in</strong> thoracic surgery has an <strong>in</strong>dependent association <strong>with</strong> lower mortality after<br />

oesophageal resection (adjusted mortality 12.4% vs. 16.5%; p = 0.01), although specialty<br />

tra<strong>in</strong><strong>in</strong>g appeared to be less important than hospital and surgeon volume [101]. Of<br />

course, specialty tra<strong>in</strong><strong>in</strong>g and high volume are closely related.<br />

Recommendations<br />

26. Surgical resection is considered standard treatment for patients <strong>with</strong><br />

resectable oesophageal cancer (1A recommendation).<br />

27. Surgery for oesophageal cancer should be aimed at achiev<strong>in</strong>g an R0<br />

resection, and should be considered preferentially through a<br />

transthoracic en bloc resection (1A recommendation).<br />

28. Extensive two-field lymphadenectomy should be standard dur<strong>in</strong>g<br />

oesophagectomy to improve stag<strong>in</strong>g, local disease control and<br />

potentially cure rate (1C recommendation).<br />

29. Three-field lymphadenectomy is strictly <strong>in</strong>vestigational (2C<br />

recommendation).<br />

30. Oesophageal cancer surgery should be carried out <strong>in</strong> high volume<br />

specialist surgical units by surgeons <strong>with</strong> experience and/or specialist<br />

tra<strong>in</strong><strong>in</strong>g <strong>in</strong> oesophageal and gastro-oesophageal junction cancer (1C<br />

recommendation).


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 27<br />

4.6.3 Adjuvant treatment<br />

CCO provided an excellent overview of the literature concern<strong>in</strong>g adjuvant treatment<br />

for resectable oesophageal cancer [84]. Three RCTs of postoperative chemotherapy (all<br />

cisplat<strong>in</strong>-based regimens) and surgery vs. surgery alone were identified. Pooled results<br />

of 2 RCTs [102, 103] showed no significant difference <strong>in</strong> the risk of death at 3 years (RR<br />

0.94; 95%CI 0.74 – 1.18; p=0.60) [84]. A third RCT also found no survival benefit for<br />

postoperative chemotherapy, but 3-year survival was not reported [104].<br />

CCO identified 5 RCTs of postoperative radiotherapy and surgery vs. surgery alone<br />

[84]. Meta-analysis of these trials showed no significant difference <strong>in</strong> the risk of death<br />

<strong>with</strong> postoperative radiotherapy and surgery at one year compared <strong>with</strong> surgery alone<br />

(RR 1.23; 95%CI 0.95 – 1.59; p=0.11). The rate of local recurrence <strong>with</strong> radiotherapy<br />

was lower <strong>in</strong> two RCTs, but this benefit was achieved at the expense of <strong>in</strong>creased<br />

morbidity [105, 106]. The most recent RCT found significantly improved 5-year survival<br />

rates <strong>with</strong> adjuvant radiotherapy for stage III patients (35% vs. 13%; p=0.0027) [107].<br />

However, this trial was hampered by some important methodological drawbacks (no<br />

<strong>in</strong>formed consent, no <strong>in</strong>tention-to-treat analysis).<br />

No RCTs on postoperative comb<strong>in</strong>ed chemoradiotherapy vs. surgery alone were<br />

identified by CCO [84]. A search for RCTs published s<strong>in</strong>ce the CCO report also did<br />

not identify new evidence.<br />

There is also no available evidence on whether adjuvant therapy is of real benefit <strong>in</strong> R1<br />

or R2 resection.<br />

Recommendations<br />

31. Postoperative adjuvant chemotherapy is not recommended for<br />

patients <strong>with</strong> oesophageal cancer (2A recommendation).<br />

32. Postoperative adjuvant radiotherapy is not recommended for<br />

patients <strong>with</strong> oesophageal cancer (2A recommendation).<br />

33. Postoperative adjuvant chemoradiotherapy is not recommended for<br />

patients <strong>with</strong> oesophageal cancer (expert op<strong>in</strong>ion).<br />

4.6.4 Non-surgical treatment <strong>with</strong> curative <strong>in</strong>tent<br />

Def<strong>in</strong>itive CRT should be considered <strong>in</strong> patients <strong>with</strong> oesophageal cancer who have<br />

locally advanced disease and are unfit for surgery, or <strong>in</strong> patients who decl<strong>in</strong>e surgery<br />

[47, 108]. Recently, a moderate-quality eastern RCT found no difference <strong>in</strong> survival at 2<br />

years between primary CRT (and salvage surgery) and primary surgery <strong>in</strong> patients <strong>with</strong><br />

potentially resectable oesophageal cancer (see above) [94]. Bedenne et al. also found no<br />

difference <strong>in</strong> survival at 2 years between CRT followed by surgery and CRT alone <strong>in</strong><br />

patients <strong>with</strong> operable oesophageal cancer that responded to CRT (see above) [93].<br />

Once aga<strong>in</strong>, the need for an adequate method to predict treatment response is<br />

highlighted.<br />

Def<strong>in</strong>itive CRT should also be considered for patients <strong>with</strong> cervical oesophageal cancer,<br />

for whom it offers the benefit of preserv<strong>in</strong>g the larynx [56]. However, no RCTs are<br />

available compar<strong>in</strong>g surgery to CRT for these patients.<br />

Concomitant CRT is recommended over radiotherapy alone. In their systematic review,<br />

CCO <strong>in</strong>cluded 8 RCTs of concomitant CRT vs. radiotherapy alone, of which 7 RCTs<br />

had 1-year mortality rates available. Pooled analysis showed an odds ratio (OR) of 0.61<br />

(95%CI 0.44 – 0.84; p


28 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Sequential radiotherapy and chemotherapy is not recommended as standard practice<br />

[108]. CCO identified 5 RCTs of sequential radiotherapy and chemotherapy vs.<br />

radiotherapy alone. Pooled analysis revealed no significant difference <strong>in</strong> survival between<br />

the treatment groups at one year.<br />

Patients should be made aware of the <strong>in</strong>creased acute toxicity associated <strong>with</strong> the use<br />

of concomitant CRT. Therefore, the decision to use concomitant CRT should only be<br />

made after careful consideration of the potential risks, benefits, and the patient’s general<br />

condition [108].<br />

In patients <strong>with</strong> locally advanced <strong>in</strong>fracar<strong>in</strong>al oesophageal tumours that do not respond<br />

to primary CRT, curative salvage surgery rema<strong>in</strong>s an option provided the patient is fit<br />

for surgery [111]. Piessen et al. found a R0 resection rate of 62.2% <strong>in</strong> these patients.<br />

Overall survival was significantly higher <strong>in</strong> the R0 resection group compared <strong>with</strong> the<br />

<strong>in</strong>complete resection group (18.4 vs. 8.6 months, p


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 29<br />

Therefore, <strong>in</strong> patients <strong>with</strong> locally advanced or metastatic cancer of the oesophagus,<br />

chemotherapy or CRT are treatment options that should be discussed <strong>in</strong> the MDT [56,<br />

114].<br />

The use of external-beam radiotherapy for the palliation of dysphagia and pa<strong>in</strong> is<br />

supported by observational studies only [47]. In 1 RCT, endolum<strong>in</strong>al brachytherapy<br />

resulted <strong>in</strong> better dysphagia control than stent placement <strong>in</strong> patients who lived longer<br />

than 140 days [47]. Therefore, it should be considered <strong>in</strong> patients <strong>with</strong> dysphagia from<br />

oesophageal cancer <strong>with</strong> the perspective of a more prolonged survival.<br />

Patients <strong>with</strong> oesophageal cancer should have access to a specialist (outpatient and/or<br />

<strong>in</strong>patient) palliative care team, <strong>in</strong> particular <strong>in</strong> relation to comfort and symptom control,<br />

and quality of life [47]. This team clearly should <strong>in</strong>volve the general practitioner, who<br />

should have a coord<strong>in</strong>at<strong>in</strong>g role <strong>in</strong> the organisation of the palliative home care.<br />

Evidence specific to control of symptoms, such as pa<strong>in</strong>, anorexia and bleed<strong>in</strong>g, <strong>in</strong> these<br />

cancers is limited. Some evidence exists for the use of celiac axis plexus block for the<br />

palliation of pa<strong>in</strong> and for the use of corticosteroids <strong>in</strong> patients <strong>with</strong> oesophageal cancer<br />

who are anorexic or who have symptoms of bowel obstruction [47]. Nutritional<br />

support <strong>in</strong> the palliative sett<strong>in</strong>g should take <strong>in</strong>to account the comfort of the patient, <strong>in</strong><br />

that quality of life is much more important than the long-term effects [56].<br />

Limited evidence (com<strong>in</strong>g from a secondary analysis of 1 RCT) is also available for<br />

psychotherapeutic support dur<strong>in</strong>g hospital stay [115].<br />

Recommendations<br />

35. Control of obstruction caused by oesophageal cancer should be<br />

obta<strong>in</strong>ed <strong>with</strong> stent placement or laser/ argon plasma coagulation<br />

(APC) therapy, depend<strong>in</strong>g on the local availability and expertise (1A<br />

recommendation).<br />

36. Partially covered self-expand<strong>in</strong>g metal stents or plastic expandable<br />

stents are the best options for palliation of dysphagia caused by<br />

oesophageal cancer (1B recommendation).<br />

37. Laser therapy, argon plasma coagulation (APC) therapy or restent<strong>in</strong>g<br />

should be considered for control of tumour <strong>in</strong>growth or<br />

overgrowth <strong>in</strong> stented patients (1C recommendation).<br />

38. The use of oesophageal dilatation alone should be avoided (2C<br />

recommendation).<br />

39. Oesophagectomy (transthoracic or transhiatal) should not be<br />

performed <strong>with</strong> palliative <strong>in</strong>tent <strong>in</strong> patients <strong>with</strong> oesophageal cancer<br />

(1C recommendation).<br />

40. Substernal bypass for oesophageal cancer should not be performed<br />

<strong>with</strong> palliative <strong>in</strong>tent (1C recommendation).<br />

41. In patients <strong>with</strong> locally advanced or metastatic cancer of the<br />

oesophagus, chemotherapy or chemoradiotherapy are treatment<br />

options that should be discussed <strong>in</strong> the multidiscipl<strong>in</strong>ary team (2A<br />

recommendation).<br />

42. Palliative external-beam radiotherapy or endolum<strong>in</strong>al brachytherapy<br />

should be considered <strong>in</strong> patients <strong>with</strong> dysphagia from oesophageal<br />

cancer and <strong>with</strong> the perspective of a more prolonged survival (2C<br />

recommendation).<br />

43. Patients <strong>with</strong> oesophageal cancer should have access to a specialist<br />

palliative care team, <strong>in</strong> particular <strong>in</strong> relation to comfort and<br />

symptom control, nutrition and quality of life (1C recommendation).


30 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

4.8 FOLLOW-UP<br />

In patients that underwent treatment for oesophageal cancer, follow-up is needed to<br />

detect recurrent disease (<strong>in</strong> case of curative treatment), progressive disease, symptoms<br />

that warrant treatment, or nutritional problems. However, evidence about the duration,<br />

frequency and type of follow-up is unavailable [56]. Therefore, published guidel<strong>in</strong>es on<br />

the follow-up of patients <strong>with</strong> oesophageal cancer are always consensus-based.<br />

A physical exam<strong>in</strong>ation is recommended every three months, and should give special<br />

attention to nutritional status, weight loss, fatigue, etc. Vitam<strong>in</strong> B12 evaluation can be<br />

done, but deficiency after oesophagectomy occurs less frequently than after total<br />

gastrectomy. A CT scan of the chest and abdomen is recommended every six months <strong>in</strong><br />

the first year and afterwards annually until the fifth year. However, it should be stressed<br />

that no evidence is available to support regular imag<strong>in</strong>g <strong>in</strong> the follow-up of these<br />

patients [47].<br />

After endoscopic treatment of dysplastic lesions or mucosal cancer, endoscopic followup<br />

is similar to that of patients <strong>with</strong> Barrett’s oesophagus [56]: three months after<br />

endoscopic treatment, then every six months <strong>in</strong> the first two years, and then annually.<br />

Recommendations<br />

44. It is recommended that the follow-up of patients treated for<br />

oesophageal cancer <strong>in</strong>cludes a physical exam<strong>in</strong>ation every three<br />

months, and a CT scan every six months <strong>in</strong> the first year and<br />

afterwards annually until the fifth year (expert op<strong>in</strong>ion).<br />

45. Patients treated <strong>with</strong> endoscopic mucosal resection (EMR) should<br />

have a follow-up endoscopy after three months, then every six<br />

months <strong>in</strong> the first two years, and then annually (expert op<strong>in</strong>ion).<br />

4.9 RECURRENT DISEASE<br />

In a small number of patients, recurrent disease can be successfully treated <strong>with</strong> curative<br />

<strong>in</strong>tent [116-118]. In patients <strong>with</strong> a localised lymph node recurrence [119-121] or a<br />

localised anastomotic recurrence [122], a comb<strong>in</strong>ed chemoradiotherapy regimen or<br />

occasionally redo-surgery can be considered after discussion by the MDT. Patients who<br />

develop a solitary lung or liver metastasis can also be considered for resection. The<br />

evidence for these treatments, however, is weak and com<strong>in</strong>g from small observational<br />

studies only.<br />

When patients are confronted <strong>with</strong> a local recurrence or a new tumour after EMR for a<br />

mucosal cancer, treatment options <strong>in</strong>clude local treatment or multimodality treatment<br />

[123]. These options should be discussed <strong>in</strong> the MDT. Aga<strong>in</strong>, only small observational<br />

studies are available to support these treatments.<br />

Recommendations<br />

46. In patients <strong>with</strong> recurrent oesophageal cancer, treatment options<br />

should be discussed <strong>in</strong> the multidiscipl<strong>in</strong>ary team (expert op<strong>in</strong>ion).<br />

47. In patients <strong>with</strong> a local recurrence or new tumour after endoscopic<br />

mucosal resection (EMR), treatment options, <strong>in</strong>clud<strong>in</strong>g local<br />

treatment, should be discussed <strong>in</strong> the multidiscipl<strong>in</strong>ary team (expert<br />

op<strong>in</strong>ion).


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 31<br />

5 FINAL RECOMMENDATIONS ON GASTRIC<br />

CANCER<br />

5.1 FLOWCHART


32 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

5.2 DIAGNOSIS<br />

As for oesophageal cancer, <strong>in</strong> addition to a history and cl<strong>in</strong>ical exam<strong>in</strong>ation, flexible<br />

upper GI endoscopy <strong>with</strong> at least biopsies of all suspicious lesions is recommended as<br />

the diagnostic procedure of choice <strong>in</strong> patients <strong>with</strong> suspected gastric cancer [47]. The<br />

same alarm symptoms are associated <strong>with</strong> gastric cancer and should lead to a referral<br />

for early endoscopy and biopsies: dysphagia, recurrent vomit<strong>in</strong>g, anorexia, weight loss<br />

and gastro<strong>in</strong>test<strong>in</strong>al blood loss (see above) [47, 49].<br />

In a prospective trial <strong>in</strong>volv<strong>in</strong>g 202 consecutive patients (<strong>in</strong>clud<strong>in</strong>g 47 <strong>with</strong> oesophageal<br />

or gastric carc<strong>in</strong>omas), seven biopsy and cytology specimens were found to yield the<br />

correct diagnosis <strong>in</strong> all patients [124]. However, <strong>in</strong> patients <strong>with</strong> large gastric folds<br />

(suggestive of l<strong>in</strong>itis plastica or gastric lymphoma), at least 10 biopsies are<br />

recommended, together <strong>with</strong> an additional technique, such as large forceps biopsy.<br />

As <strong>in</strong> patients at high risk for oesophageal cancer, patients at high risk for develop<strong>in</strong>g<br />

gastric cancer (Table 9) may also benefit from screen<strong>in</strong>g and follow-up <strong>with</strong> HRE and/or<br />

chromoendoscopy [47], although the support<strong>in</strong>g evidence is scarce. No recent RCTs<br />

were identified. Ma<strong>in</strong>ly Japanese observational studies have shown that<br />

chromoendosocpy <strong>in</strong>creases the rate of superficial and “early” lesions detection [125].<br />

Patients <strong>with</strong> a familial history of gastric cancer should be referred to a centre for<br />

genetic counsell<strong>in</strong>g.<br />

No evidence was found on the most appropriate frequency of surveillance. Frequency of<br />

surveillance endoscopy depends on specific risk factors, e.g. every 3-5 years <strong>in</strong> patients<br />

<strong>with</strong> autoimmune chronic gastritis, annual follow-up for patients <strong>with</strong> low-grade<br />

dysplasia <strong>in</strong> atrophic gastritis, etc.<br />

Table 9. Features associated <strong>with</strong> high risk for develop<strong>in</strong>g gastric cancer.<br />

Histological features:<br />

• Dysplasia<br />

• Gastric atrophy <strong>with</strong> or <strong>with</strong>out <strong>in</strong>test<strong>in</strong>al metaplasia<br />

• Adenomatous polyps<br />

Patient’s history:<br />

• Previous gastric cancer<br />

• History of partial gastrectomy<br />

• Familial history of gastric cancer<br />

Cl<strong>in</strong>ical features:<br />

• Refractory gastric ulcer<br />

• Pernicious anaemia<br />

In patients <strong>with</strong> suspected gastric cancer or at high risk for develop<strong>in</strong>g gastric cancer, H.<br />

pylori test<strong>in</strong>g should be systematically done on histology and ideally <strong>with</strong> a second test<br />

(rapid urease test, urea breath test, culture or serology) [126-128]. In case of a negative<br />

histology and negative additional test<strong>in</strong>g, a positive serological test suggests the<br />

presence of an unrecognised H. pylori <strong>in</strong>fection, and additional <strong>in</strong>vestigations to confirm<br />

whether the serological test is false positive or reflects active or recent <strong>in</strong>fection should<br />

be carried out [128].


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 33<br />

Recommendations<br />

1. Patients present<strong>in</strong>g <strong>with</strong> any of the follow<strong>in</strong>g alarm symptoms <strong>with</strong><strong>in</strong><br />

the cl<strong>in</strong>ical context of potential gastric pathology should be referred<br />

for early endoscopy and biopsies: dysphagia, recurrent vomit<strong>in</strong>g,<br />

anorexia, weight loss, gastro<strong>in</strong>test<strong>in</strong>al blood loss (1C<br />

recommendation).<br />

2. Flexible upper gastro<strong>in</strong>test<strong>in</strong>al endoscopy <strong>with</strong> at least biopsies of all<br />

suspicious lesions is recommended as the diagnostic procedure of<br />

choice <strong>in</strong> patients <strong>with</strong> suspected gastric cancer (1C<br />

recommendation).<br />

3. High-resolution endoscopy and chromoendoscopy may be of value <strong>in</strong><br />

screen<strong>in</strong>g and follow-up of high-risk patients (2C recommendation).<br />

4. H. pylori test<strong>in</strong>g should be systematically done on histology and<br />

ideally <strong>with</strong> a second test. Serology should be considered if gastric<br />

sampl<strong>in</strong>g rema<strong>in</strong>s negative (2C recommendation).<br />

5.3 WORK-UP OF DYSPLASTIC LESIONS<br />

Patients diagnosed <strong>with</strong> high-grade dysplasia should have careful endoscopic and<br />

pathological assessment [47]. Pathologists should follow a classification for report<strong>in</strong>g<br />

dysplasia that the multidiscipl<strong>in</strong>ary team is familiar <strong>with</strong>. An overview of classifications is<br />

already provided <strong>in</strong> chapter 3.2.4. Where therapeutic <strong>in</strong>tervention is contemplated on<br />

the basis of high-grade dysplasia, the diagnosis should be validated by a second<br />

pathologist experienced <strong>in</strong> this area because of the significant <strong>in</strong>ter- and <strong>in</strong>tra-observer<br />

variation [47]. Further biopsies or EMR should be done if there is uncerta<strong>in</strong>ty. Biopsies<br />

should also be reviewed at a multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong> access to the cl<strong>in</strong>ical<br />

<strong>in</strong>formation.<br />

F<strong>in</strong>ally, as the diagnosis, treatment and follow-up of patients <strong>with</strong> gastric high-grade<br />

dysplastic lesions is very specific and new techniques rapidly become available, these<br />

patients should be referred to centres or network reference centres <strong>with</strong> the<br />

appropriate endoscopic and surgical expertise and facilities [47].<br />

Recommendations<br />

5. Patients confirmed <strong>with</strong> high-grade dysplasia should have subsequent<br />

careful endoscopic and pathological assessment (1C<br />

recommendation).<br />

6. Pathologists should follow a classification for report<strong>in</strong>g dysplasia that<br />

the multidiscipl<strong>in</strong>ary team is familiar <strong>with</strong> (1C recommendation).<br />

7. Where therapeutic <strong>in</strong>tervention is contemplated on the basis of<br />

high-grade dysplasia, the diagnosis should be validated by a second<br />

pathologist experienced <strong>in</strong> this area. Further biopsies should be done<br />

if there is uncerta<strong>in</strong>ty (1C recommendation).<br />

8. Biopsies should be reviewed at a multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong><br />

access to the cl<strong>in</strong>ical <strong>in</strong>formation (expert op<strong>in</strong>ion).<br />

9. Patients <strong>with</strong> high-grade dysplasia should be referred to centres or<br />

network reference centres <strong>with</strong> the appropriate endoscopic and<br />

surgical expertise and facilities (1C recommendation).


34 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

5.4 STAGING<br />

As for patients <strong>with</strong> oesophageal cancer, the ma<strong>in</strong> contribution of CT scan to the<br />

stag<strong>in</strong>g of gastric cancer is the detection of distant metastases [47, 129]. If metastatic<br />

disease is detected <strong>with</strong> CT scan, curative treatment is excluded and additional stag<strong>in</strong>g<br />

<strong>with</strong> EUS is unnecessary. A recent systematic review identified five observational studies<br />

exam<strong>in</strong><strong>in</strong>g the diagnostic accuracy of multidetector row CT (MDCT) for the T stag<strong>in</strong>g<br />

of gastric tumours [130]. It was concluded that both EUS and MDCT achieved similar<br />

results <strong>in</strong> terms of diagnostic accuracy <strong>in</strong> T stag<strong>in</strong>g and <strong>in</strong> assess<strong>in</strong>g serosal <strong>in</strong>volvement.<br />

Only one of the five <strong>in</strong>cluded studies made a direct comparison between EUS and<br />

MDCT [130]. A more recent prospective study of MDCT (<strong>with</strong> multiplanar reformation<br />

images) found an overall diagnostic accuracy of 89% for T stag<strong>in</strong>g and of 78% for N<br />

stag<strong>in</strong>g [131].<br />

Kwee et al. identified 23 studies exam<strong>in</strong><strong>in</strong>g the diagnostic accuracy of EUS for the T<br />

stag<strong>in</strong>g of gastric tumours [130]. Diagnostic accuracy varied between 65% and 92%.<br />

Sensitivity and specificity for assess<strong>in</strong>g serosal <strong>in</strong>volvement varied between 78% and<br />

100% and between 68% and 100%, respectively.<br />

PET(/CT) is not rout<strong>in</strong>ely recommended <strong>in</strong> the stag<strong>in</strong>g of gastric cancer. However, it<br />

can be considered <strong>in</strong> locally advanced mass-form<strong>in</strong>g tumours (<strong>in</strong>test<strong>in</strong>al type a ) <strong>in</strong> a<br />

curative sett<strong>in</strong>g [134].<br />

Kwee et al. identified 3 observational studies exam<strong>in</strong><strong>in</strong>g the diagnostic accuracy of MRI<br />

for the T stag<strong>in</strong>g of gastric tumours [130]. Diagnostic accuracy varied between 71% and<br />

83%. Sensitivity and specificity for assess<strong>in</strong>g serosal <strong>in</strong>volvement varied between 90%<br />

and 93% and between 94% and 100%, respectively. Overall, there is no anatomical area<br />

where MRI is superior to CT. Therefore, MRI should be reserved for those patients<br />

who cannot undergo CT. It can also be used for additional <strong>in</strong>vestigation follow<strong>in</strong>g CT<br />

and/or EUS [47].<br />

Laparoscopy can be considered <strong>in</strong> patients <strong>with</strong> gastric tumours be<strong>in</strong>g considered for<br />

surgery where full thickness gastric wall <strong>in</strong>volvement is suspected [47]. There are<br />

<strong>in</strong>sufficient data to confirm benefit for laparoscopic ultrasound [47, 79].<br />

Sent<strong>in</strong>el lymph node mapp<strong>in</strong>g has been the subject of several observational studies [135-<br />

139]. Overall sensitivity ranged from 71 – 89% [136-138]. In patients <strong>with</strong> a pT1<br />

tumour, sensitivity ranged from 88 – 100% [136, 138].<br />

a Gastric carc<strong>in</strong>omas can be classified accord<strong>in</strong>g to different classification systems (Lauren [132],<br />

Goseki [133], WHO, etc.) based on different criteria. The Lauren classification comb<strong>in</strong>es<br />

etiopathological, cl<strong>in</strong>ical and pathological (macro- and microscopic) features [132]. The most<br />

common type, the <strong>in</strong>test<strong>in</strong>al type, generates a tumoral mass and has conv<strong>in</strong>c<strong>in</strong>gly been<br />

demonstrated to arise through a Helicobacter pylori <strong>in</strong>fection, gastritis, <strong>in</strong>test<strong>in</strong>al metaplasia,<br />

dysplasia, carc<strong>in</strong>oma sequence. The less common type, the signet r<strong>in</strong>g cell carc<strong>in</strong>oma, often<br />

presents <strong>with</strong> a characteristic macroscopic appearance called “l<strong>in</strong>itis plastica” and will often be<br />

negative on PET.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 35<br />

Recommendations<br />

10. In patients <strong>with</strong> gastric cancer, CT scan of the chest and abdomen<br />

<strong>with</strong> IV contrast and gastric distension <strong>with</strong> oral contrast or water<br />

should be performed rout<strong>in</strong>ely. The liver should at least be imaged <strong>in</strong><br />

the arterial and portal venous phase (1C recommendation).<br />

11. Endoscopic ultrasonography <strong>with</strong> or <strong>with</strong>out f<strong>in</strong>e-needle aspiration<br />

cytology can be considered <strong>in</strong> patients to be treated <strong>with</strong> curative<br />

<strong>in</strong>tent based on cl<strong>in</strong>ical presentation and/or CT (1C<br />

recommendation).<br />

12. The follow<strong>in</strong>g exam<strong>in</strong>ations can be considered for specific <strong>in</strong>dications<br />

(as expla<strong>in</strong>ed <strong>in</strong> the text above): PET scan, Magnetic Resonance<br />

Imag<strong>in</strong>g, laparoscopy (1C recommendation).<br />

5.5 TREATMENT OF MUCOSAL CANCER<br />

Treatment options for patients <strong>with</strong> mucosal cancer should be discussed at a<br />

multidiscipl<strong>in</strong>ary meet<strong>in</strong>g, tak<strong>in</strong>g <strong>in</strong>to account cl<strong>in</strong>ical and histological <strong>in</strong>formation.<br />

Superficial gastric cancer limited to the mucosa can be treated <strong>with</strong> endoscopic mucosal<br />

resection (EMR), tak<strong>in</strong>g <strong>in</strong>to account the stage, size, histological type and differentiation<br />

grade (Table 10) [47]. Large Japanese series of hundreds of patients treated<br />

endoscopically <strong>with</strong> EMR or ESD showed the advantages and success rates of<br />

endoscopic management for mucosal cancer [140-143]. However, a recent Cochrane<br />

review failed to identify RCTs of early gastric cancer patients <strong>in</strong>volv<strong>in</strong>g a treatment arm<br />

of EMR and a comparison arm of gastrectomy [144]. Therefore, the potential harms<br />

(<strong>in</strong>complete resection, recurrence, perforation) and benefits (quality of life, utility <strong>in</strong><br />

patients unfit for surgery) of EMR must be weighed aga<strong>in</strong>st the harms/benefits of surgical<br />

treatment.<br />

En-bloc resection – allow<strong>in</strong>g better pathology stag<strong>in</strong>g of deep and lateral marg<strong>in</strong>s –<br />

should be aimed at dur<strong>in</strong>g mucosectomy <strong>with</strong> the appropriate technique (EMR for<br />

lesions less than 12 mm and ESD for larger lesions) [145].<br />

Mucosal ablative techniques, such as photodynamic therapy (PDT), laser or argon<br />

plasma coagulation (APC), cannot be recommended as a curative option.<br />

Table 10. Criteria associated <strong>with</strong> low risk of lymph node metastasis <strong>in</strong><br />

patients <strong>with</strong> mucosal gastric cancer [47, 144] (based on Japanese<br />

Classification of Gastric Carc<strong>in</strong>oma [146]).<br />

• Lesion < 2 cm <strong>in</strong> size <strong>in</strong> elevated types<br />

• Lesion < 1 cm <strong>in</strong> size <strong>in</strong> depressed types<br />

• Well or moderately differentiated histology<br />

• No macroscopic ulceration<br />

• Invasion limited to mucosa and certa<strong>in</strong>ly no deeper than the superficial<br />

submucosa<br />

• No residual <strong>in</strong>vasive disease after EMR


36 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Recommendations<br />

13. Biopsies should be reviewed by an experienced pathologist <strong>in</strong> this<br />

area and discussed at a multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong> access to the<br />

cl<strong>in</strong>ical <strong>in</strong>formation (expert op<strong>in</strong>ion).<br />

14. Superficial gastric cancer limited to the mucosa can be treated <strong>with</strong><br />

endoscopic mucosal resection (EMR), tak<strong>in</strong>g <strong>in</strong>to account the stage,<br />

size, histological type and differentiation grade (2C<br />

recommendation).<br />

15. Mucosal ablative techniques, such as photodynamic therapy (PDT),<br />

laser or argon plasma coagulation (APC), cannot be recommended<br />

as a curative option (expert op<strong>in</strong>ion).<br />

5.6 TREATMENT OF CANCER BEYOND THE MUCOSA<br />

5.6.1 Neoadjuvant treatment<br />

In a recent Cochrane review, 4 RCTs were identified compar<strong>in</strong>g neoadjuvant<br />

chemotherapy and surgery to surgery alone, <strong>in</strong>clud<strong>in</strong>g 2 Japanese trials us<strong>in</strong>g oral<br />

fluoropyrimid<strong>in</strong>es [147]. Meta-analysis of these 4 trials showed no statistically significant<br />

difference <strong>in</strong> mortality between the two treatment groups (OR 1.05; 95%CI 0.73 – 1.50;<br />

p = 0.29). The MAGIC trial, which was excluded from the Cochrane review because it<br />

also <strong>in</strong>cluded postoperative chemotherapy, found a higher likelihood of overall survival<br />

<strong>in</strong> patients receiv<strong>in</strong>g perioperative chemotherapy (ECF) as compared to surgery alone<br />

(HR 0.75; 95%CI 0.60 – 0.93; p = 0.009) [148]. However, the trial received some<br />

criticism because of a lack of standardisation of surgery, the absence of separate data<br />

for gastric cancer, and some methodological shortcom<strong>in</strong>gs (e.g. lack of stag<strong>in</strong>g accuracy,<br />

no bl<strong>in</strong>d<strong>in</strong>g).<br />

One abstract was found present<strong>in</strong>g the results of an RCT compar<strong>in</strong>g preoperative<br />

chemotherapy (5-FU/cisplat<strong>in</strong>) to surgery alone <strong>in</strong> adenocarc<strong>in</strong>oma of stomach and<br />

lower oesophagus [149]. HR of death was 0.69 (95%CI 0.50 – 0.95; p=0.02) <strong>with</strong> 3 and<br />

5- year overall survival of 35% and 24% respectively <strong>in</strong> the surgery alone group vs. 48%<br />

and 38% respectively <strong>in</strong> the neoadjuvant chemotherapy group.<br />

CCO identified three RCTs compar<strong>in</strong>g preoperative radiotherapy and surgery to<br />

surgery alone [150]. However, <strong>in</strong>consistent results were found, mak<strong>in</strong>g it difficult to<br />

draw unambiguous conclusions. Three additional RCTs were identified compar<strong>in</strong>g<br />

neoadjuvant immunotherapy and surgery to surgery alone [150]. No significant survival<br />

benefit was found. A recent small trial of neoadjuvant immunotherapy <strong>with</strong> <strong>in</strong>terleuk<strong>in</strong>-2<br />

found a trend towards better overall and disease-free survival <strong>in</strong> the neoadjuvant<br />

treatment group [151].<br />

As for oesophageal cancer, s<strong>in</strong>ce the use of neoadjuvant treatment for patients <strong>with</strong><br />

gastric cancer still is controversial, it is recommended to prospectively register cl<strong>in</strong>ical<br />

outcomes and adverse events of the comb<strong>in</strong>ed treatment [92].<br />

Recommendations<br />

16. Neoadjuvant treatment is not rout<strong>in</strong>ely <strong>in</strong>dicated for patients <strong>with</strong><br />

gastric cancer, but is an option to be discussed dur<strong>in</strong>g a<br />

multidiscipl<strong>in</strong>ary meet<strong>in</strong>g (2A recommendation).<br />

17. Prospective registration of cl<strong>in</strong>ical outcomes and adverse events of<br />

comb<strong>in</strong>ed treatment is recommended (expert op<strong>in</strong>ion).


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 37<br />

5.6.2 Surgical treatment<br />

Surgical treatment rema<strong>in</strong>s the standard treatment for patients <strong>with</strong> resectable gastric<br />

cancer, and should aim at achiev<strong>in</strong>g an R0 resection [47, 129]. For distal tumours (lower<br />

third, antrum) a partial gastrectomy <strong>with</strong> spar<strong>in</strong>g of the proximal stomach is <strong>in</strong>dicated.<br />

Proximal tumours (upper two third) should be treated <strong>with</strong> a total gastrectomy [129].<br />

Figure 2 gives an overview of the extent of lymphadenectomy for subtotal gastrectomy<br />

<strong>in</strong> terms of dissection of lymph nodes. McCulloch et al. identified 2 RCTs compar<strong>in</strong>g<br />

limited (D1) versus extended (D2) node dissection (<strong>in</strong>clud<strong>in</strong>g splenectomy and distal<br />

pancreatectomy) dur<strong>in</strong>g gastrectomy for gastric cancer [152]. Meta-analysis did not<br />

reveal any survival benefit for extended lymph node dissection (RR 0.95; 95%CI 0.83 –<br />

1.09), but showed <strong>in</strong>creased postoperative mortality (RR 2.23; 95%CI 1.45 – 3.45).<br />

Figure 2. The extent of lymphadenectomy for subtotal gastrectomy <strong>in</strong> terms<br />

of dissection of lymph nodes (figure repr<strong>in</strong>ted from Roukos et al. [153].<br />

D1 requires the dissection of the first-tier nodes (N1, nodal stations no. 1-6). D2 <strong>in</strong>cludes the N1<br />

and second-tier nodes (N2, no. 7-11). D3 requires the dissection of N1, N2 and third-tier nodes<br />

(N3, no. 12-15). D4 requires the dissection of N1, N2, N3 and fourth-tier para-aortic nodes (N4,<br />

no. 16).<br />

S<strong>in</strong>ce this Cochrane review, numerous trials have been published exam<strong>in</strong><strong>in</strong>g the extent<br />

of lymph node dissection. An <strong>in</strong>terim analysis of the Italian Gastric Cancer Study Group<br />

(IGCSG) randomised surgical trial showed no statistically significant difference <strong>in</strong><br />

postoperative morbidity and mortality between D1 and D2 gastrectomy [154]. In this<br />

trial, only a m<strong>in</strong>ority of patients received a splenectomy (<strong>in</strong> case of cl<strong>in</strong>ical T > 1 on the<br />

greater curvature of the proximal and middle thirds of the stomach) or a distal<br />

pancreatectomy (suspicion of tumour <strong>in</strong>volvement). However, a subsequent<br />

multicentric phase II study by the same group evaluat<strong>in</strong>g pancreas-preserv<strong>in</strong>g D2<br />

gastrectomy suggested a survival benefit <strong>with</strong> an overall 5-year survival of 55% and a<br />

postoperative <strong>in</strong>-hospital mortality of 3.1% [155]. Hartgr<strong>in</strong>k et al. also compared D1<br />

<strong>with</strong> D2 lymph node dissection [156]. Postoperative morbidity (25% vs. 43%; p = 0.001)<br />

and mortality (4% vs. 10%; p = 0.004) were significantly higher <strong>in</strong> the D2 dissection<br />

group, but after 11 years no overall difference <strong>in</strong> survival was found (30% vs. 35%; p =<br />

0.53).


38 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

As compared to Degiuli et al. [154], the amount of splenectomies and distal<br />

pancreatectomies was higher <strong>in</strong> the Randomized <strong>Dutch</strong> Gastric Cancer Group Trial<br />

[156].<br />

Three trials recently compared standard D2 vs. extended D2 (D2+, i.e. D2 <strong>with</strong> paraaortic<br />

nodal dissection) lymph node dissection [157-159]. No statistically significant<br />

differences <strong>in</strong> postoperative morbidity and mortality were found <strong>in</strong> two trials [157, 158],<br />

while postoperative morbidity was higher <strong>in</strong> the extended D2 group <strong>in</strong> the third trial<br />

[159].<br />

Reconstruction after gastrectomy can be <strong>with</strong> or <strong>with</strong>out pouch formation and <strong>with</strong><br />

(Billroth I and II) or <strong>with</strong>out (Roux-en-Y, jejunal <strong>in</strong>terposition) ma<strong>in</strong>tenance of duodenal<br />

passage [47]. Evidence suggests that pouch procedures may be associated <strong>with</strong> a higher<br />

earlier weight ga<strong>in</strong> [160, 161] and some improvement <strong>in</strong> long-term quality of life [162].<br />

Several articles have compared laparoscopic surgery to conventional surgery for gastric<br />

cancer [163-166]. One SR identified 4 RCTs compar<strong>in</strong>g laparoscopy-assisted distal<br />

gastrectomy (LADG) to conventional open distal gastrectomy (CODG) [164]. LADG<br />

was found to be a longer procedure, but was associated <strong>with</strong> a lower postoperative<br />

morbidity. No difference was found <strong>in</strong> postoperative mortality. These results were<br />

confirmed by an RCT published subsequently [166]. Hayashi et al. also found a shorter<br />

operation time <strong>with</strong> CODG, but no differences <strong>in</strong> postoperative morbidity and<br />

mortality [163]. Huscher et al. found no differences <strong>in</strong> duration of surgery and<br />

postoperative morbidity and mortality between laparoscopic-assisted and open radical<br />

subtotal gastrectomy [165]. Overall, duration of analgesic adm<strong>in</strong>istration was shorter<br />

after laparoscopic surgery [163, 164, 166].<br />

As for oesophageal cancer, several studies have shown a relationship between patient<br />

outcomes (e.g. 30-day mortality) and surgeon or hospital volume for gastric cancer<br />

surgery [47, 98, 99]. However, this relationship is less profound than for oesophageal<br />

cancer surgery [98]. Nevertheless, based on these results, gastric cancer surgery should<br />

be carried out <strong>in</strong> high volume specialist surgical units by surgeons <strong>with</strong> experience <strong>in</strong><br />

gastric cancer [47]. Subspecialty tra<strong>in</strong><strong>in</strong>g is also associated <strong>with</strong> lower postoperative<br />

mortality, although this relationship is not statistically significant [167].<br />

Recommendations<br />

18. Surgical resection should be considered standard treatment for<br />

patients <strong>with</strong> resectable gastric cancer (1A recommendation).<br />

19. Surgery for gastric cancer should aim at achiev<strong>in</strong>g an R0 resection<br />

(1A recommendation).<br />

20. D2 lymphadenectomy (<strong>with</strong> a m<strong>in</strong>imum of 15 lymph nodes removed<br />

and exam<strong>in</strong>ed) should be standard dur<strong>in</strong>g gastrectomy to improve<br />

stag<strong>in</strong>g and local disease control (1B recommendation).<br />

21. Gastric cancer surgery should be carried out <strong>in</strong> high volume<br />

specialist surgical units by surgeons <strong>with</strong> experience and/or specialist<br />

tra<strong>in</strong><strong>in</strong>g <strong>in</strong> this area (1C recommendation).<br />

5.6.3 Adjuvant treatment<br />

Numerous RCTs and systematic reviews have been published compar<strong>in</strong>g postoperative<br />

adjuvant chemotherapy to surgery alone [47, 150]. Most meta-analyses found small<br />

statistically significant differences <strong>in</strong> survival favour<strong>in</strong>g adjuvant chemotherapy, although<br />

only a m<strong>in</strong>ority of the <strong>in</strong>cluded RCTs were of high quality. Subgroup analyses <strong>in</strong> one<br />

meta-analysis showed a trend towards a larger magnitude of the effect for trials <strong>in</strong> which<br />

at least two thirds of the patients had node-positive disease (OR 0.74; 95%CI 0.59 –<br />

0.95) [150]. Therefore, CCO concluded that adjuvant chemotherapy alone may be of<br />

benefit <strong>in</strong> particular for patients <strong>with</strong> lymph node metastases. However, this is not <strong>in</strong><br />

l<strong>in</strong>e <strong>with</strong> SIGN, that did not recommend postoperative chemotherapy outside a cl<strong>in</strong>ical<br />

trial [47].


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 39<br />

One meta-analysis of 4 Japanese RCTs compar<strong>in</strong>g adjuvant oral fluor<strong>in</strong>ated pyrimid<strong>in</strong>es<br />

to surgery alone found a survival benefit <strong>in</strong> favour of adjuvant chemotherapy (HR 0.73;<br />

95%CI 0.60 – 0.89; p = 0.002) [168]. Other trials failed to demonstrate a benefit <strong>in</strong><br />

favour of adjuvant chemotherapy [169-172]. The optimum chemotherapy regimen has<br />

also not yet been def<strong>in</strong>ed.<br />

CCO identified two RCTs compar<strong>in</strong>g adjuvant radiotherapy to surgery alone [150]. No<br />

differences <strong>in</strong> survival were found. However, addition of postoperative chemotherapy to<br />

radiotherapy may result <strong>in</strong> better outcomes. Three RCTs were identified by the CCO,<br />

of which one found no statistically significant difference <strong>in</strong> survival. The two other trials<br />

detected improved survival <strong>with</strong> postoperative CRT, but received considerable criticism<br />

[150]. For example, the surgery performed <strong>in</strong> the RCT of McDonald et al. was often not<br />

up to the desired standards [173].<br />

Several RCTs and meta-analyses compared adjuvant <strong>in</strong>traperitoneal chemotherapy to<br />

surgery alone [150, 174, 175]. A recent meta-analysis found a significant survival benefit<br />

<strong>in</strong> favour of hyperthermic <strong>in</strong>traoperative <strong>in</strong>traperitoneal chemotherapy <strong>with</strong> or <strong>with</strong>out<br />

early postoperative <strong>in</strong>traperitoneal chemotherapy [175]. However, <strong>in</strong> general survival<br />

results have been conflict<strong>in</strong>g, and some trials even reported harm from <strong>in</strong>traperitoneal<br />

therapy [150].<br />

CCO identified 9 RCTs compar<strong>in</strong>g adjuvant immunochemotherapy to surgery alone<br />

[150]; one meta-analysis on immunochemotherapy <strong>with</strong> polysaccharide K [176] and one<br />

RCT [177] were published subsequently. Overall, <strong>in</strong>consistent results were found,<br />

mak<strong>in</strong>g it difficult to draw def<strong>in</strong>ite conclusions.<br />

Recommendations<br />

22. Postoperative adjuvant chemotherapy is not recommended for<br />

patients <strong>with</strong> gastric cancer (2A recommendation).<br />

23. Postoperative adjuvant radiotherapy is not recommended for<br />

patients <strong>with</strong> gastric cancer (2A recommendation).<br />

24. Postoperative adjuvant chemoradiotherapy is not rout<strong>in</strong>ely<br />

recommended for patients <strong>with</strong> gastric cancer, but can be<br />

considered after discussion <strong>in</strong> the multidiscipl<strong>in</strong>ary team (2A<br />

recommendation).<br />

5.7 PALLIATIVE TREATMENT & METASTATIC DISEASE<br />

Palliative gastric surgery is limited to symptomatic stenoses, bleed<strong>in</strong>g tumours and<br />

perforation [129]. It should be avoided <strong>in</strong> patients who have dissem<strong>in</strong>ated peritoneal<br />

disease [47, 129]. In comparison to palliative gastrectomy, gastric bypass has a higher<br />

mortality [47]. However, reported mortality after laparoscopic gastrojejunostomy is<br />

lower than for open bypass.<br />

For patients <strong>with</strong> malignant gastric outlet obstruction, treatment options <strong>in</strong>clude<br />

endoscopic stent<strong>in</strong>g or surgical gastroenterostomy [178, 179]. Two systematic reviews,<br />

that ma<strong>in</strong>ly comprised observational studies, found a higher <strong>in</strong>itial cl<strong>in</strong>ical success after<br />

endoscopic stent<strong>in</strong>g. However, <strong>in</strong>consistent results were found for associated morbidity<br />

[178, 179]. No significant survival benefit was found. Endoscopic stent<strong>in</strong>g was associated<br />

<strong>with</strong> a shorter hospital stay.<br />

Several RCTs compared palliative chemotherapy to best supportive care for patients<br />

<strong>with</strong> advanced or metastatic gastric cancer [180, 181]. A clear survival benefit was found<br />

<strong>in</strong> favour of palliative chemotherapy. Above this, palliative chemotherapy was associated<br />

<strong>with</strong> a better quality of life [180]. Wagner et al. also found a significant survival benefit <strong>in</strong><br />

favour of comb<strong>in</strong>ation chemotherapy as compared to s<strong>in</strong>gle agent chemotherapy (HR<br />

0.83; 95%CI 0.74 – 0.93) [181]. Therefore, <strong>in</strong> patients <strong>with</strong> locally advanced or<br />

metastatic cancer of the stomach <strong>with</strong> good performance status comb<strong>in</strong>ation<br />

chemotherapy should be considered [47, 181].


40 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Patients <strong>with</strong> gastric cancer should have access to a specialist (outpatient and/or<br />

<strong>in</strong>patient) palliative care team, <strong>in</strong> particular <strong>in</strong> relation to comfort and symptom control,<br />

and quality of life [47]. This team clearly should <strong>in</strong>volve the general practitioner, who<br />

should have a coord<strong>in</strong>at<strong>in</strong>g role <strong>in</strong> the organisation of the palliative home care.<br />

However, as for oesophageal cancer, evidence specific to control of symptoms, such as<br />

pa<strong>in</strong>, anorexia and bleed<strong>in</strong>g, <strong>in</strong> these cancers is limited. Some evidence exists for the use<br />

of celiac axis plexus block for the palliation of pa<strong>in</strong> and for the use of corticosteroids <strong>in</strong><br />

patients <strong>with</strong> gastric cancer who are anorexic or who have symptoms of bowel<br />

obstruction [47]. Nutritional support <strong>in</strong> the palliative sett<strong>in</strong>g should take <strong>in</strong>to account<br />

the comfort of the patient, <strong>in</strong> that quality of life is much more important than the longterm<br />

effects.<br />

Limited evidence (com<strong>in</strong>g from a secondary analysis of 1 RCT) is also available for<br />

psychotherapeutic support dur<strong>in</strong>g hospital stay [115].<br />

Recommendations<br />

5.8 FOLLOW-UP<br />

25. Palliative gastric surgery is limited to symptomatic stenoses,<br />

bleed<strong>in</strong>g tumours and perforation (2C recommendation).<br />

26. For patients <strong>with</strong> malignant gastric outlet obstruction, treatment<br />

options <strong>in</strong>clude endoscopic stent<strong>in</strong>g or surgical gastroenterostomy<br />

(2C recommendation).<br />

27. In patients <strong>with</strong> locally advanced or metastatic cancer of the<br />

stomach <strong>with</strong> good performance status comb<strong>in</strong>ation chemotherapy<br />

should be considered (1A recommendation).<br />

28. Patients <strong>with</strong> gastric cancer should have access to a specialist<br />

palliative care team, <strong>in</strong> particular <strong>in</strong> relation to comfort and<br />

symptom control, and quality of life (1C recommendation).<br />

In patients that underwent treatment for gastric cancer, follow-up is needed to detect<br />

recurrent disease (<strong>in</strong> case of curative treatment), progressive disease, symptoms that<br />

warrant treatment, or nutritional problems result<strong>in</strong>g from total gastrectomy (e.g. iron<br />

and vitam<strong>in</strong> deficiency, maldigestion <strong>with</strong> steathorroea, etc). However, evidence about<br />

the duration, frequency and type of follow-up is lack<strong>in</strong>g [182] (one retrospective study<br />

was published after our literature search [183]). Therefore, these recommendations are<br />

made <strong>in</strong> l<strong>in</strong>e <strong>with</strong> those for oesophageal cancer.<br />

A physical exam<strong>in</strong>ation is recommended every three months, and should give special<br />

attention to nutritional status, weight loss, fatigue, etc. Above this, a blood analysis<br />

(rout<strong>in</strong>e chemistry and haematology, completed <strong>with</strong> serum ferrit<strong>in</strong>e and vitam<strong>in</strong> B12<br />

dos<strong>in</strong>g <strong>in</strong> case of anaemia) is recommended. Vitam<strong>in</strong> B12 deficiency is a frequent<br />

phenomenon after total gastrectomy, and should be adequately treated. CT scan of the<br />

abdomen is recommended every six months <strong>in</strong> the first year and afterwards annually<br />

until the fifth year. However, it should be stressed that no evidence is available to<br />

support regular imag<strong>in</strong>g <strong>in</strong> the follow-up of these patients [47]. For patients that<br />

underwent partial gastrectomy, lifelong endoscopic surveillance is <strong>in</strong>dicated because of<br />

the high risk of develop<strong>in</strong>g gastric stump carc<strong>in</strong>oma [184].<br />

Patients treated <strong>with</strong> endoscopic mucosal resection (EMR) should have strict<br />

endoscopic surveillance [144] <strong>with</strong> a follow-up endoscopy after three months, then<br />

every six months <strong>in</strong> the first two years, and then annually.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 41<br />

Recommendations<br />

29. It is recommended that the follow-up of patients treated for gastric<br />

cancer <strong>in</strong>cludes a physical exam<strong>in</strong>ation and blood analysis every<br />

three months, and a CT scan every six months <strong>in</strong> the first year and<br />

afterwards annually until the fifth year (expert op<strong>in</strong>ion).<br />

30. Patients treated <strong>with</strong> endoscopic mucosal resection (EMR) should<br />

have a follow-up endoscopy after three months, then every six<br />

months <strong>in</strong> the first two years, and then annually (expert op<strong>in</strong>ion).<br />

5.9 RECURRENT DISEASE<br />

In a small number of patients, recurrent disease can be successfully treated <strong>with</strong> curative<br />

<strong>in</strong>tent. In patients <strong>with</strong> cancer of the gastric stump after distal gastrectomy, R0 resection<br />

can be achieved <strong>in</strong> a relatively high number of patients [185, 186]. Patients who develop<br />

a solitary lung or liver metastasis can also be considered for resection [187, 188]. When<br />

patients are confronted <strong>with</strong> a local recurrence after EMR for a mucosal cancer, local<br />

treatment can be reconsidered [145, 189]. The evidence for these treatments, however,<br />

is weak and com<strong>in</strong>g from small observational studies only. Treatment options for<br />

recurrent disease should therefore be discussed <strong>in</strong> the MDT.<br />

Recommendations<br />

31. In patients <strong>with</strong> recurrent gastric cancer, treatment options should<br />

be discussed <strong>in</strong> the multidiscipl<strong>in</strong>ary team (expert op<strong>in</strong>ion).<br />

32. In patients <strong>with</strong> a local recurrence or new tumour after endoscopic<br />

mucosal resection (EMR), treatment options, <strong>in</strong>clud<strong>in</strong>g local<br />

treatment, should be discussed <strong>in</strong> the multidiscipl<strong>in</strong>ary team (expert<br />

op<strong>in</strong>ion).<br />

5.10 TREATMENT OF GASTRIC LYMPHOMA<br />

5.10.1 Introduction<br />

Primary gastric lymphoma is a rare tumour, account<strong>in</strong>g for less than 5% of primary<br />

gastric neoplasms. However, it is the most common extranodal lymphoma, represent<strong>in</strong>g<br />

4-20% of all extranodal lymphomas [190]. Helicobacter pylori <strong>in</strong>fection,<br />

immunosuppression after solid-organ transplantation, celiac disease, <strong>in</strong>flammatory bowel<br />

disease, and human immunodeficiency virus (HIV) <strong>in</strong>fection are known risk factors for<br />

GI lymphoma. A significant proportion of gastric lymphomas is of low-grade histology<br />

and arises from mucosa-associated lymphoid tissue (MALT) [191].<br />

Accord<strong>in</strong>g to the most recent WHO classification, the term ‘MALT lymphoma’ should<br />

only be applied to tumours previously def<strong>in</strong>ed as low-grade MALT lymphomas<br />

composed mostly by small cells. High-grade lymphomas are known as large B-cell<br />

lymphoma [192]. Patients <strong>with</strong> low-grade B-cell lymphoma or MALT lymphoma have a<br />

better prognosis than patients <strong>with</strong> diffuse large B-cell lymphoma (DLBCL) [193].<br />

Tumours of T-cell orig<strong>in</strong> are rare [190]. Patients <strong>with</strong> gastric lymphomas are currently<br />

staged us<strong>in</strong>g the Ann Arbor stag<strong>in</strong>g system <strong>with</strong> the Cotswold modification. This has<br />

largely replaced the older International Workshop stag<strong>in</strong>g system [194].<br />

5.10.2 Diagnosis and stag<strong>in</strong>g<br />

There is no consensus regard<strong>in</strong>g the diagnostic work-up and stag<strong>in</strong>g of primary gastric<br />

lymphomas. Evidence is scarce and exclusively based on observational studies. As for all<br />

patients <strong>with</strong> suspected gastric cancer, upper GI endoscopy is <strong>in</strong>dicated <strong>in</strong> case of<br />

suspected gastric lymphoma.


42 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Dedicated endoscopic-bioptic techniques are recommended, <strong>in</strong>clud<strong>in</strong>g a m<strong>in</strong>imum of 8 –<br />

12 biopsies from visible lesions, mapp<strong>in</strong>g of macroscopically normal-appear<strong>in</strong>g areas, and<br />

repeated exam<strong>in</strong>ations <strong>in</strong> the <strong>in</strong>dividual case. Biopsies should be preserved <strong>in</strong> such a way<br />

to allow molecular diagnostic <strong>in</strong>vestigation [195, 196].<br />

Several classifications of lymphomas are available [197, 198]. It is recommended to<br />

diagnose and classify gastric lymphomas accord<strong>in</strong>g to the most recent appropriate<br />

classification.<br />

In patients <strong>with</strong> histologically confirmed gastric lymphoma, endoscopic ultrasonography<br />

is <strong>in</strong>dicated [199, 200]. EUS-guided FNA of suspicious LN is not recommended. For<br />

other stag<strong>in</strong>g procedures, such as CT scan or PET scan, even less evidence is available<br />

[201-203]. Therefore, for patients <strong>with</strong> low-grade MALT lymphoma no further stag<strong>in</strong>g<br />

procedures are recommended, unless otherwise required for differential diagnostic<br />

reasons.<br />

Recommendations<br />

5.10.3 Treatment<br />

1. In patients <strong>with</strong> suspected gastric lymphoma, subtle endoscopicbioptic<br />

techniques are needed, <strong>in</strong>clud<strong>in</strong>g a m<strong>in</strong>imum of 8–12<br />

biopsies from visible lesions, mapp<strong>in</strong>g of macroscopically normalappear<strong>in</strong>g<br />

areas, and repeated exam<strong>in</strong>ations <strong>in</strong> the <strong>in</strong>dividual<br />

case. Biopsies should be preserved <strong>in</strong> such a way to allow<br />

molecular diagnostic <strong>in</strong>vestigation (expert op<strong>in</strong>ion).<br />

2. Lymphomas should be diagnosed and classified accord<strong>in</strong>g to the<br />

most recent appropriate classification (expert op<strong>in</strong>ion).<br />

3. In patients <strong>with</strong> histologically confirmed gastric lymphoma,<br />

endoscopic ultrasonography is <strong>in</strong>dicated. Endoscopic ultrasoundguided<br />

f<strong>in</strong>e needle aspiration of suspicious lymph nodes is not<br />

recommended (1C recommendation).<br />

4. For patients <strong>with</strong> low-grade MALT lymphoma no further stag<strong>in</strong>g<br />

procedures are recommended, unless otherwise required for<br />

differential diagnostic reasons (expert op<strong>in</strong>ion).<br />

In the past, surgery was thought to be necessary for the diagnosis, stag<strong>in</strong>g, and<br />

treatment of low-grade gastric lymphomas. However, as mentioned above, most<br />

patients now can be adequately diagnosed <strong>with</strong> biopsy and appropriately staged <strong>with</strong><br />

non-<strong>in</strong>vasive studies. Furthermore, gastrectomy can be associated <strong>with</strong> significant<br />

morbidity [194]. Numerous observational studies have demonstrated successful<br />

treatment of gastric MALT lymphomas <strong>with</strong> H. pylori eradication alone [204-208].<br />

Complete remission rates ranged from 62 – 95%. No trials were found compar<strong>in</strong>g H.<br />

pylori eradication to surgical treatment.<br />

In one RCT, 245 previously untreated patients <strong>with</strong> gastric low-grade MALT lymphoma<br />

(stage IE and IIE) were randomised to surgery, radiotherapy or chemotherapy [209]. No<br />

statistically significant differences were found <strong>in</strong> overall survival, but event-free survival<br />

was significantly higher <strong>in</strong> the group randomised to chemotherapy.<br />

Because the time from H. pylori eradication to lymphoma regression ranges from 1-28<br />

months, close follow-up is required <strong>with</strong> upper GI endoscopy <strong>in</strong>clud<strong>in</strong>g biopsy [128,<br />

194]. No evidence is available on the most appropriate duration of this follow-up, but<br />

an annual control dur<strong>in</strong>g the first three years seems rational. Patients beyond stage I<br />

have a significantly decreased response to H. pylori eradication therapy [206, 207].<br />

These patients, and patients <strong>with</strong>out tumour regression after successful H. pylori<br />

eradication, should be referred to a specialized haematology centre [205].


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 43<br />

Recommendations<br />

5. H. pylori eradication is the treatment of first choice for H. pylori<br />

<strong>in</strong>fected patients <strong>with</strong> stage I low grade gastric MALT lymphoma<br />

(1C recommendation).<br />

6. In patients <strong>with</strong> low-grade MALT lymphoma treated <strong>with</strong><br />

antibiotic eradication, close follow-up <strong>with</strong> upper GI endoscopy<br />

and biopsies is required, <strong>in</strong>clud<strong>in</strong>g evaluation of H. pylori<br />

eradication <strong>with</strong><strong>in</strong> 3 months (expert op<strong>in</strong>ion).<br />

7. Patients <strong>with</strong> successful H. pylori eradication but <strong>with</strong>out tumour<br />

regression after 1 year or <strong>with</strong> tumour progression should be<br />

referred to a specialized haematology centre (expert op<strong>in</strong>ion).<br />

5.11 TREATMENT OF GASTROINTESTINAL STROMAL<br />

TUMOURS<br />

5.11.1 Introduction<br />

Gastro<strong>in</strong>test<strong>in</strong>al stromal tumours (GIST) are relatively rare tumours, represent<strong>in</strong>g less<br />

than 1% of all tumours of the gastro<strong>in</strong>test<strong>in</strong>al tract. They are most common <strong>in</strong> the<br />

stomach (39% to 70%) and the small <strong>in</strong>test<strong>in</strong>e (20% to 32%), whereas the colon, rectum<br />

and oesophagus are affected <strong>in</strong> less than 15% of cases [210]. GISTs predom<strong>in</strong>antly occur<br />

<strong>in</strong> <strong>in</strong>dividuals over 40 years of age, <strong>with</strong> the majority occurr<strong>in</strong>g between the ages of 55<br />

to 65. Estimates of <strong>in</strong>cidence vary widely from 4 to 14 cases per million populations<br />

[211].<br />

Present<strong>in</strong>g features of these tumours <strong>in</strong>clude abdom<strong>in</strong>al discomfort or pa<strong>in</strong>, a feel<strong>in</strong>g of<br />

abdom<strong>in</strong>al fullness and the presence of a palpable mass. However, many people <strong>with</strong><br />

GISTs are asymptomatic dur<strong>in</strong>g early stages of the disease until tumours reach a large<br />

size, at which time the tumours rupture and bleed or obstruct the gastro<strong>in</strong>test<strong>in</strong>al tract<br />

[211].<br />

Overall, literature on GIST is relatively scarce and of low quality. Most studies are<br />

observational studies or case series <strong>with</strong>out an adequate control. Therefore, the<br />

recommendations presented below often have a low level of evidence or are based on<br />

expert op<strong>in</strong>ion.<br />

5.11.2 Diagnosis and stag<strong>in</strong>g<br />

In patients <strong>with</strong> cl<strong>in</strong>ical suspicion of GIST, endoscopic ultrasonography (EUS) and EUSguided<br />

FNA are recommended for differential diagnostic reasons and to confirm the<br />

presence of positive lymph nodes or malignancy <strong>in</strong> adjacent organs [212]. F<strong>in</strong>d<strong>in</strong>gs on<br />

EUS of a diameter > 3 cm, irregular extralum<strong>in</strong>al border, cystic spaces, echogenic foci,<br />

and adjacent malignant-appear<strong>in</strong>g lymph nodes are features that can suggest malignancy.<br />

Unfortunately, EUS f<strong>in</strong>d<strong>in</strong>gs do not accurately predict the malignant potential of a small<br />

GIST (< 3 cm).<br />

Most GISTs (80 – 100%) express the tyros<strong>in</strong>e k<strong>in</strong>ase growth factor receptor c-KIT,<br />

which is detected by immunosta<strong>in</strong><strong>in</strong>g <strong>with</strong> the antibody for the cell-surface marker<br />

CD117. This immunohistochemical test (performed on tissue) is now considered to be<br />

an appropriate diagnostic marker for the diagnosis of GIST [212].<br />

In patients <strong>with</strong> a (suspected) GIST tumour, a CT abdomen is recommended if<br />

treatment is considered [213-217]. However, the evidence support<strong>in</strong>g this<br />

recommendation is observational and of low quality.


44 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Recommendations<br />

1. In patients <strong>with</strong> cl<strong>in</strong>ical suspicion of GIST, endoscopic<br />

ultrasonography and endoscopic ultrasound-guided f<strong>in</strong>e needle<br />

aspiration can be recommended for differential diagnostic<br />

reasons and to confirm the presence of positive lymph nodes or<br />

malignancy <strong>in</strong> adjacent organs (2C recommendation).<br />

2. In patients <strong>with</strong> a (suspected) GIST, immunohistochemical<br />

test<strong>in</strong>g of CD117 is recommended (1C recommendation).<br />

3. In patients <strong>with</strong> a (suspected) GIST tumour, a CT abdomen is<br />

recommended if treatment is considered (expert op<strong>in</strong>ion).<br />

5.11.3 Treatment<br />

5.11.3.1 Non-metastatic resectable GIST<br />

Discrepancies were found <strong>in</strong> the literature on the strategy to predict malignant<br />

behaviour of GISTs. AGA proposed criteria <strong>in</strong>clud<strong>in</strong>g size (>3 cm), mitotic count on<br />

histology, and several EUS criteria (size; irregularity of the extralum<strong>in</strong>al border; the<br />

presence of cystic spaces, echogenic foci, and heterogeneity) [212]. The National<br />

Institutes of Health proposed a strategy for predict<strong>in</strong>g malignant behaviour of GISTs<br />

based on size (>2cm, 2-5cm, 5-10 cm, or >10) and mitotic count on histology (10 per 50 high-power field), <strong>with</strong> the understand<strong>in</strong>g that no GIST can be def<strong>in</strong>ed as<br />

benign on the basis of currently available diagnostic test<strong>in</strong>g [218].<br />

Debate also exists as to whether the potential morbidity and mortality associated <strong>with</strong><br />

surgical resection are acceptable for remov<strong>in</strong>g a lesion <strong>with</strong> low potential for<br />

malignancy. Given the overall lack of evidence to guide management, it is recommended<br />

to perform surgical resection <strong>in</strong> patients that are fit for surgery and <strong>with</strong> a histologically<br />

confirmed non-metastatic GIST or <strong>with</strong> a gastric tumour of >5 cm that is highly<br />

suspicious of a GIST (and <strong>with</strong>out evidence for metastatic disease) [211, 212, 219]. For<br />

lesions of 2 – 5 cm that are highly suspicious of GIST and <strong>with</strong>out evidence for<br />

metastatic disease, the choice between surveillance and surgical resection should be<br />

discussed at the MDT, tak<strong>in</strong>g <strong>in</strong>to consideration histology, size, marg<strong>in</strong>s, age,<br />

comorbidities and fitness for surgery. In patients <strong>with</strong> a gastric tumour of


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 45<br />

Recommendations<br />

4. In patients <strong>with</strong> a histologically confirmed non-metastatic GIST<br />

and who are fit for surgery, resectional surgery is <strong>in</strong>dicated<br />

(expert op<strong>in</strong>ion).<br />

5. In patients <strong>with</strong> a gastric tumour of >5 cm that is highly suspicious<br />

of a GIST, <strong>with</strong>out evidence for metastatic disease, and who are<br />

fit for surgery, resectional surgery is <strong>in</strong>dicated (expert op<strong>in</strong>ion).<br />

6. In patients <strong>with</strong> a gastric tumour of 2-5 cm that is highly<br />

suspicious of a GIST, <strong>with</strong>out evidence for metastatic disease, and<br />

who are fit for surgery, the choice between surveillance and<br />

resectional surgery should be discussed at the multidiscipl<strong>in</strong>ary<br />

team (expert op<strong>in</strong>ion).<br />

7. In patients <strong>with</strong> a gastric tumour of


46 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

6 IMPLEMENTATION AND UPDATING OF THE<br />

UPPER GI CANCER GUIDELINE<br />

6.1 IMPLEMENTATION<br />

The implementation of the present guidel<strong>in</strong>e will be led by the College of Oncology. An<br />

onl<strong>in</strong>e implementation tool – similar to the tools accompany<strong>in</strong>g previous guidel<strong>in</strong>es<br />

(https://portal.health.fgov.be/portal/page?_pageid=56,10338450&_dad=portal&_schema=<br />

PORTAL) – will be developed. The tool will be based on the general algorithms of this<br />

guidel<strong>in</strong>e.<br />

6.2 QUALITY CONTROL<br />

The implementation of the guidel<strong>in</strong>e has to be evaluated <strong>with</strong> appropriate quality<br />

control criteria. These criteria should at least assess the follow<strong>in</strong>g items of the general<br />

algorithms:<br />

• diagnostic work-up<br />

• stag<strong>in</strong>g<br />

• treatment accord<strong>in</strong>g to stage<br />

• follow-up<br />

• multidiscipl<strong>in</strong>ary approach<br />

For each of these steps, quality <strong>in</strong>dicators should be developed, which should be<br />

preferentially based on the recommendations <strong>with</strong> a high level of evidence. Additionally,<br />

a literature search for exist<strong>in</strong>g quality <strong>in</strong>dicators should be done. However, a preassessment<br />

of the literature (Medl<strong>in</strong>e and the National Quality Measures Clear<strong>in</strong>ghouse,<br />

http://www.qualitymeasures.ahrq.gov/) only identified a limited number of exisit<strong>in</strong>g<br />

quality <strong>in</strong>dicators (Table 11).<br />

Table 11. Exist<strong>in</strong>g quality <strong>in</strong>dicators for oesophageal and gastric cancer,<br />

identified through pre-assessment of the literature.<br />

Quality Indicator Source<br />

Oesophageal resection mortality rate Agency for Healthcare Research and Quality<br />

Oesophageal resection: volume Agency for Healthcare Research and Quality<br />

6.3 GUIDELINE UPDATE<br />

In view of the chang<strong>in</strong>g evidence, and based on a pre-assessment of the literature, this<br />

guidel<strong>in</strong>e should be fully updated <strong>in</strong> 5 years. In the meanwhile, when important evidence<br />

becomes available, this will be mentioned on the website of the College of Oncology<br />

(https://portal.health.fgov.be/portal/page?_pageid=56,10338450&_dad=portal&_schema=<br />

PORTAL).


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 47<br />

7 REFERENCES<br />

1. Park<strong>in</strong>, D.M., et al., Global cancer statistics, 2002. CA Cancer J Cl<strong>in</strong>, 2005. 55(2): p. 74-108.<br />

2. Bollschweiler, E., et al., Demographic variations <strong>in</strong> the ris<strong>in</strong>g <strong>in</strong>cidence of esophageal adenocarc<strong>in</strong>oma<br />

<strong>in</strong> white males. Cancer, 2001. 92(3): p. 549-55.<br />

3. Crane, L.M., et al., Oesophageal cancer <strong>in</strong> The Netherlands: <strong>in</strong>creas<strong>in</strong>g <strong>in</strong>cidence and mortality but<br />

improv<strong>in</strong>g survival. Eur J Cancer, 2007. 43(9): p. 1445-51.<br />

4. Qiu, D. and S. Kaneko, Comparison of esophageal cancer mortality <strong>in</strong> five countries: France, Italy,<br />

Japan, UK and USA from the WHO mortality database (1960-2000). Jpn J Cl<strong>in</strong> Oncol, 2005. 35(9):<br />

p. 564-7.<br />

5. Vizca<strong>in</strong>o, A.P., et al., Time trends <strong>in</strong>cidence of both major histologic types of esophageal carc<strong>in</strong>omas<br />

<strong>in</strong> selected countries, 1973-1995. Int J Cancer, 2002. 99(6): p. 860-8.<br />

6. Crew, K.D. and A.I. Neugut, Epidemiology of gastric cancer. World J Gastroenterol, 2006. 12(3):<br />

p. 354-62.<br />

7. Wijnhoven, B.P.L., et al., Increased <strong>in</strong>cidence of adenocarc<strong>in</strong>omas at the gastro-oesophageal junction<br />

<strong>in</strong> <strong>Dutch</strong> males s<strong>in</strong>ce the 1990s. Eur J Gastroenterol Hepatol, 2002. 14(2): p. 115-22.<br />

8. Crane, S.J., et al., The chang<strong>in</strong>g <strong>in</strong>cidence of oesophageal and gastric adenocarc<strong>in</strong>oma by anatomic<br />

sub-site. Aliment Pharmacol Ther, 2007. 25(4): p. 447-53.<br />

9. Peeters, M., et al., Nationale Richtlijnen van het College voor oncologie: A. Algemeen kader oncologish<br />

kwaliteitshandboek B. Wetenschappelijke basis voor kl<strong>in</strong>ische paden voor diagnose en behandel<strong>in</strong>g<br />

colorectale kanker en testiskanker, <strong>in</strong> <strong>KCE</strong> <strong>reports</strong>. 2006, Federaal Kenniscentrum voor de<br />

Gezondheidszorg (<strong>KCE</strong>): Brussel.<br />

10. Fervers, B., et al., Adaptation of cl<strong>in</strong>ical guidel<strong>in</strong>es: literature review and proposition for a framework<br />

and procedure. Int J Qual Health Care, 2006. 18(3): p. 167-76.<br />

11. Siewert, J.R. and H.J. Ste<strong>in</strong>, Classification of adenocarc<strong>in</strong>oma of the oesophagogastric junction. Br J<br />

Surg, 1998. 85(11): p. 1457-9.<br />

12. Spechler, S.J., et al., Adenocarc<strong>in</strong>oma of the esophago-gastric junction., <strong>in</strong> Pathology and Genetics of<br />

Tumours of the Digestive System., S.R. Hamilton and L.A. Aaltonen, Editors. 2000, IARC Press:<br />

Lyon, France. p. 31-36.<br />

13. (UICC), I.U.A.C., TNM classification of malignant tumours. 6th ed. ed. 2002, Berl<strong>in</strong>: Spr<strong>in</strong>ger-<br />

Verlag.<br />

14. Corley, D.A. and A. Kubo, Influence of site classification on cancer <strong>in</strong>cidence rates: an analysis of<br />

gastric cardia carc<strong>in</strong>omas. J Natl Cancer Inst, 2004. 96(18): p. 1383-7.<br />

15. Driessen, A., et al., Identical cytokerat<strong>in</strong> expression pattern CK7+/CK20- <strong>in</strong> esophageal and cardiac<br />

cancer: etiopathological and cl<strong>in</strong>ical implications. Mod Pathol, 2004. 17(1): p. 49-55.<br />

16. Driessen, A., et al., Are carc<strong>in</strong>omas of the cardia oesophageal or gastric adenocarc<strong>in</strong>omas? Eur J<br />

Cancer, 2003. 39(17): p. 2487-94.<br />

17. Sk<strong>in</strong>ner, D.B., et al., Barrett's esophagus. Comparison of benign and malignant cases. Ann Surg,<br />

1983. 198(4): p. 554-65.<br />

18. Paull, A., et al., The histologic spectrum of Barrett's esophagus. N Engl J Med, 1976. 295(9): p. 476-<br />

80.<br />

19. Naef, A.P., M. Savary, and L. Ozzello, Columnar-l<strong>in</strong>ed lower esophagus: an acquired lesion <strong>with</strong><br />

malignant predisposition. <strong>Report</strong> on 140 cases of Barrett's esophagus <strong>with</strong> 12 adenocarc<strong>in</strong>omas. J<br />

Thorac Cardiovasc Surg, 1975. 70(5): p. 826-35.<br />

20. Haggitt, R.C., et al., Adenocarc<strong>in</strong>oma complicat<strong>in</strong>g columnar epithelium-l<strong>in</strong>ed (Barrett's) esophagus.<br />

Am J Cl<strong>in</strong> Pathol, 1978. 70(1): p. 1-5.<br />

21. Reid, B.J., et al., Barrett's esophagus. Correlation between flow cytometry and histology <strong>in</strong> detection of<br />

patients at risk for adenocarc<strong>in</strong>oma. Gastroenterology, 1987. 93(1): p. 1-11.


48 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

22. Spechler, S.J. and R.K. Goyal, The columnar-l<strong>in</strong>ed esophagus, <strong>in</strong>test<strong>in</strong>al metaplasia, and Norman<br />

Barrett. Gastroenterology, 1996. 110(2): p. 614-21.<br />

23. Sampl<strong>in</strong>er, R.E., Practice guidel<strong>in</strong>es on the diagnosis, surveillance, and therapy of Barrett's esophagus.<br />

The Practice Parameters Committee of the American College of Gastroenterology. Am J<br />

Gastroenterol, 1998. 93(7): p. 1028-32.<br />

24. Sharma, P., et al., A critical review of the diagnosis and management of Barrett's esophagus: the AGA<br />

Chicago Workshop. Gastroenterology, 2004. 127(1): p. 310-30.<br />

25. British Society of Gastroenterology, Guidel<strong>in</strong>es for the diagnosis and management of Barrett’s<br />

columnar-l<strong>in</strong>ed oesophagus., B.S.o. Gastroenterology, Editor. 2005, British Society of<br />

Gastroenterology: London.<br />

26. Flejou, J.F. and M. Svrcek, Barrett's oesophagus--a pathologist's view. Histopathology, 2007. 50(1):<br />

p. 3-14.<br />

27. Takubo, K., et al., Double muscularis mucosae <strong>in</strong> Barrett's esophagus. Hum Pathol, 1991. 22(11): p.<br />

1158-61.<br />

28. Nigro, J.J., et al., Prevalence and location of nodal metastases <strong>in</strong> distal esophageal adenocarc<strong>in</strong>oma<br />

conf<strong>in</strong>ed to the wall: implications for therapy. J Thorac Cardiovasc Surg, 1999. 117(1): p. 16-23;<br />

discussion 23-5.<br />

29. Westerterp, M., et al., Outcome of surgical treatment for early adenocarc<strong>in</strong>oma of the esophagus or<br />

gastro-esophageal junction. Virchows Arch, 2005. 446(5): p. 497-504.<br />

30. Watanabe, H., J.R. Jass, and L.H. Sob<strong>in</strong>, Histological typ<strong>in</strong>g of oesophageal and gastric tumours. 2nd<br />

ed. ed. 1990, Berl<strong>in</strong>: Spr<strong>in</strong>ger-Verlag.<br />

31. Rubio, C.A., F.S. Liu, and H.Z. Zhao, Histological classification of <strong>in</strong>traepithelial neoplasias and<br />

micro<strong>in</strong>vasive squamous carc<strong>in</strong>oma of the esophagus. Am J Surg Pathol, 1989. 13(8): p. 685-90.<br />

32. Riddell, R.H., et al., Dysplasia <strong>in</strong> <strong>in</strong>flammatory bowel disease: standardized classification <strong>with</strong><br />

provisional cl<strong>in</strong>ical applications. Hum Pathol, 1983. 14(11): p. 931-68.<br />

33. Montgomery, E., Is there a way for pathologists to decrease <strong>in</strong>terobserver variability <strong>in</strong> the diagnosis<br />

of dysplasia? Arch Pathol Lab Med, 2005. 129(2): p. 174-6.<br />

34. Montgomery, E., et al., Reproducibility of the diagnosis of dysplasia <strong>in</strong> Barrett esophagus: a<br />

reaffirmation. Hum Pathol, 2001. 32(4): p. 368-78.<br />

35. Reid, B.J., et al., Observer variation <strong>in</strong> the diagnosis of dysplasia <strong>in</strong> Barrett's esophagus. Hum Pathol,<br />

1988. 19(2): p. 166-78.<br />

36. Schlemper, R.J., et al., International comparability of the pathological diagnosis for early cancer of the<br />

digestive tract: Munich meet<strong>in</strong>g. J Gastroenterol, 2000. 35 Suppl 12: p. 102-10.<br />

37. Schlemper, R.J., et al., Differences <strong>in</strong> diagnostic criteria for esophageal squamous cell carc<strong>in</strong>oma<br />

between Japanese and Western pathologists. Cancer, 2000. 88(5): p. 996-1006.<br />

38. Schlemper, R.J., et al., Differences <strong>in</strong> diagnostic criteria for gastric carc<strong>in</strong>oma between Japanese and<br />

western pathologists. Lancet, 1997. 349(9067): p. 1725-9.<br />

39. Antonioli, D.A. and H.H. Wang, Morphology of Barrett's esophagus and Barrett's-associated<br />

dysplasia and adenocarc<strong>in</strong>oma. Gastroenterol Cl<strong>in</strong> North Am, 1997. 26(3): p. 495-506.<br />

40. Haggitt, R.C., Barrett's esophagus, dysplasia, and adenocarc<strong>in</strong>oma. Hum Pathol, 1994. 25(10): p.<br />

982-93.<br />

41. Rugge, M., et al., Gastric dysplasia: the Padova <strong>in</strong>ternational classification. Am J Surg Pathol, 2000.<br />

24(2): p. 167-76.<br />

42. Schlemper, R.J., et al., The Vienna classification of gastro<strong>in</strong>test<strong>in</strong>al epithelial neoplasia. Gut, 2000.<br />

47(2): p. 251-5.<br />

43. Schlemper, R.J., Y. Kato, and M. Stolte, Diagnostic criteria for gastro<strong>in</strong>test<strong>in</strong>al carc<strong>in</strong>omas <strong>in</strong> Japan<br />

and Western countries: proposal for a new classification system of gastro<strong>in</strong>test<strong>in</strong>al epithelial neoplasia.<br />

J Gastroenterol Hepatol, 2000. 15 Suppl: p. G49-57.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 49<br />

44. Schlemper, R.J., Y. Kato, and M. Stolte, Review of histological classifications of gastro<strong>in</strong>test<strong>in</strong>al<br />

epithelial neoplasia: differences <strong>in</strong> diagnosis of early carc<strong>in</strong>omas between Japanese and Western<br />

pathologists. J Gastroenterol, 2001. 36(7): p. 445-56.<br />

45. Hamilton, S.R. and L.A. Aaltonen, Pathology and genetics of tumours of the digestive system., ed.<br />

S.R. Hamilton and L.A. Aaltonen. 2000, Lyon, France: IARC Press.<br />

46. Odze, R.D., Diagnosis and grad<strong>in</strong>g of dysplasia <strong>in</strong> Barrett's oesophagus. J Cl<strong>in</strong> Pathol, 2006. 59(10):<br />

p. 1029-38.<br />

47. Scottish Intercollegiate Guidel<strong>in</strong>es Network, Management of oesophageal and gastric cancer. A<br />

national cl<strong>in</strong>ical guidel<strong>in</strong>e., SIGN, Editor. 2006, SIGN: Ed<strong>in</strong>burgh.<br />

48. Wang, K.K., M. Wongkeesong, and N.S. Buttar, American Gastroenterological Association technical<br />

review on the role of the gastroenterologist <strong>in</strong> the management of esophageal carc<strong>in</strong>oma.<br />

Gastroenterology, 2005. 128(5): p. 1471-505.<br />

49. Bowrey, D.J., et al., Use of alarm symptoms to select dyspeptics for endoscopy causes patients <strong>with</strong><br />

curable esophagogastric cancer to be overlooked. Surgical Endoscopy, 2006. 20(11): p. 1725-8.<br />

50. Slee, G.R., S.M. Wagner, and F.S. McCullough, Odynophagia <strong>in</strong> patients <strong>with</strong> malignant disorders.<br />

Cancer, 1985. 55(12): p. 2877-9.<br />

51. Dubuc, J., et al., Endoscopic screen<strong>in</strong>g for esophageal squamous-cell carc<strong>in</strong>oma <strong>in</strong> high-risk patients: a<br />

prospective study conducted <strong>in</strong> 62 French endoscopy centers. Endoscopy, 2006. 38(7): p. 690-5.<br />

52. Borovicka, J., et al., Autofluorescence endoscopy <strong>in</strong> surveillance of Barrett's esophagus: a multicenter<br />

randomized trial on diagnostic efficacy. Endoscopy, 2006. 38(9): p. 867-72.<br />

53. Lim, C.H., et al., Randomized crossover study that used methylene blue or random 4-quadrant biopsy<br />

for the diagnosis of dysplasia <strong>in</strong> Barrett's esophagus.[see comment]. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy,<br />

2006. 64(2): p. 195-9.<br />

54. Kara, M.A., et al., High-resolution endoscopy plus chromoendoscopy or narrow-band imag<strong>in</strong>g <strong>in</strong><br />

Barrett's esophagus: a prospective randomized crossover study. Endoscopy, 2005. 37(10): p. 929-36.<br />

55. Corey, K.E., S.M. Schmitz, and N.J. Shaheen, Does a surgical antireflux procedure decrease the<br />

<strong>in</strong>cidence of esophageal adenocarc<strong>in</strong>oma <strong>in</strong> Barrett's esophagus? A meta-analysis.[see comment].<br />

American Journal of Gastroenterology, 2003. 98(11): p. 2390-4.<br />

56. Kwaliteits<strong>in</strong>stituut voor de Gezondheidszorg (CBO), Diagnostiek en behandel<strong>in</strong>g<br />

57.<br />

oesofaguscarc<strong>in</strong>oom., V.Z.C. B.V., Editor. 2005, CBO: Alphen aan den Rijn.<br />

Reid, B.J., et al., Optimiz<strong>in</strong>g endoscopic biopsy detection of early cancers <strong>in</strong> Barrett's high-grade<br />

dysplasia. Am J Gastroenterol, 2000. 95(11): p. 3089-96.<br />

58. Reid, B.J., et al., Predictors of progression to cancer <strong>in</strong> Barrett's esophagus: basel<strong>in</strong>e histology and flow<br />

cytometry identify low- and high-risk patient subsets. Am J Gastroenterol, 2000. 95(7): p. 1669-76.<br />

59. Lowe, V.J., et al., Comparison of positron emission tomography, computed tomography, and<br />

endoscopic ultrasound <strong>in</strong> the <strong>in</strong>itial stag<strong>in</strong>g of patients <strong>with</strong> esophageal cancer. Molecular Imag<strong>in</strong>g &<br />

Biology, 2005. 7(6): p. 422-30.<br />

60. Lerut, T., et al., Three-field lymphadenectomy for carc<strong>in</strong>oma of the esophagus and gastroesophageal<br />

junction <strong>in</strong> 174 R0 resections: impact on stag<strong>in</strong>g, disease-free survival, and outcome: a plea for<br />

adaptation of TNM classification <strong>in</strong> upper-half esophageal carc<strong>in</strong>oma. Ann Surg, 2004. 240(6): p.<br />

962-72; discussion 972-4.<br />

61. van Vliet, E.P.M., et al., Publication bias does not play a role <strong>in</strong> the report<strong>in</strong>g of the results of<br />

endoscopic ultrasound stag<strong>in</strong>g of upper gastro<strong>in</strong>test<strong>in</strong>al cancers. Endoscopy, 2007. 39(4): p. 325-32.<br />

62. Pfau, P.R., et al., Esophageal dilation for endosonographic evaluation of malignant esophageal<br />

strictures is safe and effective. Am J Gastroenterol, 2000. 95(10): p. 2813-5.<br />

63. Shimpi, R.A., et al., Stag<strong>in</strong>g of esophageal cancer by EUS: stag<strong>in</strong>g accuracy revisited. Gastro<strong>in</strong>test<br />

Endosc, 2007. 66(3): p. 475-82.<br />

64. Egan, J.V., et al., Esophageal dilation. Gastro<strong>in</strong>test Endosc, 2006. 63(6): p. 755-60.


50 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

65. Doldi, S.B., et al., Ultrasonographic evaluation of the cervical lymph nodes <strong>in</strong> preoperative stag<strong>in</strong>g of<br />

esophageal neoplasms. Abdom Imag<strong>in</strong>g, 1998. 23(3): p. 275-7.<br />

66. Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC), Recommandations<br />

pour la pratique cl<strong>in</strong>ique: Standards, Options et Recommandations 2003 pour l’utilisation de la<br />

tomographie par émission de positons au [18F]-FDG (TEP-FDG) en cancérologie (rapport <strong>in</strong>tégral). <strong>in</strong><br />

Standards, Options et Recommendations. 2003, FNCLCC: Paris.<br />

67. Cleemput, I., et al., HTA Positron Emission Tomography Imag<strong>in</strong>g <strong>in</strong> Belgium, <strong>in</strong> <strong>KCE</strong> <strong>reports</strong>. 2005,<br />

Belgian Health Care Knowledge Centre (<strong>KCE</strong>): Brussels.<br />

68. Choi, J.Y., et al., 18F-FDG PET <strong>in</strong> patients <strong>with</strong> esophageal squamous cell carc<strong>in</strong>oma undergo<strong>in</strong>g<br />

curative surgery: prognostic implications. Journal of Nuclear Medic<strong>in</strong>e, 2004. 45(11): p. 1843-50.<br />

69. Kneist, W., et al., Prospective evaluation of positron emission tomography <strong>in</strong> the preoperative stag<strong>in</strong>g<br />

of esophageal carc<strong>in</strong>oma. Archives of Surgery, 2004. 139(10): p. 1043-9.<br />

70. Leong, T., et al., A prospective study to evaluate the impact of FDG-PET on CT-based radiotherapy<br />

treatment plann<strong>in</strong>g for oesophageal cancer.[see comment]. Radiotherapy & Oncology, 2006. 78(3):<br />

p. 254-61.<br />

71. Moureau-Zabotto, L., et al., Impact of CT and 18F-deoxyglucose positron emission tomography<br />

image fusion for conformal radiotherapy <strong>in</strong> esophageal carc<strong>in</strong>oma. International Journal of Radiation<br />

Oncology, Biology, Physics, 2005. 63(2): p. 340-5.<br />

72. Ott, K., et al., Metabolic imag<strong>in</strong>g predicts response, survival, and recurrence <strong>in</strong> adenocarc<strong>in</strong>omas of<br />

the esophagogastric junction. Journal of Cl<strong>in</strong>ical Oncology, 2006. 24(29): p. 4692-8.<br />

73. Sihvo, E.I.T., et al., Adenocarc<strong>in</strong>oma of the esophagus and the esophagogastric junction: positron<br />

emission tomography improves stag<strong>in</strong>g and prediction of survival <strong>in</strong> distant but not <strong>in</strong> locoregional<br />

disease. Journal of Gastro<strong>in</strong>test<strong>in</strong>al Surgery, 2004. 8(8): p. 988-96.<br />

74. Song, S.Y., et al., FDG-PET <strong>in</strong> the prediction of pathologic response after neoadjuvant<br />

chemoradiotherapy <strong>in</strong> locally advanced, resectable esophageal cancer. International Journal of<br />

Radiation Oncology, Biology, Physics, 2005. 63(4): p. 1053-9.<br />

75. Yuan, S., et al., Additional value of PET/CT over PET <strong>in</strong> assessment of locoregional lymph nodes <strong>in</strong><br />

thoracic esophageal squamous cell cancer. Journal of Nuclear Medic<strong>in</strong>e, 2006. 47(8): p. 1255-9.<br />

76. Osugi, H., et al., Bronchoscopic ultrasonography for stag<strong>in</strong>g supracar<strong>in</strong>al esophageal squamous cell<br />

carc<strong>in</strong>oma: impact on outcome. World Journal of Surgery, 2003. 27(5): p. 590-4.<br />

77. Wakamatsu, T., et al., Usefulness of preoperative endobronchial ultrasound for airway <strong>in</strong>vasion<br />

around the trachea: esophageal cancer and thyroid cancer.[see comment]. Respiration, 2006. 73(5):<br />

p. 651-7.<br />

78. Imadahl, A., et al., [Is bronchoscopy a useful additional preoperative exam<strong>in</strong>ation <strong>in</strong> esophageal<br />

carc<strong>in</strong>oma?]. Langenbecks Arch Chir, 1990. 375(6): p. 326-9.<br />

79. Mortensen, M.B., et al., Comb<strong>in</strong>ed preoperative endoscopic and laparoscopic ultrasonography for<br />

prediction of R0 resection <strong>in</strong> upper gastro<strong>in</strong>test<strong>in</strong>al tract cancer. British Journal of Surgery, 2006.<br />

93(6): p. 720-5.<br />

80. Oka, S., et al., Advantage of endoscopic submucosal dissection compared <strong>with</strong> EMR for early gastric<br />

cancer. Gastro<strong>in</strong>test Endosc, 2006. 64(6): p. 877-83.<br />

81. Esaki, M., et al., Risk factors for local recurrence of superficial esophageal cancer after treatment by<br />

endoscopic mucosal resection. Endoscopy, 2007. 39(1): p. 41-5.<br />

82. Katada, C., et al., Local recurrence of squamous-cell carc<strong>in</strong>oma of the esophagus after EMR.<br />

Gastro<strong>in</strong>test Endosc, 2005. 61(2): p. 219-25.<br />

83. Overholt, B.F., et al., Photodynamic therapy <strong>with</strong> porfimer sodium for ablation of high-grade dysplasia<br />

<strong>in</strong> Barrett's esophagus: <strong>in</strong>ternational, partially bl<strong>in</strong>ded, randomized phase III trial.[see<br />

comment][erratum appears <strong>in</strong> Gastro<strong>in</strong>test Endosc. 2006 Feb;63(2):359]. Gastro<strong>in</strong>test<strong>in</strong>al<br />

Endoscopy, 2005. 62(4): p. 488-98.<br />

84. Malthaner, R.A., et al., Neoadjuvant or Adjuvant Therapy for Resectable Esophageal Cancer. Practice<br />

Guidel<strong>in</strong>e <strong>Report</strong> #2-11., CCO, Editor. 2005, CCO: Ottawa.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 51<br />

85. Arnott, S.J., et al., Preoperative radiotherapy for esophageal carc<strong>in</strong>oma.[update of Cochrane<br />

Database Syst Rev. 2000;(4):CD001799; PMID: 11034728]. Cochrane Database of Systematic<br />

Reviews, 2005(4): p. CD001799.<br />

86. Gebski, V., et al., Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy <strong>in</strong><br />

oesophageal carc<strong>in</strong>oma: a meta-analysis.[see comment]. Lancet Oncology, 2007. 8(3): p. 226-34.<br />

87. Malthaner, R.A., S. Coll<strong>in</strong>, and D. Fenlon, Preoperative chemotherapy for resectable thoracic<br />

esophageal cancer.[update of Cochrane Database Syst Rev. 2003;(4):CD001556; PMID: 14583936].<br />

Cochrane Database of Systematic Reviews, 2006. 3: p. CD001556.<br />

88. Graham, A.J., et al., Def<strong>in</strong><strong>in</strong>g the optimal treatment of locally advanced esophageal cancer: a<br />

systematic review and decision analysis.[see comment]. Annals of Thoracic Surgery, 2007. 83(4): p.<br />

1257-64.<br />

89. Natsugoe, S., et al., Randomized controlled study on preoperative chemoradiotherapy followed by<br />

surgery versus surgery alone for esophageal squamous cell cancer <strong>in</strong> a s<strong>in</strong>gle <strong>in</strong>stitution. Diseases of<br />

the Esophagus, 2006. 19(6): p. 468-72.<br />

90. Carstens, H., et al., A randomized trial of chemoradiotherapy versus surgery alone <strong>in</strong> patients <strong>with</strong><br />

resectable esophageal cancer. J Cl<strong>in</strong> Oncol (Meet<strong>in</strong>g Abstracts), 2007. 25(18_suppl): p. 4530-.<br />

91. Cerfolio, R.J., et al., The accuracy of endoscopic ultrasonography <strong>with</strong> f<strong>in</strong>e-needle aspiration,<br />

<strong>in</strong>tegrated positron emission tomography <strong>with</strong> computed tomography, and computed tomography <strong>in</strong><br />

restag<strong>in</strong>g patients <strong>with</strong> esophageal cancer after neoadjuvant chemoradiotherapy. Journal of Thoracic<br />

& Cardiovascular Surgery, 2005. 129(6): p. 1232-41.<br />

92. FOD Volksgezondheid Veiligheid van de voedselketen en Leefmilieu, K.B. 21 maart 2003.<br />

Kon<strong>in</strong>klijk besluit houdende vaststell<strong>in</strong>g van de normen waaraan het zorgprogramma voor oncologische<br />

basiszorg en het zorgprogramma voor oncologie moeten voldoen om te worden erkend. 2003: B.S. 25-<br />

04-2003.<br />

93. Bedenne, L., et al., Chemoradiation followed by surgery compared <strong>with</strong> chemoradiation alone <strong>in</strong><br />

squamous cancer of the esophagus: FFCD 9102.[see comment]. Journal of Cl<strong>in</strong>ical Oncology, 2007.<br />

25(10): p. 1160-8.<br />

94. Chiu, P.W.Y., et al., Multicenter prospective randomized trial compar<strong>in</strong>g standard esophagectomy<br />

<strong>with</strong> chemoradiotherapy for treatment of squamous esophageal cancer: early results from the Ch<strong>in</strong>ese<br />

University Research Group for Esophageal Cancer (CURE). Journal of Gastro<strong>in</strong>test<strong>in</strong>al Surgery,<br />

2005. 9(6): p. 794-802.<br />

95. Hulscher, J.B., et al., Extended transthoracic resection compared <strong>with</strong> limited transhiatal resection for<br />

adenocarc<strong>in</strong>oma of the esophagus. N Engl J Med, 2002. 347(21): p. 1662-9.<br />

96. Fumagalli, U., H. Akiyama, and T.R. DeMeester, Resective surgery for cancer of the thoracic<br />

esophagus: results of a consensus conference held at the VIth World congress of the <strong>in</strong>ternational<br />

society for diseases of the esophagus. Dis Esophagus, 1996. 9: p. 30-38.<br />

97. Altorki, N., et al., Three-field lymph node dissection for squamous cell and adenocarc<strong>in</strong>oma of the<br />

esophagus. Ann Surg, 2002. 236(2): p. 177-83.<br />

98. Halm, E.A., C. Lee, and M.R. Chass<strong>in</strong>, Is volume related to outcome <strong>in</strong> health care? A systematic<br />

review and methodologic critique of the literature. Ann Intern Med, 2002. 137(6): p. 511-20.<br />

99. Killeen, S.D., et al., Provider volume and outcomes for oncological procedures. Br J Surg, 2005. 92(4):<br />

p. 389-402.<br />

100. Metzger, R., et al., High volume centers for esophagectomy: what is the number needed to achieve<br />

low postoperative mortality? Diseases of the Esophagus, 2004. 17(4): p. 310-4.<br />

101. Dimick, J.B., et al., Specialty tra<strong>in</strong><strong>in</strong>g and mortality after esophageal cancer resection. Ann Thorac<br />

Surg, 2005. 80(1): p. 282-6.<br />

102. Pouliquen, X., et al., 5-Fluorouracil and cisplat<strong>in</strong> therapy after palliative surgical resection of<br />

squamous cell carc<strong>in</strong>oma of the esophagus. A multicenter randomized trial. French Associations for<br />

Surgical Research. Ann Surg, 1996. 223(2): p. 127-33.


52 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

103. Ando, N., et al., A randomized trial of surgery <strong>with</strong> and <strong>with</strong>out chemotherapy for localized squamous<br />

carc<strong>in</strong>oma of the thoracic esophagus: the Japan Cl<strong>in</strong>ical Oncology Group Study. J Thorac Cardiovasc<br />

Surg, 1997. 114(2): p. 205-9.<br />

104. Ando, N., et al., Surgery plus chemotherapy compared <strong>with</strong> surgery alone for localized squamous cell<br />

carc<strong>in</strong>oma of the thoracic esophagus: a Japan Cl<strong>in</strong>ical Oncology Group Study--JCOG9204. Journal of<br />

Cl<strong>in</strong>ical Oncology, 2003. 21(24): p. 4592-6.<br />

105. Fok, M., et al., Postoperative radiotherapy for carc<strong>in</strong>oma of the esophagus: a prospective, randomized<br />

controlled study. Surgery, 1993. 113(2): p. 138-47.<br />

106. Teniere, P., et al., Postoperative radiation therapy does not <strong>in</strong>crease survival after curative resection<br />

for squamous cell carc<strong>in</strong>oma of the middle and lower esophagus as shown by a multicenter controlled<br />

trial. French University Association for Surgical Research. Surg Gynecol Obstet, 1991. 173(2): p.<br />

123-30.<br />

107. Xiao, Z.F., et al., Value of radiotherapy after radical surgery for esophageal carc<strong>in</strong>oma: a report of<br />

495 patients.[see comment]. Annals of Thoracic Surgery, 2003. 75(2): p. 331-6.<br />

108. Wong, R.K., et al., Comb<strong>in</strong>ed Modality Radiotherapy and Chemotherapy <strong>in</strong> the Non-surgical<br />

Management of Localized Carc<strong>in</strong>oma of the Esophagus. Practice Guidel<strong>in</strong>e <strong>Report</strong> #2-12., CCO,<br />

Editor. 2005, CCO: Ottawa.<br />

109. Hao, D., et al., Plat<strong>in</strong>um-based concurrent chemoradiotherapy for tumors of the head and neck and<br />

the esophagus. Sem<strong>in</strong>ars <strong>in</strong> Radiation Oncology, 2006. 16(1): p. 10-9.<br />

110. Zhao, K.-l., et al., Late course accelerated hyperfractionated radiotherapy plus concurrent<br />

chemotherapy for squamous cell carc<strong>in</strong>oma of the esophagus: a phase III randomized study.<br />

International Journal of Radiation Oncology, Biology, Physics, 2005. 62(4): p. 1014-20.<br />

111. Piessen, G., et al., Patients <strong>with</strong> locally advanced esophageal carc<strong>in</strong>oma nonresponder to<br />

radiochemotherapy: who will benefit from surgery? Ann Surg Oncol, 2007. 14(7): p. 2036-44.<br />

112. Wenger, U., et al., Health economic evaluation of stent or endolum<strong>in</strong>al brachytherapy as a palliative<br />

strategy <strong>in</strong> patients <strong>with</strong> <strong>in</strong>curable cancer of the oesophagus or gastro-oesophageal junction: results of<br />

a randomized cl<strong>in</strong>ical trial. European Journal of Gastroenterology & Hepatology, 2005. 17(12): p.<br />

1369-77.<br />

113. Pol<strong>in</strong>der, S., et al., Cost study of metal stent placement vs s<strong>in</strong>gle-dose brachytherapy <strong>in</strong> the palliative<br />

treatment of oesophageal cancer. British Journal of Cancer, 2004. 90(11): p. 2067-72.<br />

114. Homs, M.Y.V., et al., Chemotherapy for metastatic carc<strong>in</strong>oma of the esophagus and gastroesophageal<br />

junction. Cochrane Database of Systematic Reviews, 2006(4): p. CD004063.<br />

115. Kuchler, T., et al., Impact of psychotherapeutic support for patients <strong>with</strong> gastro<strong>in</strong>test<strong>in</strong>al cancer<br />

undergo<strong>in</strong>g surgery: 10-year survival results of a randomized trial.[see comment]. Journal of Cl<strong>in</strong>ical<br />

Oncology, 2007. 25(19): p. 2702-8.<br />

116. Kunisaki, C., et al., Surgical Outcomes <strong>in</strong> Esophageal Cancer Patients <strong>with</strong> Tumor Recurrence After<br />

Curative Esophagectomy. J Gastro<strong>in</strong>test Surg, 2007.<br />

117. Natsugoe, S., et al., The role of salvage surgery for recurrence of esophageal squamous cell cancer.<br />

Eur J Surg Oncol, 2006. 32(5): p. 544-7.<br />

118. Yamashita, H., et al., Salvage radiotherapy for postoperative loco-regional recurrence of esophageal<br />

cancer. Dis Esophagus, 2005. 18(4): p. 215-20.<br />

119. Yano, M., et al., Prognosis of patients who develop cervical lymph node recurrence follow<strong>in</strong>g curative<br />

resection for thoracic esophageal cancer. Dis Esophagus, 2006. 19(2): p. 73-7.<br />

120. Komatsu, S., et al., Survival and cl<strong>in</strong>ical evaluation of salvage operation for cervical lymph node<br />

recurrence <strong>in</strong> esophageal cancer. Hepatogastroenterology, 2005. 52(63): p. 796-9.<br />

121. Nishimura, Y., et al., Concurrent chemoradiotherapy <strong>with</strong> protracted <strong>in</strong>fusion of 5-FU and cisplat<strong>in</strong> for<br />

postoperative recurrent or residual esophageal cancer. Jpn J Cl<strong>in</strong> Oncol, 2003. 33(7): p. 341-5.<br />

122. Schipper, P.H., et al., Locally recurrent esophageal carc<strong>in</strong>oma: when is re-resection <strong>in</strong>dicated? Ann<br />

Thorac Surg, 2005. 80(3): p. 1001-5; discussion 1005-6.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 53<br />

123. Nomura, T., et al., Recurrence after endoscopic mucosal resection for superficial esophageal cancer.<br />

Endoscopy, 2000. 32(4): p. 277-80.<br />

124. Graham, D.Y., et al., Prospective evaluation of biopsy number <strong>in</strong> the diagnosis of esophageal and<br />

gastric carc<strong>in</strong>oma. Gastroenterology, 1982. 82(2): p. 228-31.<br />

125. Sugano, K., K. Sato, and K. Yao, New diagnostic approaches for early detection of gastric cancer.<br />

Dig Dis, 2004. 22(4): p. 327-33.<br />

126. Wang, C., Y. Yuan, and R.H. Hunt, The association between Helicobacter pylori <strong>in</strong>fection and early<br />

gastric cancer: a meta-analysis. Am J Gastroenterol, 2007. 102(8): p. 1789-98.<br />

127. Fuccio, L., et al., Systematic review: Helicobacter pylori eradication for the prevention of gastric<br />

cancer. Aliment Pharmacol Ther, 2007. 25(2): p. 133-41.<br />

128. Malferthe<strong>in</strong>er, P., et al., Current concepts <strong>in</strong> the management of Helicobacter pylori <strong>in</strong>fection: the<br />

Maastricht III Consensus <strong>Report</strong>. Gut, 2007. 56(6): p. 772-81.<br />

129. Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC), Recommandations<br />

pour la pratique cl<strong>in</strong>ique: Standards, Options et Recommandations 2003 pour la prise en charge des<br />

patients atte<strong>in</strong>ts d’adénocarc<strong>in</strong>omes de l’estomac (cancers du cardia, autres types histologiques exclus)<br />

(rapport <strong>in</strong>tégral), <strong>in</strong> Standards, Options et Recommandations. 2004, FNCLCC: Paris.<br />

130. Kwee, R.M. and T.C. Kwee, Imag<strong>in</strong>g <strong>in</strong> local stag<strong>in</strong>g of gastric cancer: a systematic review. Journal<br />

of Cl<strong>in</strong>ical Oncology, 2007. 25(15): p. 2107-16.<br />

131. Chen, C.-Y., et al., Gastric cancer: preoperative local stag<strong>in</strong>g <strong>with</strong> 3D multi-detector row CTcorrelation<br />

<strong>with</strong> surgical and histopathologic results. Radiology, 2007. 242(2): p. 472-82.<br />

132. Lauren, P., The Two Histological Ma<strong>in</strong> Types of Gastric Carc<strong>in</strong>oma: Diffuse and So-Called Intest<strong>in</strong>al-<br />

Type Carc<strong>in</strong>oma. an Attempt at a Histo-Cl<strong>in</strong>ical Classification. Acta Pathol Microbiol Scand, 1965.<br />

64: p. 31-49.<br />

133. Goseki, N., T. Takizawa, and M. Koike, Differences <strong>in</strong> the mode of the extension of gastric cancer<br />

classified by histological type: new histological classification of gastric carc<strong>in</strong>oma. Gut, 1992. 33(5): p.<br />

606-12.<br />

134. Mukai, K., et al., Usefulness of preoperative FDG-PET for detection of gastric cancer. Gastric Cancer,<br />

2006. 9(3): p. 192-6.<br />

135. Gretschel, S., et al., Prediction of gastric cancer lymph node status by sent<strong>in</strong>el lymph node biopsy and<br />

the Maruyama computer model. European Journal of Surgical Oncology, 2005. 31(4): p. 393-400.<br />

136. Lee, J.H., et al., Sent<strong>in</strong>el node biopsy us<strong>in</strong>g dye and isotope double tracers <strong>in</strong> early gastric cancer.<br />

Annals of Surgical Oncology, 2006. 13(9): p. 1168-74.<br />

137. Miwa, K., et al., Mapp<strong>in</strong>g sent<strong>in</strong>el nodes <strong>in</strong> patients <strong>with</strong> early-stage gastric carc<strong>in</strong>oma. British Journal<br />

of Surgery, 2003. 90(2): p. 178-82.<br />

138. Mochiki, E., et al., Sent<strong>in</strong>el lymph node mapp<strong>in</strong>g <strong>with</strong> technetium-99m colloidal rhenium sulfide <strong>in</strong><br />

patients <strong>with</strong> gastric carc<strong>in</strong>oma. American Journal of Surgery, 2006. 191(4): p. 465-9.<br />

139. Simsa, J., et al., Sent<strong>in</strong>el node biopsy <strong>in</strong> gastric cancer: prelim<strong>in</strong>ary results. Acta Chirurgica Belgica,<br />

2003. 103(3): p. 270-3.<br />

140. Ono, H., et al., Endoscopic mucosal resection for treatment of early gastric cancer. Gut, 2001. 48(2):<br />

p. 225-9.<br />

141. Tajima, Y., et al., Histopathologic f<strong>in</strong>d<strong>in</strong>gs predict<strong>in</strong>g lymph node metastasis and prognosis of patients<br />

<strong>with</strong> superficial esophageal carc<strong>in</strong>oma: analysis of 240 surgically resected tumors. Cancer, 2000.<br />

88(6): p. 1285-93.<br />

142. Gotoda, T., et al., Incidence of lymph node metastasis from early gastric cancer: estimation <strong>with</strong> a<br />

large number of cases at two large centers. Gastric Cancer, 2000. 3(4): p. 219-225.<br />

143. Kitajima, K., et al., Correlations between lymph node metastasis and depth of submucosal <strong>in</strong>vasion <strong>in</strong><br />

submucosal <strong>in</strong>vasive colorectal carc<strong>in</strong>oma: a Japanese collaborative study. J Gastroenterol, 2004.<br />

39(6): p. 534-43.


54 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

144. Wang, Y.P., C. Bennett, and T. Pan, Endoscopic mucosal resection for early gastric cancer.<br />

Cochrane Database of Systematic Reviews, 2006(1): p. CD004276.<br />

145. Oka, S., et al., Endoscopic submucosal dissection for residual/local recurrence of early gastric cancer<br />

after endoscopic mucosal resection. Endoscopy, 2006. 38(10): p. 996-1000.<br />

146. Japanese Gastric Cancer, A., Japanese Classification of Gastric Carc<strong>in</strong>oma - 2nd <strong>English</strong> Edition.<br />

Gastric Cancer, 1998. 1(1): p. 10-24.<br />

147. Wu, A.W., et al., Neoadjuvant chemotherapy versus none for resectable gastric cancer. Cochrane<br />

Database of Systematic Reviews, 2007(2): p. CD005047.<br />

148. Cunn<strong>in</strong>gham, D., et al., Perioperative chemotherapy versus surgery alone for resectable<br />

gastroesophageal cancer.[see comment]. New England Journal of Medic<strong>in</strong>e, 2006. 355(1): p. 11-20.<br />

149. Boige, V., et al., F<strong>in</strong>al results of a randomized trial compar<strong>in</strong>g preoperative 5-fluorouracil (F)/cisplat<strong>in</strong><br />

(P) to surgery alone <strong>in</strong> adenocarc<strong>in</strong>oma of stomach and lower esophagus (ASLE): FNLCC ACCORD07-<br />

FFCD 9703 trial. J Cl<strong>in</strong> Oncol (Meet<strong>in</strong>g Abstracts), 2007. 25(18_suppl): p. 4510-.<br />

150. Earle, C.C., et al., Neoadjuvant or Adjuvant Therapy for Resectable Gastric Cancer. Practice Guidel<strong>in</strong>e<br />

<strong>Report</strong> #2-14., CCO, Editor. 2003, CCO: Ottawa.<br />

151. Romano, F., et al., Phase-II randomized study of preoperative IL-2 adm<strong>in</strong>istration <strong>in</strong> radically operable<br />

gastric cancer patients. Hepato-Gastroenterology, 2004. 51(60): p. 1872-6.<br />

152. McCulloch, P., et al., Extended versus limited lymph nodes dissection technique for adenocarc<strong>in</strong>oma<br />

of the stomach.[update of Cochrane Database Syst Rev. 2003;(4):CD001964; PMID: 14583942].<br />

Cochrane Database of Systematic Reviews, 2004(4): p. CD001964.<br />

153. Roukos, D.H. and A.M. Kappas, Perspectives <strong>in</strong> the treatment of gastric cancer. Nat Cl<strong>in</strong> Pract<br />

Oncol, 2005. 2(2): p. 98-107.<br />

154. Degiuli, M., et al., Morbidity and mortality after D1 and D2 gastrectomy for cancer: <strong>in</strong>terim analysis<br />

of the Italian Gastric Cancer Study Group (IGCSG) randomised surgical trial. European Journal of<br />

Surgical Oncology, 2004. 30(3): p. 303-8.<br />

155. Degiuli, M., et al., Survival results of a multicentre phase II study to evaluate D2 gastrectomy for<br />

gastric cancer. Br J Cancer, 2004. 90(9): p. 1727-32.<br />

156. Hartgr<strong>in</strong>k, H.H., et al., Extended lymph node dissection for gastric cancer: who may benefit? F<strong>in</strong>al<br />

results of the randomized <strong>Dutch</strong> gastric cancer group trial.[see comment]. Journal of Cl<strong>in</strong>ical<br />

Oncology, 2004. 22(11): p. 2069-77.<br />

157. Kulig, J., et al., Standard D2 versus extended D2 (D2+) lymphadenectomy for gastric cancer: an<br />

<strong>in</strong>terim safety analysis of a multicenter, randomized, cl<strong>in</strong>ical trial. American Journal of Surgery, 2007.<br />

193(1): p. 10-5.<br />

158. Sano, T., et al., Gastric cancer surgery: morbidity and mortality results from a prospective randomized<br />

controlled trial compar<strong>in</strong>g D2 and extended para-aortic lymphadenectomy--Japan Cl<strong>in</strong>ical Oncology<br />

Group study 9501.[see comment]. Journal of Cl<strong>in</strong>ical Oncology, 2004. 22(14): p. 2767-73.<br />

159. Yonemura, Y., et al., Operative morbidity and mortality after D2 and D4 extended dissection for<br />

advanced gastric cancer: a prospective randomized trial conducted by Asian surgeons. Hepato-<br />

Gastroenterology, 2006. 53(69): p. 389-94.<br />

160. Iwata, T., et al., Evaluation of reconstruction after proximal gastrectomy: prospective comparative<br />

study of jejunal <strong>in</strong>terposition and jejunal pouch <strong>in</strong>terposition. Hepato-Gastroenterology, 2006.<br />

53(68): p. 301-3.<br />

161. Yoo, C.H., et al., Proximal gastrectomy reconstructed by jejunal pouch <strong>in</strong>terposition for upper third<br />

gastric cancer: prospective randomized study. World Journal of Surgery, 2005. 29(12): p. 1592-9.<br />

162. Kono, K., et al., Improved quality of life <strong>with</strong> jejunal pouch reconstruction after total gastrectomy.<br />

American Journal of Surgery, 2003. 185(2): p. 150-4.<br />

163. Hayashi, H., et al., Prospective randomized study of open versus laparoscopy-assisted distal<br />

gastrectomy <strong>with</strong> extraperigastric lymph node dissection for early gastric cancer. Surgical Endoscopy,<br />

2005. 19(9): p. 1172-6.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 55<br />

164. Hosono, S., et al., Meta-analysis of short-term outcomes after laparoscopy-assisted distal gastrectomy.<br />

World Journal of Gastroenterology, 2006. 12(47): p. 7676-83.<br />

165. Huscher, C.G.S., et al., Laparoscopic versus open subtotal gastrectomy for distal gastric cancer: fiveyear<br />

results of a randomized prospective trial. Annals of Surgery, 2005. 241(2): p. 232-7.<br />

166. Lee, J.H., H.S. Han, and J.H. Lee, A prospective randomized study compar<strong>in</strong>g open vs laparoscopyassisted<br />

distal gastrectomy <strong>in</strong> early gastric cancer: early results. Surgical Endoscopy, 2005. 19(2): p.<br />

168-73.<br />

167. Callahan, M.A., et al., Influence of surgical subspecialty tra<strong>in</strong><strong>in</strong>g on <strong>in</strong>-hospital mortality for<br />

gastrectomy and colectomy patients. Ann Surg, 2003. 238(4): p. 629-36; discussion 636-9.<br />

168. Oba, K., et al., Efficacy of adjuvant chemotherapy us<strong>in</strong>g oral fluor<strong>in</strong>ated pyrimid<strong>in</strong>es for curatively<br />

resected gastric cancer: a meta-analysis of centrally randomized controlled cl<strong>in</strong>ical trials <strong>in</strong> Japan.<br />

Journal of Chemotherapy, 2006. 18(3): p. 311-7.<br />

169. Bouche, O., et al., Adjuvant chemotherapy <strong>with</strong> 5-fluorouracil and cisplat<strong>in</strong> compared <strong>with</strong> surgery<br />

alone for gastric cancer: 7-year results of the FFCD randomized phase III trial (8801). Annals of<br />

Oncology, 2005. 16(9): p. 1488-97.<br />

170. Chipponi, J., et al., Randomized trial of adjuvant chemotherapy after curative resection for gastric<br />

cancer. American Journal of Surgery, 2004. 187(3): p. 440-5.<br />

171. Nashimoto, A., et al., Randomized trial of adjuvant chemotherapy <strong>with</strong> mitomyc<strong>in</strong>, Fluorouracil, and<br />

Cytos<strong>in</strong>e arab<strong>in</strong>oside followed by oral Fluorouracil <strong>in</strong> serosa-negative gastric cancer: Japan Cl<strong>in</strong>ical<br />

Oncology Group 9206-1.[see comment]. Journal of Cl<strong>in</strong>ical Oncology, 2003. 21(12): p. 2282-7.<br />

172. Nitti, D., et al., Randomized phase III trials of adjuvant FAMTX or FEMTX compared <strong>with</strong> surgery<br />

alone <strong>in</strong> resected gastric cancer. A comb<strong>in</strong>ed analysis of the EORTC GI Group and the ICCG. Annals of<br />

Oncology, 2006. 17(2): p. 262-9.<br />

173. Macdonald, J.S., et al., Chemoradiotherapy after surgery compared <strong>with</strong> surgery alone for<br />

adenocarc<strong>in</strong>oma of the stomach or gastroesophageal junction. N Engl J Med, 2001. 345(10): p. 725-<br />

30.<br />

174. Xu, D.-Z., et al., Meta-analysis of <strong>in</strong>traperitoneal chemotherapy for gastric cancer. World Journal of<br />

Gastroenterology, 2004. 10(18): p. 2727-30.<br />

175. Yan, T.D., et al., Hp40p a systematic review and meta-analysis of the randomised controlled trials on<br />

adjuvant <strong>in</strong>traperitoneal chemotherapy for advanced gastric cancer. ANZ Journal of Surgery, 2007.<br />

77 Suppl 1: p. A49.<br />

176. Oba, K., et al., Efficacy of adjuvant immunochemotherapy <strong>with</strong> polysaccharide K for patients <strong>with</strong><br />

curative resections of gastric cancer. Cancer Immunology, Immunotherapy, 2007. 56(6): p. 905-11.<br />

177. Popiela, T., et al., Efficiency of adjuvant immunochemotherapy follow<strong>in</strong>g curative resection <strong>in</strong> patients<br />

<strong>with</strong> locally advanced gastric cancer. Gastric Cancer, 2004. 7(4): p. 240-5.<br />

178. Hosono, S., et al., Endoscopic stent<strong>in</strong>g versus surgical gastroenterostomy for palliation of malignant<br />

gastroduodenal obstruction: a meta-analysis. Journal of Gastroenterology, 2007. 42(4): p. 283-90.<br />

179. Jeurn<strong>in</strong>k, S.M., et al., Stent versus gastrojejunostomy for the palliation of gastric outlet obstruction: a<br />

systematic review. BMC Gastroenterology, 2007. 7: p. 18.<br />

180. Casaretto, L., P.L.R. Sousa, and J.J. Mari, Chemotherapy versus support cancer treatment <strong>in</strong><br />

advanced gastric cancer: a meta-analysis. Brazilian Journal of Medical & Biological Research, 2006.<br />

39(4): p. 431-40.<br />

181. Wagner, A.D., et al., Chemotherapy <strong>in</strong> advanced gastric cancer: a systematic review and metaanalysis<br />

based on aggregate data. Journal of Cl<strong>in</strong>ical Oncology, 2006. 24(18): p. 2903-9.<br />

182. Whit<strong>in</strong>g, J., et al., Follow-up of gastric cancer: a review. Gastric Cancer, 2006. 9(2): p. 74-81.<br />

183. Tan, I.T. and B.Y. So, Value of <strong>in</strong>tensive follow-up of patients after curative surgery for gastric<br />

carc<strong>in</strong>oma. J Surg Oncol, 2007. 96(6): p. 503-6.<br />

184. S<strong>in</strong>n<strong>in</strong>g, C., et al., Gastric stump carc<strong>in</strong>oma - epidemiology and current concepts <strong>in</strong> pathogenesis and<br />

treatment. Eur J Surg Oncol, 2007. 33(2): p. 133-9.


56 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

185. Ohashi, M., et al., Cancer of the gastric stump follow<strong>in</strong>g distal gastrectomy for cancer. Br J Surg,<br />

2007. 94(1): p. 92-5.<br />

186. Chen, L., et al., Surgical management of gastric stump cancer: a report of 37 cases. J Zhejiang Univ<br />

Sci B, 2005. 6(1): p. 38-42.<br />

187. Hirai, I., et al., Surgical management for metastatic liver tumors. Hepatogastroenterology, 2006.<br />

53(71): p. 757-63.<br />

188. Lehnert, T., et al., Surgical therapy for loco-regional recurrence and distant metastasis of gastric<br />

cancer. Eur J Surg Oncol, 2002. 28(4): p. 455-61.<br />

189. Yokoi, C., et al., Endoscopic submucosal dissection allows curative resection of locally recurrent early<br />

gastric cancer after prior endoscopic mucosal resection. Gastro<strong>in</strong>test Endosc, 2006. 64(2): p. 212-8.<br />

190. Al-Akwaa, A.M., N. Siddiqui, and I.A. Al-Mofleh, Primary gastric lymphoma. World J<br />

Gastroenterol, 2004. 10(1): p. 5-11.<br />

191. Crump, M., M. Gospodarowicz, and F.A. Shepherd, Lymphoma of the gastro<strong>in</strong>test<strong>in</strong>al tract. Sem<strong>in</strong><br />

Oncol, 1999. 26(3): p. 324-37.<br />

192. Zucca, E. and F. Cavalli, Are antibiotics the treatment of choice for gastric lymphoma? Curr Hematol<br />

Rep, 2004. 3(1): p. 11-6.<br />

193. B<strong>in</strong>n, M., et al., Surgical resection plus chemotherapy versus chemotherapy alone: comparison of two<br />

strategies to treat diffuse large B-cell gastric lymphoma.[see comment]. Ann Oncol, 2003. 14(12): p.<br />

1751-7.<br />

194. Yoon, S.S. and E.P. Hochberg, Chemotherapy is an effective first l<strong>in</strong>e treatment for early stage gastric<br />

mucosa-associated lymphoid tissue lymphoma. Cancer Treat Rev, 2006. 32(2): p. 139-43.<br />

195. Fischbach, W., et al., Primary gastric B-cell lymphoma: results of a prospective multicenter study. The<br />

German-Austrian Gastro<strong>in</strong>test<strong>in</strong>al Lymphoma Study Group.[see comment][erratum appears <strong>in</strong><br />

Gastroenterology 2000 Dec;119(6):1809]. Gastroenterology, 2000. 119(5): p. 1191-202.<br />

196. Strecker, P., et al., [Diagnostic value of stomach biopsy <strong>in</strong> comparison <strong>with</strong> surgical specimen <strong>in</strong><br />

gastric B-cell lymphomas of the MALT type]. Pathologe, 1998. 19(3): p. 209-13.<br />

197. Isaacson, P.G., J. Spencer, and D.H. Wright, Classify<strong>in</strong>g primary gut lymphomas. Lancet, 1988.<br />

2(8620): p. 1148-9.<br />

198. Harris, N.L., et al., A revised European-American classification of lymphoid neoplasms: a proposal<br />

from the International Lymphoma Study Group. Blood, 1994. 84(5): p. 1361-92.<br />

199. Caletti, G., et al., Accuracy of endoscopic ultrasonography <strong>in</strong> the diagnosis and stag<strong>in</strong>g of gastric<br />

cancer and lymphoma. Surgery, 1993. 113(1): p. 14-27.<br />

200. Fischbach, W., M.-E. Goebeler-Kolve, and A. Gre<strong>in</strong>er, Diagnostic accuracy of EUS <strong>in</strong> the local<br />

stag<strong>in</strong>g of primary gastric lymphoma: results of a prospective, multicenter study compar<strong>in</strong>g EUS <strong>with</strong><br />

histopathologic stage. Gastro<strong>in</strong>test Endosc, 2002. 56(5): p. 696-700.<br />

201. Rodriguez, M., et al., [18F] FDG PET <strong>in</strong> gastric non-Hodgk<strong>in</strong>'s lymphoma. Acta Oncol, 1997. 36(6):<br />

p. 577-84.<br />

202. Vorbeck, F., et al., Comparison of spiral-computed tomography <strong>with</strong> water-fill<strong>in</strong>g of the stomach and<br />

endosonography for gastric lymphoma of mucosa-associated lymphoid tissue-type. Digestion, 2002.<br />

65(4): p. 196-9.<br />

203. Hoepffner, N., et al., [Value of endosonography <strong>in</strong> diagnostic stag<strong>in</strong>g of primary gastric lymphoma<br />

(MALT type)]. Med Kl<strong>in</strong> (Munich), 2003. 98(6): p. 313-7.<br />

204. Hong, S.S., et al., A prospective analysis of low-grade gastric malt lymphoma after Helicobacter pylori<br />

eradication. Helicobacter, 2006. 11(6): p. 569-73.<br />

205. Nakamura, S., et al., Long-term cl<strong>in</strong>ical outcome of Helicobacter pylori eradication for gastric mucosaassociated<br />

lymphoid tissue lymphoma <strong>with</strong> a reference to second-l<strong>in</strong>e treatment. Cancer, 2005.<br />

104(3): p. 532-40.<br />

206. Wundisch, T., et al., Long-term follow-up of gastric MALT lymphoma after Helicobacter pylori<br />

eradication. J Cl<strong>in</strong> Oncol, 2005. 23(31): p. 8018-24.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 57<br />

207. Fischbach, W., et al., Long term outcome of patients <strong>with</strong> gastric marg<strong>in</strong>al zone B cell lymphoma of<br />

mucosa associated lymphoid tissue (MALT) follow<strong>in</strong>g exclusive Helicobacter pylori eradication therapy:<br />

experience from a large prospective series. Gut, 2004. 53(1): p. 34-7.<br />

208. Montalban, C., et al., Treatment of low grade gastric mucosa-associated lymphoid tissue lymphoma <strong>in</strong><br />

stage I <strong>with</strong> Helicobacter pylori eradication. Long-term results after sequential histologic and molecular<br />

follow-up. Haematologica, 2001. 86(6): p. 609-17.<br />

209. Aviles, A., et al., Mucosa-associated lymphoid tissue (MALT) lymphoma of the stomach: results of a<br />

controlled cl<strong>in</strong>ical trial. Medical Oncology, 2005. 22(1): p. 57-62.<br />

210. Verma, S., et al., Imat<strong>in</strong>ib Mesylate (Gleevec) for the Treatment of Adult Patients <strong>with</strong> Unresectable<br />

or Metastatic Gastro<strong>in</strong>test<strong>in</strong>al Stromal Tumours:A Cl<strong>in</strong>ical Practice Guidel<strong>in</strong>e. 2006, CCO: Ottawa.<br />

211. Wilson, J.S., et al., Imat<strong>in</strong>ib mesylate for the treatment of patients <strong>with</strong> unresectable and/or<br />

metastatic gastro-<strong>in</strong>test<strong>in</strong>al stromal tumours (GIST). 2004, West Midlands Health Technology<br />

Assessment Collaboration, University of Birm<strong>in</strong>gham: Birm<strong>in</strong>gham.<br />

212. Hwang, J.H., S.D. Rulyak, and M.B. Kimmey, American Gastroenterological Association Institute<br />

technical review on the management of gastric subepithelial masses. Gastroenterology, 2006.<br />

130(7): p. 2217-28.<br />

213. Rimond<strong>in</strong>i, A., et al., Contribution of CT to treatment plann<strong>in</strong>g <strong>in</strong> patients <strong>with</strong> GIST. Radiol Med<br />

(Tor<strong>in</strong>o), 2007. 112(5): p. 691-702.<br />

214. Lupescu, I.G., et al., Computer tomographic evaluation of digestive tract non-Hodgk<strong>in</strong> lymphomas. J<br />

Gastro<strong>in</strong>test<strong>in</strong> Liver Dis, 2007. 16(3): p. 315-9.<br />

215. De Leo, C., et al., Gastro<strong>in</strong>test<strong>in</strong>al stromal tumours: experience <strong>with</strong> multislice CT. Radiol Med<br />

(Tor<strong>in</strong>o), 2006. 111(8): p. 1103-14.<br />

216. Da Ronch, T., A. Modesto, and M. Bazzocchi, Gastro<strong>in</strong>test<strong>in</strong>al stromal tumour: spiral computed<br />

tomography features and pathologic correlation. Radiol Med (Tor<strong>in</strong>o), 2006. 111(5): p. 661-73.<br />

217. Hong, X., et al., Gastro<strong>in</strong>test<strong>in</strong>al stromal tumor: role of CT <strong>in</strong> diagnosis and <strong>in</strong> response evaluation and<br />

surveillance after treatment <strong>with</strong> imat<strong>in</strong>ib. Radiographics, 2006. 26(2): p. 481-95.<br />

218. Fletcher, C.D.M., et al., Diagnosis of gastro<strong>in</strong>test<strong>in</strong>al stromal tumors: A consensus approach. Hum<br />

Pathol, 2002. 33(5): p. 459-65.<br />

219. Blay, J.-Y., et al., [Recommendations for the management of GIST patients]. Bull Cancer, 2005.<br />

92(10): p. 907-18.<br />

220. Dematteo, R.P., et al. Adjuvant imat<strong>in</strong>ib mesylate <strong>in</strong>creases recurrence free survival (RFS) <strong>in</strong> patients<br />

<strong>with</strong> completely resected localized primary gastro<strong>in</strong>test<strong>in</strong>al stromal tumor (GIST): North American<br />

Intergroup Phase III trial ACOSOG Z9001. 2007 [cited 2008 23-01-2008]; Available from:<br />

http://www.asco.org/portal/site/ASCO/menuitem.34d60f5624ba07fd506fe310ee37a01d/?vgnext<br />

oid=76f8201eb61a7010VgnVCM100000ed730ad1RCRD&vmview=abst_detail_view&confID=4<br />

7&abstractID=100001.<br />

221. Z<strong>in</strong>cirkeser, S., et al., Early detection of response to imat<strong>in</strong>ib therapy for gastro<strong>in</strong>test<strong>in</strong>al stromal<br />

tumor by us<strong>in</strong>g 18F-FDG-positron emission tomography and computed tomography imag<strong>in</strong>g. World J<br />

Gastroenterol, 2007. 13(15): p. 2261-2.<br />

222. Goh, B.K., et al., Pathologic, radiologic and PET scan response of gastro<strong>in</strong>test<strong>in</strong>al stromal tumors after<br />

neoadjuvant treatment <strong>with</strong> imat<strong>in</strong>ib mesylate. Eur J Surg Oncol, 2006. 32(9): p. 961-3.<br />

223. He<strong>in</strong>icke, T., et al., Very early detection of response to imat<strong>in</strong>ib mesylate therapy of gastro<strong>in</strong>test<strong>in</strong>al<br />

stromal tumours us<strong>in</strong>g 18fluoro-deoxyglucose-positron emission tomography. Anticancer Res, 2005.<br />

25(6C): p. 4591-4.<br />

224. Goldste<strong>in</strong>, D., et al., Gastro<strong>in</strong>test<strong>in</strong>al stromal tumours: correlation of F-FDG gamma camera-based<br />

co<strong>in</strong>cidence positron emission tomography <strong>with</strong> CT for the assessment of treatment response--an<br />

AGITG study. Oncology, 2005. 69(4): p. 326-32.<br />

225. Demetri, G.D., et al., Efficacy and safety of sunit<strong>in</strong>ib <strong>in</strong> patients <strong>with</strong> advanced gastro<strong>in</strong>test<strong>in</strong>al<br />

stromal tumour after failure of imat<strong>in</strong>ib: a randomised controlled trial. Lancet, 2006. 368(9544): p.<br />

1329-38.


58 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

226. Jacobson, B.C., et al., The role of endoscopy <strong>in</strong> the assessment and treatment of esophageal cancer.<br />

Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2003. 57(7): p. 817-22.<br />

227. National Comprehens<strong>in</strong>e Cancer Network (NCCN), Esophageal Cancer, <strong>in</strong> NCCN Cl<strong>in</strong>ical<br />

Practice Guidel<strong>in</strong>es <strong>in</strong> Oncology, NCCN, Editor. 2007, NCCN.<br />

228. National Comprehens<strong>in</strong>e Cancer Network (NCCN), Gastric Cancer, <strong>in</strong> NCCN Cl<strong>in</strong>ical Practice<br />

Guidel<strong>in</strong>es <strong>in</strong> Oncology, NCCN, Editor. 2007, NCCN.<br />

229. Cunn<strong>in</strong>gham, D., et al., ESMO M<strong>in</strong>imum Cl<strong>in</strong>ical Recommendations for diagnosis, treatment and<br />

follow-up of gastric cancer. Annals of Oncology, 2005. 16(1).<br />

230. Stahl, M., et al., ESMO M<strong>in</strong>imal Cl<strong>in</strong>ical Recommendations for diagnosis, treatment and follow-up of<br />

esophageal cancer. Annals of Oncology, 2005. 16(1).<br />

231. Sampl<strong>in</strong>er, R.E. and G. Practice Parameters Committee of the American College of, Updated<br />

guidel<strong>in</strong>es for the diagnosis, surveillance, and therapy of Barrett's esophagus. American Journal of<br />

Gastroenterology, 2002. 97(8): p. 1888-95.<br />

232. Allum, W.H., et al., Guidel<strong>in</strong>es for the management of oesophageal and gastric cancer. Gut, 2002.<br />

1(50).<br />

233. Marmo, R., et al., Comb<strong>in</strong>ation of age and sex improves the ability to predict upper gastro<strong>in</strong>test<strong>in</strong>al<br />

malignancy <strong>in</strong> patients <strong>with</strong> uncomplicated dyspepsia: a prospective multicentre database study. Am J<br />

Gastroenterol, 2005. 100(4): p. 784-91.<br />

234. Kim, S.H., et al., Three-dimensional MDCT imag<strong>in</strong>g and CT esophagography for evaluation of<br />

esophageal tumors: prelim<strong>in</strong>ary study. European Radiology, 2006. 16(11): p. 2418-26.<br />

235. Umeoka, S., et al., Esophageal cancer: evaluation <strong>with</strong> triple-phase dynamic CT--<strong>in</strong>itial experience.<br />

Radiology, 2006. 239(3): p. 777-83.<br />

236. Panebianco, V., et al., 3D CT protocol <strong>in</strong> the assessment of the esophageal neoplastic lesions: can it<br />

improve TNM stag<strong>in</strong>g? European Radiology, 2006. 16(2): p. 414-21.<br />

237. Onbas, O., et al., Preoperative stag<strong>in</strong>g of esophageal carc<strong>in</strong>oma <strong>with</strong> multidetector CT and virtual<br />

endoscopy. Eur J Radiol, 2006. 57(1): p. 90-5.<br />

238. Weaver, S.R., et al., Comparison of special <strong>in</strong>terest computed tomography, endosonography and<br />

histopathological stage of oesophageal cancer. Cl<strong>in</strong>ical Radiology, 2004. 59(6): p. 499-504.<br />

239. Yoon, Y.C., et al., Metastasis to regional lymph nodes <strong>in</strong> patients <strong>with</strong> esophageal squamous cell<br />

carc<strong>in</strong>oma: CT versus FDG PET for presurgical detection prospective study. Radiology, 2003. 227(3):<br />

p. 764-70.<br />

240. Vazquez-Sequeiros, E., et al., Rout<strong>in</strong>e vs. selective EUS-guided FNA approach for preoperative nodal<br />

stag<strong>in</strong>g of esophageal carc<strong>in</strong>oma.[see comment]. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2006. 63(2): p. 204-<br />

11.<br />

241. May, A., et al., Accuracy of stag<strong>in</strong>g <strong>in</strong> early oesophageal cancer us<strong>in</strong>g high resolution endoscopy and<br />

high resolution endosonography: a comparative, prospective, and bl<strong>in</strong>ded trial. Gut, 2004. 53(5): p.<br />

634-40.<br />

242. Yanai, H., et al., Prognostic value and <strong>in</strong>terobserver agreement of endoscopic ultrasonography for<br />

superficial squamous cell carc<strong>in</strong>oma of the esophagus: a prospective study. International Journal of<br />

Gastro<strong>in</strong>test<strong>in</strong>al Cancer, 2003. 34(1): p. 1-8.<br />

243. Chang, K.J., et al., Impact of endoscopic ultrasound comb<strong>in</strong>ed <strong>with</strong> f<strong>in</strong>e-needle aspiration biopsy <strong>in</strong> the<br />

management of esophageal cancer. Endoscopy, 2003. 35(11): p. 962-6.<br />

244. Mariette, C., et al., Value of endoscopic ultrasonography as a predictor of long-term survival <strong>in</strong><br />

oesophageal carc<strong>in</strong>oma.[see comment]. British Journal of Surgery, 2003. 90(11): p. 1367-72.<br />

245. Menon, K.V. and T.C.B. Dehn, Multiport stag<strong>in</strong>g laparoscopy <strong>in</strong> esophageal and cardiac carc<strong>in</strong>oma.<br />

Diseases of the Esophagus, 2003. 16(4): p. 295-300.<br />

246. Riddell, A.M., et al., Potential of surface-coil MRI for stag<strong>in</strong>g of esophageal cancer. AJR, 2006.<br />

American Journal of Roentgenology. 187(5): p. 1280-7.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 59<br />

247. Meyers, B.F., et al., The utility of positron emission tomography <strong>in</strong> stag<strong>in</strong>g of potentially operable<br />

carc<strong>in</strong>oma of the thoracic esophagus: results of the American College of Surgeons Oncology Group<br />

Z0060 trial. Journal of Thoracic & Cardiovascular Surgery, 2007. 133(3): p. 738-45.<br />

248. van Baardwijk, A., et al., The current status of FDG-PET <strong>in</strong> tumour volume def<strong>in</strong>ition <strong>in</strong> radiotherapy<br />

treatment plann<strong>in</strong>g. Cancer Treatment Reviews, 2006. 32(4): p. 245-60.<br />

249. Lev<strong>in</strong>e, E.A., et al., Predictive value of 18-fluoro-deoxy-glucose-positron emission tomography (18F-<br />

FDG-PET) <strong>in</strong> the identification of responders to chemoradiation therapy for the treatment of locally<br />

advanced esophageal cancer. Annals of Surgery, 2006. 243(4): p. 472-8.<br />

250. Kato, H., et al., Comparison between whole-body positron emission tomography and bone sc<strong>in</strong>tigraphy<br />

<strong>in</strong> evaluat<strong>in</strong>g bony metastases of esophageal carc<strong>in</strong>omas. Anticancer Research, 2005. 25(6C): p.<br />

4439-44.<br />

251. van Westreenen, H.L., et al., Comparison of 18F-FLT PET and 18F-FDG PET <strong>in</strong> esophageal cancer.<br />

Journal of Nuclear Medic<strong>in</strong>e, 2005. 46(3): p. 400-4.<br />

252. Vrieze, O., et al., Is there a role for FGD-PET <strong>in</strong> radiotherapy plann<strong>in</strong>g <strong>in</strong> esophageal carc<strong>in</strong>oma?<br />

Radiotherapy & Oncology, 2004. 73(3): p. 269-75.<br />

253. Pohl, J., et al., Comparison of computed virtual chromoendoscopy and conventional chromoendoscopy<br />

<strong>with</strong> acetic acid for detection of neoplasia <strong>in</strong> Barrett's esophagus. Endoscopy, 2007. 39(7): p. 594-8.<br />

254. Ragunath, K., et al., A randomized, prospective cross-over trial compar<strong>in</strong>g methylene blue-directed<br />

biopsy and conventional random biopsy for detect<strong>in</strong>g <strong>in</strong>test<strong>in</strong>al metaplasia and dysplasia <strong>in</strong> Barrett's<br />

esophagus. Endoscopy, 2003. 35(12): p. 998-1003.<br />

255. Pech, O., et al., The impact of endoscopic ultrasound and computed tomography on the TNM stag<strong>in</strong>g<br />

of early cancer <strong>in</strong> Barrett's esophagus. American Journal of Gastroenterology, 2006. 101(10): p.<br />

2223-9.<br />

256. Larghi, A., et al., EUS followed by EMR for stag<strong>in</strong>g of high-grade dysplasia and early cancer <strong>in</strong><br />

Barrett's esophagus. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2005. 62(1): p. 16-23.<br />

257. Waxman, I., et al., High-frequency probe ultrasonography has limited accuracy for detect<strong>in</strong>g <strong>in</strong>vasive<br />

adenocarc<strong>in</strong>oma <strong>in</strong> patients <strong>with</strong> Barrett's esophagus and high-grade dysplasia or <strong>in</strong>tramucosal<br />

carc<strong>in</strong>oma: a case series. American Journal of Gastroenterology, 2006. 101(8): p. 1773-9.<br />

258. May, A., et al., A prospective randomized trial of two different endoscopic resection techniques for<br />

early stage cancer of the esophagus.[see comment]. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2003. 58(2): p.<br />

167-75.<br />

259. Geh, J.I., et al., Systematic overview of preoperative (neoadjuvant) chemoradiotherapy trials <strong>in</strong><br />

oesophageal cancer: evidence of a radiation and chemotherapy dose response. Radiotherapy &<br />

Oncology, 2006. 78(3): p. 236-44.<br />

260. Greer, S.E., et al., Neoadjuvant chemoradiotherapy for esophageal carc<strong>in</strong>oma: a meta-analysis.[see<br />

comment]. Surgery, 2005. 137(2): p. 172-7.<br />

261. Burmeister, B.H., et al., Surgery alone versus chemoradiotherapy followed by surgery for resectable<br />

cancer of the oesophagus: a randomised controlled phase III trial.[see comment]. Lancet Oncology,<br />

2005. 6(9): p. 659-68.<br />

262. Duong, C.P., et al., FDG-PET status follow<strong>in</strong>g chemoradiotherapy provides high management impact<br />

and powerful prognostic stratification <strong>in</strong> oesophageal cancer. European Journal of Nuclear Medic<strong>in</strong>e<br />

& Molecular Imag<strong>in</strong>g, 2006. 33(7): p. 770-8.<br />

263. Br<strong>in</strong>k, I., et al., Effects of neoadjuvant radio-chemotherapy on 18F-FDG-PET <strong>in</strong> esophageal carc<strong>in</strong>oma.<br />

European Journal of Surgical Oncology, 2004. 30(5): p. 544-50.<br />

264. Nakahara, T., et al., Comparison of barium swallow, CT and thallium-201 SPECT <strong>in</strong> evaluat<strong>in</strong>g<br />

responses of patients <strong>with</strong> esophageal squamous cell carc<strong>in</strong>oma to preoperative chemoradiotherapy.<br />

Annals of Nuclear Medic<strong>in</strong>e, 2003. 17(7): p. 583-91.<br />

265. Stahl, M., et al., Chemoradiation <strong>with</strong> and <strong>with</strong>out surgery <strong>in</strong> patients <strong>with</strong> locally advanced squamous<br />

cell carc<strong>in</strong>oma of the esophagus.[see comment][erratum appears <strong>in</strong> J Cl<strong>in</strong> Oncol. 2006 Jan<br />

20;24(3):531]. Journal of Cl<strong>in</strong>ical Oncology, 2005. 23(10): p. 2310-7.


60 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

266. Bhat, M.A., et al., Use of pedicled omentum <strong>in</strong> esophagogastric anastomosis for prevention of<br />

anastomotic leak.[see comment]. Annals of Thoracic Surgery, 2006. 82(5): p. 1857-62.<br />

267. Hsu, H.-H., et al., Comparison of manual and mechanical cervical esophagogastric anastomosis after<br />

esophageal resection for squamous cell carc<strong>in</strong>oma: a prospective randomized controlled trial.<br />

European Journal of Cardio-Thoracic Surgery, 2004. 25(6): p. 1097-101.<br />

268. Walther, B., et al., Cervical or thoracic anastomosis after esophageal resection and gastric tube<br />

reconstruction: a prospective randomized trial compar<strong>in</strong>g sutured neck anastomosis <strong>with</strong> stapled<br />

<strong>in</strong>trathoracic anastomosis.[see comment]. Annals of Surgery, 2003. 238(6): p. 803-12; discussion<br />

812-4.<br />

269. Ajani, J., Review of capecitab<strong>in</strong>e as oral treatment of gastric, gastroesophageal, and esophageal<br />

cancers. Cancer, 2006. 107(2): p. 221-31.<br />

270. Sumpter, K., et al., <strong>Report</strong> of two protocol planned <strong>in</strong>terim analyses <strong>in</strong> a randomised multicentre<br />

phase III study compar<strong>in</strong>g capecitab<strong>in</strong>e <strong>with</strong> fluorouracil and oxaliplat<strong>in</strong> <strong>with</strong> cisplat<strong>in</strong> <strong>in</strong> patients <strong>with</strong><br />

advanced oesophagogastric cancer receiv<strong>in</strong>g ECF. British Journal of Cancer, 2005. 92(11): p. 1976-<br />

83.<br />

271. Wenger, U., et al., An antireflux stent versus conventional stents for palliation of distal esophageal or<br />

cardia cancer: a randomized cl<strong>in</strong>ical study. Surgical Endoscopy, 2006. 20(11): p. 1675-80.<br />

272. Shim, C.S., et al., Management of malignant stricture of the esophagogastric junction <strong>with</strong> a newly<br />

designed self-expand<strong>in</strong>g metal stent <strong>with</strong> an antireflux mechanism. Endoscopy, 2005. 37(4): p. 335-<br />

9.<br />

273. Shenf<strong>in</strong>e, J., et al., A pragmatic randomised controlled trial of the cost-effectiveness of palliative<br />

therapies for patients <strong>with</strong> <strong>in</strong>operable oesophageal cancer. Health Technology Assessment, 2005.<br />

9(5): p. iii, 1-121.<br />

274. Trumper, M., et al., Efficacy and tolerability of chemotherapy <strong>in</strong> elderly patients <strong>with</strong> advanced<br />

oesophago-gastric cancer: A pooled analysis of three cl<strong>in</strong>ical trials. European Journal of Cancer,<br />

2006. 42(7): p. 827-34.<br />

275. Duffour, J., et al., Safety of cisplat<strong>in</strong> comb<strong>in</strong>ed <strong>with</strong> cont<strong>in</strong>uous 5-FU versus bolus 5-FU and leucovor<strong>in</strong>,<br />

<strong>in</strong> metastatic gastro<strong>in</strong>test<strong>in</strong>al cancer (FFCD 9404 randomised trial). Anticancer Research, 2006.<br />

26(5B): p. 3877-83.<br />

276. Moraca, R.J. and D.E. Low, Outcomes and health-related quality of life after esophagectomy for highgrade<br />

dysplasia and <strong>in</strong>tramucosal cancer. Arch Surg, 2006. 141(6): p. 545-9; discussion 549-51.<br />

277. Cense, H.A., et al., Effects of prolonged <strong>in</strong>tensive care unit stay on quality of life and long-term<br />

survival after transthoracic esophageal resection. Crit Care Med, 2006. 34(2): p. 354-62.<br />

278. Lagergren, P., et al., Health-related quality of life among patients cured by surgery for esophageal<br />

cancer. Cancer, 2007. 110(3): p. 686-93.<br />

279. Blazeby, J.M., et al., Health-related quality of life dur<strong>in</strong>g neoadjuvant treatment and surgery for<br />

localized esophageal carc<strong>in</strong>oma. Cancer, 2005. 103(9): p. 1791-9.<br />

280. Homs, M.Y.V., et al., S<strong>in</strong>gle-dose brachytherapy versus metal stent placement for the palliation of<br />

dysphagia from oesophageal cancer: multicentre randomised trial. Lancet, 2004. 364(9444): p. 1497-<br />

504.<br />

281. Lobo, D.N., et al., Early postoperative jejunostomy feed<strong>in</strong>g <strong>with</strong> an immune modulat<strong>in</strong>g diet <strong>in</strong><br />

patients undergo<strong>in</strong>g resectional surgery for upper gastro<strong>in</strong>test<strong>in</strong>al cancer: a prospective, randomized,<br />

controlled, double-bl<strong>in</strong>d study. Cl<strong>in</strong>ical Nutrition, 2006. 25(5): p. 716-26.<br />

282. Ryan, A.M., et al., Post-oesophagectomy early enteral nutrition via a needle catheter jejunostomy: 8year<br />

experience at a specialist unit. Cl<strong>in</strong> Nutr, 2006. 25(3): p. 386-93.<br />

283. Gabor, S., et al., Early enteral feed<strong>in</strong>g compared <strong>with</strong> parenteral nutrition after oesophageal or<br />

oesophagogastric resection and reconstruction. Br J Nutr, 2005. 93(4): p. 509-13.<br />

284. Lam, E.C., et al., In patients referred for <strong>in</strong>vestigation because computed tomography suggests<br />

thickened gastric folds, endoscopic ultrasound is superfluous if gastroscopy is normal. American<br />

Journal of Gastroenterology, 2007. 102(6): p. 1200-3.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 61<br />

285. Ichikawa, T., et al., Endoscopic ultrasonography <strong>with</strong> three m<strong>in</strong>iature probes of different frequency is<br />

an accurate diagnostic tool for endoscopic submucosal dissection. Hepato-Gastroenterology, 2007.<br />

54(73): p. 325-8.<br />

286. G<strong>in</strong>es, A., et al., Endoscopic ultrasonography <strong>in</strong> patients <strong>with</strong> large gastric folds at endoscopy and<br />

biopsies negative for malignancy: predictors of malignant disease and cl<strong>in</strong>ical impact. American<br />

Journal of Gastroenterology, 2006. 101(1): p. 64-9.<br />

287. Matthes, K., et al., EUS stag<strong>in</strong>g of upper GI malignancies: results of a prospective randomized<br />

trial.[see comment]. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2006. 64(4): p. 496-502.<br />

288. Chu, K.-M., et al., A prospective evaluation of catheter probe EUS for the detection of ascites <strong>in</strong><br />

patients <strong>with</strong> gastric carc<strong>in</strong>oma. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2004. 59(4): p. 471-4.<br />

289. Deogracias, M.L., et al., Absence of port-site metastases follow<strong>in</strong>g stag<strong>in</strong>g laparoscopy for gastric<br />

carc<strong>in</strong>oma. Revista Espanola de Enfermedades Digestivas, 2006. 98(10): p. 755-9.<br />

290. Lee, S.-H., et al., Cl<strong>in</strong>ical efficacy of EMR <strong>with</strong> submucosal <strong>in</strong>jection of a fibr<strong>in</strong>ogen mixture: a<br />

prospective randomized trial. Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy, 2006. 64(5): p. 691-6.<br />

291. Ates, E., et al., Perioperative immunonutrition ameliorates the postoperative immune depression <strong>in</strong><br />

patients <strong>with</strong> gastro<strong>in</strong>test<strong>in</strong>al system cancer (prospective cl<strong>in</strong>ical study <strong>in</strong> 42 patients). Acta<br />

Gastroenterologica Belgica, 2004. 67(3): p. 250-4.<br />

292. Romano, F., et al., Biological, histological, and cl<strong>in</strong>ical impact of preoperative IL-2 adm<strong>in</strong>istration <strong>in</strong><br />

radically operable gastric cancer patients. Journal of Surgical Oncology, 2004. 88(4): p. 240-7.<br />

293. Enz<strong>in</strong>ger, P.C., et al., Impact of hospital volume on recurrence and survival after surgery for gastric<br />

cancer. Annals of Surgery, 2007. 245(3): p. 426-34.<br />

294. Inaba, T., et al., Prospective randomized study of two laparotomy <strong>in</strong>cisions for gastrectomy: midl<strong>in</strong>e<br />

<strong>in</strong>cision versus transverse <strong>in</strong>cision. Gastric Cancer, 2004. 7(3): p. 167-71.<br />

295. Lee, W.J., et al., Randomized cl<strong>in</strong>ical trial of Ligasure versus conventional surgery for extended gastric<br />

cancer resection. British Journal of Surgery, 2003. 90(12): p. 1493-6.<br />

296. Sasako, M., et al., Left thoracoabdom<strong>in</strong>al approach versus abdom<strong>in</strong>al-transhiatal approach for gastric<br />

cancer of the cardia or subcardia: a randomised controlled trial.[see comment]. Lancet Oncology,<br />

2006. 7(8): p. 644-51.<br />

297. Yu, W., G.S. Choi, and H.Y. Chung, Randomized cl<strong>in</strong>ical trial of splenectomy versus splenic<br />

preservation <strong>in</strong> patients <strong>with</strong> proximal gastric cancer. British Journal of Surgery, 2006. 93(5): p. 559-<br />

63.<br />

298. Wu, C.-W., et al., Nodal dissection for patients <strong>with</strong> gastric cancer: a randomised controlled trial.[see<br />

comment]. Lancet Oncology, 2006. 7(4): p. 309-15.<br />

299. Adachi, S., et al., Subjective and functional results after total gastrectomy: prospective study for<br />

longterm comparison of reconstruction procedures. Gastric Cancer, 2003. 6(1): p. 24-9.<br />

300. Ishikawa, M., et al., Prospective randomized trial compar<strong>in</strong>g Billroth I and Roux-en-Y procedures after<br />

distal gastrectomy for gastric carc<strong>in</strong>oma. World Journal of Surgery, 2005. 29(11): p. 1415-20;<br />

discussion 1421.<br />

301. Mochiki, E., et al., Postoperative functional evaluation of jejunal <strong>in</strong>terposition <strong>with</strong> or <strong>with</strong>out a pouch<br />

after a total gastrectomy for gastric cancer. American Journal of Surgery, 2004. 187(6): p. 728-35.<br />

302. Shibata, C., et al., Outcomes after pylorus-preserv<strong>in</strong>g gastrectomy for early gastric cancer: a<br />

prospective multicenter trial.[see comment]. World Journal of Surgery, 2004. 28(9): p. 857-61.<br />

303. Berardi, R., et al., Gastric cancer treatment: a systematic review. Oncology <strong>Report</strong>s, 2004. 11(4): p.<br />

911-6.<br />

304. Casc<strong>in</strong>u, S., et al., Adjuvant treatment of high-risk, radically resected gastric cancer patients <strong>with</strong> 5fluorouracil,<br />

leucovor<strong>in</strong>, cisplat<strong>in</strong>, and epidoxorubic<strong>in</strong> <strong>in</strong> a randomized controlled trial.[see comment].<br />

Journal of the National Cancer Institute, 2007. 99(8): p. 601-7.<br />

305. Nishikawa, T. and S. Maetani, Evaluation of <strong>in</strong>tensive adjuvant chemotherapy <strong>in</strong> gastric cancer us<strong>in</strong>g<br />

life expectancy compared <strong>with</strong> log-rank test as a measure of survival benefit. Annals of Surgical<br />

Oncology, 2007. 14(2): p. 348-54.


62 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

306. Tentes, A.-A.K., et al., Intraarterial chemotherapy as an adjuvant treatment <strong>in</strong> locally advanced<br />

gastric cancer. Langenbecks Archives of Surgery, 2006. 391(2): p. 124-9.<br />

307. Ueda, Y., et al., A randomized phase III trial of postoperative adjuvant therapy <strong>with</strong> S-1 alone versus<br />

S-1 plus PSK for stage II/IIIA gastric cancer: Hokuriku-K<strong>in</strong>ki Immunochemo-Therapy Study Group-<br />

Gastric Cancer (HKIT-GC). Japanese Journal of Cl<strong>in</strong>ical Oncology, 2006. 36(8): p. 519-22.<br />

308. Tsuburaya, A., et al., A randomized phase III trial of post-operative adjuvant oral fluoropyrimid<strong>in</strong>e<br />

versus sequential paclitaxel/oral fluoropyrimid<strong>in</strong>e; and UFT versus S1 for T3/T4 gastric carc<strong>in</strong>oma: the<br />

Stomach Cancer Adjuvant Multi-<strong>in</strong>stitutional Trial Group (Samit) Trial. Japanese Journal of Cl<strong>in</strong>ical<br />

Oncology, 2005. 35(11): p. 672-5.<br />

309. Yu, W., A review of adjuvant therapy for resected primary gastric cancer <strong>with</strong> an update on Taegu's<br />

phase III trial <strong>with</strong> <strong>in</strong>traperitoneal chemotherapy. European Journal of Surgical Oncology, 2006.<br />

32(6): p. 655-60.<br />

310. Chen, D.W., et al., Role of enteral immunonutrition <strong>in</strong> patients <strong>with</strong> gastric carc<strong>in</strong>oma undergo<strong>in</strong>g<br />

major surgery. Asian Journal of Surgery, 2005. 28(2): p. 121-4.<br />

311. Farreras, N., et al., Effect of early postoperative enteral immunonutrition on wound heal<strong>in</strong>g <strong>in</strong> patients<br />

undergo<strong>in</strong>g surgery for gastric cancer. Cl<strong>in</strong>ical Nutrition, 2005. 24(1): p. 55-65.<br />

312. Sato, Y., et al., A randomized controlled study of immunochemotherapy <strong>with</strong> OK-432 after curative<br />

surgery for gastric cancer. Journal of Immunotherapy, 2004. 27(5): p. 394-7.<br />

313. Di Cosimo, S., et al., Docetaxel <strong>in</strong> advanced gastric cancer--review of the ma<strong>in</strong> cl<strong>in</strong>ical trials. Acta<br />

Oncologica, 2003. 42(7): p. 693-700.<br />

314. Wohrer, S.S., M. Raderer, and M. Hejna, Palliative chemotherapy for advanced gastric cancer.<br />

Annals of Oncology, 2004. 15(11): p. 1585-95.<br />

315. Ajani, J.A., et al., Phase II multi-<strong>in</strong>stitutional randomized trial of docetaxel plus cisplat<strong>in</strong> <strong>with</strong> or <strong>with</strong>out<br />

fluorouracil <strong>in</strong> patients <strong>with</strong> untreated, advanced gastric, or gastroesophageal adenocarc<strong>in</strong>oma.[see<br />

comment]. Journal of Cl<strong>in</strong>ical Oncology, 2005. 23(24): p. 5660-7.<br />

316. Bouche, O., et al., Randomized multicenter phase II trial of a biweekly regimen of fluorouracil and<br />

leucovor<strong>in</strong> (LV5FU2), LV5FU2 plus cisplat<strong>in</strong>, or LV5FU2 plus ir<strong>in</strong>otecan <strong>in</strong> patients <strong>with</strong> previously<br />

untreated metastatic gastric cancer: a Federation Francophone de Cancerologie Digestive Group Study-<br />

-FFCD 9803. Journal of Cl<strong>in</strong>ical Oncology, 2004. 22(21): p. 4319-28.<br />

317. Cocconi, G., et al., Cisplat<strong>in</strong>, epirubic<strong>in</strong>, leucovor<strong>in</strong> and 5-fluorouracil (PELF) is more active than 5fluorouracil,<br />

doxorubic<strong>in</strong> and methotrexate (FAMTX) <strong>in</strong> advanced gastric carc<strong>in</strong>oma. Annals of<br />

Oncology, 2003. 14(8): p. 1258-63.<br />

318. Koizumi, W., et al., Randomized phase II study compar<strong>in</strong>g mitomyc<strong>in</strong>, cisplat<strong>in</strong> plus doxiflurid<strong>in</strong>e <strong>with</strong><br />

cisplat<strong>in</strong> plus doxiflurid<strong>in</strong>e <strong>in</strong> advanced unresectable gastric cancer. Anticancer Research, 2004.<br />

24(4): p. 2465-70.<br />

319. Lutz, M.P., et al., Weekly <strong>in</strong>fusional high-dose fluorouracil (HD-FU), HD-FU plus fol<strong>in</strong>ic acid (HD-<br />

FU/FA), or HD-FU/FA plus biweekly cisplat<strong>in</strong> <strong>in</strong> advanced gastric cancer: randomized phase II trial<br />

40953 of the European Organisation for Research and Treatment of Cancer Gastro<strong>in</strong>test<strong>in</strong>al Group<br />

and the Arbeitsgeme<strong>in</strong>schaft Internistische Onkologie. Journal of Cl<strong>in</strong>ical Oncology, 2007. 25(18): p.<br />

2580-5.<br />

320. Moehler, M., et al., Randomised phase II evaluation of ir<strong>in</strong>otecan plus high-dose 5-fluorouracil and<br />

leucovor<strong>in</strong> (ILF) vs 5-fluorouracil, leucovor<strong>in</strong>, and etoposide (ELF) <strong>in</strong> untreated metastatic gastric cancer.<br />

British Journal of Cancer, 2005. 92(12): p. 2122-8.<br />

321. Ohtsu, A., et al., Randomized phase III trial of fluorouracil alone versus fluorouracil plus cisplat<strong>in</strong><br />

versus uracil and tegafur plus mitomyc<strong>in</strong> <strong>in</strong> patients <strong>with</strong> unresectable, advanced gastric cancer: The<br />

Japan Cl<strong>in</strong>ical Oncology Group Study (JCOG9205). Journal of Cl<strong>in</strong>ical Oncology, 2003. 21(1): p. 54-<br />

9.<br />

322. Park, S.H., et al., Paclitaxel versus docetaxel for advanced gastric cancer: a randomized phase II trial<br />

<strong>in</strong> comb<strong>in</strong>ation <strong>with</strong> <strong>in</strong>fusional 5-fluorouracil. Anti-Cancer Drugs, 2006. 17(2): p. 225-9.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 63<br />

323. Pozzo, C., et al., Ir<strong>in</strong>otecan <strong>in</strong> comb<strong>in</strong>ation <strong>with</strong> 5-fluorouracil and fol<strong>in</strong>ic acid or <strong>with</strong> cisplat<strong>in</strong> <strong>in</strong><br />

patients <strong>with</strong> advanced gastric or esophageal-gastric junction adenocarc<strong>in</strong>oma: results of a randomized<br />

phase II study. Annals of Oncology, 2004. 15(12): p. 1773-81.<br />

324. Sadighi, S., et al., Quality of life <strong>in</strong> patients <strong>with</strong> advanced gastric cancer: a randomized trial<br />

compar<strong>in</strong>g docetaxel, cisplat<strong>in</strong>, 5-FU (TCF) <strong>with</strong> epirubic<strong>in</strong>, cisplat<strong>in</strong>, 5-FU (ECF). BMC Cancer, 2006.<br />

6: p. 274.<br />

325. Takahashi, Y., et al., A randomized phase II cl<strong>in</strong>ical trial of tailored CPT-11 + S-1 vs S-1 <strong>in</strong> patients<br />

<strong>with</strong> advanced or recurrent gastric carc<strong>in</strong>oma as the first l<strong>in</strong>e chemotherapy. Japanese Journal of<br />

Cl<strong>in</strong>ical Oncology, 2004. 34(6): p. 342-5.<br />

326. Thuss-Patience, P.C., et al., Docetaxel and cont<strong>in</strong>uous-<strong>in</strong>fusion fluorouracil versus epirubic<strong>in</strong>, cisplat<strong>in</strong>,<br />

and fluorouracil for advanced gastric adenocarc<strong>in</strong>oma: a randomized phase II study. Journal of<br />

Cl<strong>in</strong>ical Oncology, 2005. 23(3): p. 494-501.<br />

327. Bonneta<strong>in</strong>, F., et al., Longitud<strong>in</strong>al quality of life study <strong>in</strong> patients <strong>with</strong> metastatic gastric cancer.<br />

Analysis modalities and cl<strong>in</strong>ical applicability of QoL <strong>in</strong> randomized phase II trial <strong>in</strong> a digestive oncology.<br />

Gastroenterologie Cl<strong>in</strong>ique et Biologique, 2005. 29(11): p. 1113-24.<br />

328. Van Cutsem, E., et al., Phase III study of docetaxel and cisplat<strong>in</strong> plus fluorouracil compared <strong>with</strong><br />

cisplat<strong>in</strong> and fluorouracil as first-l<strong>in</strong>e therapy for advanced gastric cancer: a report of the V325 Study<br />

Group. Journal of Cl<strong>in</strong>ical Oncology, 2006. 24(31): p. 4991-7.<br />

329. Dormann, A., et al., Self-expand<strong>in</strong>g metal stents for gastroduodenal malignancies: systematic review<br />

of their cl<strong>in</strong>ical effectiveness. Endoscopy, 2004. 36(6): p. 543-50.<br />

330. Fiori, E., et al., Palliative management of malignant antro-pyloric strictures. Gastroenterostomy vs.<br />

endoscopic stent<strong>in</strong>g. A randomized prospective trial. Anticancer Research, 2004. 24(1): p. 269-71.<br />

331. Mehta, S., et al., Prospective randomized trial of laparoscopic gastrojejunostomy versus duodenal<br />

stent<strong>in</strong>g for malignant gastric outflow obstruction. Surgical Endoscopy, 2006. 20(2): p. 239-42.<br />

332. Weston, A.P., et al., Specificity of polymerase cha<strong>in</strong> reaction monoclonality for diagnosis of gastric<br />

mucosa-associated lymphoid tissue (MALT) lymphoma: direct comparison to Southern blot gene<br />

rearrangement. Dig Dis Sci, 1998. 43(2): p. 290-9.<br />

333. Palazzo, L., et al., Endoscopic ultrasonography <strong>in</strong> the local stag<strong>in</strong>g of primary gastric lymphoma.<br />

Endoscopy, 1993. 25(8): p. 502-8.<br />

334. Ferreri, A.J., et al., Low sensitivity of computed tomography <strong>in</strong> the stag<strong>in</strong>g of gastric lymphomas of<br />

mucosa-associated lymphoid tissue: impact on prospective trials and ord<strong>in</strong>ary cl<strong>in</strong>ical practice. Am J<br />

Cl<strong>in</strong> Oncol, 1998. 21(6): p. 614-6.<br />

335. Fusaroli, P., et al., Interobserver agreement <strong>in</strong> stag<strong>in</strong>g gastric malt lymphoma by EUS. Gastro<strong>in</strong>test<br />

Endosc, 2002. 55(6): p. 662-8.<br />

336. Kataoka, M., et al., The role of gallium 67 imag<strong>in</strong>g <strong>in</strong> non-Hodgk<strong>in</strong>'s lymphoma of the gastro<strong>in</strong>test<strong>in</strong>al<br />

tract. European Journal of Nuclear Medic<strong>in</strong>e, 1990. 17(3-4): p. 142-7.<br />

337. Haim, N., et al., Intermediate and high-grade gastric non-Hodgk<strong>in</strong>'s lymphoma: a prospective study of<br />

non-surgical treatment <strong>with</strong> primary chemotherapy, <strong>with</strong> or <strong>with</strong>out radiotherapy. Leuk Lymphoma,<br />

1995. 17(3-4): p. 321-6.<br />

338. Takenaka, T., et al., A prospective study of surgery and adjuvant chemotherapy for primary gastric<br />

lymphoma stage II. Br J Cancer, 1997. 76(11): p. 1484-8.<br />

339. Kochi, M., et al., Complete remission by chemotherapy <strong>in</strong> stage IE-IIE primary gastric lymphoma.<br />

Hepatogastroenterology, 2007. 54(76): p. 1285-8.<br />

340. Willich, N.A., et al., Operative and conservative management of primary gastric lymphoma: <strong>in</strong>terim<br />

results of a German multicenter study. Int J Radiat Oncol Biol Phys, 2000. 46(4): p. 895-901.<br />

341. Chen, L.T., et al., Prospective study of Helicobacter pylori eradication therapy <strong>in</strong> stage I(E) high-grade<br />

mucosa-associated lymphoid tissue lymphoma of the stomach. J Cl<strong>in</strong> Oncol, 2001. 19(22): p. 4245-<br />

51.


64 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

342. Koch, P., et al., Treatment results <strong>in</strong> localized primary gastric lymphoma: data of patients registered<br />

<strong>with</strong><strong>in</strong> the German multicenter study (GIT NHL 02/96).[see comment]. J Cl<strong>in</strong> Oncol, 2005. 23(28):<br />

p. 7050-9.<br />

343. Ishikura, S., et al., Japanese multicenter phase II study of CHOP followed by radiotherapy <strong>in</strong> stage I-II,<br />

diffuse large B-cell lymphoma of the stomach. Cancer Sci, 2005. 96(6): p. 349-52.<br />

344. Aviles, A., et al., Surgery and chemotherapy versus chemotherapy as treatment of high-grade MALT<br />

gastric lymphoma. Medical Oncology, 2006. 23(2): p. 295-300.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 65<br />

8 APPENDICES<br />

APPENDIX 1<br />

GRADE SYSTEM<br />

Grade of Recommendation/ Benefit vs. Risk and Burdens Methodological Quality of<br />

Implications<br />

Description<br />

Support<strong>in</strong>g Evidence<br />

1A/ Strong recommendation, high Benefits clearly outweigh risk and RCTs <strong>with</strong>out important limitations or Strong recommendation, can apply to<br />

quality evidence<br />

burdens, or vice versa<br />

overwhelm<strong>in</strong>g evidence from<br />

most patients <strong>in</strong> most circumstances<br />

observational studies<br />

<strong>with</strong>out reservation<br />

1B/ Strong recommendation, moderate Benefits clearly outweigh risk and RCTs <strong>with</strong> important limitations Strong recommendation, can apply to<br />

quality evidence<br />

burdens, or vice versa<br />

(<strong>in</strong>consistent results, methodological most patients <strong>in</strong> most circumstances<br />

flaws, <strong>in</strong>direct, or imprecise) or<br />

exceptionally strong evidence from<br />

observational studies<br />

<strong>with</strong>out reservation<br />

1C/ Strong recommendation, low quality Benefits clearly outweigh risk and Observational studies or case series Strong recommendation, but may<br />

evidence<br />

burdens, or vice versa<br />

change when higher quality evidence<br />

becomes available<br />

2A/ Weak recommendation, high quality Benefits closely balanced <strong>with</strong> risks and RCTs <strong>with</strong>out important limitations or Weak recommendation, best action may<br />

evidence<br />

burden<br />

overwhelm<strong>in</strong>g evidence from<br />

differ depend<strong>in</strong>g on circumstances or<br />

observational studies<br />

patients’ or societal values<br />

2B/ Weak recommendation, moderate Benefits closely balanced <strong>with</strong> risks and RCTs <strong>with</strong> important limitations Weak recommendation, best action may<br />

quality evidence<br />

burden<br />

(<strong>in</strong>consistent results, methodological differ depend<strong>in</strong>g on circumstances or<br />

flaws, <strong>in</strong>direct, or imprecise) or<br />

exceptionally strong evidence from<br />

observational studies<br />

patients’ or societal values<br />

2C/ Weak recommendation, low quality Benefits closely balanced <strong>with</strong> risks and Observational studies or case series Very weak recommendation, other<br />

evidence<br />

burden<br />

alternatives may be equally reasonable


66 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

APPENDIX 2<br />

IDENTIFIED GUIDELINES AND THEIR QUALITY APPRAISAL<br />

Source Title Standardised Methodology Score F<strong>in</strong>al Appraisal<br />

American Gastroenterological American Gastroenterological Association Institute Technical Review on the Management 22% Not recommended<br />

Association [212]<br />

of Gastric Subepithelial Masses<br />

American Gastroenterological American Gastroenterological Association Technical Review on the Role of the<br />

31% Not recommended<br />

Association [48]<br />

Gastroenterologist <strong>in</strong> the Management of Esophageal Carc<strong>in</strong>oma<br />

American Society for<br />

Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy [226]<br />

The role of endoscopy <strong>in</strong> the assessment and treatment of esophageal cancer 33% Not recommended<br />

American Society for<br />

Gastro<strong>in</strong>test<strong>in</strong>al Endoscopy [64]<br />

Esophageal dilation 29% Not recommended<br />

National Comprehensive Cancer<br />

Network [227]<br />

Esophageal Cancer 57% Not recommended<br />

National Comprehensive Cancer<br />

Network [228]<br />

Gastric Cancer 57% Not recommended<br />

Scottish Intercollegiate Guidel<strong>in</strong>es<br />

Network [47]<br />

Management of oesophageal and gastric cancer 95% Recommended<br />

Kwaliteits<strong>in</strong>stituut voor de<br />

Gezondheidszorg [56]<br />

Diagnostiek en behandel<strong>in</strong>g oesofaguscarc<strong>in</strong>oom 93% Recommended <strong>with</strong> alterations<br />

Cancer Care Ontario [84] Neoadjuvant or Adjuvant Therapy for Resectable Esophageal Cancer. Practice Guidel<strong>in</strong>e<br />

<strong>Report</strong> #2-11.<br />

93% Recommended <strong>with</strong> alterations<br />

Cancer Care Ontario [108] Comb<strong>in</strong>ed Modality Radiotherapy and Chemotherapy <strong>in</strong> the Non-surgical Management of<br />

Localized Carc<strong>in</strong>oma of the Esophagus. Practice Guidel<strong>in</strong>e <strong>Report</strong> #2-12.<br />

93% Recommended <strong>with</strong> alterations<br />

Cancer Care Ontario [150] Neoadjuvant or Adjuvant Therapy for Resectable Gastric Cancer. Practice Guidel<strong>in</strong>e<br />

<strong>Report</strong> #2-14.<br />

93% Recommended <strong>with</strong> alterations<br />

Fédération Nationale des Centres Recommandations pour la pratique cl<strong>in</strong>ique: Standards, Options et Recommandations 2003 86% Recommended <strong>with</strong> alterations<br />

de Lutte Contre le Cancer [129] pour la prise en charge des patients atte<strong>in</strong>ts<br />

d’adénocarc<strong>in</strong>omes de l’estomac (cancers du cardia, autres types histologiques exclus)<br />

(rapport <strong>in</strong>tégral)<br />

Fédération Nationale des Centres Recommandations pour la pratique cl<strong>in</strong>ique: Standards, Options et Recommandations 2003 86% Recommended <strong>with</strong> alterations<br />

de Lutte Contre le Cancer [66] pourl’utilisation de la tomographie par émission de positons au [18F]-FDG (TEP-FDG) en<br />

cancérologie (rapport <strong>in</strong>tégral)<br />

European Society for Medical ESMO M<strong>in</strong>imum Cl<strong>in</strong>ical Recommendations for diagnosis, treatment and follow-up of 24% Not recommended<br />

Oncology [229]<br />

gastric cancer<br />

European Society for Medical ESMO M<strong>in</strong>imal Cl<strong>in</strong>ical Recommendations for diagnosis, treatment and follow-up of 24% Not recommended<br />

Oncology [230]<br />

esophageal cancer<br />

American College of<br />

Gastroenterology [231]<br />

Updated Guidel<strong>in</strong>es for the Diagnosis, Surveillance, and Therapy of Barrett’s Esophagus 27% Not recommended<br />

Allum W et al. [232] Guidel<strong>in</strong>es for the management of oesophageal and gastric cancer 46% Not recommended


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 67<br />

APPENDIX 3<br />

OVERVIEW OF SCORES OF EXTERNAL EXPERTS


68 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 69


70 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 71


72 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 73


74 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 75


76 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 77<br />

APPENDIX 4<br />

EVIDENCE TABLES OF OESOPHAGEAL CANCER BY CLINICAL QUESTION<br />

CPG ID Ref Search<br />

date<br />

Alarm symptoms<br />

Diagnosis of oesophageal cancer.<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

SIGN [47] 2004 Patients present<strong>in</strong>g <strong>with</strong> any of the follow<strong>in</strong>g alarm symptoms should be referred for early endoscopy:<br />

dysphagia, recurrent vomit<strong>in</strong>g, anorexia, weight loss, gastro<strong>in</strong>test<strong>in</strong>al blood loss.<br />

CBO [56] July 2003 Alarmsymptomen, zoals haematemesis en/of melaena, en maagklachten <strong>in</strong> comb<strong>in</strong>atie met aanhoudend<br />

braken, passagestoornissen, ongewild gewichtsverlies of anemie, vormen een <strong>in</strong>dicatie voor een<br />

endoscopie.<br />

Bij het besluit om een endoscopie te laten verrichten speelt leeftijd een beperkte rol.<br />

Oesophagogastroscopy<br />

SIGN [47] 2004 Flexible upper GI endoscopy is recommended as the diagnostic procedure of choice <strong>in</strong> patients <strong>with</strong><br />

suspected oesophageal cancer.<br />

Rout<strong>in</strong>e use of chromoendoscopy dur<strong>in</strong>g upper GI endoscopy is not recommended, but may be of value<br />

<strong>in</strong> selected patients at high risk of oesophageal malignancy.<br />

Barium X-ray<br />

SIGN [47] 2004 No recommendation, only discussion<br />

Biopsy<br />

Me<strong>in</strong>eche-Schmidt V 2002<br />

Numans ME 2001<br />

Wallace MB 2001<br />

Kapoor N 2005<br />

Gillen D 1999<br />

Me<strong>in</strong>eche-Schmidt V 2002<br />

Numans ME 2001<br />

Wallace MB 2001<br />

Graham DY 1982<br />

Abbas SZ 2004<br />

Inone H 2001<br />

Ragunath K 2003<br />

Kiesslich R 2001<br />

Canto MI 2001<br />

SIGN [47] 2004 A m<strong>in</strong>imum of eight biopsies should be taken to achieve a diagnosis of oesophageal malignancy. Lal N 1992 Low<br />

Low<br />

Low<br />

Low<br />

High


78 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Alarm symptoms<br />

Bowrey DJ 2006 [49] NA Patients referred for<br />

open-access gastroscopy<br />

Marmo R 2005 [233] NA Patients <strong>with</strong><br />

uncomplicated dyspepsia<br />

Oesophagogastroscopy<br />

No additional studies found<br />

Barium X-ray<br />

No additional studies found<br />

Biopsy<br />

No additional studies found<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Application of referral guidel<strong>in</strong>es<br />

us<strong>in</strong>g alarm symptoms<br />

Endoscopy<br />

Identification of risk factors<br />

Gastroscopy identified oesophagogastric<br />

carc<strong>in</strong>oma <strong>in</strong> 123 (3%) of the 4,018 subjects. Of<br />

these 123 patients, 104 (85%) <strong>with</strong><br />

oesophagogastric cancer had<br />

‘‘alarm’’ symptoms (anemia, mass, dysphagia,<br />

weight loss, vomit<strong>in</strong>g) and would have satisfied<br />

the referral criteria. The rema<strong>in</strong><strong>in</strong>g 15% would<br />

not have been referred for <strong>in</strong>itial endoscopic<br />

assessment because their symptoms were those<br />

of uncomplicated ‘‘benign’’ dyspepsia.<br />

The patients <strong>with</strong> ‘‘alarm’’ symptoms had a<br />

significantly more advanced tumor stage<br />

(metastatic disease <strong>in</strong> 47% vs 11%; p < 0.001),<br />

were less likely to undergo surgical resection<br />

(50% vs 95%; p


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 79<br />

CPG ID<br />

CT scan<br />

Ref Search<br />

date<br />

Stag<strong>in</strong>g of patients <strong>with</strong> oesophageal cancer.<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

SIGN [47] 2004 In patients <strong>with</strong> oesophageal cancer, CT scan of the chest and abdomen <strong>with</strong> IV contrast and gastric<br />

distention <strong>with</strong> oral contrast or water should be performed rout<strong>in</strong>ely. The liver should be imaged <strong>in</strong> the<br />

portal venous phase.<br />

CBO [56] July 2003 CT van de thoraxapertuur tot en met de bovenbuik kan worden gebruikt om de aanwezigheid van<br />

mediast<strong>in</strong>ale en/of truncale lymfkliermetastasen, alsook mogelijke metastasen <strong>in</strong> longen, lever, bijnieren of<br />

skelet vast te stellen. Hierbij dient zowel ger<strong>in</strong>g oraal contrast ter marker<strong>in</strong>g van het oesofaguslumen als<br />

<strong>in</strong>traveneus contrast te worden toegepast. De coupedikte dient kle<strong>in</strong>er dan of gelijk aan 5 mm te zijn.<br />

Endoscopic<br />

ultrasonography<br />

SIGN [47] 2004 Patients <strong>with</strong> oesophageal or oesophagogastric junction cancers who are candidates for any curative<br />

therapy should have their tumours staged <strong>with</strong> endoscopic US +/- f<strong>in</strong>e needle aspiration.<br />

CBO [56] July 2003 Endoscopische ultrasonografie is z<strong>in</strong>vol om de uitbreid<strong>in</strong>g van de laesie <strong>in</strong> de diepte en mogelijke <strong>in</strong>groei <strong>in</strong><br />

omgevende structuren (bijvoorbeeld aorta) (T-status) en de aanwezigheid van vergrote mediast<strong>in</strong>ale en/of<br />

truncale lymfklieren te bepalen (N- en M-status).<br />

Laparoscopy<br />

Kamel IR 2004 Low<br />

Romagnuolo J 2002<br />

Wakel<strong>in</strong> SJ 2002<br />

van Overhagen H 1993<br />

van den Hoed RD 1997<br />

Kelly S 2001 (SR)<br />

Vazquez-Sequeiros E 2003<br />

Weaver SR 2004<br />

Eloubeidi MA 2001<br />

Catalano MF 1995<br />

Vazquez-Sequeiros E 2003<br />

Heidemann J 2000<br />

Wiersema MJ 1997<br />

Williams DB 1999<br />

SIGN [47] 2004 Laparoscopy should be considered <strong>in</strong> patients <strong>with</strong> oesophageal tumours <strong>with</strong> a gastric component. Clements DM 2004<br />

Molloy RG 1995<br />

CBO [56] July 2003 M<strong>in</strong>imaal <strong>in</strong>vasieve chirurgische stadiër<strong>in</strong>g met behulp van thoracoscopie en laparoscopie is geïndiceerd bij<br />

afwijk<strong>in</strong>gen <strong>in</strong> de oesofagus van onduidelijke aard, gevonden bij niet-<strong>in</strong>vasieve stadiër<strong>in</strong>g. Voorts kan<br />

laparoscopie worden overwogen bij carc<strong>in</strong>omen op de gastro-oesofageale overgang en cardiacarc<strong>in</strong>omen<br />

die als T3/T4 worden gestadieerd met een niet-<strong>in</strong>vasieve techniek.<br />

Suspecte cervicale, mediast<strong>in</strong>ale en truncale klieren dienen onder beeldvorm<strong>in</strong>g ((endoscopische)<br />

ultrasonografie) cytologisch te worden gepuncteerd <strong>in</strong>dien de status van de desbetreffende lymfklieren<br />

voor beleidsbepal<strong>in</strong>g relevant is.<br />

Magnetic resonance<br />

imag<strong>in</strong>g<br />

SIGN [47] 2004 MRI should be reserved for those patients who cannot undergo CT or used for additional <strong>in</strong>vestigation<br />

follow<strong>in</strong>g CT/EUS.<br />

Wilkiemeyer MB 2004<br />

Krasna MJ 1995 & 2002<br />

Whyte RI 2001<br />

Wakel<strong>in</strong> SJ 2002<br />

Romijn MG 1998<br />

Bonvalot S 1996<br />

Natsugoe S 1999<br />

van Overhagen H 1993<br />

Sohn KM 2000<br />

Wu LF 2003<br />

Kersjes 1997<br />

Lauenste<strong>in</strong> TC 2004<br />

Wong R 2000<br />

Low<br />

Moderate<br />

Low<br />

Low<br />

Low<br />

Low<br />

Low


80 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Search<br />

date<br />

Bronchoscopy<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

SIGN [47] 2004 Bronchoscopy +/- bronchoscopic US +/- biopsy should be undertaken <strong>in</strong> patients <strong>with</strong> cl<strong>in</strong>ical or imag<strong>in</strong>g<br />

features suspicious of tracheobronchial <strong>in</strong>vasion.<br />

Aggarwal AN 2003<br />

Nguyen NT 2001<br />

Nishimura Y 2002<br />

Riedel M 2001<br />

CBO [56] July 2003 Bronchoscopie, <strong>in</strong>clusief brush-cytologie, van suspecte gebieden is onderdeel van de stadiër<strong>in</strong>g van Baisi A 1999<br />

Low<br />

oesofaguscarc<strong>in</strong>omen op of craniaal van het niveau van de car<strong>in</strong>a.<br />

Choi TK 1984<br />

Thoracoscopy<br />

SIGN [47] 2004 Thoracoscopy may be considered for patients where a tissue diagnosis of suspicious nodes (not possible<br />

by either EUS or CT guided techniques) is required to determ<strong>in</strong>e optimum management.<br />

Krasna MJ 2002 Low<br />

CBO [56] July 2003 M<strong>in</strong>imaal <strong>in</strong>vasieve chirurgische stadiër<strong>in</strong>g met behulp van thoracoscopie en laparoscopie is geïndiceerd bij Krasna MJ 1995 & 2002 Low<br />

afwijk<strong>in</strong>gen <strong>in</strong> de oesofagus van onduidelijke aard, gevonden bij niet-<strong>in</strong>vasieve stadiër<strong>in</strong>g. Voorts kan Whyte RI 2001<br />

laparoscopie worden overwogen bij carc<strong>in</strong>omen op de gastro-oesofageale overgang en cardiacarc<strong>in</strong>omen Wakel<strong>in</strong> SJ 2002<br />

die als T3/T4 worden gestadieerd met een niet-<strong>in</strong>vasieve techniek.<br />

Romijn MG 1998<br />

Suspecte cervicale, mediast<strong>in</strong>ale en truncale klieren dienen onder beeldvorm<strong>in</strong>g ((endoscopische) Bonvalot S 1996<br />

Low<br />

ultrasonografie) cytologisch te worden gepuncteerd <strong>in</strong>dien de status van de desbetreffende lymfklieren Natsugoe S 1999<br />

PET scan<br />

voor beleidsbepal<strong>in</strong>g relevant is.<br />

van Overhagen H 1993<br />

SIGN [47] 2004 PET is not rout<strong>in</strong>ely <strong>in</strong>dicated <strong>in</strong> the stag<strong>in</strong>g of oesophageal cancers. van Westreenen HL 2004 Low<br />

CBO [56] July 2003 FDG-PET bij T3-oesofaguscarc<strong>in</strong>omen kan worden overwogen om eventuele afstandsmetastasen vast te<br />

stellen.<br />

van Westreenen HL 2004 Low<br />

FNCLCC [66] October La TEP-FDG est <strong>in</strong>diquée, est complémentaire du scanner et de l’écho-endoscopie, pour l’évaluation K<strong>in</strong>kel 2002<br />

Low<br />

Neck imag<strong>in</strong>g<br />

2002 préthérapeutique du statut ganglionnaire et métastatique des cancers de l’oesophage.<br />

Depotter 2002<br />

McAteer 1999<br />

Hoffmann 1999<br />

Couper 1998<br />

SIGN [47] 2004 Neck imag<strong>in</strong>g either by US or CT is recommended as part of the stag<strong>in</strong>g of oesophageal cancer. Griffith JF 2000 Low<br />

CBO [56] July 2003 Echografie van de hals is nuttig om cervicale lymfkliermetastasen te bepalen. Bonvalot S 1996<br />

Natsugoe S 1999<br />

van Overhagen H 1993<br />

Low<br />

Suspecte cervicale, mediast<strong>in</strong>ale en truncale klieren dienen onder beeldvorm<strong>in</strong>g ((endoscopische) Bonvalot S 1996<br />

Low<br />

ultrasonografie) cytologisch te worden gepuncteerd <strong>in</strong>dien de status van de desbetreffende lymfklieren Natsugoe S 1999<br />

voor beleidsbepal<strong>in</strong>g relevant is.<br />

van Overhagen H 1993<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 81<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

CT scan<br />

Kim SH 2006 [234] NA Patients <strong>with</strong> suspected or<br />

known oesophageal<br />

neoplasms (n = 23)<br />

Umeoka S 2006 [235] NA 28 consecutive patients<br />

<strong>with</strong> oesophageal cancer<br />

histopathologically proved<br />

at endoscopic biopsy (31<br />

lesions)<br />

Panebioanco V 2006 [236] NA 39 patients <strong>with</strong><br />

oesophageal cancer<br />

proved by means of<br />

endoscopy<br />

Onbas O 2006 [237] NA 44 patients <strong>with</strong><br />

oesophageal cancer<br />

diagnosed by conventional<br />

endoscopy and biopsy<br />

specimens<br />

Lowe VJ 2005 [59] NA 69 patients <strong>with</strong> newly<br />

diagnosed oesophageal<br />

cancer<br />

3D Multidetector CT and CT<br />

oesophagography (n = 23)<br />

Conventional barium study (n<br />

= 20)<br />

Conventional endoscopy (n =<br />

23)<br />

Histology (surgery: n = 12;<br />

endoscopic biopsy: n = 11)<br />

Triple-phase dynamic<br />

contrast-enhanced CT<br />

Standard: histopathology (23<br />

lesions) or EUS (8 lesions)<br />

Dynamic CT of the chest and<br />

abdomen <strong>with</strong> the aid of 3D<br />

render<strong>in</strong>g<br />

Standard: histopathology (26<br />

patients)<br />

Multidetector CT (n = 24)<br />

Virtual endoscopy (n = 27)<br />

Standard: histopathology (n =<br />

24)<br />

FDG-PET<br />

CT<br />

EUS<br />

Histology (endoscopic biopsy:<br />

n = 55; surgery n = 14)<br />

Sensitivity MDCT: 91%<br />

Overall accuracies for T and N stag<strong>in</strong>g<br />

were 42.9% and 85.7% respectively<br />

One lesion not identifiable at CT<br />

Insufficient data to calculate accuracy<br />

T stag<strong>in</strong>g: sensitivity 92%, accuracy 88%<br />

N stag<strong>in</strong>g: sensitivity 85%, specificity<br />

58%, accuracy 69%<br />

MDCT: correct T stag<strong>in</strong>g <strong>in</strong> 22/24<br />

patients, overstag<strong>in</strong>g <strong>in</strong> 1 pt and<br />

understag<strong>in</strong>g <strong>in</strong> 1 pt; correct N stag<strong>in</strong>g <strong>in</strong><br />

20/24 patients, overstag<strong>in</strong>g <strong>in</strong> 2 pts and<br />

understag<strong>in</strong>g <strong>in</strong> 2 pts<br />

Correct T stag<strong>in</strong>g: 42% <strong>with</strong> PET and<br />

CT, 71% <strong>with</strong> EUS<br />

Sensitivity and specificity for nodal<br />

<strong>in</strong>volvement: 84% and 67% for CT, 86%<br />

and 67% for EUS, 82% and 60% for PET<br />

Sensitivity and specificity for distant<br />

metastasis: 81% and 82% for CT, 73%<br />

and 86% for EUS, and 81% and 91% for<br />

PET<br />

Not very clear if<br />

prospective<br />

Same standard not<br />

applied to all patients<br />

Standard not applied to<br />

all patients<br />

Observationa<br />

l study<br />

Prospective<br />

observational<br />

study<br />

Prospective<br />

observational<br />

study<br />

Prospective<br />

observational<br />

study<br />

Prospective<br />

observational<br />

study<br />

Very low<br />

Very low<br />

Very low<br />

Very low<br />

Low


82 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

Weaver SR 2004 [238] NA 60 consecutive patients Spiral CT<br />

Sensitivity for T and N stages was 58% Included <strong>in</strong> SIGN Prospective Low<br />

<strong>with</strong> histologically proven EUS<br />

and 79% for CT, and 72% and 91% for guidel<strong>in</strong>e<br />

observational<br />

cancer of the oesophagus, Standard: histology<br />

EUS. Specificity for T and N stages was<br />

study<br />

established by endoscopy<br />

80% and 84% for CT, and 85% and 68%<br />

and biopsy, that<br />

underwent surgery<br />

for EUS.<br />

Yoon YC 2003 [239] NA 81 patients <strong>with</strong> squamous CT and FDG-PET<br />

T stag<strong>in</strong>g: primary tumour was correctly Strong selection of 136 Prospective Very low<br />

cell carc<strong>in</strong>oma of the Standard: histopathology detected <strong>in</strong> 65 patients (80%) at CT and patients <strong>with</strong> oesophageal observational<br />

oesophagus<br />

<strong>in</strong> 74 patients (91%) at FDG PET cancer<br />

study<br />

N stag<strong>in</strong>g:<br />

Included <strong>in</strong> SR of Van<br />

CT: sensitivity 11%, specificity 95%<br />

PET: sensitivity 30%, specificity 90%<br />

Westreenen et al.<br />

Endoscopic ultrasonography<br />

van Vliet EP 2007 [61] February<br />

2006<br />

Vazquez-Sequeiros E<br />

2006<br />

Patients <strong>with</strong> oesophageal<br />

cancer<br />

[240] NA Patients (n = 144) <strong>with</strong><br />

adenocarc<strong>in</strong>oma or<br />

squamous-cell carc<strong>in</strong>oma<br />

of the oesophagus, M0<br />

(helical CT) and who<br />

were candidates for<br />

surgical resection; no<br />

neoadjuvant treatment<br />

May A 2004 [241] NA Patients <strong>with</strong> a suspicion<br />

of early oesophageal<br />

adenocarc<strong>in</strong>oma (n = 81)<br />

or squamous cell<br />

carc<strong>in</strong>oma (n = 19)<br />

Yanai H 2004 [242] NA Patients <strong>with</strong><br />

endoscopically staged<br />

EUS T stag<strong>in</strong>g: median accuracy 83% (53-<br />

94%)<br />

N stag<strong>in</strong>g: median accuracy 76% (54-<br />

94%), median sensitivity 77% (37-100%),<br />

median specificity 74% (50-90%)<br />

Helical CT<br />

EUS (standard vs. modified<br />

criteria)<br />

EUS-FNAC<br />

Standard: surgical pathology<br />

result (n = 47) or malignant<br />

cytology result (n = 97)<br />

High-resolution video<br />

endoscopy<br />

HR-EUS <strong>with</strong> a 20 MHz<br />

m<strong>in</strong>iprobe<br />

Standard: histological<br />

exam<strong>in</strong>ation of the<br />

endoscopically or surgically<br />

resected tumour<br />

20-MHz th<strong>in</strong> ultrasound probe<br />

EUS<br />

No evidence for publication bias<br />

Presence of all 7 modified criteria:<br />

specificity and PPV of 100%<br />

Presence of no modified criteria:<br />

sensitivity and NPV of 100%<br />

Accuracy of stag<strong>in</strong>g:<br />

HR-E 83.4% vs. HR-EUS 79.6%<br />

Mucosal tumours:<br />

Sensitivity HR-E 94.1% vs. HR-EUS<br />

91.2%<br />

Submucosal tumours:<br />

Sensitivity HR-E 56% vs. HR-EUS 48%<br />

Comb<strong>in</strong>ation of HR-E and HR-EUS: 60%<br />

Invasion depth correctly staged <strong>in</strong> 20/31<br />

pts (64.5%)<br />

54 articles <strong>in</strong>cluded<br />

Medl<strong>in</strong>e search only<br />

<strong>English</strong> only<br />

No quality appraisal<br />

Bl<strong>in</strong>ded evaluation<br />

Standard not the same for<br />

all patients<br />

Systematic<br />

review<br />

Prospective<br />

observational<br />

study<br />

Bl<strong>in</strong>ded evaluation Prospective<br />

observational<br />

study<br />

In calculation exclusion of<br />

2 dysplasia pts and 2 non-<br />

Prospective<br />

observational<br />

Low<br />

Low<br />

Low<br />

Very low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 83<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

superficial oesophageal Standard: histological<br />

resection cases study<br />

cancer (n = 31)<br />

exam<strong>in</strong>ation of the<br />

endoscopically or surgically<br />

resected tumour<br />

Chang KJ 2003 [243] NA Patients <strong>with</strong> oesophageal CT<br />

T stag<strong>in</strong>g: accuracy EUS 83%<br />

Standard not applied to Prospective Very low<br />

cancer (n = 60)<br />

EUS<br />

all patients<br />

observational<br />

EUS-FNAC if positive FNAC N stag<strong>in</strong>g:<br />

2 pts lost-to-follow-up study<br />

would upstage the tumour Accuracy EUS 83%<br />

Standard: surgical pathology<br />

result<br />

Accuracy EUS-FNAC 89%, PPV 100%<br />

Mariette C 2003 [244] NA 150 patients who EUS<br />

Patient survival was significantly related Doubts about prospective Prospective Very low<br />

underwent EUS for stag<strong>in</strong>g Standard: surgical pathology to tumour (EUS T), node (EUS N) and design<br />

(?)<br />

of tumours of the result<br />

Union Internacional Contra la Cancrum Not enough data to observational<br />

oesophagus (out of 372<br />

classification (EUS UICC) stage calculate stag<strong>in</strong>g accuracy study<br />

patients who underwent<br />

accord<strong>in</strong>g to sonographic f<strong>in</strong>d<strong>in</strong>gs (P =<br />

surgery <strong>with</strong> curative<br />

0·003, P = 0·009 and P = 0·004<br />

<strong>in</strong>tent)<br />

respectively), and the presence of<br />

stenosis determ<strong>in</strong>ed by EUS (P = 0·004).<br />

EUS T stage was a prognostic factor for<br />

survival (RR 1.7, 95%CI 1.1 to 3.0; P =<br />

0·046). Complete surgical resection (R0)<br />

was also significantly related to EUS T,<br />

N and UICC classification (P < 0·001).<br />

EUS UICC stage was a factor predictive<br />

of R0 resection (RR 2.6, 95%CI 1.4 to<br />

4.8; P = 0·003).<br />

Laparoscopy<br />

Mortensen MB 2006 [79] NA 411 patients <strong>with</strong> upper<br />

gastro<strong>in</strong>test<strong>in</strong>al tract<br />

cancer, of which 95 had<br />

biopsy-proven<br />

oesophageal cancer<br />

Comb<strong>in</strong>ed endoscopic<br />

ultrasonography (EUS) and<br />

laparoscopic ultrasonography<br />

(LUS)<br />

‘Standard’: treatment actually<br />

undertaken<br />

The comb<strong>in</strong>ation of EUS and LUS<br />

correctly predicted R0 resection <strong>in</strong><br />

90.6%, R1–R2 <strong>in</strong> 91% and irresectability<br />

<strong>in</strong> 91.4% of patients. Ten patients (2.4%)<br />

had explorative laparotomy only. There<br />

were no complications associated <strong>with</strong><br />

the EUS and LUS procedures.<br />

To avoid false-positive<br />

f<strong>in</strong>d<strong>in</strong>gs <strong>with</strong> regard to<br />

local irresectability<br />

ultrasonographic criteria<br />

were adjusted so that<br />

only a comb<strong>in</strong>ation of<br />

several ultrasonographic<br />

criteria <strong>with</strong> a 100 per<br />

cent predictive value for<br />

local<br />

irresectability were used<br />

Standard is not identical<br />

Prospective<br />

study<br />

Very low


84 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

for all patients<br />

Menon KV 2003 [245] NA 133 out of 320<br />

consecutive patients <strong>with</strong><br />

histologically proven<br />

carc<strong>in</strong>oma of the<br />

oesophagus or cardia,<br />

<strong>with</strong>out cl<strong>in</strong>ical or<br />

radiological evidence (CT)<br />

of metastases and deemed<br />

fit for potential excisional<br />

surgery<br />

Magnetic resonance imag<strong>in</strong>g<br />

Riddell AM 2006<br />

Bronchoscopy<br />

[246] NA 10 patients <strong>with</strong><br />

confirmed oesophageal<br />

carc<strong>in</strong>oma (7 <strong>with</strong><br />

adenocarc<strong>in</strong>oma, 2 <strong>with</strong><br />

squamous cell carc<strong>in</strong>oma,<br />

and 1 <strong>with</strong> sp<strong>in</strong>dle-cell<br />

melanoma),<br />

who were deemed<br />

medically fit for surgery<br />

and shown to have<br />

resectable disease us<strong>in</strong>g<br />

endoscopic sonography<br />

and CT<br />

Wakamatsu T 2006 [77] NA 54 surgical patients <strong>with</strong><br />

suspected thyroid or<br />

oesophageal cancer (n =<br />

Multiport laparoscopy<br />

Standard: histology or<br />

subsequent surgery<br />

High-resolution MRI of<br />

primary tumour<br />

Standard: histology<br />

CT neck/upper mediast<strong>in</strong>um<br />

MRI neck/upper mediast<strong>in</strong>um<br />

Bronchoscopy <strong>with</strong> EBUS<br />

Laparoscopy detected <strong>in</strong>curable disease<br />

<strong>in</strong> 31 patients (24%), some of whom had<br />

more than one contra<strong>in</strong>dication to<br />

surgery, <strong>in</strong>clud<strong>in</strong>g hepatic metastases (n<br />

= 10), peritoneal metastases (n = 12)<br />

and<br />

malignant small volume ascites (n = 5).<br />

Lymph node metastases were confirmed<br />

histologically by biopsy at laparoscopy <strong>in</strong><br />

26 patients (fixed nodes, n = 14; mobile<br />

nodes, n = 12). Sensitivity for the<br />

detection of liver and peritoneal<br />

metastases was 100%, and lymph node<br />

metastases were 83%. Specificity for<br />

detection of hepatic metastases was<br />

99%, 100% for peritoneal metastases<br />

and 82% for lymph node metastases.<br />

N<strong>in</strong>ety-n<strong>in</strong>e patients proceeded to<br />

def<strong>in</strong>itive surgery and only two were<br />

unresectable.<br />

50 MRI images (5 per patient)<br />

7 images underestimated, 20 images<br />

overestimated<br />

Tumor position was correctly diagnosed<br />

by MRI <strong>in</strong> 5 patients<br />

Extramural extension: 2 false negatives<br />

The sensitivity and specificity of CT, MRI<br />

and EBUS for <strong>in</strong>vasion were 59 and 56,<br />

75 and 73, and 92 and 83%, respectively.<br />

Standard is not identical<br />

for all patients<br />

Prospective<br />

study<br />

Very small sample size Prospective<br />

study<br />

No separate numbers for<br />

oesophageal cancer<br />

patients<br />

Prospective<br />

study<br />

Very low<br />

Very low<br />

Very low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 85<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

24) and a mass adjacent to Standard: histology and/or The accuracy of EBUS was significantly Standard not same for all<br />

the tracheobronchial wall cl<strong>in</strong>ical follow-up<br />

greater than that of CT <strong>in</strong> the present patients<br />

on chest or neck CT or<br />

study (p = 0.0011). The accuracy of<br />

MRI<br />

EBUS was significantly different from<br />

that of CT and MRI <strong>in</strong> the surgically<br />

treated patients (p = 0.005 and p =<br />

0.032, respectively).<br />

Osugi H 2003 [76] NA Patients <strong>with</strong> advanced BUS<br />

BUS: sensitivity 91%, specificity 89%, Sensitivity and specificity Prospective Very low<br />

squamous cell carc<strong>in</strong>oma CT<br />

accuracy 90%<br />

calculated <strong>with</strong> 50 study (?)<br />

<strong>in</strong> contact <strong>with</strong> the 50 thoracotomies<br />

CT: sensitivity 69%, specificity 55%, thoracotomy patients<br />

Thoracoscopy<br />

trachea, the ma<strong>in</strong> bronchi,<br />

or both (n = 66)<br />

accuracy 62%<br />

No additional diagnostic studies found<br />

PET scan<br />

Meyers BF 2007 [247] NA Patients <strong>with</strong> resectable,<br />

biopsy-proven carc<strong>in</strong>oma<br />

of the thoracic<br />

oesophagus (n = 189)<br />

Ott K 2006 [72] NA 65 patients <strong>with</strong> biopsy<br />

proven adenocarc<strong>in</strong>oma<br />

of the distal oesophagus<br />

or cardia (stage uT3)<br />

FDG-PET<br />

Chest and abdom<strong>in</strong>al CT<br />

Additional study or biopsy if<br />

positive PET f<strong>in</strong>d<strong>in</strong>g<br />

PET before preoperative<br />

chemotherapy and on day 14<br />

of first chemotherapy cycle<br />

The reasons for no resection <strong>in</strong>cluded<br />

the follow<strong>in</strong>g: M1 disease found by PET<br />

and confirmed (9), M1 disease found by<br />

PET and not confirmed (2), M1 disease<br />

at exploration not found by PET (7),<br />

decl<strong>in</strong>e or death before surgery (10),<br />

patient refusal of surgery (7),<br />

unresectable local tumour at<br />

exploration (5), and extensive N1<br />

disease preclud<strong>in</strong>g operation (2). Eight<br />

(4.2%) patients undergo<strong>in</strong>g resection had<br />

a recurrence <strong>in</strong> the first 6 months.<br />

Metabolic responders showed a high<br />

histopathologic response rate (44%)<br />

<strong>with</strong> a 3-year survival rate of 70%. In<br />

contrast, prognosis was poor for<br />

metabolic nonresponders <strong>with</strong> a<br />

histopathologic response rate of 5% (P =<br />

0.001) and a 3-year survival rate of 35%<br />

(P = 0.01). A multivariate analysis<br />

(covariates: ypT-, ypN-category,<br />

histopathologic response) demonstrated<br />

that metabolic response was the only<br />

factor predict<strong>in</strong>g recurrence (P = 0.018)<br />

Standard not applied to<br />

all patients<br />

9 patients excluded from<br />

analysis (8 too low FDG<br />

uptake, 1 <strong>with</strong><br />

hyperglycemia)<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Very low<br />

Low


86 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

<strong>in</strong> the subgroup of completely resected<br />

Yuan S 2006 [75] NA Patients <strong>with</strong> a first<br />

diagnosis of biopsy-proven<br />

squamous cell cancer of<br />

the thoracic oesophagus,<br />

operable (<strong>in</strong>clud<strong>in</strong>g M1a),<br />

not refus<strong>in</strong>g surgery, and<br />

will<strong>in</strong>g to pay at least 30%<br />

of the charge for PET/CT<br />

(n = 45)<br />

Van Baardwijk A<br />

2006<br />

[248] August 2005 Patients <strong>with</strong> oesophageal<br />

cancer<br />

Leong T 2006 [70] NA Patients <strong>with</strong> histologically<br />

proven squamous or<br />

adenocarc<strong>in</strong>oma of the<br />

oesophagus stages I–III<br />

and ECOG performance<br />

status 0–2, where the<br />

treatment <strong>in</strong>tent was<br />

radical chemoradiotherapy<br />

(radiation dose 50 Gy)<br />

follow<strong>in</strong>g completion of all<br />

non-PET stag<strong>in</strong>g<br />

<strong>in</strong>vestigations that did not<br />

reveal evidence of<br />

metastatic disease (n =<br />

23)<br />

PET/CT<br />

Standard: histology<br />

PET for radiotherapy<br />

treatment plann<strong>in</strong>g<br />

PET/CT for radiotherapy<br />

plann<strong>in</strong>g vs. CT<br />

(R0) patients.<br />

The sensitivity, specificity, accuracy,<br />

positive predictive value, and negative<br />

predictive value of PET/CT were 93.90%<br />

(77/82 nodal groups), 92.06% (290/315),<br />

92.44% (367/397), 75.49% (77/102), and<br />

98.31% (290/295), respectively, whereas<br />

those of PET were 81.71% (67/82),<br />

87.30% (275/315), 86.15% (342/397),<br />

62.62% (67/107), and 94.83% (275/290),<br />

respectively. P values were 0.032, 0.067,<br />

0.006, 0.063, and 0.037, respectively.<br />

The differences <strong>in</strong> sensitivity, accuracy,<br />

and negative predictive value between<br />

PET and PET/CT were statistically<br />

significant.<br />

In oesophageal cancer, PET scan can be<br />

used to <strong>in</strong>clude PET positive lymph<br />

nodes <strong>in</strong> the target volume.<br />

The addition of PET <strong>in</strong>formation altered<br />

the cl<strong>in</strong>ical stage <strong>in</strong> 8 of 21 eligible<br />

patients enrolled on the study (38%); 4<br />

patients had distant metastatic disease<br />

and 4 had unsuspected regional nodal<br />

disease. Sixteen patients proceeded to<br />

the radiotherapy plann<strong>in</strong>g phase of the<br />

study and received def<strong>in</strong>itive<br />

chemoradiation planned <strong>with</strong> the<br />

PET/CT data set. The GTV based on CT<br />

<strong>in</strong>formation alone excluded PET-avid<br />

disease <strong>in</strong> 11 patients (69%), and <strong>in</strong> five<br />

patients (31%) this would have resulted<br />

<strong>in</strong> a geographic miss of gross tumour.<br />

The discordance between CT and<br />

PET/CT was due ma<strong>in</strong>ly to differences <strong>in</strong><br />

def<strong>in</strong><strong>in</strong>g the longitud<strong>in</strong>al extent of<br />

disease <strong>in</strong> the oesophagus. The cranial<br />

extent of the primary tumour as def<strong>in</strong>ed<br />

Medl<strong>in</strong>e only<br />

No <strong>in</strong>formation on study<br />

selection and quality<br />

appraisal<br />

Patients <strong>with</strong> PET<br />

detected metastatic<br />

disease came off study<br />

<strong>with</strong> no further data<br />

collection, while patients<br />

who did not have<br />

metastatic disease on PET<br />

and were deemed suitable<br />

for radical treatment<br />

proceeded to the<br />

radiotherapy plann<strong>in</strong>g<br />

phase of the study.<br />

Two patients excluded<br />

Prospective<br />

study<br />

Low<br />

SR Low<br />

Prospective<br />

study<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 87<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

by CT vs. PET/CT differed <strong>in</strong> 75% of<br />

cases, while the caudal extent differed <strong>in</strong><br />

Lev<strong>in</strong>e EA 2006 [249] NA 64 consecutive<br />

patients, <strong>with</strong> potentially<br />

curable, locally advanced<br />

oesophageal cancer<br />

Kato H 2005 [250] NA 44 patients <strong>with</strong> thoracic<br />

oesophageal cancer<br />

Moureau-Zabotto L<br />

2005<br />

[71] NA Thirty-four oesophageal<br />

carc<strong>in</strong>oma patients<br />

referred for concomitant<br />

radiotherapy and<br />

chemotherapy <strong>with</strong> radical<br />

<strong>in</strong>tent<br />

Song SY 2005 [74] NA Patients <strong>with</strong> locally<br />

advanced but resectable<br />

oesophageal cancer (n =<br />

32)<br />

PET pre- and postchemoradiation<br />

(of the 44<br />

patients undergo<strong>in</strong>g<br />

oesophagectomy follow<strong>in</strong>g<br />

chemoradiation, 31 patients<br />

had both a prechemoradiation<br />

and postchemoradiation<br />

PET, while 13<br />

patients had a s<strong>in</strong>gle PET scan:<br />

10 patients of which<br />

had a pre-chemoradiation<br />

study and 3 of which had postchemoradiation<br />

imag<strong>in</strong>g)<br />

FDG-PET<br />

Bone sc<strong>in</strong>tigraphy<br />

PET-CT vs. CT for<br />

radiotherapy treatment<br />

plann<strong>in</strong>g<br />

PET before and after<br />

neoadjuvant chemoradiation<br />

81%.<br />

Response was as follows: pCR 27%,<br />

pathologic residual microscopic<br />

(pCRmicro) 14.5%, partial response<br />

19%, and stable or progressive disease<br />

39.5%. A pretreatment standardized<br />

uptake value<br />

(SUVmax1 hour) ≥15 was associated<br />

<strong>with</strong> an observed 77.8% significant<br />

response (pCR + pCRmicro) compared<br />

<strong>with</strong> 24.2% for patients <strong>with</strong> a<br />

pretreatment SUVmax1 hour


88 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

oesophagus and lymph nodes.<br />

Van Westreenen HL<br />

2005<br />

[251] NA Ten patients <strong>with</strong> biopsyproven<br />

cancer of the<br />

oesophagus or gastrooesophageal<br />

junction<br />

Vrieze O 2004 [252] NA 30 patients <strong>with</strong> an<br />

advanced oesophageal<br />

carc<strong>in</strong>oma (cT4 based on<br />

EUS and/or CT), M0<br />

Sihvo EI 2004 [73] NA Patients <strong>with</strong> histologically<br />

proves adenocarc<strong>in</strong>oma of<br />

the oesophagus (n = 20)<br />

or the EG junction (n =<br />

Choi JY 2004 [68] NA Eighty-n<strong>in</strong>e consecutive<br />

patients hav<strong>in</strong>g newly<br />

diagnosed oesophageal<br />

cancer, undergo<strong>in</strong>g<br />

curative surgery<br />

35)<br />

18F-FLT PET vs.<br />

18F-FDG PET<br />

Standard: histology<br />

PET before neoadjuvant<br />

chemoradiation therapy (25<br />

patients treated <strong>with</strong> surgery)<br />

Rout<strong>in</strong>e stag<strong>in</strong>g <strong>with</strong><br />

endoscopy, CT and EUS<br />

FDG-PET<br />

Histopathology<br />

18F-FDG PET was able to detect all<br />

oesophageal cancers, whereas 18FFLT<br />

PET visualized the tumor <strong>in</strong> 8 of 10<br />

patients. Both 18F-FDG PET and 18F-<br />

FLT PET detected lymph node<br />

metastases <strong>in</strong> 2 of 8 patients. 18F-FDG<br />

PET detected 1 cervical lymph node that<br />

was missed on 18F-FLT PET, whereas<br />

18F-FDG PET showed uptake <strong>in</strong> benign<br />

lesions <strong>in</strong> 2 patients.<br />

In 14 of the 30 patients (47%)<br />

discordances were found <strong>in</strong> detection of<br />

the pathological lymph nodes between<br />

CT/EUS and FDGPET. In 8 patients, 9<br />

lymph node regions were found <strong>with</strong><br />

pathologic nodes on conventional<br />

imag<strong>in</strong>g only. In three of these patients<br />

the <strong>in</strong>fluence of FDG-PET f<strong>in</strong>d<strong>in</strong>gs would<br />

have led to a decrease of the irradiated<br />

volume. In 6 patients, 8 lymph node<br />

regions were found <strong>with</strong> a normal<br />

CT/EUS and pathologic nodes on FDG-<br />

PET. In three of these patients (10%) the<br />

<strong>in</strong>fluence of the FDG-PET would have<br />

led to enlargement of the irradiated<br />

volume.<br />

Accuracy N stag<strong>in</strong>g: PET 60%, EUS 72%,<br />

CT 58%<br />

Accuracy M stag<strong>in</strong>g: PET 76%, CT 75%<br />

PET preoperatively There were 130 malignant nodal groups<br />

of 554 dissected nodal groups <strong>in</strong> 43 of<br />

69 patients on pathologic exam<strong>in</strong>ation.<br />

N<strong>in</strong>ety-six positive lymph nodes <strong>in</strong> 43<br />

patients were evident on 18F-FDG PET.<br />

Only 30 lymph nodes were positive <strong>in</strong><br />

24 patients on CT. EUS revealed lymph<br />

Small sample size Prospective<br />

study<br />

Included <strong>in</strong> SR of Van<br />

Baardwijk et al.<br />

One centre analysis of<br />

prospective study of 36<br />

patients<br />

No comparison <strong>with</strong><br />

histology<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Very low<br />

Very low<br />

Low<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 89<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

node metastases <strong>in</strong> 27 patients.<br />

Kneist W 2004 [69] NA Patients <strong>with</strong> biopsyproved<br />

oesophageal<br />

carc<strong>in</strong>oma (n = 81)<br />

Neck imag<strong>in</strong>g<br />

No additional diagnostic studies found<br />

PET and computer<br />

tomography<br />

(CT) of the chest and<br />

abdomen (and of the neck <strong>in</strong><br />

45 patients) <strong>with</strong><strong>in</strong> 45 days.<br />

The f<strong>in</strong>d<strong>in</strong>gs of the<br />

histopathological exam<strong>in</strong>ation<br />

for 31 suspicious lesions from<br />

28 patients served as the basis<br />

for the diagnosis of distant<br />

metastasis.<br />

The PET f<strong>in</strong>d<strong>in</strong>gs had a higher specificity<br />

(89% vs 11%) but a lower sensitivity<br />

(38% vs 63%) than CT f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> the<br />

detection of metastatic sites. The CT<br />

results showed greater agreement <strong>with</strong><br />

histopathological f<strong>in</strong>d<strong>in</strong>gs than did PET<br />

results. In 8 patients (10%), PET<br />

detected distant metastases that were<br />

not identified <strong>with</strong> CT. In 4 patients<br />

(5%), PET detected bone metastases<br />

only, but <strong>in</strong> all of these patients<br />

metastases <strong>in</strong> other locations were<br />

detected by CT. Although PET led to<br />

upstag<strong>in</strong>g (M1) <strong>in</strong> 2 patients (2%), it did<br />

not enable the exclusion of oesophageal<br />

resection.<br />

Prospective<br />

study<br />

Low


90 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Diagnosis and treatment of early lesions.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

SIGN [47] 2004 Reduction of risk of progression to adenocarc<strong>in</strong>oma is not an <strong>in</strong>dication for anti-reflux surgery <strong>in</strong> patients Spechler SJ 2001<br />

High<br />

<strong>with</strong> Barrett’s oesophagus.<br />

Corey KE 2003<br />

Parrilla P 2003<br />

Rout<strong>in</strong>e use of chromoendoscopy dur<strong>in</strong>g upper GI endoscopy is not recommended, but may be of value Inone H 2001<br />

Moderate<br />

<strong>in</strong> selected patients at high risk of oesophageal malignancy.<br />

Ragunath K 2003<br />

Kiesslich R 2001<br />

Canto MI 2001<br />

Mitooka H 1995<br />

In patients <strong>with</strong> Barrett’s oesophagus there should be a structured biopsy protocol <strong>with</strong> quadrantic Garside R 2006<br />

Low<br />

biopsies every two centimetres and biopsy of any visible lesion.<br />

Fitzgerald RC 2001<br />

Conio M 2003<br />

Macdonald CE 2000<br />

Pathologists should follow the revised Vienna classification for report<strong>in</strong>g dysplasia. Montgomery E 2001a<br />

Schlemper RJ 2000<br />

Low<br />

Where radical <strong>in</strong>tervention is contemplated on the basis of high-grade dysplasia or early adenocarc<strong>in</strong>oma Montgomery E 2001a Low<br />

the diagnosis should be validated by a second pathologist experienced <strong>in</strong> this area and further biopsies Baak JP 2002<br />

should be taken if there is uncerta<strong>in</strong>ty.<br />

Montgomery E 2001b<br />

Evaluation of suspected high-grade dysplasia <strong>in</strong> Barrett’s oesophagus biopsies should be undertaken <strong>with</strong> Baak JP 2002<br />

Low<br />

knowledge of the cl<strong>in</strong>ical and endoscopic background and biopsies should be reviewed at a<br />

multidiscipl<strong>in</strong>ary meet<strong>in</strong>g <strong>with</strong> access to the cl<strong>in</strong>ical <strong>in</strong>formation.<br />

Montgomery E 2001b<br />

Patients diagnosed <strong>with</strong> high-grade dysplasia should have careful assessment (CT, EUS, rigorous biopsy Schnell TG 2001<br />

Low<br />

protocol +/- EMR) to exclude coexist<strong>in</strong>g cancer.<br />

Lev<strong>in</strong>e DS 1993<br />

May A 2002<br />

In the absence of <strong>in</strong>vasive cancer, patients <strong>with</strong> high-grade dysplasia should be offered endoscopic May A 2002<br />

Low<br />

treatment.<br />

Ell C 2000<br />

Pacifico RJ 2003<br />

The assessment and management of patients <strong>with</strong> high-grade dysplasia should be centralised to units <strong>with</strong><br />

the appropriate endoscopic facilities and expertise.<br />

Ell C 2000 Low<br />

Superficial oesophageal cancer limited to the mucosa should be treated <strong>with</strong> EMR. Fujita H 2001<br />

Inoue H 1991<br />

Takekoshi T 1994<br />

Low<br />

Mucosal ablative techniques, such as PDT, APC, or laser should be reserved for the management of May A 2002<br />

Low<br />

residual disease after EMR and not for <strong>in</strong>itial management if <strong>in</strong>vasive disease is present <strong>in</strong> patients fit for Ell C 2000<br />

surgery.<br />

Pacifico RJ 2003<br />

Attwood SE 2003<br />

CBO [56] July 2003 Ondanks de relatief lage <strong>in</strong>terobserveerdersvariatie verdient het aanbevel<strong>in</strong>g dat de histopathologische Schlemper 2000a<br />

Low<br />

diagnose van hooggradige dysplasie (carc<strong>in</strong>oma <strong>in</strong> situ) en vroegcarc<strong>in</strong>oom <strong>in</strong> de oesofagus door een Schlemper 2000b<br />

patholoog met ervar<strong>in</strong>g op dit gebied wordt gesteld.<br />

Montgomery 2001


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 91<br />

CPG ID Ref Search<br />

date<br />

Recommendation Support<strong>in</strong>g evidence<br />

Skacel 2000<br />

Level of<br />

evidence<br />

Chromoscopie met lugolkleur<strong>in</strong>g is geïndiceerd bij patiënten met een neoplastische afwijk<strong>in</strong>g <strong>in</strong> het Dawsey 1998 Low<br />

plaveiselepitheel van de oesofagus waarbij endoscopische therapie wordt overwogen.<br />

Bij patiënten met een neoplastische afwijk<strong>in</strong>g <strong>in</strong> een Barrett-oesofagus gelden de volgende aanbevel<strong>in</strong>gen <strong>in</strong><br />

de endoscopische work-up voor eventuele endoscopische therapie:<br />

- Endoscopische <strong>in</strong>spectie geschiedt bij voorkeur <strong>in</strong> een centrum met endoscopische expertise <strong>in</strong><br />

de detectie van vroege afwijk<strong>in</strong>gen <strong>in</strong> Barrett-oesofagus.<br />

- Hierbij wordt bij voorkeur gebruikgemaakt van hoge-resolutie-endoscopie, eventueel aangevuld<br />

met chromoscopie.<br />

- Geadviseerd wordt om biopten te nemen uit alle zichtbare mucosale onregelmatigheden en at<br />

random uit vier kwadranten voor elke centimeter Barrett-oesofagus, te beg<strong>in</strong>nen 1 cm boven de<br />

bovenrand van de maagplooien en doorlopend tot aan de overgang van cil<strong>in</strong>drisch naar<br />

plaveiselepitheel.<br />

Bij alle patiënten met een neoplastische afwijk<strong>in</strong>g <strong>in</strong> de oesofagus bij wie endoscopische therapie wordt<br />

overwogen, dient een endoscopische ultrasonografie te worden verricht om diepte-<strong>in</strong>filtratie van de<br />

afwijk<strong>in</strong>g en verdachte lokale lymfklieren uit te sluiten.<br />

Dave U 2001<br />

Endo T 2002<br />

Guelrud M 2001<br />

Sharma P 2003<br />

Reid BJ 2000<br />

Scot<strong>in</strong>iotis IA 2001<br />

Hasegawa N 1996<br />

Natsugoe S 1996<br />

Aanvullende stadiër<strong>in</strong>g met CT-scan is optioneel. Bergman JJ 1999<br />

Scot<strong>in</strong>iotis IA 2001<br />

Low<br />

Voor patiënten met hooggradige dysplasie en/of vroegcarc<strong>in</strong>oom <strong>in</strong> plaveiselepitheel van de oesofagus Shimizu Y 2002<br />

Low<br />

heeft endoscopische behandel<strong>in</strong>g de voorkeur. Hierbij zijn de volgende voorwaarden van belang: Fujita H 2001<br />

- Voorafgaande aan de behandel<strong>in</strong>g worden een endoscopische beoordel<strong>in</strong>g en stadiër<strong>in</strong>g verricht. Narahara H 2000<br />

- Het betreft een solitaire, vlakke afwijk<strong>in</strong>g met een maximale diameter van 2-3 cm.<br />

Nijhawan PK 2000<br />

Als endoscopische resectietechniek verdient de endoscopische mucosale resectie-cap-techniek de<br />

voorkeur, zodat histologisch onderzoek van de verwijderde afwijk<strong>in</strong>g mogelijk is.<br />

Het besluit om de endoscopische behandel<strong>in</strong>g als curatief te beschouwen, kan pas worden genomen nadat<br />

het endoscopische mucosale resectiepreparaat histologisch is beoordeeld.<br />

Bij patiënten met evidente submucosale <strong>in</strong>groei en/of positieve resectieranden van het endoscopische<br />

mucosale resectiepreparaat dient <strong>in</strong> pr<strong>in</strong>cipe een chirurgische resectie te worden verricht.<br />

Inoue H 1991 Low<br />

Er v<strong>in</strong>dt <strong>in</strong>itieel een endoscopisch onderzoek plaats dat specifiek dient om <strong>in</strong> de Barrett-oesofagus een<br />

eventueel <strong>in</strong>vasief carc<strong>in</strong>oom te detecteren:<br />

- Hierbij geniet hoge-resolutie-endoscopie, eventueel aangevuld met chromoscopie, de voorkeur.<br />

- Er worden biopten genomen uit alle zichtbare mucosale onregelmatigheden en random uit vier<br />

kwadranten voor elke centimeter Barrett-oesofagus, te beg<strong>in</strong>nen 1 cm boven de bovenrand van de<br />

maagplooien en doorlopend tot aan de overgang tussen cil<strong>in</strong>der- en plaveiselepitheel.<br />

Cfr supra<br />

Na de work-up wordt de endoscopische surveillance als volgt verricht:<br />

- Frequentie surveillance: <strong>in</strong> het eerste jaar elke drie maanden, <strong>in</strong> het tweede jaar elke zes maanden en<br />

daarna tenm<strong>in</strong>ste jaarlijks.<br />

- Indien er tijdens surveillance sprake is van nieuwe afwijk<strong>in</strong>gen zoals erosies of een onregelmatig<br />

oppervlak, dienen opnieuw biopten te worden genomen en/of dient dit te worden uitgebreid met een<br />

No evidence<br />

Low<br />

Low


92 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Search<br />

date<br />

Recommendation<br />

diagnostische endoscopische mucosale resectie.<br />

Support<strong>in</strong>g evidence Level of<br />

evidence<br />

Endoscopische surveillance (ten m<strong>in</strong>ste gedurende het eerste jaar) wordt bij voorkeur verricht <strong>in</strong> een Cfr supra<br />

centrum met ervar<strong>in</strong>g.<br />

Voor geselecteerde patiënten met hooggradige dysplasie en/of vroegcarc<strong>in</strong>oom <strong>in</strong> een Barrett-oesofagus<br />

heeft endoscopische behandel<strong>in</strong>g de voorkeur. Hierbij gelden de volgende voorwaarden:<br />

- Voorafgaande aan de behandel<strong>in</strong>g zijn endoscopische beoordel<strong>in</strong>g en stadiër<strong>in</strong>g verricht.<br />

- Het betreft een solitaire verheven en/of vlakke afwijk<strong>in</strong>g met een maximale grootte van 2 cm.<br />

De endoscopische behandel<strong>in</strong>g omvat als primaire behandel<strong>in</strong>g een endoscopische resectietechniek (bij<br />

voorkeur de endoscopische mucosale resectie-cap-techniek), waardoor histologisch onderzoek van de<br />

verwijderde afwijk<strong>in</strong>g mogelijk is.<br />

Buttar NS 2001<br />

May A 2002<br />

Buttar NS 2001<br />

May A 2002<br />

Het besluit om endoscopische behandel<strong>in</strong>g als <strong>in</strong> opzet curatieve behandel<strong>in</strong>g te beschouwen, kan pas<br />

worden genomen nadat het endoscopische mucosale resectiepreparaat is beoordeeld.<br />

Bij geselecteerde patiënten kan aanvullende ablatietherapie na voorafgaande endoscopische mucosale<br />

resectie worden overwogen.<br />

Bij afwijk<strong>in</strong>gen groter dan 2 cm en/of de helft van de circumferentie is ‘piece-meal-resectie vereist. Ell C 2000 Low<br />

De histopathologische beoordel<strong>in</strong>g van de endoscopische mucosale resectiepreparaten dient door een<br />

patholoog met ervar<strong>in</strong>g op dit gebied te geschieden. Referentiepanels voor een ‘second op<strong>in</strong>ion’ zijn <strong>in</strong><br />

Nederland aanwezig.<br />

Bij submucosale <strong>in</strong>groei van een vroegcarc<strong>in</strong>oom <strong>in</strong> plaveiselcelepitheel dient alsnog een chirurgische<br />

resectie te worden overwogen gezien de hoge kans (26-47%) op lokale lymfkliermetastasen.<br />

Bij submucosale <strong>in</strong>groei van een vroegcarc<strong>in</strong>oom <strong>in</strong> een Barrett-oesofagus dient alsnog een chirurgische<br />

resectie te worden overwogen gezien de hoge kans (tot 50%) op lokale lymfkliermetastasen.<br />

Na endoscopische behandel<strong>in</strong>g van hooggradige dysplasie/vroegcarc<strong>in</strong>oom <strong>in</strong> de oesofagus gelden de<br />

aanbevel<strong>in</strong>gen voor de endoscopische follow-up conform de aanbevel<strong>in</strong>gen voor endoscopische<br />

surveillance van Barrett-oesofagus.<br />

Schlemper 2000a<br />

Schlemper 2000b<br />

Low<br />

Low<br />

Low<br />

Multiple studies Low<br />

Multiple studies Low<br />

May A 2002 Low<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

Diagnosis<br />

Pohl J 2007 [253] NA Patients <strong>with</strong>Barrett’s<br />

oesophagus and possible<br />

high-grade <strong>in</strong>traepithelial<br />

neoplasia or early cancer,<br />

hav<strong>in</strong>g discrete mucosal<br />

alterations or<br />

macroscopically occult<br />

lesions (n = 57)<br />

Conventional<br />

chromoendoscopy <strong>with</strong> acetic<br />

acid vs. computed virtual<br />

chromoendoscopy<br />

At each exam<strong>in</strong>ation targeted<br />

biopsies from all detected<br />

lesions, followed by random<br />

four-quadrant biopsies<br />

In 24/57 patients, 30 lesions <strong>with</strong><br />

HGIN/early cancer were detected. The<br />

sensitivity of targeted biopsies for<br />

HGIN/early cancer on a 'per lesion'<br />

basis was 87 % (26/30) for both CAA<br />

and CVC. The positive predictive value<br />

was 39 % (26/66) for CAA and 37 %<br />

(26/70) for CVC. In the 'per patient'<br />

analysis, sensitivity was 83 % (20/24) and<br />

Cross-over design<br />

Randomization <strong>with</strong><br />

sealed envelopes<br />

(bl<strong>in</strong>ded??)<br />

Bl<strong>in</strong>ded assessors<br />

RCT Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 93<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

92 % (22/24) for CAA and CVC,<br />

respectively ( P = 0.617). Stepwise<br />

random four-quadrant biopsies identified<br />

only one patient <strong>with</strong> HGIN/early<br />

cancer that was missed by both, CAA<br />

Borovicka J 2006 [52] NA Patients <strong>with</strong><br />

endoscopically<br />

documented Barrett’s<br />

oesophagus (n = 187)<br />

Lim CH 2006 [53] NA Patients <strong>with</strong> a diagnosis<br />

of dysplasia identified <strong>in</strong><br />

Barrett's oesophagus (n =<br />

35)<br />

Phase 1: Autofluorescence<br />

endoscopy <strong>with</strong> targeted<br />

biopsy followed by fourquadrant<br />

biopsies vs.<br />

conventional endoscopic<br />

surveillance, also <strong>in</strong>clud<strong>in</strong>g<br />

four-quadrant biopsies.<br />

Phase 2: After exclusion of<br />

patients <strong>with</strong> early cancer or<br />

high-grade dysplasia, who<br />

underwent endoscopic or<br />

surgical treatment, as well as<br />

those who decl<strong>in</strong>ed to<br />

participate <strong>in</strong> phase 2 of the<br />

study, 130 patients rema<strong>in</strong>ed.<br />

These patients were exam<strong>in</strong>ed<br />

aga<strong>in</strong> <strong>with</strong> the alternative<br />

method after a mean of 10<br />

weeks, us<strong>in</strong>g the same<br />

methods described.<br />

4 random quadrant biopsies<br />

taken every 2 cm through the<br />

length of the Barrett's<br />

esophagus (n = 18) vs.<br />

methylene blue (MB)<br />

chromoendoscopy directed<br />

biopsies from unsta<strong>in</strong>ed or<br />

heterogenously sta<strong>in</strong>ed<br />

mucosa (n = 17)<br />

and CVC.<br />

In study phase 1, the AFE and<br />

conventional approaches yielded<br />

adenocarc<strong>in</strong>oma/HGD rates of 12% and<br />

5.3%, respectively, on a per-patient<br />

basis. With AFE, four previously<br />

unrecognized adenocarc<strong>in</strong>oma/HGD<br />

lesions were identified (4.3% of the<br />

patients); <strong>with</strong> the conventional<br />

approach, one new lesion (1.1%) was<br />

identified. Of the 19<br />

adenocarc<strong>in</strong>oma/HGD lesions detected<br />

dur<strong>in</strong>g AFE endoscopy <strong>in</strong> study phase 1,<br />

eight were visualized, while 11 were<br />

only detected us<strong>in</strong>g untargeted fourquadrant<br />

biopsies (sensitivity 42%). Of<br />

the 766 biopsies classified at histology as<br />

be<strong>in</strong>g non-neoplastic, 58 appeared<br />

suspicious (specificity 92%, positive<br />

predictive value 12%, negative predictive<br />

value 98.5%). In study phase 2, AFE<br />

detected two further lesions <strong>in</strong> addition<br />

to the <strong>in</strong>itial alternative approach <strong>in</strong><br />

3.2% of cases, <strong>in</strong> comparison <strong>with</strong> one<br />

lesion <strong>with</strong> conventional endoscopy<br />

(1.7%).<br />

Overall, dysplasia was identified <strong>in</strong> 17 of<br />

18 patients by RB and <strong>in</strong> 9 of 18 by<br />

MBDB (McNemar test, p = 0.02).<br />

Cross-over design<br />

Bl<strong>in</strong>ded assessors<br />

Computer-generated<br />

randomization list <strong>with</strong><br />

block randomization<br />

Cross-over design<br />

No ITT<br />

RCT High<br />

RCT Moderate


94 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

Kara MA 2005 [54] NA Patients <strong>with</strong> Barrett’s High-resolution endoscopy Fourteen patients were diagnosed <strong>with</strong> Cross-over design RCT Moderate<br />

oesophagus and recently <strong>with</strong> <strong>in</strong>digo carm<strong>in</strong>e<br />

HGD/EC. The sensitivity for HGD/EC Randomization <strong>with</strong><br />

diagnosed but<br />

chromoendoscopy or narrow- was 93 % and 86 % for HRE-ICC and sealed opaque envelopes<br />

endoscopically<br />

band imag<strong>in</strong>g<br />

HRE-NBI, respectively. Targeted (bl<strong>in</strong>ded??)<br />

<strong>in</strong>conspicuous high-grade<br />

biopsies had a sensitivity of 79 % <strong>with</strong> Bl<strong>in</strong>ded assessors<br />

dysplasia or early cancer<br />

HRE alone. HGD was diagnosed from<br />

(n = 28)<br />

random biopsies alone <strong>in</strong> only one<br />

patient. ICC and NBI detected a limited<br />

number of additional lesions occult to<br />

HRE, but these lesions did not alter the<br />

sensitivity for identify<strong>in</strong>g patients <strong>with</strong><br />

HGD/EC.<br />

Ragunath K [254] NA Patients <strong>with</strong> Barrett's Methylene blue-directed Analysis of the results by per-biopsy Cross-over design RCT High<br />

2003<br />

oesophagus segments 3 biopsies vs. random biopsy (4 protocol showed that the MBDB Bl<strong>in</strong>d<strong>in</strong>g<br />

cm or more <strong>in</strong> length random quadrant biopsies technique diagnosed significantly more<br />

<strong>with</strong>out macroscopic taken every 2 cm)<br />

specialized <strong>in</strong>test<strong>in</strong>al metaplasia (75 %)<br />

evidence of dysplasia or<br />

compared to the random biopsy<br />

cancer (n = 75)<br />

technique (68 %; P = 0.032). The<br />

sensitivity and specificity rates of MBDB<br />

for diagnos<strong>in</strong>g specialized <strong>in</strong>test<strong>in</strong>al<br />

metaplasia were 91 % (95 % CI, 88 - 93<br />

%) and 43 % (95 % CI, 36 - 51 %),<br />

respectively. The sensitivity and<br />

specificity rates of MBDB for diagnos<strong>in</strong>g<br />

dysplasia or carc<strong>in</strong>oma were 49 % (95 %<br />

CI, 38 - 61 %) and 85 % (95 % CI, 82 -<br />

88 %), respectively. There were no<br />

significant differences <strong>in</strong> the diagnosis of<br />

dysplasia and carc<strong>in</strong>oma - MBDB 12 %,<br />

random biopsy 10 %. The methylene<br />

blue sta<strong>in</strong><strong>in</strong>g pattern appeared to have<br />

an <strong>in</strong>fluence on the detection of<br />

specialized <strong>in</strong>test<strong>in</strong>al metaplasia and<br />

dysplasia/carc<strong>in</strong>oma. Dark blue sta<strong>in</strong><strong>in</strong>g<br />

was associated <strong>with</strong> <strong>in</strong>creased detection<br />

of specialized <strong>in</strong>test<strong>in</strong>al metaplasia (P <<br />

0.0001), and heterogeneous sta<strong>in</strong><strong>in</strong>g (P =<br />

0.137) or no sta<strong>in</strong><strong>in</strong>g (P = 0.005) were<br />

associated <strong>with</strong> dysplasia and/or<br />

carc<strong>in</strong>oma detection. The MBDB


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 95<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

technique prolonged the endoscopy<br />

exam<strong>in</strong>ation by an average of 6 m<strong>in</strong>.<br />

Stag<strong>in</strong>g<br />

Pech O 2006 [255] NA Patients <strong>with</strong> suspected<br />

early cancer <strong>in</strong> Barrett’s<br />

oesophagus referred for<br />

endoscopic treatment (n<br />

= 100)<br />

Larghi A 2005 [256] NA Patients (n = 48) <strong>with</strong><br />

Barrett’s oesophagus and<br />

biopsy specimen proven<br />

high-grade dysplasia and<br />

adenocarc<strong>in</strong>oma <strong>in</strong> focal<br />

nodular lesions or <strong>in</strong><br />

endoscopically unapparent<br />

flat lesions <strong>in</strong> shortsegment<br />

Barrett’s<br />

oesophagus<br />

Waxman I 2006<br />

Treatment<br />

[257] NA 9 consecutive patients<br />

<strong>with</strong> Barrett’s oesophagus<br />

and high-grade dysplasia (n<br />

= 7) or <strong>in</strong>tra-mucosal<br />

carc<strong>in</strong>oma (n = 2)<br />

Overholt BF<br />

2005<br />

[83] NA Patients <strong>with</strong> Barrett’s<br />

oesophagus and highgrade<br />

dysplasia<br />

EUS<br />

Helical CT of chest and upper<br />

abdom<strong>in</strong>al organs<br />

Abdom<strong>in</strong>al US exam<strong>in</strong>ation<br />

Histology (n = 62)<br />

EUS<br />

Standard: surgical pathology<br />

result (n = 8) or EMR<br />

pathology result<br />

High-frequency (20 MHz)<br />

probe ultrasonography<br />

(HFPUS)<br />

Standard: histology<br />

Photodynamic therapy <strong>with</strong><br />

porfimer sodium +<br />

omeprazole (n = 138) vs.<br />

omeprazole only (n = 70)<br />

For stag<strong>in</strong>g of T1 tumours, the<br />

sensitivity of CT was 100%, but its<br />

specificity was 0%; PPV 89%, NPV 0%.<br />

Differentiation between T1 and>T1<br />

us<strong>in</strong>g EUS was possible <strong>in</strong> all 62 patients.<br />

Sensitivity of CT for N stag<strong>in</strong>g was not<br />

acceptable compared <strong>with</strong> EUS (38% vs.<br />

75%). No extranodal metastases were<br />

found on CT.<br />

Accurate stag<strong>in</strong>g <strong>in</strong> 41/48 patients (85%)<br />

Overstag<strong>in</strong>g <strong>in</strong> 1 pt, understag<strong>in</strong>g <strong>in</strong> 6<br />

pts<br />

Sensitivity 60%<br />

Specificity 75%<br />

Of the 3 true-positives: 1 overstaged<br />

and 1 understaged<br />

There was a significant difference<br />

(p=0.0001) <strong>in</strong> favour of PORPDT<br />

(106/138 [77%]) compared <strong>with</strong> OM<br />

(27/70 [39%]) <strong>in</strong> complete ablation of<br />

HGD at any time dur<strong>in</strong>g the study<br />

period. The occurrence of<br />

adenocarc<strong>in</strong>oma <strong>in</strong> the PORPDT group<br />

(13%) (n=18) was significantly lower<br />

(p=0.006) compared <strong>with</strong> the OM group<br />

(20%) (n=20). The safety profile showed<br />

94% of patients <strong>in</strong> the PORPDT group<br />

Histology not available <strong>in</strong><br />

all patients<br />

Prospective<br />

observational<br />

study<br />

S<strong>in</strong>gle-center study Prospective<br />

observational<br />

study<br />

Small sample size Prospective<br />

case series<br />

No <strong>in</strong>formation on<br />

randomization procedure<br />

Only partially bl<strong>in</strong>ded<br />

(pathology)<br />

Low<br />

Low<br />

Very low<br />

RCT Moderate


96 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

and 13% of patients <strong>in</strong> the OM group<br />

May A 2003 [258] NA 72 patients <strong>with</strong> early<br />

stage oesophageal cancer<br />

Endoscopic resection <strong>with</strong><br />

"suck-and-ligate" device<br />

<strong>with</strong>out prior submucosa<br />

<strong>in</strong>jection (n = 50) vs. cap<br />

technique <strong>with</strong> prior<br />

submucosa <strong>in</strong>jection of a dilute<br />

sal<strong>in</strong>e solution of ep<strong>in</strong>ephr<strong>in</strong>e<br />

(n = 50)<br />

had treatment-related adverse effects.<br />

No significant differences were observed<br />

between the two groups <strong>with</strong> regard to<br />

the maximum diameters and calculated<br />

area of the resected specimens (ligation<br />

group: 16.4 [4.0] x 11 [3.1] mm/185 [84]<br />

mm(2) vs. cap group: 15.5 [4.1] x 10.7<br />

[2.7] mm/168 [83] mm(2)), or the<br />

maximum diameters and calculated area<br />

of the endoscopic resection ulcers after<br />

24 hours (ligation group: 20.6 [4.8] x<br />

14.3 [4.5] mm/314 [160] mm(2) vs. cap<br />

group: 18.9 [5.1] x 12.9 [3.8] mm/260<br />

[145] mm(2)). There was only a slight<br />

advantage (greater diameter of resection<br />

specimens) for the ligation group <strong>in</strong><br />

patients who had prior endoscopic<br />

treatment. There was one m<strong>in</strong>or<br />

episode of bleed<strong>in</strong>g <strong>in</strong> each group; there<br />

was no severe complication. In 41 of 72<br />

patients (57%), further endoscopic<br />

therapy after endoscopic resection was<br />

necessary because of residual neoplasia<br />

at the first follow-up endoscopy after<br />

resection (61 of 100 resection<br />

specimens [61%] had lateral marg<strong>in</strong>s that<br />

could not be evaluated because of<br />

coagulation artifact or conta<strong>in</strong>ed<br />

malignancy but <strong>with</strong> the base of the<br />

lesion free of tumor).<br />

Bl<strong>in</strong>ded randomization<br />

Bl<strong>in</strong>ded evaluation?<br />

RCT Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 97<br />

CPG ID Ref Search<br />

date<br />

Chemotherapy<br />

CCO [84] January<br />

2005<br />

Neoadjuvant treatment.<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

(In adult patients <strong>with</strong> resectable and potentially curable thoracic (lower two-thirds of oesophagus)<br />

oesophageal cancer) if surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or<br />

adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.<br />

SIGN [47] 2004 Patients <strong>with</strong> operable oesophageal cancer, who are treated surgically, should be considered for two<br />

cycles of preoperative chemotherapy <strong>with</strong> cisplat<strong>in</strong> and 5-fluorouracil or offered entry <strong>in</strong>to a cl<strong>in</strong>ical trial.<br />

CBO [56] July 2003 Patiënten met een potentieel resectabel oesofaguscarc<strong>in</strong>oom dienen alleen <strong>in</strong> onderzoeksverband<br />

voorafgaande aan een operatie chemotherapie te krijgen.<br />

Radiotherapy<br />

CCO [84] January<br />

2005<br />

(In adult patients <strong>with</strong> resectable and potentially curable thoracic (lower two-thirds of oesophagus)<br />

oesophageal cancer) if surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or<br />

adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.<br />

Numerous RCTs and SRs High<br />

Malthaner 2003 High<br />

Malthaner 2003 High<br />

Arnott SJ 2000<br />

Fok 1994<br />

SIGN [47] 2004 Preoperative radiotherapy is not recommended for patients <strong>with</strong> oesophageal cancer. Arnott SJ 2000 High<br />

CBO [56] July 2003 De werkgroep raadt af om patiënten met een potentieel resectabel oesofaguscarc<strong>in</strong>oom voorafgaand aan<br />

de operatie te bestralen.<br />

Arnott SJ 2000 High<br />

Chemoradiotherapy<br />

CCO [84] January<br />

2005<br />

(In adult patients <strong>with</strong> resectable and potentially curable thoracic (lower two-thirds of oesophagus)<br />

oesophageal cancer) if surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or<br />

adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.<br />

SIGN [47] 2004 Preoperative chemoradiotherapy for patients <strong>with</strong> oesophageal cancer is not recommended outside<br />

cl<strong>in</strong>ical trials.<br />

CBO [56] July 2003 De werkgroep adviseert om patiënten met een potentieel resectabel oesofaguscarc<strong>in</strong>oom voorafgaand<br />

aan een operatie uitsluitend <strong>in</strong> onderzoeksverband chemoradiotherapie te geven.<br />

High<br />

Numerous RCTs and SRs High<br />

Urschel 2003 High<br />

Walsh TN 1996 (RCT)<br />

Urba SG 2001 (RCT)<br />

High


98 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID<br />

Chemotherapy<br />

Ref Search<br />

date<br />

Graham AH<br />

2007<br />

[88] October<br />

2004<br />

Population Intervention Results Comments Study<br />

type<br />

Patients <strong>with</strong> locally<br />

advanced oesophageal<br />

cancer (T2-3N0M0 or T1-<br />

3N1M0)<br />

Gebski V 2007 [86] 2006 Patients <strong>with</strong> local<br />

operable oesophageal<br />

cancer<br />

Malthaner RA<br />

2006<br />

[87] 2006 Patients <strong>with</strong> localized<br />

potentially resectable<br />

thoracic oesophageal<br />

carc<strong>in</strong>omas<br />

Surgery vs.<br />

chemoradiotherapy followed<br />

by surgery vs. chemotherapy<br />

followed by surgery vs.<br />

surgery followed by<br />

chemoradiotherapy<br />

Neoadjuvant<br />

chemoradiotherapy or<br />

neoadjuvant<br />

chemotherapy followed by<br />

surgery vs. surgery alone<br />

Neoadjuvant chemotherapy<br />

followed by surgery vs.<br />

surgery alone<br />

For the first year, the relative risk (95%<br />

confidence <strong>in</strong>terval) of death for treatments<br />

compared <strong>with</strong> surgery were 0.87 (0.75 to<br />

1.02) for chemoradiotherapy followed by<br />

surgery, 0.94 (0.82 to 1.08) for chemotherapy<br />

followed by surgery, and 1.33 (0.93 to 1.93)<br />

for surgery <strong>with</strong> adjuvant chemoradiotherapy.<br />

The QALYs ga<strong>in</strong>ed for surgery alone,<br />

chemoradiotherapy followed by surgery,<br />

chemotherapy followed by surgery, and<br />

surgery <strong>with</strong> adjuvant chemoradiotherapy<br />

strategies were 2.07, 2.18, 2.14, and 1.99,<br />

respectively. If the reduction <strong>in</strong> utility for<br />

multimodality treatment was <strong>in</strong>creased to 21%,<br />

the QALYs ga<strong>in</strong>ed for surgery alone,<br />

chemoradiotherapy followed by surgery,<br />

chemotherapy followed by surgery, and<br />

surgery <strong>with</strong> adjuvant chemoradiotherapy<br />

were 2.07, 2.03, 1.99, and 1.85, respectively.<br />

The hazard ratio for neoadjuvant<br />

chemotherapy was 0.90 (0.81-1.00; p=0.05),<br />

which <strong>in</strong>dicates a 2-year absolute survival<br />

benefit of 7%. There was no significant effect<br />

on all-cause mortality of chemotherapy for<br />

patients <strong>with</strong> SCC (hazard ratio 0.88 [0.75-<br />

1.03]; p=0.12), although there was a significant<br />

benefit for those <strong>with</strong> adenocarc<strong>in</strong>oma (0.78<br />

[0.64-0.95]; p=0.014).<br />

There was some evidence to suggest that<br />

preoperative chemotherapy improves survival,<br />

but this was <strong>in</strong>conclusive (HR 0.88; 95% CI<br />

0.75 to 1.04).<br />

There was no evidence to suggest that the<br />

14 RCTs <strong>in</strong>cluded, of<br />

which 6 RCTs on<br />

neoadjuvant<br />

chemotherapy<br />

NO NEW EVIDENCE<br />

SINCE CCO<br />

GUIDELINE!<br />

Only <strong>in</strong>clusion of RCTs<br />

<strong>with</strong> ITT analysis<br />

No results of quality<br />

appraisal reported<br />

8 RCTs on neoadjuvant<br />

chemotherapy<br />

2 RCTs <strong>in</strong> addition to<br />

Graham et al. (Roth<br />

1998 and Schlag 1992)<br />

NO NEW EVIDENCE<br />

SINCE CCO<br />

GUIDELINE!<br />

11 RCTs <strong>in</strong>cluded<br />

NO NEW EVIDENCE<br />

SINCE CCO<br />

GUIDELINE!<br />

SR High<br />

SR High<br />

Level of<br />

evidence<br />

SR High


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 99<br />

Study ID Ref Search<br />

date<br />

Cunn<strong>in</strong>gham D<br />

2006<br />

Radiotherapy<br />

Arnott SJ 2005 [85] June<br />

2006<br />

Chemoradiotherapy<br />

Population Intervention Results Comments Study Level of<br />

overall rate of resections (RR 0.96, 95% CI<br />

0.92 to 1.01) or the rate of complete<br />

resections (R0) (RR 1.05; 95% CI 0.97 to 1.15)<br />

differ between the preoperative chemotherapy<br />

arm and surgery alone. There is no evidence<br />

that tumour recurrence (RR 0.81, 95% CI 0.54<br />

to 1.22) or non-fatal complication rates (RR<br />

0.90; 95% CI 0.76 to 1.06) differ for<br />

preoperative chemotherapy compared to<br />

surgery alone. Trials reported risks of toxicity<br />

<strong>with</strong> chemotherapy that ranged from 11% to<br />

90%.<br />

type evidence<br />

[148] NA Patients <strong>with</strong> histologically Perioperative chemotherapy As compared <strong>with</strong> the surgery group, the Randomization RCT Moderate<br />

proven adenocarc<strong>in</strong>oma and surgery (n = 250) vs. perioperative-chemotherapy group had a procedure?<br />

of the stomach or lower surgery alone (n = 253) higher likelihood of overall survival (HR for No <strong>in</strong>formation of<br />

third of the oesophagus<br />

death, 0.75; 95%CI 0.60 to 0.93; P=0.009; five- bl<strong>in</strong>d<strong>in</strong>g<br />

that was considered to be<br />

year survival rate, 36 percent vs. 23 percent)<br />

stage II (through the<br />

and of progression-free survival (HR for<br />

submucosa) or higher,<br />

progression, 0.66; 95%CI 0.53 to 0.81;<br />

<strong>with</strong> no evidence of<br />

P


Graham AH<br />

2007<br />

100 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

[88] October<br />

2004<br />

Patients <strong>with</strong> locally<br />

advanced oesophageal<br />

cancer (T2-3N0M0 or T1-<br />

3N1M0)<br />

Gebski V 2007 [86] 2006 Patients <strong>with</strong> local<br />

operable oesophageal<br />

cancer<br />

Geh JI 2006 [259] 2000 Patients <strong>with</strong> local<br />

operable oesophageal<br />

cancer<br />

Hao D 2006 [109] January<br />

2005<br />

Patients <strong>with</strong> local<br />

operable oesophageal<br />

cancer<br />

Surgery vs.<br />

chemoradiotherapy followed<br />

by surgery vs. chemotherapy<br />

followed by surgery vs.<br />

surgery followed by<br />

chemoradiotherapy<br />

Neoadjuvant<br />

chemoradiotherapy or<br />

neoadjuvant<br />

chemotherapy followed by<br />

surgery vs. surgery alone<br />

Neoadjuvant<br />

chemoradiotherapy followed<br />

by surgery vs. surgery alone<br />

Plat<strong>in</strong>um-based neoadjuvant<br />

chemoradiotherapy<br />

See above 14 RCTs <strong>in</strong>cluded, of<br />

which 6 RCTs on<br />

neoadjuvant<br />

chemoradiotherapy<br />

NO NEW EVIDENCE<br />

SINCE CCO<br />

The hazard ratio for all-cause mortality <strong>with</strong><br />

neoadjuvant chemoradiotherapy versus<br />

surgery alone was 0.81 (95% CI 0.70-0.93;<br />

p=0.002), correspond<strong>in</strong>g to a 13% absolute<br />

difference <strong>in</strong> survival at 2 years, <strong>with</strong> similar<br />

results for different histological tumour types:<br />

0.84 (0.71-0.99; p=0.04) for squamous-cell<br />

carc<strong>in</strong>oma (SCC), and 0.75 (0.59-0.95; p=0.02)<br />

for adenocarc<strong>in</strong>oma.<br />

The probability of pCR improved <strong>with</strong><br />

<strong>in</strong>creas<strong>in</strong>g dose of radiotherapy (P=0.006), 5FU<br />

(P=0.003) and cisplat<strong>in</strong> (P=0.018). Increas<strong>in</strong>g<br />

radiotherapy treatment time (P=0.035) and<br />

<strong>in</strong>creas<strong>in</strong>g median age (P=0.019) reduced the<br />

probability of pCR. The estimated alpha/beta<br />

ratio of oesophageal cancer was 4.9 Gy (95%<br />

confidence <strong>in</strong>terval (CI) 1.5-17 Gy) and the<br />

estimated radiotherapy dose lost per day was<br />

0.59 Gy (95% CI 0.18-0.99 Gy). One gram per<br />

square metre of 5FU was estimated to be<br />

equivalent to 1.9 Gy (95% CI 0.8-5.2 Gy) of<br />

radiation and 100mg/m2 of cisplat<strong>in</strong> was<br />

estimated to be equivalent to 7.2 Gy (95% CI<br />

2.1-28 Gy). Mitomyc<strong>in</strong> C dose did not appear<br />

to <strong>in</strong>fluence pCR rates (P=0.60).<br />

The addition of surgery to CT/RT appeared to<br />

improve local tumour control but not overall<br />

survival <strong>in</strong> patients <strong>with</strong> locally advanced<br />

oesophageal SCC<br />

GUIDELINE!<br />

Only <strong>in</strong>clusion of RCTs<br />

<strong>with</strong> ITT analysis<br />

No results of quality<br />

appraisal reported<br />

10 RCTs on neoadjuvant<br />

chemoradiotherapy<br />

3 RCTs <strong>in</strong> addition to<br />

Graham et al. (Tepper<br />

2006, Burmeister 2005,<br />

Lee 2004)<br />

Tepper 2006 also not<br />

<strong>in</strong>cluded <strong>in</strong> CCO<br />

guidel<strong>in</strong>e, but published<br />

as an abstract<br />

Medl<strong>in</strong>e and meet<strong>in</strong>g<br />

abstracts<br />

Quality appraisal??<br />

26 RCTs + non-RCTs<br />

Same search as for CCO<br />

guidel<strong>in</strong>e<br />

SR High<br />

SR High<br />

SR Moderate<br />

SR High


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 101<br />

Greer SE 2005 [260] January<br />

2003<br />

Patients undergo<strong>in</strong>g<br />

surgery for oesophageal<br />

cancer<br />

Natsugoe S 2006 [89] NA Patients <strong>with</strong> local<br />

operable squamous cell<br />

oesophageal cancer<br />

Burmeister BH<br />

2005<br />

[261] NA Patients <strong>with</strong> histologically<br />

confirmed <strong>in</strong>vasive<br />

cancer of the thoracic<br />

oesophagus<br />

Restag<strong>in</strong>g after neoadjuvant treatment.<br />

Neoadjuvant<br />

chemoradiotherapy followed<br />

by surgery vs. surgery alone<br />

Neoadjuvant<br />

chemoradiotherapy (40 Gy,<br />

cisplat<strong>in</strong> + 5FU) (n = 22) vs.<br />

surgery alone (n = 23)<br />

Neoadjuvant<br />

chemoradiotherapy (35 Gy,<br />

cisplat<strong>in</strong> + 5FU) (n = 128) vs.<br />

surgery alone (n = 128)<br />

Small, non-statistically significant trend toward<br />

improved long-term survival <strong>in</strong> the NCRT<br />

followed by surgery group (relative risk of<br />

death <strong>in</strong> the NCRT group 0.86; 95%CI 0.74 to<br />

1.01; P = .07).<br />

Surgical treatment was performed <strong>in</strong> 20<br />

patients <strong>in</strong> the CRT group except for two<br />

patients <strong>with</strong> bone metastasis after CRT.<br />

Accord<strong>in</strong>g to histological effects of primary<br />

tumors, the number of patient <strong>with</strong> Grades 1,<br />

2 and 3 was 11, 7 and 3, respectively.<br />

Frequency of lymphatic and venous <strong>in</strong>vasion<br />

was significantly lower <strong>in</strong> the CRT group than<br />

<strong>in</strong> the Surgery group. The 5-year survival rate<br />

was 57% <strong>in</strong> the CRT group and 41% <strong>in</strong> the<br />

Surgery group (P = 0.58). Accord<strong>in</strong>g to the<br />

histological effect <strong>in</strong> the CRT group, 5-year<br />

survival was 30% for Grade 1, 83% for Grade 2<br />

and 100% for Grade 3 (P = 0.0069).<br />

Neither progression-free survival nor overall<br />

survival differed between groups (HR 0.82<br />

[95%CI 0.61-1.10] and 0.89 [0.67-1.19],<br />

respectively). The chemoradiotherapy-andsurgery<br />

group had more complete resections<br />

<strong>with</strong> clear marg<strong>in</strong>s than did the surgery-alone<br />

group (103 of 128 [80%] vs 76 of 128 [59%],<br />

p=0.0002), and had fewer positive lymph<br />

nodes (44 of 103 [43%] vs 69 of 103 [67%],<br />

p=0.003).<br />

6 RCTs (same as<br />

Graham 2007)<br />

Quality appraisal??<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

Included <strong>in</strong> CCO<br />

guidel<strong>in</strong>e as an abstract<br />

No bl<strong>in</strong>d<strong>in</strong>g of patients<br />

and cl<strong>in</strong>icians<br />

SR Moderate<br />

RCT Moderate<br />

RCT Moderate<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

FNCLCC [66] La place de la TEP-FDG dans l’évaluation de la réponse au traitement reste à déterm<strong>in</strong>er par des études<br />

prospectives.<br />

Flamen 2002 Low<br />

SIGN [47] 2004 Patients <strong>with</strong> locally advanced disease hav<strong>in</strong>g chemotherapy/chemoradiotherapy should have their<br />

response assessed for an impact on the potential to operate ; follow<strong>in</strong>g a good response the patient<br />

should be restaged and the role restaged and the role of surgery re-evaluated by the multidiscipl<strong>in</strong>ary<br />

team.<br />

Ross P 1996 Low


102 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Duong CP 2006 [262] NA 53 patients <strong>with</strong><br />

FDG-PET<br />

High PET impact (change <strong>in</strong> treatment <strong>in</strong>tent or Standard not Prospective Very low<br />

oesophageal cancer Histology (n = 22) or serial imag<strong>in</strong>g modality) was observed <strong>in</strong><br />

the same for study<br />

referred for PET and cl<strong>in</strong>ical follow up (n = 26) 19 patients (36%). When PET f<strong>in</strong>d<strong>in</strong>gs could be all patients<br />

evaluation of tumour<br />

verified, they were confirmed to be correct <strong>in</strong> 79%<br />

response to CRT<br />

(38 of 48 cases).<br />

Cerfolio RJ 2005 [91] NA Patients <strong>with</strong> suspected or Chest, abdomen, pelvis CT The accuracy of each test for dist<strong>in</strong>guish<strong>in</strong>g pathologic<br />

Prospective Low<br />

biopsy-proved<br />

EUS-FNA<br />

T4 from T1 to T3 disease is 76%, 80%, and 80% for<br />

study<br />

oesophageal carc<strong>in</strong>oma FDG-PET/CT<br />

CT scan, EUS-FNA and FDG-PET/CT, respectively.<br />

treated <strong>with</strong> neoadjuvant 41 patients underwent surgery The accuracy for nodal disease was 78%, 78%, and<br />

chemotherapy (n = 48)<br />

93% for CT scan, EUS-FNA and FDG-PET/CT,<br />

respectively (p=0.04). FDGPET/<br />

CT correctly identified M1b disease <strong>in</strong> 4 patients,<br />

falsely suggested it <strong>in</strong> 4 patients, and missed it <strong>in</strong> 2<br />

patients, whereas for CT, it was 3, 3, and 3 patients.<br />

Fifteen (31%) patients were complete responders, and<br />

FDG-PET/CT accurately predicted complete response<br />

<strong>in</strong> 89% compared <strong>with</strong> 67% for EUS-FNA (p=0.045)<br />

and 71% for CT (p=0.05).<br />

Song SY 2005 [74] NA Patients <strong>with</strong> locally FDG-PET<br />

Sensitivity, specificity, positive predictive value (PPV),<br />

Prospective Low<br />

advanced but resectable Histology<br />

and negative predictive value (NPV) <strong>in</strong> the primary<br />

study<br />

oesophageal cancer<br />

tumours of the preoperative FDG-PET were 27%,<br />

treated <strong>with</strong> neoadjuvant<br />

95%, 75%, and 71%, respectively. In regional lymph<br />

chemoradiotherapy (n =<br />

nodes, these values were 16%, 98%, 36%, and 93%,<br />

32)<br />

respectively.<br />

Br<strong>in</strong>k I 2004 [263] NA 20 patients <strong>with</strong><br />

FDG-PET<br />

Not enough <strong>in</strong>formation to calculate accuracy Prospective Very low<br />

oesophageal cancer CT<br />

study<br />

treated <strong>with</strong> neoadjuvant<br />

chemoradiotherapy<br />

Histology<br />

Nakahara T [264] NA 28 patients <strong>with</strong> squamous Tl-201 SPECT<br />

Although thallium-201 SPECT cannot be used to<br />

Prospective Low<br />

2003<br />

oesophageal cancer Barium exam<strong>in</strong>ation<br />

evaluate the therapeutic effect <strong>with</strong> acceptable<br />

study<br />

CT<br />

accuracy, SPECT may be of additional value to barium<br />

Ga-67 SPECT<br />

swallow and CT <strong>in</strong> assess<strong>in</strong>g the response of AESCC<br />

All studies before and after<br />

preoperative chemoradiotherapy<br />

All patients underwent<br />

oesophagectomy (bl<strong>in</strong>ded<br />

pathologist)<br />

to preoperative chemoradiotherapy.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 103<br />

Surgical treatment.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

SIGN [47] 2004 Oesophageal resectional surgery should be carried out <strong>in</strong> high volume specialist surgical units by frequent van Lanschot JJ 2001 Low<br />

operators.<br />

Bachmann MO 2002<br />

Surgery for oesophageal cancer should be aimed at achiev<strong>in</strong>g an R0 resection. Hulscher JB 2002 High<br />

Follow<strong>in</strong>g oesopagectomy, the route of reconstruction and potential use of pyloric dra<strong>in</strong>age procedure Urschel JD 2001a High<br />

should be determ<strong>in</strong>ed by the surgeon based on their <strong>in</strong>dividual experience.<br />

Urschel JD 2001b<br />

Urschel JD 2002<br />

Walther B 2003<br />

Two-field lymphadenectomy should be considered dur<strong>in</strong>g oesophagectomy to improve stag<strong>in</strong>g and local<br />

disease control.<br />

Dresner SM 2000 Low<br />

Rout<strong>in</strong>e extension of lymphadenectomy <strong>in</strong>to the superior mediast<strong>in</strong>um or neck should not be carried out. Nishihira T 1998 High<br />

CBO [56] July 2003 Totdat vergelijkende data tussen chirurgie en andere behandel<strong>in</strong>gsmodaliteiten of data van grote series<br />

van een andere modaliteit dan chirurgie beschikbaar zijn, kan chirurgische resectie als de<br />

standaardbehandel<strong>in</strong>g voor het <strong>in</strong> opzet curatief resectabele oesofaguscarc<strong>in</strong>oom worden beschouwd.<br />

Hulscher JB 2001 High<br />

Bij grote tumoren zonder metastasen moet worden gestreefd naar een radicale resectie van het<br />

Hulscher JB 2002<br />

High<br />

oesofaguscarc<strong>in</strong>oom. Een palliatieve resectie is <strong>in</strong> het algemeen gecontra<strong>in</strong>diceerd.<br />

van Lanschot JJ 2001<br />

- Tumoren van de proximale oesofagus (gelegen boven de car<strong>in</strong>a) kunnen uitsluitend via een<br />

thoracotomie worden gereseceerd.<br />

Sagar PM 1993<br />

- Voor type-I-tumoren, die gelegen zijn distaal van de car<strong>in</strong>a, heeft een radicale transthoracale<br />

benader<strong>in</strong>g de voorkeur, mits hiermee voldoende ervar<strong>in</strong>g bestaat en de algemene conditie van<br />

de patiënt dit toelaat.<br />

- Gastro-oesofageale type-II-tumoren worden bij voorkeur transhiataal verwijderd.<br />

- Voor een proximaal maagcarc<strong>in</strong>oom type III dat m<strong>in</strong>der dan 1 cm de distale oesofagus <strong>in</strong>groeit,<br />

kan een totale maagresectie worden verricht.<br />

Na subtotale en macroscopisch radicale oesofagusresectie gevolgd door reconstructie met een buismaag Urschel JD 2001<br />

High<br />

en cervicale anastomose heeft positioner<strong>in</strong>g van de buismaag <strong>in</strong> het achterste mediast<strong>in</strong>um de voorkeur. In van Lanschot JJ 1994<br />

geval van macroscopische irradicaliteit heeft reconstructie via het voorste mediast<strong>in</strong>um de voorkeur.<br />

Voor tumoren van de cardia of van de cardio-oesofageale overgang is transhiatale resectie met<br />

Hulscher JB 2002<br />

High<br />

medenemen van de peri-oesofageale en -cardiale klieren voldoende. Bij carc<strong>in</strong>omen van de distale Nishihira T 1998<br />

oesofagus bestaat een lichte voorkeur voor een gecomb<strong>in</strong>eerde abdom<strong>in</strong>ale en transthoracale resectie Matsubara T 1998<br />

met tweevelds-lymfklierdissectie van de abdom<strong>in</strong>ale en thoracale lymfklierstations.<br />

Oesofagustumoren boven car<strong>in</strong>aniveau worden door middel van een gecomb<strong>in</strong>eerde abdom<strong>in</strong>ale en<br />

transthoracale benader<strong>in</strong>g gereseceerd.<br />

Altorki N 2002<br />

Na subtotale oesofagus- (en cardia)resectie heeft de maag als <strong>in</strong>terponaat de voorkeur. Het colon is Urschel JD 2001<br />

High/low<br />

tweede keuze. Slechts <strong>in</strong> uitzonder<strong>in</strong>gsgevallen kan eventueel van het jejunum gebruik worden gemaakt.<br />

Het is op grond van literatuurgegevens niet mogelijk een keuze te maken tussen een totale maag of een<br />

buismaag als <strong>in</strong>terponaat. Om oncologische redenen kan bij een cardia- of distale oesofagustumor slechts<br />

een buismaag worden gebruikt.<br />

Collard JM 1995


104 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

Er is geen duidelijke voorkeur uit te spreken voor een cervicale of thoracale anastomose na<br />

Hulscher JB 2001<br />

High<br />

oesofagusresectie en buismaagreconstructie.<br />

Chasseray VM 1989<br />

- Bij de meeste patiënten is de cervicale anastomose de meest aangewezen procedure. Met name Ribet M 1992<br />

is dit het geval bij patiënten bij wie de chirurg, <strong>in</strong> geval van een tumor onder het car<strong>in</strong>aniveau, Goldm<strong>in</strong>c M 1993<br />

kiest voor een transhiatale resectie en halsexploratie voor het verrichten van de<br />

oesofagusresectie. Er is dan geen reden om tot thoracotomie over te gaan voor het leggen van<br />

een <strong>in</strong>trathoracale anastomose. Wanneer de chirurg bij dit soort tumoren de voorkeur geeft<br />

aan een thoracotomie voor de dissectiefase, kan de anastomose <strong>in</strong>trathoracaal worden<br />

aangelegd en kan de halsfase achterwege worden gelaten.<br />

Chu KM 1997<br />

- Wanneer bij tumoren boven het car<strong>in</strong>aniveau wordt gekozen voor een thoracotomie, is het om<br />

oncologische redenen ter verkrijg<strong>in</strong>g van voldoende proximale marge verstandig ook <strong>in</strong> dat geval<br />

voor een subtotale oesofagusresectie met cervicale anastomose te kiezen.<br />

Zowel handgelegde als mechanische (gestapelde) anastomosen kunnen worden verricht na subtotale Urschel JD 2001<br />

High<br />

oesofagectomie, maag<strong>in</strong>terponaat en oesofagogastrische anastomose.<br />

Craig SR 1996<br />

Valverde A 1996<br />

Law S 1997<br />

Bij een handgelegde oesofagogastrische anastomose na een subtotale oesofagusresectie met<br />

Bard<strong>in</strong>i R 1994a<br />

High<br />

buismaagreconstructie verdient een enkelrijige doorlopende hechttechniek de voorkeur.<br />

Zieren HU 1993<br />

Bard<strong>in</strong>i R 1994b<br />

Muller JM 1990<br />

Na oesofagusresectie gevolgd door buismaag<strong>in</strong>terponaat en oesofagogastrische anastomose is een<br />

pyloroplastiek niet geïndiceerd.<br />

Zieren HU 1995 High<br />

CCO [84] January If surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or adjuvant therapy) is<br />

2005 recommended as the standard practice for resectable thoracic oesophageal cancer.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 105<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Graham AH [88] October Patients <strong>with</strong> locally Surgery vs.<br />

For the first year, the relative risk (95% 14 RCTs <strong>in</strong>cluded SR High<br />

2007<br />

2004 advanced oesophageal chemoradiotherapy followed confidence <strong>in</strong>terval) of death for The 1 RCT on adjuvant<br />

cancer (T2-3N0M0 or T1- by surgery vs. chemotherapy treatments compared <strong>with</strong> surgery were CRT vs. surgery alone<br />

3N1M0)<br />

followed by surgery vs. 0.87 (0.75 to 1.02) for<br />

<strong>in</strong>cluded patients <strong>with</strong><br />

surgery followed by<br />

chemoradiotherapy followed by surgery, gastric cancer or cancer<br />

chemoradiotherapy<br />

0.94 (0.82 to 1.08) for chemotherapy<br />

followed by surgery, and 1.33 (0.93 to<br />

1.93) for surgery <strong>with</strong> adjuvant<br />

chemoradiotherapy. The QALYs ga<strong>in</strong>ed<br />

for surgery alone, chemoradiotherapy<br />

followed by surgery, chemotherapy<br />

followed by surgery, and surgery <strong>with</strong><br />

adjuvant chemoradiotherapy strategies<br />

were 2.07, 2.18, 2.14, and 1.99,<br />

respectively. If the reduction <strong>in</strong> utility<br />

for multimodality treatment was<br />

<strong>in</strong>creased to 21%, the QALYs ga<strong>in</strong>ed for<br />

surgery alone, chemoradiotherapy<br />

followed by surgery, chemotherapy<br />

followed by surgery, and surgery <strong>with</strong><br />

adjuvant chemoradiotherapy were 2.07,<br />

2.03, 1.99, and 1.85, respectively.<br />

of the GEJ<br />

Bedenne L 2007 [93] NA Patients <strong>with</strong> operable Arm A: Surgery (n = 129) Two-year survival rate: 34% <strong>in</strong> arm A Two types of<br />

RCT Moderate<br />

Bonneta<strong>in</strong> F<br />

T3N0-1M0 thoracic Arm B: Cont<strong>in</strong>uation of versus 40% <strong>in</strong> arm B (HR for arm B vs. radiotherapy<br />

2006<br />

oesophageal cancer, and chemoradiation (three cycles arm A = 0.90; adjusted P = .44). Not clear if bl<strong>in</strong>ded<br />

<strong>with</strong> response to of FU/cisplat<strong>in</strong> and either Median survival time: 17.7 months <strong>in</strong> randomization<br />

chemoradiation (two conventional [20 Gy] or split- arm A, 19.3 months <strong>in</strong> arm B.<br />

No <strong>in</strong>formation on<br />

cycles of FU/cisplat<strong>in</strong> and course [15 Gy] radiotherapy) Two-year local control rate: 66.4% <strong>in</strong> bl<strong>in</strong>d<strong>in</strong>g of patients,<br />

either conventional (46 (n = 130)<br />

arm A, 57.0% <strong>in</strong> arm B<br />

cl<strong>in</strong>icians and assessors<br />

Gy <strong>in</strong> 4.5 weeks) or split- vs.<br />

Stents were less required <strong>in</strong> the surgery<br />

course (15 Gy, days 1 to 5<br />

arm (5% <strong>in</strong> arm A vs. 32% <strong>in</strong> arm B; P <<br />

and 22 to 26) concomitant<br />

.001).<br />

radiotherapy) and no<br />

3-month mortality rate: 9.3% <strong>in</strong> arm A,<br />

contra<strong>in</strong>dication to either<br />

0.8% <strong>in</strong> arm B (P = .002).<br />

treatment (n = 444, of<br />

Cumulative hospital stay: 68 days <strong>in</strong> arm<br />

which 259 were randomly<br />

assigned)<br />

A, 52 days <strong>in</strong> arm B (P = .02).


106 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Stahl M 2005 [265] NA Patients <strong>with</strong> locally Induction chemotherapy Overall survival was equivalent between Unbl<strong>in</strong>ded study RCT Moderate<br />

advanced squamous cell followed by<br />

the two treatment groups (log-rank test<br />

carc<strong>in</strong>oma of the chemoradiotherapy (40 Gy) for equivalence, P < .05). Local<br />

oesophagus<br />

followed by surgery (n = 86), progression-free survival was better <strong>in</strong><br />

or the same <strong>in</strong>duction the surgery group (2-year progression-<br />

chemotherapy followed by free survival, 64.3%; 95%CI 52.1% to<br />

chemoradiotherapy (at least 76.5%) than <strong>in</strong> the chemoradiotherapy<br />

65 Gy) <strong>with</strong>out surgery (n = group (2-year progression-free survival,<br />

86).<br />

40.7%; 95%CI 28.9% to 52.5%; hazard<br />

ratio [HR] for arm B v arm A, 2.1; 95%<br />

CI, 1.3 to 3.5; P = .003). Treatmentrelated<br />

mortality was significantly<br />

<strong>in</strong>creased <strong>in</strong> the surgery group than <strong>in</strong><br />

the chemoradiotherapy group (12.8% v<br />

3.5%, respectively; P = .03).<br />

Chiu PWY 2005 [94] NA Patients <strong>with</strong> potentially Oesophagectomy (n = 44) vs. No difference <strong>in</strong> the early cumulative No <strong>in</strong>formation on RCT Moderate<br />

(CURE)<br />

resectable squamous cell chemoradiotherapy<br />

survival (RR = 0.89; 95%CI 0.37-2.17; randomization procedure<br />

carc<strong>in</strong>oma of the mid or (cont<strong>in</strong>uous 5-FU (200 log-rank test P = 0.45).<br />

No <strong>in</strong>formation on<br />

lower thoracic<br />

mg/m2/day) from day 1 to 42 No difference <strong>in</strong> the disease-free bl<strong>in</strong>d<strong>in</strong>g of patients,<br />

oesophagus<br />

and cisplat<strong>in</strong> (60 mg/m2) on<br />

days 1 and 22; tumour and<br />

regional lymphatics were<br />

concomitantly irradiated to a<br />

total of 50-60 Gy) (n = 36)<br />

survival.<br />

cl<strong>in</strong>icians and assessors<br />

Bhat MA 2006 [266] NA 238 patients treated for Oesophagogastrectomy <strong>with</strong> Anastomotic leaks occurred <strong>in</strong> 3 group No <strong>in</strong>formation on RCT Moderate<br />

oesophageal carc<strong>in</strong>oma wrapp<strong>in</strong>g of the pedicled A patients (3.09%) and <strong>in</strong> 14 (14.43%) bl<strong>in</strong>d<strong>in</strong>g of assessors<br />

(44 excluded because omentum around the group B patients. The difference <strong>in</strong> the<br />

<strong>in</strong>operable)<br />

esophagogastric anastomosis <strong>in</strong>cidence of leakage was statistically<br />

(n = 97) vs.<br />

significant (p = 0.005). There was no<br />

oesophagogastrectomy complication related to the omental<br />

<strong>with</strong>out us<strong>in</strong>g the omental graft technique nor was there a<br />

graft (n = 97)<br />

significant difference <strong>in</strong> the mortality<br />

between the two groups.<br />

Hsu HH 2004 [267] NA Patients <strong>with</strong> curatively Hand-sewn (n = 32) or The mean operat<strong>in</strong>g time was longer Randomization<br />

RCT Moderate<br />

resectable squamous cell circular stapled (n = 31) when the hand-sewn method was used procedure??<br />

cancer of the thoracic cervical oesophagogastric (524 vs. 447 m<strong>in</strong>, P < 0.001).<br />

Bl<strong>in</strong>d<strong>in</strong>g??<br />

oesophagus (T1-3 N0-1) anastomosis<br />

Anastomotic leakage was noted <strong>in</strong> seven<br />

patients (22%) <strong>in</strong> the hand-sewn group<br />

and eight patients (26%) <strong>in</strong> the stapler<br />

group (P = NS). Hospital mortality


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 107<br />

Study ID Ref Search<br />

date<br />

Walther B 2003 [268] NA Patients <strong>with</strong> benign or<br />

malignant oesophageal<br />

disease for which a tube<br />

gastroplasty <strong>with</strong><br />

anastomotic site <strong>in</strong> the<br />

proximal chest or <strong>in</strong> the<br />

neck was deemed suitable<br />

(n = 83, of which 74 had<br />

malignant disease)<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

occurred <strong>in</strong> four patients (13%) of the<br />

hand-sewn group and <strong>in</strong> three patients<br />

(10%) of the stapler group (P = NS).<br />

After the operation, four patients (14%)<br />

<strong>in</strong> the hand-sewn group and five patients<br />

(18%) <strong>in</strong> the stapler group developed a<br />

benign oesophageal stricture (P = NS).<br />

The mean follow-up time was 24<br />

months, and the rates of freedom from<br />

benign stricture and survival were<br />

Manually sutured<br />

oesophagogastric anastomosis<br />

<strong>in</strong> the neck or an anastomosis<br />

stapled <strong>in</strong> the right chest<br />

comparable <strong>in</strong> each group.<br />

The genu<strong>in</strong>e 5-year survival rate was<br />

29% for chest anastomoses and 30% for<br />

neck anastomoses. The overall leakage<br />

rate was 1.8% (2 cases of 112) <strong>with</strong> no<br />

relation to mortality or anastomotic<br />

method. All patients <strong>in</strong> the randomized<br />

group had tumor-free proximal and<br />

distal resection l<strong>in</strong>es, but 1 patient <strong>in</strong> the<br />

nonrandomized group had tumor<br />

<strong>in</strong>filtrates <strong>in</strong> the proximal resection<br />

marg<strong>in</strong>. At 3, 6, and 12 months after<br />

operation, there was no difference <strong>in</strong><br />

anastomotic diameter between the<br />

esophagogastric anastomosis <strong>in</strong> the neck<br />

and <strong>in</strong> the thorax (P = 0.771), and both<br />

<strong>in</strong>creased <strong>with</strong> time (P = 0.004, ANOVA<br />

repeated measures). Body weight<br />

development was the same <strong>in</strong> the two<br />

groups. With similar results <strong>in</strong><br />

randomized and nonrandomized<br />

patients, study bias was elim<strong>in</strong>ated.<br />

Randomization <strong>with</strong><br />

sealed envelopes<br />

Also 29 non-randomized<br />

patients <strong>in</strong>cluded <strong>in</strong><br />

results<br />

RCT Moderate


108 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Adjuvant treatment.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

Chemotherapy<br />

SIGN [47] 2004 Postoperative adjuvant chemotherapy is not recommended for patients <strong>with</strong> oesophageal cancer. Lehnert T 1999 High<br />

CBO [56] July Patiënten met een oesofaguscarc<strong>in</strong>oom komen niet <strong>in</strong> aanmerk<strong>in</strong>g voor postoperatieve chemotherapie. Ando N 1997<br />

High<br />

2003<br />

Pouliquen X 1996<br />

CCO [84] January If surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or adjuvant therapy) is Ando N 1997<br />

High<br />

2005 recommended as the standard practice for resectable thoracic oesophageal cancer.<br />

Pouliquen X 1996<br />

Ando N 2003<br />

Radiotherapy<br />

SIGN [47] 2004 No recommendation, only discussion<br />

CBO [56] July<br />

2003<br />

CCO [84] January<br />

2005<br />

Chemoradiotherapy<br />

Patiënten met een oesofaguscarc<strong>in</strong>oom komen niet <strong>in</strong> aanmerk<strong>in</strong>g voor postoperatieve radiotherapie. Fok M 1993<br />

Teniere P 1991<br />

Zieren HU 1995<br />

If surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or adjuvant therapy) is<br />

recommended as the standard practice for resectable thoracic oesophageal cancer.<br />

Xiao ZF 2003<br />

Fok M 1993<br />

Fok M 1994<br />

Teniere P 1991<br />

Zieren HU 1995<br />

Xiao ZF 2003<br />

SIGN [47] 2004 Postoperative adjuvant chemoradiotherapy is not recommended for patients <strong>with</strong> oesophageal cancer. No evidence<br />

CCO [84] January If surgery is considered appropriate, then surgery alone (i.e., <strong>with</strong>out neoadjuvant or adjuvant therapy) is No evidence<br />

2005 recommended as the standard practice for resectable thoracic oesophageal cancer.<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence<br />

Chemotherapy<br />

Ajani J 2006 [269] October<br />

2005<br />

Radiotherapy<br />

No additional studies found<br />

Chemoradiotherapy<br />

No additional studies found<br />

Patients oesophageal<br />

cancer<br />

Oral capecitab<strong>in</strong>e 4 trials identified that <strong>in</strong>cluded patients<br />

<strong>with</strong> oesophageal cancer<br />

No conclusions to be drawn<br />

Medl<strong>in</strong>e search only<br />

No <strong>in</strong>formation on<br />

selection, quality<br />

appraisal, etc<br />

High<br />

High<br />

SR Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 109<br />

Non-surgical treatment <strong>with</strong> curative <strong>in</strong>tent.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

SIGN [47] 2004 Chemoradiotherapy should be considered <strong>in</strong> patients <strong>with</strong> esophageal cancer who have locally Wong R '03<br />

High<br />

advanced disease, those unfit for surgery or those who decl<strong>in</strong>e surgery.<br />

Cooper JS '99<br />

CCO [108] February (In adult patients <strong>with</strong> localized (T1-3, small volume N1, M0) carc<strong>in</strong>oma of the esophagus and good Wong R ‘06 High<br />

2005 performance status who are consider<strong>in</strong>g a non-surgical approach and for whom comb<strong>in</strong>ed<br />

radiotherapy and chemotherapy can be tolerated <strong>in</strong> the judgment of the treat<strong>in</strong>g oncologist)<br />

concomitant radiotherapy and chemotherapy is recommended over radiotherapy alone. Based on<br />

considerations of the current cl<strong>in</strong>ical practice pattern and the currently available research evidence, a<br />

cisplat<strong>in</strong>-based chemotherapy regimen is a reasonable chemotherapy regimen to use when<br />

concomitant radiotherapy and chemotherapy is used.<br />

Patients should be made aware of the <strong>in</strong>creased acute toxicity associated <strong>with</strong> this approach. The<br />

decision to use concomitant radiotherapy and chemotherapy should only be made after careful<br />

consideration of the potential risks, benefits, and the patient’s general condition.<br />

Wong R ‘06 High<br />

Sequential radiotherapy and chemotherapy is not recommended as standard practice. Wong R ‘06 High<br />

Future cl<strong>in</strong>ical trials to better def<strong>in</strong>e the optimal chemoradiotherapy comb<strong>in</strong>ation that would improve<br />

outcomes while limit<strong>in</strong>g toxicities are strongly encouraged.<br />

Wong R ‘06 High<br />

CBO [56] July 2003 De werkgroep adviseert om patiënten met een oesofaguscarc<strong>in</strong>oom die niet voor een resectie <strong>in</strong> Wong R '03<br />

High<br />

aanmerk<strong>in</strong>g komen, <strong>in</strong>dien hun conditie dit toelaat, te behandelen met concomitante<br />

chemoradiotherapie, zo mogelijk <strong>in</strong> onderzoeksverband.<br />

Cooper JS '99


110 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Bedenne L 2007 [93] NA Patients <strong>with</strong> operable Arm B: Cont<strong>in</strong>uation of Two-year survival rate: 34% <strong>in</strong> arm A Two types of<br />

RCT Moderate<br />

T3N0-1M0 thoracic chemoradiation (three cycles versus 40% <strong>in</strong> arm B (HR for arm B vs. radiotherapy<br />

oesophageal cancer, and of FU/cisplat<strong>in</strong> and either arm A = 0.90; adjusted P = .44). Not clear of bl<strong>in</strong>ded<br />

<strong>with</strong> response to conventional [20 Gy] or split- Median survival time: 17.7 months <strong>in</strong> randomization<br />

chemoradiation (two course [15 Gy] radiotherapy) arm A, 19.3 months <strong>in</strong> arm B.<br />

No <strong>in</strong>formation on<br />

cycles of FU/cisplat<strong>in</strong> and (n = 130)<br />

Two-year local control rate: 66.4% <strong>in</strong> bl<strong>in</strong>d<strong>in</strong>g of patients,<br />

either conventional (46 vs.<br />

arm A, 57.0% <strong>in</strong> arm B<br />

cl<strong>in</strong>icians and assessors<br />

Gy <strong>in</strong> 4.5 weeks) or split- Arm A: surgery (n = 129) Stents were less required <strong>in</strong> the surgery<br />

course (15 Gy, days 1 to 5<br />

arm (5% <strong>in</strong> arm A vs. 32% <strong>in</strong> arm B; P <<br />

and 22 to 26) concomitant<br />

.001).<br />

radiotherapy) and no<br />

3-month mortality rate: 9.3% <strong>in</strong> arm A,<br />

contra<strong>in</strong>dication to either<br />

0.8% <strong>in</strong> arm B (P = .002).<br />

treatment (n = 444, of<br />

Cumulative hospital stay: 68 days <strong>in</strong> arm<br />

which 259 were randomly<br />

assigned)<br />

A, 52 days <strong>in</strong> arm B (P = .02).<br />

Hao D 2006 [109] January Patients <strong>with</strong> local Plat<strong>in</strong>um-based<br />

To date, no randomized trials have Same search as for CCO SR High<br />

2005 operable oesophageal chemoradiotherapy<br />

directly compared CT/RT <strong>with</strong> guidel<strong>in</strong>e<br />

cancer<br />

oesophagectomy as primary<br />

locoregional treatment of oesophageal<br />

carc<strong>in</strong>oma.<br />

Chiu PW 2005 [94] NA Patients <strong>with</strong> potentially Oesophagectomy (n = 44) vs. No difference <strong>in</strong> the early cumulative No <strong>in</strong>formation on RCT Moderate<br />

(CURE)<br />

resectable squamous cell chemoradiotherapy<br />

survival (RR = 0.89; 95%CI 0.37-2.17; randomization procedure<br />

carc<strong>in</strong>oma of the mid or (cont<strong>in</strong>uous 5-FU (200 log-rank test P = 0.45).<br />

No <strong>in</strong>formation on<br />

lower thoracic<br />

mg/m2/day) from day 1 to 42 No difference <strong>in</strong> the disease-free bl<strong>in</strong>d<strong>in</strong>g of patients,<br />

oesophagus<br />

and cisplat<strong>in</strong> (60 mg/m2) on<br />

days 1 and 22; tumour and<br />

regional lymphatics were<br />

concomitantly irradiated to a<br />

total of 50-60 Gy) <strong>with</strong> salvage<br />

surgery (n = 36)<br />

survival.<br />

cl<strong>in</strong>icians and assessors<br />

Zhao KL 2005 [110] NA Patients <strong>with</strong> confirmed LCAF-RT (n = 57) vs. LCAF- Median survival: 23.9 months (95%CI No <strong>in</strong>formation on RCT Moderate<br />

SCC of the oesophagus RT + chemotherapy (four 20.1-27.7) for the LCAF arm and 30.8 bl<strong>in</strong>d<strong>in</strong>g of patients,<br />

<strong>with</strong> cl<strong>in</strong>ical stage T1-4, cycles of cisplat<strong>in</strong> 25 mg/m(2) months (95%CI 17.6-44.1) for the cl<strong>in</strong>icians and assessors<br />

N0-1, M0 (n = 111) daily and fluorouracil (5-FU) LCAF+CT arm.<br />

600 mg/m(2) daily on Days 1-3 Survival rates at 1, 3, and 5 years of the<br />

every 4 weeks start<strong>in</strong>g on the LCAF arm were 77%, 39%, and 28%,<br />

same day that LCAF was respectively, while those of the<br />

delivered) (n = 54)<br />

LCAF+CT arm were 67%, 44%, and


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 111<br />

Study ID Ref Search<br />

date<br />

Sumpter K 2005 [270] NA Patients <strong>with</strong> <strong>in</strong>operable,<br />

histologically verified<br />

locally advanced or<br />

metastatic<br />

adenocarc<strong>in</strong>oma,<br />

squamous cell or<br />

undifferentiated<br />

carc<strong>in</strong>oma of the<br />

oespohagus,<br />

oesophagogastric junction<br />

(OGJ) or stomach<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

40%, respectively (p = 0.310).<br />

Grades 3 and 4 acute toxicities<br />

occurred <strong>in</strong> 46% and 25% of the patients<br />

<strong>in</strong> the LCAF arm and the LCAF+CT<br />

arm, respectively; 6% of the patients <strong>in</strong><br />

the comb<strong>in</strong>ed arm had Grade 5 acute<br />

toxicities, whereas none was noted <strong>in</strong><br />

ECF (n = 53)<br />

ECX (n = 48)<br />

EOF (n = 55)<br />

EOX (n = 48)<br />

(E = epirubuc<strong>in</strong>, C = cisplat<strong>in</strong>,<br />

F = 5FU, O = oxaliplat<strong>in</strong>, X =<br />

capecitab<strong>in</strong>)<br />

the LCAF alone arm.<br />

First <strong>in</strong>terim analysis was performed<br />

when 80 pts had been randomised.<br />

Doselimit<strong>in</strong>g fluoropyrimid<strong>in</strong>e toxicities<br />

were stomatitis, palmar plantar<br />

erythema (PPE) and diarrhoea; 5.1% of<br />

X-treated pts experienced grade 3/4<br />

toxicity. Protocol planned dose<br />

escalation of X to 625 mg/m² b.i.d. was<br />

<strong>in</strong>stituted and a second <strong>in</strong>terim analysis<br />

has been performed. A total of 204 pts<br />

were randomised at the time of the<br />

protocol planned 2nd <strong>in</strong>terim analysis.<br />

Grade 3/4 fluoropyrimid<strong>in</strong>e-related<br />

toxicity was seen <strong>in</strong> 13.7% pts receiv<strong>in</strong>g<br />

F, 8.4% pts receiv<strong>in</strong>g X 500 mg/m² b.i.d.<br />

and 14.7% pts receiv<strong>in</strong>g X 625 mg/m²<br />

b.i.d. Comb<strong>in</strong>ed complete and partial<br />

response rates were ECF 31% (95% CI<br />

18.7–46.3), EOF 39% (95% CI 25.9–<br />

53.1), ECX 35% (95% CI 21.4–50.3),<br />

EOX 48% (95% CI 33.3–62.8). Grade<br />

3/4 fluoropyrimid<strong>in</strong>e toxicity affected<br />

14.7% of pts treated <strong>with</strong> X 625 mg/m²<br />

b.i.d., which is similar to that observed<br />

<strong>with</strong> F, confirm<strong>in</strong>g this to be the optimal<br />

dose. The replacement of C by O and F<br />

by X does not appear to impair efficacy.<br />

The trial cont<strong>in</strong>ues to total accrual of<br />

1000 pts.<br />

Interim analysis<br />

6 orig<strong>in</strong>ally randomized<br />

pts not <strong>in</strong>cluded <strong>in</strong> results<br />

Non<strong>in</strong>feriority study<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g of randomization<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g of patients,<br />

cl<strong>in</strong>icians and assessors<br />

RCT Moderate


112 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG<br />

ID<br />

General<br />

Palliative treatment.<br />

Ref Search<br />

date<br />

CBO [56] July<br />

2003<br />

Endoscopic ablation<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

Afhankelijk van de patiëntkarakteristieken en de tumoruitbreid<strong>in</strong>g zal bij patiënten met een T4oesofaguscarc<strong>in</strong>oom<br />

bij voorkeur worden gekozen voor een korte palliatieve behandel<strong>in</strong>g of een<br />

behandel<strong>in</strong>g gericht op palliatie op de langere termijn, zoals (chemo)radiotherapie. Patiënten met een<br />

fistel komen, afhankelijk van de plaats en de grootte van de fistel, <strong>in</strong> aanmerk<strong>in</strong>g voor stentplaats<strong>in</strong>g.<br />

SIGN [47] 2004 Laser or photodynamic therapy should be used for <strong>in</strong>itial control of obstructive symptoms caused by<br />

exophytic tumours <strong>in</strong> the oesophagus <strong>in</strong>clud<strong>in</strong>g tumours near the upper oesophageal sph<strong>in</strong>cter.<br />

Stent<strong>in</strong>g<br />

Laser or photodynamic therapy should be considered for control of tumour overgrowth <strong>in</strong> stented<br />

patients.<br />

CBO [56] July Indien de levensverwacht<strong>in</strong>g korter is dan zes weken, kan allereerst stentplaats<strong>in</strong>g worden overwogen,<br />

2003 gezien het snelle effect op de verbeter<strong>in</strong>g van passageklachten.<br />

Bij patiënten met een fistel tussen de oesofagus en de luchtwegen, patiënten met een <strong>in</strong>operabel<br />

oesofaguscarc<strong>in</strong>oom nabij de bovenste oesofagussf<strong>in</strong>cter en patiënten met een recidief na eerdere<br />

oesofagusresectie <strong>in</strong> verband met een oesofaguscarc<strong>in</strong>oom heeft stentplaats<strong>in</strong>g de voorkeur.<br />

Het verdient aanbevel<strong>in</strong>g om, na stentplaats<strong>in</strong>g <strong>in</strong> verband met passageklachten ten gevolge van een<br />

<strong>in</strong>operabel oesofaguscarc<strong>in</strong>oom, rustig te eten, goed te kauwen en veel te dr<strong>in</strong>ken om verstopp<strong>in</strong>g van<br />

de stent te voorkomen. Bij verstopp<strong>in</strong>g van de stent kan deze snel worden gere<strong>in</strong>igd door middel van<br />

een endoscopie.<br />

SIGN [47] 2004 Partially covered self-expand<strong>in</strong>g metal stents are the <strong>in</strong>tubation of choice for patients <strong>with</strong> obstructive<br />

oesophageal symptoms.<br />

Dilatation<br />

Partially covered self-expand<strong>in</strong>g metal stents should be used to control obstructive oesophageal<br />

symptoms either follow<strong>in</strong>g or <strong>in</strong>stead of laser therapy, depend<strong>in</strong>g on the availability of local expertise.<br />

Allum WH 2002<br />

O’Donnell CA 2002<br />

Dallal HJ 2001<br />

Lightdale CJ 1995<br />

Carazzone A 1999<br />

Heier SK 1995<br />

Moghissi K 2000<br />

Litle VR 2003<br />

Homs MY 2004<br />

May A 1998<br />

Dumonceau JM 1999<br />

Siersema PD 2001<br />

Homs MY 2004a<br />

Sabharwal T 2003<br />

Vakil N 2001<br />

Bethge N 1996<br />

Homs MY 2004b<br />

Low<br />

High<br />

Low<br />

High<br />

Moderate<br />

Moderate<br />

SIGN [47] 2004 The use of oesophageal dilatation alone should be avoided. Allum WH 2002 Very low<br />

Surgery<br />

SIGN [47] 2004 Oesophagectomy (transthoracic or transhiatal) should not be performed <strong>with</strong> palliative <strong>in</strong>tent <strong>in</strong><br />

patients <strong>with</strong> oesophageal cancer.<br />

Blazeby JM 2000 Low<br />

Substernal bypass for oesophageal cancer should not be performed <strong>with</strong> palliative <strong>in</strong>tent. Whooley BP 2002<br />

Alcantara PS 1997<br />

Low<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 113<br />

CPG Ref Search<br />

ID<br />

date<br />

Chemotherapy<br />

CBO [56] July<br />

2003<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

Chemotherapie is geen standaardbehandel<strong>in</strong>g voor het irresectabele of gemetastaseerde<br />

oesofaguscarc<strong>in</strong>oom.<br />

SIGN [47] 2004 In patients <strong>with</strong> locally advanced or metastatic cancer of the oesophagus <strong>with</strong> good performance status<br />

comb<strong>in</strong>ation chemotherapy <strong>in</strong>clud<strong>in</strong>g cisplat<strong>in</strong> and <strong>in</strong>fusional 5FU (such as ECF or MCF) should be<br />

considered.<br />

Radiotherapy<br />

CBO [56] July<br />

2003<br />

Bij oesofaguscarc<strong>in</strong>oompatiënten met een matige algemene conditie en/of met metastasen op afstand<br />

bestaat een voorkeur voor brachytherapie, eventueel gevolgd door een stentplaats<strong>in</strong>g <strong>in</strong>dien<br />

brachytherapie onvoldoende effect heeft.<br />

Bij <strong>in</strong>operabele patiënten met een levensverwacht<strong>in</strong>g langer dan drie maanden kan uitwendige<br />

radiotherapie <strong>in</strong> comb<strong>in</strong>atie met brachytherapie worden overwogen.<br />

Schmid EU 1993<br />

Moderate<br />

Levard H 1998<br />

Mannell A 1986<br />

Webb A 1997<br />

Ross P 2002<br />

Bleiberg H 1997<br />

Janunger KG 2002 High<br />

Homs MY 2004 High<br />

Flores AD 1989<br />

Low<br />

Hishikawa Y 1991<br />

Taal BG 1996<br />

Allum WH 2002 Very low<br />

SIGN [47] 2004 Palliative external-beam radiotherapy is an appropriate option for the treatment of mild dysphagia <strong>in</strong><br />

patients <strong>with</strong> oesophageal cancer.<br />

Endolum<strong>in</strong>al brachytherapy is an option for patients <strong>with</strong> dysphagia from oesophageal cancer. Brewster AE 1995<br />

Homs MYV 2003<br />

Supportive care<br />

CBO [56] July<br />

2003<br />

De werkgroep is van men<strong>in</strong>g dat de verpleegkundige zorg voor patiënten met een oesofaguscarc<strong>in</strong>oom<br />

het beste door een verpleegkundige met specifieke kennis en ervar<strong>in</strong>g kan worden verricht.<br />

Dieetmaatregelen bij patiënten met een <strong>in</strong>operabel oesofaguscarc<strong>in</strong>oom kunnen het beste zijn gericht<br />

op het welbev<strong>in</strong>den van de patiënt, dat wil zeggen dat de kwaliteit van leven belangrijker is dan de<br />

effecten van voed<strong>in</strong>g op lange termijn.<br />

SIGN [47] 2004 Patients <strong>with</strong> oesophageal cancer should have access to a specialist palliative care team. Smeenk FW 1998<br />

Micc<strong>in</strong>esi G 2003<br />

Costant<strong>in</strong>i M 2003<br />

Costant<strong>in</strong>i M 1993<br />

Low<br />

Doornik N 2000 Very low<br />

Low


114 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

Endoscopic ablation<br />

No additional studies found<br />

Stent<strong>in</strong>g<br />

Wenger U 2006 [271] NA Patients <strong>with</strong> <strong>in</strong>curable<br />

cancer of the distal<br />

oesophagus or gastric<br />

cardia (n = 41)<br />

Wenger U 2005<br />

Bergquist H<br />

2005<br />

[112] NA Patients <strong>with</strong> <strong>in</strong>curable<br />

cancer of the oesophagus<br />

or gastro-oesophageal<br />

junction<br />

Antireflux stent (n = 19) vs.<br />

standard stent (n = 22)<br />

Self-expandable metallic stent<br />

(n = 30) or 3 x 7 Gy<br />

brachytherapy (n = 30)<br />

Fewer patients <strong>with</strong> complications were<br />

observed <strong>in</strong> the antireflux stent group (n<br />

= 3) than <strong>in</strong> the standard group (n = 8),<br />

but no statistically significant difference<br />

was shown (p = 0.14). The survival rates<br />

were similar <strong>in</strong> the two groups (p =<br />

0.99; hazard ratio, 1.0; 95% confidence<br />

<strong>in</strong>terval, 0.5-2.0). The groups did not<br />

differ significantly <strong>in</strong> terms of studied<br />

oesophageal or respiratory symptoms<br />

or quality of life. Cl<strong>in</strong>ically relevant<br />

improvement <strong>in</strong> dysphagia occurred <strong>in</strong><br />

both groups. Dyspnea decreased after<br />

antireflux stent <strong>in</strong>sertion (mean score<br />

change, -11), and <strong>in</strong>creased after<br />

<strong>in</strong>sertion of standard stent (mean score<br />

change, +21).<br />

The group of patients treated <strong>with</strong> stent<br />

reported significantly better HRQL<br />

scores for dysphagia (P < 0.05) at the 1month<br />

follow-up, but most other HRQL<br />

scores, <strong>in</strong>clud<strong>in</strong>g function<strong>in</strong>g and<br />

symptom scales, deteriorated.<br />

There was no difference <strong>in</strong> survival or<br />

complication rates between the two<br />

treatment strategies. There was a<br />

significant difference <strong>in</strong> the change of<br />

dysphagia scores between the time of<br />

<strong>in</strong>clusion and the 1-month follow-up<br />

visit, <strong>in</strong> favour of the stented group (P =<br />

0.03). This difference had disappeared at<br />

3 months.<br />

Median total lifetime costs were 17,690<br />

for the stented group compared <strong>with</strong><br />

Interim report<br />

Central randomization,<br />

but no <strong>in</strong>formation on<br />

procedure<br />

Patients were bl<strong>in</strong>ded, but<br />

no <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g of cl<strong>in</strong>icians<br />

Orig<strong>in</strong>ally 65 randomized,<br />

but 5 <strong>with</strong>drew consent<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

RCT Moderate<br />

RCT<br />

Cost study<br />

Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 115<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

33,171 for the brachytherapy group (P =<br />

0.005). This difference was due to higher<br />

costs for the <strong>in</strong>itial treatment (4,615<br />

versus 23,857, P < 0.0001). Sensitivity<br />

analyses showed that the charges for a<br />

brachytherapy session had to be<br />

reduced from 6,092 to 4,222 (31%) to<br />

make this therapeutic concept cost-<br />

Shim CS 2005 [272] NA Patients <strong>with</strong> cancer at the<br />

oesophagogastric junction<br />

(n = 36)<br />

Shenf<strong>in</strong>e J 2005 [273] NA Patients <strong>with</strong> oesophageal<br />

cancer deemed unsuitable<br />

for surgery (n = 217)<br />

Newly designed antireflux<br />

stent (n = 12), a Dostent (n =<br />

12), or a standard open stent<br />

(n = 12)<br />

SEMS 18 mm (n = 54)<br />

SEMS 24 mm (n = 54)<br />

Rigid <strong>in</strong>tubation (n = 57)<br />

All other non-SEMS<br />

treatments (n = 52)<br />

competitive.<br />

After 1 week, dysphagia had improved <strong>in</strong><br />

all patient groups (P < 0.05), but the<br />

degree of improvement did not differ<br />

between the three groups. The<br />

DeMeester score was significantly lower<br />

<strong>in</strong> the group <strong>with</strong> the newly designed<br />

antireflux stent than <strong>in</strong> the other groups.<br />

The fraction of the total record<strong>in</strong>g time<br />

dur<strong>in</strong>g which oesophageal pH was below<br />

4 was 3.14 +/- 5.78 % us<strong>in</strong>g the newly<br />

designed antireflux stent, <strong>in</strong> comparison<br />

<strong>with</strong> 29.25 +/- 15.41 % <strong>in</strong> the Dostent<br />

group and 15.01 +/- 11.72 % <strong>in</strong> the<br />

standard open stent group (P < 0.001).<br />

Fewer reflux episodes occurred <strong>with</strong> the<br />

newly designed antireflux stent than<br />

<strong>with</strong> the Dostent or standard open<br />

stent. There were no complications <strong>with</strong><br />

any of the three stents.<br />

There was no difference <strong>in</strong> cost or<br />

effectiveness between SEMS and non-<br />

SEMS therapies, and 18-mm SEMS had<br />

equal effectiveness to, but less<br />

associated pa<strong>in</strong> than, 24-mm SEMS. Rigid<br />

<strong>in</strong>tubation was associated <strong>with</strong> a worse<br />

quality of swallow<strong>in</strong>g and <strong>in</strong>creased late<br />

morbidity. Bipolar electrocoagulation<br />

and ethanol tumour necrosis were poor<br />

<strong>in</strong> primary palliation. A survival<br />

advantage was found for non-stent<br />

therapies, but there was a significant<br />

No <strong>in</strong>formation on<br />

randomization procedure<br />

or bl<strong>in</strong>d<strong>in</strong>g<br />

Secondary randomization<br />

possible after treatment<br />

failure follow<strong>in</strong>g non-<br />

SEMS treatment<br />

Correct randomization<br />

protocol<br />

RCT Moderate<br />

RCT<br />

Cost study<br />

High


116 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

delay to treatment. The length of stay<br />

accounts for the majority of the cost to<br />

the NHS. Patients were found to have<br />

Pol<strong>in</strong>der S 2004<br />

(SIREC trial)<br />

Dilatation<br />

No additional studies found<br />

Surgery<br />

No additional studies found<br />

Chemotherapy<br />

Homs MY 2006 [114] February<br />

2006<br />

[113] NA Patients <strong>with</strong> <strong>in</strong>operable<br />

oesophagogastric cancer<br />

Patients <strong>with</strong> metastatic<br />

carc<strong>in</strong>oma of the<br />

oesophagus or GEjunction<br />

Siewert type I<br />

Ultraflex stent (n=108) or<br />

s<strong>in</strong>gle-dose brachytherapy (12<br />

Gy, n=101)<br />

RCTs compar<strong>in</strong>g<br />

chemotherapy <strong>with</strong> best<br />

supportive care for patients<br />

<strong>with</strong> metastatic carc<strong>in</strong>oma of<br />

the oesophagus and GEjunction,<br />

as well as RCTs<br />

compar<strong>in</strong>g different<br />

chemotherapeutic therapies.<br />

Treatments <strong>with</strong> s<strong>in</strong>gle<br />

chemotherapeutic agents as<br />

dist<strong>in</strong>ct <strong>in</strong>dividual treatment preferences.<br />

The <strong>in</strong>itial costs of stent placement were<br />

higher than the costs of brachytherapy<br />

(1500 euro vs 570 euro; P0.20). Total hospital stay<br />

dur<strong>in</strong>g follow-up was 11.5 days after<br />

stent placement vs 12.4 days after<br />

brachytherapy, which was responsible<br />

for the high <strong>in</strong>tramural costs <strong>in</strong> both<br />

treatment groups (stent 6512 euro vs<br />

brachytherapy 7982 euro, P>0.20).<br />

Costs for medical procedures dur<strong>in</strong>g<br />

follow-up were higher after stent<br />

placement (stent 249 euro vs<br />

brachytherapy 168 euro, P=0.002), while<br />

the costs of extramural care were<br />

similar (1278 euro vs 1046 euro,<br />

P>0.20).<br />

Only two RCTs <strong>with</strong> a total of 42<br />

participants compared chemotherapy<br />

<strong>with</strong> best supportive care for metastatic<br />

oesophageal cancer. No survival benefit<br />

was shown for chemotherapy treatment<br />

<strong>in</strong> these RCTs. Five RCTs <strong>with</strong> a total of<br />

1242 participants compared different<br />

chemotherapy regimes. Due to variation<br />

<strong>in</strong> patient population and chemotherapy<br />

regimes, it was not possible to perform<br />

See also Homs MY 2004,<br />

Lancet<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

RCT<br />

Cost study<br />

Moderate<br />

7 RCTs <strong>in</strong>cluded SR High


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 117<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

well as comb<strong>in</strong>ation regimes <strong>in</strong> a formal pooled analysis. There was no<br />

all doses and schedules were consistent benefit of any specific<br />

<strong>in</strong>cluded. Studies <strong>in</strong> which<br />

patients received additional<br />

treatment for dysphagia such<br />

as stent placement or<br />

brachytherapy were not<br />

excluded.<br />

chemotherapy regimen.<br />

Ajani J 2006 [269] October Patients <strong>with</strong> oesophageal Oral capecitab<strong>in</strong>e 4 trials identified that <strong>in</strong>cluded patients Medl<strong>in</strong>e search only SR Low<br />

2005 cancer<br />

<strong>with</strong> oesophageal cancer<br />

No <strong>in</strong>formation on<br />

selection, quality<br />

appraisal, etc<br />

Trumper M [274] NA Patients <strong>with</strong> gastro- Plat<strong>in</strong>um-conta<strong>in</strong><strong>in</strong>g regimen There were no significant differences <strong>in</strong> Pooled analysis of 3 RCTs MA Low<br />

2006<br />

oesophageal cancer, older (ECF, MCF), PVI 5-FU the <strong>in</strong>cidence of grades 3/4 toxicity (retrospective<br />

than 70 years (n = 257) (protracted venous <strong>in</strong>fusion of between the two cohorts. Objective and subanalysis): which trials??<br />

5-fluorouracil) ± mitomyc<strong>in</strong> C symptomatic response rates, failure-free<br />

(MMC), or FAMTX<br />

and overall survival were not<br />

significantly different. In a multivariate<br />

analysis, <strong>in</strong>dependent prognostic factors<br />

for survival were performance status<br />

and locally advanced disease, not age.<br />

Patients >70 years <strong>with</strong> OGC obta<strong>in</strong>ed<br />

similar benefits from palliative<br />

chemotherapy <strong>with</strong> respect to<br />

symptomatic response, tumour<br />

regression and survival, <strong>with</strong>out<br />

<strong>in</strong>creased toxicities.<br />

Duffour J 2006 [275] NA 232 patients <strong>with</strong> Cisplat<strong>in</strong> <strong>with</strong> cont<strong>in</strong>uous 5FU All tumours:<br />

Mixed patient population RCT Moderate<br />

histologically proven (n = 19) vs. bolus 5FU and Safety rema<strong>in</strong>ed acceptable and Bl<strong>in</strong>ded assessors?<br />

carc<strong>in</strong>oma of the Leucovor<strong>in</strong> (n = 19)<br />

comparable <strong>in</strong> the two arms except for<br />

oesophagus (n = 38),<br />

the severe grade 3-4 mucositis, which<br />

stomach or pancreas, and<br />

was lower <strong>in</strong> arm B (4.5 vs. 16.4%, p <<br />

metastatic disease<br />

0.009). Efficacy <strong>in</strong> terms of tumour<br />

response and survival was similar <strong>in</strong> the<br />

two arms, show<strong>in</strong>g an objective<br />

response rate (after external review) of<br />

18.6% (95% confidence <strong>in</strong>terval (CI)<br />

11.4-25.8%) <strong>in</strong> arm A vs. 15% (95% CI<br />

8.5-21.6%) <strong>in</strong> arm B, an overall median<br />

survival of 24 weeks <strong>in</strong> arm A vs. 24.7 <strong>in</strong>


118 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

arm B (p = 0.83) and a progression-free<br />

median survival of 12.4 weeks vs. 12.1 <strong>in</strong><br />

Sumpter K 2005 [270] NA Patients <strong>with</strong> <strong>in</strong>operable,<br />

histologically verified<br />

locally advanced or<br />

metastatic<br />

adenocarc<strong>in</strong>oma,<br />

squamous cell or<br />

undifferentiated carc<strong>in</strong>oma<br />

of the oespohagus,<br />

oesophagogastric junction<br />

(OGJ) or stomach<br />

Radiotherapy<br />

Wenger U 2005<br />

Bergquist H<br />

2005<br />

[112] NA Patients <strong>with</strong> <strong>in</strong>curable<br />

cancer of the oesophagus<br />

or gastro-oesophageal<br />

junction<br />

ECF (n = 53)<br />

ECX (n = 48)<br />

EOF (n = 55)<br />

EOX (n = 48)<br />

(E = epirubuc<strong>in</strong>, C = cisplat<strong>in</strong>,<br />

F = 5FU, O = oxaliplat<strong>in</strong>, X =<br />

capecitab<strong>in</strong>)<br />

Self-expandable metallic stent<br />

(n = 30) or 3 x 7 Gy<br />

brachytherapy (n = 30)<br />

arms A and B, respectively (p = 0.91).<br />

First <strong>in</strong>terim analysis was performed<br />

when 80 pts had been randomised.<br />

Doselimit<strong>in</strong>g fluoropyrimid<strong>in</strong>e toxicities<br />

were stomatitis, palmar plantar<br />

erythema (PPE) and diarrhoea; 5.1% of<br />

X-treated pts experienced grade 3/4<br />

toxicity. Protocol planned dose<br />

escalation of X to 625 mg/m² b.i.d. was<br />

<strong>in</strong>stituted and a second <strong>in</strong>terim analysis<br />

has been performed. A total of 204 pts<br />

were randomised at the time of the<br />

protocol planned 2nd <strong>in</strong>terim analysis.<br />

Grade 3/4 fluoropyrimid<strong>in</strong>e-related<br />

toxicity was seen <strong>in</strong> 13.7% pts receiv<strong>in</strong>g<br />

F, 8.4% pts receiv<strong>in</strong>g X 500 mg/m² b.i.d.<br />

and 14.7% pts receiv<strong>in</strong>g X 625 mg/m²<br />

b.i.d. Comb<strong>in</strong>ed complete and partial<br />

response rates were ECF 31% (95% CI<br />

18.7–46.3), EOF 39% (95% CI 25.9–<br />

53.1), ECX 35% (95% CI 21.4–50.3),<br />

EOX 48% (95% CI 33.3–62.8). Grade<br />

3/4 fluoropyrimid<strong>in</strong>e toxicity affected<br />

14.7% of pts treated <strong>with</strong> X 625<br />

mgm 2 b.i.d. 1, which is similar to<br />

that observed <strong>with</strong> F, confirm<strong>in</strong>g this to<br />

be the optimal dose. The replacement of<br />

C by O and F by X does not appear to<br />

impair efficacy. The trial cont<strong>in</strong>ues to<br />

total accrual of 1000 pts.<br />

The group of patients treated <strong>with</strong> stent<br />

reported significantly better HRQL<br />

scores for dysphagia (P < 0.05) at the 1month<br />

follow-up, but most other HRQL<br />

scores, <strong>in</strong>clud<strong>in</strong>g function<strong>in</strong>g and<br />

symptom scales, deteriorated.<br />

Interim analysis<br />

6 orig<strong>in</strong>ally randomized<br />

pts not <strong>in</strong>cluded <strong>in</strong> results<br />

Non<strong>in</strong>feriority study<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g of randomization<br />

Orig<strong>in</strong>ally 65 randomized,<br />

but 5 <strong>with</strong>drew consent<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

RCT Moderate<br />

RCT<br />

Cost study<br />

Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 119<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

There was no difference <strong>in</strong> survival or<br />

complication rates between the two<br />

treatment strategies. There was a<br />

significant difference <strong>in</strong> the change of<br />

dysphagia scores between the time of<br />

<strong>in</strong>clusion and the 1-month follow-up<br />

visit, <strong>in</strong> favour of the stented group (P =<br />

0.03). This difference had disappeared at<br />

3 months. Median total lifetime costs<br />

were 17,690 for the stented group<br />

compared <strong>with</strong> 33 171 for the<br />

brachytherapy group (P = 0.005). This<br />

difference was due to higher costs for<br />

the <strong>in</strong>itial treatment (4615 versus 23<br />

857, P < 0.0001). Sensitivity analyses<br />

showed that the charges for a<br />

brachytherapy session had to be<br />

reduced from 6092 to 4222 (31%) to<br />

make this therapeutic concept cost-<br />

Pol<strong>in</strong>der S 2004<br />

(SIREC trial)<br />

[113] NA Patients <strong>with</strong> <strong>in</strong>operable<br />

oesophagogastric cancer<br />

Ultraflex stent (n=108) or<br />

s<strong>in</strong>gle-dose brachytherapy (12<br />

Gy, n=101)<br />

competitive.<br />

The <strong>in</strong>itial costs of stent placement were<br />

higher than the costs of brachytherapy<br />

(1500 euro vs 570 euro; P0.20). Total hospital stay<br />

dur<strong>in</strong>g follow-up was 11.5 days after<br />

stent placement vs 12.4 days after<br />

brachytherapy, which was responsible<br />

for the high <strong>in</strong>tramural costs <strong>in</strong> both<br />

treatment groups (stent 6512 euro vs<br />

brachytherapy 7982 euro, P>0.20).<br />

Costs for medical procedures dur<strong>in</strong>g<br />

follow-up were higher after stent<br />

placement (stent 249 euro vs<br />

brachytherapy 168 euro, P=0.002), while<br />

the costs of extramural care were<br />

similar (1278 euro vs 1046 euro,<br />

P>0.20).<br />

See also Homs MY 2004,<br />

Lancet<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

RCT<br />

Cost study<br />

Moderate


120 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Follow up.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

CBO [56] July 2003 In het eerste jaar kan bij patiënten na behandel<strong>in</strong>g van een oesofaguscarc<strong>in</strong>oom worden gekozen voor Virgo KS 1999<br />

Very low<br />

frequente follow-up<strong>in</strong>tervals (drie en eventueel zes weken en daarna driemaandelijks), waarna <strong>in</strong> de Goodnight J 1996<br />

daarop volgende jaren deze frequentie teruggaat naar elke zes maanden <strong>in</strong> het tweede jaar en daarna Johnson FE 1997<br />

jaarlijks. Na vijf jaar is er geen <strong>in</strong>dicatie meer voor rout<strong>in</strong>ematige follow-up. Er is slechts plaats voor<br />

aanvullend<br />

onderzoek op geleide van klachten.<br />

Marsh JC 1997<br />

SIGN [47] 2004 Follow up of patients <strong>with</strong> oesophageal cancer should monitor symptoms, signs and nutritional status. Allum WH 2002 Very low<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

No additional studies found<br />

Quality of life<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

SIGN [47] 2004 The possibility of reduction <strong>in</strong> quality of life after surgery should be considered when discuss<strong>in</strong>g treatment<br />

options, particularly when preoperative stag<strong>in</strong>g suggests that surgery would be unlikely to be curative.<br />

Blazeby 2000 Low<br />

Study ID<br />

Early lesions<br />

Ref Search<br />

date<br />

Moraca R 2006 [276] 36 patients <strong>with</strong> highgrade<br />

dysplasia (HGD) or<br />

<strong>in</strong>tramucosal cancer (IMC)<br />

Surgery<br />

Cense HA 2006 [277] 109 patients undergo<strong>in</strong>g a<br />

transthoracic resection<br />

Lagergren<br />

2007<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

[278] 90 patients who<br />

underwent surgery for<br />

oesophageal cancer.<br />

52% survivors >/=3 years.<br />

Esophagectomy 28 survivors, significant differences <strong>in</strong><br />

postesophagectomy gastro<strong>in</strong>test<strong>in</strong>al<br />

symptoms (decrease on heartburn and<br />

slower speed of eat<strong>in</strong>g)<br />

Esophagectomy For survivors, no difference <strong>in</strong> long-term<br />

(at 2 years) quality of life or survival<br />

between those submitted to the<br />

<strong>in</strong>tensive care unit for a short period vs.<br />

a long period.<br />

Esophagectomy Recovery except that scores for physical<br />

function, breathlessness, diarrhea, and<br />

reflux significantly worse than at basel<strong>in</strong>e<br />

(P < .01)<br />

Not specific scale :<br />

Medical Outcomes Study<br />

36-Item Short-Form<br />

Health Survey<br />

Scale : Medical Outcome<br />

Studies Short Form-20<br />

and the Rotterdam<br />

Symptom Check List<br />

It’s necessary to <strong>in</strong>form<br />

patients of the long-term<br />

consequences of surgery<br />

Prospective<br />

cohort study<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Low<br />

Low<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 121<br />

Study ID Ref Search<br />

date<br />

Neoadjuvant treatment<br />

Blazeby 2005 [279] 34 patients undergo<strong>in</strong>g<br />

chemoradiotherapy +<br />

surgery<br />

28 patiens undergo<strong>in</strong>g<br />

chemotherapy + surgery<br />

34 patients undergo<strong>in</strong>g<br />

surgery alone<br />

Palliative treatment<br />

Homs 2004 [280] Patients <strong>with</strong> dysphagia<br />

from <strong>in</strong>operable<br />

oesophageal carc<strong>in</strong>oma<br />

Psychological support<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Quality of life assessment <strong>with</strong><br />

QLQ-C30 and QLQ-OES24<br />

(dysphagia scale from the<br />

EORTC)<br />

Ultraflex stent (n = 108) or<br />

s<strong>in</strong>gle dose (12 Gy)<br />

brachytherapy (n = 101).<br />

After neoadjuvant treatment, but before<br />

surgery, HRQL returned to basel<strong>in</strong>e<br />

levels. Recovery of HRQL was not<br />

hampered by preoperative treatment,<br />

and fewer problems <strong>with</strong> postoperative<br />

nausea, emesis, and dysphagia were<br />

reported by patients who had<br />

undergone neoadjuvant treatment<br />

compared <strong>with</strong> patients who had<br />

undergone surgery alone.<br />

Significant difference <strong>in</strong> favour of<br />

brachytherapy on four out of five<br />

functional scales of the EORTC QLQ-<br />

C30 (role, emotional, cognitive and<br />

social) (P < 0.05).<br />

Major improvements were seen on the<br />

dysphagia and eat<strong>in</strong>g scales of the<br />

EORTC OES-23.<br />

Recovery of HRQL after<br />

esophagectomy was not<br />

impaired by neoadjuvant<br />

treatment<br />

The effects of s<strong>in</strong>gle dose<br />

brachytherapy on HRQoL<br />

compared favourably to<br />

those of stent placement<br />

for the palliation of<br />

oesophageal cancer.<br />

Inlcuded <strong>in</strong> SIGN<br />

guidel<strong>in</strong>e.<br />

Prospective<br />

study<br />

Low<br />

RCT High<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

Kuchler T [115] Patients (N = 271) <strong>with</strong> a Standard care as provided on Kaplan-Meier survival curves<br />

Not specifically for RCT Moderate<br />

prelim<strong>in</strong>ary diagnosis of the surgical wards vs. formal demonstrated better survival for the oesophageal cancer.<br />

cancer of the oesophagus psychotherapeutic support <strong>in</strong> experimental group than the control<br />

(n = 31), stomach, addition to rout<strong>in</strong>e care dur<strong>in</strong>g group. The unadjusted significance level<br />

liver/gallbladder, pancreas, the hospital stay<br />

for group differences was P = .0006 for<br />

or colon/rectum<br />

survival to 10 years. Cox regression<br />

models that took TNM stag<strong>in</strong>g or the<br />

residual tumor classification and tumor<br />

site <strong>in</strong>to account also found significant<br />

differences at 10 years. Secondary<br />

analyses found that differences <strong>in</strong> favor<br />

of the experimental group occurred <strong>in</strong><br />

patients <strong>with</strong> stomach, pancreatic,<br />

primary liver, or colorectal cancer.


122 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Searc<br />

h<br />

Nutritional support<br />

date<br />

CBO [56] July<br />

2003<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

Het verdient aanbevel<strong>in</strong>g gedurende de behandel<strong>in</strong>g met gecomb<strong>in</strong>eerde chemoradiotherapie bij een<br />

patiënt met oesofaguscarc<strong>in</strong>oom de voedsel<strong>in</strong>name te evalueren en zo nodig te starten met<br />

dieetmaatregelen.<br />

Het verdient aanbevel<strong>in</strong>g om reeds <strong>in</strong> een vroeg stadium bij patiënten met een oesofaguscarc<strong>in</strong>oom het<br />

lichaamsgewicht te controleren, de voedsel<strong>in</strong>name te evalueren en zo nodig te starten met<br />

voed<strong>in</strong>gs<strong>in</strong>terventie.<br />

Het verdient aanbevel<strong>in</strong>g bij electieve gastro-<strong>in</strong>test<strong>in</strong>ale chirurgie terughoudend te zijn met het stimuleren<br />

van orale dr<strong>in</strong>kvoed<strong>in</strong>g gezien het gebrek aan aangetoonde effectiviteit.<br />

Op grond van de beschikbare literatuur is er mogelijk plaats voor het gebruik van immunonutritie <strong>in</strong> de<br />

perioperatieve fase bij stabiele patiënten met een oesofaguscarc<strong>in</strong>oom.<br />

Bij ernstig zieke patiënten is terughoudendheid geboden met arg<strong>in</strong><strong>in</strong>erijke immunonutritie.<br />

In de postoperatieve fase kan men overwegen om gedurende metabole stress te streven naar een <strong>in</strong>name<br />

van 1,5-1,7 gram eiwitten per kilogram actueel lichaamsgewicht per dag en 25-30 kcal voor mannen en 20-<br />

25 kcal voor vrouwen per kilogram actueel lichaamsgewicht per dag. Hyperalimentatie dient te worden<br />

voorkomen, omdat dit samen kan gaan met metabole ontregel<strong>in</strong>gen.<br />

Het verdient aanbevel<strong>in</strong>g <strong>in</strong> de postoperatieve fase langdurig het lichaamsgewicht, de voedsel<strong>in</strong>name en de<br />

aanwezigheid van aan voed<strong>in</strong>g gerelateerde klachten te evalueren. Bij gewichtsverlies, onvoldoende <strong>in</strong>name<br />

en bij klachten, kan door middel van dieetadviezen zo nodig met orale dr<strong>in</strong>kvoed<strong>in</strong>g dan wel (nachtelijke)<br />

enterale voed<strong>in</strong>g worden geprobeerd gewichtsverlies te voorkomen en de voed<strong>in</strong>gstoestand<br />

te verbeteren.<br />

Het verdient aanbevel<strong>in</strong>g na een oesofagusresectie gebruik te maken van enterale voed<strong>in</strong>g (sondevoed<strong>in</strong>g)<br />

wanneer orale voed<strong>in</strong>g onvoldoende mogelijk is.<br />

Het verdient aanbevel<strong>in</strong>g om <strong>in</strong> de perioperatieve fase bij patiënten met een oesofaguscarc<strong>in</strong>oom slechts<br />

gebruik te maken van parenterale voed<strong>in</strong>g <strong>in</strong>dien enterale voed<strong>in</strong>g niet mogelijk is.<br />

Dieetmaatregelen bij patiënten met een <strong>in</strong>operabel oesofaguscarc<strong>in</strong>oom kunnen het beste zijn gericht op<br />

het welbev<strong>in</strong>den van de patiënt, dat wil zeggen dat de kwaliteit van leven belangrijker is dan de effecten<br />

van voed<strong>in</strong>g op lange termijn.<br />

SIGN [47] 2004 Patients undergo<strong>in</strong>g surgery for oesophageal cancer who are identified as be<strong>in</strong>g at high nutritional risk<br />

should be considered for preoperative nutritional support.<br />

All patients undergo<strong>in</strong>g surgery for oesophageal cancer should be considered for early postoperative<br />

nutritional support preferably by the enteral route.<br />

Corticosteroids or megestrol acetate should be considered for patients <strong>with</strong> advanced oesophageal<br />

cancer who are anorexic.<br />

Jeremic B 1998<br />

Safran H 2001<br />

Sikora SS 1998<br />

Low<br />

Gianotti L 2002<br />

High<br />

Bozzetti F 2000<br />

Braga M 1999 & 2002<br />

Several RCTs High<br />

Observational studies Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 123<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

Lobo DN 2006 [281] 120 patients undergo<strong>in</strong>g Jejunostomy feed<strong>in</strong>g <strong>with</strong> an Feed delivery, although less than targeted, was 12 excluded from RCT High<br />

resection for cancers of immune modulat<strong>in</strong>g diet similar <strong>in</strong> both groups. There were 6 (11%) analysis (3 protocol<br />

the pancreas, oesophagus (Group A, n = 60) or an deaths <strong>in</strong> each group. Median (IQR)<br />

violoations, 9<br />

and stomach<br />

isonitrogenous, isocaloric feed postoperative hospital stay was 14.5 (12-23) unresectable<br />

(Group B, n = 60) for 10-15 days <strong>in</strong> Group A and 17.5 (13-23) days <strong>in</strong> disease)<br />

days<br />

Group B (P=0.48). A total of 24 (44%) patients<br />

<strong>in</strong> each group had <strong>in</strong>fective complications<br />

(P=1.0). A total of 21 (39%) patients <strong>in</strong> Group<br />

A and 28 (52%) <strong>in</strong> Group B had non-<strong>in</strong>fective<br />

complications (P=0.18). Jejunostomy-related<br />

complications occurred <strong>in</strong> 26 (48%) patients <strong>in</strong><br />

Group A and 30 (56%) <strong>in</strong> Group B (P=0.3).<br />

Ryan AM 2006 [282] 205 consecutive patients Early enteral nutrition via a N<strong>in</strong>ety-two per cent of patients were<br />

Prospective Low<br />

who underwent<br />

needle catheter jejunostomy successfully fed exclusively by NCJ post-<br />

study<br />

oesophagectomy for<br />

oesophagectomy, and 94% of patients were<br />

malignancy<br />

tolerat<strong>in</strong>g a ma<strong>in</strong>tenance regimen of 2000 ml<br />

feed over 20 h by day 2 post-operatively.<br />

Patients spent a median of 15 days on<br />

jejunostomy feed<strong>in</strong>g post-surgery (range 2-112<br />

days); however, 26% required prolonged<br />

jejunostomy feed<strong>in</strong>g (>20 days). M<strong>in</strong>or<br />

gastro<strong>in</strong>test<strong>in</strong>al complications were effectively<br />

managed by slow<strong>in</strong>g the rate of <strong>in</strong>fusion, or<br />

adm<strong>in</strong>ister<strong>in</strong>g medication. Three (1.4%) serious<br />

complications of jejunostomy feed<strong>in</strong>g occurred,<br />

all requir<strong>in</strong>g re-laparotomy, one result<strong>in</strong>g <strong>in</strong><br />

death. NCJ feed<strong>in</strong>g was extremely effective <strong>in</strong><br />

prevent<strong>in</strong>g severe post-operative weight loss <strong>in</strong><br />

the majority of oesophagectomy patients postop.<br />

However, oral <strong>in</strong>take was generally poor at<br />

discharge <strong>with</strong> only 65% of requirements be<strong>in</strong>g<br />

met orally. Sixteen patients (8%) patients<br />

required home jejunostomy feed<strong>in</strong>g. By the<br />

first post-operative month, a further 6% (12)<br />

patients were recommenced on jejunostomy<br />

feed<strong>in</strong>g.<br />

Gabor S 2005 [283] 44 consecutive patients Early enteral feed<strong>in</strong>g<br />

There was a significant difference <strong>in</strong> the<br />

Observationa Low<br />

(38 males and 6 females; compared <strong>with</strong> parenteral <strong>in</strong>terval until the first bowel movements. No<br />

l study<br />

mean age 62, range 30-82) nutrition<br />

difference <strong>in</strong> overall 30 d mortality was<br />

<strong>with</strong> oesophageal<br />

identified. A poor nutritional status was a


124 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search date Population Intervention Results Comments Study type Level of<br />

evidence<br />

carc<strong>in</strong>oma (stages I-III),<br />

significant prognostic factor for an <strong>in</strong>creased<br />

who had undergone<br />

mortality. Early enteral feed<strong>in</strong>g significantly<br />

radical resection and<br />

reduces the duration of ICU treatment and<br />

reconstruction.<br />

total hospital stay <strong>in</strong> patients who undergo<br />

Historical control of 44<br />

oesophagectomy or oesophagogastrectomy for<br />

patients<br />

oesophageal carc<strong>in</strong>oma. The mortality rate is<br />

not affected.<br />

Treatment of recurrent disease<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

CBO [56] July 2003 Bij patiënten met … een recidief na eerdere oesofagusresectie <strong>in</strong> verband met een oesofaguscarc<strong>in</strong>oom heeft<br />

stentplaats<strong>in</strong>g de voorkeur.<br />

Only observational studies were found, but no prospective trials.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 125<br />

APPENDIX 5<br />

EVIDENCE TABLES OF GASTRIC CANCER BY CLINICAL QUESTION<br />

CPG ID Ref Search<br />

date<br />

Alarm symptoms<br />

Diagnosis of gastric cancer.<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

SIGN [47] 2004 Patients present<strong>in</strong>g <strong>with</strong> any of the follow<strong>in</strong>g alarm symptoms should be referred for early endoscopy:<br />

dysphagia, recurrent vomit<strong>in</strong>g, anorexia, weight loss, gastro<strong>in</strong>test<strong>in</strong>al blood loss.<br />

FNCLCC [129] January<br />

2002<br />

Oesophagogastroscopy<br />

Le bilan diagnostique repose sur la recherche des signes cl<strong>in</strong>iques d’appel (douleur abdom<strong>in</strong>ale,<br />

amaigrissement, dyspepsie), … (standard).<br />

Me<strong>in</strong>eche-Schmidt V 2002<br />

Numans ME 2001<br />

Wallace MB 2001<br />

Kapoor N 2005<br />

Potet 1993<br />

Faivre 1997<br />

Low<br />

Very low<br />

SIGN [47] 2004 Flexible upper GI endoscopy is recommended as the diagnostic procedure of choice <strong>in</strong> patients <strong>with</strong> Graham DY 1982 Low<br />

suspected gastric cancer.<br />

Abbas SZ 2004<br />

Rout<strong>in</strong>e use of chromoendoscopy dur<strong>in</strong>g upper GI endoscopy is not recommended, but may be of value<br />

<strong>in</strong> selected patients at high risk of gastric malignancy.<br />

Mitooka H 1995 Very low<br />

FNCLCC [129]<br />

Barium X-ray<br />

January<br />

2002<br />

Le bilan diagnostique repose sur … la gastroscopie … (standard). No evidence provided<br />

SIGN [47] 2004 No recommendation, only discussion<br />

Biopsy<br />

FNCLCC [129] January<br />

2002<br />

Study ID Ref Search<br />

date<br />

Alarm symptoms<br />

Bowrey DJ 2006 [49] NA Patients referred for<br />

open-access gastroscopy<br />

Le bilan diagnostique … doit être confirmé par des biopsies multiples. Ces biopsies (nombre m<strong>in</strong>imum 5<br />

à 8) doivent être réalisées sur toutes les anomalies du relief muqueux permettant un examen d’anatomie<br />

et cytologie pathologique (standard).<br />

No evidence provided<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Application of referral<br />

guidel<strong>in</strong>es us<strong>in</strong>g alarm<br />

symptoms<br />

Gastroscopy identified oesophagogastric<br />

carc<strong>in</strong>oma <strong>in</strong> 123 (3%) of the 4,018<br />

subjects. Of these 123 patients, 104<br />

(85%) <strong>with</strong> oesophagogastric cancer had<br />

‘‘alarm’’ symptoms (anemia, mass,<br />

dysphagia, weight loss, vomit<strong>in</strong>g) and<br />

Prospective<br />

study<br />

Low


126 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Marmo R 2005 [233] NA Patients <strong>with</strong><br />

uncomplicated dyspepsia<br />

Oesophagogastroscopy<br />

Lam EC 2007 [284] Patients <strong>with</strong> thicken<strong>in</strong>g of<br />

the gastric wall on CT (n<br />

= 71)<br />

Barium X-ray<br />

No additional studies found<br />

Biopsy<br />

No additional studies found<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

would have satisfied the referral criteria.<br />

The rema<strong>in</strong><strong>in</strong>g 15% would not have been<br />

referred for <strong>in</strong>itial endoscopic<br />

assessment because their symptoms<br />

were those of uncomplicated ‘‘benign’’<br />

dyspepsia.<br />

The patients <strong>with</strong> ‘‘alarm’’ symptoms<br />

had a significantly more advanced tumor<br />

stage (metastatic disease <strong>in</strong> 47% vs 11%;<br />

p < 0.001), were less likely to undergo<br />

surgical resection (50% vs 95%;<br />

p


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 127<br />

Stag<strong>in</strong>g of patients <strong>with</strong> gastric cancer.<br />

CPG ID<br />

CT scan<br />

Ref Search date Recommendation Support<strong>in</strong>g<br />

evidence<br />

SIGN [47] 2004 In patients <strong>with</strong> gastric cancer, CT scan of the chest and abdomen <strong>with</strong> IV contrast and gastric distention <strong>with</strong><br />

oral contrast or water should be performed rout<strong>in</strong>ely. The liver should be imaged <strong>in</strong> the portal venous phase.<br />

FNCLCC [129] January 2002 Le bilan d’extension doit <strong>in</strong>clure au m<strong>in</strong>imum un scanner abdom<strong>in</strong>opelvien et une radiographie du thorax<br />

(standard).<br />

Endoscopic ultrasonography<br />

Le scanner thoracique peut être proposé pour le bilan d’extension (option).<br />

FNCLCC [129] January 2002 Une écho-endoscopie peut être proposée pour aff<strong>in</strong>er le diagnostic en cas d’hypertrophie des plis gastriques<br />

sans histologie, et pour le diagnostic des tumeurs classées T4 et pour mieux apprécier l’envahissement pariétal<br />

et ganglionnaire si un traitement néoadjuvant est envisagé (option).<br />

Laparoscopy<br />

SIGN [47] 2004 Laparoscopy should be considered <strong>in</strong> patients <strong>with</strong> oesophageal tumours <strong>with</strong> a gastric component, and <strong>in</strong><br />

patients <strong>with</strong> gastric tumours be<strong>in</strong>g considered for surgery where full thickness gastric wall <strong>in</strong>volvement is<br />

suspected.<br />

FNCLCC [129] January 2002 Une laparoscopie peut être proposée pour améliorer l’établissement du stade préthérapeutique dans le cas où<br />

un risque d’extension péritonéale existe, notamment dans les tumeurs suspectées comme T3 et T4 (option).<br />

Magnetic resonance imag<strong>in</strong>g<br />

SIGN [47] 2004 MRI should be reserved for those patients who cannot undergo CT or used for additional <strong>in</strong>vestigation<br />

follow<strong>in</strong>g CT/EUS.<br />

PET scan<br />

Kamel IR 2004 Low<br />

Lee 2001<br />

Delia 2000<br />

Mani 2001<br />

Dux 1999<br />

Davies 1997<br />

Kelly 2001<br />

Manc<strong>in</strong>o 2000<br />

Willis 2000<br />

Tseng 2000<br />

Matsumoto 2000<br />

Fujimara T 2002<br />

Clements DM 2004<br />

Molloy RG 1995<br />

Wilkiemeyer MB 2004<br />

Numerous<br />

observational studies<br />

Sohn KM 2000<br />

Wu LF 2003<br />

Kersjes 1997<br />

Lauenste<strong>in</strong> TC 2004<br />

Wong R 2000<br />

SIGN [47] 2004 PET is not rout<strong>in</strong>ely <strong>in</strong>dicated <strong>in</strong> the stag<strong>in</strong>g of gastric cancers. Mochiki E 2004<br />

Yeung HW 1998<br />

FNCLCC [66] October L’impact de la TEP-FDG sur la prise en charge thérapeutique des patients atte<strong>in</strong>ts d’un cancer de l’estomac McAteer 1999<br />

2002 reste à déterm<strong>in</strong>er par des études prospectives (recommendation).<br />

Hoffmann 1999<br />

Couper 1998<br />

Depotter 2002<br />

K<strong>in</strong>kel 2002<br />

Level of<br />

evidence<br />

Low<br />

Low<br />

Low<br />

Low<br />

Low<br />

Low


128 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID<br />

CT scan<br />

Ref Searc<br />

h date<br />

Kwee RM 2007 [130] August<br />

2006<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Patients <strong>with</strong> gastric<br />

carc<strong>in</strong>oma <strong>with</strong>out<br />

neoadjuvant treatment<br />

Chen CY 2007 [131] Patients <strong>with</strong> gastric<br />

cancer (M0) (n = 69)<br />

Kikuchi S 2006 Patients <strong>with</strong> gastric<br />

adenocarc<strong>in</strong>oma (n = 74)<br />

Endoscopic ultrasonography<br />

Kwee RM 2007 [130] August<br />

2006<br />

Patients <strong>with</strong> gastric<br />

carc<strong>in</strong>oma <strong>with</strong>out<br />

neoadjuvant treatment<br />

Ichikawa T 2007 [285] Patients <strong>with</strong> early gastric<br />

cancer scheduled for<br />

endoscopic submucosal<br />

dissection<br />

G<strong>in</strong>es A 2006 [286] Patients <strong>with</strong> large gastric<br />

folds at endoscopy and<br />

biopsies negative for<br />

malignancy (n = 61)<br />

MDCT<br />

Pathologic exam<strong>in</strong>ation after<br />

surgery as standard<br />

MDCT<br />

Pathologic exam<strong>in</strong>ation after<br />

surgery as standard (n = 55)<br />

MDCT<br />

Standard: histopathological<br />

diagnosis (n = 70); endoscopic<br />

biopsies and cl<strong>in</strong>ical results (n<br />

= 4)<br />

EUS<br />

Pathologic exam<strong>in</strong>ation after<br />

surgery as standard<br />

EUS, three different m<strong>in</strong>iature<br />

probes<br />

EUS<br />

Standard: histology or cl<strong>in</strong>ical<br />

follow-up of at least 2 years<br />

6 studies on MDCT identified<br />

Diagnostic accuracy of overall T stag<strong>in</strong>g:<br />

77% - 89%<br />

Sensitivity for assess<strong>in</strong>g serosal<br />

<strong>in</strong>volvement: 83% - 100%<br />

Specificity for assess<strong>in</strong>g serosal<br />

<strong>in</strong>volvement: 80% - 97%<br />

Overall accuracy of T stag<strong>in</strong>g: 89% (<strong>with</strong><br />

MPR images), or 49/55 correctly staged<br />

Overall accuracy of N stag<strong>in</strong>g: 78% (<strong>with</strong><br />

MPR images), or 43/55 correctly staged<br />

Tumor detected <strong>in</strong> only 47% of patients.<br />

Insufficient data to calculate diagnostic<br />

measures.<br />

23 studies on EUS identified<br />

Diagnostic accuracy of overall T stag<strong>in</strong>g:<br />

65% - 92%<br />

Sensitivity for assess<strong>in</strong>g serosal<br />

<strong>in</strong>volvement: 78% - 100%<br />

Specificity for assess<strong>in</strong>g serosal<br />

<strong>in</strong>volvement: 68% - 100%<br />

30-MHz probes most accurate of<br />

<strong>in</strong>vasion depth (92%)<br />

Enlargement of deep layers was only<br />

<strong>in</strong>dependent predictive factor for<br />

malignancy.<br />

<strong>English</strong> articles only<br />

Also retrospective studies<br />

<strong>in</strong>cluded<br />

Overall moderate quality<br />

of <strong>in</strong>cluded studies<br />

Standard not identical for<br />

all patients<br />

<strong>English</strong> articles only<br />

Also retrospective studies<br />

<strong>in</strong>cluded<br />

Overall moderate quality<br />

of <strong>in</strong>cluded studies<br />

Matthes K 2006 [287] TA-EUS vs. L-EUS Excluded: only 4 patients<br />

<strong>with</strong> gastric cancer<br />

SR Low<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Low<br />

SR Low<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

RCT<br />

Very low<br />

Low<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 129<br />

Study ID Ref Searc Population Intervention Results Comments Study type Level of<br />

h date<br />

evidence<br />

Chu KM 2004 [288] Patients <strong>with</strong><br />

EUS for detection of ascites Sensitivity 61%, specificity 99% Prospective Low<br />

Laparoscopy<br />

histopathologically<br />

confirmed<br />

adenocarc<strong>in</strong>oma of the<br />

stomach<br />

Standard: stag<strong>in</strong>g laparoscopy<br />

study<br />

Mortensen MB 2006 [79] 411 patients <strong>with</strong> upper<br />

gastro<strong>in</strong>test<strong>in</strong>al tract<br />

cancer, of which 134 had<br />

biopsy-proven gastric<br />

cancer<br />

Deogracias ML 2006 [289] Patients operated on for<br />

gastric carc<strong>in</strong>oma (n = 41)<br />

Magnetic resonance imag<strong>in</strong>g<br />

Kwee RM 2007 [130] August<br />

2006<br />

PET scan<br />

No additional studies found<br />

Sent<strong>in</strong>el lymph node mapp<strong>in</strong>g<br />

Patients <strong>with</strong> gastric<br />

carc<strong>in</strong>oma <strong>with</strong>out<br />

neoadjuvant treatment<br />

Comb<strong>in</strong>ed endoscopic<br />

ultrasonography (EUS) and<br />

laparoscopic ultrasonography<br />

(LUS)<br />

‘Standard’: treatment actually<br />

undertaken<br />

Stag<strong>in</strong>g laparoscopy<br />

Open gastrectomy <strong>in</strong> 33 cases<br />

MRI<br />

Pathologic exam<strong>in</strong>ation after<br />

surgery as standard<br />

The comb<strong>in</strong>ation of EUS and LUS<br />

correctly predicted R0 resection <strong>in</strong><br />

90.6%, R1–R2 <strong>in</strong> 91% and irresectability<br />

<strong>in</strong> 91.4% of patients. Ten patients (2.4%)<br />

had explorative laparotomy only. There<br />

were no complications associated <strong>with</strong><br />

the EUS and LUS procedures.<br />

No port-site metastases or port-site<br />

recurrence<br />

3 studies on MRI identified<br />

Diagnostic accuracy of overall T stag<strong>in</strong>g:<br />

71% - 83%<br />

Sensitivity for assess<strong>in</strong>g serosal<br />

<strong>in</strong>volvement: 90% - 93%<br />

Specificity for assess<strong>in</strong>g serosal<br />

<strong>in</strong>volvement: 91% - 100%<br />

To avoid false-positive<br />

f<strong>in</strong>d<strong>in</strong>gs <strong>with</strong> regard to<br />

local irresectability<br />

ultrasonographic criteria<br />

were adjusted so that<br />

only a comb<strong>in</strong>ation of<br />

several ultrasonographic<br />

criteria <strong>with</strong> a 100 per<br />

cent predictive value for<br />

local<br />

irresectability were used<br />

Standard is not identical<br />

for all patients<br />

Selection bias: study<br />

sample is taken from a<br />

prospective database of<br />

surgically treated patients<br />

<strong>English</strong> articles only<br />

Also retrospective studies<br />

<strong>in</strong>cluded<br />

Overall moderate quality<br />

of <strong>in</strong>cluded studies<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

SR Low<br />

Lee JH 2006 [136] Patients <strong>with</strong> cT1N0 Dye and isotope double Overall sensitivity 71%; pT1 subset: 88% Prospective Low<br />

gastric adenocarc<strong>in</strong>oma (T tracers to improve SLNB<br />

study<br />

< 5 cm) (n = 64) Histology as standard<br />

Mochiki E 2006 [138] Patients <strong>with</strong> cN0M0 SLN mapp<strong>in</strong>g <strong>with</strong> Tc-99m Overall sensitivity 83% and specificity Prospective Low<br />

Very low<br />

Very low


130 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Searc Population Intervention Results Comments Study type Level of<br />

h date<br />

evidence<br />

gastric adenocarc<strong>in</strong>oma (n colloidal rhenium sulphide 100%; pT1 subset 100% sensitivity study<br />

= 59)<br />

Histology as standard<br />

Gretschel S 2005 [135] Patients <strong>with</strong> stage I-IV SLN mapp<strong>in</strong>g <strong>with</strong> 99mTc- Difficult to <strong>in</strong>terpret Only 33 patients <strong>in</strong>cluded Prospective Very low<br />

gastric cancer<br />

colloid (n = 15) or dye (n =<br />

19)<br />

Maruyama computer model<br />

Histology as standard<br />

<strong>in</strong> calculation of accuracy study<br />

Miwa K 2003 [137] Patients <strong>with</strong> early gastric Sent<strong>in</strong>el node biopsy after The assay was successful <strong>in</strong> mapp<strong>in</strong>g the<br />

Prospective Low<br />

carc<strong>in</strong>oma treated <strong>with</strong> mapp<strong>in</strong>g <strong>with</strong> vital dye lymphatic bas<strong>in</strong>s <strong>in</strong> 203 (96%) of 211<br />

study<br />

surgery (n = 211)<br />

patients. The dye sta<strong>in</strong>ed one or more<br />

metastatic nodes <strong>in</strong> 31 patients, but<br />

failed to <strong>in</strong>dicate a metastatic node <strong>in</strong><br />

four patients <strong>with</strong> a large <strong>in</strong>volved node.<br />

Meticulous postoperative exam<strong>in</strong>ation of<br />

all resected nodes <strong>in</strong> the standard<br />

paraff<strong>in</strong> slices revealed no new<br />

metastases.<br />

Sensitivity 89%, specificity 100%.<br />

Simsa J 2003 [139] Patients <strong>with</strong> gastric Sent<strong>in</strong>el node biopsy after Success rate of sent<strong>in</strong>el node detection<br />

Prospective Very low<br />

cancer treated <strong>with</strong> mapp<strong>in</strong>g <strong>with</strong> vital dye <strong>with</strong> a good relation between metastatic<br />

study<br />

surgery (D2<br />

<strong>in</strong>volvement of the sent<strong>in</strong>el node and<br />

lymphadenectomy) (n =<br />

other nodes was 56% (13 out of 22<br />

22)<br />

patients).<br />

Two false-negatives.<br />

Insufficient data to calculate diagnostic<br />

measures.


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 131<br />

Diagnosis and treatment of early lesions.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g Level of<br />

date<br />

evidence evidence<br />

SIGN [47] 2004 Rout<strong>in</strong>e use of chromoendoscopy dur<strong>in</strong>g upper GI endoscopy is not recommended, but may be of value <strong>in</strong><br />

selected patients at high risk of gastric malignancy.<br />

Mitooka H 1995 Very low<br />

Pathologists should follow the revised Vienna classification for report<strong>in</strong>g dysplasia.<br />

Where radical <strong>in</strong>tervention is contemplated on the basis of high-grade dysplasia or early adenocarc<strong>in</strong>oma the<br />

diagnosis should be validated by a second pathologist experienced <strong>in</strong> this area and further biopsies should be<br />

taken if there is uncerta<strong>in</strong>ty.<br />

Patients diagnosed <strong>with</strong> high-grade dysplasia should have careful assessment (CT, EUS, rigorous biopsy<br />

protocol +/- EMR) to exclude coexist<strong>in</strong>g cancer.<br />

Schlemper RJ 2000<br />

In the absence of <strong>in</strong>vasive cancer, patients <strong>with</strong> high-grade dysplasia should be offered endoscopic treatment.<br />

The assessment and management of patients <strong>with</strong> high-grade dysplasia should be centralised to units <strong>with</strong> the<br />

appropriate endoscopic facilities and expertise.<br />

Takekoshi T 1994 Very Low<br />

Early gastric cancer limited to the superficial submucosa should be treated by EMR. Wang YP 2006 Low<br />

Mucosal ablative techniques, such as PDT, APC, or laser should be reserved for the management of residual No evidence<br />

disease after EMR and not for <strong>in</strong>itial management if <strong>in</strong>vasive disease is present <strong>in</strong> patients fit for surgery. provide for gastric<br />

cancer<br />

FNCLCC [129] January<br />

2002<br />

La mucosectomie n’est pas une attitude standard (standard).<br />

La mucosectomie endoscopique peut être proposée pour les patients à risques opératoires présentant une<br />

lésion usT1N0 limitée à la muqueuse sans envahissement de la muscularis mucosae (option).<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Lee SH 2006 [290] Patients <strong>with</strong> early gastric EMR <strong>with</strong> submucosal No differences <strong>in</strong> hard outcomes. Randomization <strong>with</strong> RCT Moderate<br />

cancer (n = 72)<br />

<strong>in</strong>jection of normal sal<strong>in</strong>e Shorter mean procedure time <strong>in</strong> FM sealed, opaque envelopes<br />

solution (n = 36) vs. fibr<strong>in</strong>ogen group.<br />

Not clear if bl<strong>in</strong>ded<br />

mixture solution (n = 36)<br />

assessors


132 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Search<br />

date<br />

Chemotherapy<br />

Neoadjuvant treatment.<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

SIGN [47] 2004 The neoadjuvant use of either chemotherapy or radiotherapy for patients <strong>with</strong> gastric cancer is not<br />

recommended outside cl<strong>in</strong>ical trials.<br />

CCO [150] April There is <strong>in</strong>sufficient evidence from randomized trials to recommend neoadjuvant chemotherapy outside<br />

2003 of a cl<strong>in</strong>ical trial.<br />

FNCLCC [129] January<br />

2002<br />

Radiotherapy<br />

La chimiothérapie néoadjuvante n’est pas <strong>in</strong>diquée dans les cancers de l’estomac résécables en dehors<br />

d’essais thérapeutiques.<br />

SIGN [47] 2004 The neoadjuvant use of either chemotherapy or radiotherapy for patients <strong>with</strong> gastric cancer is not<br />

recommended outside cl<strong>in</strong>ical trials.<br />

CCO [150] April There is <strong>in</strong>sufficient evidence from randomized trials to recommend neoadjuvant radiation therapy<br />

2003 outside of a cl<strong>in</strong>ical trial.<br />

FNCLCC [129] January<br />

2002<br />

Immunotherapy<br />

CCO [150] April<br />

2003<br />

Study ID<br />

Chemotherapy<br />

Ref Search<br />

date<br />

Wu AN 2007 [147] June<br />

2005<br />

No recommendations, only discussion<br />

There is <strong>in</strong>sufficient evidence from randomized trials to recommend neoadjuvant immunotherapy outside<br />

of a cl<strong>in</strong>ical trial.<br />

Janunger KG 2002<br />

Earle CC 2003<br />

Songun I 1999<br />

Fujii M 1999 (abstract)<br />

Kang YK 1999 (abstract)<br />

Songun I 1999<br />

Fujii M 1999 (abstract)<br />

Kang YK 1999 (abstract)<br />

Janunger KG 2002<br />

Earle CC 2003<br />

Zang ZX 1998<br />

Skoropad V 1999a<br />

Skoropad V 1999b<br />

Skoropad V 2002<br />

Gouchi A 1997<br />

Peters KM 1990<br />

Terashima M 1998<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Patients <strong>with</strong> resectable<br />

gastric cancer<br />

Neoadjuvant<br />

chemotherapy followed by<br />

surgery vs. surgery alone<br />

Of the four cl<strong>in</strong>ical trials enrolled, there were 250<br />

and 332 cases <strong>in</strong> total, <strong>with</strong> 106 and 126 deaths at<br />

the end of follow-up <strong>in</strong> the NAC and control group,<br />

respectively. The OR (odds ratio) was 1.05 (95%CI:<br />

0.73-1.50), which was not statistically significant. Of<br />

the evaluable 129 patients receiv<strong>in</strong>g NAC, 28.7%<br />

demonstrated either a complete or a partial<br />

response. Two studies of NAC <strong>in</strong> resectable gastric<br />

cancer had resection rate data available for analysis<br />

The R0 resection rate <strong>in</strong> the NAC group was<br />

High<br />

High<br />

High<br />

High<br />

High<br />

High<br />

SR High


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 133<br />

Study ID Ref Search<br />

date<br />

Cunn<strong>in</strong>gham D<br />

2006<br />

Radiotherapy<br />

No additional studies found<br />

Immunotherapy<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

comparable to that <strong>in</strong> the control (OR: 0.96 (95%CI:<br />

0.51-1.83)). The morbidity and mortality of NAC<br />

varied <strong>with</strong> the regimens used preoperatively. Of<br />

the 129 patients <strong>in</strong>cluded <strong>in</strong> the analyzed studies,<br />

[148] Patients <strong>with</strong> histologically<br />

proven adenocarc<strong>in</strong>oma of<br />

the stomach or lower third of<br />

the oesophagus that was<br />

considered to be stage II<br />

(through the submucosa) or<br />

higher, <strong>with</strong> no evidence of<br />

distant metastases, or locally<br />

advanced <strong>in</strong>operable disease,<br />

as evaluated by computed<br />

tomography, chest<br />

radiography, ultrasonography,<br />

or laparoscopy.<br />

Perioperative<br />

chemotherapy and surgery<br />

(n = 250) vs. surgery alone<br />

(n = 253)<br />

some acceptable toxicity was observed.<br />

As compared <strong>with</strong> the surgery group, the<br />

perioperative-chemotherapy group had a higher<br />

likelihood of overall survival (HR for death, 0.75;<br />

95%CI 0.60 to 0.93; P=0.009; five-year survival rate,<br />

36 percent vs. 23 percent) and of progression-free<br />

survival (HR for progression, 0.66; 95%CI 0.53 to<br />

0.81; P


134 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Restag<strong>in</strong>g after neoadjuvant treatment.<br />

CPG ID Ref Searc Recommendation Support<strong>in</strong>g evidence Level of<br />

h date<br />

evidence<br />

SIGN [47] 2004 Patients <strong>with</strong> locally advanced disease hav<strong>in</strong>g chemotherapy/chemoradiotherapy should have their Ross P 1996<br />

Low<br />

response assessed for an impact on the potential to operate ; follow<strong>in</strong>g a good response the patient<br />

should be restaged and the role restaged and the role of surgery re-evaluated by the multidiscipl<strong>in</strong>ary<br />

team.<br />

Wilke H 1995<br />

Surgical treatment.<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

SIGN [47] 2004 Gastric cancer resectional surgery should be carried out <strong>in</strong> high volume specialist surgical units by<br />

frequent operators.<br />

Bachmann MO 2002 Low<br />

Surgery for gastric cancer should be aimed at achiev<strong>in</strong>g an R0 resection. Marub<strong>in</strong>i E 2002<br />

Bozzetti F 1997<br />

High<br />

Consideration should be given to pouch formation after total gastrectomy.<br />

D2 lymphadenectomy, <strong>with</strong> a m<strong>in</strong>imum of 25 lymph nodes removed, should be considered for patients<br />

undergo<strong>in</strong>g gastrectomy. Rout<strong>in</strong>e resection of additional organs (spleen and pancreas) dur<strong>in</strong>g<br />

gastrectomy is not recommended.<br />

Lehnert T 2004 High?<br />

FNCLCC [129] January Une chirurgie d’exérèse à visée curative doit être réalisée (standard). Maruyama 1987<br />

2002<br />

Jakl 1995<br />

Allum 1989<br />

Pour les tumeurs distales (1/3 <strong>in</strong>férieur, antre), il est <strong>in</strong>diqué de pratiquer une gastrectomie des 4/5 et Gouzi 1989<br />

High<br />

curage ganglionnaire monobloc avec élargissement monobloc de nécessité aux organes de vois<strong>in</strong>age<br />

atte<strong>in</strong>ts (standard).<br />

Bozzetti 1999<br />

Pour les tumeurs proximales (2/3 supérieurs), il est <strong>in</strong>diqué de pratiquer une gastrectomie totale et<br />

curage ganglionnaire monobloc avec élargissement monobloc de nécessité aux organes de vois<strong>in</strong>age<br />

atte<strong>in</strong>ts (standard).<br />

No evidence provided<br />

Pour les cancers superficiels et pour des petites tumeurs limitées, les techniques de chirurgie<br />

laparoscopique sont applicables dans le cadre d’études prospectives (option).<br />

Observational studies Low<br />

Un curage ganglionnaire de type D1 (ganglions périgastriques, groupes 1 à 6 soit ≥ 15 ganglions) doit Dent 1988<br />

High<br />

être réalisé selon la topographie de la tumeur (standard).<br />

Robertson 1994<br />

Cischieri 1996<br />

Bonenkamp 1999<br />

Wu 2004<br />

Un curage de type D2 (D1 + ganglions pédiculaires (groupes 1 à 11 soit ≥ 25 ganglions)) sans<br />

Dent 1988<br />

splénectomie ni pancréatectomie peut être envisagé (option).<br />

Robertson 1994<br />

Cischieri 1996<br />

Bonenkamp 1999<br />

Wu 2004


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 135<br />

CPG ID Ref Search<br />

date<br />

Study ID Ref Search<br />

date<br />

Volume-outcome relation<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

Csendes 2002<br />

Des essais randomisés évaluant le curage D2 sans splénectomie sont recommandés. Une splénectomie<br />

est <strong>in</strong>diquée en cas d'adénopathies de l'artère splénique ou de cancer de la grosse tubérosité atteignant<br />

la séreuse (recommandation).<br />

Csendes 2002<br />

Subanalyses of RCTs<br />

Il n’y a pas de modalités standards de reconstruction après gastrectomie distale subtotale (standard).<br />

La reconstruction peut être effectuée par rétablissement de cont<strong>in</strong>uité par anastomose gastrojéjunale ou<br />

par établissement de cont<strong>in</strong>uité par anastomose gastroduodénale (option).<br />

Chareton 1996 High<br />

Il est recommandé de privilégier le rétablissement de cont<strong>in</strong>uité par la technique habituelle du chirurgien No evidence provided<br />

(recommandation).<br />

Enz<strong>in</strong>ger P 2007 [293] Patients <strong>with</strong> stage IB<br />

through IV (M0) gastric<br />

and gastroesophageal<br />

junction adenocarc<strong>in</strong>oma,<br />

previously randomized to<br />

adjuvant chemoradiation<br />

after surgery or surgery<br />

alone (n = 448)<br />

Type of surgery<br />

Hosono S 2006 [164] August<br />

2006<br />

Après gastrectomie totale, le rétablissement de la cont<strong>in</strong>uité digestive peut se faire par anse jéjunale<br />

selon la technique en Y de Roux, avec ou sans reconstitution d’un réservoir (J Pouch), ou par<br />

constitution d’un réservoir avec <strong>in</strong>terposition entre oesophage et duodénum d’une anse jéjunale en S ou<br />

du côlon transverse gauche (option).<br />

5 RCTs<br />

4 prospective trials<br />

Moderate<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Patients <strong>with</strong> gastric<br />

cancer<br />

Effect of hospital surgical<br />

volume, as assessed by<br />

Medicare claims data, on<br />

overall survival and gastric<br />

cancer recurrence<br />

Laparoscopy assisted<br />

distal gastrectomy<br />

(LADG) vs. conventional<br />

open distal gastrectomy<br />

(CODG)<br />

Hospital surgical volume was not predictive of<br />

overall survival (P = 0.46) or disease-free survival<br />

(P = 0.43) at a median follow-up of 8.9 years.<br />

However, patients who underwent either a D1<br />

or D2 dissection at a high- or moderate-volume<br />

center experienced an adjusted hazard ratio of<br />

0.80 (95% CI, 0.53-1.20) for overall survival and<br />

0.78 (95% CI, 0.53-1.14) for disease-free survival<br />

compared <strong>with</strong> those patients resected at a lowvolume<br />

hospital; these results were not<br />

statistically significant. When a D0 resection was<br />

performed, hospital procedure volume showed<br />

no impact on survival.<br />

Compared to CODG, LADG was a longer<br />

procedure (weighted mean difference 54.3;<br />

95%CI 38.8 to 69.8; P < 0.001), but<br />

was associated <strong>with</strong> a lower associated morbidity<br />

(OR 0.54; 95%CI 0.37 to 0.77; P < 0.001); this<br />

was most significant for postoperative ileus (OR<br />

Retrospective selection of<br />

patients from a RCT (n =<br />

556)<br />

Inclusion of 4 RCTs (only 1<br />

Western study) and 12<br />

retrospective studies<br />

<strong>English</strong> only<br />

No <strong>in</strong>formation on how<br />

quality appraisal was done<br />

Low<br />

Observation<br />

al study<br />

Very low<br />

SR Moderate


136 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Hayashi H 2005 [163] Patients <strong>with</strong> early gastric<br />

cancers <strong>in</strong> the lower half<br />

of the stomach (n = 28)<br />

Huscher C 2005 [165] Patients <strong>with</strong> distal gastric<br />

cancer<br />

Inaba T 2004 [294] Patients <strong>with</strong> gastric<br />

cancer undergo<strong>in</strong>g distal<br />

gastrectomy or total<br />

gastrectomy<br />

Lee JH 2005 [166] Patients <strong>with</strong> early gastric<br />

cancer<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

0.27; 95%CI 0.09 to 0.84; P = 0.02). There was<br />

no significant difference between the two groups<br />

<strong>in</strong> anastomotic, pulmonary, and wound<br />

Open distal gastrectomy<br />

(n = 14) vs. laparoscopyassisted<br />

distal gastectomy<br />

(n = 14)<br />

Laparoscopic-assisted (n<br />

= 30) vs. open radical<br />

subtotal gastrectomy (n =<br />

29)<br />

Upper midl<strong>in</strong>e <strong>in</strong>cision vs.<br />

transverse <strong>in</strong>cision<br />

Laparoscopy-assisted<br />

distal gastrectomy<br />

(LADG) <strong>in</strong>clud<strong>in</strong>g<br />

lymphadenectomy vs.<br />

complications and mortality.<br />

The LADG group required a significantly shorter<br />

period of postoperative epidural anesthesia and<br />

had no major postsurgery complications.<br />

Pathologic exam<strong>in</strong>ations showed that surgery was<br />

equally radical <strong>in</strong> the two groups.<br />

Operative mortality rates were 6.7% (2 patients)<br />

<strong>in</strong> the OG and 3.3% (1 patient) <strong>in</strong> the LG (P =<br />

not significant); morbidity rates were 27.6% and<br />

26.7%, respectively (P = not significant). Five-year<br />

overall and disease-free survival rates were 55.7%<br />

and 54.8% and 58.9% and 57.3% <strong>in</strong> the OG and<br />

the LG, respectively (P = not significant).<br />

Times for both open<strong>in</strong>g and clos<strong>in</strong>g the<br />

abdom<strong>in</strong>al cavity were longer <strong>with</strong> a transverse<br />

<strong>in</strong>cision, <strong>in</strong> both the distal gastrectomy group and<br />

total gastrectomy group. In the patients <strong>in</strong> whom<br />

cont<strong>in</strong>uous epidural analgesia was used<br />

postoperatively, the number of additional doses<br />

of analgesics was smaller <strong>in</strong> the transverse<strong>in</strong>cision<br />

group after distal gastrectomy. The<br />

<strong>in</strong>cidence of postoperative pneumonia was lower<br />

<strong>in</strong> the transverse-<strong>in</strong>cision group after distal<br />

gastrectomy. The number of patients <strong>with</strong><br />

postoperative <strong>in</strong>test<strong>in</strong>al obstruction was smaller<br />

<strong>in</strong> the transverse-<strong>in</strong>cision group than <strong>in</strong> the<br />

midl<strong>in</strong>e-<strong>in</strong>cision group after distal gastrectomy. In<br />

contrast to distal gastrectomy, there was no<br />

significant difference <strong>in</strong> the number of doses of<br />

postoperative analgesics, <strong>in</strong>cidence of<br />

postoperative pneumonia, or <strong>in</strong>cidence of<br />

postoperative <strong>in</strong>test<strong>in</strong>al obstruction between the<br />

two study groups after total gastrectomy.<br />

The mean postoperative hospital stay and the<br />

duration of analgesic adm<strong>in</strong>istration were shorter<br />

for group L but not significantly. Postoperative<br />

pulmonary complications occurred more<br />

Randomization <strong>with</strong> sealed<br />

envelope<br />

Bl<strong>in</strong>d<strong>in</strong>g???<br />

Not clear if randomization<br />

was bl<strong>in</strong>ded<br />

Bl<strong>in</strong>ded assessment??<br />

No <strong>in</strong>formation about<br />

randomization procedure<br />

Bl<strong>in</strong>d<strong>in</strong>g???<br />

RCT Moderate<br />

RCT Moderate<br />

RCT Moderate<br />

Bl<strong>in</strong>d<strong>in</strong>g?? RCT Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 137<br />

Study ID Ref Search<br />

date<br />

Lee WJ 2003 [295] Patients <strong>with</strong> gastric<br />

cancer treated <strong>with</strong><br />

extended lymph node<br />

dissection<br />

(D2) dur<strong>in</strong>g gastrectomy<br />

Sasako M 2006 [296] Patients <strong>with</strong> gastric<br />

cancer of the cardia or<br />

subcardia<br />

Yu W 2006 [297] Patients <strong>with</strong> gastric<br />

adenocarc<strong>in</strong>oma who<br />

underwent total<br />

gastrectomy (n = 216)<br />

Lymphadenectomy<br />

McCulloch 2004 [152] April<br />

2001<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

open distal gastrectomy frequently <strong>in</strong> the O group (p = 0.043). At a<br />

median follow-up of 14 months, there has been<br />

Patients <strong>with</strong> gastric<br />

cancer<br />

D2 gastric resection<br />

performed <strong>with</strong> the<br />

LigasureTM system (n =<br />

40) vs. resection us<strong>in</strong>g<br />

conventional haemostatic<br />

methods (n = 40)<br />

Left thoracoabdom<strong>in</strong>al<br />

approach (LTA) (n = 85)<br />

vs. abdom<strong>in</strong>al-transhiatal<br />

approach (TH) (n = 82)<br />

Total gastrectomy either<br />

<strong>with</strong> (n = 104) or <strong>with</strong>out<br />

(n = 103) splenectomy<br />

Limited vs. extended<br />

lymph node removal<br />

dur<strong>in</strong>g gastrectomy<br />

no recurrence of disease <strong>in</strong> either group.<br />

Ligasure was associated <strong>with</strong> less <strong>in</strong>traoperative<br />

blood loss (mean (s.d.) 142(73) versus 239(124)<br />

ml; P = 0.001) and a shorter operat<strong>in</strong>g time<br />

(mean(s.d.) 169(25) versus 222(28) m<strong>in</strong>; P =<br />

0.001) than conventional operation.<br />

Postoperative dra<strong>in</strong>age fluid volumes were<br />

greater <strong>in</strong> the Ligasure group (mean (s.d.)<br />

1577(940) versus 886(542) ml; P = 0.020). There<br />

were no differences <strong>in</strong> postoperative<br />

complications or hospital stay.<br />

5-year overall survival was 52.3% (95% CI 40.4-<br />

64.1) <strong>in</strong> the TH group and 37.9% (26.1-49.6) <strong>in</strong><br />

the LTA group. The hazard ratio of death for<br />

LTA compared <strong>with</strong> TH was 1.36 (0.89-2.08,<br />

p=0.92). Three patients died <strong>in</strong> hospital after LTA<br />

but none after TH. Morbidity was worse after<br />

LTA than after TH.<br />

Gastrectomy comb<strong>in</strong>ed <strong>with</strong> splenectomy tended<br />

to be associated <strong>with</strong> slightly higher morbidity<br />

and mortality rates, a slightly greater <strong>in</strong>cidence of<br />

lymph node metastasis at the splenic hilum and<br />

along the splenic artery, and marg<strong>in</strong>ally better<br />

survival, but there were no statistically significant<br />

differences between the groups. Splenectomy had<br />

no impact on survival <strong>in</strong> patients <strong>with</strong> metastatic<br />

lymph nodes at the hilum of the spleen or <strong>in</strong><br />

those <strong>with</strong> metastatic lymph nodes along the<br />

splenic artery.<br />

Meta-analysis of randomised trials did not reveal<br />

any survival benefit for extended lymph node<br />

dissection (Risk ratio = 0.95 (95%CI 0.83 - 1.09),<br />

but showed <strong>in</strong>creased postoperative mortality<br />

(RR 2.23, 95% CI 1.45 - 3.45). Pre-specified<br />

subgroup analysis suggested a possible benefit <strong>in</strong><br />

stage T3+ tumours (RR = 0.68, 95% CI 0.42-<br />

No <strong>in</strong>formation on<br />

randomization procedure<br />

RCT Moderate<br />

No bl<strong>in</strong>d<strong>in</strong>g RCT Moderate<br />

9 patients excluded from<br />

analysis<br />

No <strong>in</strong>formation on bl<strong>in</strong>d<strong>in</strong>g<br />

Two randomised and two<br />

non-randomised<br />

comparisons of limited<br />

(D1) versus extended (D2)<br />

node dissection and 11<br />

cohort studies of either D1<br />

or D2 resection were<br />

RCT Moderate<br />

SR High


138 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

1.10). Non-randomised comparisons showed no<br />

significant survival benefit for extended dissection<br />

(RR 0.92, 95% CI 0.83 -1.02), but decreased<br />

mortality (RR 0.65, 95% CI 0.45-0.93). Subgroup<br />

analysis showed apparent benefit <strong>in</strong> UICC stage II<br />

and IIIa. Observational studies of D2 resection<br />

reported much better mortality and survival than<br />

those of D1 surgery, but the sett<strong>in</strong>gs were<br />

strik<strong>in</strong>gly different.<br />

analysed.<br />

Degiuli M 2004 [154] Patients <strong>with</strong> potentially D1 (n = 76) and D2 Complications developed <strong>in</strong> 10.5% of patients No <strong>in</strong>formation on bl<strong>in</strong>d<strong>in</strong>g RCT Moderate<br />

curable gastric cancer gastrectomy (n = 86) after D1 and <strong>in</strong> 16.3% of patients after D2<br />

gastrectomy (p


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 139<br />

Study ID Ref Search<br />

date<br />

Wu C 2004 &<br />

2006<br />

Yonemura Y<br />

2006<br />

Reconstruction<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Hospital mortality was reported at 0.80%: one<br />

patient <strong>in</strong> each group died of operative<br />

complications, while one from each group died of<br />

[298] Patients <strong>with</strong> gastric<br />

adenocarc<strong>in</strong>oma treated<br />

<strong>with</strong> curative <strong>in</strong>tent<br />

[159] Patients <strong>with</strong> advanced<br />

gastric adenocarc<strong>in</strong>oma<br />

Adachi S 2003 [299] Patients <strong>with</strong> gastric<br />

cancer (n = 30)<br />

D1 (ie, level 1)<br />

lymphadenectomy (n =<br />

110) vs. D3 (ie, levels 1,<br />

2, and 3) dissection (n =<br />

111)<br />

D2 (level 1 and 2<br />

lymphadenectomy) (n =<br />

128) vs. D4 (D2 plus<br />

lymphadenectomy of<br />

para-aortic lymph nodes)<br />

dissection (n = 128)<br />

Roux-en-Y<br />

esophagojejunostomy (R-<br />

Y), R-Y <strong>with</strong> pouch (P-Y),<br />

and jejunal <strong>in</strong>terposition<br />

<strong>with</strong> pouch (P-I) after<br />

total gastrectomy<br />

rapid progressive cancer while <strong>in</strong>patient.<br />

Overall 5-year survival was significantly higher <strong>in</strong><br />

patients assigned D3 surgery than <strong>in</strong> those<br />

assigned D1 surgery (59.5% [95% CI 50.3-68.7] vs<br />

53.6% [44.2-63.0]; difference beteween groups<br />

5.9% [-7.3 to 19.1], log-rank p=0.041). 215<br />

patients who had R0 resection (ie, no<br />

microscopic evidence of residual disease) had<br />

recurrence at 5 years of 50.6% [41.1-60.2] for D1<br />

surgery and 40.3% [30.9-49.7] for D3 surgery<br />

(difference between groups 10.3% [-3.2 to 23.7],<br />

log-rank p=0.197).<br />

Five (4%) and two (2%) medical complications<br />

developed <strong>in</strong> the D2 and D4 groups, respectively.<br />

Surgical complications developed <strong>in</strong> 28 (22%) and<br />

48 patients (38%) after D2 and D4 gastrectomy.<br />

The most common complications were<br />

anastomotic leakage, pancreatic fistula, and<br />

abdom<strong>in</strong>al abscess. Pancreatic fistula developed <strong>in</strong><br />

6 (19%) of 32 patients after D4 plus<br />

pancreatosplenectomy, but the <strong>in</strong>cidence of<br />

pancreatic fistula after D2 gastrectomy plus<br />

pancreatosplenectomy was low (6%, 1/16). Two<br />

patients died <strong>with</strong><strong>in</strong> 30 days of operation (0.8%,<br />

2/256), and each patient belonged to the D2 and<br />

D4 group.<br />

No <strong>in</strong>formation on hard endpo<strong>in</strong>ts.<br />

Reconstruction should be performed <strong>with</strong> pouch<br />

formation after total gastrectomy <strong>with</strong> curative<br />

<strong>in</strong>tent.<br />

No bl<strong>in</strong>d<strong>in</strong>g RCT Moderate<br />

No <strong>in</strong>formation on<br />

randomization procedure.<br />

No bl<strong>in</strong>ded assessment<br />

Randomization <strong>with</strong> sealed<br />

envelope<br />

In case or recurrence or<br />

impossible follow-up <strong>with</strong><strong>in</strong><br />

2 years after surgery:<br />

replacement by other<br />

patient<br />

Bl<strong>in</strong>d<strong>in</strong>g???<br />

RCT Moderate<br />

RCT Moderate<br />

Ishikawa M 2005 [300] Patients <strong>with</strong> gastric Roux-en-Y 5 of 24 patients <strong>with</strong> RY reconstruction Randomization <strong>with</strong> sealed RCT Moderate


140 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

carc<strong>in</strong>oma who<br />

reconstruction vs. developed gastrojejunal stasis <strong>in</strong> the early envelopes<br />

underwent distal conventional Billroth I postoperative period, which led to a longer No <strong>in</strong>formation on bl<strong>in</strong>d<strong>in</strong>g<br />

gastrectomy (n = 50) reconstruction<br />

postoperative hospital stay as compared <strong>with</strong> the<br />

B-I group (mean +/- S.D; B-I; 19.0 +/- 6.2, RY;<br />

31.8 +/- 21.7 days) (P < 0.05). Endoscopic<br />

exam<strong>in</strong>ation revealed that the frequency of bile<br />

reflux (P < 0.01) and degree of <strong>in</strong>flammation <strong>in</strong><br />

the remnant stomach (P < 0.05) were less <strong>in</strong> the<br />

RY group than <strong>in</strong> the B-I group. However,<br />

<strong>in</strong>flammatory f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> the lower esophagus<br />

were observed <strong>in</strong> 7 (27%) of B-I, and 8 (35%) of<br />

the RY group, suggest<strong>in</strong>g that late phase<br />

esophagitis was not improved <strong>in</strong> the RY group.<br />

Iwata T 2006 [160] Patients <strong>with</strong> gastric Reconstruction by jejunal The JPI group showed a significant dietary Very small sample size RCT Moderate<br />

cancer <strong>in</strong> the upper third pouch <strong>in</strong>terposition (JPI advantage. Three months after surgery, JPI No <strong>in</strong>formation on<br />

of the stomach who group) (n = 4) vs. patients could eat more than 80% of the volume randomization procedure<br />

underwent proximal reconstruction by jejunal of their preoperative meals, whereas JI patients or bl<strong>in</strong>d<strong>in</strong>g<br />

gastrectomy (n = 9) <strong>in</strong>terposition (JI group) (n ate less than 50%. The percentage of<br />

= 5)<br />

postoperative body weight loss was higher <strong>in</strong> the<br />

JI group than <strong>in</strong> the JPI group because the volume<br />

of the remnant stomach was more adequate <strong>in</strong><br />

the latter. Moreover, it was easier to enter the<br />

remnant stomach and duodenum for endoscopic<br />

fiberscopy <strong>in</strong> the JPI group for the treatment of<br />

hepato-biliary pancreatic disease.<br />

Kono K 2003 [162] Patients receiv<strong>in</strong>g total Roux-en-Y<br />

Jejunal pouch reconstruction provided the better No <strong>in</strong>formation on RCT Moderate<br />

gastrectomy for early reconstruction group QOL than Roux-en-Y reconstruction <strong>with</strong>out randomization procedure<br />

gastric cancer (n = 50) <strong>with</strong>out pouch (RY pouch both at short-term and long-term periods. or bl<strong>in</strong>d<strong>in</strong>g<br />

group) or the jejunal Pouch reconstruction provided less bile reflux<br />

pouch reconstruction <strong>in</strong>to the esophagus compared <strong>with</strong> Roux-en-Y<br />

group (pouch group) reconstruction.<br />

Mochiki E 2004 [301] Patients <strong>with</strong> gastric Jejunal <strong>in</strong>terposition The <strong>in</strong>terposed jejunum <strong>with</strong> a pouch shows No <strong>in</strong>formation on RCT Moderate<br />

cancer treated by total <strong>with</strong>out a pouch (n = 12) marked disturbances from the motor pattern of a randomization procedure<br />

gastrectomy (n = 12) vs. jejunal <strong>in</strong>terposition normal jejunum dur<strong>in</strong>g the fast<strong>in</strong>g and fed states. or bl<strong>in</strong>d<strong>in</strong>g<br />

<strong>with</strong> a pouch (n = 14) These motor abnormalities may be responsible<br />

for <strong>in</strong>sufficient food <strong>in</strong>take of the J-P group.<br />

Shibata C 2004 [302] Patients <strong>with</strong> early gastric Pylorus-preserv<strong>in</strong>g There were no differences <strong>in</strong> early results Randomization <strong>with</strong> sealed RCT Moderate<br />

cancer (n = 81)<br />

gastrectomy (PPG) (n = between PPG and CDG. The <strong>in</strong>cidence of early envelope<br />

36) vs. conventional distal dump<strong>in</strong>g syndrome was lower <strong>in</strong> PPG (8%) than No <strong>in</strong>formation on bl<strong>in</strong>d<strong>in</strong>g<br />

gastrectomy (CDG) <strong>with</strong> <strong>in</strong> CDG (33%). Other late results <strong>in</strong>clud<strong>in</strong>g the 7 patients excluded from


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 141<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Billroth I anastomosis (n <strong>in</strong>cidence of gallstones were not different analysis<br />

= 38)<br />

between the 2 groups.<br />

Yoo C 2005 [161] Patients <strong>with</strong> upper third Proximal gastrectomy No significant differences <strong>in</strong> operat<strong>in</strong>g time,<br />

RCT High<br />

gastric cancer<br />

<strong>with</strong> jejunal pouch hospital stay, and postoperative complications.<br />

<strong>in</strong>terposition (PGJP) (n = Blood loss was significantly less <strong>in</strong> the PGJP group<br />

25) vs. Roux-en-Y (P = 0.036). N<strong>in</strong>eteen patients (73%) <strong>in</strong> the<br />

esophagojejunostomy TGRY group had one or more postgastrectomy<br />

(TGRY) (n = 26) symptoms, which was significantly more frequent<br />

than <strong>in</strong> the PGJP group (32%; P = 0.012). There<br />

were also significant differences between the two<br />

groups <strong>with</strong> regard to food <strong>in</strong>take, weight<br />

recovery, hemoglob<strong>in</strong>, and serum vitam<strong>in</strong> B12<br />

levels <strong>in</strong> favour of PGJP.<br />

CPG ID Ref Search<br />

date<br />

Chemotherapy<br />

Adjuvant treatment.<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

SIGN [47] 2004 Postoperative chemotherapy for patients <strong>with</strong> gastric cancer is not recommended outside a cl<strong>in</strong>ical trial. Janunger 2002<br />

Earle 2003<br />

High<br />

CCO [150] April For patients unable to undergo radiation, adjuvant chemotherapy alone may be of benefit, particularly for Numerous MA and RCTs High<br />

2003 patients <strong>with</strong> lymph node metastases. The optimal regimen rema<strong>in</strong>s to be def<strong>in</strong>ed.<br />

FNCLCC [129] January La chimiothérapie adjuvante n’est pas <strong>in</strong>diquée dans le traitement des cancers de l’estomac en dehors Numerous MA and RCTs High<br />

Radiotherapy<br />

2002 d’essais thérapeutiques (standard).<br />

Il est recommandé de poursuivre les essais thérapeutiques, en améliorant les protocoles de<br />

chimiothérapie, comparant la chimiothérapie adjuvante à la chirurgie seule ou à la chirurgie +<br />

radiochimiothérapie postopératoire (recommandation).<br />

CCO [150] April<br />

2003<br />

FNCLCC [129] January<br />

2002<br />

Chemoradiotherapy<br />

There is <strong>in</strong>sufficient evidence from randomized trials to recommend adjuvant radiation therapy outside<br />

of a cl<strong>in</strong>ical trial.<br />

La radiothérapie externe adjuvante n'est pas une attitude thérapeutique standard dans les cancers de<br />

l'estomac (standard).<br />

SIGN [47] 2004 Postoperative chemoradiation for patients <strong>with</strong> gastric cancer is not recommended outside a cl<strong>in</strong>ical<br />

trial.<br />

CCO [150] April Follow<strong>in</strong>g surgical resection, patients whose tumours penetrated the muscularis propria or <strong>in</strong>volved<br />

2003 regional lymph nodes should be considered for adjuvant comb<strong>in</strong>ed chemoradiotherapy. The current<br />

standard protocol consists of one cycle of 5-FU (425 mg/m2/day) and leucovor<strong>in</strong> (20 mg/m2/day) <strong>in</strong> a<br />

Hallissey 1994<br />

Kraml<strong>in</strong>g 1996<br />

High<br />

Hallissey 1994 High<br />

McDonald JS 2001 Moderate<br />

Dent 1979<br />

Moertel 1984<br />

McDonald 2001<br />

High


142 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Search<br />

date<br />

FNCLCC [129] January<br />

2002<br />

Recommendation Support<strong>in</strong>g evidence Level of<br />

evidence<br />

daily regimen for five days, followed one month later by 4,500 cGy (180 cGy/day) of radiation given <strong>with</strong><br />

5-FU (400 mg/m2/day) and leucovor<strong>in</strong> (20 mg/m2/day) on days 1 through 4 and the last three days of<br />

radiation. One month after completion of radiation, two cycles of 5-FU (425 mg/m2/day) and leucovor<strong>in</strong><br />

(20 mg/m2/day) <strong>in</strong> a daily regimen for five days are given at monthly <strong>in</strong>tervals.<br />

Chez tous les patients opérés d’un cancer de l’estomac R0 avec un curage D1 ou D2 et de stade II ou III,<br />

la radiochimiothérapie adjuvante n’est pas considérée comme un traitement standard. En cas de curage <<br />

D1 (mo<strong>in</strong>s de 15 ganglions exam<strong>in</strong>és) et chez des patients T3 et/ou N+, la radiochimiothérapie peut être<br />

proposée chez les patients non dénutris selon les modalités de l’essai de Macdonald (standard).<br />

En cas de chirurgie avec curage D1 ou D2 et avec un envahissement ganglionnaire majeur (N2 ou N3),<br />

une radiochimiothérapie adjuvante peut être proposée dans l’attente d’autres résultats, chez des patients<br />

non dénutris (option).<br />

Il est recommandé de réaliser des essais thérapeutiques évaluant la place de la radiochimiothérapie<br />

adjuvante dans le traitement des cancers de l’estomac (accord d’experts). Le choix entre ces différentes<br />

options thérapeutiques doit prendre en compte l'avis ou le souhait des patients (recommandation).<br />

Intraperitoneal<br />

chemotherapy<br />

SIGN [47] 2004 Intraperitoneal chemotherapy and immunotherapy for patients <strong>with</strong> gastric cancer are not<br />

recommended outside a cl<strong>in</strong>ical trial.<br />

Immunotherapy<br />

SIGN [47] 2004 Intraperitoneal chemotherapy and immunotherapy for patients <strong>with</strong> gastric cancer are not<br />

recommended outside a cl<strong>in</strong>ical trial.<br />

CCO [150] April There is <strong>in</strong>sufficient evidence from randomized trials to recommend adjuvant immunotherapy outside of<br />

2003 a cl<strong>in</strong>ical trial.<br />

FNCLCC [129] January<br />

2002<br />

L’immunochimiothérapie adjuvante n’est pas un standard dans les cancers de l’estomac en occident<br />

(standard).<br />

Il est recommandé de poursuivre les essais thérapeutiques comparant l’immunochimiothérapie adjuvante<br />

à la chirurgie seule (recommandation).<br />

Bleiberg 1989<br />

Dent 1979<br />

Moertel 1984<br />

McDonald 2001<br />

High<br />

Yonemura Y 2001 Moderate<br />

Earle 2003 High<br />

Ochiai 1983<br />

Yamamura 1986<br />

Bonfanti 1988<br />

Jakesz 1988<br />

Kim 1992<br />

Popeila 1982<br />

Imaizumi 1990<br />

Kim 1997<br />

Jakesz 1988<br />

IGTSG 1988<br />

Primrose 1998<br />

Tonnesen 1988<br />

Langman 1999<br />

High<br />

High


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 143<br />

Study ID<br />

Chemotherapy<br />

Ref Search<br />

date<br />

Ajani J 2006 [269] October<br />

2005<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Patients <strong>with</strong> gastric<br />

cancer<br />

Oral capecitab<strong>in</strong>e Narrative presentation of results, no<br />

conclusions to be drawn.<br />

Medl<strong>in</strong>e search only<br />

No <strong>in</strong>formation on<br />

selection, quality<br />

appraisal, etc<br />

No <strong>in</strong>formation on search<br />

SR Low<br />

Berrardi R 2004 [303] Not Patients <strong>with</strong> gastric Adjuvant chemotherapy 3 RCTs <strong>in</strong> favour, 3 RCTs aga<strong>in</strong>st.<br />

SR? Low<br />

stated cancer<br />

Narrative presentation of results. at all.<br />

Casc<strong>in</strong>u S 2007 [304] Gastric cancer patients at PELFw regimen (consist<strong>in</strong>g of 5-year survival rates were 52% <strong>in</strong> the No <strong>in</strong>formation on RCT Moderate<br />

high risk for recurrence eight weekly adm<strong>in</strong>istrations PELFw arm and 50% <strong>in</strong> the 5-FU/LV arm. bl<strong>in</strong>d<strong>in</strong>g<br />

<strong>in</strong>clud<strong>in</strong>g patients <strong>with</strong> of cisplat<strong>in</strong> (40 mg/m2), LV Compared <strong>with</strong> the 5-FU/LV regimen,<br />

serosal <strong>in</strong>vasion (stage (250 mg/m2), epidoxorubic<strong>in</strong> the PELFw regimen did not reduce the<br />

pT3 N0) and/or lymph (35 mg/m2), 5-FU (500 risk of death (HR = 0.95, 95%<br />

node metastasis (stage mg/m2), and glutathione (1.5 confidence <strong>in</strong>terval [CI] = 0.70 to 1.29)<br />

pT2 or pT3 N1, N2, or g/m2) <strong>with</strong> the support of or relapse (HR = 0.98, 95% CI = 0.75 to<br />

N3)<br />

filgrastim) (n = 201) vs. 5- 1.29). Less than 10% of patients <strong>in</strong> either<br />

FU/LV regimen (consist<strong>in</strong>g of arm experienced a grade 3 or 4 toxic<br />

six monthly adm<strong>in</strong>istrations of episode. Neutropenia (occurr<strong>in</strong>g more<br />

a 5-day course of 5-FU (375 often <strong>in</strong> the PELFw arm) and diarrhea<br />

mg/m2 daily) and LV (20 and mucositis (more prevalent <strong>in</strong> the 5-<br />

mg/m2 daily, 5-FU/LV)) (n = FU/LV arm) were the most common<br />

196)<br />

serious side effects. Nevertheless, only<br />

19 patients (9.4%) completed the<br />

treatment <strong>in</strong> the PELFw arm and 85<br />

(43%) patients completed the treatment<br />

<strong>in</strong> the 5-FU/LV arm.<br />

Nishikawa T [305] Excluded: re-analysis of RCT<br />

2007<br />

previous RCT (2001)<br />

Nitti D 2006 [172] NA Patients <strong>with</strong><br />

FAMTX or FEMTX vs. control In a planned comb<strong>in</strong>ed analysis of the Prospective meta-analysis MA Moderate<br />

adenocarc<strong>in</strong>oma of the<br />

two trials, no significant differences of 2 RCTs<br />

stomach or esophago-<br />

were found between the treatment and Bl<strong>in</strong>d<strong>in</strong>g??<br />

gastric junction treated<br />

control arms for either DFS (hazards<br />

<strong>with</strong> curative resection<br />

ratio: 0.98, P=0.87) or OS (hazards<br />

ratio: 0.98, P=0.86). The 5-year OS was<br />

43% <strong>in</strong> the treatment arm and 44% <strong>in</strong><br />

the control arm and the 5-year DFS was<br />

41% and 42%, respectively.<br />

Oba K 2006 [168] March Patients <strong>with</strong> gastric Adjuvant oral fluor<strong>in</strong>ated For the 4 trials that met the eligibility Meta-analysis of 4 MA High<br />

2005 cancer<br />

pyrimid<strong>in</strong>es vs. surgery alone criteria, the estimated hazard ratio was Japanese centrally<br />

0.73 (95%CI=0.60-0.89) <strong>in</strong> favour of oral randomized controlled


144 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Tentes A 2006 [306] Patients <strong>with</strong> locally<br />

advanced gastric cancer<br />

after potentially curative<br />

gastrectomy<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

fluor<strong>in</strong>ated pyrimid<strong>in</strong>es. cl<strong>in</strong>ical trials conducted<br />

Adjuvant <strong>in</strong>traarterial<br />

chemotherapy (IARC) vs.<br />

surgery alone<br />

Five-year survival rate for the study and<br />

the control group was 52 and 54%,<br />

respectively (p>0.05). Mean survival for<br />

the study and the control group was<br />

50+/-8 and 62+/-10 months, respectively<br />

(p>0.05). The number of recurrences<br />

and the failure sites were comparable<br />

(p>0.05).<br />

between 1980 and 2005<br />

No <strong>in</strong>formation on<br />

randomization procedure<br />

or bl<strong>in</strong>d<strong>in</strong>g<br />

RCT Moderate<br />

Ueda Y 2006 [307] Excluded: protocol<br />

Bouche O 2005 [169] Stage II-III-IVM0 gastric Postoperative chemotherapy 5- and 7-year overall survival were Bl<strong>in</strong>d<strong>in</strong>g??? RCT Moderate<br />

cancer patients treated (n = 127) vs. surgery alone (n 41.9% and 34.9% <strong>in</strong> the control group<br />

<strong>with</strong> curative resection (n = 133).<br />

versus 46.6% and 44.6% <strong>in</strong> the<br />

= 260)<br />

5-Fluorouracil (5-FU) 800 chemotherapy group (P=0.22). Cox<br />

mg/m(2) daily (5-day<br />

model hazard ratios were 0.74 [95%CI<br />

cont<strong>in</strong>uous <strong>in</strong>fusion) was 0.54-1.02; P=0.063] for death and 0.70<br />

<strong>in</strong>itiated before day 14 after (95%CI 0.51-0.97; P=0.032) for<br />

resection. One month later,<br />

four 5-day cycles of 5-FU (1<br />

g/m² per day) plus cisplat<strong>in</strong><br />

(100 mg/m(2) on day 2) were<br />

adm<strong>in</strong>istered every 4 weeks.<br />

recurrence.<br />

Tsuburaya A<br />

2005<br />

[308] Excluded: protocol<br />

Chipponi J 2004 [170] Patients <strong>with</strong> gastric Surgery alone (n = 104) vs. Because of toxicity, 54% of the patients No <strong>in</strong>formation on RCT Moderate<br />

cancer and lymph node or surgery and adjuvant stopped the protocol before the end of bl<strong>in</strong>d<strong>in</strong>g<br />

serosal <strong>in</strong>volvement or chemotherapy (daily<br />

the n<strong>in</strong>e courses, and 46% of the<br />

both<br />

adm<strong>in</strong>istration of 200 mg/m² patients received the n<strong>in</strong>e courses<br />

of fol<strong>in</strong>ic acid, 5-fluorouracil <strong>in</strong>clud<strong>in</strong>g 32% <strong>with</strong> a decreased dose and<br />

(375 mg/m² dur<strong>in</strong>g the first 14% <strong>with</strong> a full dose. The 5-year survival<br />

session <strong>in</strong>creas<strong>in</strong>g 25 mg by rate was 39% <strong>in</strong> the control group and<br />

session until reach<strong>in</strong>g 500<br />

mg/m²) and CDDP 15 mg/m²<br />

(n = 101)<br />

39% <strong>in</strong> the chemotherapy group.<br />

Nashimoto A [171] Serosa-negative gastric Adjuvant chemotherapy There was no significant difference <strong>in</strong> No <strong>in</strong>formation on RCT Moderate<br />

2003<br />

cancer patients (exclud<strong>in</strong>g (<strong>in</strong>travenous mitomyc<strong>in</strong> 1.33 relapse-free and overall survival bl<strong>in</strong>d<strong>in</strong>g<br />

patients who were T1N0) mg/m², fluorouracil (FU) 166.7 between the arms (5-year relapse-free<br />

(n = 252)<br />

mg/m², and cytarab<strong>in</strong>e 13.3 survival 88.8% chemotherapy v 83.7%


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 145<br />

Study ID Ref Search<br />

date<br />

Radiotherapy<br />

No additional studies found<br />

Chemoradiotherapy<br />

No additional studies found<br />

Intraperitoneal chemotherapy<br />

Yan TD 2007 [175] Decemb<br />

er 2006<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

mg/m² twice weekly for the surgery alone; P =.14 and 5-year survival<br />

first 3 weeks after surgery, 91.2% chemotherapy v 86.1% surgery<br />

and oral FU 134 mg/m² daily alone; P =.13, respectively). N<strong>in</strong>e<br />

for the next 18 months for a patients (7.1%) <strong>in</strong> the chemotherapy arm<br />

total dose of 67 g/m²) vs. and 17 patients (13.8%) <strong>in</strong> the surgery-<br />

surgery alone<br />

alone arm had cancer recurrence.<br />

Patients <strong>with</strong> locally<br />

advanced resectable<br />

gastric cancer<br />

Adjuvant <strong>in</strong>traperitoneal<br />

chemotherapy<br />

A significant improvement <strong>in</strong> survival<br />

was associated <strong>with</strong> hyperthermic<br />

<strong>in</strong>traoperative <strong>in</strong>traperitoneal<br />

chemotherapy (HIIC) alone (HR = 0.60;<br />

95% CI = 0.43 to 0.83; p = 0.002) or<br />

comb<strong>in</strong>ed <strong>with</strong> early postoperative<br />

<strong>in</strong>traperitoneal chemotherapy (EPIC)<br />

(HR = 0.45; 95% CI = 0.29 to 0.68; p =<br />

0.0002). Survival improvement was<br />

marg<strong>in</strong>ally significant (p = 0.06) <strong>with</strong><br />

normothermic <strong>in</strong>traoperative<br />

<strong>in</strong>traperitoneal chemotherapy, but not<br />

significant <strong>with</strong> EPIC alone or delayed<br />

postoperative <strong>in</strong>traperitoneal<br />

chemotherapy. Intraperitoneal<br />

chemotherapy was also found to be<br />

associated <strong>with</strong> higher risks for <strong>in</strong>traabdom<strong>in</strong>al<br />

abscess (RR = 2.37; 95% CI =<br />

1.32 to 4.26; p = 0.003) and neutropenia<br />

(RR = 4.33; 95% CI = 1.49 to 12.61; p =<br />

0.007).<br />

13 RCTs of which 10<br />

were of good quality<br />

Very good SR<br />

Yu W 2006 [309] Excluded narrative review<br />

Xu DZ 2004 [174] January<br />

2003<br />

Patients undergo<strong>in</strong>g<br />

curative resection<br />

for gastric cancer<br />

Adjuvant <strong>in</strong>traperitoneal<br />

chemotherapy<br />

Intraperitoneal chemotherapy may<br />

benefit the patients after curative<br />

resection for locally advanced gastric<br />

cancer, and the comb<strong>in</strong>ation of<br />

<strong>in</strong>traperitoneal chemotherapy <strong>with</strong><br />

Also identified by Yan et<br />

al.<br />

Ch<strong>in</strong>ese and <strong>English</strong> only<br />

11 RCTs identified, of<br />

which 2 were not<br />

SR High<br />

SR High


146 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Immunotherapy<br />

Chen DW 2005 [310] Patients <strong>with</strong> gastric<br />

carc<strong>in</strong>oma who had<br />

undergone major surgery<br />

Oba K 2007 [176] Not<br />

stated<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

hyperthermia or activated carbon identified by Yan et al.<br />

particles may provide more benefits to (Chen ZX 1996, Tan CQ<br />

patients due to the enhanced antitumor<br />

activity of drugs. Sensitivity analysis and<br />

fail-safe number suggested that the<br />

result was comparatively reliable.<br />

However, of 11 trials, only 3 studies<br />

were of high quality.<br />

2000)<br />

Patients <strong>with</strong> curative<br />

resections of gastric<br />

cancers<br />

Farreras N 2005 [311] Patients <strong>with</strong> gastric<br />

cancer (n = 66)<br />

Popiela T 2004 [177] Patients <strong>with</strong> stage III or<br />

IV gastric cancer who had<br />

undergone curative<br />

resection (n = 156)<br />

Immunonutrition (n = 20) vs.<br />

standard nutrition (n = 20)<br />

Immunochemotherapy <strong>with</strong><br />

polysaccharide K (PSK)<br />

Early postoperative enteral<br />

immunonutrition (formula<br />

supplemented <strong>with</strong> arg<strong>in</strong><strong>in</strong>e,<br />

omega-3 fatty acids and<br />

ribonucleic acid (RNA)) vs. an<br />

isocaloric-isonitrogenous<br />

control<br />

BCG + FAM<br />

(immunochemotherapy) vs.<br />

FAM (chemotherapy) vs.<br />

control (surgery only)<br />

No serious adverse effects were<br />

recorded <strong>with</strong> the two formulas.<br />

Postoperative procedures were smooth<br />

<strong>in</strong> both groups.<br />

The overall hazard ratio for eligible<br />

patients was 0.88 (95% confidence<br />

<strong>in</strong>terval, 0.79-0.98; P = 0.018) <strong>with</strong> no<br />

significant heterogeneity [chi (2)(8) for<br />

heterogeneity = 11.7; P = 0.16]. The<br />

results of this meta-analysis suggest that<br />

adjuvant immunochemotherapy <strong>with</strong><br />

PSK improves the survival of patients<br />

after curative gastric cancer resection.<br />

Patients fed <strong>with</strong> immunonutrition<br />

(n=30) showed significantly lower<br />

episodes of surgical wound heal<strong>in</strong>g<br />

complications (0 vs. 8 (26.7%) P=0.005)<br />

when compared to patients fed <strong>with</strong> the<br />

control formula (n=30).<br />

Overall 10-year survival was 47.1% for<br />

the immunochemotherapy group (P <<br />

0.037 vs FAM and P < 0.0006 vs<br />

control), 30% for the chemotherapy<br />

group (vs control, NS), and 15.2% for<br />

the control group. In patients <strong>with</strong><br />

pT2/T3 primary tumors, 10-year survival<br />

was 55.3% for BCG + FAM vs 28.2% for<br />

FAM (P < 0.01) and 14.6% for the<br />

control group (P < 0.00018). BCG +<br />

No full-text RCT<br />

8 RCTs <strong>in</strong>cluded SR High<br />

No ITT anlysis (6 patients<br />

excluded, 3 <strong>in</strong> both arms)<br />

Randomization <strong>with</strong><br />

envelope<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

RCT Moderate<br />

RCT Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 147<br />

Study ID Ref Search<br />

date<br />

Sato Y 2004 [312] Patients who underwent<br />

curative resection of<br />

gastric cancer<br />

Palliative treatment.<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

FAM significantly improved the survival<br />

of patients <strong>with</strong> <strong>in</strong>test<strong>in</strong>al-type but not<br />

diffuse-type cancer.<br />

Immunochemotherapy was well<br />

800 mg/d 5'-DFUR (Furtulon)<br />

for 2 years from 2 weeks after<br />

the operation (n = 143) vs.<br />

OK-432 plus 5'-DFUR by the<br />

same regimen (n = 144)<br />

tolerated.<br />

The 5-year survival rates for groups A<br />

and B were 62.9% and 63.8%,<br />

respectively, show<strong>in</strong>g no significant<br />

difference (P = 0.7996).<br />

No full-text RCT<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

Surgery<br />

date<br />

evidence<br />

SIGN [47] 2004 Palliative gastrectomy should be avoided <strong>in</strong> patients <strong>with</strong> gastric cancer who have dissem<strong>in</strong>ated<br />

peritoneal disease.<br />

Green D 2002 Low<br />

D2 lymphadenectomy should be avoided <strong>in</strong> patients <strong>with</strong> gastric cancer <strong>in</strong> the palliative sett<strong>in</strong>g. Hanazaki K 1999 Low<br />

Health professionals should take the follow<strong>in</strong>g factors <strong>in</strong>to account when consider<strong>in</strong>g palliative gastric Green D 2002<br />

Low<br />

resection:<br />

Kunisaki C 2003<br />

- peritoneal disease<br />

- tumour diameter<br />

- histological type<br />

- degree of differentiation<br />

Laparoscopic bypass or gastric outlet stent<strong>in</strong>g are alternatives to palliative gastric bypass. Brune IB 1997 Low<br />

Palliative gastric bypass should be avoided when malignant ascites or small bowel obstruction are<br />

present.<br />

Blair S 2001 Low<br />

Exploratory laparotomy alone should be avoided. Ouchi K 1998 Low<br />

FNCLCC [129] January La chirurgie palliative est <strong>in</strong>diquée pour les sténoses symptomatiques, les tumeurs qui saignent et parfois<br />

Low<br />

2002 en urgence pour perforation. Il n’y a pas d’<strong>in</strong>dication à la chirurgie s’il existe une atte<strong>in</strong>te péritonéale, une<br />

ascite néoplasique ou des métastases hépatiques et si la tumeur est peu symptomatique (standard).<br />

L’exérèse ou la dérivation peuvent être proposes (option). Low<br />

L’<strong>in</strong>dication de chirurgie palliative dépend de l’<strong>in</strong>tensité des signes fonctionnels, de l’état général et<br />

nutritionnel, de l’âge, des ressources thérapeutiques complémentaires utilisables et surtout de la<br />

résécabilité et d’une espérance de vie supérieure à 6 mois. L’exérèse est préférable à la derivation<br />

(recommendation).<br />

Low<br />

Chemotherapy


148 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

CPG ID Ref Search Recommendation Support<strong>in</strong>g evidence Level of<br />

date<br />

evidence<br />

SIGN [47] 2004 In patients <strong>with</strong> locally advanced or metastatic cancer of the stomach <strong>with</strong> good performance status<br />

comb<strong>in</strong>ation chemotherapy <strong>in</strong>clud<strong>in</strong>g cisplat<strong>in</strong> and <strong>in</strong>fusional 5FU (such as ECF or MCF) should be<br />

considered.<br />

Janunger K 2002 High<br />

FNCLCC [129] January La chimiothérapie doit être proposée aux patients en bon état général dans le but d’améliorer leur Murad 1993<br />

High<br />

2002 survie (standard).<br />

Pyrhonen 1995<br />

Glimelius 1997<br />

Modalités (option):<br />

Cull<strong>in</strong>an 1995<br />

High<br />

- monochimiothérapie par 5FU - acide fol<strong>in</strong>ique<br />

Kim 1993<br />

- polychimiothérapie par des comb<strong>in</strong>aisons à base de 5FU et de cisplat<strong>in</strong>e<br />

Cull<strong>in</strong>an 1994<br />

Many other RCTs<br />

Il est recommandé de poursuivre les essais thérapeutiques pour préciser l’<strong>in</strong>térêt de nouveaux<br />

médicaments en première ligne et en seconde ligne. Le choix de la chimiothérapie doit être fait en<br />

fonction de l’état général et de l’âge. Une polychimiothérapie est recommandée pour les patients en bon<br />

état général. Le choix entre ces différentes options thérapeutiques doit prendre en compte l'avis ou le<br />

souhait des patients (recommendation).<br />

No evidence provided<br />

Supportive care<br />

SIGN [47] 2004 Patients <strong>with</strong> gastric cancer should have access to a specialist palliative care team. Smeenk FW 1998<br />

Micc<strong>in</strong>esi G 2003<br />

Costant<strong>in</strong>i M 2003<br />

Costant<strong>in</strong>i M 1993<br />

Low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 149<br />

Study ID<br />

Chemotherapy<br />

Ref Search<br />

date<br />

Ajani J 2006 [269] October<br />

2005<br />

Trumper M<br />

2006<br />

Di Cosimo S<br />

2003<br />

Wagner AD<br />

2005 & 2006<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Patients <strong>with</strong> gastric<br />

cancer<br />

Oral capecitab<strong>in</strong>e Narrative presentation of results, no<br />

conclusions to be drawn.<br />

Medl<strong>in</strong>e search only<br />

No <strong>in</strong>formation on<br />

selection, quality<br />

appraisal, etc<br />

SR Low<br />

[274] Excluded Retrospective<br />

subanalysis of<br />

[313] February<br />

2003<br />

[181] February<br />

2005<br />

Patients <strong>with</strong> advanced<br />

gastric cancer<br />

Patients <strong>with</strong> advanced<br />

gastric cancer<br />

Docetaxel Eight phase II trials focused on docetaxel<br />

as a s<strong>in</strong>gle agent. Consider<strong>in</strong>g<br />

collectively the 262 evaluable patients<br />

enrolled <strong>in</strong> these studies, the mean<br />

response rate (RR) was 19% (CI 95%<br />

14-24%). Docetaxel was well tolerated<br />

<strong>with</strong> a dose-limit<strong>in</strong>g myelosuppression<br />

(grade 3-4 neutropenia <strong>in</strong> 36-95% of<br />

cases). Add<strong>in</strong>g fluorouracil, an RR<br />

rang<strong>in</strong>g from 22% to 86% was<br />

registered, due to differences <strong>in</strong><br />

populations studied (young vs elderly)<br />

and modalities of drug adm<strong>in</strong>istration<br />

(cont<strong>in</strong>uous vs. bolus <strong>in</strong>fusion). RRs for<br />

docetaxel-cisplat<strong>in</strong> comb<strong>in</strong>ation were<br />

56%, 37% and 36% <strong>in</strong> three phase II trials<br />

and 35% <strong>in</strong> a phase III trial. The addition<br />

of both cisplat<strong>in</strong> and fluorouracil to<br />

docetaxel did not <strong>in</strong>crease toxicity.<br />

Randomized trials compar<strong>in</strong>g docetaxelcisplat<strong>in</strong>-fluorouracil<br />

<strong>with</strong> ciplat<strong>in</strong>fluorouoracil<br />

or epirubic<strong>in</strong>-cisplat<strong>in</strong>fluorouracil,<br />

the most commonly used<br />

regimens, are ongo<strong>in</strong>g.<br />

Chemotherapy Analysis of chemotherapy versus best<br />

supportive care (HR = 0.39; 95% CI,<br />

0.28 to 0.52) and comb<strong>in</strong>ation versus<br />

s<strong>in</strong>gle agent, ma<strong>in</strong>ly fluorouracil (FU) -<br />

based chemotherapy (HR = 0.83; 95%<br />

No <strong>in</strong>formation on<br />

selection process or<br />

quality appraisal<br />

3 RCTs<br />

SR Low<br />

27 RCTs <strong>in</strong>cluded SR High


150 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Wohrer SS 2004 [314] Patients <strong>with</strong> advanced<br />

gastric cancer<br />

Ajani J 2005 [315] Untreated patients <strong>with</strong><br />

confirmed advanced<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

CI = 0.74 to 0.93) showed significant<br />

overall survival benefits <strong>in</strong> favor of<br />

chemotherapy and comb<strong>in</strong>ation<br />

chemotherapy, respectively. In addition,<br />

comparisons of FU/cisplat<strong>in</strong>-conta<strong>in</strong><strong>in</strong>g<br />

regimens <strong>with</strong> versus <strong>with</strong>out<br />

anthracycl<strong>in</strong>es (HR = 0.77; 95% CI, 0.62<br />

to 0.95) and FU/anthracycl<strong>in</strong>e-conta<strong>in</strong><strong>in</strong>g<br />

comb<strong>in</strong>ations <strong>with</strong> versus <strong>with</strong>out<br />

cisplat<strong>in</strong> (HR = 0.83; 95% CI, 0.76 to<br />

0.91) both demonstrated a significant<br />

survival benefit for the three-drug<br />

comb<strong>in</strong>ation. Compar<strong>in</strong>g ir<strong>in</strong>otecanconta<strong>in</strong><strong>in</strong>g<br />

versus nonir<strong>in</strong>otecanconta<strong>in</strong><strong>in</strong>g<br />

comb<strong>in</strong>ations (ma<strong>in</strong>ly<br />

FU/cisplat<strong>in</strong>) resulted <strong>in</strong> a nonsignificant<br />

survival benefit <strong>in</strong> favor of the<br />

ir<strong>in</strong>otecan-conta<strong>in</strong><strong>in</strong>g regimens (HR =<br />

0.88; 95% CI, 0.73 to 1.06), but they<br />

have never been compared aga<strong>in</strong>st a<br />

three-drug comb<strong>in</strong>ation.<br />

Chemotherapy Analysis of results shows chemotherapy<br />

to be superior to best supportive care<br />

alone. Comb<strong>in</strong>ation chemotherapy<br />

compared <strong>with</strong> monochemotherapy is<br />

associated <strong>with</strong> significantly higher<br />

overall (complete plus partial) response<br />

rates but nevertheless results <strong>in</strong> similar<br />

survival. ECF (epirubic<strong>in</strong>, cisplat<strong>in</strong> and 5fluorouracil)<br />

currently represents one of<br />

the most effective regimens for<br />

advanced gastric cancer, whereas among<br />

the newer comb<strong>in</strong>ations, ir<strong>in</strong>otecan- or<br />

taxane-based regimens have also given<br />

promis<strong>in</strong>g results. In patients <strong>with</strong> a<br />

poor performance status, consideration<br />

could be given to leucovor<strong>in</strong>-modulated<br />

DCF (docetaxel 75 mg/m2,<br />

cisplat<strong>in</strong> 75 mg/m² on day 1,<br />

5-fluorouracil alone.<br />

Of 158 randomly assigned patients, 155<br />

(DCF, n = 79; DC, n = 76) received<br />

Medl<strong>in</strong>e and <strong>English</strong> only<br />

No <strong>in</strong>formation on<br />

selection process or<br />

quality appraisal<br />

SR Low<br />

RCT High


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 151<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

gastric or<br />

and fluorouracil 750 mg/m²/d treatment. The confirmed ORR was<br />

gastroesophageal as cont<strong>in</strong>uous <strong>in</strong>fusion on days 43% for DCF (n = 79) and 26% for DC<br />

adenocarc<strong>in</strong>oma 1 to 5) (n = 79) vs. DC (n = 76). Median time to progression<br />

(docetaxel 85 mg/m² and was 5.9 months for DCF and 5.0 months<br />

cisplat<strong>in</strong> 75 mg/m² on day 1) for DC. Median overall survival time was<br />

every 3 weeks (n = 76) 9.6 months for DCF and 10.5 months<br />

for DC. The most frequent grade 3 and<br />

4 events per patient <strong>in</strong>cluded<br />

neutropenia (DCF = 86%; DC = 87%)<br />

and GI (DCF = 56%; DC = 30%).<br />

Bouche O 2004 [316] Patients <strong>with</strong> histologically LV 200 mg/m² (2-hour The overall response rates, which were No <strong>in</strong>formation on exact RCT Moderate<br />

proven metastatic gastric <strong>in</strong>fusion) followed by FU 400 confirmed by an <strong>in</strong>dependent expert randomization procedure<br />

or cardial adenocarc<strong>in</strong>oma mg/m² (bolus) and FU 600 panel, were 13% (95%CI, 3.4% to or bl<strong>in</strong>d<strong>in</strong>g<br />

<strong>with</strong>out l<strong>in</strong>itis<br />

mg/m² (22-hour cont<strong>in</strong>uous 23.3%), 27% (95%CI, 14.1% to 40.4%), Included <strong>in</strong> Wagner 2006<br />

<strong>in</strong>fusion) on days 1 and 2 and 40% (95%CI, 25.7% to 54.3%) for<br />

every 14 days (LV5FU2; arm arms A, B, and C, respectively. Median<br />

A) (n = 45) vs. LV5FU2 plus progression-free survival and overall<br />

cisplat<strong>in</strong> 50 mg/m² (1-hour survival times were 3.2 months (95%CI,<br />

<strong>in</strong>fusion) on day 1 or 2 (arm 1.8 to 4.6 months) and 6.8 months<br />

B) (n = 44), vs. LV5FU2 plus (95%CI, 2.6 to 11.1 months) <strong>with</strong><br />

ir<strong>in</strong>otecan 180 mg/m² (2-hour LV5FU2, respectively; 4.9 months<br />

<strong>in</strong>fusion) on day 1 (arm C) (n (95%CI, 3.5 to 6.3 months) and 9.5<br />

= 45)<br />

months (95%CI, 6.9 to 12.2 months)<br />

<strong>with</strong> LV5FU2-cisplat<strong>in</strong>, respectively; and<br />

6.9 months (95%CI, 5.5 to 8.3 months)<br />

and 11.3 months (95%CI, 9.3 to 13.3<br />

months) <strong>with</strong> LV5FU2-ir<strong>in</strong>otecan,<br />

respectively.<br />

Cocconi G 2003 [317] Patients <strong>with</strong> untreated 5-Fluorouracil (5-FU), The complete response (CR) rates to No <strong>in</strong>formation on RCT Moderate<br />

advanced gastric doxorubic<strong>in</strong> and methotrexate PELF and FAMTX were, respectively, bl<strong>in</strong>d<strong>in</strong>g<br />

carc<strong>in</strong>oma<br />

(FAMTX) (n = 97) vs. cisplat<strong>in</strong>, 13% [95% confidence <strong>in</strong>tervals (CI) 6% Included <strong>in</strong> Wagner 2006<br />

epirubic<strong>in</strong>, leucovor<strong>in</strong> and 5- to 20%] and 2% (95% CI 0% to 5%; P =<br />

FU (PELF) (n = 98)<br />

0.003), and the objective response rates<br />

[CR plus partial response (PR) rates]<br />

39% (95% CI 29% to 49%) and 22% (95%<br />

CI 13% to 30%; P = 0.009), thus<br />

significantly favour<strong>in</strong>g the PELF<br />

comb<strong>in</strong>ation. The survival rates after 12<br />

months (30.8% versus 22.4%) and 24<br />

months (15.7% versus 9.5%) were also


152 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Koizumi W<br />

2004<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

higher among patients receiv<strong>in</strong>g PELF,<br />

but these differences were not<br />

statistically significant. The toxicities<br />

were qualitatively different but<br />

quantitatively similar. Both regimens<br />

seem to be feasible provided that careful<br />

[318] Patients <strong>with</strong> advanced<br />

unresectable gastric<br />

cancer<br />

Lutz M 2007 [319] Patients <strong>with</strong> advanced<br />

gastric cancer<br />

Moehler M 2005 [320] Patients <strong>with</strong> untreated<br />

metastatic or locally<br />

advanced gastric cancer<br />

Cisplat<strong>in</strong> + mitomyc<strong>in</strong> + oral<br />

5’-DFUR (n = 32) vs. Cisplat<strong>in</strong><br />

+ oral 5’-DFUR (n = 29)<br />

Weekly FU 3000 mg/m²/24<br />

hours (HD-FU) (n = 37), FU<br />

2600 mg/m²/24 hours plus dl-<br />

FA 500 mg/m² or l-FA 250<br />

mg/m² (HD-FU/FA) (n = 53),<br />

vs. FU 2000 mg/m²/24 hours<br />

plus FA plus biweekly Cis 50<br />

mg/m² (n = 51), each<br />

adm<strong>in</strong>istered for 6 weeks <strong>with</strong><br />

a 1-week rest<br />

Ir<strong>in</strong>otecan plus high-dose 5fluorouracil<br />

(5-FU) and<br />

leucovor<strong>in</strong> (LV) (ILF) (n = 56)<br />

vs. etoposide plus 5-FU/LV<br />

(ELF) (n = 58)<br />

patient monitor<strong>in</strong>g is assured.<br />

No significant differences <strong>in</strong> response<br />

rate and median survival time.<br />

Confirmed responses were observed <strong>in</strong><br />

6.1% (two of 33) of the eligible patients<br />

treated <strong>with</strong> HD-FU, <strong>in</strong> 25% (12 of 48,<br />

<strong>in</strong>clud<strong>in</strong>g one complete remission [CR])<br />

<strong>with</strong> HD-FU/FA, and <strong>in</strong> 45.7% (21 of 46,<br />

<strong>in</strong>clud<strong>in</strong>g four CRs) <strong>with</strong> HD-FU/FA/Cis.<br />

The HD-FU arm was closed after stage<br />

1 because the required m<strong>in</strong>imum<br />

number of responses was not met. The<br />

median progression-free survival of all<br />

patients <strong>in</strong> the HD-FU, HD-FU/FA, and<br />

HD-FU/FA/Cis arm was 1.9, 4.0, and 6.1<br />

months, respectively. The median<br />

overall survival was 7.1, 8.9, and 9.7<br />

months, and the survival rate at 1 year<br />

was 24.3%, 30.3%, and 45.3%,<br />

respectively. Grade 4 toxicities were<br />

rare. The most relevant grade 3/4<br />

toxicities were neutropenia <strong>in</strong> 1.9%,<br />

5.4%, and 19.6%, and diarrhea <strong>in</strong> 2.7%,<br />

1.9%, and 3.9% of the cycles <strong>in</strong> the HD-<br />

FU, HD-FU/FA, and HD-/FU/Cis arms,<br />

respectively.<br />

The objective cl<strong>in</strong>ical response rates<br />

after 14 weeks treatment were 30% for<br />

ILF and 17% for ELF (RR 0.57, 95%CI<br />

0.29–1.13, P=0.077). Overall response<br />

rates over the entire treatment period<br />

for ILF and ELF were 43 and 24%,<br />

respectively (RR 0.56, 95%CI 0.33–0.97;<br />

No <strong>in</strong>formation on exact<br />

randomization procedure<br />

Bl<strong>in</strong>d<strong>in</strong>g??<br />

No <strong>in</strong>formation on exact<br />

randomization procedure<br />

No <strong>in</strong>formation on exact<br />

randomization procedure<br />

Bl<strong>in</strong>d<strong>in</strong>g???<br />

Included <strong>in</strong> Wagner 2006<br />

RCT Moderate<br />

RCT Moderate<br />

RCT Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 153<br />

Study ID Ref Search<br />

date<br />

Ohtsu A 2003 [321] Patients <strong>with</strong> advanced<br />

gastric cancer<br />

Park SH 2006 [322] Patients <strong>with</strong> measurable<br />

metastatic gastric cancer<br />

Pozzo C 2004 [323] Patients <strong>with</strong> advanced<br />

gastric or oesophagealgastric<br />

junction<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

P=0.047). For ILF and ELF, respectively,<br />

median progression-free survival was 4.5<br />

vs 2.3 months, time to treatment failure<br />

was 3.6 vs 2.2 months (P=0.45), and<br />

overall survival was 10.8 vs 8.3 months<br />

(P=0.28). Both regimens were well<br />

tolerated, the ma<strong>in</strong> grade 3/4 toxicities<br />

be<strong>in</strong>g diarrhoea (18%, ILF) and<br />

Fluorouracil (FU) alone vs. FU<br />

plus cisplat<strong>in</strong> (FP) vs. uracil<br />

and tegafur plus mitomyc<strong>in</strong><br />

(UFTM)<br />

Paclitaxel + 5FU (n = 38) vs.<br />

docetaxel + 5FU (n = 38)<br />

Ir<strong>in</strong>otecan [80 mg/m2² IV], FA<br />

(500 mg/m² IV) and a 22-h<br />

<strong>in</strong>fusion of 5-FU (2000 mg/m²<br />

neutropenia (57%, ELF).<br />

At the <strong>in</strong>terim analysis, the UFTM arm<br />

showed a significantly <strong>in</strong>ferior survival<br />

<strong>with</strong> higher <strong>in</strong>cidences of hematologic<br />

toxic effects than did control arm FU<br />

alone, and the registration to UFTM was<br />

term<strong>in</strong>ated. Both <strong>in</strong>vestigational<br />

regimens, FP and UFTM, had a<br />

significantly higher <strong>in</strong>cidence of<br />

hematologic toxic effects than FU alone,<br />

although the effects were manageable.<br />

The overall response rates of the FUalone,<br />

FP, and UFTM arms were 11%,<br />

34%, and 9%, respectively. The median<br />

progression-free survival was 1.9<br />

months <strong>with</strong> FU alone, 3.9 months <strong>with</strong><br />

FP, and 2.4 months <strong>with</strong> UFTM,<br />

respectively. Although FP demonstrated<br />

a higher response rate (P < .001) and<br />

longer progression free survival than did<br />

FU alone (P < .001), no differences <strong>in</strong><br />

overall survival were observed between<br />

the arms. The median survival times and<br />

1-year survival rates were 7.1 months<br />

and 28% <strong>with</strong> FU, 7.3 months and 29%<br />

<strong>with</strong> FP, and 6.0 months and 16% <strong>with</strong><br />

UFTM, respectively.<br />

No <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g<br />

Included <strong>in</strong> Wagner 2006<br />

RCT Moderate<br />

No significant differences No <strong>in</strong>formation on<br />

randomization procedure<br />

RCT Moderate<br />

The overall response rate <strong>in</strong> the<br />

ir<strong>in</strong>otecan/5-FU/FA arm was 42.4%, <strong>with</strong><br />

a complete response rate of 5.1%.<br />

No ITT RCT Moderate


154 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

adenocarc<strong>in</strong>oma IV), weekly for 6 weeks <strong>with</strong> Correspond<strong>in</strong>g figures for the<br />

a 1-week rest (n = 59), vs. ir<strong>in</strong>otecan/cisplat<strong>in</strong> arm were 32.1% and<br />

ir<strong>in</strong>otecan (200 mg/m² IV) and 1.8%, respectively. The median time to<br />

cisplat<strong>in</strong> (60 mg/m² IV), on day progression was 6.5 months<br />

1 for 3 weeks (n = 58) (ir<strong>in</strong>otecan/5-FU/FA) and 4.2 months<br />

(ir<strong>in</strong>otecan/<br />

cisplat<strong>in</strong>) (P < 0.0001), <strong>with</strong> median<br />

survival times of 10.7 and 6.9 months,<br />

respectively (P = 0.0018). The major<br />

toxicity was grade 3/4 neutropenia,<br />

which was more pronounced <strong>with</strong><br />

ir<strong>in</strong>otecan/cisplat<strong>in</strong> than <strong>with</strong><br />

ir<strong>in</strong>otecan/5-FU/FA (65.7% versus 27%).<br />

Diarrhea was the ma<strong>in</strong> grade 3/4 nonhematological<br />

toxicity <strong>with</strong> both<br />

ir<strong>in</strong>otecan/5-FU/FA (27.0%) and<br />

ir<strong>in</strong>otecan/cisplat<strong>in</strong> (18.1%).<br />

Sadighi S 2006 [324] Patients <strong>with</strong> advanced Docetaxel, cisplat<strong>in</strong>, 5-FU Only the TCF group showed statistically No <strong>in</strong>formation on RCT Moderate<br />

gastric cancer<br />

(TCF) (n = 44) vs. epirubic<strong>in</strong>, and cl<strong>in</strong>ically mean<strong>in</strong>gful improvement <strong>in</strong> randomization procedure<br />

cisplat<strong>in</strong>, 5-FU (ECF) (n = 42) global QOL (P = 0.001). Surgical and<br />

pathologic response were better <strong>with</strong><br />

TCF, but there was no difference <strong>in</strong><br />

survival rate between two groups.<br />

or bl<strong>in</strong>d<strong>in</strong>g<br />

Sumpter K 2005 [270] Patients <strong>with</strong> <strong>in</strong>operable, ECF (n = 53)<br />

First <strong>in</strong>terim analysis was performed Interim analysis<br />

RCT Moderate<br />

histologically verified ECX (n = 48)<br />

when 80 pts had been randomised. 6 orig<strong>in</strong>ally randomized<br />

locally advanced or EOF (n = 55)<br />

Doselimit<strong>in</strong>g fluoropyrimid<strong>in</strong>e toxicities pts not <strong>in</strong>cluded <strong>in</strong> results<br />

metastatic<br />

EOX (n = 48)<br />

were stomatitis, palmar plantar Non<strong>in</strong>feriority study<br />

adenocarc<strong>in</strong>oma, (E = epirubuc<strong>in</strong>, C = cisplat<strong>in</strong>, erythema (PPE) and diarrhoea; 5.1% of No <strong>in</strong>formation on<br />

squamous cell or F = 5FU, O = oxaliplat<strong>in</strong>, X = X-treated pts experienced grade 3/4 bl<strong>in</strong>d<strong>in</strong>g of randomization<br />

undifferentiated<br />

capecitab<strong>in</strong>)<br />

toxicity. Protocol planned dose<br />

carc<strong>in</strong>oma of the<br />

escalation of X to 625 mg/m² b.i.d. was<br />

oespohagus,<br />

<strong>in</strong>stituted and a second <strong>in</strong>terim analysis<br />

oesophagogastric junction<br />

has been performed. A total of 204 pts<br />

(OGJ) or stomach<br />

were randomised at the time of the<br />

protocol planned 2nd <strong>in</strong>terim analysis.<br />

Grade 3/4 fluoropyrimid<strong>in</strong>e-related<br />

toxicity was seen <strong>in</strong> 13.7% pts receiv<strong>in</strong>g<br />

F, 8.4% pts receiv<strong>in</strong>g X 500 mg/m² b.i.d.<br />

and 14.7% pts receiv<strong>in</strong>g X 625 mg/m²<br />

b.i.d. Comb<strong>in</strong>ed complete and partial


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 155<br />

Study ID Ref Search<br />

date<br />

Takahashi Y<br />

2004<br />

Thuss-Patience P<br />

2005<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

response rates were ECF 31% (95% CI<br />

18.7–46.3), EOF 39% (95% CI 25.9–<br />

53.1), ECX 35% (95% CI 21.4–50.3),<br />

EOX 48% (95% CI 33.3–62.8). Grade<br />

3/4 fluoropyrimid<strong>in</strong>e toxicity affected<br />

14.7% of pts treated <strong>with</strong> X 625<br />

mgm 2 b.i.d. 1, which is similar to<br />

that observed <strong>with</strong> F, confirm<strong>in</strong>g this to<br />

be the optimal dose. The replacement of<br />

C by O and F by X does not appear to<br />

impair efficacy. The trial cont<strong>in</strong>ues to<br />

[325] Patients <strong>with</strong> advanced or<br />

recurrent gastric<br />

carc<strong>in</strong>oma<br />

[326] Patients <strong>with</strong> metastatic<br />

or locally advanced gastric<br />

adenocarc<strong>in</strong>oma <strong>with</strong>out<br />

prior chemotherapy (n =<br />

94)<br />

Tailored CPT-11 + S-1 vs S-1<br />

total accrual of 1000 pts.<br />

Excluded: protocol RCT<br />

ECF (epirubic<strong>in</strong> 50 mg/m² day<br />

1, cisplat<strong>in</strong> 60 mg/m² day 1,<br />

and fluorouracil 200 mg/m²<br />

days 1 through 21, every 3<br />

weeks) (n = 45) or DF<br />

(docetaxel 75 mg/m² day 1,<br />

and fluorouracil 200 mg/m²<br />

days 1 through 21, every 3<br />

weeks) (n = 45)<br />

In the DF arm, two patients (4.4%,<br />

<strong>in</strong>tent to treat) experienced a confirmed<br />

complete tumor remission as best<br />

response, and 15 patients (33.3%)<br />

experienced a confirmed partial<br />

remission (overall response rate [ORR],<br />

37.8%; 95% CI, 25.9% to 51.9%). Two<br />

patients (4.4%) <strong>in</strong> the ECF arm showed<br />

confirmed complete remission, and 14<br />

(31.1%) showed confirmed partial<br />

remission<br />

(ORR, 35.6%; 95% CI, 24.8% to 48.7%).<br />

For the DF and ECF arms, the median<br />

survival was 9.5 and 9.7 months, and the<br />

median time to tumor progression 5.5<br />

and 5.3 months, respectively.<br />

No <strong>in</strong>formation on<br />

randomization procedure<br />

Four patients excluded<br />

because of violation of<br />

<strong>in</strong>clusion criteria<br />

RCT Moderate<br />

Tsuburaya A<br />

2005<br />

[308] Excluded: protocol RCT<br />

Berardi R 2004 [303] Not Patients <strong>with</strong> gastric<br />

No <strong>in</strong>formation on search SR? Low<br />

stated cancer<br />

at all: excluded<br />

Casaretto L [180] Not Patients diagnosed <strong>with</strong> Chemotherapy versus support The 1-year survival rate was 8% for 5 RCTs <strong>in</strong>cluded SR High<br />

2006<br />

stated gastric adenocarc<strong>in</strong>oma cancer treatment<br />

support care and 20% for chemotherapy<br />

(RR = 2.14, 95%CI = 1.00-4.57, P =<br />

0.05); 30% of the patients <strong>in</strong> the<br />

chemotherapy group and 12% <strong>in</strong> the


156 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Bonneta<strong>in</strong> F<br />

2005<br />

Van Cutsem E<br />

2006<br />

Stent<strong>in</strong>g<br />

Dormann A<br />

2004<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

support care group atta<strong>in</strong>ed a 6-month<br />

symptom-free period (RR = 2.33, 95%CI<br />

= 1.41-3.87, P < 0.01). Quality of life<br />

evaluated after 4 months was<br />

significantly better for the chemotherapy<br />

patients (34%; RR = 2.07, 95%CI = 1.31-<br />

3.28, P < 0.01) <strong>with</strong> tumor mass<br />

reduction (RR = 3.32, 95%CI = 0.77-<br />

[327] Patients <strong>with</strong> metastatic<br />

gastric adenocarc<strong>in</strong>oma<br />

[328] Patients <strong>with</strong> advanced<br />

gastric cancer<br />

[329] Septemb<br />

er 2003<br />

Patients <strong>with</strong> malignant<br />

symptomatic<br />

gastroduodenal<br />

obstruction<br />

LV5FU2-ir<strong>in</strong>otecan vs. LV5FU2<br />

alone vs. LV5FU2-cisplat<strong>in</strong><br />

Docetaxel 75 mg/m² and<br />

cisplat<strong>in</strong> 75 mg/m² (day 1) plus<br />

fluorouracil 750 mg/m²/d (days<br />

1 to 5) every 3 weeks (n =<br />

221) vs. cisplat<strong>in</strong> 100 mg/m²<br />

(day 1) plus fluorouracil 1,000<br />

mg/m²/d (days 1 to 5) every 4<br />

weeks (n = 224)<br />

14.24, P = 0.1).<br />

Patients <strong>with</strong> a stable or improved global<br />

health ranged from 11% <strong>in</strong> the LV5FU2cisplat<strong>in</strong><br />

arm to 18% <strong>in</strong> the LV5FU2ir<strong>in</strong>otecan<br />

arm. The ir<strong>in</strong>otecan-basedtherapy<br />

presented 14 to 15 scores <strong>with</strong><br />

a better QoL.<br />

TTP was longer <strong>with</strong> DCFvs. CF (32%<br />

risk reduction; log-rank P < .001).<br />

Overall survival was longer <strong>with</strong> DCF<br />

versus CF<br />

(23% risk reduction; log-rank P = .02).<br />

Two-year survival rate was 18% <strong>with</strong><br />

DCF and 9% <strong>with</strong> CF. Overall response<br />

rate was higher <strong>with</strong> DCF (P = .01).<br />

Grade 3 to 4 treatment-related adverse<br />

events occurred <strong>in</strong> 69% (DCF) v 59%<br />

(CF) of patients. Frequent grade 3 to 4<br />

toxicities for DCF vs. CF were:<br />

neutropenia (82% vs. 57%), stomatitis<br />

(21% vs. 27%), diarrhea (19% vs. 8%),<br />

lethargy (19% vs. 14%). Complicated<br />

neutropenia was more frequent <strong>with</strong><br />

DCF than CF (29% vs. 12%).<br />

Self-expand<strong>in</strong>g metal stents Cl<strong>in</strong>ical success was achieved <strong>in</strong> 526<br />

patients <strong>in</strong> the group <strong>in</strong> which technical<br />

success was reported (89 %; 87 % of the<br />

entire group undergo<strong>in</strong>g stent<strong>in</strong>g).<br />

Disease-related factors accounted for<br />

the majority of cl<strong>in</strong>ical failures. Oral<br />

No <strong>in</strong>formation on<br />

randomization procedure<br />

or bl<strong>in</strong>d<strong>in</strong>g<br />

No <strong>in</strong>formation on exact<br />

randomization procedure<br />

32 case series <strong>in</strong>cluded<br />

(only 10 prospective)<br />

No <strong>in</strong>formation on quality<br />

appraisal or selection<br />

process (two reviewers?)<br />

RCT Moderate<br />

RCT Moderate<br />

SR Very low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 157<br />

Study ID Ref Search<br />

date<br />

Hosono S 2007 [178] Septemb<br />

er 2006<br />

Jeurn<strong>in</strong>k S 2007 [179] Decemb<br />

er 2005<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

<strong>in</strong>take became possible <strong>in</strong> all of the<br />

patients <strong>in</strong> whom a successful procedure<br />

was carried out, <strong>with</strong> 87 % tak<strong>in</strong>g soft<br />

solids or a full diet, <strong>with</strong> f<strong>in</strong>al resolution<br />

of symptoms occurr<strong>in</strong>g after a mean of 4<br />

days. There was no procedure-related<br />

mortality. Severe complications<br />

(bleed<strong>in</strong>g and perforation) were<br />

observed <strong>in</strong> seven patients (1.2 %). Stent<br />

migration was reported <strong>in</strong> 31 patients (5<br />

%). Stent obstruction occurred <strong>in</strong> 104<br />

cases (18 %), ma<strong>in</strong>ly due to tumor<br />

<strong>in</strong>filtration. The mean survival period<br />

Patients <strong>with</strong> malignant<br />

gastroduodenal<br />

obstruction<br />

Patients <strong>with</strong> gastric<br />

outlet obstruction<br />

Endoscopic stent<strong>in</strong>g vs.<br />

surgical gastroenterostomy<br />

was 12.1 weeks.<br />

Endoscopic stent<strong>in</strong>g was found to be<br />

associated <strong>with</strong> higher cl<strong>in</strong>ical success (P<br />

= 0.007), a shorter time from the<br />

procedure to start<strong>in</strong>g oral <strong>in</strong>take (P <<br />

0.001), less morbidity (P = 0.02), lower<br />

<strong>in</strong>cidence of delayed gastric empty<strong>in</strong>g (P<br />

= 0.002), and a shorter hospital stay<br />

(P < 0.001) than surgical gastroenterostomy.<br />

There was no significant<br />

difference between the two groups <strong>in</strong><br />

the analysis of 30-day mortality.<br />

Stent vs. gastrojejunostomy No differences between stent placement<br />

and gastrojejunostomy were found <strong>in</strong><br />

technical success (96% vs. 100%), early<br />

and late major complications 7% vs. 6%<br />

and 18% vs. 17%, respectively) and<br />

persist<strong>in</strong>g symptoms (8% vs. 9%). Initial<br />

cl<strong>in</strong>ical success was higher after stent<br />

placement (89% vs. 72%). M<strong>in</strong>or<br />

complications were less frequently seen<br />

after stent placement <strong>in</strong> the patient<br />

series (9% vs. 33%), however the pooled<br />

analysis showed no differences (OR:<br />

0.75, p = 0.8). Recurrent obstructive<br />

symptoms were more common after<br />

stent placement (18% vs. 1%). Hospital<br />

<strong>English</strong> only<br />

1 RCT and 8 nonrandomized<br />

retrospective<br />

studies <strong>in</strong>cluded<br />

Medl<strong>in</strong>e only<br />

No <strong>in</strong>formation on study<br />

selection<br />

Inclusion of 44 studies (2<br />

RCTs)<br />

SR Low<br />

SR Low


158 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

stay was prolonged after GJJ compared<br />

to stent placement (13 days vs. 7 days).<br />

The mean survival was 105 days after<br />

stent placement and 164 days after GJJ.<br />

Fiori E 2004 [330] Included <strong>in</strong> Jeurn<strong>in</strong>k S RCT<br />

Mehta S 2006 [331] Included <strong>in</strong> Jeurn<strong>in</strong>k S RCT<br />

Wenger U 2006 [271] Patients <strong>with</strong> <strong>in</strong>curable<br />

cancer of the distal<br />

oesophagus or gastric<br />

cardia (n = 41)<br />

Follow up.<br />

No evidence identified.<br />

Antireflux stent (n = 19) vs.<br />

standard stent (n = 22)<br />

Fewer patients <strong>with</strong> complications were<br />

observed <strong>in</strong> the antireflux stent group (n<br />

= 3) than <strong>in</strong> the standard group (n = 8),<br />

but no statistically significant difference<br />

was shown (p = 0.14). The survival rates<br />

were similar <strong>in</strong> the two groups (p =<br />

0.99; hazard ratio, 1.0; 95% confidence<br />

<strong>in</strong>terval, 0.5-2.0). The groups did not<br />

differ significantly <strong>in</strong> terms of studied<br />

oesophageal or respiratory symptoms<br />

or quality of life. Cl<strong>in</strong>ically relevant<br />

improvement <strong>in</strong> dysphagia occurred <strong>in</strong><br />

both groups. Dyspnea decreased after<br />

antireflux stent <strong>in</strong>sertion (mean score<br />

change, -11), and <strong>in</strong>creased after<br />

<strong>in</strong>sertion of standard stent (mean score<br />

change, +21).<br />

Interim report<br />

Central randomization,<br />

but no <strong>in</strong>formation on<br />

procedure<br />

Patients were bl<strong>in</strong>ded, but<br />

no <strong>in</strong>formation on<br />

bl<strong>in</strong>d<strong>in</strong>g of cl<strong>in</strong>icians<br />

RCT Moderate


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 159<br />

APPENDIX 6<br />

EVIDENCE TABLES OF GASTRIC LYMPHOMA BY CLINICAL QUESTION<br />

Study ID<br />

Molecular diagnosis<br />

Ref Search<br />

date<br />

Diagnosis and stag<strong>in</strong>g of gastric lymphoma.<br />

Weston 1998 [332] Mix of 50 patients of which<br />

15 had gastric lymphoma<br />

Endoscopy<br />

Palazzo 1993 [333] Patients <strong>with</strong> primary<br />

gastric lymphoma (n = 50)<br />

Ultrasonography<br />

Hoepffner 2003 [203] Patients <strong>with</strong> gastric NHL<br />

(n = 44)<br />

CT scan<br />

Ferreri 1998 [334] Patients <strong>with</strong> gastric MALT<br />

lymphoma (n = 20)<br />

Low-grade: n = 11; high-<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

grade: n = 9<br />

Vorbeck 2002 [202] Patients <strong>with</strong> gastric MALT<br />

lymphoma (n = 14)<br />

Hoepffner 2003 [203] Patients <strong>with</strong> gastric NHL<br />

(n = 44)<br />

Endoscopic ultrasonography<br />

Caletti 1993 [199] Patients <strong>with</strong> endoscopic<br />

gross appearance<br />

suspicious for gastric<br />

cancer of lymphoma<br />

PCR monoclonality<br />

Standard: histology<br />

(endoscopy)<br />

EUS vs. endoscopy<br />

Standard: histology (surgery) (n<br />

= 24)<br />

EUS (n = 44) vs. CT (n = 24)<br />

vs. US (n = 34)<br />

Standard: histology (surgery)<br />

CT for detection of perigastric<br />

adenopathy<br />

Standard: histology (surgery)<br />

EUS vs. CT<br />

Standard: histology<br />

(endoscopy)<br />

EUS (n = 44) vs. CT (n = 24)<br />

vs. US (n = 34)<br />

Standard: histology (surgery)<br />

EUS<br />

Standard: histology (surgery) (n<br />

= 86)<br />

Sensitivity 73%, specificity 46%<br />

However, correct <strong>in</strong>terpretation of these<br />

values is impossible<br />

Endoscopy correctly assessed tumour<br />

extent <strong>in</strong> 25% of cases<br />

Sensitivity for diagnosis: 27% (specificity<br />

unknown)<br />

Sensitivity to dist<strong>in</strong>guish II1 from I: 27%<br />

Specificity 100%<br />

Sensitivity for diagnosis: 57% (specificity<br />

unknown)<br />

Sensitivity for diagnosis: 42% (specificity<br />

unknown)<br />

42 gastric cancers, 44 primary<br />

lymphomas, 142 patients <strong>with</strong> benign<br />

gastric lesions or suspected pancreatic<br />

diseases.<br />

Sensitivity (lymphoma): 89%<br />

Specificity: 97%<br />

PPV: 87%; NPV: 97%<br />

Selection of patients?? Prospective<br />

study<br />

Decision for surgery not<br />

based on EUS<br />

Selection of patients??<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

CT not performed <strong>in</strong> all<br />

patients<br />

Selection of patients??<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

No <strong>in</strong>formation on how<br />

patients were selected<br />

Small study sample<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

Selection of patients??<br />

Small study sample<br />

CT not performed <strong>in</strong> all<br />

patients<br />

Selection of patients??<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

No <strong>in</strong>formation on bl<strong>in</strong>d<strong>in</strong>g<br />

dur<strong>in</strong>g surgery<br />

Standard not applied to all<br />

patients, no <strong>in</strong>formation on<br />

follow-up of 142 patients<br />

<strong>with</strong>out gastric malignancy<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Retrospective<br />

(?) study<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Very low<br />

Very low<br />

Very low<br />

Very low<br />

Very low<br />

Very low<br />

Low


160 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search Population Intervention Results Comments Study type Level of<br />

date<br />

evidence<br />

Palazzo 1993 [333] Patients <strong>with</strong> primary EUS vs. endoscopy<br />

Diagnosis of LN metastasis: sensitivity Decision for surgery not Prospective Very low<br />

gastric lymphoma (n = 50) Standard: histology (surgery) (n 100%, specificity 80%<br />

based on EUS<br />

study<br />

= 24)<br />

Selection of patients??<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

Fischbach 2002 [200] Patients <strong>with</strong> primary EUS<br />

Sensitivity to dist<strong>in</strong>guish II1 from I: 59% 10 patients excluded from Prospective Low<br />

gastric B-cell lymphoma Standard: histology (surgery) Specificity 71%<br />

statistical analysis, study<br />

stage I and II (n = 80)<br />

because EUS was<br />

considered <strong>in</strong>appropriate<br />

for evaluation of nonregional<br />

lymph nodes<br />

Fusaroli 2002 [335] Patients <strong>with</strong> gastric MALT EUS<br />

Inter-observer agreement T-stage: kappa<br />

Prospective Very low<br />

lymphoma (n = 54; n = 42, Bl<strong>in</strong>ded assessment of 0.38 before and 0.37 after treatment<br />

study<br />

six months after treatment) photographs by 9 other Inter-observer agreement N-stage: kappa<br />

endosonographers<br />

0.63 before and 0.34 after treatment<br />

Vorbeck 2002 [202] Patients <strong>with</strong> gastric MALT EUS vs. CT<br />

Sensitivity for diagnosis: 93% (specificity Selection of patients?? Prospective Very low<br />

lymphoma (n = 14) Standard: histology<br />

(endoscopy)<br />

unknown)<br />

Small study sample study<br />

Hoepffner 2003 [203] Patients <strong>with</strong> gastric NHL EUS (n = 44) vs. CT (n = 24) Sensitivity for diagnosis: 95% (specificity CT not performed <strong>in</strong> all Prospective Very low<br />

(n = 44)<br />

vs. US (n = 34)<br />

unknown)<br />

patients<br />

study<br />

PET scan<br />

Standard: histology (surgery)<br />

Selection of patients??<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

Rodriguez 1997<br />

Gallium 67 imag<strong>in</strong>g<br />

[201] Mix of 15 patients, of which<br />

8 had primary gastric NHL<br />

and 7 others served as<br />

control<br />

Kataoka 1990 [336] Patients <strong>with</strong> NHL of the GI<br />

tract (n = 24, of which 20<br />

had gastric NHL), selected<br />

out of 189 consecutive<br />

patients<br />

PET vs. CT or MRI<br />

Standard: endoscopic biopsies<br />

Gallium 67 (n = 24) vs. barium<br />

study (n = 24) vs. CT (n = 16)<br />

Sensitivity 88% Patient selection??<br />

Comparator not applied to<br />

all patients<br />

Sensitivity for diagnosis: 95% (specificity<br />

unknown)<br />

Not only patients <strong>with</strong><br />

gastric NHL<br />

Bl<strong>in</strong>d<strong>in</strong>g?<br />

Prospective<br />

study<br />

Retrospective<br />

study<br />

Very low<br />

Very low


<strong>KCE</strong> <strong>Report</strong>s 75 Upper GI Cancer 161<br />

Treatment of low-grade (Ie and IIE) MALT lymphoma<br />

Study ID Ref Search<br />

date<br />

Helicobacter eradication<br />

Montalban 2001 [208] Patients <strong>with</strong> stage I lowgrade<br />

gastric MALT<br />

lymphoma<br />

Fischbach 2004 [207] Patients <strong>with</strong> low grade<br />

gastric MALT lymphoma<br />

Chemotherapy<br />

Aviles 2005 [209] 241 patients <strong>with</strong> low-grade<br />

MALT lymphoma <strong>in</strong> early<br />

stage (IE and IIE)<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

Eradication treatment 91% achieved disappearance of the<br />

lymphoma<br />

Exclusive H pylori<br />

eradication<br />

surgery (80 cases),<br />

radiotherapy (78 cases), and<br />

chemotherapy (83 cases)<br />

(CHOP)<br />

Treatment of high-grade gastric lymphoma IE and IIE<br />

Complete regression of lymphoma <strong>in</strong> 56<br />

patients (62%), m<strong>in</strong>imal residual disease<br />

<strong>in</strong> 17 patients (18%), partial<br />

remission <strong>in</strong> 11 patients (12%), no<br />

change <strong>in</strong> four patients (4%), and<br />

progressive disease <strong>in</strong> two patients<br />

(2%)<br />

Event-free survival at follow-up (median<br />

follow-up was 7.5 y) :<br />

Surgery : 52 %<br />

Radiotherapy : 52%<br />

Chemotherapy : 87% ( p


162 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Study ID Ref Search<br />

date<br />

Chen LT 2001 [341] patients suffer<strong>in</strong>g from<br />

high-grade gastric<br />

lymphomas (DLBCL)<br />

B<strong>in</strong>n M 2003 [193] patients suffer<strong>in</strong>g from<br />

localized gastric<br />

lymphomas (DLBCL)<br />

Koch P 2005 [342] 393 patients <strong>with</strong><br />

aggressive localized PGL<br />

were treated between<br />

December 1996 and<br />

Population Intervention Results Comments Study type Level of<br />

evidence<br />

(chemotherapy) after 5 years for all<br />

stages and histologies.<br />

December 2003<br />

Ishikura S 2005 [343] 52 patients <strong>with</strong> aggressive<br />

localized PGL<br />

were treated between<br />

December 1999 and<br />

December 2003<br />

Aviles A 2006 [344] 101 patients <strong>with</strong> high<br />

grade lymphoma<br />

were treated between<br />

December 1999 and<br />

December 2003<br />

16 patients received a brief<br />

antibiotic therapy as first-l<strong>in</strong>e<br />

treatment<br />

58 patients underwent<br />

chemotherapy<br />

48 patients underwent surgery<br />

+chemotherapy<br />

332 patients underwent<br />

“conservative treatment”<br />

(radiotherapy +/-chemotherapy)<br />

61 patients received surgery<br />

and conservative treatment.<br />

Chemotherapy (CHOP) and<br />

radiotherapy (40.5Gy)<br />

52 patients underwent surgery<br />

+chemotherapy (CEOP-Bleo)<br />

49 patients underwent<br />

chemotherapy alone<br />

Total : 101<br />

Eradication of H pylori: 15 patients<br />

Rapid gross tumor regression: 10<br />

patients<br />

Complete remission rate: 62.5% (CI<br />

35.8% - 89.1%)<br />

Median duration of complete response<br />

was 31.2 months (range, 14.4 to 49.1<br />

months)<br />

Overall survival at follow-up (59 months<br />

(range 3–128) :<br />

Best prognosis : 90.5%(chemotherapy<br />

alone) versus 91.1% (surgery<br />

+chemotherapy) (P = 0.303)<br />

Mild Prognosis : 85.9%(chemotherapy<br />

alone) versus 91.6% (surgery<br />

+chemotherapy) (P = 0.187)<br />

Overall survival at 42 months :<br />

For surgery was 86% versus<br />

91.0% for patients <strong>with</strong>out surgery<br />

Complete remission rate: 48 on 52 (CI<br />

82-98%%)<br />

Overall survival at follow-up (median<br />

follow-up was 2y) : 94%<br />

Overall survival at follow-up (median<br />

follow-up was 5 y) :<br />

Surgery + chemotherapy: 78% (CI 70-<br />

88%)<br />

Chemotherapy: 76% (CI 70-87%)<br />

P=0.8<br />

a similar 5-year survival<br />

rate<br />

(>90%) is to be expected<br />

<strong>with</strong> either surgery plus<br />

chemotherapy or<br />

chemotherapy alone<br />

Treatment decision was<br />

left to each participat<strong>in</strong>g<br />

centre<br />

Prospective<br />

study<br />

Prospective<br />

comparative<br />

study<br />

Prospective<br />

study<br />

Prospective<br />

study<br />

Low<br />

Low<br />

Low<br />

Low<br />

RCT High


<strong>KCE</strong> <strong>reports</strong> 75 Upper GI Cancer 163<br />

APPENDIX 7<br />

TNM CLASSIFICATION<br />

Oesophageal cancer<br />

(source: Digestive System Tumours. In: Sob<strong>in</strong> LH, Wittek<strong>in</strong>g Ch, editors. TNM<br />

Classification of Malignant Tumours. 6 th ed. New York: Wiley-Liss; 2002. p. 57-96.)<br />

PRIMARY TUMOUR (T)<br />

TX Primary tumour cannot be assessed<br />

T0 No evidence of primary tumour<br />

Tis Carc<strong>in</strong>oma <strong>in</strong> situ<br />

T1 Tumour <strong>in</strong>vades lam<strong>in</strong>a propria or submucosa<br />

T2 Tumour <strong>in</strong>vades muscularis propria<br />

T3 Tumour <strong>in</strong>vades adventitia<br />

T4 Tumour <strong>in</strong>vades adjacent structures<br />

REGIONAL LYMPH NODES (N)<br />

NX Regional lymph nodes cannot be assessed<br />

N0 No regional lymph node metastasis<br />

N1 Regional lymph node metastasis<br />

DISTANT METASTASIS (M)<br />

MX Distant metastasis cannot be assessed<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

Tumours of the lower thoracic oesophagus:<br />

M1a Metastasis <strong>in</strong> celiac lymph nodes<br />

M1b Other distant metastasis<br />

Tumours of the midthoracic oesophagus:<br />

M1a Not applicable<br />

M1b Nonregional lymph nodes and/or other distant metastasis<br />

Tumours of the upper thoracic oesophagus:<br />

M1a Metastasis <strong>in</strong> cervical nodes<br />

M1b Other distant metastasis<br />

STAGE GROUPING<br />

Stage 0 Tis N0 M0<br />

Stage I T1 N0 M0<br />

Stage IIA T2 N0 M0<br />

T3 N0 M0<br />

Stage IIB T1 N1 M0<br />

T2 N1 M0<br />

Stage III T3 N1 M0<br />

T4 Any N M0<br />

Stage IV Any T Any N M1<br />

Stage IVA Any T Any N M1a<br />

Stage IVB Any T Any N M1b


164 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Gastric cancer<br />

PRIMARY TUMOUR (T)<br />

TX Primary tumour cannot be assessed<br />

T0 No evidence of primary tumour<br />

Tis Carc<strong>in</strong>oma <strong>in</strong> situ : <strong>in</strong>traepithelial tumour <strong>with</strong>out <strong>in</strong>vasion of the lam<strong>in</strong>a propria<br />

T1 Tumour <strong>in</strong>vades lam<strong>in</strong>a propria or submucosa<br />

T2 Tumour <strong>in</strong>vades muscularis propria or subserosa<br />

T2a Tumour <strong>in</strong>vades muscularis propria<br />

T2b Tumour <strong>in</strong>vades subserosa<br />

T3 Tumour penetrates serosa (visceral peritoneum) <strong>with</strong>out <strong>in</strong>vasion of adjacent<br />

structures<br />

T4 Tumour <strong>in</strong>vades adjacent structures<br />

REGIONAL LYMPH NODES (N)<br />

NX Regional lymph nodes cannot be assessed<br />

N0 No regional lymph node metastasis<br />

N1 Metastasis <strong>in</strong> 1 to 6 regional lymph nodes<br />

N2 Metastasis <strong>in</strong> 7 to 15 regional lymph nodes<br />

N3 Metastasis <strong>in</strong> more than 15 regional lymph nodes<br />

DISTANT METASTASIS (M)<br />

MX Distant metastasis cannot be assessed<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

STAGE GROUPING<br />

Stage 0 Tis N0 M0<br />

Stage IA T1 N0 M0<br />

Stage IB T1 N1 M0<br />

T2a/b N0 M0<br />

Stage II T1 N2 M0<br />

T2a/b N1 M0<br />

T3 N0 M0<br />

Stage IIIA T2a/b N2 M0<br />

T3 N1 M0<br />

T4 N0 M0<br />

Stage IIIB T3 N2 M0<br />

Stage IV T4 N1-3 M0<br />

T1-3 N3 M0<br />

Any T Any N M1


<strong>KCE</strong> <strong>reports</strong> 75 Upper GI Cancer 165<br />

APPENDIX 8<br />

USED MESH AND SEARCH TERMS FOR ADDITIONAL EVIDENCE<br />

SEARCH IN MEDLINE (OVID)<br />

Oeophageal cancer:<br />

1 exp esophageal neoplasms/<br />

2 (esophag$ adj5 neoplas$).tw.<br />

3 (oesophag$ adj5 neoplas$).tw.<br />

4 (esophag$ adj5 cancer$).tw.<br />

5 (oesophag$ adj5 cancer$).tw.<br />

6 (esophag$ adj5 carc<strong>in</strong>$).tw.<br />

7 (oesophag$ adj5 carc<strong>in</strong>$).tw.<br />

8 (esophag$ adj5 tumo$).tw.<br />

9 (oesophag$ adj5 tumo$).tw.<br />

10 (esophag$ adj5 metasta$).tw.<br />

11 (oesophag$ adj5 metasta$).tw.<br />

12 (esophag$ adj5 malig$).tw.<br />

13 (oesophag$ adj5 malig$).tw.<br />

14 or/1-13<br />

Gastric cancer:<br />

1 exp stomach neoplasms/<br />

2 (stomach adj5 neoplas$).tw.<br />

3 (stomach adj5 cancer$).tw.<br />

4 (stomach adj5 carc<strong>in</strong>$).tw.<br />

5 (stomach adj5 tumo$).tw.<br />

6 (stomach adj5 metasta$).tw.<br />

7 (stomach adj5 malig$).tw.<br />

8 (gastric adj5 neoplas$).tw.<br />

9 (gastric adj5 cancer$).tw.<br />

10 (gastric adj5 carc<strong>in</strong>$).tw.<br />

11 (gastric adj5 tumo$).tw.<br />

12 (gastric adj5 metasta$).tw.<br />

13 (gastric adj5 malig$).tw.<br />

14 or/1-13


166 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Systematic reviews / meta-analyses:<br />

1 meta-analysis.pt,ti,ab,sh.<br />

2 1 or (meta anal$ or metaanal$).ti,ab,sh.<br />

3 (methodol$ or systematic$ or quantitativ$).ti,ab,sh.<br />

((methodol$ or systematic$ or quantitativ$) adj (review$ or overview$<br />

4<br />

or survey$)).ti,ab,sh.<br />

5 (medl<strong>in</strong>e or embase or <strong>in</strong>dex medicus).ti,ab.<br />

((pool$ or comb<strong>in</strong>ed or comb<strong>in</strong><strong>in</strong>g) adj (data or trials or studies or<br />

6<br />

results)).ti,ab.<br />

7 3 or 4 or 5 or 6<br />

8 7 and review.pt,sh.<br />

9 2 or 8<br />

Randomized controlled trials:<br />

1 Randomized controlled trials/<br />

2 Randomized controlled trial.pt.<br />

3 Random allocation/<br />

4 Double bl<strong>in</strong>d method/<br />

5 S<strong>in</strong>gle bl<strong>in</strong>d method/<br />

6 Cl<strong>in</strong>ical trial.pt.<br />

7 exp cl<strong>in</strong>ical trials/<br />

8 or/1-7<br />

9 (cl<strong>in</strong>ic$ adj trial$1).tw.<br />

10 ((s<strong>in</strong>gl$ or doubl$ or treb$ or tripl$) adj (bl<strong>in</strong>d$3 or mask$3)).tw.<br />

11 Placebos/<br />

12 Placebo$.tw.<br />

13 Randomly allocated.tw.<br />

14 (allocated adj2 random).tw.<br />

15 or/9-14<br />

16 8 or 15<br />

17 Case report.tw.<br />

18 Letter.pt.<br />

19 Historical article.pt.<br />

20 Review of reported cases.pt.<br />

21 Review, multicase.pt.<br />

22 or/17-21<br />

23 16 not 22


<strong>KCE</strong> <strong>reports</strong> 75 Upper GI Cancer 167<br />

Chemotherapy:<br />

1 exp drug therapy/<br />

2 exp chemotherapy adjuvant/<br />

3 exp drug therapy comb<strong>in</strong>ation/<br />

4 exp ant<strong>in</strong>eoplastic agents comb<strong>in</strong>ed/<br />

5 chemothera$.tw.<br />

6 exp chemotherapy/<br />

7 or/1-6<br />

Endoscopy:<br />

1 endoscopy/ or endoscopy, digestive system/ or endoscopy, gastro<strong>in</strong>test<strong>in</strong>al/<br />

2 Gastroscopy/<br />

3 Esophagoscopy/<br />

4 or/1-3<br />

Stag<strong>in</strong>g (e.g. gastric cancer):<br />

1 exp stomach neoplasms/<br />

2 (stomach adj5 neoplas$).tw.<br />

3 (stomach adj5 cancer$).tw.<br />

4 (stomach adj5 carc<strong>in</strong>$).tw.<br />

5 (stomach adj5 tumo$).tw.<br />

6 (stomach adj5 metasta$).tw.<br />

7 (stomach adj5 malig$).tw.<br />

8 (gastric adj5 neoplas$).tw.<br />

9 (gastric adj5 cancer$).tw.<br />

10 (gastric adj5 carc<strong>in</strong>$).tw.<br />

11 (gastric adj5 tumo$).tw.<br />

12 (gastric adj5 metasta$).tw.<br />

13 (gastric adj5 malig$).tw.<br />

14 or/1-13<br />

tomography scanners, x-ray computed/ or tomography, x-ray computed/<br />

15 or magnetic resonance imag<strong>in</strong>g/ or positron-emission tomography/ or<br />

tomography, spiral computed/ or tomography/<br />

16 Endosonography/<br />

17 Laparoscopy/<br />

18 Bronchoscopy/<br />

19 Thoracoscopy/<br />

20 Ultrasonography/<br />

21 15 or 16 or 17 or 18 or 19 or 20<br />

22 Neoplasm Stag<strong>in</strong>g/<br />

23 "Tumor Markers, Biological"/<br />

24 "Immunoenzyme Techniques"/<br />

25 23 or 24<br />

26 Stomach Neoplasms/an, pa, di, ra, ri, us [Analysis, Pathology, Diagnosis,


168 Upper GI Cancer <strong>KCE</strong> <strong>reports</strong> 75<br />

Radiography, Radionuclide Imag<strong>in</strong>g, Ultrasonography]<br />

27 21 or 22 or 25<br />

28 14 and 27<br />

29 26 or 28<br />

30 Prospective Studies/<br />

31 29 and 30<br />

Recurrence (e.g. oesophageal cancer):<br />

1 Salvage Therapy/<br />

2 neoplasm recurrence, local/ or recurrence/<br />

3 exp esophageal neoplasms/<br />

4 (esophag$ adj5 neoplas$).tw.<br />

5 (oesophag$ adj5 neoplas$).tw.<br />

6 (esophag$ adj5 cancer$).tw.<br />

7 (oesophag$ adj5 cancer$).tw.<br />

8 (esophag$ adj5 carc<strong>in</strong>$).tw.<br />

9 (oesophag$ adj5 carc<strong>in</strong>$).tw.<br />

10 (esophag$ adj5 tumo$).tw.<br />

11 (oesophag$ adj5 tumo$).tw.<br />

12 (esophag$ adj5 metasta$).tw.<br />

13 (oesophag$ adj5 metasta$).tw.<br />

14 (esophag$ adj5 malig$).tw.<br />

15 (oesophag$ adj5 malig$).tw.<br />

16 or/3-15<br />

17 1 or 2<br />

18 16 and 17


This page is left <strong>in</strong>tentionally blank.


Wettelijk depot : D/2008/10.273/16


<strong>KCE</strong> <strong>reports</strong><br />

1. Effectiviteit en kosten-effectiviteit van behandel<strong>in</strong>gen voor rookstop. D/2004/10.273/1.<br />

2. Studie naar de mogelijke kosten van een eventuele wijzig<strong>in</strong>g van de rechtsregels <strong>in</strong>zake<br />

medische aansprakelijkheid (fase 1). D/2004/10.273/2.<br />

3. Antibioticagebruik <strong>in</strong> ziekenhuizen bij acute pyelonefritis. D/2004/10.273/5.<br />

4. Leukoreductie. Een mogelijke maatregel <strong>in</strong> het kader van een nationaal beleid voor<br />

bloedtransfusieveiligheid. D/2004/10.273/7.<br />

5. Het preoperatief onderzoek. D/2004/10.273/9.<br />

6. Validatie van het rapport van de Onderzoekscommissie over de onderf<strong>in</strong>ancier<strong>in</strong>g van de<br />

ziekenhuizen. D/2004/10.273/11.<br />

7. Nationale richtlijn prenatale zorg. Een basis voor een kl<strong>in</strong>isch pad voor de opvolg<strong>in</strong>g van<br />

zwangerschappen. D/2004/10.273/13.<br />

8. F<strong>in</strong>ancier<strong>in</strong>gssystemen van ziekenhuisgeneesmiddelen: een beschrijvende studie van een<br />

aantal Europese landen en Canada. D/2004/10.273/15.<br />

9. Feedback: onderzoek naar de impact en barrières bij implementatie – Onderzoeksrapport:<br />

deel 1. D/2005/10.273/01.<br />

10. De kost van tandprothesen. D/2005/10.273/03.<br />

11. Borstkankerscreen<strong>in</strong>g. D/2005/10.273/05.<br />

12. Studie naar een alternatieve f<strong>in</strong>ancier<strong>in</strong>g van bloed en labiele bloedderivaten <strong>in</strong> de<br />

ziekenhuizen. D/2005/10.273/07.<br />

13. Endovasculaire behandel<strong>in</strong>g van Carotisstenose. D/2005/10.273/09.<br />

14. Variaties <strong>in</strong> de ziekenhuispraktijk bij acuut myocard<strong>in</strong>farct <strong>in</strong> België. D/2005/10.273/11.<br />

15. Evolutie van de uitgaven voor gezondheidszorg. D/2005/10.273/13.<br />

16. Studie naar de mogelijke kosten van een eventuele wijzig<strong>in</strong>g van de rechtsregels <strong>in</strong>zake<br />

medische aansprakelijkheid. Fase II : ontwikkel<strong>in</strong>g van een actuarieel model en eerste<br />

schatt<strong>in</strong>gen. D/2005/10.273/15.<br />

17. Evaluatie van de referentiebedragen. D/2005/10.273/17.<br />

18. Prospectief bepalen van de honoraria van ziekenhuisartsen op basis van kl<strong>in</strong>ische paden en<br />

guidel<strong>in</strong>es: makkelijker gezegd dan gedaan.. D/2005/10.273/19.<br />

19. Evaluatie van forfaitaire persoonlijk bijdrage op het gebruik van spoedgevallendienst.<br />

D/2005/10.273/21.<br />

20. HTA Moleculaire Diagnostiek <strong>in</strong> België. D/2005/10.273/23, D/2005/10.273/25.<br />

21. HTA Stomamateriaal <strong>in</strong> België. D/2005/10.273/27.<br />

22. HTA Positronen Emissie Tomografie <strong>in</strong> België. D/2005/10.273/29.<br />

23. HTA De electieve endovasculaire behandel<strong>in</strong>g van het abdom<strong>in</strong>ale aorta aneurysma (AAA).<br />

D/2005/10.273/32.<br />

24. Het gebruik van natriuretische peptides <strong>in</strong> de diagnostische aanpak van patiënten met<br />

vermoeden van hartfalen. D/2005/10.273/34.<br />

25. Capsule endoscopie. D/2006/10.273/01.<br />

26. Medico–legale aspecten van kl<strong>in</strong>ische praktijkrichtlijnen. D2006/10.273/05.<br />

27. De kwaliteit en de organisatie van type 2 diabeteszorg. D2006/10.273/07.<br />

28. Voorlopige richtlijnen voor farmaco-economisch onderzoek <strong>in</strong> België. D2006/10.273/10.<br />

29. Nationale Richtlijnen College voor Oncologie: A. algemeen kader oncologisch<br />

kwaliteitshandboek B. wetenschappelijke basis voor kl<strong>in</strong>ische paden voor diagnose en<br />

behandel<strong>in</strong>g colorectale kanker en testiskanker. D2006/10.273/12.<br />

30. Inventaris van databanken gezondheidszorg. D2006/10.273/14.<br />

31. Health Technology Assessment prostate-specific-antigen (PSA) voor<br />

prostaatkankerscreen<strong>in</strong>g. D2006/10.273/17.<br />

32. Feedback : onderzoek naar de impact en barrières bij implementatie – Onderzoeksrapport :<br />

deel II. D/2006/10.273/19.<br />

33. Effecten en kosten van de vacc<strong>in</strong>atie van Belgische k<strong>in</strong>deren met geconjugeerd<br />

pneumokokkenvacc<strong>in</strong>. D/2006/10.273/21.<br />

34. Trastuzumab bij vroegtijdige stadia van borstkanker. D/2006/10.273/23.<br />

35. Studie naar de mogelijke kosten van een eventuele wijzig<strong>in</strong>g van de rechtsregels <strong>in</strong>zake<br />

medische aansprakelijkheid (fase III)- preciser<strong>in</strong>g van de kostenram<strong>in</strong>g. D/2006/10.273/26.<br />

36. Farmacologische en chirurgische behandel<strong>in</strong>g van obesitas. Residentiële zorg voor ernstig<br />

obese k<strong>in</strong>deren <strong>in</strong> België. D/2006/10.273/28.<br />

37. HTA Magnetische Resonantie Beeldvorm<strong>in</strong>g. D/2006/10.273/32.


38. Baarmoederhalskankerscreen<strong>in</strong>g en testen op Human Papillomavirus (HPV).<br />

D/2006/10.273/35<br />

39. Rapid assessment van nieuwe wervelzuil technologieën : totale discusprothese en<br />

vertebro/ballon kyfoplastie. D/2006/10.273/38.<br />

40. Functioneel bilan van de patiënt als mogelijke basis voor nomenclatuur van k<strong>in</strong>esitherapie <strong>in</strong><br />

België? D/2006/10.273/40.<br />

41. Kl<strong>in</strong>ische kwaliteits<strong>in</strong>dicatoren. D/2006/10.273/43.<br />

42. Studie naar praktijkverschillen bij electieve chirurgische <strong>in</strong>grepen <strong>in</strong> België. D/2006/10.273/45.<br />

43. Herzien<strong>in</strong>g bestaande praktijkrichtlijnen. D/2006/10.273/48.<br />

44. Een procedure voor de beoordel<strong>in</strong>g van nieuwe medische hulpmiddelen. D/2006/10.273/50.<br />

45. HTA Colorectale Kankerscreen<strong>in</strong>g: wetenschappelijke stand van zaken en budgetimpact<br />

voor België. D/2006/10.273/53.<br />

46. Health Technology Assessment. Polysomnografie en thuismonitor<strong>in</strong>g van zuigel<strong>in</strong>gen voor de<br />

preventie van wiegendood. D/2006/10.273/59.<br />

47. Geneesmiddelengebruik <strong>in</strong> de belgische rusthuizen en rust- en verzorg<strong>in</strong>gstehuizen.<br />

D/2006/10.273/61<br />

48. Chronische lage rugpijn. D/2006/10.273/63.<br />

49. Antivirale middelen bij seizoensgriep en grieppandemie. Literatuurstudie en ontwikkel<strong>in</strong>g van<br />

praktijkrichtlijnen. D/2006/10.273/65.<br />

50. Eigen betal<strong>in</strong>gen <strong>in</strong> de Belgische gezondheidszorg. De impact van supplementen.<br />

D/2006/10.273/68.<br />

51. Chronische zorgbehoeften bij personen met een niet- aangeboren hersenletsel (NAH)<br />

tussen 18 en 65 jaar. D/2007/10.273/01.<br />

52. Rapid Assessment: Cardiovasculaire Primaire Preventie <strong>in</strong> de Belgische Huisartspraktijk.<br />

D/2007/10.273/03.<br />

53. F<strong>in</strong>ancier<strong>in</strong>g van verpleegkundige zorg <strong>in</strong> ziekenhuizen. D/2007/10 273/06<br />

54. Kosten-effectiviteitsanalyse van rotavirus vacc<strong>in</strong>atie van zuigel<strong>in</strong>gen <strong>in</strong> België<br />

55. Evidence-based <strong>in</strong>houd van geschreven <strong>in</strong>formatie vanuit de farmaceutische <strong>in</strong>dustrie aan<br />

huisartsen. D/2007/10.273/12.<br />

56. Orthopedisch Materiaal <strong>in</strong> België: Health Technology Assessment. D/2007/10.273/14.<br />

57. Organisatie en F<strong>in</strong>ancier<strong>in</strong>g van Musculoskeletale en Neurologische Revalidatie <strong>in</strong> België.<br />

D/2007/10.273/18.<br />

58. De Implanteerbare Defibrillator: een Health Technology Assessment. D/2007/10.273/21.<br />

59. Laboratoriumtesten <strong>in</strong> de huisartsgeneeskunde. D2007/10.273/24.<br />

60. Longfunctie testen bij volwassenen. D/2007/10.273/27.<br />

61. Vacuümgeassisteerde Wondbehandel<strong>in</strong>g: een Rapid Assessment. D/2007/10.273/30<br />

62. Intensiteitsgemoduleerde Radiotherapie (IMRT). D/2007/10.273/32.<br />

63. Wetenschappelijke ondersteun<strong>in</strong>g van het College voor Oncologie: een nationale<br />

praktijkrichtlijn voor de aanpak van borstkanker. D/2007/10.273/35.<br />

64. HPV Vacc<strong>in</strong>atie ter Preventie van Baarmoederhalskanker <strong>in</strong> België: Health Technology<br />

Assessment. D/2007/10.273/41.<br />

65. Organisatie en f<strong>in</strong>ancier<strong>in</strong>g van genetische diagnostiek <strong>in</strong> België. D/2007/10.273/44.<br />

66. Health Technology Assessment: Drug-Elut<strong>in</strong>g Stents <strong>in</strong> België. D/2007/10.273/47.<br />

67. Hadrontherapie. D/2007/10.273/50.<br />

68. Vergoed<strong>in</strong>g van schade als gevolg van gezondheidszorg – Fase IV : Verdeelsleutel tussen het<br />

Fonds en de verzekeraars. D/2007/10.273/52.<br />

69. Kwaliteit van rectale kankerzorg – Fase 1: een praktijkrichtlijn voor rectale kanker<br />

D/2007/10.273/54.<br />

70. Vergelijkende studie van ziekenhuisaccrediter<strong>in</strong>gs-programma ’ s <strong>in</strong> Europa<br />

D/2008/10.273/57.<br />

71. Aanbevel<strong>in</strong>gen voor het gebruik van vijf oftalmologische testen <strong>in</strong> de kl<strong>in</strong>ische<br />

praktijk .D/2008/10.273/04<br />

72. Het aanbod van artsen <strong>in</strong> België. Huidige toestand en toekomstige uitdag<strong>in</strong>gen.<br />

D/2008/10.273/07<br />

73. F<strong>in</strong>ancier<strong>in</strong>g van het zorgprogramma voor de geriatrische patiënt <strong>in</strong> algemene ziekenhuizen:<br />

def<strong>in</strong>itie en evaluatie van een geriatrische patiënt, def<strong>in</strong>itie van de <strong>in</strong>terne liaisongeriatrie en<br />

evaluatie van de middelen voor een goede f<strong>in</strong>ancier<strong>in</strong>g. D/2008/10.273/11<br />

74. Hyperbare Zuurstoftherapie: Rapid Assessment. D/2008/10.273/13.<br />

75. Wetenschappelijke ondersteun<strong>in</strong>g van het College voor Oncologie: een nationale<br />

praktijkrichtlijn voor de aanpak van slokdarm- en maagkanker D/2008/10.273/16

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!