Guideline pour la prise en charge du cancer oesophagien et ...

190
Guideline pour la prise en charge du cancer oesophagien et gastrique : éléments scientifiques à destination du Collège d’Oncologie KCE reports 75B Federaal Kenniscentrum voor de Gezondheidszorg Centre fédéral d’expertise des soins de santé 2008

Transcript of Guideline pour la prise en charge du cancer oesophagien et ...

Page 1: Guideline pour la prise en charge du cancer oesophagien et ...

Guideline pour la prise en charge du cancer oesophagien et gastrique :

éléments scientifiques à destination du Collège d’Oncologie

KCE reports 75B

Federaal Kenniscentrum voor de Gezondheidszorg Centre fédéral d’expertise des soins de santé

2008

Page 2: Guideline pour la prise en charge du cancer oesophagien et ...

Le Centre fédéral d’expertise des soins de santé

Présentation : Le Centre fédéral d’expertise des soins de santé est un parastatal, créé le 24 décembre 2002 par la loi-programme (articles 262 à 266), sous tutelle du Ministre de la Santé publique et des Affaires sociales, qui est chargé de réaliser des études éclairant la décision politique dans le domaine des soins de santé et de l’assurance maladie.

Conseil d’administration

Membres effectifs : Gillet Pierre (Président), Cuypers Dirk (Vice-Président), Avontroodt Yolande, De Cock Jo (Vice-Président), Demeyere Frank, De Ridder Henri, Gillet Jean-Bernard, Godin Jean-Noël, Goyens Floris, Maes Jef, Mertens Pascal, Mertens Raf, Moens Marc, Perl François, Van Massenhove Frank, Vandermeeren Philippe, Verertbruggen Patrick, Vermeyen Karel.

Membres suppléants : Annemans Lieven, Bertels Jan, Collin Benoît, Cuypers Rita, Decoster Christiaan, Dercq Jean-Paul, Désir Daniel, Laasman Jean-Marc, Lemye Roland, Morel Amanda, Palsterman Paul, Ponce Annick, Remacle Anne, Schrooten Renaat, Vanderstappen Anne.

Commissaire du gouvernement : Roger Yves

Direction

Directeur général : Dirk Ramaekers

Directeur général adjoint : Jean-Pierre Closon

Contact

Centre fédéral d’expertise des soins de santé (KCE). Rue de la Loi 62 B-1040 Bruxelles Belgium

Tel: +32 [0]2 287 33 88 Fax: +32 [0]2 287 33 85

Email : [email protected] Web : http://www.kce.fgov.be

Page 3: Guideline pour la prise en charge du cancer oesophagien et ...

Guideline pour la prise en charge du cancer oesophagien

et gastrique : éléments scientifiques à destination du Collège

d’Oncologie

KCE reports 75B

M. PEETERS, T. LERUT, J. VLAYEN, F. MAMBOURG, N. ECTORS, P. DEPREZ, T. BOTERBERG, J. DE MEY,

P. FLAMEN, J.-L. VAN LAETHEM, B. NEYNS, P. PATTYN

Federaal Kenniscentrum voor de Gezondheidszorg

Centre fédéral d’expertise des soins de santé 2008

Page 4: Guideline pour la prise en charge du cancer oesophagien et ...

KCE REPORTS 75B

Titre : Guideline pour la prise en charge du cancer oesophagien et gastrique: éléments scientifiques à destination du Collège d’Oncologie

Auteurs : M. Peeters (UZ Gent; College Oncologie), T. Lerut (UZ Leuven), J. Vlayen (KCE), F. Mambourg (KCE), N. Ectors (UZ Leuven), P. Deprez (UCL), T. Boterberg (UZ Gent), J. De Mey (UZ Brussel), P. Flamen (Institut Jules Bordet), J.-L. Van Laethem (ULB), B. Neyns (UZ Brussel), P. Pattyn (UZ Gent)

Experts externes : Michel Buset (Belgian Society of Gastrointestinal Endoscopy), Wim Ceelen (Belgian Society of Surgical Oncology), Paul Cheyns (Upper GI sectie van het Koninklijk Belgisch Genootschap Heelkunde), Jean-Marie Collard (Belgian Society of Surgical Oncology), Jochen Decaestecker (Vlaamse Vereniging voor Gastro-enterologie), Jacques De Grève (Président Working Party Manual and Guidelines, College Oncologie), Pierre Demetter (Belgian Digestive Pathology Club), Louis Ferrant (Domus Medica), Roland Hustinx (Belgische Genootschap voor Nucleaire Geneeskunde), Anne Jouret-Mourin (Belgian Digestive Pathology Club), Cathy Mahin (Belgische Vereniging voor Radiotherapie-Oncologie), Max Mano (Société Belge d’Oncologie Medicale), Hubert Piessevaux (Société Royale Belge de Gastro-enterologie), Daniel Urbain (Belgian Society of Gastrointestinal Endoscopy), Eric Van Cutsem (Vlaamse Vereniging voor Gastro-enterologie), Bart Van den Eynden (Domus Medica), Didier Verhoeven (Belgische Vereniging voor Medische Oncologie), Joseph Weerts (Upper GI section de l’Association Royale Belge de Chirurgie)

Acknowledgements Liesbet Van Eycken (Stichting Kankerregister), Kris Henau (Stichting Kankerregister)

Validateurs : Harry Bleiberg (Jules Bordet Instituut), Marc De Man (OLV Ziekenhuis Aalst), Hugo W. Tilanus (Erasmus MC Rotterdam)

Conflict of interest : La plupart des auteurs, des experts externes et des validateurs travaillent pour un centre spécialisé dans le traitement des cancers de l'oesophage et de l'estomac. Joseph Weerts a reçu une bourse destinée à couvrir les frais de participation à cours organisé dans le cadre d'un postgraduat (IRCAD Stasbourg). D'autres conflits d'intérêt n'ont pas été mentionnés

Disclaimer: Les experts externes ont collaboré au rapport scientifique qui a ensuite été soumis aux validateurs. La validation du rapport résulte d’un consensus ou d’un vote majoritaire entre les validateurs. Le KCE reste seul responsable des erreurs ou omissions qui pourraient subsister de même que des recommandations faites aux autorités publiques.

Mise en Page : Ine Verhulst

Bruxelles, 21 mars 2008

Etude n° 2007-28-01

Domaine : Good Clinical Practice (GCP)

MeSH : Esophageal Neoplasms ; Stomach Neoplasms ; Gastrointestinal Stromal Tumors ; Lymphoma, B-Cell, Marginal Zone

NLM classification : WI 149

Langage : français, anglais

Format : Adobe® PDF™ (A4)

Dépôt légal : D/2008/10.273/17

La reproduction partielle de ce document est autorisée à condition que la source soit mentionnée. Ce document est disponible en téléchargement sur le site Web du Centre fédéral d’expertise des soins de santé.

Page 5: Guideline pour la prise en charge du cancer oesophagien et ...

Comment citer ce rapport ?

Peeters M, Lerut T, Vlayen J, Mambourg F, Ectors N, Deprez P, et al. Guideline pour la prise en charge du cancer oesophagien et gastrique : éléments scientifiques à destination du Collège d’Oncologie. Bruxelles: Centre fédéral d'expertise des soins de santé (KCE); 2008. KCE Reports 75B (D/2008/10.273/17)

Page 6: Guideline pour la prise en charge du cancer oesophagien et ...
Page 7: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75B Cancer oesophagien et gastrique i

PREFACE Chaque année, environ deux mille belges sont confrontés au diagnostic de cancer de l’oesophage ou de l’estomac. Le pronostic défavorable de ces cancers leur donne la cinquième place dans les causes de mortalité par cancer les plus fréquentes après le cancer du poumon, du colon, du sein et de la prostate. Une importante proportion des patients ne peut espérer guérir et bénéficiera d’emblée d’un traitement palliatif.

Il était donc justifié de présenter des recommandations de bonne pratique à l’intention des praticiens et des patients concernés. Ils trouveront dans ce rapport les meilleures recommandations de bonne pratique internationales mises à jour en fonction de la littérature récente.

Ce rapport est le quatrième à s’inscrire dans une collaboration de plus en plus confiante entre le Collège d’Oncologie et le KCE. De nombreux experts parmi lesquels des représentants de la société scientifique néerlandophone des médecins généralistes ont collaboré à ce rapport. Leur enthousiasme et leur participation furent à nouveau remarquables et ont largement contribué à la bonne qualité du document. Qu’ils en soient remerciés.

La rédaction de ces recommandations constitue la première étape d’un processus long et continu. Leur mise à disposition en ligne via le site web du Collège d’Oncologie contribuera à leur diffusion et à leur mise en pratique. La toute première pierre de la construction d’indicateurs de qualité a également été posée dans ce rapport. Certains projets en cours, comme le projet PROCARE, apporteront incontestablement un éclairage utile pour la stratégie à suivre dans ce domaine

La prise en charge du cancer est actuellement d’actualité puisqu’un plan national de lutte contre le cancer est en cours de développement. Puissent les recommandations présentées ci-dessous, s’intégrer dans les développements ultérieurs de ce plan.

Jean-Pierre Closon Dirk Ramaekers

Directeur general adjoint Directeur général

Page 8: Guideline pour la prise en charge du cancer oesophagien et ...

ii Cancer oesophagien et gastrique KCE Reports 75B

Résumé exécutif

INTRODUCTION Des recommandations décrivant la prise en charge du cancer de l’œsophage et du cancer de l’estomac (y compris le lymphome primaire de l’estomac et les tumeurs stromales gastro-intestinales [GIST]) ont été élaborées dans le cadre de la collaboration entre le Collègue d’Oncologie et le Centre Fédéral d’Expertise des Soins de Santé (KCE). Ces recommandations concernent l’intégralité du trajet de ces patients, du diagnostic au follow-up. Elles s’adressent à l’ensemble des prestataires de soins impliqués dans leur prise en charge.

MÉTHODOLOGIE La méthodologie ADAPTE a été utilisée pour le développer cette directive. Les membres de l’équipe formulent d’abord les questions cliniques principales avec l’aide des experts. Les directives (inter)nationales existantes sont ensuite recherchées dans Medline, Embase, National Guideline Clearinghouse et les sites Web des organisations qui rédigent des directives et sont actives en oncologie. La qualité des 17 directives répertoriées a été évaluée au moyen de l’instrument AGREE par deux évaluateurs travaillant de manière indépendante. Sept directives ont été sélectionnées sur base de cette évaluation. Elles ont été actualisées pour chaque question clinique, en recherchant les données probantes les plus récentes dans Medline et la Cochrane Database of Systematic Reviews. Un niveau d’évidence a été attribué à chaque recommandation originelle et à chaque étude au moyen de la classification GRADE.

Ensuite, des recommandations ont été formulées par le groupe de développement multidisciplinaire sur base des données probantes. Elles ont finalement été évaluées de manière formelle par des représentants des associations professionnelles et scientifiques. Les conflits d’intérêt des experts ont été répertoriés.

DÉFINITIONS

DÉFINITIONS TOPOGRAPHIQUES De nombreuses discussions concernant la classification des tumeurs de la jonction gastro-oesophagienne sont en cours. Les définitions suivantes sont utilisées dans ce rapport :

• Si plus de 50 % de la tumeur se situe dans la partie gastrique du cardia, celle-ci est classée comme tumeur de l'estomac.

• Si la masse tumorale se situe principalement dans l'œsophage, elle est classée comme tumeur de l’œsophage.

• Les tumeurs de la jonction gastro-oesophagienne doivent être considérées comme des tumeurs de l’œsophage et traitées comme telles.

LÉSIONS PRÉCOCES La définition d’une lésion de Barrett ne fait pas l’objet d’aucun consensus. La définition la plus courante est la suivante : il s’agit d’une modification de l’épithélium œsophagien visible à l’endoscopie et présentant des lésions de métaplasie intestinale confirmées par la biopsie.

Plusieurs classifications de la dysplasie sont disponibles. La pertinence clinique est le critère principal à prendre en compte pour choisir l’une d’entre elles.

Page 9: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75B Cancer oesophagien et gastrique iii

RECOMMANDATIONS FINALES

1. CANCER DE L’ŒSOPHAGE

COM: Concertation Multidisciplinaire

PET : Tomographie par Emission de Positrons

Page 10: Guideline pour la prise en charge du cancer oesophagien et ...

iv Cancer oesophagien et gastrique KCE Reports 75B

Diagnostic

Après l’anamnèse et l’examen clinique, le diagnostic du cancer de l’œsophage est établi par une endoscopie de l’œsophage comportant des biopsies. Les patients présentant les symptômes d’alarme suivants devraient être orientés le plus rapidement possible vers une endoscopie : dysphagie, vomissements incessants, anorexie, perte de poids et hémorragies gastro-intestinales.

Une endoscopie haute résolution et une chromo-endoscopie ne sont pas indiquées en routine, mais peuvent être utiles pour le suivi des patients présentant un risque élevé de cancer de l’œsophage, comme les patients ayant un œsophage de Barrett ou une dysplasie de haut grade.

Approche des lésions de dysplasie

Chez les patients présentant une lésion de Barrett, le protocole de biopsie sera structuré ainsi : réaliser des biopsies tous les 2 centimètres en 4 quadrants et biopsier chaque lésion visible. En cas de présomption de dysplasie de haut grade, il convient de tenir compte en outre des conclusions cliniques et endoscopiques. En cas de confirmation d’une dysplasie de haut grade et si une intervention thérapeutique est envisagée, une évaluation endoscopique (endoscopie haute résolution +/- chromo-endoscopie, avec biopsies tous les centimètres en 4 quadrants) et une évaluation pathologique (confirmation du diagnostic par un deuxième pathologiste, avec biopsies additionnelles éventuelles ou une résection endoscopique (REM) de la muqueuse à visée diagnostique seront réalisées. Le traitement doit être discuté en concertation pluridisciplinaire et avoir lieu de préférence dans un centre possédant des installations endoscopiques et chirurgicales ainsi que l’expertise requises.

Staging

Les patients atteints d’un cancer de l’œsophage bénéficieront d’un CT du thorax (incluant la région basse du cou) et de l’abdomen pour exclure les métastases. Si un traitement curatif est jugé possible, la détermination du stade au moyen d’une écho-endoscopie (EUS) avec ou sans aspiration cytologique à l’aiguille fine (FNAC) sera réalisée. En complément, un PET(/CT) scan peut être envisagé pour l’évaluation des métastases lymphatiques et autres. Des analyses additionnelles, comme une résonance magnétique nucléaire (RMN), une bronchoscopie (+/- échographie des bronches +/- biopsie), une thoracoscopie ou une laparoscopie sont envisageables en cas d’indication précise.

Traitement des cancers de la muqueuse

Chez les patients présentant une lésion T1a, le diagnostic doit être confirmé par un deuxième pathologiste. Des biopsies additionnelles ou une RMN à visée diagnostique peuvent être indiqués en cas de doute. Le traitement doit être discuté en concertation pluridisciplinaire où la REM sera préférée à la chirurgie, compte tenu du stade, de la taille, de la longueur de la lésion de Barrett, du type histologique, du degré de différenciation et de l’invasion lymphovasculaire.

Traitement des cancers sous-muqueux

Traitement néoadjuvant

Le traitement néoadjuvant n’est pas indiqué en routine chez les patients atteints d’un cancer de l’œsophage et doit être discuté durant une concertation pluridisciplinaire. La radiothérapie néoadjuvante n'est pas indiquée.

Si, toutefois, un traitement combiné (traitement néoadjuvant et chirurgie) était réalisé, ses résultats cliniques et ses effets secondaires doivent être enregistrés de manière prospective.

Page 11: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75B Cancer oesophagien et gastrique v

Chirurgie

La chirurgie est le traitement standard des patients atteints d’un cancer de l’œsophage résécable et a lieu de préférence au moyen d’une résection transthoracique ‘en bloc’ avec lymphadénectomie étendue à deux champs. L’objectif de la chirurgie consiste en l’élimination totale de la tumeur (résection R0). La chirurgie du cancer de l’œsophage est effectuée de préférence au sein de services spécialisés ayant un volume d’actes élevé et par un chirurgien ayant l’expérience et/ou la formation requises.

Traitement adjuvant

Le traitement adjuvant n’est pas indiqué chez les patients atteints d’un cancer de l’œsophage.

Traitement non chirurgical à but curatif

Un traitement par radio et chimiothérapie exclusive est envisageable pour les patients présentant un cancer de l'œsophage étendu non résécable ou pour les patients ayant un cancer de l’œsophage localement étendu, inopérables pour des raisons médicales ou qui refusent la chirurgie.

Traitement palliatif et maladie métastatique

Le placement d’un stent, le traitement au laser ou la coagulation au plasma d’argon (APC) sont indiqués pour lever l’obstruction de l’œsophage par une tumeur. Le choix de la technique sera fait en fonction de sa disponibilité et de l’expertise locales. Le traitement de la dysphagie causée par une tumeur œsophagienne sera effectué de préférence avec des stents en métal auto-déployables ou des stents déployables en plastique. En cas de recouvrement du stent par la tumeur, un traitement par laser, l’APC ou un re-stenting sont envisageables. La radiothérapie (externe ou endoluminale) sera utilisée pour les patients atteints de dysphagie ayant une espérance de vie relativement longue.

La chimiothérapie avec ou sans radiothérapie est une solution qui doit être discutée en concertation pluridisciplinaire pour les patients atteints d’un cancer de l'œsophage localement étendu ou métastasé. La chirurgie palliative, comme l’oesophagectomie ou le bypass substernal, n’est plus indiquée dans ces situations.

Il convient que les patients atteints d’un cancer de l’œsophage aient accès à une équipe palliative spécialisée, laquelle apportera une attention spécifique au contrôle des symptômes, à l’alimentation et à la qualité de vie. Le médecin traitant assurera le rôle de coordinateur dans l’organisation des soins palliatifs à domicile.

Follow-up

Chez les patients qui ont été traités pour un cancer de l’œsophage, un examen clinique tous les 3 mois sera réalisé. Il est recommandé de réaliser un CT scan tous les 6 mois au cours de la première année, puis chaque année par la suite. Les patients qui ont été traités par REM subiront une endoscopie de contrôle après 3 mois, puis tous les 6 mois pendant 2 ans, et ensuite chaque année.

Traitement de la récidive

Les options thérapeutiques envisageables pour les patients présentant une récidive de cancer de l’œsophage seront discutées en concertation pluridisciplinaire. La répétition d’un traitement local fait partie des options envisageables pour les patients ayant une récidive localisée ou une deuxième tumeur révélées par la RMN.

Page 12: Guideline pour la prise en charge du cancer oesophagien et ...

vi Cancer oesophagien et gastrique KCE Reports 75B

2. CANCER DE L’ESTOMAC

Page 13: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75B Cancer oesophagien et gastrique vii

Diagnostic

Après une anamnèse et un examen clinique, le diagnostic du cancer de l’estomac est établi par une endoscopie de l’œsophage comportant des biopsies. Les patients présentant les symptômes d’alarme suivants devraient être orientés le plus rapidement possible vers une endoscopie : dysphagie, vomissements incessants, anorexie, perte de poids et hémorragie gastro-intestinales. Un test de positivité au H. pylori doit être réalisé de manière systématique et confirmé par un deuxième test.

Une endoscopie haute résolution et une chromo-endoscopie ne sont pas indiquées en routine, mais peuvent se révéler utiles dans le suivi des patients présentant un risque élevé de cancer de l’estomac, comme c’est le cas pour les patients présentant une dysplasie de haut grade.

Approche des lésions de dysplasie

Chez les patients présentant une dysplasie de haut grade pour lesquels une intervention thérapeutique est envisagée, une évaluation endoscopique et pathologique approfondie est indiquée (confirmation du diagnostic par un deuxième pathologiste, biopsies additionnelles éventuelles). Le traitement doit faire l’objet de discussions en concertation pluridisciplinaire et sera effectué de préférence au sein de services spécialisés ayant un volume d’actes endoscopiques élevé et par un chirurgien ayant l’expérience et/ou la formation requises.

Staging

Les patients atteints d’un cancer de l’estomac, bénéficieront d’un CT du thorax et de l’abdomen pour exclure les métastases. Si un traitement curatif est jugé possible, la déterminations du stade par EUS (écho-endoscopie) avec ou sans FNAC est indiquée. Des examens additionnels comme un PET scan, une RMN ou une laparoscopie seront envisagés en fonction des nécessités.

Traitement du cancer de la muqueuse

Les biopsies d’un cancer des muqueuses seront évaluées par un pathologiste ayant une expérience suffisante. Le traitement doit être discuté en concertation pluridisciplinaire où la REM sera préférée à la chirurgie, compte tenu du stade, de la taille, du type histologique, du degré de différentiation.

Traitement du cancer sous-muqueux

Traitement néoadjuvant

Le traitement néoadjuvant n’est pas indiqué en routine chez les patients atteints d’un cancer de l’estomac, mais constitue une option devant être discutée en concertation pluridisciplinaire.

En cas de traitement combiné (traitement néoadjuvant et chirurgie), les résultats cliniques et les effets secondaires seront enregistrés de manière prospective.

Chirurgie

La chirurgie est le traitement standard des patients atteints d’un cancer de l’estomac résécable. Les tumeurs distales sont traitées au moyen d’une gastrectomie partielle, les tumeurs proximales, par gastrectomie totale. Une lymphadénectomie D2 doit être réalisée de manière standard. L’objectif de la chirurgie consiste en l’élimination totale de la tumeur (résection R0). La chirurgie du cancer de l’estomac a lieu de préférence au sein de services spécialisés avec un volume élevé par un chirurgien ayant l’expérience et/ou la formation requises.

Page 14: Guideline pour la prise en charge du cancer oesophagien et ...

viii Cancer oesophagien et gastrique KCE Reports 75B

Traitement adjuvant

Le traitement adjuvant n’est pas indiqué chez les patients atteints d’un cancer de l’estomac. La chimiothérapie adjuvante n’est pas conseillée de manière routinière, mais peut être envisagée après discussion en concertation pluridisciplinaire.

Traitement palliatif et maladie métastatique

La chirurgie palliative est limitée aux patients présentant des sténoses symptomatiques, des tumeurs qui saignent ou une perforation. Chez les patients présentant une obstruction maligne de l’estomac, le choix existe entre un stenting endoscopique ou une gastroentérostomie chirurgicale.

Chez les patients atteints d’un cancer étendu ou métastatique ayant une bonne condition physique générale, une chimiothérapie combinée est envisageable.

Il convient que les patients atteints d’un cancer de l’estomac aient accès à une équipe palliative spécialisée, laquelle apportera une attention spécifique au contrôle des symptômes, à l’alimentation et à la qualité de vie. Le médecin traitant devra remplir un rôle de coordination dans l’organisation des soins palliatifs à domicile.

Follow-up

Chez les patients qui ont été traités pour un cancer de l’estomac, un examen clinique et une analyse de sang sont conseillés tous les 3 mois. Un CT scan est indiqué tous les 6 mois au cours de la première année, et ensuite chaque année. Les patients qui ont été traités par REM subiront une endoscopie de contrôle après 3 mois, puis tous les 6 mois durant 2 ans, et ensuite chaque année.

Traitement de la récidive

Les options thérapeutiques envisageables pour les patients présentant une récidive de cancer de l’estomac seront discutées en concertation pluridisciplinaire. Pour les patients présentant une récidive locale ou une nouvelle tumeur après la REM, un deuxième traitement local fait partie des options.

3. LYMPHOME PRIMAIRE DE L’ESTOMAC

Diagnostic et staging

En cas de suspicion d’un lymphome primaire de l’estomac, la réalisation de 8 à 12 biopsies (au minimum) est indiquée. Les biopsies seront conservées de manière telle qu’un diagnostic moléculaire soit possible. Chez les patients pour lesquels un lymphome primaire de l’estomac a été confirmé par l’histologie, un EUS (sans FNAC) est indiqué. Il est inutile de procéder à d’autres investigations pour les patients présentant un lymphome de type MALT (mucosa-associated lymphoid tissue) de faible malignité, à moins qu’il ne soit nécessaire d’établir un diagnostic différentiel.

Traitement

L’éradication de l’H. pylori est le traitement de premier choix des patients présentant un lymphome MALT de faible malignité à petites cellules. Ensuite, un suivi rigoureux avec endoscopie et biopsies est nécessaire. L’évaluation de l’éradication de l’ H. pylori aura lieu dans les 3 mois. Les patients présentant une éradication réussie de l’ H. pylori, mais pas de régression tumorale après 1 an ou présentant une progression tumorale seront dirigés vers un centre spécialisé en hématologie.

Page 15: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75B Cancer oesophagien et gastrique ix

4. TUMEURS STROMALES GASTRO-INTESTINALES DE L’ESTOMAC

Diagnostic et staging

Un EUS avec FNAC et un test immunohistochimique du CD117 sont recommandés pour les patients présentant une suspicion de tumeur stromale gastro-intestinale (GIST). . Si une thérapie est envisagée, un CT de l’abdomen est conseillé..

Traitement

GIST résécable non-métastatique

La résection chirurgicale est indiquée pour les patients présentant une bonne condition physique générale avec un GIST non-métastatique histologiquement confirmé. Celle-ci est également indiquée pour les patients présentant une bonne condition physique générale et une tumeur de l’estomac > à 5 cm suggestive d’un GIST sans métastases.

Le choix entre une résection chirurgicale et l’expectative sera discuté en concertation pluridisciplinaire pour les patients présentant une bonne condition physique générale et une tumeur de l’estomac de 2 à 5 cm suggestive d’un GIST sans métastases. L’expectative est indiquée pour les patients présentant une tumeur de l’estomac de < 2 cm suggestive d’un GIST sans métastases.

GIST métastatique et/ou non-résécable

Le traitement à l’imatinib est conseillé pour les patients atteints d’un GIST métastatique et/ou non-résécable. Un PET/CT est recommandé pour évaluer la réponse à l’imatinib. Pour les patients résistants à l’imatinib ou présentant une intolérance à ce produit, le sunitinib peut être considéré comme traitement de deuxième ligne.

IMPLEMENTATION ET REVISION DE LA RECOMMANDATION

Il convient que l’implémentation de cette directive soit facilitée par la mise en ligne d’un algorithme basé sur ces recommandations. Cet outil sera mis à disposition sur le site web du Collège d’Oncologie.

Des indicateurs de qualité appropriés seront développés sur base des principales recommandations.

En raison de l’évolution des données probantes, une actualisation de cette recommandation sera probablement nécessaire dans 5 ans, au terme d’une pré-évaluation de la littérature.

Page 16: Guideline pour la prise en charge du cancer oesophagien et ...
Page 17: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 1

Scientific summary

Table of contents

1 INTRODUCTION ................................................................................................... 5 1.1 SCOPE.................................................................................................................................................. 5 1.2 EPIDEMIOLOGY .................................................................................................................................. 5

1.2.1 Oesophageal cancer ............................................................................................................ 5 1.2.2 Gastric cancer ...................................................................................................................... 6

2 METHODOLOGY................................................................................................... 8 2.1 GENERAL APPROACH........................................................................................................................ 8 2.2 CLINICAL QUESTIONS ....................................................................................................................... 8 2.3 SEARCH FOR EVIDENCE..................................................................................................................... 9

2.3.1 Clinical practice guidelines................................................................................................. 9 2.3.2 Additional evidence...........................................................................................................10

2.4 QUALITY APPRAISAL........................................................................................................................10 2.4.1 Clinical practice guidelines...............................................................................................10 2.4.2 Additional evidence...........................................................................................................10

2.5 DATA EXTRACTION AND SUMMARY ............................................................................................10 2.6 FORMULATION OF RECOMMENDATIONS.....................................................................................10 2.7 EXTERNAL REVIEW ..........................................................................................................................10 3 DEFINITIONS...................................................................................................... 12 3.1 TOPOGRAPHIC DEFINITIONS .........................................................................................................12 3.2 EARLY LESIONS.................................................................................................................................13

3.2.1 Histology of the normal oesophagus ............................................................................13 3.2.2 Barrett’s oesophagus ........................................................................................................13 3.2.3 Dysplasia in squamous epithelium .................................................................................15 3.2.4 Dysplasia in columnar epithelium ..................................................................................15

4 FINAL RECOMMENDATIONS ON OESOPHAGEAL CANCER ................................. 18 4.1 FLOWCHART....................................................................................................................................18 4.2 DIAGNOSIS .......................................................................................................................................19 4.3 WORK-UP OF DYSPLASTIC LESIONS..............................................................................................20 4.4 STAGING...........................................................................................................................................22 4.5 TREATMENT OF MUCOSAL CANCER .............................................................................................23 4.6 TREATMENT OF CANCER BEYOND THE MUCOSA .......................................................................24

4.6.1 Neoadjuvant treatment....................................................................................................24 4.6.2 Surgical treatment .............................................................................................................25 4.6.3 Adjuvant treatment ...........................................................................................................27

Page 18: Guideline pour la prise en charge du cancer oesophagien et ...

2 Upper GI Cancer KCE reports 75

4.6.4 Non-surgical treatment with curative intent ..............................................................27 4.7 PALLIATIVE TREATMENT & METASTATIC DISEASE ........................................................................28 4.8 FOLLOW-UP .....................................................................................................................................30 4.9 RECURRENT DISEASE .......................................................................................................................30 5 FINAL RECOMMENDATIONS ON GASTRIC CANCER............................................ 31 5.1 FLOWCHART....................................................................................................................................31 5.2 DIAGNOSIS .......................................................................................................................................32 5.3 WORK-UP OF DYSPLASTIC LESIONS..............................................................................................33 5.4 STAGING...........................................................................................................................................34 5.5 TREATMENT OF MUCOSAL CANCER .............................................................................................35 5.6 TREATMENT OF CANCER BEYOND THE MUCOSA .......................................................................36

5.6.1 Neoadjuvant treatment....................................................................................................36 5.6.2 Surgical treatment .............................................................................................................37 5.6.3 Adjuvant treatment ...........................................................................................................38

5.7 PALLIATIVE TREATMENT & METASTATIC DISEASE ........................................................................39 5.8 FOLLOW-UP .....................................................................................................................................40 5.9 RECURRENT DISEASE .......................................................................................................................41 5.10 TREATMENT OF GASTRIC LYMPHOMA ..........................................................................................41

5.10.1 Introduction ........................................................................................................................41 5.10.2 Diagnosis and staging ........................................................................................................41 5.10.3 Treatment............................................................................................................................42

5.11 TREATMENT OF GASTROINTESTINAL STROMAL TUMOURS ........................................................43 5.11.1 Introduction ........................................................................................................................43 5.11.2 Diagnosis and staging ........................................................................................................43 5.11.3 Treatment............................................................................................................................44

6 IMPLEMENTATION AND UPDATING OF THE UPPER GI CANCER GUIDELINE...... 46 6.1 IMPLEMENTATION............................................................................................................................46 6.2 QUALITY CONTROL........................................................................................................................46 6.3 GUIDELINE UPDATE.........................................................................................................................46 7 REFERENCES ...................................................................................................... 47 8 APPENDICES ...................................................................................................... 65

Page 19: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 3

ABBREVIATIONS 95% CI 95 percent confidence interval

5-FU 5-fluorouracil

AFE Autofluorescence endoscopy

AGA American Gastroenterological Association

APC Argon plasma coagulation

ASCO American Society of Clinical Oncology

BUS Bronchoscopic ultrasound

CBO Dutch Institute for Healthcare Improvement

CCO Cancer Care Ontario

CODG Conventional open distal gastrectomy

CPG Clinical Practice Guideline

CRT Chemoradiotherapy

CT Computed tomography

EMR Endoscopic mucosal resection

ESD Endoscopic submucosal dissection

EUS Endoscopic ultrasound

FNA(C Fine needle aspiration (cytology)

FNCLCC Fédération Nationale des Centres de Lutte Contre le Cancer

GI Gastrointestinal

GIST Gastrointestinal stromal tumours

GOJ Gastro-oesophageal junction

GRADE Grading of Recommendations Assessment, Development and Evaluation

HR Hazard ratio

HRE High-resolution endoscopy

IARC International Agency for Research on Cancer

IBD Inflammatory bowel disease

IUAC International Union Against Cancer

LN Lymph node

MALT Mucosa-associated lymphoid tissue

MDCT Multidetector row CT

MDT Multidisciplinary team

MeSH Medical Subject Headings

MRI Magnetic resonance imaging

NBI Narrow band imaging

NICE National Institute for Health and Clinical Excellence

OR Odds ratio

PDT Photodynamic therapy

PET Positron-emission tomography

Page 20: Guideline pour la prise en charge du cancer oesophagien et ...

4 Upper GI Cancer KCE reports 75

RCT Randomised Controlled Trial

RR Risk ratio

SCJ Squamo-columnar junction

SEMS Self-expanding metal stents

SIGN Scottish Intercollegiate Guidelines Network

SSBE Short-segment Barrett’s oesophagus

UK United Kingdom

US United States of America

WHO World Health Organisation

Page 21: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 5

1 INTRODUCTION

1.1 SCOPE In the present report, a clinical practice guideline (CPG) on upper gastrointestinal cancer is presented, which is the result of a collaboration of the College of Physicians for Oncology and the KCE. This clinical practice guideline will cover a broad range of topics: diagnosis, staging, treatment, supportive therapy, and follow-up. The guideline primarily concerns individuals with oesophageal or gastric cancer, and is intended to be used by all care providers involved in the care for these patients.

1.2 EPIDEMIOLOGY

1.2.1 Oesophageal cancer

Oesophageal cancer is the eighth most common cancer in the world and one of the most lethal [1]. Incidence rates of oesophageal cancer show well-known regional disparities. Overall, incidence rates for all types of oesophageal cancer range from four to nine cases per 100.000 males per year (1975 – 1997) in Western countries [2]. Lower incidence rates are found in Northern Europe (Finland, Norway, and Sweden), whereas the French regions of Burgundy and Calvados have incidence rates of > 14 per 100.000 males per year.

In the Netherlands, Crane et al. found an increase in age standardised incidence by 3.4% and 1.9% per year (1989 – 2003) for males and females respectively [3]. This increase was almost exclusively caused by oesophageal adenocarcinomas. In 14 years, age standardised mortality increased 2.5% per year among males and 1.7% per year among females. Similar trends were found in the UK and the US, but not in France [4]. Relative survival in the Netherlands improved significantly from 8.1% in 1989-1993 to 12.6% in 1999-2003 (p<0.001) [3].

Differences in incidence trends of the two main histological types of oesophageal cancer – squamous cell carcinoma and adenocarcinoma – are noteworthy. The incidence rate of squamous cell carcinoma of the oesophagus has been relatively stable in most countries from 1975 to 1995 according to the International Agency for Research on Cancer (IARC), although increasing trends were observed in Denmark and the Netherlands among men and in Canada, Scotland and Switzerland among women [5]. A significant increase in the incidence of oesophageal adenocarcinomas was found in both sexes in Canada and South Australia and in 6 European countries (Scotland, Denmark, Iceland, Finland, Sweden and Norway). In France the increase was limited to men and in Switzerland the increase was observed only in women [5].

In Belgium, the crude incidence rate of oesophageal cancer rose from 8.8 per 100.000 males in 1997 to 10.8 per 100.000 males in 2003, and from 2.2 per 100.000 females in 1997 to 3.5 per 100.000 females in 2003 (Belgian Cancer Registry, personal communication). Age standardised incidence increased by 2.9% and 11.7% per year (1997 – 2003) for males and females respectively (Table 1).

Table 1. Age standardised incidence$ of oesophageal cancer in Belgium, 1997 – 2003 (n/100.000 person years) (Belgian Cancer Registry, personal communication).

Males Females Year Belgium Flanders Belgium Flanders

1997 5.8 6.5 1.0 1.2 1998 5.5 6.2 1.1 1.2 1999 5.8 5.7 1.4 1.5 2000 6.3 6.8 1.6 1.7 2001 7.0 8.0 1.6 1.5 2002 6.9 7.3 1.6 1.3 2003 6.8 7.0 1.7 1.9

$using a world standard population.

Page 22: Guideline pour la prise en charge du cancer oesophagien et ...

6 Upper GI Cancer KCE reports 75

The differences in incidence trends of oesophageal squamous cell carcinoma and adenocarcinoma are less pronounced in Belgium (Table 2 and Figure 1). This is probably due to the fact that the Belgian cancer registration is of recent years (only going back until 1997). During the first registration years a high percentage of tumours with unspecified histology were registered. However, recently the quality of the cancer registration markedly improved. This lead to an increase in incidence of both squamous cell carcinoma and adenocarcinoma and a decrease in the incidence of tumours with unspecified histology (Table 2 and Figure 1) (Belgian Cancer Registry, personal communication).

Table 2. Age standardised incidence$ of oesophageal cancer in Belgium according to histological type, 1997 – 2003 (n/100.000 person years) (Belgian Cancer Registry, personal communication).

1997 1998 1999 2000 2001 2002 2003 Males • SCC# 4.0 3.9 4.2 4.4 4.9 5.2 4.7 • Adenocarcinoma 2.7 2.8 2.9 3.6 4.5 4.0 4.3 • Unspecified tumour 1.0 0.6 0.5 0.4 0.2 0.1 0.2 Females • SCC# 0.9 1.0 1.3 1.4 1.6 1.5 1.5 • Adenocarcinoma 0.4 0.4 0.4 0.7 0.6 0.5 0.8 • Unspecified tumour 0.1 0.1 0.2 0.0 0.1 0.1 0.1

$using a European standard population. #SCC: squamous cell carcinoma.

Figure 1. Age standardised incidence$ of oesophageal cancer in Belgium for males according to histological type, 1997 – 2003 (n/100.000 person years) (Belgian Cancer Registry, personal communication).

$using a European standard population.

1.2.2 Gastric cancer

With an estimated 934.000 new cases per year in 2002 worldwide (8.6% of all new cancer cases), gastric cancer is in fourth place behind cancers of the lung, breast, and colon and rectum, with almost two-third of the cases occurring in developing countries [1]. It is the second most common cause of death from cancer.

Gastric cancer incidence rates vary by up to ten-fold throughout the world. Japan and Korea have the highest gastric cancer incidence rates in the world.

Page 23: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 7

High-incidence areas for non-cardia gastric adenocarcinoma include East Asia, Eastern Europe, and Central and South America. Low incidence rates are found in South Asia, North and East Africa, North America, Australia, and New Zealand [6]. Survival is moderately good only in Japan (52%), where mass screening by photofluoroscopy has been practiced since the 1960s. Survival is also relatively high in North America (approximatively 21%), possibly due to early diagnosis following a higher number of endoscopic examinations performed for gastric disorders. Estimated survival is 27% in Western Europe [1].

In the Netherlands, age standardised incidence of gastric cancer declined from 21.6 to 15.9 per 100.000 person years in males and from 10.4 to 6.2 per 100.000 person years in females since 1978 [7]. The age standardised mortality rates decreased from 20.7 to 12.8 per 100.000 person years in males and from 8.2 to 4.2 per 100.000 person years in females.

In Belgium, the crude incidence rate of gastric cancer rose from 12.9 per 100.000 males in 1997 to 14.9 per 100.000 males in 2003, and from 8.0 per 100.000 females in 1997 to 8.4 per 100.000 females in 2003 (Belgian Cancer Registry, personal communication). Age standardised incidence increased by 2.6% and 0.8% per year (1997 – 2003) for males and females respectively (Table 3). However, in these rates tumours of the gastro-oesophageal junction (GOJ) are also included.

While the incidence rates of these GOJ tumours recently increased, the incidence rates of ‘real’ gastric tumours (see chapter 3.1) declined [8].

Table 3. Age standardised incidence$ of gastric cancer in Belgium, 1997 – 2003 (n/100.000 person years) (Belgian Cancer Registry, personal communication).

Males Females Year Belgium Flanders Belgium Flanders

1997 7.4 9.2 3.1 4.3 1998 7.3 8.3 3.4 4.1 1999 7.9 9.1 3.5 4.4 2000 7.5 9.0 3.9 4.5 2001 8.2 9.8 3.5 4.2 2002 8.5 9.8 3.9 4.5 2003 8.2 9.1 3.2 3.6

$using a world standard population.

Page 24: Guideline pour la prise en charge du cancer oesophagien et ...

8 Upper GI Cancer KCE reports 75

2 METHODOLOGY

2.1 GENERAL APPROACH As for the previous CPGs developed within the collaboration between the College and the KCE, the present CPG was developed by adapting (inter)national CPGs to the Belgian context [9]. This approach is currently being structured in a formal methodology by the ADAPTE group, an international group of guideline developers and researchers [10]. The ADAPTE methodology generally consists of three major phases:

1. Set-up Phase: Outlines the necessary tasks to be completed prior to beginning the adaptation process (e.g., identifying necessary skills and resources).

2. Adaptation Phase: Assists guideline developers in moving from selection of a topic to identification of specific clinical questions; searching for and retrieving guidelines; assessing the consistency of the evidence therein, their quality, currency, content and applicability; decision making around adaptation; and preparing the draft adapted guideline.

3. Finalization Phase: Guides guideline developers through getting feedback on the document from stakeholders who will be impacted by the guideline, consulting with the source developers of guidelines used in the adaptation process, establishing a process for review and updating of the adapted guideline and the process of creating a final document.

2.2 CLINICAL QUESTIONS The clinical practice guideline addresses the following clinical questions:

1. Which diagnostic techniques should be used for the diagnosis of oesophageal and gastric cancer?

2. What are the best treatment options for dysplastic lesions of the oesophagus and stomach?

3. What staging techniques should be used for oesophageal and gastric cancer?

4. What are the best treatment options for mucosal oesophageal and gastric cancer?

5. What are the best treatment options for oesophageal and gastric cancer beyond the mucosa?

a. neoadjuvant treatment

b. surgical treatment

c. adjuvant treatment

d. non-surgical treatment with curative intent

6. What are the best palliative treatment options for extensive disease?

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

8. What are the best treatment options for gastric lymphoma?

9. What are the best treatment options for gastrointestinal stromal tumours?

Page 25: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 9

2.3 SEARCH FOR EVIDENCE

2.3.1 Clinical practice guidelines

2.3.1.1 Sources

A broad search of electronic databases (Medline, EMBASE), specific guideline websites and websites of oncologic organisations (Table 4) was conducted in July 2007.

Table 4: Searched guideline websites and websites of oncologic organisations.

Alberta Heritage Foundation For Medical Research (AHFMR)

http://www.ahfmr.ab.ca/

American Society of Clinical Oncology (ASCO) http://www.asco.org/ American College of Surgeons (ACS) http://www.facs.org/cancer/coc/ CMA Infobase http://mdm.ca/cpgsnew/cpgs/index.asp Guidelines International Network (GIN) http://www.g-i-n.net/ National Comprehensive Cancer Network (NCCN)

http://www.nccn.org/

National Guideline Clearinghouse http://www.guideline.gov/ National Cancer Institute http://www.cancer.gov/ Haute Autorité de Santé (HAS) http://bfes.has-

sante.fr/HTML/indexBFES_HAS.html BC Cancer Agency http://www.bccancer.bc.ca/default.htm Institute for Clinical Systems Improvement (ICSI) http://www.icsi.org/index.asp National Health and Medical Research Council (NHMRC)

http://www.nhmrc.gov.au/

Scottish Intercollegiate Guidelines Network (SIGN)

http://www.sign.ac.uk/

New Zealand Guidelines Group (NZGG) http://www.nzgg.org.nz/ Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC)

http://www.fnclcc.fr/sor/structure/index-sorspecialistes.html

National Institute for Health and Clinical Excellence (NICE)

http://www.nice.org.uk/

2.3.1.2 Search terms

For Medline the following MeSH terms were used in combination: stomach neoplasms, esophageal neoplasms. For EMBASE the following Emtree terms were used in combination: stomach tumor, esophagus tumor. These MeSH and Emtree terms were combined with a standardised search strategy to identify CPGs (Table 5).

Table 5: Standardised search strategy for CPGs.

Database Search strategy Medline guideline [pt] OR practice guideline [pt] OR

recommendation* [ti] OR standard* [ti] OR guideline* [ti]

EMBASE 'practice guideline'/exp

2.3.1.3 In- and exclusion criteria

Both national and international CPGs on oesophageal and gastric cancer were searched. A language (English, Dutch, French) and date restriction (2001 – 2007) were used. CPGs without references were excluded, as were CPGs without clear recommendations.

Page 26: Guideline pour la prise en charge du cancer oesophagien et ...

10 Upper GI Cancer KCE reports 75

2.3.2 Additional evidence

For each clinical question, the evidence – identified through the included CPGs – was updated by searching Medline and the Cochrane Database of Systematic Reviews from the search date of the CPG on (search date August – September 2007). A combination of appropriate MeSH terms and free text words was used (see appendix).

For therapeutic interventions, only systematic reviews and randomized controlled trials (RCT) were included. However, for diagnostic interventions we also searched for observational studies in case no systematic review or RCT was found. The identified studies were selected based on title and abstract. For all eligible studies, the full-text was retrieved. In case no full-text was available, the study was not taken into account for the final recommendations.

2.4 QUALITY APPRAISAL

2.4.1 Clinical practice guidelines

In total, 17 CPGs were identified. All were quality appraised by two independent reviewers (JV, FM) using the AGREE instrument. Disagreement was discussed face-to-face. At the end, agreement was reached for all CPGs, and 7 CPGs were included (see appendix).

2.4.2 Additional evidence

The quality of the retrieved systematic reviews and RCTs was assessed using the checklists of the Dutch Cochrane Centre (www.cochrane.nl).

2.5 DATA EXTRACTION AND SUMMARY For each included CPG the following data were extracted: search date & publication year, searched databases, availability of evidence tables, recommendations and referenced evidence.

For each systematic review, the search date, publication year, included studies and main results were extracted. For RCTs, the following data were extracted: publication year, study population, study intervention, and outcomes.

For each clinical question, the recommendations from the identified CPGs and the additional evidence were summarized in evidence tables. A level of evidence was assigned to each recommendation and additional study using the GRADE system (see appendix).

2.6 FORMULATION OF RECOMMENDATIONS Based on the retrieved evidence, a first draft of recommendations was prepared by a small working group (JV, FM). This first draft together with the evidence tables was circulated to the guideline development group 2 weeks prior to the first face-to-face meeting. The guideline development group met on several occasions (October 1st 2007, November 5th 2007, December 10th 2007 and January 9th 2008) to discuss the first draft. Recommendations were changed if important evidence supported this change. Based on the discussion meetings a second draft of recommendations was prepared. A grade of recommendation was assigned to each recommendation using the GRADE system (see appendix). The second draft was once more circulated to the guideline development group for final approval.

2.7 EXTERNAL REVIEW The recommendations prepared by the guideline development group were circulated to the Professional Associations (Table 6). Each association was asked to assign 2 key persons to discuss the recommendations during an open meeting.

Page 27: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 11

These panellists received the recommendations one week prior to this open meeting. As a preparation of the meeting all invited panellists were asked to score each recommendation on a 5-point Likert-scale to indicate their agreement with the recommendation, with a score of ‘1’ indicating ‘completely disagree’, ‘2’ indicating ‘somewhat disagree’, ‘3’ indicating ‘unsure’, ‘4’ indicating ‘somewhat agree’, and ‘5’ indicating ‘completely agree’ (the panellists were also able to answer ‘not applicable’ in case they were not familiar with the underlying evidence). In case a panellist disagreed with the recommendation (score ‘1’ or ‘2’), (s)he was asked to provide appropriate evidence. All scores (n = 15) were than anonymized and summarized into a mean score, standard deviation and % of ‘agree’-scores (score ‘4’ and ‘5’) to allow a targeted discussion (see appendix). The recommendations were then discussed during a face-to-face meeting on January 23rd 2008. Based on this discussion a final draft of the recommendations was prepared, and discussed by the guideline development group by email. In appendix, an overview is provided of how the comments of the experts were taken into account.

Table 6: List of Professional Associations to which the recommendations were communicated.

Belgian Society of Medical Oncology (BSMO)

Belgian Society of Radiotherapy (BVRO – ABRO)

Belgian Society of Nuclear Medicine

Belgian Society of Surgical Oncology (BSSO)

Upper GI section of the Royal Belgian Society of Surgery

Flemish Society of Gastroenterology (VVGE)

Belgian Group of Digestive Oncology (BGDO)

Royal Belgian Society of Gastroenterology (SRBGE)

Domus Medica (Scientific association of Flemish general practitioners)

Belgian Society of Gastrointestinal Endoscopy (BSGIE)

Belgian Digestive Pathology Club

Belgian Society of Pathology

Royal Belgian Radiological Society

Scientific Society of General Medicine (SSMG)

Belgian Group for Endoscopic Surgery (BGES)

Page 28: Guideline pour la prise en charge du cancer oesophagien et ...

12 Upper GI Cancer KCE reports 75

3 DEFINITIONS

3.1 TOPOGRAPHIC DEFINITIONS Traditionally, when discussing cancer of the oesophagus, cancer of the gastro-oesophageal junction (GOJ) is also included. Clinicians are very often confronted with adenocarcinomas that straddle the GOJ. Various criteria have been used to categorize tumours situated at the GOJ. In most classification systems, the anatomic location of the epicentre or predominant mass of the tumour is used to determine whether the neoplasm is oesophageal or gastric (cardia) in origin.

Siewert and Stein proposed a topographic classification for cardia carcinomas [11]. According to these authors, epidemiologic, clinical, and pathologic data support a subclassification of adenocarcinomas arising into the vicinity (i.e. that have their centre within 5 cm proximal and distal of the anatomical cardia) of the GOJ into:

1. adenocarcinoma of the distal oesophagus, which usually arises from an area with specialized intestinal metaplasia (i.e. Barrett oesophagus) and may infiltrate the GOJ from above (type I);

2. true carcinoma of the cardia arising immediately at the GOJ (type II);

3. subcardial carcinoma that infiltrates the GOJ and distal oesophagus from below (type III).

In contrast to previously described classification systems, Siewert and Stein attempted to solve the problem of splitting up GOJ tumours into oesophageal and gastric tumours by creating a third entity [11]. This third entity, the so-called cardiacarcinoma, is lumping a large group of tumours and ‘squeezes’ the true GOJ tumours between the type I and type II tumours. Their effort seems rather adding to the confusion than helping to solve the true problem. This classification is entirely based on identifying the “anatomical” cardia and measuring the centre of the tumour in relation to this anatomical cardia on the resected specimen (i.e. pathological staging). However, measuring the centre of the tumour is impractical if not impossible for clinical staging purposes, which need to be as accurate as possible for making appropriate therapeutic decisions.

In 2000, the World Health Organization Classification of Tumours published Pathology and Genetics of Tumours of the Digestive System [12]. The authors formulated diagnostic criteria based on the following definition of the GOJ: “the GOJ is the anatomical region at which the tubular oesophagus joins the stomach”. According to these authors, adenocarcinomas that cross the GOJ are called adenocarcinomas of the GOJ, regardless of where the bulk of the tumour lies. Adenocarcinomas located entirely above the GOJ, as defined above, are considered oesophageal carcinomas. Adenocarcinomas located entirely below the GOJ are considered gastric in origin. The use of the ambiguous and often misleading term ‘carcinoma of the gastric cardia’ is discouraged. Depending on their size, these tumours should instead be referred to as carcinoma of the body of the stomach [12].

In the most recent recommendations of the International Union Against Cancer (IUAC) [13], according to the advice formulated in the supplement on the TNM classification, adenocarcinomas situated at the GOJ are to be classified into oesophageal, GOJ or cardiac adenocarcinomas using a single major criterion, i.e. the localization of the bulk of the tumour. If more than 50% of the mass of the tumour is situated in the cardia, the tumour should be considered to be of cardiac origin and classified as a gastric tumour. If the mass of the tumour is predominantly found in the oesophagus, it is to be classified as an oesophageal tumour. Furthermore, it is specified that a tumour situated on the GOJ is likely to be of oesophageal origin when the neoplastic lesion was associated with a Barrett oesophagus of the specialized or intestinal type. Unfortunately, the recommendations in the most recent Cancer Staging Manual on how to handle these tumours may not always be compatible with this classification, again creating confusion.

Page 29: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 13

The chapter on stomach refers to the 50% rule, whereas the chapter on oesophagus indicates that “tumours arising within the GOJ and gastric cardia that have minimal involvement (2 cm or less) of the oesophagus are considered primary gastric cancers”.

A continuing increase in the incidence of cardia cancer has been reported since the mid 1970s [14]. The output of scientific publications on cardia and cardiac cancer has evolved in parallel internationally. Today, the vast majority of data available on cardia and cardiac cancer are not comparable because of lack/variability of diagnostic criteria. This may result in therapeutic approaches based on loose (and non-scientific) grounds. Uniformity in classification, terminology and diagnostic criteria is urgently needed. At present, at least in our experience, it would appear that the similarities between adenocarcinoma of the GOJ or cardia and Barrett adenocarcinoma outnumber the dissimilarities and are to be differentiated from gastric (including “subcardia”) cancer [15, 16].

Since the ‘50% rule’ as proposed by the IUAC [13] is practical from a clinical staging point of view, easily being applicable on endoscopic ultrasonography (EUS) (the top of the longitudinal gastric mucosal folds being the endoscopic landmark between oesophagus and stomach), this rule will be used in the present CPG. “True” GOJ tumours should be classified and treated as oesophageal tumours.

Key points

• If more than 50% of the mass of the tumour is situated in the cardia, the tumour should be considered to be of cardiac origin and classified as a gastric tumour.

• If the mass of the tumour is predominantly found in the oesophagus, it should be classified as an oesophageal tumour.

• Tumours of the gastro-oesophageal junction should be classified and have the same concept of treatment as oesophageal tumours.

3.2 EARLY LESIONS

3.2.1 Histology of the normal oesophagus

The luminal side of the normal oesophagus is lined by mucosa composed of epithelium, lamina propria and the muscularis mucosae. Except for a short segment of columnar epithelium in the distal oesophagus at the gastro-oesophageal junction the normal oesophageal epithelium is a tough non-keratinizing stratified squamous epithelium. This epithelium consists of a dynamic cell population which is renewed continuously. The different cell layers in the squamous epithelium - basal, intermediate or prickle cell layers and superficial layers (functional and surface) - are the morphological expression of processes of proliferation, differentiation or maturation and dying cells. A variety of cell types such as neuroendocrine cells (Merkel cells), rare melanocytes, lymphocytes and Langerhans cells are normally present within the squamous epithelium of the oesophagus. The lamina propria rests on a muscularis mucosae. The lamina propria contains lymphatics, blood vessels, nerve fibres and occasional inflammatory cells. The three remaining layers of the oesophageal wall are the submucosa, an area of loose connective tissue containing mucus-secreting glands that open into the lumen via ducts, the muscularis propria with an inner circular and an outer longitudinal layer, and the adventitia. The oesophagus lacks a defining layer of mesothelial cells.

3.2.2 Barrett’s oesophagus

3.2.2.1 Anatomy

The muscular GOJ is the site at which the most distal portion of the oesophagus (the most distal segment of the lower oesophageal sphincter [LOS]) meets the proximal stomach. Endoscopically, one can closely approximate the muscular GOJ by identifying the proximal margin of the gastric folds.

Page 30: Guideline pour la prise en charge du cancer oesophagien et ...

14 Upper GI Cancer KCE reports 75

The mucosal GOJ, also known as the mucosal squamo-columnar junction (SCJ) or Z-line, is the site at which the squamous mucosa of the oesophagus meets columnar- lined mucosa. It is important to understand, however, that the SCJ may be at the same level as the muscular GOJ or may lie 1-2 cm above the muscular GOJ in ‘normal’ individuals.

In order to avoid confusion between Barrett’s mucosa and normal – gastric – junctional columnar mucosa, especially in cases further complicated by the presence of hiatus hernia, an arbitrary minimal length of 3 cm of Barrett’s mucosa from the GOJ was required before the diagnosis of Barrett’s mucosa could be made [17]. Short-segment Barrett’s oesophagus (SSBE) was later defined as Barrett’s mucosa <2-3 cm in length and an ultrashort segment as a microscopically Barrett’s mucosa at a normal looking GOJ, both in contrast to the classical ‘long’ segment Barrett’s oeosophagus.

3.2.2.2 Histology

THE EPITHELIUM

Barrett’s mucosa is a type of metaplasia (replacement of one mature tissue type by another mature tissue type) aimed at better withstanding the gastro-oesophageal reflux. Since its first description, three types of columnar epithelium were described in Barrett’s mucosa: the specialized intestinal epithelium (SIM), the junctional epithelium (or cardia-antral type) and the fundic (or oxyntic) epithelium, the two latter both being gastric types [18]. The malignant potential of Barrett’s mucosa was subsequently described and specifically attributed to the specialized intestinal epithelium and not to the two gastric types of metaplasia [19-22].

Over time, the following ‘adapted’ definitions of Barrett’s mucosa – combining endoscopy and histology – were proposed:

- A change in the oesophageal epithelium of any length that can be recognized at endoscopy and is confirmed to have intestinal metaplasia by biopsy [23] (which is the most commonly accepted definition);

- A displacement of the SCJ proximal to the GOJ with the presence of specialized intestinal epithelium [24];

- An apparent area above the GOJ that is suggestive of Barrett’s, which is supported by the finding of columnar lined oesophagus on histology. The finding of intestinal metaplasia, although often present, is not a requirement for diagnosis. If a sufficient number of biopsies are taken over an adequate period of time, intestinal metaplasia can usually be demonstrated in the majority of these patients [25].

The fact that the definition of Barrett’s mucosa has evolved and is adapted over time and that these ‘adapted’ definitions show (minor) differences, does hamper the interpretation of data published in the literature.

Conceptually, intestinal metaplasia in Barrett’s mucosa is an integral part of the ‘metaplasia-dysplasia-carcinoma’ sequence. Pragmatically, it was thought that the finding of specialized intestinal mucosa was the ultimate hallmark of biopsies taken in a Barrett’s mucosa. Therefore, these biopsies would have to be taken in the oesophagus and not in the stomach, i.e. taken in the cardia (especially, when considering an ultrashort Barrett’s oesophagus). In the meantime, it has been shown that intestinal metaplasia does develop in the cardia too. Thus, the finding of intestinal cells at the GOJ is an abnormal feature. It is not clear whether this metaplastic epithelium originates from the oesophagus (so-called ultrashort Barrett) or from the stomach

(intestinal metaplasia of the gastric cardia), the latter arguably being the result of gastro-oesophageal reflux disease and subsequent carditis [26].

THE MUSCULARIS MUCOSAE

Patients with Barrett’s oesophagus often develop a new (superficial), more luminally situated, layer of muscularis mucosae [27]. Important to know is that endoscopic biopsies may contain some limited fragments of muscle. These may originate from a newly formed muscularis mucosae or from the original muscularis mucosae. However, light microscopic distinction towards origin is impossible.

Page 31: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 15

The ‘double’ muscularis mucosae can only be visualized in (good quality) endoscopic mucosal resection (EMR) and operation specimens. Only invasion through the original, deeper muscularis mucosae is defined as “submucosal” invasion [28, 29].

3.2.3 Dysplasia in squamous epithelium

In squamous epithelium dysplasia is classified as mild, moderate, or severe. With increasing grades of dysplasia there is a progressive increase in dysplastic cells from the basal layer onwards untill the entire thickness of the epithelium is replaced. The latter state is described as carcinoma in situ [30]. When dysplastic cells reach the luminal aspect of the epithelium without cytoplasmic maturation the term carcinoma in situ can be used.

Some authors use a simplified classification for non-invasive squamous neoplastic lesions: low-grade intraepithelial neoplasia, which includes mild and moderate dysplasia, and high-grade intraepithelial neoplasia, which includes severe dysplasia and carcinoma in situ.

By definition, the basement membrane is intact in dysplasia. However, the junction of the epithelium with the underlying stroma may become irregular [31].

3.2.4 Dysplasia in columnar epithelium

From a biological point of view, the progression of precursors and precursor lesions into carcinomas is driven by the evolution and proliferation of clones of cells with accumulated genetic errors related to control of cell proliferation, intercellular adhesion, tumour suppression, etc., resulting in genomic instability.

Dysplasia is defined as ‘an unequivocal neoplastic epithelium confined within its basement membrane [32]. Furthermore, dysplasia has the potential to progress into invasive malignancy. Although morphological detection in mucosal biopsy specimens is still the best method of detecting patients at risk of developing cancer, it has its limitations:

INTRA-OBSERVER AND INTER-OBSERVER VARIABILITY

Numerous articles have been published on this topic demonstrating specific areas of discordance at the lower and higher end of the spectrum of early lesions. There is a significant degree of intra-observer and inter-observer variability in the diagnosis of dysplasia (unequivocal neoplastic atypia) versus reactive atypia even among experienced gastrointestinal pathologists [33-35]. Similarly, it has become apparent, especially from comparative studies between Western and Japanese pathologists, that the differential diagnosis between high-grade dysplasia, carcinoma in situ and intramucosal carcinoma is prone to intra-observer and inter-observer variability [36-38].

CLASSIFICATIONS

Diagnosis of dysplasia is based on the detection of morphological changes. According to the severity of histological changes, dysplasia has been graded using either a three tier system or a two tier system (Table 7). The changes were initially described as mild, moderate and severe dysplasia (morphological classification, 3-tier). In 1983, the Inflammatory Bowel Disease (IBD) study group classified dysplasia as negative, indefinite or positive, i.e. low and high-grade (clinical classification, 2-tier: low-grade = mild and moderate dysplasia; high-grade = severe dysplasia) (Table 7)) [32].

Grading in a two tier system would seem to be easier and more reproducible, and moreover to correlate with clinical implications. In the US and Western Europe, there was a general agreement that this clinically based classification was applicable to neoplastic changes in Barrett’s mucosa too [35, 39, 40]. In 2000, three new classifications for gastrointestinal dysplasia have been proposed: the Padova classification (gastric) [41], the Vienna classification [42] and the revised Vienna classification [43] (Table 7).

Page 32: Guideline pour la prise en charge du cancer oesophagien et ...

16 Upper GI Cancer KCE reports 75

These new classifications aimed at : 1) changing the terminology used, i.e. replacing dysplasia by (intra)epithelial neoplasia; 2) including not only dysplasia but also (invasive) carcinomas; and 3) distinguishing ‘mucosal high-grade neoplasia’ into high-grade dysplasia, suspicion for non-invasive or invasive carcinoma, non-invasive carcinoma, intramucosal carcinoma and carcinoma invading submucosa or beyond. The major differences amongst these classifications reside in the subcategories included/grouped and the figures attributed. Comparative studies using the new classifications have shown an improvement of the inter-observer variability especially between the Western and Japanese pathologists [36-38, 44]. Finally, a revision of the WHO classification of tumours of the digestive system was published at the end of 2000 [45]. The latter introduced ‘high-grade intraepithelial neoplasia’ (including severe dysplasia and carcinoma in situ), but did not recommend one or another of the previously mentioned classifications. Up till now, these new classifications have not yet gained widespread acceptance [25, 46]. Moreover, the authors of the Vienna classification stressed that the subdivisions related to ‘mucosal high-grade neoplasia’ (high-grade dysplasia, suspicion for invasive carcinoma, non-invasive carcinoma and intramucosal carcinoma) may be important for research purposes and may not be needed for clinical purposes [42]. Moreover, for resection specimens, only specific histological diagnoses should be given. Group classifications such as ‘mucosal high-grade neoplasia’ should not be used [44].

In conclusion, especially concerning the higher end of the spectrum of early lesions and in view of the importance of a multidisciplinary approach, it is important for a pathologist to have a clear understanding of the particular treatment regimens available and to be applied under particular circumstances. Classification is a chosen arrangement of elements in relation to a purpose. For the physician, a classification should be clinically relevant. Currently, the main clinical options are no follow-up, follow-up, local treatment by endoscopy, minimally invasive (laparoscopic) surgery and extensive surgery including lymph node dissection (see below). Many studies have evaluated the potential utility of immunohistochemical or molecular markers as additional techniques in detecting dysplasia, however with limited success. Additional confirmation by an expert pathologist (second opinion) is advocated, especially when therapeutic intervention is considered [25]. Today, the new classifications should be taken for what they are, i.e. attempts at an international level to reach consensus on histological diagnosis of dysplasia in ‘chosen arrangements of elements’ which may eventually generate guidelines for the development of diagnostic and management strategies.

Page 33: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 17

Table 7. Overview of classifications for dysplasia.

IBD Padova Vienna Revised Vienna TNM Negative for dysplasia

1. Negative for dysplasia 1.0. Normal 1.1. Reactive foveolar

hyperplasia 1.2. Intestinal metaplasia (IM) 1.2.1. IM complete type 1.2.1. IM incomplete type

1. No neoplasia 1. No neoplasia

Indefinite for dysplasia

2. Indefinite for dysplasia 2.1. Foveolar hyperplasia 2.2. Hyperproliferative IM

2. Indefinite for dysplasia 2. Indefinite for dysplasia

3. Non-invasive neoplasia (flat or elevated)

Low-grade dysplasia

3.1. Low-grade 3. Low-grade adenoma/dysplasia

3. Low-grade adenoma/dysplasia

High-grade dysplasia

3.2. High-grade 4. Non-invasive high-grade neoplasia

4. High-grade neoplasia

4.1. High-grade adenoma/dysplasia

4.1. High-grade adenoma/dysplasia

3.2.1. High-grade including suspicious carcinoma without invasion (intraglandular)

3.2.2. High-grade including carcinoma without invasion (intraglandular)

4.2. Non-invasive carcinoma (carcinoma in situ)

4.2. Non-invasive carcinoma (carcinoma in situ)

Tis

4. Suspicious for invasive carcinoma

4.3. Suspicious for invasive carcinoma

4.3. Suspicious for invasive carcinoma

5. Invasive adenocarcinoma 5. Invasive neoplasia 5.1. Intramucosal

carcinoma 4.4. Intramucosal carcinoma

T1a

5.2. Submucosal carcinoma (or deeper infiltration)

T1b

Key points

• There is no consensus about the definition of Barrett’s oesophagus.

• Several classifications are available for dysplasia. For the physician, the used classification should be clinically relevant.

Page 34: Guideline pour la prise en charge du cancer oesophagien et ...

18 Upper GI Cancer KCE reports 75

4 FINAL RECOMMENDATIONS ON OESOPHAGEAL CANCER

4.1 FLOWCHART

Page 35: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 19

4.2 DIAGNOSIS The diagnosis of oesophageal cancer should include a history and clinical examination. However, a diagnosis purely based on clinical grounds is unreliable [47]. Two diagnostic procedures, i.e. flexible upper gastrointestinal (GI) endoscopy and barium contrast radiology, are available. However, barium studies are less sensitive than endoscopy for the diagnosis of early malignancy and they cannot reliably diagnose premalignant lesions including dysplasia. Therefore, flexible upper GI endoscopy with at least biopsies of all suspicious lesions is recommended as the diagnostic procedure of choice in patients with suspected oesophageal cancer [47]. Nevertheless, barium studies can be useful to determine the oesophageal or gastric extent of a tumour, to confirm the presence of a tracheo-oesophageal fistula or to document complete luminal obstruction [48].

The following alarm symptoms are associated with oesophageal (and gastric) cancer and should lead to a referral for early endoscopy and biopsies: dysphagia, recurrent vomiting, anorexia, weight loss and gastrointestinal blood loss [47, 49]. The general practitioner has an important role in recognizing these symptoms. In a recent prospective study, Bowrey et al. found a sensitivity of 85% using alarm symptoms for the referral for endoscopy [49]. This means that 15% of patients with oesophageal cancer would not have been referred for initial endoscopic assessment because of the absence of alarm symptoms. Importantly, patients with alarm symptoms had a significantly more advanced tumour stage (metastatic disease in 47% vs. 11%; p < 0.001), were less likely to undergo surgical resection (50% vs. 95%; p<0.001), and had a poorer survival (median, 11 vs. 39 months; p = 0.01) than patients without such symptoms [49]. Although odynophagia was not discussed in this study, it can be used as an additional alarm symptom of oesophageal cancer warranting endoscopic evaluation [50].

Patients at high risk for developing oesophageal cancer (Table 8) may benefit from screening and follow-up with chromoendoscopy (methylene blue, indigo carmine, lugol, etc.) [47, 51]. In a recent RCT, diagnostic accuracy of autofluorescence endoscopy (AFE) combined with four-quadrant biopsies was higher than the diagnostic accuracy of conventional endoscopy with four-quadrant biopsies (one additional high�grade dysplasia or adenocarcinoma for every 23 patients undergoing AFE vs. 1 in 93 patients undergoing conventional endoscopy) [52]. On the other hand, methylene-blue directed biopsies were found to be less sensitive in detecting dysplasia than random four-quadrant biopsies in Barrett’s esophagus [53].

High-resolution endoscopy (HRE) is a fairly new endoscopic imaging technique with the potential to improve the detection of early neoplasia in Barrett’s oesophagus [48]. One RCT was identified comparing indigo carmine chromoendoscopy and narrow�band imaging as adjuncts to HRE [54]. Both techniques were found to have a comparable diagnostic accuracy. No studies were found comparing HRE to conventional techniques [48].

Page 36: Guideline pour la prise en charge du cancer oesophagien et ...

20 Upper GI Cancer KCE reports 75

Table 8. Features associated with high risk for developing oesophageal cancer.

Clinical features:

• Alcohol and tobacco abuse

• Longstanding gastro-oesophageal reflux disease

• Neck, mouth and head cancer

• Previous oesophageal cancer

• Barrett’s oesophagus

• Caustic injury

• Papilloma

• Genetic diseases

• Various: Achalasia, Plummer-Vinson, Celiac disease

Endoscopic features:

• stricture

• ulceration

• nodularity

Histological features:

• high-grade dysplasia

• multifocal dysplastic lesions, persisting after adequate conservative treatment for 6-8 weeks

Recommendations

1. Patients presenting with any of the following alarm symptoms within the clinical context of potential oesophageal pathology should be referred for early endoscopy and biopsies: dysphagia, recurrent vomiting, anorexia, weight loss, gastrointestinal blood loss (1C recommendation).

2. Flexible upper gastrointestinal endoscopy with at least biopsies of all suspicious lesions is recommended as the diagnostic procedure of choice in patients with suspected oesophageal cancer (1C recommendation).

3. High-resolution endoscopy (HRE) and chromoendoscopy is not routinely recommended, but may be of value in screening and follow-up of high-risk patients (2C recommendation).

4.3 WORK-UP OF DYSPLASTIC LESIONS Evidence from a few RCTs [47] and one low-quality systematic review [55] suggests that anti-reflux surgery is not indicated in patients with Barrett’s oesophagus to reduce the risk of progression to adenocarcinoma.

Patients diagnosed with high-grade dysplasia should have careful endoscopic assessment [47]. However, dysplastic lesions in patients with Barrett’s oesophagus are difficult to detect using conventional endoscopy. Recent studies suggest that high-resolution endoscopy (HRE) – with or without chromoendoscopy –, narrow band imaging (NBI) or autofluorescence may improve the detection of dysplastic lesions (see above) [56].

Page 37: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 21

In patients with Barrett's oesophagus, a structured biopsy protocol with quadrantic biopsies every two centimetres and biopsy of any visible lesion is recommended. In the presence of dysplasia, quadrantic biopsies should be taken every 1 cm during surveillance and control endoscopies [57, 58]. In addition to the endoscopic assessment, patients with high-grade dysplasia should have a careful pathological assessment [47].

Evaluation of suspected high-grade dysplasia in Barrett's oesophagus biopsies should be undertaken with knowledge of the clinical and endoscopic background. Pathologists should follow a classification for reporting dysplasia that the multidisciplinary team is familiar with. An overview of classifications is already provided in chapter 3.2.4. Where therapeutic intervention is contemplated on the basis of high-grade dysplasia, the diagnosis should be validated by a second pathologist experienced in this area because of the significant inter- and intra-observer variation [47, 56]. Further biopsies or diagnostic endoscopic mucosal resection (EMR) should be done if there is uncertainty. Biopsies should also be reviewed at a multidisciplinary meeting with access to the clinical information.

Finally, as the diagnosis, treatment and follow-up of patients with oesophageal high-grade dysplastic lesions is very specific and new techniques rapidly become available, these patients should be referred to centres or network reference centres with the appropriate endoscopic and surgical expertise and facilities [47]

Recommendations

4. Reduction of risk of progression to adenocarcinoma is not an indication for anti-reflux surgery in patients with Barrett's oesophagus (2A recommendation).

5. In patients with Barrett's oesophagus there should be a structured biopsy protocol with quadrantic biopsies every two centimetres and biopsy of any visible lesion (1C recommendation).

6. Pathologists should follow a classification for reporting dysplasia that the multidisciplinary team is familiar with (1C recommendation).

7. Evaluation of suspected high-grade dysplasia in Barrett's oesophagus biopsies should be undertaken with knowledge of the clinical and endoscopic background (1C recommendation).

8. Patients confirmed with high-grade dysplasia should have careful endoscopic and pathological assessment (1C recommendation).

9. High-resolution endoscopy +/- chromoendoscopy as well as every 1 cm qaudrantic biopsies is recommended in patients with a dysplastic or neoplastic lesion in a Barrett's oesophagus (1C recommendation).

10. Where therapeutic intervention is contemplated on the basis of high-grade dysplasia, the diagnosis should be validated by a second pathologist experienced in this area and further biopsies or eventually diagnostic endoscopic mucosal resection (EMR) should be done if there is uncertainty (1C recommendation).

11. Treatment options for patients with high-grade dysplasia should be discussed at a multidisciplinary meeting with access to the clinical and pathological information (expert opinion).

12. Patients with high-grade dysplasia should be referred to centres or network reference centres with the appropriate endoscopic and surgical expertise and facilities (1C recommendation).

Page 38: Guideline pour la prise en charge du cancer oesophagien et ...

22 Upper GI Cancer KCE reports 75

4.4 STAGING In patients with oesophageal cancer, CT scan of the chest and abdomen with intravenous contrast and gastric distension with oral contrast or water should be performed routinely. The liver should at least be imaged in the arterial and portal venous phase [47]. Slice thickness should be 5 mm or less [56]. The main contribution of CT scan to the staging of oesophageal cancer is the detection of distant metastases and gross invasion of adjacent structures/organs [47, 56, 59]. If metastatic disease is detected with CT scan, curative treatment is excluded and additional staging with EUS and/or PET scan is unnecessary. CT scan has also an acceptable diagnostic accuracy for locoregional staging (T and N), although EUS is clearly more sensitive (see below) [47, 56]. Given the high incidence of cervical lymph node metastasis [60], CT scan should also include the cervical region.

EUS has emerged as the imaging technique of choice for locoregional staging [47, 56], with a diagnostic accuracy ranging from 53-94% (median 83%) for T-staging and from 54-94% for N-staging (median 76%) [61]. In stenotic tumours, the accuracy of EUS is further improved with the use of dilation, which in most cases permits passage of the endoscope [62, 63]. However, this is associated with a risk of perforation [64]. Importantly, most studies dealing with the accuracy of N-staging only assess the yes-no possibility of positive lymph nodes, without correlating the EUS findings of a positive lymph node to the histopathological findings of that particular node. As a result, although a final pathology report may conclude N1 disease to be present, a node with positive EUS findings may be histopathologically negative and vice versa. Therefore, the diagnostic accuracy of EUS for N-staging may be overestimated.

The accuracy of N staging with EUS is further improved with fine needle aspiration cytology (FNAC) [47, 56]. FNAC needs to be interpreted by an experienced pathologist.

Neck imaging with ultrasonography or CT enables the detection of metastatic lymph nodes that are clinically not palpable [47] or can be used to guide fine needle aspiration of suspicious lymph nodes with very high accuracy and specificity [56, 65].

In the absence of clear metastatic disease on conventional CT/EUS/cervical US, PET(/CT) may provide valuable additional information, in particular when considering multimodality treatment with curative option. In such cases, baseline PET(/CT) allows better response assessment as compared to conventional CT/EUS/cervical US. FNCLCC made specific recommendations on the use of PET scan in the diagnosis and staging of oesophageal cancer [66]. In addition, the KCE recently published an HTA report on the use of PET scan [67]. Since this HTA report, numerous observational studies of variable quality have been published [68-75]. For initial staging of patients without apparent metastases on CT scan, especially in locally advanced disease (cT>2 or cN1 or cN doubtful), PET(/CT) is useful for detecting positive lymph nodes and distant metastatic disease [67]. However, in early-stage disease (cT1-2N0) the probability that PET(/CT) will significantly upstage disease is low.

Magnetic resonance imaging (MRI) was not found to be superior to conventional imaging techniques such as CT scan ([47, 56]. Therefore, MRI should be reserved for those patients who cannot undergo CT or used for additional investigation following CT and/or EUS.

In patients with clinical or imaging features suspicious of tracheobronchial invasion (e.g. cough aggravated by swallowing in case of a fistula) or extrinsic compression, bronchoscopy combined with bronchoscopic ultrasound (BUS)

and/or biopsy can be useful [47, 76, 77]. The underlying evidence, however, is weak to very weak. Observational data also provide weak evidence for the occurrence of synchronous neoplasms of the bronchial tree in patients with squamous cell carcinoma of the oropharynx and the oesophagus, which is an indication for bronchoscopy [78].

Few studies are available to support the use of invasive staging with thoracoscopy and laparoscopy (+/- peritoneal cytology and/or laparoscopic ultrasound) [47, 56, 79].

Page 39: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 23

Thoracoscopy can be useful for the detection of positive mediastinal lymph nodes, while laparoscopy can be considered in patients with oesophageal tumours with a gastric component [47]. Laparoscopy has a higher specificity for M staging in comparison with CT scan, but obviously carries a higher risk of morbidity [56].

Recommendations

13. In patients with oesophageal cancer, CT scan of the chest (including lower neck region) and abdomen with intravenous contrast and gastric distension with oral contrast or water should be performed routinely. The liver should at least be imaged in the arterial and portal venous phase (1C recommendation).

14. Patients with oesophageal or gastro-oesophageal junction cancers who are candidates for any curative therapy should have their tumours staged with endoscopic ultrasonography +/- fine needle aspiration cytology (FNAC) and ultrasonography of the neck (1B recommendation).

15. Fine needle aspiration cytology (FNAC) needs to be interpreted by an experienced pathologist (expert opinion).

16. In patients with oesophageal cancer and an option for curative treatment after conventional staging (CT/endoscopic ultrasonography), PET(/CT) scan may be considered for the staging of lymph nodes (loco-regional, distal or all lymph nodes) and distant sites other than lymph nodes (1C recommendation).

17. The following examinations can be considered for specific indications: MRI, bronchoscopy +/- bronchial ultrasonography (BUS) +/- biopsy, thoracoscopy, or laparoscopy (1C recommendation).

4.5 TREATMENT OF MUCOSAL CANCER The principles of anatomopathological evaluation of mucosal cancer are similar to those of high-grade dysplasia, including validation of the diagnosis by a second pathologist, additional biopsies or diagnostic EMR in case of uncertainty, and discussion of treatment options at the MDT [47].

Only observational studies are available comparing endoscopic mucosal resection (EMR) to surgery for the treatment of superficial oesophageal cancer [56]. Both treatments were found to be equally effective, but EMR was associated with less complications. Therefore, superficial oesophageal cancer limited to the mucosa should be treated with EMR, taking into account the stage, size, length of Barrett, histological type, differentiation grade, and lymphovascular invasion [47]. For example, in case of mucosal cancer in a long Barrett’s segment, complete resection of the Barrett’s area with EMR is unlikely. In these cases, surgery (e.g. gastric pull-up or vagal sparing oesophagectomy with colon interposition) may remain the treatment of choice.

En-bloc resection – allowing better pathology staging of deep and lateral margins – should be aimed at with the appropriate technique (EMR for lesions less than 12 mm) [80-82].

Mucosal ablative techniques, such as argon plasma coagulation (APC), photodynamic therapy (PDT) or laser, are investigational [47, 56]. Only small observational studies showed the benefits of these techniques, and they should therefore be limited to units with appropriate expertise.

Moreover, APC was associated with buried neoplastic glands and should not be considered for curative treatment. Importantly, PDT is associated with a risk of stricture formation in up to 30% of patients [83]. Above this, mucosal ablative techniques make an anatomopathological control of the staging results impossible.

Page 40: Guideline pour la prise en charge du cancer oesophagien et ...

24 Upper GI Cancer KCE reports 75

Recommendations

18. Where therapeutic intervention is considered for a supposedly T1a mucosal cancer, the diagnosis should be validated by a second pathologist experienced in this area. Further biopsies or eventually diagnostic endoscopic mucosal resection (EMR) should be done if there is uncertainty (1C recommendation).

19. Treatment options for patients with mucosal cancer should be discussed at a multidisciplinary meeting with access to the clinical and pathological information (expert opinion).

20. Superficial oesophageal cancer limited to the mucosa should be treated with endoscopic mucosal resection (EMR), taking into account well-defined criteria relating to stage, size, length of Barrett, histological type, differentiation grade, and lymphovascular invasion (1C recommendation).

21. Mucosal ablative techniques, such as argon plasma coagulation (APC), photodynamic therapy (PDT) or laser, are investigational and should be limited to units with appropriate expertise (1C recommendation).

4.6 TREATMENT OF CANCER BEYOND THE MUCOSA

4.6.1 Neoadjuvant treatment

In 2006, Cancer Care Ontario (CCO) published a high-quality CPG (including meta-analyses) on the use of neoadjuvant treatment for oesophageal cancer [84]. Three additional meta-analyses [85-87] and one systematic review [88] have been published since then. Only Gebski et al. found new evidence in addition to that presented by CCO [86].

CCO identified 6 RCTs comparing preoperative radiotherapy and surgery vs. surgery alone [84]. No significant difference was detected in the risk of death at one year (RR 1.01; 95%CI 0.88 – 1.16; p=0.90). These results are in line with those of Arnott et al., who found a hazard ratio (HR) of 0.98 (95%CI 0.78 – 1.01), suggesting a small but non-significant absolute survival benefit of 3% at 2 years and 4% at 5 years in favour of preoperative radiotherapy [85].

Nine different RCTs were identified by CCO comparing preoperative chemotherapy and surgery vs. surgery alone [84]. In addition, two RCTs were identified comparing preoperative and postoperative chemotherapy and surgery vs. surgery alone. All eleven trials were also included in a recent Cochrane review [87]. A meta-analysis of the first 9 RCTs found a pooled RR of 1.00 (95%CI 0.83 – 1.19; p=0.97), detecting no difference in survival at one year [84]. These results are in line with the Cochrane review (HR 0.88; 95%CI 0.75 – 1.04). Trials also reported risks of toxicity with chemotherapy that ranged from 11% to 90% [87].

Gebski et al. identified 10 RCTs (of which one was an unpublished thesis, and two were published as an abstract) comparing preoperative CRT and surgery vs. surgery alone [86]. A HR for all-cause mortality of 0.81 (95%CI 0.70 – 0.93; p=0.002) in favour of preoperative CRT was found, corresponding to a 13% absolute difference in survival at 2 years. Patients with adenocarcinoma seemed to obtain the highest benefit, although conflicting evidence is available.

These results are not in line with those presented by CCO, who found no difference in one-year survival between the 2 treatment arms (RR 0.91; 95%CI 0.79 – 1.06; p=0.21) [84]. CCO also identified 1 RCT comparing preoperative CRT (bleomycin + radiotherapy) to preoperative radiotherapy alone. No significant difference in survival between the two treatment groups was found [84].

Page 41: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 25

In a systematic review of 6 RCTs by Graham et al., a RR of 0.81 (95%CI 0.64 – 1.02) compared to surgery alone was found. This corresponded to an additional 40 days of quality-adjusted life expectancy [88].

Since the literature search of CCO and Gebski et al., results of two additional small RCTs became available, comparing preoperative CRT (cisplatin + 5-FU) to surgery alone [89, 90]. No significant difference in survival was found.

Based on the evidence presented, the advantage of neoadjuvant treatment is small and although neoadjuvant treatment is frequently used, it cannot be considered as a routine practice in patients with oesophageal cancer. It is clear that for each individual patient the need for neoadjuvant treatment should be discussed during a multidisciplinary meeting. If neoadjuvant treatment is part of the treatment plan, it should be administered according to a predefined protocol. This protocol should also specify the role of PET(/CT) scan in the evaluation of treatment response to neoadjuvant treatment. In a previous KCE report, some evidence was identified supporting the role of PET scan in the assessment of treatment response to neoadjuvant treatment, using the initial staging PET results as a comparison [67]. These conclusions are confirmed by subsequent prospective trials [74, 91]. Finally, in all trials, a complete pathological response was associated with the most favourable prognosis.

Since the use of neoadjuvant treatment for patients with oesophageal cancer still is controversial, it is recommended to prospectively register clinical outcomes and adverse events of the combined treatment. In fact, besides the mandatory cancer registration, prospective registration of clinical outcomes and adverse events is mandatory for all oncologic patients according to the Royal Decree of March 21st 2003 settling the norms for the healthcare programme in oncology [92].

Recommendations

22. Preoperative radiotherapy alone is not recommended for patients with oesophageal cancer (2A recommendation).

23. Neoadjuvant treatment is not routinely indicated for patients with oesophageal cancer (2A recommendation).

24. The need for neoadjuvant treatment should be discussed during a multidisciplinary meeting (expert opinion).

25. Prospective registration of clinical outcomes and adverse events of combined treatment is recommended (expert opinion).

4.6.2 Surgical treatment

Surgical resection remains the standard treatment for patients with resectable oesophageal cancer [47, 56, 84, 88]. Patients are considered to have unresectable disease in case of metastatic disease, positive lymph nodes outside the region of surgery and/or radiotherapy (i.e. the cervical, thoracic and upper abdominal compartment), advanced locoregional disease (e.g. tracheo-oesophageal fistula), or severe comorbidity.

Until now, only few studies are available comparing surgery to other treatment modalities [56, 93, 94]. One moderate-quality eastern RCT compared primary chemoradiotherapy (CRT) and salvage surgery to primary ‘standard’ oesophagectomy [94]. No significant difference in 2-year survival and disease-free survival was found between the 2 treatment groups. Six out of 36 patients (17%) assigned to primary CRT were treated with salvage oesophagectomy [94]. A French study compared CRT followed by surgery to CRT alone in patients with operable oesophageal cancer that responded to CRT [93]. Two-year survival and 2-year local recurrence rate did not differ significantly.

The results of these two RCTs need to be interpreted with caution, since they had important methodological limitations (no accurate information on randomisation procedure, absence of blinding). Above this, as in many other trials, surgery was not standardized [93, 94].

Page 42: Guideline pour la prise en charge du cancer oesophagien et ...

26 Upper GI Cancer KCE reports 75

Surgery for oesophageal cancer should be aimed at achieving an R0 resection, and should be considered preferentially through an en bloc resection (i.e. with systematic abdominal and mediastinal lymph node dissection, the so-called two field lymphadenectomy) [47, 56]. This recommendation is mainly based on the results of 1 RCT that compared extended transthoracic resection with two-field lymphadenectomy to limited transhiatal resection without two-field lymphadenectomy [95]. A trend towards improved 5-year survival was found in favour of the extended thoracic group.

The goal of lymphadenectomy is to optimize staging and to improve long-term outcomes by decreasing the risk for local recurrence and increasing the probability of an R0 resection [47, 56]. Two-field lymphadenectomy involves the clearance of mediastinal (1st field) and abdominal (2nd field) lymph nodes, while three-field lymphadenectomy also involves cervical lymph node clearance (3rd field). The extent of two-field lymphadenectomy is specified in relation to the extent of the mediastinal part of the lymphadenectomy. Standard two-field lymphadenectomy involves the clearance of lymph nodes from the carina down to the diaphragm. In extended two-field lymphadenectomy, clearance is expanded to the right paratracheal and recurrent nerve, while in total two-field lymphadenectomy clearance is further extended to the left recurrent nerve [96]. Extensive two-field lymphadenectomy should be standard during oesophagectomy [47]. Three-field lymphadenectomy is considered strictly investigational, especially in distal third adenocarcinoma [60]. Indeed, data from Western studies with three-field lymphadenectomy are scarce and are based on relatively small sample sizes [60, 97].

Many studies have shown a relationship between patient outcomes (e.g. 30-day mortality) and surgeon or hospital volume [47, 56, 98, 99]. Recently, a meta-analysis of 13 studies calculated that the experience of 20 oesophagectomies per year is needed to significantly reduce postoperative mortality [100]. Therefore, oesophageal cancer surgery should be carried out in high volume specialist surgical units by surgeons with experience in oesophageal and GOJ cancer [47]. Recently, it was shown that specialty training in thoracic surgery has an independent association with lower mortality after oesophageal resection (adjusted mortality 12.4% vs. 16.5%; p = 0.01), although specialty training appeared to be less important than hospital and surgeon volume [101]. Of course, specialty training and high volume are closely related.

Recommendations

26. Surgical resection is considered standard treatment for patients with resectable oesophageal cancer (1A recommendation).

27. Surgery for oesophageal cancer should be aimed at achieving an R0 resection, and should be considered preferentially through a transthoracic en bloc resection (1A recommendation).

28. Extensive two-field lymphadenectomy should be standard during oesophagectomy to improve staging, local disease control and potentially cure rate (1C recommendation).

29. Three-field lymphadenectomy is strictly investigational (2C recommendation).

30. Oesophageal cancer surgery should be carried out in high volume specialist surgical units by surgeons with experience and/or specialist training in oesophageal and gastro-oesophageal junction cancer (1C recommendation).

Page 43: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 27

4.6.3 Adjuvant treatment

CCO provided an excellent overview of the literature concerning adjuvant treatment for resectable oesophageal cancer [84]. Three RCTs of postoperative chemotherapy (all cisplatin-based regimens) and surgery vs. surgery alone were identified. Pooled results of 2 RCTs [102, 103] showed no significant difference in the risk of death at 3 years (RR 0.94; 95%CI 0.74 – 1.18; p=0.60) [84]. A third RCT also found no survival benefit for postoperative chemotherapy, but 3-year survival was not reported [104].

CCO identified 5 RCTs of postoperative radiotherapy and surgery vs. surgery alone [84]. Meta-analysis of these trials showed no significant difference in the risk of death with postoperative radiotherapy and surgery at one year compared with surgery alone (RR 1.23; 95%CI 0.95 – 1.59; p=0.11). The rate of local recurrence with radiotherapy was lower in two RCTs, but this benefit was achieved at the expense of increased morbidity [105, 106]. The most recent RCT found significantly improved 5-year survival rates with adjuvant radiotherapy for stage III patients (35% vs. 13%; p=0.0027) [107]. However, this trial was hampered by some important methodological drawbacks (no informed consent, no intention-to-treat analysis).

No RCTs on postoperative combined chemoradiotherapy vs. surgery alone were identified by CCO [84]. A search for RCTs published since the CCO report also did not identify new evidence.

There is also no available evidence on whether adjuvant therapy is of real benefit in R1 or R2 resection.

Recommendations

31. Postoperative adjuvant chemotherapy is not recommended for patients with oesophageal cancer (2A recommendation).

32. Postoperative adjuvant radiotherapy is not recommended for patients with oesophageal cancer (2A recommendation).

33. Postoperative adjuvant chemoradiotherapy is not recommended for patients with oesophageal cancer (expert opinion).

4.6.4 Non-surgical treatment with curative intent

Definitive CRT should be considered in patients with oesophageal cancer who have locally advanced disease and are unfit for surgery, or in patients who decline surgery [47, 108]. Recently, a moderate-quality eastern RCT found no difference in survival at 2 years between primary CRT (and salvage surgery) and primary surgery in patients with potentially resectable oesophageal cancer (see above) [94]. Bedenne et al. also found no difference in survival at 2 years between CRT followed by surgery and CRT alone in patients with operable oesophageal cancer that responded to CRT (see above) [93]. Once again, the need for an adequate method to predict treatment response is highlighted.

Definitive CRT should also be considered for patients with cervical oesophageal cancer, for whom it offers the benefit of preserving the larynx [56]. However, no RCTs are available comparing surgery to CRT for these patients.

Concomitant CRT is recommended over radiotherapy alone. In their systematic review, CCO included 8 RCTs of concomitant CRT vs. radiotherapy alone, of which 7 RCTs had 1-year mortality rates available. Pooled analysis showed an odds ratio (OR) of 0.61 (95%CI 0.44 – 0.84; p<0.00001) in favour of concomitant CRT [108]. When only concomitant CRT trials using cisplatin-based chemotherapy regimens were included, a statistically significant survival benefit was detected at one year (OR 0.54; 95%CI 0.36 – 0.82; p=0.003) [108, 109]. Zhao et al. recently published 5-year results of a trial comparing late course accelerated hyperfractionation (LCAF) radiotherapy combined with chemotherapy vs. LCAF radiotherapy alone [110]. A trend toward better survival was found in patients who received combination therapy.

Page 44: Guideline pour la prise en charge du cancer oesophagien et ...

28 Upper GI Cancer KCE reports 75

Sequential radiotherapy and chemotherapy is not recommended as standard practice [108]. CCO identified 5 RCTs of sequential radiotherapy and chemotherapy vs. radiotherapy alone. Pooled analysis revealed no significant difference in survival between the treatment groups at one year.

Patients should be made aware of the increased acute toxicity associated with the use of concomitant CRT. Therefore, the decision to use concomitant CRT should only be made after careful consideration of the potential risks, benefits, and the patient’s general condition [108].

In patients with locally advanced infracarinal oesophageal tumours that do not respond to primary CRT, curative salvage surgery remains an option provided the patient is fit for surgery [111]. Piessen et al. found a R0 resection rate of 62.2% in these patients. Overall survival was significantly higher in the R0 resection group compared with the incomplete resection group (18.4 vs. 8.6 months, p<0.001). Tumours < 5 cm and with < 90° aortic contact had the highest chance of obtaining an R0 resection.

Recommendation

34. Definitive chemoradiotherapy should be considered in patients with oesophageal cancer who have locally advanced disease that is considered unresectable, in patients who are unfit for surgery, or in patients who decline surgery (1A recommendation). It can also be considered for patients with cervical oesophageal cancer in order to preserve the larynx (1C recommendation).

4.7 PALLIATIVE TREATMENT & METASTATIC DISEASE Endoscopic therapy has an important role in the control of obstruction caused by oesophageal cancer. Different treatment options are available, such as stent placement, laser or argon plasma coagulation (APC) therapy. Treatment must be individualized and should depend on the local availability and expertise [47].

Partially covered self-expanding metal stents (SEMS) or plastic expandable stents are the best options for palliation of dysphagia caused by oesophageal cancer, especially in patients with a short life expectancy [47, 112, 113]. Both types have a highly successful insertion rate, but SEMS are associated with fewer procedure-related complications and a trend toward better quality of life [48]. On the other hand, plastic stents are associated with lower costs. In case of tumour ingrowth or overgrowth in stented patients, laser therapy, APC therapy or re-stenting should be considered [47]. However, the use of APC therapy is supported by observational studies only.

The use of oesophageal dilatation alone should be avoided [47]. Dilation can be used as an adjunct to other treatment modalities, such as by facilitating placement of an oesophageal stent.

No RCTs are available to support the use of palliative surgery in patients with oesophageal cancer [47]. Observational studies showed associated high morbidity and decreased quality of life in selected patients. Therefore, oesophagectomy (transthoracic or transhiatal) should not be performed routinely with palliative intent in patients with oesophageal cancer [47]. In carefully selected patients that are fit for surgery and having recurrent upper GI bleeding or a covered perforation that induces severe back pain that is unlikely to be relieved by any other therapy, oesophagectomy can be considered. Since substernal bypass is also associated with high morbidity and mortality, it should not be performed with palliative intent in patients with oesophageal cancer [47].

In a recent Cochrane review, 7 RCTs on chemotherapy for patients with metastatic oesophageal cancer were identified [114]. No survival benefit was found in the 2 RCTs comparing chemotherapy with best supportive care. In the five RCTs comparing different chemotherapy regimens, no consistent benefit of any specific regimen was found [114]. No RCTs or systematic reviews were identified supporting the use of CRT in the palliative setting of oesophageal cancer [47].

Page 45: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 29

Therefore, in patients with locally advanced or metastatic cancer of the oesophagus, chemotherapy or CRT are treatment options that should be discussed in the MDT [56, 114].

The use of external-beam radiotherapy for the palliation of dysphagia and pain is supported by observational studies only [47]. In 1 RCT, endoluminal brachytherapy resulted in better dysphagia control than stent placement in patients who lived longer than 140 days [47]. Therefore, it should be considered in patients with dysphagia from oesophageal cancer with the perspective of a more prolonged survival.

Patients with oesophageal cancer should have access to a specialist (outpatient and/or inpatient) palliative care team, in particular in relation to comfort and symptom control, and quality of life [47]. This team clearly should involve the general practitioner, who should have a coordinating role in the organisation of the palliative home care.

Evidence specific to control of symptoms, such as pain, anorexia and bleeding, in these cancers is limited. Some evidence exists for the use of celiac axis plexus block for the palliation of pain and for the use of corticosteroids in patients with oesophageal cancer who are anorexic or who have symptoms of bowel obstruction [47]. Nutritional support in the palliative setting should take into account the comfort of the patient, in that quality of life is much more important than the long-term effects [56].

Limited evidence (coming from a secondary analysis of 1 RCT) is also available for psychotherapeutic support during hospital stay [115].

Recommendations

35. Control of obstruction caused by oesophageal cancer should be obtained with stent placement or laser/ argon plasma coagulation (APC) therapy, depending on the local availability and expertise (1A recommendation).

36. Partially covered self-expanding metal stents or plastic expandable stents are the best options for palliation of dysphagia caused by oesophageal cancer (1B recommendation).

37. Laser therapy, argon plasma coagulation (APC) therapy or re-stenting should be considered for control of tumour ingrowth or overgrowth in stented patients (1C recommendation).

38. The use of oesophageal dilatation alone should be avoided (2C recommendation).

39. Oesophagectomy (transthoracic or transhiatal) should not be performed with palliative intent in patients with oesophageal cancer (1C recommendation).

40. Substernal bypass for oesophageal cancer should not be performed with palliative intent (1C recommendation).

41. In patients with locally advanced or metastatic cancer of the oesophagus, chemotherapy or chemoradiotherapy are treatment options that should be discussed in the multidisciplinary team (2A recommendation).

42. Palliative external-beam radiotherapy or endoluminal brachytherapy should be considered in patients with dysphagia from oesophageal cancer and with the perspective of a more prolonged survival (2C recommendation).

43. Patients with oesophageal cancer should have access to a specialist palliative care team, in particular in relation to comfort and symptom control, nutrition and quality of life (1C recommendation).

Page 46: Guideline pour la prise en charge du cancer oesophagien et ...

30 Upper GI Cancer KCE reports 75

4.8 FOLLOW-UP In patients that underwent treatment for oesophageal cancer, follow-up is needed to detect recurrent disease (in case of curative treatment), progressive disease, symptoms that warrant treatment, or nutritional problems. However, evidence about the duration, frequency and type of follow-up is unavailable [56]. Therefore, published guidelines on the follow-up of patients with oesophageal cancer are always consensus-based.

A physical examination is recommended every three months, and should give special attention to nutritional status, weight loss, fatigue, etc. Vitamin B12 evaluation can be done, but deficiency after oesophagectomy occurs less frequently than after total gastrectomy. A CT scan of the chest and abdomen is recommended every six months in the first year and afterwards annually until the fifth year. However, it should be stressed that no evidence is available to support regular imaging in the follow-up of these patients [47].

After endoscopic treatment of dysplastic lesions or mucosal cancer, endoscopic follow-up is similar to that of patients with Barrett’s oesophagus [56]: three months after endoscopic treatment, then every six months in the first two years, and then annually.

Recommendations

44. It is recommended that the follow-up of patients treated for oesophageal cancer includes a physical examination every three months, and a CT scan every six months in the first year and afterwards annually until the fifth year (expert opinion).

45. Patients treated with endoscopic mucosal resection (EMR) should have a follow-up endoscopy after three months, then every six months in the first two years, and then annually (expert opinion).

4.9 RECURRENT DISEASE In a small number of patients, recurrent disease can be successfully treated with curative intent [116-118]. In patients with a localised lymph node recurrence [119-121] or a localised anastomotic recurrence [122], a combined chemoradiotherapy regimen or occasionally redo-surgery can be considered after discussion by the MDT. Patients who develop a solitary lung or liver metastasis can also be considered for resection. The evidence for these treatments, however, is weak and coming from small observational studies only.

When patients are confronted with a local recurrence or a new tumour after EMR for a mucosal cancer, treatment options include local treatment or multimodality treatment [123]. These options should be discussed in the MDT. Again, only small observational studies are available to support these treatments.

Recommendations

46. In patients with recurrent oesophageal cancer, treatment options should be discussed in the multidisciplinary team (expert opinion).

47. In patients with a local recurrence or new tumour after endoscopic mucosal resection (EMR), treatment options, including local treatment, should be discussed in the multidisciplinary team (expert opinion).

Page 47: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 31

5 FINAL RECOMMENDATIONS ON GASTRIC CANCER

5.1 FLOWCHART

Page 48: Guideline pour la prise en charge du cancer oesophagien et ...

32 Upper GI Cancer KCE reports 75

5.2 DIAGNOSIS As for oesophageal cancer, in addition to a history and clinical examination, flexible upper GI endoscopy with at least biopsies of all suspicious lesions is recommended as the diagnostic procedure of choice in patients with suspected gastric cancer [47]. The same alarm symptoms are associated with gastric cancer and should lead to a referral for early endoscopy and biopsies: dysphagia, recurrent vomiting, anorexia, weight loss and gastrointestinal blood loss (see above) [47, 49].

In a prospective trial involving 202 consecutive patients (including 47 with oesophageal or gastric carcinomas), seven biopsy and cytology specimens were found to yield the correct diagnosis in all patients [124]. However, in patients with large gastric folds (suggestive of linitis plastica or gastric lymphoma), at least 10 biopsies are recommended, together with an additional technique, such as large forceps biopsy.

As in patients at high risk for oesophageal cancer, patients at high risk for developing gastric cancer (Table 9) may also benefit from screening and follow-up with HRE and/or chromoendoscopy [47], although the supporting evidence is scarce. No recent RCTs were identified. Mainly Japanese observational studies have shown that chromoendosocpy increases the rate of superficial and “early” lesions detection [125].

Patients with a familial history of gastric cancer should be referred to a centre for genetic counselling.

No evidence was found on the most appropriate frequency of surveillance. Frequency of surveillance endoscopy depends on specific risk factors, e.g. every 3-5 years in patients with autoimmune chronic gastritis, annual follow-up for patients with low-grade dysplasia in atrophic gastritis, etc.

Table 9. Features associated with high risk for developing gastric cancer.

Histological features:

• Dysplasia

• Gastric atrophy with or without intestinal metaplasia

• Adenomatous polyps

Patient’s history:

• Previous gastric cancer

• History of partial gastrectomy

• Familial history of gastric cancer

Clinical features:

• Refractory gastric ulcer

• Pernicious anaemia

In patients with suspected gastric cancer or at high risk for developing gastric cancer, H. pylori testing should be systematically done on histology and ideally with a second test (rapid urease test, urea breath test, culture or serology) [126-128]. In case of a negative histology and negative additional testing, a positive serological test suggests the presence of an unrecognised H. pylori infection, and additional investigations to confirm whether the serological test is false positive or reflects active or recent infection should be carried out [128].

Page 49: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 33

Recommendations

1. Patients presenting with any of the following alarm symptoms within the clinical context of potential gastric pathology should be referred for early endoscopy and biopsies: dysphagia, recurrent vomiting, anorexia, weight loss, gastrointestinal blood loss (1C recommendation).

2. Flexible upper gastrointestinal endoscopy with at least biopsies of all suspicious lesions is recommended as the diagnostic procedure of choice in patients with suspected gastric cancer (1C recommendation).

3. High-resolution endoscopy and chromoendoscopy may be of value in screening and follow-up of high-risk patients (2C recommendation).

4. H. pylori testing should be systematically done on histology and ideally with a second test. Serology should be considered if gastric sampling remains negative (2C recommendation).

5.3 WORK-UP OF DYSPLASTIC LESIONS Patients diagnosed with high-grade dysplasia should have careful endoscopic and pathological assessment [47]. Pathologists should follow a classification for reporting dysplasia that the multidisciplinary team is familiar with. An overview of classifications is already provided in chapter 3.2.4. Where therapeutic intervention is contemplated on the basis of high-grade dysplasia, the diagnosis should be validated by a second pathologist experienced in this area because of the significant inter- and intra-observer variation [47]. Further biopsies or EMR should be done if there is uncertainty. Biopsies should also be reviewed at a multidisciplinary meeting with access to the clinical information.

Finally, as the diagnosis, treatment and follow-up of patients with gastric high-grade dysplastic lesions is very specific and new techniques rapidly become available, these patients should be referred to centres or network reference centres with the appropriate endoscopic and surgical expertise and facilities [47].

Recommendations

5. Patients confirmed with high-grade dysplasia should have subsequent careful endoscopic and pathological assessment (1C recommendation).

6. Pathologists should follow a classification for reporting dysplasia that the multidisciplinary team is familiar with (1C recommendation).

7. Where therapeutic intervention is contemplated on the basis of high-grade dysplasia, the diagnosis should be validated by a second pathologist experienced in this area. Further biopsies should be done if there is uncertainty (1C recommendation).

8. Biopsies should be reviewed at a multidisciplinary meeting with access to the clinical information (expert opinion).

9. Patients with high-grade dysplasia should be referred to centres or network reference centres with the appropriate endoscopic and surgical expertise and facilities (1C recommendation).

Page 50: Guideline pour la prise en charge du cancer oesophagien et ...

34 Upper GI Cancer KCE reports 75

5.4 STAGING As for patients with oesophageal cancer, the main contribution of CT scan to the staging of gastric cancer is the detection of distant metastases [47, 129]. If metastatic disease is detected with CT scan, curative treatment is excluded and additional staging with EUS is unnecessary. A recent systematic review identified five observational studies examining the diagnostic accuracy of multidetector row CT (MDCT) for the T staging of gastric tumours [130]. It was concluded that both EUS and MDCT achieved similar results in terms of diagnostic accuracy in T staging and in assessing serosal involvement. Only one of the five included studies made a direct comparison between EUS and MDCT [130]. A more recent prospective study of MDCT (with multiplanar reformation images) found an overall diagnostic accuracy of 89% for T staging and of 78% for N staging [131].

Kwee et al. identified 23 studies examining the diagnostic accuracy of EUS for the T staging of gastric tumours [130]. Diagnostic accuracy varied between 65% and 92%. Sensitivity and specificity for assessing serosal involvement varied between 78% and 100% and between 68% and 100%, respectively.

PET(/CT) is not routinely recommended in the staging of gastric cancer. However, it can be considered in locally advanced mass-forming tumours (intestinal typea ) in a curative setting [134].

Kwee et al. identified 3 observational studies examining the diagnostic accuracy of MRI for the T staging of gastric tumours [130]. Diagnostic accuracy varied between 71% and 83%. Sensitivity and specificity for assessing serosal involvement varied between 90% and 93% and between 94% and 100%, respectively. Overall, there is no anatomical area where MRI is superior to CT. Therefore, MRI should be reserved for those patients who cannot undergo CT. It can also be used for additional investigation following CT and/or EUS [47].

Laparoscopy can be considered in patients with gastric tumours being considered for surgery where full thickness gastric wall involvement is suspected [47]. There are insufficient data to confirm benefit for laparoscopic ultrasound [47, 79].

Sentinel lymph node mapping has been the subject of several observational studies [135-139]. Overall sensitivity ranged from 71 – 89% [136-138]. In patients with a pT1 tumour, sensitivity ranged from 88 – 100% [136, 138].

a Gastric carcinomas can be classified according to different classification systems (Lauren [132],

Goseki [133], WHO, etc.) based on different criteria. The Lauren classification combines etiopathological, clinical and pathological (macro- and microscopic) features [132]. The most common type, the intestinal type, generates a tumoral mass and has convincingly been demonstrated to arise through a Helicobacter pylori infection, gastritis, intestinal metaplasia, dysplasia, carcinoma sequence. The less common type, the signet ring cell carcinoma, often presents with a characteristic macroscopic appearance called “linitis plastica” and will often be negative on PET.

Page 51: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 35

Recommendations

10. In patients with gastric cancer, CT scan of the chest and abdomen with IV contrast and gastric distension with oral contrast or water should be performed routinely. The liver should at least be imaged in the arterial and portal venous phase (1C recommendation).

11. Endoscopic ultrasonography with or without fine-needle aspiration cytology can be considered in patients to be treated with curative intent based on clinical presentation and/or CT (1C recommendation).

12. The following examinations can be considered for specific indications (as explained in the text above): PET scan, Magnetic Resonance Imaging, laparoscopy (1C recommendation).

5.5 TREATMENT OF MUCOSAL CANCER Treatment options for patients with mucosal cancer should be discussed at a multidisciplinary meeting, taking into account clinical and histological information. Superficial gastric cancer limited to the mucosa can be treated with endoscopic mucosal resection (EMR), taking into account the stage, size, histological type and differentiation grade (Table 10) [47]. Large Japanese series of hundreds of patients treated endoscopically with EMR or ESD showed the advantages and success rates of endoscopic management for mucosal cancer [140-143]. However, a recent Cochrane review failed to identify RCTs of early gastric cancer patients involving a treatment arm of EMR and a comparison arm of gastrectomy [144]. Therefore, the potential harms (incomplete resection, recurrence, perforation) and benefits (quality of life, utility in patients unfit for surgery) of EMR must be weighed against the harms/benefits of surgical treatment.

En-bloc resection – allowing better pathology staging of deep and lateral margins – should be aimed at during mucosectomy with the appropriate technique (EMR for lesions less than 12 mm and ESD for larger lesions) [145].

Mucosal ablative techniques, such as photodynamic therapy (PDT), laser or argon plasma coagulation (APC), cannot be recommended as a curative option.

Table 10. Criteria associated with low risk of lymph node metastasis in patients with mucosal gastric cancer [47, 144] (based on Japanese Classification of Gastric Carcinoma [146]).

• Lesion < 2 cm in size in elevated types

• Lesion < 1 cm in size in depressed types

• Well or moderately differentiated histology

• No macroscopic ulceration

• Invasion limited to mucosa and certainly no deeper than the superficial submucosa

• No residual invasive disease after EMR

Page 52: Guideline pour la prise en charge du cancer oesophagien et ...

36 Upper GI Cancer KCE reports 75

Recommendations

13. Biopsies should be reviewed by an experienced pathologist in this area and discussed at a multidisciplinary meeting with access to the clinical information (expert opinion).

14. Superficial gastric cancer limited to the mucosa can be treated with endoscopic mucosal resection (EMR), taking into account the stage, size, histological type and differentiation grade (2C recommendation).

15. Mucosal ablative techniques, such as photodynamic therapy (PDT), laser or argon plasma coagulation (APC), cannot be recommended as a curative option (expert opinion).

5.6 TREATMENT OF CANCER BEYOND THE MUCOSA

5.6.1 Neoadjuvant treatment

In a recent Cochrane review, 4 RCTs were identified comparing neoadjuvant chemotherapy and surgery to surgery alone, including 2 Japanese trials using oral fluoropyrimidines [147]. Meta-analysis of these 4 trials showed no statistically significant difference in mortality between the two treatment groups (OR 1.05; 95%CI 0.73 – 1.50; p = 0.29). The MAGIC trial, which was excluded from the Cochrane review because it also included postoperative chemotherapy, found a higher likelihood of overall survival in patients receiving perioperative chemotherapy (ECF) as compared to surgery alone (HR 0.75; 95%CI 0.60 – 0.93; p = 0.009) [148]. However, the trial received some criticism because of a lack of standardisation of surgery, the absence of separate data for gastric cancer, and some methodological shortcomings (e.g. lack of staging accuracy, no blinding).

One abstract was found presenting the results of an RCT comparing preoperative chemotherapy (5-FU/cisplatin) to surgery alone in adenocarcinoma of stomach and lower oesophagus [149]. HR of death was 0.69 (95%CI 0.50 – 0.95; p=0.02) with 3 and 5- year overall survival of 35% and 24% respectively in the surgery alone group vs. 48% and 38% respectively in the neoadjuvant chemotherapy group.

CCO identified three RCTs comparing preoperative radiotherapy and surgery to surgery alone [150]. However, inconsistent results were found, making it difficult to draw unambiguous conclusions. Three additional RCTs were identified comparing neoadjuvant immunotherapy and surgery to surgery alone [150]. No significant survival benefit was found. A recent small trial of neoadjuvant immunotherapy with interleukin-2 found a trend towards better overall and disease-free survival in the neoadjuvant treatment group [151].

As for oesophageal cancer, since the use of neoadjuvant treatment for patients with gastric cancer still is controversial, it is recommended to prospectively register clinical outcomes and adverse events of the combined treatment [92].

Recommendations

16. Neoadjuvant treatment is not routinely indicated for patients with gastric cancer, but is an option to be discussed during a multidisciplinary meeting (2A recommendation).

17. Prospective registration of clinical outcomes and adverse events of combined treatment is recommended (expert opinion).

Page 53: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 37

5.6.2 Surgical treatment

Surgical treatment remains the standard treatment for patients with resectable gastric cancer, and should aim at achieving an R0 resection [47, 129]. For distal tumours (lower third, antrum) a partial gastrectomy with sparing of the proximal stomach is indicated. Proximal tumours (upper two third) should be treated with a total gastrectomy [129].

Figure 2 gives an overview of the extent of lymphadenectomy for subtotal gastrectomy in terms of dissection of lymph nodes. McCulloch et al. identified 2 RCTs comparing limited (D1) versus extended (D2) node dissection (including splenectomy and distal pancreatectomy) during gastrectomy for gastric cancer [152]. Meta-analysis did not reveal any survival benefit for extended lymph node dissection (RR 0.95; 95%CI 0.83 – 1.09), but showed increased postoperative mortality (RR 2.23; 95%CI 1.45 – 3.45).

Figure 2. The extent of lymphadenectomy for subtotal gastrectomy in terms of dissection of lymph nodes (figure reprinted from Roukos et al. [153].

D1 requires the dissection of the first-tier nodes (N1, nodal stations no. 1-6). D2 includes the N1 and second-tier nodes (N2, no. 7-11). D3 requires the dissection of N1, N2 and third-tier nodes (N3, no. 12-15). D4 requires the dissection of N1, N2, N3 and fourth-tier para-aortic nodes (N4, no. 16).

Since this Cochrane review, numerous trials have been published examining the extent of lymph node dissection. An interim analysis of the Italian Gastric Cancer Study Group (IGCSG) randomised surgical trial showed no statistically significant difference in postoperative morbidity and mortality between D1 and D2 gastrectomy [154]. In this trial, only a minority of patients received a splenectomy (in case of clinical T > 1 on the greater curvature of the proximal and middle thirds of the stomach) or a distal pancreatectomy (suspicion of tumour involvement). However, a subsequent multicentric phase II study by the same group evaluating pancreas-preserving D2 gastrectomy suggested a survival benefit with an overall 5-year survival of 55% and a postoperative in-hospital mortality of 3.1% [155]. Hartgrink et al. also compared D1 with D2 lymph node dissection [156]. Postoperative morbidity (25% vs. 43%; p = 0.001) and mortality (4% vs. 10%; p = 0.004) were significantly higher in the D2 dissection group, but after 11 years no overall difference in survival was found (30% vs. 35%; p = 0.53).

Page 54: Guideline pour la prise en charge du cancer oesophagien et ...

38 Upper GI Cancer KCE reports 75

As compared to Degiuli et al. [154], the amount of splenectomies and distal pancreatectomies was higher in the Randomized Dutch Gastric Cancer Group Trial [156].

Three trials recently compared standard D2 vs. extended D2 (D2+, i.e. D2 with para-aortic nodal dissection) lymph node dissection [157-159]. No statistically significant differences in postoperative morbidity and mortality were found in two trials [157, 158], while postoperative morbidity was higher in the extended D2 group in the third trial [159].

Reconstruction after gastrectomy can be with or without pouch formation and with (Billroth I and II) or without (Roux-en-Y, jejunal interposition) maintenance of duodenal passage [47]. Evidence suggests that pouch procedures may be associated with a higher earlier weight gain [160, 161] and some improvement in long-term quality of life [162].

Several articles have compared laparoscopic surgery to conventional surgery for gastric cancer [163-166]. One SR identified 4 RCTs comparing laparoscopy-assisted distal gastrectomy (LADG) to conventional open distal gastrectomy (CODG) [164]. LADG was found to be a longer procedure, but was associated with a lower postoperative morbidity. No difference was found in postoperative mortality. These results were confirmed by an RCT published subsequently [166]. Hayashi et al. also found a shorter operation time with CODG, but no differences in postoperative morbidity and mortality [163]. Huscher et al. found no differences in duration of surgery and postoperative morbidity and mortality between laparoscopic-assisted and open radical subtotal gastrectomy [165]. Overall, duration of analgesic administration was shorter after laparoscopic surgery [163, 164, 166].

As for oesophageal cancer, several studies have shown a relationship between patient outcomes (e.g. 30-day mortality) and surgeon or hospital volume for gastric cancer surgery [47, 98, 99]. However, this relationship is less profound than for oesophageal cancer surgery [98]. Nevertheless, based on these results, gastric cancer surgery should be carried out in high volume specialist surgical units by surgeons with experience in gastric cancer [47]. Subspecialty training is also associated with lower postoperative mortality, although this relationship is not statistically significant [167].

Recommendations

18. Surgical resection should be considered standard treatment for patients with resectable gastric cancer (1A recommendation).

19. Surgery for gastric cancer should aim at achieving an R0 resection (1A recommendation).

20. D2 lymphadenectomy (with a minimum of 15 lymph nodes removed and examined) should be standard during gastrectomy to improve staging and local disease control (1B recommendation).

21. Gastric cancer surgery should be carried out in high volume specialist surgical units by surgeons with experience and/or specialist training in this area (1C recommendation).

5.6.3 Adjuvant treatment

Numerous RCTs and systematic reviews have been published comparing postoperative adjuvant chemotherapy to surgery alone [47, 150]. Most meta-analyses found small statistically significant differences in survival favouring adjuvant chemotherapy, although only a minority of the included RCTs were of high quality. Subgroup analyses in one meta-analysis showed a trend towards a larger magnitude of the effect for trials in which at least two thirds of the patients had node-positive disease (OR 0.74; 95%CI 0.59 – 0.95) [150]. Therefore, CCO concluded that adjuvant chemotherapy alone may be of benefit in particular for patients with lymph node metastases. However, this is not in line with SIGN, that did not recommend postoperative chemotherapy outside a clinical trial [47].

Page 55: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 39

One meta-analysis of 4 Japanese RCTs comparing adjuvant oral fluorinated pyrimidines to surgery alone found a survival benefit in favour of adjuvant chemotherapy (HR 0.73; 95%CI 0.60 – 0.89; p = 0.002) [168]. Other trials failed to demonstrate a benefit in favour of adjuvant chemotherapy [169-172]. The optimum chemotherapy regimen has also not yet been defined.

CCO identified two RCTs comparing adjuvant radiotherapy to surgery alone [150]. No differences in survival were found. However, addition of postoperative chemotherapy to radiotherapy may result in better outcomes. Three RCTs were identified by the CCO, of which one found no statistically significant difference in survival. The two other trials detected improved survival with postoperative CRT, but received considerable criticism [150]. For example, the surgery performed in the RCT of McDonald et al. was often not up to the desired standards [173].

Several RCTs and meta-analyses compared adjuvant intraperitoneal chemotherapy to surgery alone [150, 174, 175]. A recent meta-analysis found a significant survival benefit in favour of hyperthermic intraoperative intraperitoneal chemotherapy with or without early postoperative intraperitoneal chemotherapy [175]. However, in general survival results have been conflicting, and some trials even reported harm from intraperitoneal therapy [150].

CCO identified 9 RCTs comparing adjuvant immunochemotherapy to surgery alone [150]; one meta-analysis on immunochemotherapy with polysaccharide K [176] and one RCT [177] were published subsequently. Overall, inconsistent results were found, making it difficult to draw definite conclusions.

Recommendations

22. Postoperative adjuvant chemotherapy is not recommended for patients with gastric cancer (2A recommendation).

23. Postoperative adjuvant radiotherapy is not recommended for patients with gastric cancer (2A recommendation).

24. Postoperative adjuvant chemoradiotherapy is not routinely recommended for patients with gastric cancer, but can be considered after discussion in the multidisciplinary team (2A recommendation).

5.7 PALLIATIVE TREATMENT & METASTATIC DISEASE Palliative gastric surgery is limited to symptomatic stenoses, bleeding tumours and perforation [129]. It should be avoided in patients who have disseminated peritoneal disease [47, 129]. In comparison to palliative gastrectomy, gastric bypass has a higher mortality [47]. However, reported mortality after laparoscopic gastrojejunostomy is lower than for open bypass.

For patients with malignant gastric outlet obstruction, treatment options include endoscopic stenting or surgical gastroenterostomy [178, 179]. Two systematic reviews, that mainly comprised observational studies, found a higher initial clinical success after endoscopic stenting. However, inconsistent results were found for associated morbidity [178, 179]. No significant survival benefit was found. Endoscopic stenting was associated with a shorter hospital stay.

Several RCTs compared palliative chemotherapy to best supportive care for patients with advanced or metastatic gastric cancer [180, 181]. A clear survival benefit was found in favour of palliative chemotherapy. Above this, palliative chemotherapy was associated with a better quality of life [180]. Wagner et al. also found a significant survival benefit in favour of combination chemotherapy as compared to single agent chemotherapy (HR 0.83; 95%CI 0.74 – 0.93) [181]. Therefore, in patients with locally advanced or metastatic cancer of the stomach with good performance status combination chemotherapy should be considered [47, 181].

Page 56: Guideline pour la prise en charge du cancer oesophagien et ...

40 Upper GI Cancer KCE reports 75

Patients with gastric cancer should have access to a specialist (outpatient and/or inpatient) palliative care team, in particular in relation to comfort and symptom control, and quality of life [47]. This team clearly should involve the general practitioner, who should have a coordinating role in the organisation of the palliative home care. However, as for oesophageal cancer, evidence specific to control of symptoms, such as pain, anorexia and bleeding, in these cancers is limited. Some evidence exists for the use of celiac axis plexus block for the palliation of pain and for the use of corticosteroids in patients with gastric cancer who are anorexic or who have symptoms of bowel obstruction [47]. Nutritional support in the palliative setting should take into account the comfort of the patient, in that quality of life is much more important than the long-term effects.

Limited evidence (coming from a secondary analysis of 1 RCT) is also available for psychotherapeutic support during hospital stay [115].

Recommendations

25. Palliative gastric surgery is limited to symptomatic stenoses, bleeding tumours and perforation (2C recommendation).

26. For patients with malignant gastric outlet obstruction, treatment options include endoscopic stenting or surgical gastroenterostomy (2C recommendation).

27. In patients with locally advanced or metastatic cancer of the stomach with good performance status combination chemotherapy should be considered (1A recommendation).

28. Patients with gastric cancer should have access to a specialist palliative care team, in particular in relation to comfort and symptom control, and quality of life (1C recommendation).

5.8 FOLLOW-UP In patients that underwent treatment for gastric cancer, follow-up is needed to detect recurrent disease (in case of curative treatment), progressive disease, symptoms that warrant treatment, or nutritional problems resulting from total gastrectomy (e.g. iron and vitamin deficiency, maldigestion with steathorroea, etc). However, evidence about the duration, frequency and type of follow-up is lacking [182] (one retrospective study was published after our literature search [183]). Therefore, these recommendations are made in line with those for oesophageal cancer.

A physical examination is recommended every three months, and should give special attention to nutritional status, weight loss, fatigue, etc. Above this, a blood analysis (routine chemistry and haematology, completed with serum ferritine and vitamin B12 dosing in case of anaemia) is recommended. Vitamin B12 deficiency is a frequent phenomenon after total gastrectomy, and should be adequately treated. CT scan of the abdomen is recommended every six months in the first year and afterwards annually until the fifth year. However, it should be stressed that no evidence is available to support regular imaging in the follow-up of these patients [47]. For patients that underwent partial gastrectomy, lifelong endoscopic surveillance is indicated because of the high risk of developing gastric stump carcinoma [184].

Patients treated with endoscopic mucosal resection (EMR) should have strict endoscopic surveillance [144] with a follow-up endoscopy after three months, then every six months in the first two years, and then annually.

Page 57: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 41

Recommendations

29. It is recommended that the follow-up of patients treated for gastric cancer includes a physical examination and blood analysis every three months, and a CT scan every six months in the first year and afterwards annually until the fifth year (expert opinion).

30. Patients treated with endoscopic mucosal resection (EMR) should have a follow-up endoscopy after three months, then every six months in the first two years, and then annually (expert opinion).

5.9 RECURRENT DISEASE In a small number of patients, recurrent disease can be successfully treated with curative intent. In patients with cancer of the gastric stump after distal gastrectomy, R0 resection can be achieved in a relatively high number of patients [185, 186]. Patients who develop a solitary lung or liver metastasis can also be considered for resection [187, 188]. When patients are confronted with a local recurrence after EMR for a mucosal cancer, local treatment can be reconsidered [145, 189]. The evidence for these treatments, however, is weak and coming from small observational studies only. Treatment options for recurrent disease should therefore be discussed in the MDT.

Recommendations

31. In patients with recurrent gastric cancer, treatment options should be discussed in the multidisciplinary team (expert opinion).

32. In patients with a local recurrence or new tumour after endoscopic mucosal resection (EMR), treatment options, including local treatment, should be discussed in the multidisciplinary team (expert opinion).

5.10 TREATMENT OF GASTRIC LYMPHOMA

5.10.1 Introduction

Primary gastric lymphoma is a rare tumour, accounting for less than 5% of primary gastric neoplasms. However, it is the most common extranodal lymphoma, representing 4-20% of all extranodal lymphomas [190]. Helicobacter pylori infection, immunosuppression after solid-organ transplantation, celiac disease, inflammatory bowel disease, and human immunodeficiency virus (HIV) infection are known risk factors for GI lymphoma. A significant proportion of gastric lymphomas is of low-grade histology and arises from mucosa-associated lymphoid tissue (MALT) [191].

According to the most recent WHO classification, the term ‘MALT lymphoma’ should only be applied to tumours previously defined as low-grade MALT lymphomas composed mostly by small cells. High-grade lymphomas are known as large B-cell lymphoma [192]. Patients with low-grade B-cell lymphoma or MALT lymphoma have a better prognosis than patients with diffuse large B-cell lymphoma (DLBCL) [193].

Tumours of T-cell origin are rare [190]. Patients with gastric lymphomas are currently staged using the Ann Arbor staging system with the Cotswold modification. This has largely replaced the older International Workshop staging system [194].

5.10.2 Diagnosis and staging

There is no consensus regarding the diagnostic work-up and staging of primary gastric lymphomas. Evidence is scarce and exclusively based on observational studies. As for all patients with suspected gastric cancer, upper GI endoscopy is indicated in case of suspected gastric lymphoma.

Page 58: Guideline pour la prise en charge du cancer oesophagien et ...

42 Upper GI Cancer KCE reports 75

Dedicated endoscopic-bioptic techniques are recommended, including a minimum of 8 – 12 biopsies from visible lesions, mapping of macroscopically normal-appearing areas, and repeated examinations in the individual case. Biopsies should be preserved in such a way to allow molecular diagnostic investigation [195, 196].

Several classifications of lymphomas are available [197, 198]. It is recommended to diagnose and classify gastric lymphomas according to the most recent appropriate classification.

In patients with histologically confirmed gastric lymphoma, endoscopic ultrasonography is indicated [199, 200]. EUS-guided FNA of suspicious LN is not recommended. For other staging procedures, such as CT scan or PET scan, even less evidence is available [201-203]. Therefore, for patients with low-grade MALT lymphoma no further staging procedures are recommended, unless otherwise required for differential diagnostic reasons.

Recommendations

1. In patients with suspected gastric lymphoma, subtle endoscopic-bioptic techniques are needed, including a minimum of 8–12 biopsies from visible lesions, mapping of macroscopically normal-appearing areas, and repeated examinations in the individual case. Biopsies should be preserved in such a way to allow molecular diagnostic investigation (expert opinion).

2. Lymphomas should be diagnosed and classified according to the most recent appropriate classification (expert opinion).

3. In patients with histologically confirmed gastric lymphoma, endoscopic ultrasonography is indicated. Endoscopic ultrasound-guided fine needle aspiration of suspicious lymph nodes is not recommended (1C recommendation).

4. For patients with low-grade MALT lymphoma no further staging procedures are recommended, unless otherwise required for differential diagnostic reasons (expert opinion).

5.10.3 Treatment

In the past, surgery was thought to be necessary for the diagnosis, staging, and treatment of low-grade gastric lymphomas. However, as mentioned above, most patients now can be adequately diagnosed with biopsy and appropriately staged with non-invasive studies. Furthermore, gastrectomy can be associated with significant morbidity [194]. Numerous observational studies have demonstrated successful treatment of gastric MALT lymphomas with H. pylori eradication alone [204-208]. Complete remission rates ranged from 62 – 95%. No trials were found comparing H. pylori eradication to surgical treatment.

In one RCT, 245 previously untreated patients with gastric low-grade MALT lymphoma (stage IE and IIE) were randomised to surgery, radiotherapy or chemotherapy [209]. No statistically significant differences were found in overall survival, but event-free survival was significantly higher in the group randomised to chemotherapy.

Because the time from H. pylori eradication to lymphoma regression ranges from 1-28 months, close follow-up is required with upper GI endoscopy including biopsy [128, 194]. No evidence is available on the most appropriate duration of this follow-up, but an annual control during the first three years seems rational. Patients beyond stage I have a significantly decreased response to H. pylori eradication therapy [206, 207]. These patients, and patients without tumour regression after successful H. pylori eradication, should be referred to a specialized haematology centre [205].

Page 59: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 43

Recommendations

5. H. pylori eradication is the treatment of first choice for H. pylori infected patients with stage I low grade gastric MALT lymphoma (1C recommendation).

6. In patients with low-grade MALT lymphoma treated with antibiotic eradication, close follow-up with upper GI endoscopy and biopsies is required, including evaluation of H. pylori eradication within 3 months (expert opinion).

7. Patients with successful H. pylori eradication but without tumour regression after 1 year or with tumour progression should be referred to a specialized haematology centre (expert opinion).

5.11 TREATMENT OF GASTROINTESTINAL STROMAL TUMOURS

5.11.1 Introduction

Gastrointestinal stromal tumours (GIST) are relatively rare tumours, representing less than 1% of all tumours of the gastrointestinal tract. They are most common in the stomach (39% to 70%) and the small intestine (20% to 32%), whereas the colon, rectum and oesophagus are affected in less than 15% of cases [210]. GISTs predominantly occur in individuals over 40 years of age, with the majority occurring between the ages of 55 to 65. Estimates of incidence vary widely from 4 to 14 cases per million populations [211].

Presenting features of these tumours include abdominal discomfort or pain, a feeling of abdominal fullness and the presence of a palpable mass. However, many people with GISTs are asymptomatic during early stages of the disease until tumours reach a large size, at which time the tumours rupture and bleed or obstruct the gastrointestinal tract [211].

Overall, literature on GIST is relatively scarce and of low quality. Most studies are observational studies or case series without an adequate control. Therefore, the recommendations presented below often have a low level of evidence or are based on expert opinion.

5.11.2 Diagnosis and staging

In patients with clinical suspicion of GIST, endoscopic ultrasonography (EUS) and EUS-guided FNA are recommended for differential diagnostic reasons and to confirm the presence of positive lymph nodes or malignancy in adjacent organs [212]. Findings on EUS of a diameter > 3 cm, irregular extraluminal border, cystic spaces, echogenic foci, and adjacent malignant-appearing lymph nodes are features that can suggest malignancy. Unfortunately, EUS findings do not accurately predict the malignant potential of a small GIST (< 3 cm).

Most GISTs (80 – 100%) express the tyrosine kinase growth factor receptor c-KIT, which is detected by immunostaining with the antibody for the cell-surface marker CD117. This immunohistochemical test (performed on tissue) is now considered to be an appropriate diagnostic marker for the diagnosis of GIST [212].

In patients with a (suspected) GIST tumour, a CT abdomen is recommended if treatment is considered [213-217]. However, the evidence supporting this recommendation is observational and of low quality.

Page 60: Guideline pour la prise en charge du cancer oesophagien et ...

44 Upper GI Cancer KCE reports 75

Recommendations

1. In patients with clinical suspicion of GIST, endoscopic ultrasonography and endoscopic ultrasound-guided fine needle aspiration can be recommended for differential diagnostic reasons and to confirm the presence of positive lymph nodes or malignancy in adjacent organs (2C recommendation).

2. In patients with a (suspected) GIST, immunohistochemical testing of CD117 is recommended (1C recommendation).

3. In patients with a (suspected) GIST tumour, a CT abdomen is recommended if treatment is considered (expert opinion).

5.11.3 Treatment

5.11.3.1 Non-metastatic resectable GIST

Discrepancies were found in the literature on the strategy to predict malignant behaviour of GISTs. AGA proposed criteria including size (>3 cm), mitotic count on histology, and several EUS criteria (size; irregularity of the extraluminal border; the presence of cystic spaces, echogenic foci, and heterogeneity) [212]. The National Institutes of Health proposed a strategy for predicting malignant behaviour of GISTs based on size (>2cm, 2-5cm, 5-10 cm, or >10) and mitotic count on histology (<5, 6–10, or >10 per 50 high-power field), with the understanding that no GIST can be defined as benign on the basis of currently available diagnostic testing [218].

Debate also exists as to whether the potential morbidity and mortality associated with surgical resection are acceptable for removing a lesion with low potential for malignancy. Given the overall lack of evidence to guide management, it is recommended to perform surgical resection in patients that are fit for surgery and with a histologically confirmed non-metastatic GIST or with a gastric tumour of >5 cm that is highly suspicious of a GIST (and without evidence for metastatic disease) [211, 212, 219]. For lesions of 2 – 5 cm that are highly suspicious of GIST and without evidence for metastatic disease, the choice between surveillance and surgical resection should be discussed at the MDT, taking into consideration histology, size, margins, age, comorbidities and fitness for surgery. In patients with a gastric tumour of <2 cm that is highly suspicious of a GIST and without evidence for metastatic disease, surveillance is indicated.

The use of imatinib (see below), a targeted therapy aimed specifically at blocking the phosphorylation of tyrosine kinase receptors, as adjuvant treatment is investigational [210]. One abstract was identified comparing adjuvant imatinib to surgery alone for primary GIST measuring at least 3 cm and expressing c-kit [220]. Recurrence-free survival was significantly higher in the group assigned to adjuvant imatinib (HR 0.33; 95%CI 0.20 – 0.53).

Page 61: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 45

Recommendations

4. In patients with a histologically confirmed non-metastatic GIST and who are fit for surgery, resectional surgery is indicated (expert opinion).

5. In patients with a gastric tumour of >5 cm that is highly suspicious of a GIST, without evidence for metastatic disease, and who are fit for surgery, resectional surgery is indicated (expert opinion).

6. In patients with a gastric tumour of 2-5 cm that is highly suspicious of a GIST, without evidence for metastatic disease, and who are fit for surgery, the choice between surveillance and resectional surgery should be discussed at the multidisciplinary team (expert opinion).

7. In patients with a gastric tumour of <2 cm that is highly suspicious of a GIST and without evidence for metastatic disease, surveillance is indicated (expert opinion).

8. The use of imatinib as adjuvant treatment is investigational (expert opinion).

5.11.3.2 Metastatic or unresectable GIST

Imatinib clearly represents an important advance in the treatment of metastatic GISTs. A recent systematic review identified 2 RCTs comparing 2 doses of imatinib [210]. An initial dose of 400 mg daily is recommended, as a higher dose (800 mg) is not associated with a survival benefit, but leads to a significant increase in side effects [210]. Nevertheless, a dose of 400 mg twice daily may be considered in patients who demonstrate progression with 400 mg daily [211]. No RCTs comparing imatinib to no treatment or best supportive care have been published so far, making it difficult to state with statistical certainty whether treatment with imatinib confers a definite survival advantage as such. However, worldwide imatinib is considered standard treatment for these patients.

The optimal duration of imatinib treatment in responding patients or in those patients who achieve a complete clinical and/or radiological remission has not yet been defined, but continuation seems to be more favourable [219]. Low-quality observational studies show that PET/CT can be used to evaluate treatment response to imatinib [221-224]. In patients with imatinib resistance or intolerance sunitinib can be considered as second-line treatment [225]. So far, no RCTs have been published to compare sunitinib to imatinib or no treatment.

Recommendations

9. In patients with inoperable or metastatic (suspected) GIST imatinib is recommended (1C recommendation).

10. PET/CT is indicated to evaluate treatment response to imatinib (expert opinion).

11. In patients with imatinib resistance or intolerance sunitinib can be considered as second-line treatment (2A recommendation).

Page 62: Guideline pour la prise en charge du cancer oesophagien et ...

46 Upper GI Cancer KCE reports 75

6 IMPLEMENTATION AND UPDATING OF THE UPPER GI CANCER GUIDELINE

6.1 IMPLEMENTATION The implementation of the present guideline will be led by the College of Oncology. An online implementation tool – similar to the tools accompanying previous guidelines (https://portal.health.fgov.be/portal/page?_pageid=56,10338450&_dad=portal&_schema=PORTAL) – will be developed. The tool will be based on the general algorithms of this guideline.

6.2 QUALITY CONTROL The implementation of the guideline has to be evaluated with appropriate quality control criteria. These criteria should at least assess the following items of the general algorithms:

• diagnostic work-up

• staging

• treatment according to stage

• follow-up

• multidisciplinary approach

For each of these steps, quality indicators should be developed, which should be preferentially based on the recommendations with a high level of evidence. Additionally, a literature search for existing quality indicators should be done. However, a pre-assessment of the literature (Medline and the National Quality Measures Clearinghouse, http://www.qualitymeasures.ahrq.gov/) only identified a limited number of exisiting quality indicators (Table 11).

Table 11. Existing quality indicators for oesophageal and gastric cancer, identified through pre-assessment of the literature.

Quality Indicator Source Oesophageal resection mortality rate Agency for Healthcare Research and Quality Oesophageal resection: volume Agency for Healthcare Research and Quality

6.3 GUIDELINE UPDATE In view of the changing evidence, and based on a pre-assessment of the literature, this guideline should be fully updated in 5 years. In the meanwhile, when important evidence becomes available, this will be mentioned on the website of the College of Oncology (https://portal.health.fgov.be/portal/page?_pageid=56,10338450&_dad=portal&_schema=PORTAL).

Page 63: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 47

7 REFERENCES 1. Parkin, D.M., et al., Global cancer statistics, 2002. CA Cancer J Clin, 2005. 55(2): p. 74-108.

2. Bollschweiler, E., et al., Demographic variations in the rising incidence of esophageal adenocarcinoma in white males. Cancer, 2001. 92(3): p. 549-55.

3. Crane, L.M., et al., Oesophageal cancer in The Netherlands: increasing incidence and mortality but improving survival. Eur J Cancer, 2007. 43(9): p. 1445-51.

4. Qiu, D. and S. Kaneko, Comparison of esophageal cancer mortality in five countries: France, Italy, Japan, UK and USA from the WHO mortality database (1960-2000). Jpn J Clin Oncol, 2005. 35(9): p. 564-7.

5. Vizcaino, A.P., et al., Time trends incidence of both major histologic types of esophageal carcinomas in selected countries, 1973-1995. Int J Cancer, 2002. 99(6): p. 860-8.

6. Crew, K.D. and A.I. Neugut, Epidemiology of gastric cancer. World J Gastroenterol, 2006. 12(3): p. 354-62.

7. Wijnhoven, B.P.L., et al., Increased incidence of adenocarcinomas at the gastro-oesophageal junction in Dutch males since the 1990s. Eur J Gastroenterol Hepatol, 2002. 14(2): p. 115-22.

8. Crane, S.J., et al., The changing incidence of oesophageal and gastric adenocarcinoma by anatomic sub-site. Aliment Pharmacol Ther, 2007. 25(4): p. 447-53.

9. Peeters, M., et al., Nationale Richtlijnen van het College voor oncologie: A. Algemeen kader oncologish kwaliteitshandboek B. Wetenschappelijke basis voor klinische paden voor diagnose en behandeling colorectale kanker en testiskanker, in KCE reports. 2006, Federaal Kenniscentrum voor de Gezondheidszorg (KCE): Brussel.

10. Fervers, B., et al., Adaptation of clinical guidelines: literature review and proposition for a framework and procedure. Int J Qual Health Care, 2006. 18(3): p. 167-76.

11. Siewert, J.R. and H.J. Stein, Classification of adenocarcinoma of the oesophagogastric junction. Br J Surg, 1998. 85(11): p. 1457-9.

12. Spechler, S.J., et al., Adenocarcinoma of the esophago-gastric junction., in Pathology and Genetics of Tumours of the Digestive System., S.R. Hamilton and L.A. Aaltonen, Editors. 2000, IARC Press: Lyon, France. p. 31-36.

13. (UICC), I.U.A.C., TNM classification of malignant tumours. 6th ed. ed. 2002, Berlin: Springer-Verlag.

14. Corley, D.A. and A. Kubo, Influence of site classification on cancer incidence rates: an analysis of gastric cardia carcinomas. J Natl Cancer Inst, 2004. 96(18): p. 1383-7.

15. Driessen, A., et al., Identical cytokeratin expression pattern CK7+/CK20- in esophageal and cardiac cancer: etiopathological and clinical implications. Mod Pathol, 2004. 17(1): p. 49-55.

16. Driessen, A., et al., Are carcinomas of the cardia oesophageal or gastric adenocarcinomas? Eur J Cancer, 2003. 39(17): p. 2487-94.

17. Skinner, D.B., et al., Barrett's esophagus. Comparison of benign and malignant cases. Ann Surg, 1983. 198(4): p. 554-65.

18. Paull, A., et al., The histologic spectrum of Barrett's esophagus. N Engl J Med, 1976. 295(9): p. 476-80.

19. Naef, A.P., M. Savary, and L. Ozzello, Columnar-lined lower esophagus: an acquired lesion with malignant predisposition. Report on 140 cases of Barrett's esophagus with 12 adenocarcinomas. J Thorac Cardiovasc Surg, 1975. 70(5): p. 826-35.

20. Haggitt, R.C., et al., Adenocarcinoma complicating columnar epithelium-lined (Barrett's) esophagus. Am J Clin Pathol, 1978. 70(1): p. 1-5.

21. Reid, B.J., et al., Barrett's esophagus. Correlation between flow cytometry and histology in detection of patients at risk for adenocarcinoma. Gastroenterology, 1987. 93(1): p. 1-11.

Page 64: Guideline pour la prise en charge du cancer oesophagien et ...

48 Upper GI Cancer KCE reports 75

22. Spechler, S.J. and R.K. Goyal, The columnar-lined esophagus, intestinal metaplasia, and Norman Barrett. Gastroenterology, 1996. 110(2): p. 614-21.

23. Sampliner, R.E., Practice guidelines on the diagnosis, surveillance, and therapy of Barrett's esophagus. The Practice Parameters Committee of the American College of Gastroenterology. Am J Gastroenterol, 1998. 93(7): p. 1028-32.

24. Sharma, P., et al., A critical review of the diagnosis and management of Barrett's esophagus: the AGA Chicago Workshop. Gastroenterology, 2004. 127(1): p. 310-30.

25. British Society of Gastroenterology, Guidelines for the diagnosis and management of Barrett’s columnar-lined oesophagus., B.S.o. Gastroenterology, Editor. 2005, British Society of Gastroenterology: London.

26. Flejou, J.F. and M. Svrcek, Barrett's oesophagus--a pathologist's view. Histopathology, 2007. 50(1): p. 3-14.

27. Takubo, K., et al., Double muscularis mucosae in Barrett's esophagus. Hum Pathol, 1991. 22(11): p. 1158-61.

28. Nigro, J.J., et al., Prevalence and location of nodal metastases in distal esophageal adenocarcinoma confined to the wall: implications for therapy. J Thorac Cardiovasc Surg, 1999. 117(1): p. 16-23; discussion 23-5.

29. Westerterp, M., et al., Outcome of surgical treatment for early adenocarcinoma of the esophagus or gastro-esophageal junction. Virchows Arch, 2005. 446(5): p. 497-504.

30. Watanabe, H., J.R. Jass, and L.H. Sobin, Histological typing of oesophageal and gastric tumours. 2nd ed. ed. 1990, Berlin: Springer-Verlag.

31. Rubio, C.A., F.S. Liu, and H.Z. Zhao, Histological classification of intraepithelial neoplasias and microinvasive squamous carcinoma of the esophagus. Am J Surg Pathol, 1989. 13(8): p. 685-90.

32. Riddell, R.H., et al., Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol, 1983. 14(11): p. 931-68.

33. Montgomery, E., Is there a way for pathologists to decrease interobserver variability in the diagnosis of dysplasia? Arch Pathol Lab Med, 2005. 129(2): p. 174-6.

34. Montgomery, E., et al., Reproducibility of the diagnosis of dysplasia in Barrett esophagus: a reaffirmation. Hum Pathol, 2001. 32(4): p. 368-78.

35. Reid, B.J., et al., Observer variation in the diagnosis of dysplasia in Barrett's esophagus. Hum Pathol, 1988. 19(2): p. 166-78.

36. Schlemper, R.J., et al., International comparability of the pathological diagnosis for early cancer of the digestive tract: Munich meeting. J Gastroenterol, 2000. 35 Suppl 12: p. 102-10.

37. Schlemper, R.J., et al., Differences in diagnostic criteria for esophageal squamous cell carcinoma between Japanese and Western pathologists. Cancer, 2000. 88(5): p. 996-1006.

38. Schlemper, R.J., et al., Differences in diagnostic criteria for gastric carcinoma between Japanese and western pathologists. Lancet, 1997. 349(9067): p. 1725-9.

39. Antonioli, D.A. and H.H. Wang, Morphology of Barrett's esophagus and Barrett's-associated dysplasia and adenocarcinoma. Gastroenterol Clin North Am, 1997. 26(3): p. 495-506.

40. Haggitt, R.C., Barrett's esophagus, dysplasia, and adenocarcinoma. Hum Pathol, 1994. 25(10): p. 982-93.

41. Rugge, M., et al., Gastric dysplasia: the Padova international classification. Am J Surg Pathol, 2000. 24(2): p. 167-76.

42. Schlemper, R.J., et al., The Vienna classification of gastrointestinal epithelial neoplasia. Gut, 2000. 47(2): p. 251-5.

43. Schlemper, R.J., Y. Kato, and M. Stolte, Diagnostic criteria for gastrointestinal carcinomas in Japan and Western countries: proposal for a new classification system of gastrointestinal epithelial neoplasia. J Gastroenterol Hepatol, 2000. 15 Suppl: p. G49-57.

Page 65: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 49

44. Schlemper, R.J., Y. Kato, and M. Stolte, Review of histological classifications of gastrointestinal epithelial neoplasia: differences in diagnosis of early carcinomas between Japanese and Western pathologists. J Gastroenterol, 2001. 36(7): p. 445-56.

45. Hamilton, S.R. and L.A. Aaltonen, Pathology and genetics of tumours of the digestive system., ed. S.R. Hamilton and L.A. Aaltonen. 2000, Lyon, France: IARC Press.

46. Odze, R.D., Diagnosis and grading of dysplasia in Barrett's oesophagus. J Clin Pathol, 2006. 59(10): p. 1029-38.

47. Scottish Intercollegiate Guidelines Network, Management of oesophageal and gastric cancer. A national clinical guideline., SIGN, Editor. 2006, SIGN: Edinburgh.

48. Wang, K.K., M. Wongkeesong, and N.S. Buttar, American Gastroenterological Association technical review on the role of the gastroenterologist in the management of esophageal carcinoma. Gastroenterology, 2005. 128(5): p. 1471-505.

49. Bowrey, D.J., et al., Use of alarm symptoms to select dyspeptics for endoscopy causes patients with curable esophagogastric cancer to be overlooked. Surgical Endoscopy, 2006. 20(11): p. 1725-8.

50. Slee, G.R., S.M. Wagner, and F.S. McCullough, Odynophagia in patients with malignant disorders. Cancer, 1985. 55(12): p. 2877-9.

51. Dubuc, J., et al., Endoscopic screening for esophageal squamous-cell carcinoma in high-risk patients: a prospective study conducted in 62 French endoscopy centers. Endoscopy, 2006. 38(7): p. 690-5.

52. Borovicka, J., et al., Autofluorescence endoscopy in surveillance of Barrett's esophagus: a multicenter randomized trial on diagnostic efficacy. Endoscopy, 2006. 38(9): p. 867-72.

53. Lim, C.H., et al., Randomized crossover study that used methylene blue or random 4-quadrant biopsy for the diagnosis of dysplasia in Barrett's esophagus.[see comment]. Gastrointestinal Endoscopy, 2006. 64(2): p. 195-9.

54. Kara, M.A., et al., High-resolution endoscopy plus chromoendoscopy or narrow-band imaging in Barrett's esophagus: a prospective randomized crossover study. Endoscopy, 2005. 37(10): p. 929-36.

55. Corey, K.E., S.M. Schmitz, and N.J. Shaheen, Does a surgical antireflux procedure decrease the incidence of esophageal adenocarcinoma in Barrett's esophagus? A meta-analysis.[see comment]. American Journal of Gastroenterology, 2003. 98(11): p. 2390-4.

56. Kwaliteitsinstituut voor de Gezondheidszorg (CBO), Diagnostiek en behandeling oesofaguscarcinoom., V.Z.C. B.V., Editor. 2005, CBO: Alphen aan den Rijn.

57. Reid, B.J., et al., Optimizing endoscopic biopsy detection of early cancers in Barrett's high-grade dysplasia. Am J Gastroenterol, 2000. 95(11): p. 3089-96.

58. Reid, B.J., et al., Predictors of progression to cancer in Barrett's esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am J Gastroenterol, 2000. 95(7): p. 1669-76.

59. Lowe, V.J., et al., Comparison of positron emission tomography, computed tomography, and endoscopic ultrasound in the initial staging of patients with esophageal cancer. Molecular Imaging & Biology, 2005. 7(6): p. 422-30.

60. Lerut, T., et al., Three-field lymphadenectomy for carcinoma of the esophagus and gastroesophageal junction in 174 R0 resections: impact on staging, disease-free survival, and outcome: a plea for adaptation of TNM classification in upper-half esophageal carcinoma. Ann Surg, 2004. 240(6): p. 962-72; discussion 972-4.

61. van Vliet, E.P.M., et al., Publication bias does not play a role in the reporting of the results of endoscopic ultrasound staging of upper gastrointestinal cancers. Endoscopy, 2007. 39(4): p. 325-32.

62. Pfau, P.R., et al., Esophageal dilation for endosonographic evaluation of malignant esophageal strictures is safe and effective. Am J Gastroenterol, 2000. 95(10): p. 2813-5.

63. Shimpi, R.A., et al., Staging of esophageal cancer by EUS: staging accuracy revisited. Gastrointest Endosc, 2007. 66(3): p. 475-82.

64. Egan, J.V., et al., Esophageal dilation. Gastrointest Endosc, 2006. 63(6): p. 755-60.

Page 66: Guideline pour la prise en charge du cancer oesophagien et ...

50 Upper GI Cancer KCE reports 75

65. Doldi, S.B., et al., Ultrasonographic evaluation of the cervical lymph nodes in preoperative staging of esophageal neoplasms. Abdom Imaging, 1998. 23(3): p. 275-7.

66. Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC), Recommandations pour la pratique clinique: Standards, Options et Recommandations 2003 pour l’utilisation de la tomographie par émission de positons au [18F]-FDG (TEP-FDG) en cancérologie (rapport intégral). in Standards, Options et Recommendations. 2003, FNCLCC: Paris.

67. Cleemput, I., et al., HTA Positron Emission Tomography Imaging in Belgium, in KCE reports. 2005, Belgian Health Care Knowledge Centre (KCE): Brussels.

68. Choi, J.Y., et al., 18F-FDG PET in patients with esophageal squamous cell carcinoma undergoing curative surgery: prognostic implications. Journal of Nuclear Medicine, 2004. 45(11): p. 1843-50.

69. Kneist, W., et al., Prospective evaluation of positron emission tomography in the preoperative staging of esophageal carcinoma. Archives of Surgery, 2004. 139(10): p. 1043-9.

70. Leong, T., et al., A prospective study to evaluate the impact of FDG-PET on CT-based radiotherapy treatment planning for oesophageal cancer.[see comment]. Radiotherapy & Oncology, 2006. 78(3): p. 254-61.

71. Moureau-Zabotto, L., et al., Impact of CT and 18F-deoxyglucose positron emission tomography image fusion for conformal radiotherapy in esophageal carcinoma. International Journal of Radiation Oncology, Biology, Physics, 2005. 63(2): p. 340-5.

72. Ott, K., et al., Metabolic imaging predicts response, survival, and recurrence in adenocarcinomas of the esophagogastric junction. Journal of Clinical Oncology, 2006. 24(29): p. 4692-8.

73. Sihvo, E.I.T., et al., Adenocarcinoma of the esophagus and the esophagogastric junction: positron emission tomography improves staging and prediction of survival in distant but not in locoregional disease. Journal of Gastrointestinal Surgery, 2004. 8(8): p. 988-96.

74. Song, S.Y., et al., FDG-PET in the prediction of pathologic response after neoadjuvant chemoradiotherapy in locally advanced, resectable esophageal cancer. International Journal of Radiation Oncology, Biology, Physics, 2005. 63(4): p. 1053-9.

75. Yuan, S., et al., Additional value of PET/CT over PET in assessment of locoregional lymph nodes in thoracic esophageal squamous cell cancer. Journal of Nuclear Medicine, 2006. 47(8): p. 1255-9.

76. Osugi, H., et al., Bronchoscopic ultrasonography for staging supracarinal esophageal squamous cell carcinoma: impact on outcome. World Journal of Surgery, 2003. 27(5): p. 590-4.

77. Wakamatsu, T., et al., Usefulness of preoperative endobronchial ultrasound for airway invasion around the trachea: esophageal cancer and thyroid cancer.[see comment]. Respiration, 2006. 73(5): p. 651-7.

78. Imadahl, A., et al., [Is bronchoscopy a useful additional preoperative examination in esophageal carcinoma?]. Langenbecks Arch Chir, 1990. 375(6): p. 326-9.

79. Mortensen, M.B., et al., Combined preoperative endoscopic and laparoscopic ultrasonography for prediction of R0 resection in upper gastrointestinal tract cancer. British Journal of Surgery, 2006. 93(6): p. 720-5.

80. Oka, S., et al., Advantage of endoscopic submucosal dissection compared with EMR for early gastric cancer. Gastrointest Endosc, 2006. 64(6): p. 877-83.

81. Esaki, M., et al., Risk factors for local recurrence of superficial esophageal cancer after treatment by endoscopic mucosal resection. Endoscopy, 2007. 39(1): p. 41-5.

82. Katada, C., et al., Local recurrence of squamous-cell carcinoma of the esophagus after EMR. Gastrointest Endosc, 2005. 61(2): p. 219-25.

83. Overholt, B.F., et al., Photodynamic therapy with porfimer sodium for ablation of high-grade dysplasia in Barrett's esophagus: international, partially blinded, randomized phase III trial.[see comment][erratum appears in Gastrointest Endosc. 2006 Feb;63(2):359]. Gastrointestinal Endoscopy, 2005. 62(4): p. 488-98.

84. Malthaner, R.A., et al., Neoadjuvant or Adjuvant Therapy for Resectable Esophageal Cancer. Practice Guideline Report #2-11., CCO, Editor. 2005, CCO: Ottawa.

Page 67: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 51

85. Arnott, S.J., et al., Preoperative radiotherapy for esophageal carcinoma.[update of Cochrane Database Syst Rev. 2000;(4):CD001799; PMID: 11034728]. Cochrane Database of Systematic Reviews, 2005(4): p. CD001799.

86. Gebski, V., et al., Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: a meta-analysis.[see comment]. Lancet Oncology, 2007. 8(3): p. 226-34.

87. Malthaner, R.A., S. Collin, and D. Fenlon, Preoperative chemotherapy for resectable thoracic esophageal cancer.[update of Cochrane Database Syst Rev. 2003;(4):CD001556; PMID: 14583936]. Cochrane Database of Systematic Reviews, 2006. 3: p. CD001556.

88. Graham, A.J., et al., Defining the optimal treatment of locally advanced esophageal cancer: a systematic review and decision analysis.[see comment]. Annals of Thoracic Surgery, 2007. 83(4): p. 1257-64.

89. Natsugoe, S., et al., Randomized controlled study on preoperative chemoradiotherapy followed by surgery versus surgery alone for esophageal squamous cell cancer in a single institution. Diseases of the Esophagus, 2006. 19(6): p. 468-72.

90. Carstens, H., et al., A randomized trial of chemoradiotherapy versus surgery alone in patients with resectable esophageal cancer. J Clin Oncol (Meeting Abstracts), 2007. 25(18_suppl): p. 4530-.

91. Cerfolio, R.J., et al., The accuracy of endoscopic ultrasonography with fine-needle aspiration, integrated positron emission tomography with computed tomography, and computed tomography in restaging patients with esophageal cancer after neoadjuvant chemoradiotherapy. Journal of Thoracic & Cardiovascular Surgery, 2005. 129(6): p. 1232-41.

92. FOD Volksgezondheid Veiligheid van de voedselketen en Leefmilieu, K.B. 21 maart 2003. Koninklijk besluit houdende vaststelling van de normen waaraan het zorgprogramma voor oncologische basiszorg en het zorgprogramma voor oncologie moeten voldoen om te worden erkend. 2003: B.S. 25-04-2003.

93. Bedenne, L., et al., Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102.[see comment]. Journal of Clinical Oncology, 2007. 25(10): p. 1160-8.

94. Chiu, P.W.Y., et al., Multicenter prospective randomized trial comparing standard esophagectomy with chemoradiotherapy for treatment of squamous esophageal cancer: early results from the Chinese University Research Group for Esophageal Cancer (CURE). Journal of Gastrointestinal Surgery, 2005. 9(6): p. 794-802.

95. Hulscher, J.B., et al., Extended transthoracic resection compared with limited transhiatal resection for adenocarcinoma of the esophagus. N Engl J Med, 2002. 347(21): p. 1662-9.

96. Fumagalli, U., H. Akiyama, and T.R. DeMeester, Resective surgery for cancer of the thoracic esophagus: results of a consensus conference held at the VIth World congress of the international society for diseases of the esophagus. Dis Esophagus, 1996. 9: p. 30-38.

97. Altorki, N., et al., Three-field lymph node dissection for squamous cell and adenocarcinoma of the esophagus. Ann Surg, 2002. 236(2): p. 177-83.

98. Halm, E.A., C. Lee, and M.R. Chassin, Is volume related to outcome in health care? A systematic review and methodologic critique of the literature. Ann Intern Med, 2002. 137(6): p. 511-20.

99. Killeen, S.D., et al., Provider volume and outcomes for oncological procedures. Br J Surg, 2005. 92(4): p. 389-402.

100. Metzger, R., et al., High volume centers for esophagectomy: what is the number needed to achieve low postoperative mortality? Diseases of the Esophagus, 2004. 17(4): p. 310-4.

101. Dimick, J.B., et al., Specialty training and mortality after esophageal cancer resection. Ann Thorac Surg, 2005. 80(1): p. 282-6.

102. Pouliquen, X., et al., 5-Fluorouracil and cisplatin therapy after palliative surgical resection of squamous cell carcinoma of the esophagus. A multicenter randomized trial. French Associations for Surgical Research. Ann Surg, 1996. 223(2): p. 127-33.

Page 68: Guideline pour la prise en charge du cancer oesophagien et ...

52 Upper GI Cancer KCE reports 75

103. Ando, N., et al., A randomized trial of surgery with and without chemotherapy for localized squamous carcinoma of the thoracic esophagus: the Japan Clinical Oncology Group Study. J Thorac Cardiovasc Surg, 1997. 114(2): p. 205-9.

104. Ando, N., et al., Surgery plus chemotherapy compared with surgery alone for localized squamous cell carcinoma of the thoracic esophagus: a Japan Clinical Oncology Group Study--JCOG9204. Journal of Clinical Oncology, 2003. 21(24): p. 4592-6.

105. Fok, M., et al., Postoperative radiotherapy for carcinoma of the esophagus: a prospective, randomized controlled study. Surgery, 1993. 113(2): p. 138-47.

106. Teniere, P., et al., Postoperative radiation therapy does not increase survival after curative resection for squamous cell carcinoma of the middle and lower esophagus as shown by a multicenter controlled trial. French University Association for Surgical Research. Surg Gynecol Obstet, 1991. 173(2): p. 123-30.

107. Xiao, Z.F., et al., Value of radiotherapy after radical surgery for esophageal carcinoma: a report of 495 patients.[see comment]. Annals of Thoracic Surgery, 2003. 75(2): p. 331-6.

108. Wong, R.K., et al., Combined Modality Radiotherapy and Chemotherapy in the Non-surgical Management of Localized Carcinoma of the Esophagus. Practice Guideline Report #2-12., CCO, Editor. 2005, CCO: Ottawa.

109. Hao, D., et al., Platinum-based concurrent chemoradiotherapy for tumors of the head and neck and the esophagus. Seminars in Radiation Oncology, 2006. 16(1): p. 10-9.

110. Zhao, K.-l., et al., Late course accelerated hyperfractionated radiotherapy plus concurrent chemotherapy for squamous cell carcinoma of the esophagus: a phase III randomized study. International Journal of Radiation Oncology, Biology, Physics, 2005. 62(4): p. 1014-20.

111. Piessen, G., et al., Patients with locally advanced esophageal carcinoma nonresponder to radiochemotherapy: who will benefit from surgery? Ann Surg Oncol, 2007. 14(7): p. 2036-44.

112. Wenger, U., et al., Health economic evaluation of stent or endoluminal brachytherapy as a palliative strategy in patients with incurable cancer of the oesophagus or gastro-oesophageal junction: results of a randomized clinical trial. European Journal of Gastroenterology & Hepatology, 2005. 17(12): p. 1369-77.

113. Polinder, S., et al., Cost study of metal stent placement vs single-dose brachytherapy in the palliative treatment of oesophageal cancer. British Journal of Cancer, 2004. 90(11): p. 2067-72.

114. Homs, M.Y.V., et al., Chemotherapy for metastatic carcinoma of the esophagus and gastro-esophageal junction. Cochrane Database of Systematic Reviews, 2006(4): p. CD004063.

115. Kuchler, T., et al., Impact of psychotherapeutic support for patients with gastrointestinal cancer undergoing surgery: 10-year survival results of a randomized trial.[see comment]. Journal of Clinical Oncology, 2007. 25(19): p. 2702-8.

116. Kunisaki, C., et al., Surgical Outcomes in Esophageal Cancer Patients with Tumor Recurrence After Curative Esophagectomy. J Gastrointest Surg, 2007.

117. Natsugoe, S., et al., The role of salvage surgery for recurrence of esophageal squamous cell cancer. Eur J Surg Oncol, 2006. 32(5): p. 544-7.

118. Yamashita, H., et al., Salvage radiotherapy for postoperative loco-regional recurrence of esophageal cancer. Dis Esophagus, 2005. 18(4): p. 215-20.

119. Yano, M., et al., Prognosis of patients who develop cervical lymph node recurrence following curative resection for thoracic esophageal cancer. Dis Esophagus, 2006. 19(2): p. 73-7.

120. Komatsu, S., et al., Survival and clinical evaluation of salvage operation for cervical lymph node recurrence in esophageal cancer. Hepatogastroenterology, 2005. 52(63): p. 796-9.

121. Nishimura, Y., et al., Concurrent chemoradiotherapy with protracted infusion of 5-FU and cisplatin for postoperative recurrent or residual esophageal cancer. Jpn J Clin Oncol, 2003. 33(7): p. 341-5.

122. Schipper, P.H., et al., Locally recurrent esophageal carcinoma: when is re-resection indicated? Ann Thorac Surg, 2005. 80(3): p. 1001-5; discussion 1005-6.

Page 69: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 53

123. Nomura, T., et al., Recurrence after endoscopic mucosal resection for superficial esophageal cancer. Endoscopy, 2000. 32(4): p. 277-80.

124. Graham, D.Y., et al., Prospective evaluation of biopsy number in the diagnosis of esophageal and gastric carcinoma. Gastroenterology, 1982. 82(2): p. 228-31.

125. Sugano, K., K. Sato, and K. Yao, New diagnostic approaches for early detection of gastric cancer. Dig Dis, 2004. 22(4): p. 327-33.

126. Wang, C., Y. Yuan, and R.H. Hunt, The association between Helicobacter pylori infection and early gastric cancer: a meta-analysis. Am J Gastroenterol, 2007. 102(8): p. 1789-98.

127. Fuccio, L., et al., Systematic review: Helicobacter pylori eradication for the prevention of gastric cancer. Aliment Pharmacol Ther, 2007. 25(2): p. 133-41.

128. Malfertheiner, P., et al., Current concepts in the management of Helicobacter pylori infection: the Maastricht III Consensus Report. Gut, 2007. 56(6): p. 772-81.

129. Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC), Recommandations pour la pratique clinique: Standards, Options et Recommandations 2003 pour la prise en charge des patients atteints d’adénocarcinomes de l’estomac (cancers du cardia, autres types histologiques exclus) (rapport intégral), in Standards, Options et Recommandations. 2004, FNCLCC: Paris.

130. Kwee, R.M. and T.C. Kwee, Imaging in local staging of gastric cancer: a systematic review. Journal of Clinical Oncology, 2007. 25(15): p. 2107-16.

131. Chen, C.-Y., et al., Gastric cancer: preoperative local staging with 3D multi-detector row CT--correlation with surgical and histopathologic results. Radiology, 2007. 242(2): p. 472-82.

132. Lauren, P., The Two Histological Main Types of Gastric Carcinoma: Diffuse and So-Called Intestinal-Type Carcinoma. an Attempt at a Histo-Clinical Classification. Acta Pathol Microbiol Scand, 1965. 64: p. 31-49.

133. Goseki, N., T. Takizawa, and M. Koike, Differences in the mode of the extension of gastric cancer classified by histological type: new histological classification of gastric carcinoma. Gut, 1992. 33(5): p. 606-12.

134. Mukai, K., et al., Usefulness of preoperative FDG-PET for detection of gastric cancer. Gastric Cancer, 2006. 9(3): p. 192-6.

135. Gretschel, S., et al., Prediction of gastric cancer lymph node status by sentinel lymph node biopsy and the Maruyama computer model. European Journal of Surgical Oncology, 2005. 31(4): p. 393-400.

136. Lee, J.H., et al., Sentinel node biopsy using dye and isotope double tracers in early gastric cancer. Annals of Surgical Oncology, 2006. 13(9): p. 1168-74.

137. Miwa, K., et al., Mapping sentinel nodes in patients with early-stage gastric carcinoma. British Journal of Surgery, 2003. 90(2): p. 178-82.

138. Mochiki, E., et al., Sentinel lymph node mapping with technetium-99m colloidal rhenium sulfide in patients with gastric carcinoma. American Journal of Surgery, 2006. 191(4): p. 465-9.

139. Simsa, J., et al., Sentinel node biopsy in gastric cancer: preliminary results. Acta Chirurgica Belgica, 2003. 103(3): p. 270-3.

140. Ono, H., et al., Endoscopic mucosal resection for treatment of early gastric cancer. Gut, 2001. 48(2): p. 225-9.

141. Tajima, Y., et al., Histopathologic findings predicting lymph node metastasis and prognosis of patients with superficial esophageal carcinoma: analysis of 240 surgically resected tumors. Cancer, 2000. 88(6): p. 1285-93.

142. Gotoda, T., et al., Incidence of lymph node metastasis from early gastric cancer: estimation with a large number of cases at two large centers. Gastric Cancer, 2000. 3(4): p. 219-225.

143. Kitajima, K., et al., Correlations between lymph node metastasis and depth of submucosal invasion in submucosal invasive colorectal carcinoma: a Japanese collaborative study. J Gastroenterol, 2004. 39(6): p. 534-43.

Page 70: Guideline pour la prise en charge du cancer oesophagien et ...

54 Upper GI Cancer KCE reports 75

144. Wang, Y.P., C. Bennett, and T. Pan, Endoscopic mucosal resection for early gastric cancer. Cochrane Database of Systematic Reviews, 2006(1): p. CD004276.

145. Oka, S., et al., Endoscopic submucosal dissection for residual/local recurrence of early gastric cancer after endoscopic mucosal resection. Endoscopy, 2006. 38(10): p. 996-1000.

146. Japanese Gastric Cancer, A., Japanese Classification of Gastric Carcinoma - 2nd English Edition. Gastric Cancer, 1998. 1(1): p. 10-24.

147. Wu, A.W., et al., Neoadjuvant chemotherapy versus none for resectable gastric cancer. Cochrane Database of Systematic Reviews, 2007(2): p. CD005047.

148. Cunningham, D., et al., Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer.[see comment]. New England Journal of Medicine, 2006. 355(1): p. 11-20.

149. Boige, V., et al., Final results of a randomized trial comparing preoperative 5-fluorouracil (F)/cisplatin (P) to surgery alone in adenocarcinoma of stomach and lower esophagus (ASLE): FNLCC ACCORD07-FFCD 9703 trial. J Clin Oncol (Meeting Abstracts), 2007. 25(18_suppl): p. 4510-.

150. Earle, C.C., et al., Neoadjuvant or Adjuvant Therapy for Resectable Gastric Cancer. Practice Guideline Report #2-14., CCO, Editor. 2003, CCO: Ottawa.

151. Romano, F., et al., Phase-II randomized study of preoperative IL-2 administration in radically operable gastric cancer patients. Hepato-Gastroenterology, 2004. 51(60): p. 1872-6.

152. McCulloch, P., et al., Extended versus limited lymph nodes dissection technique for adenocarcinoma of the stomach.[update of Cochrane Database Syst Rev. 2003;(4):CD001964; PMID: 14583942]. Cochrane Database of Systematic Reviews, 2004(4): p. CD001964.

153. Roukos, D.H. and A.M. Kappas, Perspectives in the treatment of gastric cancer. Nat Clin Pract Oncol, 2005. 2(2): p. 98-107.

154. Degiuli, M., et al., Morbidity and mortality after D1 and D2 gastrectomy for cancer: interim analysis of the Italian Gastric Cancer Study Group (IGCSG) randomised surgical trial. European Journal of Surgical Oncology, 2004. 30(3): p. 303-8.

155. Degiuli, M., et al., Survival results of a multicentre phase II study to evaluate D2 gastrectomy for gastric cancer. Br J Cancer, 2004. 90(9): p. 1727-32.

156. Hartgrink, H.H., et al., Extended lymph node dissection for gastric cancer: who may benefit? Final results of the randomized Dutch gastric cancer group trial.[see comment]. Journal of Clinical Oncology, 2004. 22(11): p. 2069-77.

157. Kulig, J., et al., Standard D2 versus extended D2 (D2+) lymphadenectomy for gastric cancer: an interim safety analysis of a multicenter, randomized, clinical trial. American Journal of Surgery, 2007. 193(1): p. 10-5.

158. Sano, T., et al., Gastric cancer surgery: morbidity and mortality results from a prospective randomized controlled trial comparing D2 and extended para-aortic lymphadenectomy--Japan Clinical Oncology Group study 9501.[see comment]. Journal of Clinical Oncology, 2004. 22(14): p. 2767-73.

159. Yonemura, Y., et al., Operative morbidity and mortality after D2 and D4 extended dissection for advanced gastric cancer: a prospective randomized trial conducted by Asian surgeons. Hepato-Gastroenterology, 2006. 53(69): p. 389-94.

160. Iwata, T., et al., Evaluation of reconstruction after proximal gastrectomy: prospective comparative study of jejunal interposition and jejunal pouch interposition. Hepato-Gastroenterology, 2006. 53(68): p. 301-3.

161. Yoo, C.H., et al., Proximal gastrectomy reconstructed by jejunal pouch interposition for upper third gastric cancer: prospective randomized study. World Journal of Surgery, 2005. 29(12): p. 1592-9.

162. Kono, K., et al., Improved quality of life with jejunal pouch reconstruction after total gastrectomy. American Journal of Surgery, 2003. 185(2): p. 150-4.

163. Hayashi, H., et al., Prospective randomized study of open versus laparoscopy-assisted distal gastrectomy with extraperigastric lymph node dissection for early gastric cancer. Surgical Endoscopy, 2005. 19(9): p. 1172-6.

Page 71: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 55

164. Hosono, S., et al., Meta-analysis of short-term outcomes after laparoscopy-assisted distal gastrectomy. World Journal of Gastroenterology, 2006. 12(47): p. 7676-83.

165. Huscher, C.G.S., et al., Laparoscopic versus open subtotal gastrectomy for distal gastric cancer: five-year results of a randomized prospective trial. Annals of Surgery, 2005. 241(2): p. 232-7.

166. Lee, J.H., H.S. Han, and J.H. Lee, A prospective randomized study comparing open vs laparoscopy-assisted distal gastrectomy in early gastric cancer: early results. Surgical Endoscopy, 2005. 19(2): p. 168-73.

167. Callahan, M.A., et al., Influence of surgical subspecialty training on in-hospital mortality for gastrectomy and colectomy patients. Ann Surg, 2003. 238(4): p. 629-36; discussion 636-9.

168. Oba, K., et al., Efficacy of adjuvant chemotherapy using oral fluorinated pyrimidines for curatively resected gastric cancer: a meta-analysis of centrally randomized controlled clinical trials in Japan. Journal of Chemotherapy, 2006. 18(3): p. 311-7.

169. Bouche, O., et al., Adjuvant chemotherapy with 5-fluorouracil and cisplatin compared with surgery alone for gastric cancer: 7-year results of the FFCD randomized phase III trial (8801). Annals of Oncology, 2005. 16(9): p. 1488-97.

170. Chipponi, J., et al., Randomized trial of adjuvant chemotherapy after curative resection for gastric cancer. American Journal of Surgery, 2004. 187(3): p. 440-5.

171. Nashimoto, A., et al., Randomized trial of adjuvant chemotherapy with mitomycin, Fluorouracil, and Cytosine arabinoside followed by oral Fluorouracil in serosa-negative gastric cancer: Japan Clinical Oncology Group 9206-1.[see comment]. Journal of Clinical Oncology, 2003. 21(12): p. 2282-7.

172. Nitti, D., et al., Randomized phase III trials of adjuvant FAMTX or FEMTX compared with surgery alone in resected gastric cancer. A combined analysis of the EORTC GI Group and the ICCG. Annals of Oncology, 2006. 17(2): p. 262-9.

173. Macdonald, J.S., et al., Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med, 2001. 345(10): p. 725-30.

174. Xu, D.-Z., et al., Meta-analysis of intraperitoneal chemotherapy for gastric cancer. World Journal of Gastroenterology, 2004. 10(18): p. 2727-30.

175. Yan, T.D., et al., Hp40p a systematic review and meta-analysis of the randomised controlled trials on adjuvant intraperitoneal chemotherapy for advanced gastric cancer. ANZ Journal of Surgery, 2007. 77 Suppl 1: p. A49.

176. Oba, K., et al., Efficacy of adjuvant immunochemotherapy with polysaccharide K for patients with curative resections of gastric cancer. Cancer Immunology, Immunotherapy, 2007. 56(6): p. 905-11.

177. Popiela, T., et al., Efficiency of adjuvant immunochemotherapy following curative resection in patients with locally advanced gastric cancer. Gastric Cancer, 2004. 7(4): p. 240-5.

178. Hosono, S., et al., Endoscopic stenting versus surgical gastroenterostomy for palliation of malignant gastroduodenal obstruction: a meta-analysis. Journal of Gastroenterology, 2007. 42(4): p. 283-90.

179. Jeurnink, S.M., et al., Stent versus gastrojejunostomy for the palliation of gastric outlet obstruction: a systematic review. BMC Gastroenterology, 2007. 7: p. 18.

180. Casaretto, L., P.L.R. Sousa, and J.J. Mari, Chemotherapy versus support cancer treatment in advanced gastric cancer: a meta-analysis. Brazilian Journal of Medical & Biological Research, 2006. 39(4): p. 431-40.

181. Wagner, A.D., et al., Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. Journal of Clinical Oncology, 2006. 24(18): p. 2903-9.

182. Whiting, J., et al., Follow-up of gastric cancer: a review. Gastric Cancer, 2006. 9(2): p. 74-81.

183. Tan, I.T. and B.Y. So, Value of intensive follow-up of patients after curative surgery for gastric carcinoma. J Surg Oncol, 2007. 96(6): p. 503-6.

184. Sinning, C., et al., Gastric stump carcinoma - epidemiology and current concepts in pathogenesis and treatment. Eur J Surg Oncol, 2007. 33(2): p. 133-9.

Page 72: Guideline pour la prise en charge du cancer oesophagien et ...

56 Upper GI Cancer KCE reports 75

185. Ohashi, M., et al., Cancer of the gastric stump following distal gastrectomy for cancer. Br J Surg, 2007. 94(1): p. 92-5.

186. Chen, L., et al., Surgical management of gastric stump cancer: a report of 37 cases. J Zhejiang Univ Sci B, 2005. 6(1): p. 38-42.

187. Hirai, I., et al., Surgical management for metastatic liver tumors. Hepatogastroenterology, 2006. 53(71): p. 757-63.

188. Lehnert, T., et al., Surgical therapy for loco-regional recurrence and distant metastasis of gastric cancer. Eur J Surg Oncol, 2002. 28(4): p. 455-61.

189. Yokoi, C., et al., Endoscopic submucosal dissection allows curative resection of locally recurrent early gastric cancer after prior endoscopic mucosal resection. Gastrointest Endosc, 2006. 64(2): p. 212-8.

190. Al-Akwaa, A.M., N. Siddiqui, and I.A. Al-Mofleh, Primary gastric lymphoma. World J Gastroenterol, 2004. 10(1): p. 5-11.

191. Crump, M., M. Gospodarowicz, and F.A. Shepherd, Lymphoma of the gastrointestinal tract. Semin Oncol, 1999. 26(3): p. 324-37.

192. Zucca, E. and F. Cavalli, Are antibiotics the treatment of choice for gastric lymphoma? Curr Hematol Rep, 2004. 3(1): p. 11-6.

193. Binn, M., et al., Surgical resection plus chemotherapy versus chemotherapy alone: comparison of two strategies to treat diffuse large B-cell gastric lymphoma.[see comment]. Ann Oncol, 2003. 14(12): p. 1751-7.

194. Yoon, S.S. and E.P. Hochberg, Chemotherapy is an effective first line treatment for early stage gastric mucosa-associated lymphoid tissue lymphoma. Cancer Treat Rev, 2006. 32(2): p. 139-43.

195. Fischbach, W., et al., Primary gastric B-cell lymphoma: results of a prospective multicenter study. The German-Austrian Gastrointestinal Lymphoma Study Group.[see comment][erratum appears in Gastroenterology 2000 Dec;119(6):1809]. Gastroenterology, 2000. 119(5): p. 1191-202.

196. Strecker, P., et al., [Diagnostic value of stomach biopsy in comparison with surgical specimen in gastric B-cell lymphomas of the MALT type]. Pathologe, 1998. 19(3): p. 209-13.

197. Isaacson, P.G., J. Spencer, and D.H. Wright, Classifying primary gut lymphomas. Lancet, 1988. 2(8620): p. 1148-9.

198. Harris, N.L., et al., A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood, 1994. 84(5): p. 1361-92.

199. Caletti, G., et al., Accuracy of endoscopic ultrasonography in the diagnosis and staging of gastric cancer and lymphoma. Surgery, 1993. 113(1): p. 14-27.

200. Fischbach, W., M.-E. Goebeler-Kolve, and A. Greiner, Diagnostic accuracy of EUS in the local staging of primary gastric lymphoma: results of a prospective, multicenter study comparing EUS with histopathologic stage. Gastrointest Endosc, 2002. 56(5): p. 696-700.

201. Rodriguez, M., et al., [18F] FDG PET in gastric non-Hodgkin's lymphoma. Acta Oncol, 1997. 36(6): p. 577-84.

202. Vorbeck, F., et al., Comparison of spiral-computed tomography with water-filling of the stomach and endosonography for gastric lymphoma of mucosa-associated lymphoid tissue-type. Digestion, 2002. 65(4): p. 196-9.

203. Hoepffner, N., et al., [Value of endosonography in diagnostic staging of primary gastric lymphoma (MALT type)]. Med Klin (Munich), 2003. 98(6): p. 313-7.

204. Hong, S.S., et al., A prospective analysis of low-grade gastric malt lymphoma after Helicobacter pylori eradication. Helicobacter, 2006. 11(6): p. 569-73.

205. Nakamura, S., et al., Long-term clinical outcome of Helicobacter pylori eradication for gastric mucosa-associated lymphoid tissue lymphoma with a reference to second-line treatment. Cancer, 2005. 104(3): p. 532-40.

206. Wundisch, T., et al., Long-term follow-up of gastric MALT lymphoma after Helicobacter pylori eradication. J Clin Oncol, 2005. 23(31): p. 8018-24.

Page 73: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 57

207. Fischbach, W., et al., Long term outcome of patients with gastric marginal zone B cell lymphoma of mucosa associated lymphoid tissue (MALT) following exclusive Helicobacter pylori eradication therapy: experience from a large prospective series. Gut, 2004. 53(1): p. 34-7.

208. Montalban, C., et al., Treatment of low grade gastric mucosa-associated lymphoid tissue lymphoma in stage I with Helicobacter pylori eradication. Long-term results after sequential histologic and molecular follow-up. Haematologica, 2001. 86(6): p. 609-17.

209. Aviles, A., et al., Mucosa-associated lymphoid tissue (MALT) lymphoma of the stomach: results of a controlled clinical trial. Medical Oncology, 2005. 22(1): p. 57-62.

210. Verma, S., et al., Imatinib Mesylate (Gleevec™) for the Treatment of Adult Patients with Unresectable or Metastatic Gastrointestinal Stromal Tumours:A Clinical Practice Guideline. 2006, CCO: Ottawa.

211. Wilson, J.S., et al., Imatinib mesylate for the treatment of patients with unresectable and/or metastatic gastro-intestinal stromal tumours (GIST). 2004, West Midlands Health Technology Assessment Collaboration, University of Birmingham: Birmingham.

212. Hwang, J.H., S.D. Rulyak, and M.B. Kimmey, American Gastroenterological Association Institute technical review on the management of gastric subepithelial masses. Gastroenterology, 2006. 130(7): p. 2217-28.

213. Rimondini, A., et al., Contribution of CT to treatment planning in patients with GIST. Radiol Med (Torino), 2007. 112(5): p. 691-702.

214. Lupescu, I.G., et al., Computer tomographic evaluation of digestive tract non-Hodgkin lymphomas. J Gastrointestin Liver Dis, 2007. 16(3): p. 315-9.

215. De Leo, C., et al., Gastrointestinal stromal tumours: experience with multislice CT. Radiol Med (Torino), 2006. 111(8): p. 1103-14.

216. Da Ronch, T., A. Modesto, and M. Bazzocchi, Gastrointestinal stromal tumour: spiral computed tomography features and pathologic correlation. Radiol Med (Torino), 2006. 111(5): p. 661-73.

217. Hong, X., et al., Gastrointestinal stromal tumor: role of CT in diagnosis and in response evaluation and surveillance after treatment with imatinib. Radiographics, 2006. 26(2): p. 481-95.

218. Fletcher, C.D.M., et al., Diagnosis of gastrointestinal stromal tumors: A consensus approach. Hum Pathol, 2002. 33(5): p. 459-65.

219. Blay, J.-Y., et al., [Recommendations for the management of GIST patients]. Bull Cancer, 2005. 92(10): p. 907-18.

220. Dematteo, R.P., et al. Adjuvant imatinib mesylate increases recurrence free survival (RFS) in patients with completely resected localized primary gastrointestinal stromal tumor (GIST): North American Intergroup Phase III trial ACOSOG Z9001. 2007 [cited 2008 23-01-2008]; Available from: http://www.asco.org/portal/site/ASCO/menuitem.34d60f5624ba07fd506fe310ee37a01d/?vgnextoid=76f8201eb61a7010VgnVCM100000ed730ad1RCRD&vmview=abst_detail_view&confID=47&abstractID=100001.

221. Zincirkeser, S., et al., Early detection of response to imatinib therapy for gastrointestinal stromal tumor by using 18F-FDG-positron emission tomography and computed tomography imaging. World J Gastroenterol, 2007. 13(15): p. 2261-2.

222. Goh, B.K., et al., Pathologic, radiologic and PET scan response of gastrointestinal stromal tumors after neoadjuvant treatment with imatinib mesylate. Eur J Surg Oncol, 2006. 32(9): p. 961-3.

223. Heinicke, T., et al., Very early detection of response to imatinib mesylate therapy of gastrointestinal stromal tumours using 18fluoro-deoxyglucose-positron emission tomography. Anticancer Res, 2005. 25(6C): p. 4591-4.

224. Goldstein, D., et al., Gastrointestinal stromal tumours: correlation of F-FDG gamma camera-based coincidence positron emission tomography with CT for the assessment of treatment response--an AGITG study. Oncology, 2005. 69(4): p. 326-32.

225. Demetri, G.D., et al., Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet, 2006. 368(9544): p. 1329-38.

Page 74: Guideline pour la prise en charge du cancer oesophagien et ...

58 Upper GI Cancer KCE reports 75

226. Jacobson, B.C., et al., The role of endoscopy in the assessment and treatment of esophageal cancer. Gastrointestinal Endoscopy, 2003. 57(7): p. 817-22.

227. National Comprehensine Cancer Network (NCCN), Esophageal Cancer, in NCCN Clinical Practice Guidelines in Oncology™, NCCN, Editor. 2007, NCCN.

228. National Comprehensine Cancer Network (NCCN), Gastric Cancer, in NCCN Clinical Practice Guidelines in Oncology™, NCCN, Editor. 2007, NCCN.

229. Cunningham, D., et al., ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up of gastric cancer. Annals of Oncology, 2005. 16(1).

230. Stahl, M., et al., ESMO Minimal Clinical Recommendations for diagnosis, treatment and follow-up of esophageal cancer. Annals of Oncology, 2005. 16(1).

231. Sampliner, R.E. and G. Practice Parameters Committee of the American College of, Updated guidelines for the diagnosis, surveillance, and therapy of Barrett's esophagus. American Journal of Gastroenterology, 2002. 97(8): p. 1888-95.

232. Allum, W.H., et al., Guidelines for the management of oesophageal and gastric cancer. Gut, 2002. 1(50).

233. Marmo, R., et al., Combination of age and sex improves the ability to predict upper gastrointestinal malignancy in patients with uncomplicated dyspepsia: a prospective multicentre database study. Am J Gastroenterol, 2005. 100(4): p. 784-91.

234. Kim, S.H., et al., Three-dimensional MDCT imaging and CT esophagography for evaluation of esophageal tumors: preliminary study. European Radiology, 2006. 16(11): p. 2418-26.

235. Umeoka, S., et al., Esophageal cancer: evaluation with triple-phase dynamic CT--initial experience. Radiology, 2006. 239(3): p. 777-83.

236. Panebianco, V., et al., 3D CT protocol in the assessment of the esophageal neoplastic lesions: can it improve TNM staging? European Radiology, 2006. 16(2): p. 414-21.

237. Onbas, O., et al., Preoperative staging of esophageal carcinoma with multidetector CT and virtual endoscopy. Eur J Radiol, 2006. 57(1): p. 90-5.

238. Weaver, S.R., et al., Comparison of special interest computed tomography, endosonography and histopathological stage of oesophageal cancer. Clinical Radiology, 2004. 59(6): p. 499-504.

239. Yoon, Y.C., et al., Metastasis to regional lymph nodes in patients with esophageal squamous cell carcinoma: CT versus FDG PET for presurgical detection prospective study. Radiology, 2003. 227(3): p. 764-70.

240. Vazquez-Sequeiros, E., et al., Routine vs. selective EUS-guided FNA approach for preoperative nodal staging of esophageal carcinoma.[see comment]. Gastrointestinal Endoscopy, 2006. 63(2): p. 204-11.

241. May, A., et al., Accuracy of staging in early oesophageal cancer using high resolution endoscopy and high resolution endosonography: a comparative, prospective, and blinded trial. Gut, 2004. 53(5): p. 634-40.

242. Yanai, H., et al., Prognostic value and interobserver agreement of endoscopic ultrasonography for superficial squamous cell carcinoma of the esophagus: a prospective study. International Journal of Gastrointestinal Cancer, 2003. 34(1): p. 1-8.

243. Chang, K.J., et al., Impact of endoscopic ultrasound combined with fine-needle aspiration biopsy in the management of esophageal cancer. Endoscopy, 2003. 35(11): p. 962-6.

244. Mariette, C., et al., Value of endoscopic ultrasonography as a predictor of long-term survival in oesophageal carcinoma.[see comment]. British Journal of Surgery, 2003. 90(11): p. 1367-72.

245. Menon, K.V. and T.C.B. Dehn, Multiport staging laparoscopy in esophageal and cardiac carcinoma. Diseases of the Esophagus, 2003. 16(4): p. 295-300.

246. Riddell, A.M., et al., Potential of surface-coil MRI for staging of esophageal cancer. AJR, 2006. American Journal of Roentgenology. 187(5): p. 1280-7.

Page 75: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 59

247. Meyers, B.F., et al., The utility of positron emission tomography in staging of potentially operable carcinoma of the thoracic esophagus: results of the American College of Surgeons Oncology Group Z0060 trial. Journal of Thoracic & Cardiovascular Surgery, 2007. 133(3): p. 738-45.

248. van Baardwijk, A., et al., The current status of FDG-PET in tumour volume definition in radiotherapy treatment planning. Cancer Treatment Reviews, 2006. 32(4): p. 245-60.

249. Levine, E.A., et al., Predictive value of 18-fluoro-deoxy-glucose-positron emission tomography (18F-FDG-PET) in the identification of responders to chemoradiation therapy for the treatment of locally advanced esophageal cancer. Annals of Surgery, 2006. 243(4): p. 472-8.

250. Kato, H., et al., Comparison between whole-body positron emission tomography and bone scintigraphy in evaluating bony metastases of esophageal carcinomas. Anticancer Research, 2005. 25(6C): p. 4439-44.

251. van Westreenen, H.L., et al., Comparison of 18F-FLT PET and 18F-FDG PET in esophageal cancer. Journal of Nuclear Medicine, 2005. 46(3): p. 400-4.

252. Vrieze, O., et al., Is there a role for FGD-PET in radiotherapy planning in esophageal carcinoma? Radiotherapy & Oncology, 2004. 73(3): p. 269-75.

253. Pohl, J., et al., Comparison of computed virtual chromoendoscopy and conventional chromoendoscopy with acetic acid for detection of neoplasia in Barrett's esophagus. Endoscopy, 2007. 39(7): p. 594-8.

254. Ragunath, K., et al., A randomized, prospective cross-over trial comparing methylene blue-directed biopsy and conventional random biopsy for detecting intestinal metaplasia and dysplasia in Barrett's esophagus. Endoscopy, 2003. 35(12): p. 998-1003.

255. Pech, O., et al., The impact of endoscopic ultrasound and computed tomography on the TNM staging of early cancer in Barrett's esophagus. American Journal of Gastroenterology, 2006. 101(10): p. 2223-9.

256. Larghi, A., et al., EUS followed by EMR for staging of high-grade dysplasia and early cancer in Barrett's esophagus. Gastrointestinal Endoscopy, 2005. 62(1): p. 16-23.

257. Waxman, I., et al., High-frequency probe ultrasonography has limited accuracy for detecting invasive adenocarcinoma in patients with Barrett's esophagus and high-grade dysplasia or intramucosal carcinoma: a case series. American Journal of Gastroenterology, 2006. 101(8): p. 1773-9.

258. May, A., et al., A prospective randomized trial of two different endoscopic resection techniques for early stage cancer of the esophagus.[see comment]. Gastrointestinal Endoscopy, 2003. 58(2): p. 167-75.

259. Geh, J.I., et al., Systematic overview of preoperative (neoadjuvant) chemoradiotherapy trials in oesophageal cancer: evidence of a radiation and chemotherapy dose response. Radiotherapy & Oncology, 2006. 78(3): p. 236-44.

260. Greer, S.E., et al., Neoadjuvant chemoradiotherapy for esophageal carcinoma: a meta-analysis.[see comment]. Surgery, 2005. 137(2): p. 172-7.

261. Burmeister, B.H., et al., Surgery alone versus chemoradiotherapy followed by surgery for resectable cancer of the oesophagus: a randomised controlled phase III trial.[see comment]. Lancet Oncology, 2005. 6(9): p. 659-68.

262. Duong, C.P., et al., FDG-PET status following chemoradiotherapy provides high management impact and powerful prognostic stratification in oesophageal cancer. European Journal of Nuclear Medicine & Molecular Imaging, 2006. 33(7): p. 770-8.

263. Brink, I., et al., Effects of neoadjuvant radio-chemotherapy on 18F-FDG-PET in esophageal carcinoma. European Journal of Surgical Oncology, 2004. 30(5): p. 544-50.

264. Nakahara, T., et al., Comparison of barium swallow, CT and thallium-201 SPECT in evaluating responses of patients with esophageal squamous cell carcinoma to preoperative chemoradiotherapy. Annals of Nuclear Medicine, 2003. 17(7): p. 583-91.

265. Stahl, M., et al., Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus.[see comment][erratum appears in J Clin Oncol. 2006 Jan 20;24(3):531]. Journal of Clinical Oncology, 2005. 23(10): p. 2310-7.

Page 76: Guideline pour la prise en charge du cancer oesophagien et ...

60 Upper GI Cancer KCE reports 75

266. Bhat, M.A., et al., Use of pedicled omentum in esophagogastric anastomosis for prevention of anastomotic leak.[see comment]. Annals of Thoracic Surgery, 2006. 82(5): p. 1857-62.

267. Hsu, H.-H., et al., Comparison of manual and mechanical cervical esophagogastric anastomosis after esophageal resection for squamous cell carcinoma: a prospective randomized controlled trial. European Journal of Cardio-Thoracic Surgery, 2004. 25(6): p. 1097-101.

268. Walther, B., et al., Cervical or thoracic anastomosis after esophageal resection and gastric tube reconstruction: a prospective randomized trial comparing sutured neck anastomosis with stapled intrathoracic anastomosis.[see comment]. Annals of Surgery, 2003. 238(6): p. 803-12; discussion 812-4.

269. Ajani, J., Review of capecitabine as oral treatment of gastric, gastroesophageal, and esophageal cancers. Cancer, 2006. 107(2): p. 221-31.

270. Sumpter, K., et al., Report of two protocol planned interim analyses in a randomised multicentre phase III study comparing capecitabine with fluorouracil and oxaliplatin with cisplatin in patients with advanced oesophagogastric cancer receiving ECF. British Journal of Cancer, 2005. 92(11): p. 1976-83.

271. Wenger, U., et al., An antireflux stent versus conventional stents for palliation of distal esophageal or cardia cancer: a randomized clinical study. Surgical Endoscopy, 2006. 20(11): p. 1675-80.

272. Shim, C.S., et al., Management of malignant stricture of the esophagogastric junction with a newly designed self-expanding metal stent with an antireflux mechanism. Endoscopy, 2005. 37(4): p. 335-9.

273. Shenfine, J., et al., A pragmatic randomised controlled trial of the cost-effectiveness of palliative therapies for patients with inoperable oesophageal cancer. Health Technology Assessment, 2005. 9(5): p. iii, 1-121.

274. Trumper, M., et al., Efficacy and tolerability of chemotherapy in elderly patients with advanced oesophago-gastric cancer: A pooled analysis of three clinical trials. European Journal of Cancer, 2006. 42(7): p. 827-34.

275. Duffour, J., et al., Safety of cisplatin combined with continuous 5-FU versus bolus 5-FU and leucovorin, in metastatic gastrointestinal cancer (FFCD 9404 randomised trial). Anticancer Research, 2006. 26(5B): p. 3877-83.

276. Moraca, R.J. and D.E. Low, Outcomes and health-related quality of life after esophagectomy for high-grade dysplasia and intramucosal cancer. Arch Surg, 2006. 141(6): p. 545-9; discussion 549-51.

277. Cense, H.A., et al., Effects of prolonged intensive care unit stay on quality of life and long-term survival after transthoracic esophageal resection. Crit Care Med, 2006. 34(2): p. 354-62.

278. Lagergren, P., et al., Health-related quality of life among patients cured by surgery for esophageal cancer. Cancer, 2007. 110(3): p. 686-93.

279. Blazeby, J.M., et al., Health-related quality of life during neoadjuvant treatment and surgery for localized esophageal carcinoma. Cancer, 2005. 103(9): p. 1791-9.

280. Homs, M.Y.V., et al., Single-dose brachytherapy versus metal stent placement for the palliation of dysphagia from oesophageal cancer: multicentre randomised trial. Lancet, 2004. 364(9444): p. 1497-504.

281. Lobo, D.N., et al., Early postoperative jejunostomy feeding with an immune modulating diet in patients undergoing resectional surgery for upper gastrointestinal cancer: a prospective, randomized, controlled, double-blind study. Clinical Nutrition, 2006. 25(5): p. 716-26.

282. Ryan, A.M., et al., Post-oesophagectomy early enteral nutrition via a needle catheter jejunostomy: 8-year experience at a specialist unit. Clin Nutr, 2006. 25(3): p. 386-93.

283. Gabor, S., et al., Early enteral feeding compared with parenteral nutrition after oesophageal or oesophagogastric resection and reconstruction. Br J Nutr, 2005. 93(4): p. 509-13.

284. Lam, E.C., et al., In patients referred for investigation because computed tomography suggests thickened gastric folds, endoscopic ultrasound is superfluous if gastroscopy is normal. American Journal of Gastroenterology, 2007. 102(6): p. 1200-3.

Page 77: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 61

285. Ichikawa, T., et al., Endoscopic ultrasonography with three miniature probes of different frequency is an accurate diagnostic tool for endoscopic submucosal dissection. Hepato-Gastroenterology, 2007. 54(73): p. 325-8.

286. Gines, A., et al., Endoscopic ultrasonography in patients with large gastric folds at endoscopy and biopsies negative for malignancy: predictors of malignant disease and clinical impact. American Journal of Gastroenterology, 2006. 101(1): p. 64-9.

287. Matthes, K., et al., EUS staging of upper GI malignancies: results of a prospective randomized trial.[see comment]. Gastrointestinal Endoscopy, 2006. 64(4): p. 496-502.

288. Chu, K.-M., et al., A prospective evaluation of catheter probe EUS for the detection of ascites in patients with gastric carcinoma. Gastrointestinal Endoscopy, 2004. 59(4): p. 471-4.

289. Deogracias, M.L., et al., Absence of port-site metastases following staging laparoscopy for gastric carcinoma. Revista Espanola de Enfermedades Digestivas, 2006. 98(10): p. 755-9.

290. Lee, S.-H., et al., Clinical efficacy of EMR with submucosal injection of a fibrinogen mixture: a prospective randomized trial. Gastrointestinal Endoscopy, 2006. 64(5): p. 691-6.

291. Ates, E., et al., Perioperative immunonutrition ameliorates the postoperative immune depression in patients with gastrointestinal system cancer (prospective clinical study in 42 patients). Acta Gastroenterologica Belgica, 2004. 67(3): p. 250-4.

292. Romano, F., et al., Biological, histological, and clinical impact of preoperative IL-2 administration in radically operable gastric cancer patients. Journal of Surgical Oncology, 2004. 88(4): p. 240-7.

293. Enzinger, P.C., et al., Impact of hospital volume on recurrence and survival after surgery for gastric cancer. Annals of Surgery, 2007. 245(3): p. 426-34.

294. Inaba, T., et al., Prospective randomized study of two laparotomy incisions for gastrectomy: midline incision versus transverse incision. Gastric Cancer, 2004. 7(3): p. 167-71.

295. Lee, W.J., et al., Randomized clinical trial of Ligasure versus conventional surgery for extended gastric cancer resection. British Journal of Surgery, 2003. 90(12): p. 1493-6.

296. Sasako, M., et al., Left thoracoabdominal approach versus abdominal-transhiatal approach for gastric cancer of the cardia or subcardia: a randomised controlled trial.[see comment]. Lancet Oncology, 2006. 7(8): p. 644-51.

297. Yu, W., G.S. Choi, and H.Y. Chung, Randomized clinical trial of splenectomy versus splenic preservation in patients with proximal gastric cancer. British Journal of Surgery, 2006. 93(5): p. 559-63.

298. Wu, C.-W., et al., Nodal dissection for patients with gastric cancer: a randomised controlled trial.[see comment]. Lancet Oncology, 2006. 7(4): p. 309-15.

299. Adachi, S., et al., Subjective and functional results after total gastrectomy: prospective study for longterm comparison of reconstruction procedures. Gastric Cancer, 2003. 6(1): p. 24-9.

300. Ishikawa, M., et al., Prospective randomized trial comparing Billroth I and Roux-en-Y procedures after distal gastrectomy for gastric carcinoma. World Journal of Surgery, 2005. 29(11): p. 1415-20; discussion 1421.

301. Mochiki, E., et al., Postoperative functional evaluation of jejunal interposition with or without a pouch after a total gastrectomy for gastric cancer. American Journal of Surgery, 2004. 187(6): p. 728-35.

302. Shibata, C., et al., Outcomes after pylorus-preserving gastrectomy for early gastric cancer: a prospective multicenter trial.[see comment]. World Journal of Surgery, 2004. 28(9): p. 857-61.

303. Berardi, R., et al., Gastric cancer treatment: a systematic review. Oncology Reports, 2004. 11(4): p. 911-6.

304. Cascinu, S., et al., Adjuvant treatment of high-risk, radically resected gastric cancer patients with 5-fluorouracil, leucovorin, cisplatin, and epidoxorubicin in a randomized controlled trial.[see comment]. Journal of the National Cancer Institute, 2007. 99(8): p. 601-7.

305. Nishikawa, T. and S. Maetani, Evaluation of intensive adjuvant chemotherapy in gastric cancer using life expectancy compared with log-rank test as a measure of survival benefit. Annals of Surgical Oncology, 2007. 14(2): p. 348-54.

Page 78: Guideline pour la prise en charge du cancer oesophagien et ...

62 Upper GI Cancer KCE reports 75

306. Tentes, A.-A.K., et al., Intraarterial chemotherapy as an adjuvant treatment in locally advanced gastric cancer. Langenbecks Archives of Surgery, 2006. 391(2): p. 124-9.

307. Ueda, Y., et al., A randomized phase III trial of postoperative adjuvant therapy with S-1 alone versus S-1 plus PSK for stage II/IIIA gastric cancer: Hokuriku-Kinki Immunochemo-Therapy Study Group-Gastric Cancer (HKIT-GC). Japanese Journal of Clinical Oncology, 2006. 36(8): p. 519-22.

308. Tsuburaya, A., et al., A randomized phase III trial of post-operative adjuvant oral fluoropyrimidine versus sequential paclitaxel/oral fluoropyrimidine; and UFT versus S1 for T3/T4 gastric carcinoma: the Stomach Cancer Adjuvant Multi-institutional Trial Group (Samit) Trial. Japanese Journal of Clinical Oncology, 2005. 35(11): p. 672-5.

309. Yu, W., A review of adjuvant therapy for resected primary gastric cancer with an update on Taegu's phase III trial with intraperitoneal chemotherapy. European Journal of Surgical Oncology, 2006. 32(6): p. 655-60.

310. Chen, D.W., et al., Role of enteral immunonutrition in patients with gastric carcinoma undergoing major surgery. Asian Journal of Surgery, 2005. 28(2): p. 121-4.

311. Farreras, N., et al., Effect of early postoperative enteral immunonutrition on wound healing in patients undergoing surgery for gastric cancer. Clinical Nutrition, 2005. 24(1): p. 55-65.

312. Sato, Y., et al., A randomized controlled study of immunochemotherapy with OK-432 after curative surgery for gastric cancer. Journal of Immunotherapy, 2004. 27(5): p. 394-7.

313. Di Cosimo, S., et al., Docetaxel in advanced gastric cancer--review of the main clinical trials. Acta Oncologica, 2003. 42(7): p. 693-700.

314. Wohrer, S.S., M. Raderer, and M. Hejna, Palliative chemotherapy for advanced gastric cancer. Annals of Oncology, 2004. 15(11): p. 1585-95.

315. Ajani, J.A., et al., Phase II multi-institutional randomized trial of docetaxel plus cisplatin with or without fluorouracil in patients with untreated, advanced gastric, or gastroesophageal adenocarcinoma.[see comment]. Journal of Clinical Oncology, 2005. 23(24): p. 5660-7.

316. Bouche, O., et al., Randomized multicenter phase II trial of a biweekly regimen of fluorouracil and leucovorin (LV5FU2), LV5FU2 plus cisplatin, or LV5FU2 plus irinotecan in patients with previously untreated metastatic gastric cancer: a Federation Francophone de Cancerologie Digestive Group Study--FFCD 9803. Journal of Clinical Oncology, 2004. 22(21): p. 4319-28.

317. Cocconi, G., et al., Cisplatin, epirubicin, leucovorin and 5-fluorouracil (PELF) is more active than 5-fluorouracil, doxorubicin and methotrexate (FAMTX) in advanced gastric carcinoma. Annals of Oncology, 2003. 14(8): p. 1258-63.

318. Koizumi, W., et al., Randomized phase II study comparing mitomycin, cisplatin plus doxifluridine with cisplatin plus doxifluridine in advanced unresectable gastric cancer. Anticancer Research, 2004. 24(4): p. 2465-70.

319. Lutz, M.P., et al., Weekly infusional high-dose fluorouracil (HD-FU), HD-FU plus folinic acid (HD-FU/FA), or HD-FU/FA plus biweekly cisplatin in advanced gastric cancer: randomized phase II trial 40953 of the European Organisation for Research and Treatment of Cancer Gastrointestinal Group and the Arbeitsgemeinschaft Internistische Onkologie. Journal of Clinical Oncology, 2007. 25(18): p. 2580-5.

320. Moehler, M., et al., Randomised phase II evaluation of irinotecan plus high-dose 5-fluorouracil and leucovorin (ILF) vs 5-fluorouracil, leucovorin, and etoposide (ELF) in untreated metastatic gastric cancer. British Journal of Cancer, 2005. 92(12): p. 2122-8.

321. Ohtsu, A., et al., Randomized phase III trial of fluorouracil alone versus fluorouracil plus cisplatin versus uracil and tegafur plus mitomycin in patients with unresectable, advanced gastric cancer: The Japan Clinical Oncology Group Study (JCOG9205). Journal of Clinical Oncology, 2003. 21(1): p. 54-9.

322. Park, S.H., et al., Paclitaxel versus docetaxel for advanced gastric cancer: a randomized phase II trial in combination with infusional 5-fluorouracil. Anti-Cancer Drugs, 2006. 17(2): p. 225-9.

Page 79: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 63

323. Pozzo, C., et al., Irinotecan in combination with 5-fluorouracil and folinic acid or with cisplatin in patients with advanced gastric or esophageal-gastric junction adenocarcinoma: results of a randomized phase II study. Annals of Oncology, 2004. 15(12): p. 1773-81.

324. Sadighi, S., et al., Quality of life in patients with advanced gastric cancer: a randomized trial comparing docetaxel, cisplatin, 5-FU (TCF) with epirubicin, cisplatin, 5-FU (ECF). BMC Cancer, 2006. 6: p. 274.

325. Takahashi, Y., et al., A randomized phase II clinical trial of tailored CPT-11 + S-1 vs S-1 in patients with advanced or recurrent gastric carcinoma as the first line chemotherapy. Japanese Journal of Clinical Oncology, 2004. 34(6): p. 342-5.

326. Thuss-Patience, P.C., et al., Docetaxel and continuous-infusion fluorouracil versus epirubicin, cisplatin, and fluorouracil for advanced gastric adenocarcinoma: a randomized phase II study. Journal of Clinical Oncology, 2005. 23(3): p. 494-501.

327. Bonnetain, F., et al., Longitudinal quality of life study in patients with metastatic gastric cancer. Analysis modalities and clinical applicability of QoL in randomized phase II trial in a digestive oncology. Gastroenterologie Clinique et Biologique, 2005. 29(11): p. 1113-24.

328. Van Cutsem, E., et al., Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. Journal of Clinical Oncology, 2006. 24(31): p. 4991-7.

329. Dormann, A., et al., Self-expanding metal stents for gastroduodenal malignancies: systematic review of their clinical effectiveness. Endoscopy, 2004. 36(6): p. 543-50.

330. Fiori, E., et al., Palliative management of malignant antro-pyloric strictures. Gastroenterostomy vs. endoscopic stenting. A randomized prospective trial. Anticancer Research, 2004. 24(1): p. 269-71.

331. Mehta, S., et al., Prospective randomized trial of laparoscopic gastrojejunostomy versus duodenal stenting for malignant gastric outflow obstruction. Surgical Endoscopy, 2006. 20(2): p. 239-42.

332. Weston, A.P., et al., Specificity of polymerase chain reaction monoclonality for diagnosis of gastric mucosa-associated lymphoid tissue (MALT) lymphoma: direct comparison to Southern blot gene rearrangement. Dig Dis Sci, 1998. 43(2): p. 290-9.

333. Palazzo, L., et al., Endoscopic ultrasonography in the local staging of primary gastric lymphoma. Endoscopy, 1993. 25(8): p. 502-8.

334. Ferreri, A.J., et al., Low sensitivity of computed tomography in the staging of gastric lymphomas of mucosa-associated lymphoid tissue: impact on prospective trials and ordinary clinical practice. Am J Clin Oncol, 1998. 21(6): p. 614-6.

335. Fusaroli, P., et al., Interobserver agreement in staging gastric malt lymphoma by EUS. Gastrointest Endosc, 2002. 55(6): p. 662-8.

336. Kataoka, M., et al., The role of gallium 67 imaging in non-Hodgkin's lymphoma of the gastrointestinal tract. European Journal of Nuclear Medicine, 1990. 17(3-4): p. 142-7.

337. Haim, N., et al., Intermediate and high-grade gastric non-Hodgkin's lymphoma: a prospective study of non-surgical treatment with primary chemotherapy, with or without radiotherapy. Leuk Lymphoma, 1995. 17(3-4): p. 321-6.

338. Takenaka, T., et al., A prospective study of surgery and adjuvant chemotherapy for primary gastric lymphoma stage II. Br J Cancer, 1997. 76(11): p. 1484-8.

339. Kochi, M., et al., Complete remission by chemotherapy in stage IE-IIE primary gastric lymphoma. Hepatogastroenterology, 2007. 54(76): p. 1285-8.

340. Willich, N.A., et al., Operative and conservative management of primary gastric lymphoma: interim results of a German multicenter study. Int J Radiat Oncol Biol Phys, 2000. 46(4): p. 895-901.

341. Chen, L.T., et al., Prospective study of Helicobacter pylori eradication therapy in stage I(E) high-grade mucosa-associated lymphoid tissue lymphoma of the stomach. J Clin Oncol, 2001. 19(22): p. 4245-51.

Page 80: Guideline pour la prise en charge du cancer oesophagien et ...

64 Upper GI Cancer KCE reports 75

342. Koch, P., et al., Treatment results in localized primary gastric lymphoma: data of patients registered within the German multicenter study (GIT NHL 02/96).[see comment]. J Clin Oncol, 2005. 23(28): p. 7050-9.

343. Ishikura, S., et al., Japanese multicenter phase II study of CHOP followed by radiotherapy in stage I-II, diffuse large B-cell lymphoma of the stomach. Cancer Sci, 2005. 96(6): p. 349-52.

344. Aviles, A., et al., Surgery and chemotherapy versus chemotherapy as treatment of high-grade MALT gastric lymphoma. Medical Oncology, 2006. 23(2): p. 295-300.

Page 81: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 65

8 APPENDICES

APPENDIX 1

GRADE SYSTEM Grade of Recommendation/ Description

Benefit vs. Risk and Burdens Methodological Quality of Supporting Evidence

Implications

1A/ Strong recommendation, high quality evidence

Benefits clearly outweigh risk and burdens, or vice versa

RCTs without important limitations or overwhelming evidence from observational studies

Strong recommendation, can apply to most patients in most circumstances without reservation

1B/ Strong recommendation, moderate quality evidence

Benefits clearly outweigh risk and burdens, or vice versa

RCTs with important limitations (inconsistent results, methodological flaws, indirect, or imprecise) or exceptionally strong evidence from observational studies

Strong recommendation, can apply to most patients in most circumstances without reservation

1C/ Strong recommendation, low quality evidence

Benefits clearly outweigh risk and burdens, or vice versa

Observational studies or case series Strong recommendation, but may change when higher quality evidence becomes available

2A/ Weak recommendation, high quality evidence

Benefits closely balanced with risks and burden

RCTs without important limitations or overwhelming evidence from observational studies

Weak recommendation, best action may differ depending on circumstances or patients’ or societal values

2B/ Weak recommendation, moderate quality evidence

Benefits closely balanced with risks and burden

RCTs with important limitations (inconsistent results, methodological flaws, indirect, or imprecise) or exceptionally strong evidence from observational studies

Weak recommendation, best action may differ depending on circumstances or patients’ or societal values

2C/ Weak recommendation, low quality evidence

Benefits closely balanced with risks and burden

Observational studies or case series Very weak recommendation, other alternatives may be equally reasonable

Page 82: Guideline pour la prise en charge du cancer oesophagien et ...

66 Upper GI Cancer KCE reports 75

APPENDIX 2

IDENTIFIED GUIDELINES AND THEIR QUALITY APPRAISAL Source Title Standardised Methodology Score Final Appraisal American Gastroenterological Association [212]

American Gastroenterological Association Institute Technical Review on the Management of Gastric Subepithelial Masses

22% Not recommended

American Gastroenterological Association [48]

American Gastroenterological Association Technical Review on the Role of the Gastroenterologist in the Management of Esophageal Carcinoma

31% Not recommended

American Society for Gastrointestinal Endoscopy [226]

The role of endoscopy in the assessment and treatment of esophageal cancer 33% Not recommended

American Society for Gastrointestinal Endoscopy [64]

Esophageal dilation 29% Not recommended

National Comprehensive Cancer Network [227]

Esophageal Cancer 57% Not recommended

National Comprehensive Cancer Network [228]

Gastric Cancer 57% Not recommended

Scottish Intercollegiate Guidelines Network [47]

Management of oesophageal and gastric cancer 95% Recommended

Kwaliteitsinstituut voor de Gezondheidszorg [56]

Diagnostiek en behandeling oesofaguscarcinoom 93% Recommended with alterations

Cancer Care Ontario [84] Neoadjuvant or Adjuvant Therapy for Resectable Esophageal Cancer. Practice Guideline Report #2-11.

93% Recommended with alterations

Cancer Care Ontario [108] Combined Modality Radiotherapy and Chemotherapy in the Non-surgical Management of Localized Carcinoma of the Esophagus. Practice Guideline Report #2-12.

93% Recommended with alterations

Cancer Care Ontario [150] Neoadjuvant or Adjuvant Therapy for Resectable Gastric Cancer. Practice Guideline Report #2-14.

93% Recommended with alterations

Fédération Nationale des Centres de Lutte Contre le Cancer [129]

Recommandations pour la pratique clinique: Standards, Options et Recommandations 2003 pour la prise en charge des patients atteints d’adénocarcinomes de l’estomac (cancers du cardia, autres types histologiques exclus) (rapport intégral)

86% Recommended with alterations

Fédération Nationale des Centres de Lutte Contre le Cancer [66]

Recommandations pour la pratique clinique: Standards, Options et Recommandations 2003 pourl’utilisation de la tomographie par émission de positons au [18F]-FDG (TEP-FDG) en cancérologie (rapport intégral)

86% Recommended with alterations

European Society for Medical Oncology [229]

ESMO Minimum Clinical Recommendations for diagnosis, treatment and follow-up of gastric cancer

24% Not recommended

European Society for Medical Oncology [230]

ESMO Minimal Clinical Recommendations for diagnosis, treatment and follow-up of esophageal cancer

24% Not recommended

American College of Gastroenterology [231]

Updated Guidelines for the Diagnosis, Surveillance, and Therapy of Barrett’s Esophagus 27% Not recommended

Allum W et al. [232] Guidelines for the management of oesophageal and gastric cancer 46% Not recommended

Page 83: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 67

APPENDIX 3

OVERVIEW OF SCORES OF EXTERNAL EXPERTS

Page 84: Guideline pour la prise en charge du cancer oesophagien et ...

68 Upper GI Cancer KCE reports 75

Page 85: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 69

Page 86: Guideline pour la prise en charge du cancer oesophagien et ...

70 Upper GI Cancer KCE reports 75

Page 87: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 71

Page 88: Guideline pour la prise en charge du cancer oesophagien et ...

72 Upper GI Cancer KCE reports 75

Page 89: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 73

Page 90: Guideline pour la prise en charge du cancer oesophagien et ...

74 Upper GI Cancer KCE reports 75

Page 91: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 75

Page 92: Guideline pour la prise en charge du cancer oesophagien et ...

76 Upper GI Cancer KCE reports 75

Page 93: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 77

APPENDIX 4

EVIDENCE TABLES OF OESOPHAGEAL CANCER BY CLINICAL QUESTION Diagnosis of oesophageal cancer.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Alarm symptoms

SIGN [47] 2004 Patients presenting with any of the following alarm symptoms should be referred for early endoscopy: dysphagia, recurrent vomiting, anorexia, weight loss, gastrointestinal blood loss.

Meineche-Schmidt V 2002 Numans ME 2001 Wallace MB 2001 Kapoor N 2005

Low

CBO [56] July 2003 Alarmsymptomen, zoals haematemesis en/of melaena, en maagklachten in combinatie met aanhoudend braken, passagestoornissen, ongewild gewichtsverlies of anemie, vormen een indicatie voor een endoscopie. Bij het besluit om een endoscopie te laten verrichten speelt leeftijd een beperkte rol.

Gillen D 1999 Meineche-Schmidt V 2002 Numans ME 2001 Wallace MB 2001

Low

Oesophagogastroscopy

SIGN [47] 2004 Flexible upper GI endoscopy is recommended as the diagnostic procedure of choice in patients with suspected oesophageal cancer.

Graham DY 1982 Abbas SZ 2004

Low

Routine use of chromoendoscopy during upper GI endoscopy is not recommended, but may be of value in selected patients at high risk of oesophageal malignancy.

Inone H 2001 Ragunath K 2003 Kiesslich R 2001 Canto MI 2001

High

Barium X-ray

SIGN [47] 2004 No recommendation, only discussion

Biopsy

SIGN [47] 2004 A minimum of eight biopsies should be taken to achieve a diagnosis of oesophageal malignancy. Lal N 1992 Low

Page 94: Guideline pour la prise en charge du cancer oesophagien et ...

78 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Alarm symptoms

Bowrey DJ 2006 [49] NA Patients referred for open-access gastroscopy

Application of referral guidelines using alarm symptoms

Gastroscopy identified oesophagogastric carcinoma in 123 (3%) of the 4,018 subjects. Of these 123 patients, 104 (85%) with oesophagogastric cancer had ‘‘alarm’’ symptoms (anemia, mass, dysphagia, weight loss, vomiting) and would have satisfied the referral criteria. The remaining 15% would not have been referred for initial endoscopic assessment because their symptoms were those of uncomplicated ‘‘benign’’ dyspepsia. The patients with ‘‘alarm’’ symptoms had a significantly more advanced tumor stage (metastatic disease in 47% vs 11%; p < 0.001), were less likely to undergo surgical resection (50% vs 95%; p<0.001), and had a poorer survival (median, 11 vs 39 months; p = 0.01) than their counterparts without such symptoms.

Prospective study

Low

Marmo R 2005 [233] NA Patients with uncomplicated dyspepsia

Endoscopy Identification of risk factors

A total of 5,224 patients with uncomplicated dyspepsia were considered (training sample). Twenty-two (16 males) had malignancy at endoscopy. These patients were about 20 yr older than patients with no malignancy (p < 0.001). The mean age of females with cancer was almost 10 yr higher compared to males (p= 0.08). Such differences in age were confirmed in a split sample of 3,684 patients (p < 0.001 and p < 0.05, respectively). The age cut-offs identified were 35 yr for males and 56 yr for females.

Prospective study

Low

Oesophagogastroscopy

No additional studies found

Barium X-ray

No additional studies found

Biopsy

No additional studies found

Page 95: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 79

Staging of patients with oesophageal cancer.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

CT scan

SIGN [47] 2004 In patients with oesophageal cancer, CT scan of the chest and abdomen with IV contrast and gastric distention with oral contrast or water should be performed routinely. The liver should be imaged in the portal venous phase.

Kamel IR 2004 Low

CBO [56] July 2003 CT van de thoraxapertuur tot en met de bovenbuik kan worden gebruikt om de aanwezigheid van mediastinale en/of truncale lymfkliermetastasen, alsook mogelijke metastasen in longen, lever, bijnieren of skelet vast te stellen. Hierbij dient zowel gering oraal contrast ter markering van het oesofaguslumen als intraveneus contrast te worden toegepast. De coupedikte dient kleiner dan of gelijk aan 5 mm te zijn.

Romagnuolo J 2002 Wakelin SJ 2002 van Overhagen H 1993 van den Hoed RD 1997

Low

Endoscopic ultrasonography

SIGN [47] 2004 Patients with oesophageal or oesophagogastric junction cancers who are candidates for any curative therapy should have their tumours staged with endoscopic US +/- fine needle aspiration.

Kelly S 2001 (SR) Vazquez-Sequeiros E 2003 Weaver SR 2004

Moderate

CBO [56] July 2003 Endoscopische ultrasonografie is zinvol om de uitbreiding van de laesie in de diepte en mogelijke ingroei in omgevende structuren (bijvoorbeeld aorta) (T-status) en de aanwezigheid van vergrote mediastinale en/of truncale lymfklieren te bepalen (N- en M-status).

Eloubeidi MA 2001 Catalano MF 1995 Vazquez-Sequeiros E 2003 Heidemann J 2000 Wiersema MJ 1997 Williams DB 1999

Low

Laparoscopy

SIGN [47] 2004 Laparoscopy should be considered in patients with oesophageal tumours with a gastric component. Clements DM 2004 Molloy RG 1995 Wilkiemeyer MB 2004

Low

CBO [56] July 2003 Minimaal invasieve chirurgische stadiëring met behulp van thoracoscopie en laparoscopie is geïndiceerd bij afwijkingen in de oesofagus van onduidelijke aard, gevonden bij niet-invasieve stadiëring. Voorts kan laparoscopie worden overwogen bij carcinomen op de gastro-oesofageale overgang en cardiacarcinomen die als T3/T4 worden gestadieerd met een niet-invasieve techniek.

Krasna MJ 1995 & 2002 Whyte RI 2001 Wakelin SJ 2002 Romijn MG 1998

Low

Suspecte cervicale, mediastinale en truncale klieren dienen onder beeldvorming ((endoscopische) ultrasonografie) cytologisch te worden gepuncteerd indien de status van de desbetreffende lymfklieren voor beleidsbepaling relevant is.

Bonvalot S 1996 Natsugoe S 1999 van Overhagen H 1993

Low

Magnetic resonance imaging

SIGN [47] 2004 MRI should be reserved for those patients who cannot undergo CT or used for additional investigation following CT/EUS.

Sohn KM 2000 Wu LF 2003 Kersjes 1997 Lauenstein TC 2004 Wong R 2000

Low

Page 96: Guideline pour la prise en charge du cancer oesophagien et ...

80 Upper GI Cancer KCE reports 75

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Bronchoscopy

SIGN [47] 2004 Bronchoscopy +/- bronchoscopic US +/- biopsy should be undertaken in patients with clinical or imaging features suspicious of tracheobronchial invasion.

Aggarwal AN 2003 Nguyen NT 2001 Nishimura Y 2002 Riedel M 2001

Low

CBO [56] July 2003 Bronchoscopie, inclusief brush-cytologie, van suspecte gebieden is onderdeel van de stadiëring van oesofaguscarcinomen op of craniaal van het niveau van de carina.

Baisi A 1999 Choi TK 1984

Low

Thoracoscopy

SIGN [47] 2004 Thoracoscopy may be considered for patients where a tissue diagnosis of suspicious nodes (not possible by either EUS or CT guided techniques) is required to determine optimum management.

Krasna MJ 2002 Low

CBO [56] July 2003 Minimaal invasieve chirurgische stadiëring met behulp van thoracoscopie en laparoscopie is geïndiceerd bij afwijkingen in de oesofagus van onduidelijke aard, gevonden bij niet-invasieve stadiëring. Voorts kan laparoscopie worden overwogen bij carcinomen op de gastro-oesofageale overgang en cardiacarcinomen die als T3/T4 worden gestadieerd met een niet-invasieve techniek.

Krasna MJ 1995 & 2002 Whyte RI 2001 Wakelin SJ 2002 Romijn MG 1998

Low

Suspecte cervicale, mediastinale en truncale klieren dienen onder beeldvorming ((endoscopische) ultrasonografie) cytologisch te worden gepuncteerd indien de status van de desbetreffende lymfklieren voor beleidsbepaling relevant is.

Bonvalot S 1996 Natsugoe S 1999 van Overhagen H 1993

Low

PET scan

SIGN [47] 2004 PET is not routinely indicated in the staging of oesophageal cancers. van Westreenen HL 2004 Low CBO [56] July 2003 FDG-PET bij T3-oesofaguscarcinomen kan worden overwogen om eventuele afstandsmetastasen vast te

stellen. van Westreenen HL 2004

Low

FNCLCC [66] October 2002

La TEP-FDG est indiquée, est complémentaire du scanner et de l’écho-endoscopie, pour l’évaluation préthérapeutique du statut ganglionnaire et métastatique des cancers de l’oesophage.

Kinkel 2002 Depotter 2002 McAteer 1999 Hoffmann 1999 Couper 1998

Low

Neck imaging

SIGN [47] 2004 Neck imaging either by US or CT is recommended as part of the staging of oesophageal cancer. Griffith JF 2000 Low CBO [56] July 2003 Echografie van de hals is nuttig om cervicale lymfkliermetastasen te bepalen. Bonvalot S 1996

Natsugoe S 1999 van Overhagen H 1993

Low

Suspecte cervicale, mediastinale en truncale klieren dienen onder beeldvorming ((endoscopische) ultrasonografie) cytologisch te worden gepuncteerd indien de status van de desbetreffende lymfklieren voor beleidsbepaling relevant is.

Bonvalot S 1996 Natsugoe S 1999 van Overhagen H 1993

Low

Page 97: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 81

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

CT scan

Kim SH 2006 [234] NA Patients with suspected or known oesophageal neoplasms (n = 23)

3D Multidetector CT and CT oesophagography (n = 23) Conventional barium study (n = 20) Conventional endoscopy (n = 23) Histology (surgery: n = 12; endoscopic biopsy: n = 11)

Sensitivity MDCT: 91% Overall accuracies for T and N staging were 42.9% and 85.7% respectively

Not very clear if prospective

Observational study

Very low

Umeoka S 2006 [235] NA 28 consecutive patients with oesophageal cancer histopathologically proved at endoscopic biopsy (31 lesions)

Triple-phase dynamic contrast-enhanced CT Standard: histopathology (23 lesions) or EUS (8 lesions)

One lesion not identifiable at CT Insufficient data to calculate accuracy

Same standard not applied to all patients

Prospective observational study

Very low

Panebioanco V 2006 [236] NA 39 patients with oesophageal cancer proved by means of endoscopy

Dynamic CT of the chest and abdomen with the aid of 3D rendering Standard: histopathology (26 patients)

T staging: sensitivity 92%, accuracy 88% N staging: sensitivity 85%, specificity 58%, accuracy 69%

Standard not applied to all patients

Prospective observational study

Very low

Onbas O 2006 [237] NA 44 patients with oesophageal cancer diagnosed by conventional endoscopy and biopsy specimens

Multidetector CT (n = 24) Virtual endoscopy (n = 27) Standard: histopathology (n = 24)

MDCT: correct T staging in 22/24 patients, overstaging in 1 pt and understaging in 1 pt; correct N staging in 20/24 patients, overstaging in 2 pts and understaging in 2 pts

Prospective observational study

Very low

Lowe VJ 2005 [59] NA 69 patients with newly diagnosed oesophageal cancer

FDG-PET CT EUS Histology (endoscopic biopsy: n = 55; surgery n = 14)

Correct T staging: 42% with PET and CT, 71% with EUS Sensitivity and specificity for nodal involvement: 84% and 67% for CT, 86% and 67% for EUS, 82% and 60% for PET Sensitivity and specificity for distant metastasis: 81% and 82% for CT, 73% and 86% for EUS, and 81% and 91% for PET

Prospective observational study

Low

Page 98: Guideline pour la prise en charge du cancer oesophagien et ...

82 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Weaver SR 2004 [238] NA 60 consecutive patients with histologically proven cancer of the oesophagus, established by endoscopy and biopsy, that underwent surgery

Spiral CT EUS Standard: histology

Sensitivity for T and N stages was 58% and 79% for CT, and 72% and 91% for EUS. Specificity for T and N stages was 80% and 84% for CT, and 85% and 68% for EUS.

Included in SIGN guideline

Prospective observational study

Low

Yoon YC 2003 [239] NA 81 patients with squamous cell carcinoma of the oesophagus

CT and FDG-PET Standard: histopathology

T staging: primary tumour was correctly detected in 65 patients (80%) at CT and in 74 patients (91%) at FDG PET N staging: CT: sensitivity 11%, specificity 95% PET: sensitivity 30%, specificity 90%

Strong selection of 136 patients with oesophageal cancer Included in SR of Van Westreenen et al.

Prospective observational study

Very low

Endoscopic ultrasonography

van Vliet EP 2007 [61] February 2006

Patients with oesophageal cancer

EUS T staging: median accuracy 83% (53-94%) N staging: median accuracy 76% (54-94%), median sensitivity 77% (37-100%), median specificity 74% (50-90%) No evidence for publication bias

54 articles included Medline search only English only No quality appraisal

Systematic review

Low

Vazquez-Sequeiros E 2006

[240] NA Patients (n = 144) with adenocarcinoma or squamous-cell carcinoma of the oesophagus, M0 (helical CT) and who were candidates for surgical resection; no neoadjuvant treatment

Helical CT EUS (standard vs. modified criteria) EUS-FNAC Standard: surgical pathology result (n = 47) or malignant cytology result (n = 97)

Presence of all 7 modified criteria: specificity and PPV of 100% Presence of no modified criteria: sensitivity and NPV of 100%

Blinded evaluation Standard not the same for all patients

Prospective observational study

Low

May A 2004 [241] NA Patients with a suspicion of early oesophageal adenocarcinoma (n = 81) or squamous cell carcinoma (n = 19)

High-resolution video endoscopy HR-EUS with a 20 MHz miniprobe Standard: histological examination of the endoscopically or surgically resected tumour

Accuracy of staging: HR-E 83.4% vs. HR-EUS 79.6% Mucosal tumours: Sensitivity HR-E 94.1% vs. HR-EUS 91.2% Submucosal tumours: Sensitivity HR-E 56% vs. HR-EUS 48% Combination of HR-E and HR-EUS: 60%

Blinded evaluation Prospective observational study

Low

Yanai H 2004 [242] NA Patients with endoscopically staged

20-MHz thin ultrasound probe EUS

Invasion depth correctly staged in 20/31 pts (64.5%)

In calculation exclusion of 2 dysplasia pts and 2 non-

Prospective observational

Very low

Page 99: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 83

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

superficial oesophageal cancer (n = 31)

Standard: histological examination of the endoscopically or surgically resected tumour

resection cases study

Chang KJ 2003 [243] NA Patients with oesophageal cancer (n = 60)

CT EUS EUS-FNAC if positive FNAC would upstage the tumour Standard: surgical pathology result

T staging: accuracy EUS 83% N staging: Accuracy EUS 83% Accuracy EUS-FNAC 89%, PPV 100%

Standard not applied to all patients 2 pts lost-to-follow-up

Prospective observational study

Very low

Mariette C 2003 [244] NA 150 patients who underwent EUS for staging of tumours of the oesophagus (out of 372 patients who underwent surgery with curative intent)

EUS Standard: surgical pathology result

Patient survival was significantly related to tumour (EUS T), node (EUS N) and Union Internacional Contra la Cancrum classification (EUS UICC) stage according to sonographic findings (P = 0·003, P = 0·009 and P = 0·004 respectively), and the presence of stenosis determined by EUS (P = 0·004). EUS T stage was a prognostic factor for survival (RR 1.7, 95%CI 1.1 to 3.0; P = 0·046). Complete surgical resection (R0) was also significantly related to EUS T, N and UICC classification (P < 0·001). EUS UICC stage was a factor predictive of R0 resection (RR 2.6, 95%CI 1.4 to 4.8; P = 0·003).

Doubts about prospective design Not enough data to calculate staging accuracy

Prospective (?) observational study

Very low

Laparoscopy

Mortensen MB 2006 [79] NA 411 patients with upper gastrointestinal tract cancer, of which 95 had biopsy-proven oesophageal cancer

Combined endoscopic ultrasonography (EUS) and laparoscopic ultrasonography (LUS) ‘Standard’: treatment actually undertaken

The combination of EUS and LUS correctly predicted R0 resection in 90.6%, R1–R2 in 91% and irresectability in 91.4% of patients. Ten patients (2.4%) had explorative laparotomy only. There were no complications associated with the EUS and LUS procedures.

To avoid false-positive findings with regard to local irresectability ultrasonographic criteria were adjusted so that only a combination of several ultrasonographic criteria with a 100 per cent predictive value for local irresectability were used Standard is not identical

Prospective study

Very low

Page 100: Guideline pour la prise en charge du cancer oesophagien et ...

84 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

for all patients Menon KV 2003 [245] NA 133 out of 320

consecutive patients with histologically proven carcinoma of the oesophagus or cardia, without clinical or radiological evidence (CT) of metastases and deemed fit for potential excisional surgery

Multiport laparoscopy Standard: histology or subsequent surgery

Laparoscopy detected incurable disease in 31 patients (24%), some of whom had more than one contraindication to surgery, including hepatic metastases (n = 10), peritoneal metastases (n = 12) and malignant small volume ascites (n = 5). Lymph node metastases were confirmed histologically by biopsy at laparoscopy in 26 patients (fixed nodes, n = 14; mobile nodes, n = 12). Sensitivity for the detection of liver and peritoneal metastases was 100%, and lymph node metastases were 83%. Specificity for detection of hepatic metastases was 99%, 100% for peritoneal metastases and 82% for lymph node metastases. Ninety-nine patients proceeded to definitive surgery and only two were unresectable.

Standard is not identical for all patients

Prospective study

Very low

Magnetic resonance imaging

Riddell AM 2006 [246] NA 10 patients with confirmed oesophageal carcinoma (7 with adenocarcinoma, 2 with squamous cell carcinoma, and 1 with spindle-cell melanoma), who were deemed medically fit for surgery and shown to have resectable disease using endoscopic sonography and CT

High-resolution MRI of primary tumour Standard: histology

50 MRI images (5 per patient) 7 images underestimated, 20 images overestimated Tumor position was correctly diagnosed by MRI in 5 patients Extramural extension: 2 false negatives

Very small sample size Prospective study

Very low

Bronchoscopy

Wakamatsu T 2006 [77] NA 54 surgical patients with suspected thyroid or oesophageal cancer (n =

CT neck/upper mediastinum MRI neck/upper mediastinum Bronchoscopy with EBUS

The sensitivity and specificity of CT, MRI and EBUS for invasion were 59 and 56, 75 and 73, and 92 and 83%, respectively.

No separate numbers for oesophageal cancer patients

Prospective study

Very low

Page 101: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 85

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

24) and a mass adjacent to the tracheobronchial wall on chest or neck CT or MRI

Standard: histology and/or clinical follow-up

The accuracy of EBUS was significantly greater than that of CT in the present study (p = 0.0011). The accuracy of EBUS was significantly different from that of CT and MRI in the surgically treated patients (p = 0.005 and p = 0.032, respectively).

Standard not same for all patients

Osugi H 2003 [76] NA Patients with advanced squamous cell carcinoma in contact with the trachea, the main bronchi, or both (n = 66)

BUS CT 50 thoracotomies

BUS: sensitivity 91%, specificity 89%, accuracy 90% CT: sensitivity 69%, specificity 55%, accuracy 62%

Sensitivity and specificity calculated with 50 thoracotomy patients

Prospective study (?)

Very low

Thoracoscopy

No additional diagnostic studies found

PET scan

Meyers BF 2007 [247] NA Patients with resectable, biopsy-proven carcinoma of the thoracic oesophagus (n = 189)

FDG-PET Chest and abdominal CT Additional study or biopsy if positive PET finding

The reasons for no resection included the following: M1 disease found by PET and confirmed (9), M1 disease found by PET and not confirmed (2), M1 disease at exploration not found by PET (7), decline or death before surgery (10), patient refusal of surgery (7), unresectable local tumour at exploration (5), and extensive N1 disease precluding operation (2). Eight (4.2%) patients undergoing resection had a recurrence in the first 6 months.

Standard not applied to all patients

Prospective study

Very low

Ott K 2006 [72] NA 65 patients with biopsy proven adenocarcinoma of the distal oesophagus or cardia (stage uT3)

PET before preoperative chemotherapy and on day 14 of first chemotherapy cycle

Metabolic responders showed a high histopathologic response rate (44%) with a 3-year survival rate of 70%. In contrast, prognosis was poor for metabolic nonresponders with a histopathologic response rate of 5% (P = 0.001) and a 3-year survival rate of 35% (P = 0.01). A multivariate analysis (covariates: ypT-, ypN-category, histopathologic response) demonstrated that metabolic response was the only factor predicting recurrence (P = 0.018)

9 patients excluded from analysis (8 too low FDG uptake, 1 with hyperglycemia)

Prospective study

Low

Page 102: Guideline pour la prise en charge du cancer oesophagien et ...

86 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

in the subgroup of completely resected (R0) patients.

Yuan S 2006 [75] NA Patients with a first diagnosis of biopsy-proven squamous cell cancer of the thoracic oesophagus, operable (including M1a), not refusing surgery, and willing to pay at least 30% of the charge for PET/CT (n = 45)

PET/CT Standard: histology

The sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of PET/CT were 93.90% (77/82 nodal groups), 92.06% (290/315), 92.44% (367/397), 75.49% (77/102), and 98.31% (290/295), respectively, whereas those of PET were 81.71% (67/82), 87.30% (275/315), 86.15% (342/397), 62.62% (67/107), and 94.83% (275/290), respectively. P values were 0.032, 0.067, 0.006, 0.063, and 0.037, respectively. The differences in sensitivity, accuracy, and negative predictive value between PET and PET/CT were statistically significant.

Prospective study

Low

Van Baardwijk A 2006

[248] August 2005 Patients with oesophageal cancer

PET for radiotherapy treatment planning

In oesophageal cancer, PET scan can be used to include PET positive lymph nodes in the target volume.

Medline only No information on study selection and quality appraisal

SR Low

Leong T 2006 [70] NA Patients with histologically proven squamous or adenocarcinoma of the oesophagus stages I–III and ECOG performance status 0–2, where the treatment intent was radical chemoradiotherapy (radiation dose 50 Gy) following completion of all non-PET staging investigations that did not reveal evidence of metastatic disease (n = 23)

PET/CT for radiotherapy planning vs. CT

The addition of PET information altered the clinical stage in 8 of 21 eligible patients enrolled on the study (38%); 4 patients had distant metastatic disease and 4 had unsuspected regional nodal disease. Sixteen patients proceeded to the radiotherapy planning phase of the study and received definitive chemoradiation planned with the PET/CT data set. The GTV based on CT information alone excluded PET-avid disease in 11 patients (69%), and in five patients (31%) this would have resulted in a geographic miss of gross tumour. The discordance between CT and PET/CT was due mainly to differences in defining the longitudinal extent of disease in the oesophagus. The cranial extent of the primary tumour as defined

Patients with PET detected metastatic disease came off study with no further data collection, while patients who did not have metastatic disease on PET and were deemed suitable for radical treatment proceeded to the radiotherapy planning phase of the study. Two patients excluded

Prospective study

Low

Page 103: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 87

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

by CT vs. PET/CT differed in 75% of cases, while the caudal extent differed in 81%.

Levine EA 2006 [249] NA 64 consecutive patients, with potentially curable, locally advanced oesophageal cancer

PET pre- and post-chemoradiation (of the 44 patients undergoing oesophagectomy following chemoradiation, 31 patients had both a pre-chemoradiation and post-chemoradiation PET, while 13 patients had a single PET scan: 10 patients of which had a pre-chemoradiation study and 3 of which had post-chemoradiation imaging)

Response was as follows: pCR 27%, pathologic residual microscopic (pCRmicro) 14.5%, partial response 19%, and stable or progressive disease 39.5%. A pretreatment standardized uptake value (SUVmax1 hour) ≥15 was associated with an observed 77.8% significant response (pCR + pCRmicro) compared with 24.2% for patients with a pretreatment SUVmax1 hour <15 (P = 0.005). Significant response was observed in 71.4% of patients with a decrease in SUVmax1 hour ≥10 compared with 33.3% when the SUVmax1 hour decreased<10 (P = 0.004).

Patients not treated similarly (different chemotherapy schedule, surgery not for all patients, etc.) Standard not similar for all patients

Prospective study (?)

Very low

Kato H 2005 [250] NA 44 patients with thoracic oesophageal cancer

FDG-PET Bone scintigraphy

PET: sensitivity 92%, specificity 94% Bone scintigraphy: sensitivity 77%, specificity 84%

Not clear if prospective Prospective (?) study

Very low

Moureau-Zabotto L 2005

[71] NA Thirty-four oesophageal carcinoma patients referred for concomitant radiotherapy and chemotherapy with radical intent

PET-CT vs. CT for radiotherapy treatment planning

PET identified previously undetected distant metastatic disease in 2 patients, making them ineligible for curative conformal radiotherapy. Modifications of the GTV affected the planning treatment volume in 18 patients. Modifications of the delineation of the GTV and displacement of the isocenter of the planning treatment volume by FDG-PET also affected the percentage of total lung volume receiving >20 Gy in 25 patients (74%), with a dose reduction in 12 patients and dose increase in 13.

Prospective study

Low

Song SY 2005 [74] NA Patients with locally advanced but resectable oesophageal cancer (n = 32)

PET before and after neoadjuvant chemoradiation

Five patients (16%) were PET negative before chemoradiotherapy, which indicates a sensitivity of 84% as a diagnostic procedure for both the

Prospective study

Low

Page 104: Guideline pour la prise en charge du cancer oesophagien et ...

88 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

oesophagus and lymph nodes. Van Westreenen HL 2005

[251] NA Ten patients with biopsy-proven cancer of the oesophagus or gastro-oesophageal junction

18F-FLT PET vs. 18F-FDG PET Standard: histology

18F-FDG PET was able to detect all oesophageal cancers, whereas 18FFLT PET visualized the tumor in 8 of 10 patients. Both 18F-FDG PET and 18F-FLT PET detected lymph node metastases in 2 of 8 patients. 18F-FDG PET detected 1 cervical lymph node that was missed on 18F-FLT PET, whereas 18F-FDG PET showed uptake in benign lesions in 2 patients.

Small sample size Prospective study

Very low

Vrieze O 2004 [252] NA 30 patients with an advanced oesophageal carcinoma (cT4 based on EUS and/or CT), M0

PET before neoadjuvant chemoradiation therapy (25 patients treated with surgery)

In 14 of the 30 patients (47%) discordances were found in detection of the pathological lymph nodes between CT/EUS and FDGPET. In 8 patients, 9 lymph node regions were found with pathologic nodes on conventional imaging only. In three of these patients the influence of FDG-PET findings would have led to a decrease of the irradiated volume. In 6 patients, 8 lymph node regions were found with a normal CT/EUS and pathologic nodes on FDG-PET. In three of these patients (10%) the influence of the FDG-PET would have led to enlargement of the irradiated volume.

Included in SR of Van Baardwijk et al. One centre analysis of prospective study of 36 patients No comparison with histology

Prospective study

Very low

Sihvo EI 2004 [73] NA Patients with histologically proves adenocarcinoma of the oesophagus (n = 20) or the EG junction (n = 35)

Routine staging with endoscopy, CT and EUS FDG-PET Histopathology

Accuracy N staging: PET 60%, EUS 72%, CT 58% Accuracy M staging: PET 76%, CT 75%

Prospective study

Low

Choi JY 2004 [68] NA Eighty-nine consecutive patients having newly diagnosed oesophageal cancer, undergoing curative surgery

PET preoperatively There were 130 malignant nodal groups of 554 dissected nodal groups in 43 of 69 patients on pathologic examination. Ninety-six positive lymph nodes in 43 patients were evident on 18F-FDG PET. Only 30 lymph nodes were positive in 24 patients on CT. EUS revealed lymph

Prospective study

Low

Page 105: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 89

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

node metastases in 27 patients. Kneist W 2004 [69] NA Patients with biopsy-

proved oesophageal carcinoma (n = 81)

PET and computer tomography (CT) of the chest and abdomen (and of the neck in 45 patients) within 45 days. The findings of the histopathological examination for 31 suspicious lesions from 28 patients served as the basis for the diagnosis of distant metastasis.

The PET findings had a higher specificity (89% vs 11%) but a lower sensitivity (38% vs 63%) than CT findings in the detection of metastatic sites. The CT results showed greater agreement with histopathological findings than did PET results. In 8 patients (10%), PET detected distant metastases that were not identified with CT. In 4 patients (5%), PET detected bone metastases only, but in all of these patients metastases in other locations were detected by CT. Although PET led to upstaging (M1) in 2 patients (2%), it did not enable the exclusion of oesophageal resection.

Prospective study

Low

Neck imaging

No additional diagnostic studies found

Page 106: Guideline pour la prise en charge du cancer oesophagien et ...

90 Upper GI Cancer KCE reports 75

Diagnosis and treatment of early lesions.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 Reduction of risk of progression to adenocarcinoma is not an indication for anti-reflux surgery in patients with Barrett’s oesophagus.

Spechler SJ 2001 Corey KE 2003 Parrilla P 2003

High

Routine use of chromoendoscopy during upper GI endoscopy is not recommended, but may be of value in selected patients at high risk of oesophageal malignancy.

Inone H 2001 Ragunath K 2003 Kiesslich R 2001 Canto MI 2001 Mitooka H 1995

Moderate

In patients with Barrett’s oesophagus there should be a structured biopsy protocol with quadrantic biopsies every two centimetres and biopsy of any visible lesion.

Garside R 2006 Fitzgerald RC 2001 Conio M 2003 Macdonald CE 2000

Low

Pathologists should follow the revised Vienna classification for reporting dysplasia. Montgomery E 2001a Schlemper RJ 2000

Low

Where radical intervention is contemplated on the basis of high-grade dysplasia or early adenocarcinoma the diagnosis should be validated by a second pathologist experienced in this area and further biopsies should be taken if there is uncertainty.

Montgomery E 2001a Baak JP 2002 Montgomery E 2001b

Low

Evaluation of suspected high-grade dysplasia in Barrett’s oesophagus biopsies should be undertaken with knowledge of the clinical and endoscopic background and biopsies should be reviewed at a multidisciplinary meeting with access to the clinical information.

Baak JP 2002 Montgomery E 2001b

Low

Patients diagnosed with high-grade dysplasia should have careful assessment (CT, EUS, rigorous biopsy protocol +/- EMR) to exclude coexisting cancer.

Schnell TG 2001 Levine DS 1993 May A 2002

Low

In the absence of invasive cancer, patients with high-grade dysplasia should be offered endoscopic treatment.

May A 2002 Ell C 2000 Pacifico RJ 2003

Low

The assessment and management of patients with high-grade dysplasia should be centralised to units with the appropriate endoscopic facilities and expertise.

Ell C 2000 Low

Superficial oesophageal cancer limited to the mucosa should be treated with EMR. Fujita H 2001 Inoue H 1991 Takekoshi T 1994

Low

Mucosal ablative techniques, such as PDT, APC, or laser should be reserved for the management of residual disease after EMR and not for initial management if invasive disease is present in patients fit for surgery.

May A 2002 Ell C 2000 Pacifico RJ 2003 Attwood SE 2003

Low

CBO [56] July 2003 Ondanks de relatief lage interobserveerdersvariatie verdient het aanbeveling dat de histopathologische diagnose van hooggradige dysplasie (carcinoma in situ) en vroegcarcinoom in de oesofagus door een patholoog met ervaring op dit gebied wordt gesteld.

Schlemper 2000a Schlemper 2000b Montgomery 2001

Low

Page 107: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 91

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Skacel 2000 Chromoscopie met lugolkleuring is geïndiceerd bij patiënten met een neoplastische afwijking in het

plaveiselepitheel van de oesofagus waarbij endoscopische therapie wordt overwogen. Dawsey 1998 Low

Bij patiënten met een neoplastische afwijking in een Barrett-oesofagus gelden de volgende aanbevelingen in de endoscopische work-up voor eventuele endoscopische therapie:

- Endoscopische inspectie geschiedt bij voorkeur in een centrum met endoscopische expertise in de detectie van vroege afwijkingen in Barrett-oesofagus.

- Hierbij wordt bij voorkeur gebruikgemaakt van hoge-resolutie-endoscopie, eventueel aangevuld met chromoscopie.

- Geadviseerd wordt om biopten te nemen uit alle zichtbare mucosale onregelmatigheden en at random uit vier kwadranten voor elke centimeter Barrett-oesofagus, te beginnen 1 cm boven de bovenrand van de maagplooien en doorlopend tot aan de overgang van cilindrisch naar plaveiselepitheel.

Dave U 2001 Endo T 2002 Guelrud M 2001 Sharma P 2003 Reid BJ 2000

Low

Bij alle patiënten met een neoplastische afwijking in de oesofagus bij wie endoscopische therapie wordt overwogen, dient een endoscopische ultrasonografie te worden verricht om diepte-infiltratie van de afwijking en verdachte lokale lymfklieren uit te sluiten.

Scotiniotis IA 2001 Hasegawa N 1996 Natsugoe S 1996

Low

Aanvullende stadiëring met CT-scan is optioneel. Bergman JJ 1999 Scotiniotis IA 2001

Low

Voor patiënten met hooggradige dysplasie en/of vroegcarcinoom in plaveiselepitheel van de oesofagus heeft endoscopische behandeling de voorkeur. Hierbij zijn de volgende voorwaarden van belang:

- Voorafgaande aan de behandeling worden een endoscopische beoordeling en stadiëring verricht. - Het betreft een solitaire, vlakke afwijking met een maximale diameter van 2-3 cm.

Shimizu Y 2002 Fujita H 2001 Narahara H 2000 Nijhawan PK 2000

Low

Als endoscopische resectietechniek verdient de endoscopische mucosale resectie-cap-techniek de voorkeur, zodat histologisch onderzoek van de verwijderde afwijking mogelijk is.

Inoue H 1991 Low

Het besluit om de endoscopische behandeling als curatief te beschouwen, kan pas worden genomen nadat het endoscopische mucosale resectiepreparaat histologisch is beoordeeld.

Bij patiënten met evidente submucosale ingroei en/of positieve resectieranden van het endoscopische mucosale resectiepreparaat dient in principe een chirurgische resectie te worden verricht.

Er vindt initieel een endoscopisch onderzoek plaats dat specifiek dient om in de Barrett-oesofagus een eventueel invasief carcinoom te detecteren: - Hierbij geniet hoge-resolutie-endoscopie, eventueel aangevuld met chromoscopie, de voorkeur. - Er worden biopten genomen uit alle zichtbare mucosale onregelmatigheden en random uit vier kwadranten voor elke centimeter Barrett-oesofagus, te beginnen 1 cm boven de bovenrand van de maagplooien en doorlopend tot aan de overgang tussen cilinder- en plaveiselepitheel.

Cfr supra

Na de work-up wordt de endoscopische surveillance als volgt verricht: - Frequentie surveillance: in het eerste jaar elke drie maanden, in het tweede jaar elke zes maanden en daarna tenminste jaarlijks. - Indien er tijdens surveillance sprake is van nieuwe afwijkingen zoals erosies of een onregelmatig oppervlak, dienen opnieuw biopten te worden genomen en/of dient dit te worden uitgebreid met een

No evidence

Page 108: Guideline pour la prise en charge du cancer oesophagien et ...

92 Upper GI Cancer KCE reports 75

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

diagnostische endoscopische mucosale resectie. Endoscopische surveillance (ten minste gedurende het eerste jaar) wordt bij voorkeur verricht in een

centrum met ervaring. Cfr supra

Voor geselecteerde patiënten met hooggradige dysplasie en/of vroegcarcinoom in een Barrett-oesofagus heeft endoscopische behandeling de voorkeur. Hierbij gelden de volgende voorwaarden:

- Voorafgaande aan de behandeling zijn endoscopische beoordeling en stadiëring verricht. - Het betreft een solitaire verheven en/of vlakke afwijking met een maximale grootte van 2 cm.

Buttar NS 2001 May A 2002

Low

De endoscopische behandeling omvat als primaire behandeling een endoscopische resectietechniek (bij voorkeur de endoscopische mucosale resectie-cap-techniek), waardoor histologisch onderzoek van de verwijderde afwijking mogelijk is.

Buttar NS 2001 May A 2002

Low

Het besluit om endoscopische behandeling als in opzet curatieve behandeling te beschouwen, kan pas worden genomen nadat het endoscopische mucosale resectiepreparaat is beoordeeld.

Bij geselecteerde patiënten kan aanvullende ablatietherapie na voorafgaande endoscopische mucosale resectie worden overwogen.

Bij afwijkingen groter dan 2 cm en/of de helft van de circumferentie is ‘piece-meal-resectie vereist. Ell C 2000 Low De histopathologische beoordeling van de endoscopische mucosale resectiepreparaten dient door een

patholoog met ervaring op dit gebied te geschieden. Referentiepanels voor een ‘second opinion’ zijn in Nederland aanwezig.

Schlemper 2000a Schlemper 2000b

Low

Bij submucosale ingroei van een vroegcarcinoom in plaveiselcelepitheel dient alsnog een chirurgische resectie te worden overwogen gezien de hoge kans (26-47%) op lokale lymfkliermetastasen.

Multiple studies Low

Bij submucosale ingroei van een vroegcarcinoom in een Barrett-oesofagus dient alsnog een chirurgische resectie te worden overwogen gezien de hoge kans (tot 50%) op lokale lymfkliermetastasen.

Multiple studies Low

Na endoscopische behandeling van hooggradige dysplasie/vroegcarcinoom in de oesofagus gelden de aanbevelingen voor de endoscopische follow-up conform de aanbevelingen voor endoscopische surveillance van Barrett-oesofagus.

May A 2002 Low

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Diagnosis

Pohl J 2007 [253] NA Patients withBarrett’s oesophagus and possible high-grade intraepithelial neoplasia or early cancer, having discrete mucosal alterations or macroscopically occult lesions (n = 57)

Conventional chromoendoscopy with acetic acid vs. computed virtual chromoendoscopy At each examination targeted biopsies from all detected lesions, followed by random four-quadrant biopsies

In 24/57 patients, 30 lesions with HGIN/early cancer were detected. The sensitivity of targeted biopsies for HGIN/early cancer on a 'per lesion' basis was 87 % (26/30) for both CAA and CVC. The positive predictive value was 39 % (26/66) for CAA and 37 % (26/70) for CVC. In the 'per patient' analysis, sensitivity was 83 % (20/24) and

Cross-over design Randomization with sealed envelopes (blinded??) Blinded assessors

RCT Moderate

Page 109: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 93

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

92 % (22/24) for CAA and CVC, respectively ( P = 0.617). Stepwise random four-quadrant biopsies identified only one patient with HGIN/early cancer that was missed by both, CAA and CVC.

Borovicka J 2006 [52] NA Patients with endoscopically documented Barrett’s oesophagus (n = 187)

Phase 1: Autofluorescence endoscopy with targeted biopsy followed by four-quadrant biopsies vs. conventional endoscopic surveillance, also including four-quadrant biopsies. Phase 2: After exclusion of patients with early cancer or high-grade dysplasia, who underwent endoscopic or surgical treatment, as well as those who declined to participate in phase 2 of the study, 130 patients remained. These patients were examined again with the alternative method after a mean of 10 weeks, using the same methods described.

In study phase 1, the AFE and conventional approaches yielded adenocarcinoma/HGD rates of 12% and 5.3%, respectively, on a per-patient basis. With AFE, four previously unrecognized adenocarcinoma/HGD lesions were identified (4.3% of the patients); with the conventional approach, one new lesion (1.1%) was identified. Of the 19 adenocarcinoma/HGD lesions detected during AFE endoscopy in study phase 1, eight were visualized, while 11 were only detected using untargeted four-quadrant biopsies (sensitivity 42%). Of the 766 biopsies classified at histology as being non-neoplastic, 58 appeared suspicious (specificity 92%, positive predictive value 12%, negative predictive value 98.5%). In study phase 2, AFE detected two further lesions in addition to the initial alternative approach in 3.2% of cases, in comparison with one lesion with conventional endoscopy (1.7%).

Cross-over design Blinded assessors Computer-generated randomization list with block randomization

RCT High

Lim CH 2006 [53] NA Patients with a diagnosis of dysplasia identified in Barrett's oesophagus (n = 35)

4 random quadrant biopsies taken every 2 cm through the length of the Barrett's esophagus (n = 18) vs. methylene blue (MB) chromoendoscopy directed biopsies from unstained or heterogenously stained mucosa (n = 17)

Overall, dysplasia was identified in 17 of 18 patients by RB and in 9 of 18 by MBDB (McNemar test, p = 0.02).

Cross-over design No ITT

RCT Moderate

Page 110: Guideline pour la prise en charge du cancer oesophagien et ...

94 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Kara MA 2005 [54] NA Patients with Barrett’s oesophagus and recently diagnosed but endoscopically inconspicuous high-grade dysplasia or early cancer (n = 28)

High-resolution endoscopy with indigo carmine chromoendoscopy or narrow-band imaging

Fourteen patients were diagnosed with HGD/EC. The sensitivity for HGD/EC was 93 % and 86 % for HRE-ICC and HRE-NBI, respectively. Targeted biopsies had a sensitivity of 79 % with HRE alone. HGD was diagnosed from random biopsies alone in only one patient. ICC and NBI detected a limited number of additional lesions occult to HRE, but these lesions did not alter the sensitivity for identifying patients with HGD/EC.

Cross-over design Randomization with sealed opaque envelopes (blinded??) Blinded assessors

RCT Moderate

Ragunath K 2003

[254] NA Patients with Barrett's oesophagus segments 3 cm or more in length without macroscopic evidence of dysplasia or cancer (n = 75)

Methylene blue-directed biopsies vs. random biopsy (4 random quadrant biopsies taken every 2 cm)

Analysis of the results by per-biopsy protocol showed that the MBDB technique diagnosed significantly more specialized intestinal metaplasia (75 %) compared to the random biopsy technique (68 %; P = 0.032). The sensitivity and specificity rates of MBDB for diagnosing specialized intestinal metaplasia were 91 % (95 % CI, 88 - 93 %) and 43 % (95 % CI, 36 - 51 %), respectively. The sensitivity and specificity rates of MBDB for diagnosing dysplasia or carcinoma were 49 % (95 % CI, 38 - 61 %) and 85 % (95 % CI, 82 - 88 %), respectively. There were no significant differences in the diagnosis of dysplasia and carcinoma - MBDB 12 %, random biopsy 10 %. The methylene blue staining pattern appeared to have an influence on the detection of specialized intestinal metaplasia and dysplasia/carcinoma. Dark blue staining was associated with increased detection of specialized intestinal metaplasia (P < 0.0001), and heterogeneous staining (P = 0.137) or no staining (P = 0.005) were associated with dysplasia and/or carcinoma detection. The MBDB

Cross-over design Blinding

RCT High

Page 111: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 95

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

technique prolonged the endoscopy examination by an average of 6 min.

Staging

Pech O 2006 [255] NA Patients with suspected early cancer in Barrett’s oesophagus referred for endoscopic treatment (n = 100)

EUS Helical CT of chest and upper abdominal organs Abdominal US examination Histology (n = 62)

For staging of T1 tumours, the sensitivity of CT was 100%, but its specificity was 0%; PPV 89%, NPV 0%. Differentiation between T1 and>T1 using EUS was possible in all 62 patients. Sensitivity of CT for N staging was not acceptable compared with EUS (38% vs. 75%). No extranodal metastases were found on CT.

Histology not available in all patients

Prospective observational study

Low

Larghi A 2005 [256] NA Patients (n = 48) with Barrett’s oesophagus and biopsy specimen proven high-grade dysplasia and adenocarcinoma in focal nodular lesions or in endoscopically unapparent flat lesions in short-segment Barrett’s oesophagus

EUS Standard: surgical pathology result (n = 8) or EMR pathology result

Accurate staging in 41/48 patients (85%) Overstaging in 1 pt, understaging in 6 pts

Single-center study Prospective observational study

Low

Waxman I 2006 [257] NA 9 consecutive patients with Barrett’s oesophagus and high-grade dysplasia (n = 7) or intra-mucosal carcinoma (n = 2)

High-frequency (20 MHz) probe ultrasonography (HFPUS) Standard: histology

Sensitivity 60% Specificity 75% Of the 3 true-positives: 1 overstaged and 1 understaged

Small sample size Prospective case series

Very low

Treatment

Overholt BF 2005

[83] NA Patients with Barrett’s oesophagus and high-grade dysplasia

Photodynamic therapy with porfimer sodium + omeprazole (n = 138) vs. omeprazole only (n = 70)

There was a significant difference (p=0.0001) in favour of PORPDT (106/138 [77%]) compared with OM (27/70 [39%]) in complete ablation of HGD at any time during the study period. The occurrence of adenocarcinoma in the PORPDT group (13%) (n=18) was significantly lower (p=0.006) compared with the OM group (20%) (n=20). The safety profile showed 94% of patients in the PORPDT group

No information on randomization procedure Only partially blinded (pathology)

RCT Moderate

Page 112: Guideline pour la prise en charge du cancer oesophagien et ...

96 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

and 13% of patients in the OM group had treatment-related adverse effects.

May A 2003 [258] NA 72 patients with early stage oesophageal cancer

Endoscopic resection with "suck-and-ligate" device without prior submucosa injection (n = 50) vs. cap technique with prior submucosa injection of a dilute saline solution of epinephrine (n = 50)

No significant differences were observed between the two groups with regard to the maximum diameters and calculated area of the resected specimens (ligation group: 16.4 [4.0] x 11 [3.1] mm/185 [84] mm(2) vs. cap group: 15.5 [4.1] x 10.7 [2.7] mm/168 [83] mm(2)), or the maximum diameters and calculated area of the endoscopic resection ulcers after 24 hours (ligation group: 20.6 [4.8] x 14.3 [4.5] mm/314 [160] mm(2) vs. cap group: 18.9 [5.1] x 12.9 [3.8] mm/260 [145] mm(2)). There was only a slight advantage (greater diameter of resection specimens) for the ligation group in patients who had prior endoscopic treatment. There was one minor episode of bleeding in each group; there was no severe complication. In 41 of 72 patients (57%), further endoscopic therapy after endoscopic resection was necessary because of residual neoplasia at the first follow-up endoscopy after resection (61 of 100 resection specimens [61%] had lateral margins that could not be evaluated because of coagulation artifact or contained malignancy but with the base of the lesion free of tumor).

Blinded randomization Blinded evaluation?

RCT Moderate

Page 113: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 97

Neoadjuvant treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Chemotherapy

CCO [84] January 2005

(In adult patients with resectable and potentially curable thoracic (lower two-thirds of oesophagus) oesophageal cancer) if surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

Numerous RCTs and SRs High

SIGN [47] 2004 Patients with operable oesophageal cancer, who are treated surgically, should be considered for two cycles of preoperative chemotherapy with cisplatin and 5-fluorouracil or offered entry into a clinical trial.

Malthaner 2003 High

CBO [56] July 2003 Patiënten met een potentieel resectabel oesofaguscarcinoom dienen alleen in onderzoeksverband voorafgaande aan een operatie chemotherapie te krijgen.

Malthaner 2003 High

Radiotherapy

CCO [84] January 2005

(In adult patients with resectable and potentially curable thoracic (lower two-thirds of oesophagus) oesophageal cancer) if surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

Arnott SJ 2000 Fok 1994

High

SIGN [47] 2004 Preoperative radiotherapy is not recommended for patients with oesophageal cancer. Arnott SJ 2000 High CBO [56] July 2003 De werkgroep raadt af om patiënten met een potentieel resectabel oesofaguscarcinoom voorafgaand aan

de operatie te bestralen. Arnott SJ 2000 High

Chemoradiotherapy

CCO [84] January 2005

(In adult patients with resectable and potentially curable thoracic (lower two-thirds of oesophagus) oesophageal cancer) if surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

Numerous RCTs and SRs High

SIGN [47] 2004 Preoperative chemoradiotherapy for patients with oesophageal cancer is not recommended outside clinical trials.

Urschel 2003 High

CBO [56] July 2003 De werkgroep adviseert om patiënten met een potentieel resectabel oesofaguscarcinoom voorafgaand aan een operatie uitsluitend in onderzoeksverband chemoradiotherapie te geven.

Walsh TN 1996 (RCT) Urba SG 2001 (RCT)

High

Page 114: Guideline pour la prise en charge du cancer oesophagien et ...

98 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type

Level of evidence

Chemotherapy

Graham AH 2007

[88] October 2004

Patients with locally advanced oesophageal cancer (T2-3N0M0 or T1-3N1M0)

Surgery vs. chemoradiotherapy followed by surgery vs. chemotherapy followed by surgery vs. surgery followed by chemoradiotherapy

For the first year, the relative risk (95% confidence interval) of death for treatments compared with surgery were 0.87 (0.75 to 1.02) for chemoradiotherapy followed by surgery, 0.94 (0.82 to 1.08) for chemotherapy followed by surgery, and 1.33 (0.93 to 1.93) for surgery with adjuvant chemoradiotherapy. The QALYs gained for surgery alone, chemoradiotherapy followed by surgery, chemotherapy followed by surgery, and surgery with adjuvant chemoradiotherapy strategies were 2.07, 2.18, 2.14, and 1.99, respectively. If the reduction in utility for multimodality treatment was increased to 21%, the QALYs gained for surgery alone, chemoradiotherapy followed by surgery, chemotherapy followed by surgery, and surgery with adjuvant chemoradiotherapy were 2.07, 2.03, 1.99, and 1.85, respectively.

14 RCTs included, of which 6 RCTs on neoadjuvant chemotherapy NO NEW EVIDENCE SINCE CCO GUIDELINE!

SR High

Gebski V 2007 [86] 2006 Patients with local operable oesophageal cancer

Neoadjuvant chemoradiotherapy or neoadjuvant chemotherapy followed by surgery vs. surgery alone

The hazard ratio for neoadjuvant chemotherapy was 0.90 (0.81-1.00; p=0.05), which indicates a 2-year absolute survival benefit of 7%. There was no significant effect on all-cause mortality of chemotherapy for patients with SCC (hazard ratio 0.88 [0.75-1.03]; p=0.12), although there was a significant benefit for those with adenocarcinoma (0.78 [0.64-0.95]; p=0.014).

Only inclusion of RCTs with ITT analysis No results of quality appraisal reported 8 RCTs on neoadjuvant chemotherapy 2 RCTs in addition to Graham et al. (Roth 1998 and Schlag 1992) NO NEW EVIDENCE SINCE CCO GUIDELINE!

SR High

Malthaner RA 2006

[87] 2006 Patients with localized potentially resectable thoracic oesophageal carcinomas

Neoadjuvant chemotherapy followed by surgery vs. surgery alone

There was some evidence to suggest that preoperative chemotherapy improves survival, but this was inconclusive (HR 0.88; 95% CI 0.75 to 1.04). There was no evidence to suggest that the

11 RCTs included NO NEW EVIDENCE SINCE CCO GUIDELINE!

SR High

Page 115: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 99

Study ID Ref Search date

Population Intervention Results Comments Study type

Level of evidence

overall rate of resections (RR 0.96, 95% CI 0.92 to 1.01) or the rate of complete resections (R0) (RR 1.05; 95% CI 0.97 to 1.15) differ between the preoperative chemotherapy arm and surgery alone. There is no evidence that tumour recurrence (RR 0.81, 95% CI 0.54 to 1.22) or non-fatal complication rates (RR 0.90; 95% CI 0.76 to 1.06) differ for preoperative chemotherapy compared to surgery alone. Trials reported risks of toxicity with chemotherapy that ranged from 11% to 90%.

Cunningham D 2006

[148] NA Patients with histologically proven adenocarcinoma of the stomach or lower third of the oesophagus that was considered to be stage II (through the submucosa) or higher, with no evidence of distant metastases, or locally advanced inoperable disease, as evaluated by computed tomography, chest radiography, ultrasonography, or laparoscopy.

Perioperative chemotherapy and surgery (n = 250) vs. surgery alone (n = 253)

As compared with the surgery group, the perioperative-chemotherapy group had a higher likelihood of overall survival (HR for death, 0.75; 95%CI 0.60 to 0.93; P=0.009; five-year survival rate, 36 percent vs. 23 percent) and of progression-free survival (HR for progression, 0.66; 95%CI 0.53 to 0.81; P<0.001). No separate date for oesophageal cancer, but no clear evidence of heterogeneity of treatment effect according to the site of the primary tumour

Randomization procedure? No information of blinding

RCT Moderate

Radiotherapy

Arnott SJ 2005 [85] June 2006

Patients with potentially resectable carcinoma of the oesophagus

Neoadjuvant radiotherapy followed by surgery vs. surgery alone

The hazard ratio (HR) of 0.89 (95%CI 0.78-1.01) suggests an overall reduction in the risk of death of 11% and an absolute survival benefit of 3% at 2 years and 4% at 5 years. This result is not conventionally statistically significant (p=0.062). No clear differences in the size of the effect by sex, age or tumor location were apparent.

Meta-analysis of 5 RCTs SR High

Chemoradiotherapy

Page 116: Guideline pour la prise en charge du cancer oesophagien et ...

100 Upper GI Cancer KCE reports 75

Graham AH 2007

[88] October 2004

Patients with locally advanced oesophageal cancer (T2-3N0M0 or T1-3N1M0)

Surgery vs. chemoradiotherapy followed by surgery vs. chemotherapy followed by surgery vs. surgery followed by chemoradiotherapy

See above 14 RCTs included, of which 6 RCTs on neoadjuvant chemoradiotherapy NO NEW EVIDENCE SINCE CCO GUIDELINE!

SR High

Gebski V 2007 [86] 2006 Patients with local operable oesophageal cancer

Neoadjuvant chemoradiotherapy or neoadjuvant chemotherapy followed by surgery vs. surgery alone

The hazard ratio for all-cause mortality with neoadjuvant chemoradiotherapy versus surgery alone was 0.81 (95% CI 0.70-0.93; p=0.002), corresponding to a 13% absolute difference in survival at 2 years, with similar results for different histological tumour types: 0.84 (0.71-0.99; p=0.04) for squamous-cell carcinoma (SCC), and 0.75 (0.59-0.95; p=0.02) for adenocarcinoma.

Only inclusion of RCTs with ITT analysis No results of quality appraisal reported 10 RCTs on neoadjuvant chemoradiotherapy 3 RCTs in addition to Graham et al. (Tepper 2006, Burmeister 2005, Lee 2004) Tepper 2006 also not included in CCO guideline, but published as an abstract

SR High

Geh JI 2006 [259] 2000 Patients with local operable oesophageal cancer

Neoadjuvant chemoradiotherapy followed by surgery vs. surgery alone

The probability of pCR improved with increasing dose of radiotherapy (P=0.006), 5FU (P=0.003) and cisplatin (P=0.018). Increasing radiotherapy treatment time (P=0.035) and increasing median age (P=0.019) reduced the probability of pCR. The estimated alpha/beta ratio of oesophageal cancer was 4.9 Gy (95% confidence interval (CI) 1.5-17 Gy) and the estimated radiotherapy dose lost per day was 0.59 Gy (95% CI 0.18-0.99 Gy). One gram per square metre of 5FU was estimated to be equivalent to 1.9 Gy (95% CI 0.8-5.2 Gy) of radiation and 100mg/m2 of cisplatin was estimated to be equivalent to 7.2 Gy (95% CI 2.1-28 Gy). Mitomycin C dose did not appear to influence pCR rates (P=0.60).

Medline and meeting abstracts Quality appraisal?? 26 RCTs + non-RCTs

SR Moderate

Hao D 2006 [109] January 2005

Patients with local operable oesophageal cancer

Platinum-based neoadjuvant chemoradiotherapy

The addition of surgery to CT/RT appeared to improve local tumour control but not overall survival in patients with locally advanced oesophageal SCC

Same search as for CCO guideline

SR High

Page 117: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 101

Greer SE 2005 [260] January 2003

Patients undergoing surgery for oesophageal cancer

Neoadjuvant chemoradiotherapy followed by surgery vs. surgery alone

Small, non-statistically significant trend toward improved long-term survival in the NCRT followed by surgery group (relative risk of death in the NCRT group 0.86; 95%CI 0.74 to 1.01; P = .07).

6 RCTs (same as Graham 2007) Quality appraisal??

SR Moderate

Natsugoe S 2006 [89] NA Patients with local operable squamous cell oesophageal cancer

Neoadjuvant chemoradiotherapy (40 Gy, cisplatin + 5FU) (n = 22) vs. surgery alone (n = 23)

Surgical treatment was performed in 20 patients in the CRT group except for two patients with bone metastasis after CRT. According to histological effects of primary tumors, the number of patient with Grades 1, 2 and 3 was 11, 7 and 3, respectively. Frequency of lymphatic and venous invasion was significantly lower in the CRT group than in the Surgery group. The 5-year survival rate was 57% in the CRT group and 41% in the Surgery group (P = 0.58). According to the histological effect in the CRT group, 5-year survival was 30% for Grade 1, 83% for Grade 2 and 100% for Grade 3 (P = 0.0069).

No information on blinding

RCT Moderate

Burmeister BH 2005

[261] NA Patients with histologically confirmed invasive cancer of the thoracic oesophagus

Neoadjuvant chemoradiotherapy (35 Gy, cisplatin + 5FU) (n = 128) vs. surgery alone (n = 128)

Neither progression-free survival nor overall survival differed between groups (HR 0.82 [95%CI 0.61-1.10] and 0.89 [0.67-1.19], respectively). The chemoradiotherapy-and-surgery group had more complete resections with clear margins than did the surgery-alone group (103 of 128 [80%] vs 76 of 128 [59%], p=0.0002), and had fewer positive lymph nodes (44 of 103 [43%] vs 69 of 103 [67%], p=0.003).

Included in CCO guideline as an abstract No blinding of patients and clinicians

RCT Moderate

Restaging after neoadjuvant treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

FNCLCC [66] La place de la TEP-FDG dans l’évaluation de la réponse au traitement reste à déterminer par des études prospectives.

Flamen 2002 Low

SIGN [47] 2004 Patients with locally advanced disease having chemotherapy/chemoradiotherapy should have their response assessed for an impact on the potential to operate ; following a good response the patient should be restaged and the role restaged and the role of surgery re-evaluated by the multidisciplinary team.

Ross P 1996 Low

Page 118: Guideline pour la prise en charge du cancer oesophagien et ...

102 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Duong CP 2006 [262] NA 53 patients with oesophageal cancer referred for PET evaluation of tumour response to CRT

FDG-PET Histology (n = 22) or serial imaging and clinical follow up (n = 26)

High PET impact (change in treatment intent or modality) was observed in 19 patients (36%). When PET findings could be verified, they were confirmed to be correct in 79% (38 of 48 cases).

Standard not the same for all patients

Prospective study

Very low

Cerfolio RJ 2005 [91] NA Patients with suspected or biopsy-proved oesophageal carcinoma treated with neoadjuvant chemotherapy (n = 48)

Chest, abdomen, pelvis CT EUS-FNA FDG-PET/CT 41 patients underwent surgery

The accuracy of each test for distinguishing pathologic T4 from T1 to T3 disease is 76%, 80%, and 80% for CT scan, EUS-FNA and FDG-PET/CT, respectively. The accuracy for nodal disease was 78%, 78%, and 93% for CT scan, EUS-FNA and FDG-PET/CT, respectively (p=0.04). FDGPET/ CT correctly identified M1b disease in 4 patients, falsely suggested it in 4 patients, and missed it in 2 patients, whereas for CT, it was 3, 3, and 3 patients. Fifteen (31%) patients were complete responders, and FDG-PET/CT accurately predicted complete response in 89% compared with 67% for EUS-FNA (p=0.045) and 71% for CT (p=0.05).

Prospective study

Low

Song SY 2005 [74] NA Patients with locally advanced but resectable oesophageal cancer treated with neoadjuvant chemoradiotherapy (n = 32)

FDG-PET Histology

Sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) in the primary tumours of the preoperative FDG-PET were 27%, 95%, 75%, and 71%, respectively. In regional lymph nodes, these values were 16%, 98%, 36%, and 93%, respectively.

Prospective study

Low

Brink I 2004 [263] NA 20 patients with oesophageal cancer treated with neoadjuvant chemoradiotherapy

FDG-PET CT Histology

Not enough information to calculate accuracy Prospective study

Very low

Nakahara T 2003

[264] NA 28 patients with squamous oesophageal cancer

Tl-201 SPECT Barium examination CT Ga-67 SPECT All studies before and after preoperative chemoradiotherapy All patients underwent oesophagectomy (blinded pathologist)

Although thallium-201 SPECT cannot be used to evaluate the therapeutic effect with acceptable accuracy, SPECT may be of additional value to barium swallow and CT in assessing the response of AESCC to preoperative chemoradiotherapy.

Prospective study

Low

Page 119: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 103

Surgical treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 Oesophageal resectional surgery should be carried out in high volume specialist surgical units by frequent operators.

van Lanschot JJ 2001 Bachmann MO 2002

Low

Surgery for oesophageal cancer should be aimed at achieving an R0 resection. Hulscher JB 2002 High Following oesopagectomy, the route of reconstruction and potential use of pyloric drainage procedure

should be determined by the surgeon based on their individual experience. Urschel JD 2001a Urschel JD 2001b Urschel JD 2002 Walther B 2003

High

Two-field lymphadenectomy should be considered during oesophagectomy to improve staging and local disease control.

Dresner SM 2000 Low

Routine extension of lymphadenectomy into the superior mediastinum or neck should not be carried out. Nishihira T 1998 High CBO [56] July 2003 Totdat vergelijkende data tussen chirurgie en andere behandelingsmodaliteiten of data van grote series

van een andere modaliteit dan chirurgie beschikbaar zijn, kan chirurgische resectie als de standaardbehandeling voor het in opzet curatief resectabele oesofaguscarcinoom worden beschouwd.

Hulscher JB 2001 High

Bij grote tumoren zonder metastasen moet worden gestreefd naar een radicale resectie van het oesofaguscarcinoom. Een palliatieve resectie is in het algemeen gecontraindiceerd.

- Tumoren van de proximale oesofagus (gelegen boven de carina) kunnen uitsluitend via een thoracotomie worden gereseceerd.

- Voor type-I-tumoren, die gelegen zijn distaal van de carina, heeft een radicale transthoracale benadering de voorkeur, mits hiermee voldoende ervaring bestaat en de algemene conditie van de patiënt dit toelaat.

- Gastro-oesofageale type-II-tumoren worden bij voorkeur transhiataal verwijderd. - Voor een proximaal maagcarcinoom type III dat minder dan 1 cm de distale oesofagus ingroeit,

kan een totale maagresectie worden verricht.

Hulscher JB 2002 van Lanschot JJ 2001 Sagar PM 1993

High

Na subtotale en macroscopisch radicale oesofagusresectie gevolgd door reconstructie met een buismaag en cervicale anastomose heeft positionering van de buismaag in het achterste mediastinum de voorkeur. In geval van macroscopische irradicaliteit heeft reconstructie via het voorste mediastinum de voorkeur.

Urschel JD 2001 van Lanschot JJ 1994

High

Voor tumoren van de cardia of van de cardio-oesofageale overgang is transhiatale resectie met medenemen van de peri-oesofageale en -cardiale klieren voldoende. Bij carcinomen van de distale oesofagus bestaat een lichte voorkeur voor een gecombineerde abdominale en transthoracale resectie met tweevelds-lymfklierdissectie van de abdominale en thoracale lymfklierstations. Oesofagustumoren boven carinaniveau worden door middel van een gecombineerde abdominale en transthoracale benadering gereseceerd.

Hulscher JB 2002 Nishihira T 1998 Matsubara T 1998 Altorki N 2002

High

Na subtotale oesofagus- (en cardia)resectie heeft de maag als interponaat de voorkeur. Het colon is tweede keuze. Slechts in uitzonderingsgevallen kan eventueel van het jejunum gebruik worden gemaakt. Het is op grond van literatuurgegevens niet mogelijk een keuze te maken tussen een totale maag of een buismaag als interponaat. Om oncologische redenen kan bij een cardia- of distale oesofagustumor slechts een buismaag worden gebruikt.

Urschel JD 2001 Collard JM 1995

High/low

Page 120: Guideline pour la prise en charge du cancer oesophagien et ...

104 Upper GI Cancer KCE reports 75

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Er is geen duidelijke voorkeur uit te spreken voor een cervicale of thoracale anastomose na oesofagusresectie en buismaagreconstructie.

- Bij de meeste patiënten is de cervicale anastomose de meest aangewezen procedure. Met name is dit het geval bij patiënten bij wie de chirurg, in geval van een tumor onder het carinaniveau, kiest voor een transhiatale resectie en halsexploratie voor het verrichten van de oesofagusresectie. Er is dan geen reden om tot thoracotomie over te gaan voor het leggen van een intrathoracale anastomose. Wanneer de chirurg bij dit soort tumoren de voorkeur geeft aan een thoracotomie voor de dissectiefase, kan de anastomose intrathoracaal worden aangelegd en kan de halsfase achterwege worden gelaten.

- Wanneer bij tumoren boven het carinaniveau wordt gekozen voor een thoracotomie, is het om oncologische redenen ter verkrijging van voldoende proximale marge verstandig ook in dat geval voor een subtotale oesofagusresectie met cervicale anastomose te kiezen.

Hulscher JB 2001 Chasseray VM 1989 Ribet M 1992 Goldminc M 1993 Chu KM 1997

High

Zowel handgelegde als mechanische (gestapelde) anastomosen kunnen worden verricht na subtotale oesofagectomie, maaginterponaat en oesofagogastrische anastomose.

Urschel JD 2001 Craig SR 1996 Valverde A 1996 Law S 1997

High

Bij een handgelegde oesofagogastrische anastomose na een subtotale oesofagusresectie met buismaagreconstructie verdient een enkelrijige doorlopende hechttechniek de voorkeur.

Bardini R 1994a Zieren HU 1993 Bardini R 1994b Muller JM 1990

High

Na oesofagusresectie gevolgd door buismaaginterponaat en oesofagogastrische anastomose is een pyloroplastiek niet geïndiceerd.

Zieren HU 1995 High

CCO [84] January 2005

If surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

Page 121: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 105

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Graham AH 2007

[88] October 2004

Patients with locally advanced oesophageal cancer (T2-3N0M0 or T1-3N1M0)

Surgery vs. chemoradiotherapy followed by surgery vs. chemotherapy followed by surgery vs. surgery followed by chemoradiotherapy

For the first year, the relative risk (95% confidence interval) of death for treatments compared with surgery were 0.87 (0.75 to 1.02) for chemoradiotherapy followed by surgery, 0.94 (0.82 to 1.08) for chemotherapy followed by surgery, and 1.33 (0.93 to 1.93) for surgery with adjuvant chemoradiotherapy. The QALYs gained for surgery alone, chemoradiotherapy followed by surgery, chemotherapy followed by surgery, and surgery with adjuvant chemoradiotherapy strategies were 2.07, 2.18, 2.14, and 1.99, respectively. If the reduction in utility for multimodality treatment was increased to 21%, the QALYs gained for surgery alone, chemoradiotherapy followed by surgery, chemotherapy followed by surgery, and surgery with adjuvant chemoradiotherapy were 2.07, 2.03, 1.99, and 1.85, respectively.

14 RCTs included The 1 RCT on adjuvant CRT vs. surgery alone included patients with gastric cancer or cancer of the GEJ

SR High

Bedenne L 2007 Bonnetain F 2006

[93] NA Patients with operable T3N0-1M0 thoracic oesophageal cancer, and with response to chemoradiation (two cycles of FU/cisplatin and either conventional (46 Gy in 4.5 weeks) or split-course (15 Gy, days 1 to 5 and 22 to 26) concomitant radiotherapy) and no contraindication to either treatment (n = 444, of which 259 were randomly assigned)

Arm A: Surgery (n = 129) Arm B: Continuation of chemoradiation (three cycles of FU/cisplatin and either conventional [20 Gy] or split-course [15 Gy] radiotherapy) (n = 130) vs.

Two-year survival rate: 34% in arm A versus 40% in arm B (HR for arm B vs. arm A = 0.90; adjusted P = .44). Median survival time: 17.7 months in arm A, 19.3 months in arm B. Two-year local control rate: 66.4% in arm A, 57.0% in arm B Stents were less required in the surgery arm (5% in arm A vs. 32% in arm B; P < .001). 3-month mortality rate: 9.3% in arm A, 0.8% in arm B (P = .002). Cumulative hospital stay: 68 days in arm A, 52 days in arm B (P = .02).

Two types of radiotherapy Not clear if blinded randomization No information on blinding of patients, clinicians and assessors

RCT Moderate

Page 122: Guideline pour la prise en charge du cancer oesophagien et ...

106 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Stahl M 2005 [265] NA Patients with locally advanced squamous cell carcinoma of the oesophagus

Induction chemotherapy followed by chemoradiotherapy (40 Gy) followed by surgery (n = 86), or the same induction chemotherapy followed by chemoradiotherapy (at least 65 Gy) without surgery (n = 86).

Overall survival was equivalent between the two treatment groups (log-rank test for equivalence, P < .05). Local progression-free survival was better in the surgery group (2-year progression-free survival, 64.3%; 95%CI 52.1% to 76.5%) than in the chemoradiotherapy group (2-year progression-free survival, 40.7%; 95%CI 28.9% to 52.5%; hazard ratio [HR] for arm B v arm A, 2.1; 95% CI, 1.3 to 3.5; P = .003). Treatment-related mortality was significantly increased in the surgery group than in the chemoradiotherapy group (12.8% v 3.5%, respectively; P = .03).

Unblinded study RCT Moderate

Chiu PWY 2005 (CURE)

[94] NA Patients with potentially resectable squamous cell carcinoma of the mid or lower thoracic oesophagus

Oesophagectomy (n = 44) vs. chemoradiotherapy (continuous 5-FU (200 mg/m2/day) from day 1 to 42 and cisplatin (60 mg/m2) on days 1 and 22; tumour and regional lymphatics were concomitantly irradiated to a total of 50-60 Gy) (n = 36)

No difference in the early cumulative survival (RR = 0.89; 95%CI 0.37-2.17; log-rank test P = 0.45). No difference in the disease-free survival.

No information on randomization procedure No information on blinding of patients, clinicians and assessors

RCT Moderate

Bhat MA 2006 [266] NA 238 patients treated for oesophageal carcinoma (44 excluded because inoperable)

Oesophagogastrectomy with wrapping of the pedicled omentum around the esophagogastric anastomosis (n = 97) vs. oesophagogastrectomy without using the omental graft (n = 97)

Anastomotic leaks occurred in 3 group A patients (3.09%) and in 14 (14.43%) group B patients. The difference in the incidence of leakage was statistically significant (p = 0.005). There was no complication related to the omental graft technique nor was there a significant difference in the mortality between the two groups.

No information on blinding of assessors

RCT Moderate

Hsu HH 2004 [267] NA Patients with curatively resectable squamous cell cancer of the thoracic oesophagus (T1-3 N0-1)

Hand-sewn (n = 32) or circular stapled (n = 31) cervical oesophagogastric anastomosis

The mean operating time was longer when the hand-sewn method was used (524 vs. 447 min, P < 0.001). Anastomotic leakage was noted in seven patients (22%) in the hand-sewn group and eight patients (26%) in the stapler group (P = NS). Hospital mortality

Randomization procedure?? Blinding??

RCT Moderate

Page 123: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 107

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

occurred in four patients (13%) of the hand-sewn group and in three patients (10%) of the stapler group (P = NS). After the operation, four patients (14%) in the hand-sewn group and five patients (18%) in the stapler group developed a benign oesophageal stricture (P = NS). The mean follow-up time was 24 months, and the rates of freedom from benign stricture and survival were comparable in each group.

Walther B 2003 [268] NA Patients with benign or malignant oesophageal disease for which a tube gastroplasty with anastomotic site in the proximal chest or in the neck was deemed suitable (n = 83, of which 74 had malignant disease)

Manually sutured oesophagogastric anastomosis in the neck or an anastomosis stapled in the right chest

The genuine 5-year survival rate was 29% for chest anastomoses and 30% for neck anastomoses. The overall leakage rate was 1.8% (2 cases of 112) with no relation to mortality or anastomotic method. All patients in the randomized group had tumor-free proximal and distal resection lines, but 1 patient in the nonrandomized group had tumor infiltrates in the proximal resection margin. At 3, 6, and 12 months after operation, there was no difference in anastomotic diameter between the esophagogastric anastomosis in the neck and in the thorax (P = 0.771), and both increased with time (P = 0.004, ANOVA repeated measures). Body weight development was the same in the two groups. With similar results in randomized and nonrandomized patients, study bias was eliminated.

Randomization with sealed envelopes Also 29 non-randomized patients included in results

RCT Moderate

Page 124: Guideline pour la prise en charge du cancer oesophagien et ...

108 Upper GI Cancer KCE reports 75

Adjuvant treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Chemotherapy

SIGN [47] 2004 Postoperative adjuvant chemotherapy is not recommended for patients with oesophageal cancer. Lehnert T 1999 High CBO [56] July

2003 Patiënten met een oesofaguscarcinoom komen niet in aanmerking voor postoperatieve chemotherapie. Ando N 1997

Pouliquen X 1996 High

CCO [84] January 2005

If surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

Ando N 1997 Pouliquen X 1996 Ando N 2003

High

Radiotherapy

SIGN [47] 2004 No recommendation, only discussion CBO [56] July

2003 Patiënten met een oesofaguscarcinoom komen niet in aanmerking voor postoperatieve radiotherapie. Fok M 1993

Teniere P 1991 Zieren HU 1995 Xiao ZF 2003

High

CCO [84] January 2005

If surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

Fok M 1993 Fok M 1994 Teniere P 1991 Zieren HU 1995 Xiao ZF 2003

High

Chemoradiotherapy

SIGN [47] 2004 Postoperative adjuvant chemoradiotherapy is not recommended for patients with oesophageal cancer. No evidence CCO [84] January

2005 If surgery is considered appropriate, then surgery alone (i.e., without neoadjuvant or adjuvant therapy) is recommended as the standard practice for resectable thoracic oesophageal cancer.

No evidence

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Chemotherapy

Ajani J 2006 [269] October 2005

Patients oesophageal cancer

Oral capecitabine 4 trials identified that included patients with oesophageal cancer No conclusions to be drawn

Medline search only No information on selection, quality appraisal, etc

SR Low

Radiotherapy

No additional studies found

Chemoradiotherapy

No additional studies found

Page 125: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 109

Non-surgical treatment with curative intent.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 Chemoradiotherapy should be considered in patients with esophageal cancer who have locally advanced disease, those unfit for surgery or those who decline surgery.

Wong R '03 Cooper JS '99

High

CCO [108] February 2005

(In adult patients with localized (T1-3, small volume N1, M0) carcinoma of the esophagus and good performance status who are considering a non-surgical approach and for whom combined radiotherapy and chemotherapy can be tolerated in the judgment of the treating oncologist) concomitant radiotherapy and chemotherapy is recommended over radiotherapy alone. Based on considerations of the current clinical practice pattern and the currently available research evidence, a cisplatin-based chemotherapy regimen is a reasonable chemotherapy regimen to use when concomitant radiotherapy and chemotherapy is used.

Wong R ‘06 High

Patients should be made aware of the increased acute toxicity associated with this approach. The decision to use concomitant radiotherapy and chemotherapy should only be made after careful consideration of the potential risks, benefits, and the patient’s general condition.

Wong R ‘06 High

Sequential radiotherapy and chemotherapy is not recommended as standard practice. Wong R ‘06 High Future clinical trials to better define the optimal chemoradiotherapy combination that would improve

outcomes while limiting toxicities are strongly encouraged. Wong R ‘06 High

CBO [56] July 2003 De werkgroep adviseert om patiënten met een oesofaguscarcinoom die niet voor een resectie in aanmerking komen, indien hun conditie dit toelaat, te behandelen met concomitante chemoradiotherapie, zo mogelijk in onderzoeksverband.

Wong R '03 Cooper JS '99

High

Page 126: Guideline pour la prise en charge du cancer oesophagien et ...

110 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Bedenne L 2007 [93] NA Patients with operable T3N0-1M0 thoracic oesophageal cancer, and with response to chemoradiation (two cycles of FU/cisplatin and either conventional (46 Gy in 4.5 weeks) or split-course (15 Gy, days 1 to 5 and 22 to 26) concomitant radiotherapy) and no contraindication to either treatment (n = 444, of which 259 were randomly assigned)

Arm B: Continuation of chemoradiation (three cycles of FU/cisplatin and either conventional [20 Gy] or split-course [15 Gy] radiotherapy) (n = 130) vs. Arm A: surgery (n = 129)

Two-year survival rate: 34% in arm A versus 40% in arm B (HR for arm B vs. arm A = 0.90; adjusted P = .44). Median survival time: 17.7 months in arm A, 19.3 months in arm B. Two-year local control rate: 66.4% in arm A, 57.0% in arm B Stents were less required in the surgery arm (5% in arm A vs. 32% in arm B; P < .001). 3-month mortality rate: 9.3% in arm A, 0.8% in arm B (P = .002). Cumulative hospital stay: 68 days in arm A, 52 days in arm B (P = .02).

Two types of radiotherapy Not clear of blinded randomization No information on blinding of patients, clinicians and assessors

RCT Moderate

Hao D 2006 [109] January 2005

Patients with local operable oesophageal cancer

Platinum-based chemoradiotherapy

To date, no randomized trials have directly compared CT/RT with oesophagectomy as primary locoregional treatment of oesophageal carcinoma.

Same search as for CCO guideline

SR High

Chiu PW 2005 (CURE)

[94] NA Patients with potentially resectable squamous cell carcinoma of the mid or lower thoracic oesophagus

Oesophagectomy (n = 44) vs. chemoradiotherapy (continuous 5-FU (200 mg/m2/day) from day 1 to 42 and cisplatin (60 mg/m2) on days 1 and 22; tumour and regional lymphatics were concomitantly irradiated to a total of 50-60 Gy) with salvage surgery (n = 36)

No difference in the early cumulative survival (RR = 0.89; 95%CI 0.37-2.17; log-rank test P = 0.45). No difference in the disease-free survival.

No information on randomization procedure No information on blinding of patients, clinicians and assessors

RCT Moderate

Zhao KL 2005 [110] NA Patients with confirmed SCC of the oesophagus with clinical stage T1-4, N0-1, M0 (n = 111)

LCAF-RT (n = 57) vs. LCAF-RT + chemotherapy (four cycles of cisplatin 25 mg/m(2) daily and fluorouracil (5-FU) 600 mg/m(2) daily on Days 1-3 every 4 weeks starting on the same day that LCAF was delivered) (n = 54)

Median survival: 23.9 months (95%CI 20.1-27.7) for the LCAF arm and 30.8 months (95%CI 17.6-44.1) for the LCAF+CT arm. Survival rates at 1, 3, and 5 years of the LCAF arm were 77%, 39%, and 28%, respectively, while those of the LCAF+CT arm were 67%, 44%, and

No information on blinding of patients, clinicians and assessors

RCT Moderate

Page 127: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 111

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

40%, respectively (p = 0.310). Grades 3 and 4 acute toxicities occurred in 46% and 25% of the patients in the LCAF arm and the LCAF+CT arm, respectively; 6% of the patients in the combined arm had Grade 5 acute toxicities, whereas none was noted in the LCAF alone arm.

Sumpter K 2005 [270] NA Patients with inoperable, histologically verified locally advanced or metastatic adenocarcinoma, squamous cell or undifferentiated carcinoma of the oespohagus, oesophagogastric junction (OGJ) or stomach

ECF (n = 53) ECX (n = 48) EOF (n = 55) EOX (n = 48) (E = epirubucin, C = cisplatin, F = 5FU, O = oxaliplatin, X = capecitabin)

First interim analysis was performed when 80 pts had been randomised. Doselimiting fluoropyrimidine toxicities were stomatitis, palmar plantar erythema (PPE) and diarrhoea; 5.1% of X-treated pts experienced grade 3/4 toxicity. Protocol planned dose escalation of X to 625 mg/m² b.i.d. was instituted and a second interim analysis has been performed. A total of 204 pts were randomised at the time of the protocol planned 2nd interim analysis. Grade 3/4 fluoropyrimidine-related toxicity was seen in 13.7% pts receiving F, 8.4% pts receiving X 500 mg/m² b.i.d. and 14.7% pts receiving X 625 mg/m² b.i.d. Combined complete and partial response rates were ECF 31% (95% CI 18.7–46.3), EOF 39% (95% CI 25.9–53.1), ECX 35% (95% CI 21.4–50.3), EOX 48% (95% CI 33.3–62.8). Grade 3/4 fluoropyrimidine toxicity affected 14.7% of pts treated with X 625 mg/m² b.i.d., which is similar to that observed with F, confirming this to be the optimal dose. The replacement of C by O and F by X does not appear to impair efficacy. The trial continues to total accrual of 1000 pts.

Interim analysis 6 originally randomized pts not included in results Noninferiority study No information on blinding of randomization No information on blinding of patients, clinicians and assessors

RCT Moderate

Page 128: Guideline pour la prise en charge du cancer oesophagien et ...

112 Upper GI Cancer KCE reports 75

Palliative treatment.

CPG ID

Ref Search date

Recommendation Supporting evidence Level of evidence

General

CBO [56] July 2003

Afhankelijk van de patiëntkarakteristieken en de tumoruitbreiding zal bij patiënten met een T4-oesofaguscarcinoom bij voorkeur worden gekozen voor een korte palliatieve behandeling of een behandeling gericht op palliatie op de langere termijn, zoals (chemo)radiotherapie. Patiënten met een fistel komen, afhankelijk van de plaats en de grootte van de fistel, in aanmerking voor stentplaatsing.

Low

Endoscopic ablation

SIGN [47] 2004 Laser or photodynamic therapy should be used for initial control of obstructive symptoms caused by exophytic tumours in the oesophagus including tumours near the upper oesophageal sphincter.

Allum WH 2002 O’Donnell CA 2002 Dallal HJ 2001 Lightdale CJ 1995 Carazzone A 1999 Heier SK 1995

High

Laser or photodynamic therapy should be considered for control of tumour overgrowth in stented patients.

Moghissi K 2000 Litle VR 2003

Low

Stenting

CBO [56] July 2003

Indien de levensverwachting korter is dan zes weken, kan allereerst stentplaatsing worden overwogen, gezien het snelle effect op de verbetering van passageklachten.

Homs MY 2004

High

Bij patiënten met een fistel tussen de oesofagus en de luchtwegen, patiënten met een inoperabel oesofaguscarcinoom nabij de bovenste oesofagussfincter en patiënten met een recidief na eerdere oesofagusresectie in verband met een oesofaguscarcinoom heeft stentplaatsing de voorkeur.

May A 1998 Dumonceau JM 1999 Siersema PD 2001

Moderate

Het verdient aanbeveling om, na stentplaatsing in verband met passageklachten ten gevolge van een inoperabel oesofaguscarcinoom, rustig te eten, goed te kauwen en veel te drinken om verstopping van de stent te voorkomen. Bij verstopping van de stent kan deze snel worden gereinigd door middel van een endoscopie.

SIGN [47] 2004 Partially covered self-expanding metal stents are the intubation of choice for patients with obstructive oesophageal symptoms.

Homs MY 2004a Sabharwal T 2003 Vakil N 2001

Moderate

Partially covered self-expanding metal stents should be used to control obstructive oesophageal symptoms either following or instead of laser therapy, depending on the availability of local expertise.

Bethge N 1996 Homs MY 2004b

Low

Dilatation

SIGN [47] 2004 The use of oesophageal dilatation alone should be avoided. Allum WH 2002 Very low

Surgery

SIGN [47] 2004 Oesophagectomy (transthoracic or transhiatal) should not be performed with palliative intent in patients with oesophageal cancer.

Blazeby JM 2000 Low

Substernal bypass for oesophageal cancer should not be performed with palliative intent. Whooley BP 2002 Alcantara PS 1997

Low

Page 129: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 113

CPG ID

Ref Search date

Recommendation Supporting evidence Level of evidence

Chemotherapy

CBO [56] July 2003

Chemotherapie is geen standaardbehandeling voor het irresectabele of gemetastaseerde oesofaguscarcinoom.

Schmid EU 1993 Levard H 1998 Mannell A 1986 Webb A 1997 Ross P 2002 Bleiberg H 1997

Moderate

SIGN [47] 2004 In patients with locally advanced or metastatic cancer of the oesophagus with good performance status combination chemotherapy including cisplatin and infusional 5FU (such as ECF or MCF) should be considered.

Janunger KG 2002 High

Radiotherapy

CBO [56] July 2003

Bij oesofaguscarcinoompatiënten met een matige algemene conditie en/of met metastasen op afstand bestaat een voorkeur voor brachytherapie, eventueel gevolgd door een stentplaatsing indien brachytherapie onvoldoende effect heeft.

Homs MY 2004 High

Bij inoperabele patiënten met een levensverwachting langer dan drie maanden kan uitwendige radiotherapie in combinatie met brachytherapie worden overwogen.

Flores AD 1989 Hishikawa Y 1991 Taal BG 1996

Low

SIGN [47] 2004 Palliative external-beam radiotherapy is an appropriate option for the treatment of mild dysphagia in patients with oesophageal cancer.

Allum WH 2002 Very low

Endoluminal brachytherapy is an option for patients with dysphagia from oesophageal cancer. Brewster AE 1995 Homs MYV 2003

Low

Supportive care

CBO [56] July 2003

De werkgroep is van mening dat de verpleegkundige zorg voor patiënten met een oesofaguscarcinoom het beste door een verpleegkundige met specifieke kennis en ervaring kan worden verricht.

Dieetmaatregelen bij patiënten met een inoperabel oesofaguscarcinoom kunnen het beste zijn gericht op het welbevinden van de patiënt, dat wil zeggen dat de kwaliteit van leven belangrijker is dan de effecten van voeding op lange termijn.

Doornik N 2000 Very low

SIGN [47] 2004 Patients with oesophageal cancer should have access to a specialist palliative care team. Smeenk FW 1998 Miccinesi G 2003 Costantini M 2003 Costantini M 1993

Low

Page 130: Guideline pour la prise en charge du cancer oesophagien et ...

114 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Endoscopic ablation

No additional studies found

Stenting

Wenger U 2006 [271] NA Patients with incurable cancer of the distal oesophagus or gastric cardia (n = 41)

Antireflux stent (n = 19) vs. standard stent (n = 22)

Fewer patients with complications were observed in the antireflux stent group (n = 3) than in the standard group (n = 8), but no statistically significant difference was shown (p = 0.14). The survival rates were similar in the two groups (p = 0.99; hazard ratio, 1.0; 95% confidence interval, 0.5-2.0). The groups did not differ significantly in terms of studied oesophageal or respiratory symptoms or quality of life. Clinically relevant improvement in dysphagia occurred in both groups. Dyspnea decreased after antireflux stent insertion (mean score change, -11), and increased after insertion of standard stent (mean score change, +21).

Interim report Central randomization, but no information on procedure Patients were blinded, but no information on blinding of clinicians

RCT Moderate

Wenger U 2005 Bergquist H 2005

[112] NA Patients with incurable cancer of the oesophagus or gastro-oesophageal junction

Self-expandable metallic stent (n = 30) or 3 x 7 Gy brachytherapy (n = 30)

The group of patients treated with stent reported significantly better HRQL scores for dysphagia (P < 0.05) at the 1-month follow-up, but most other HRQL scores, including functioning and symptom scales, deteriorated. There was no difference in survival or complication rates between the two treatment strategies. There was a significant difference in the change of dysphagia scores between the time of inclusion and the 1-month follow-up visit, in favour of the stented group (P = 0.03). This difference had disappeared at 3 months. Median total lifetime costs were 17,690 for the stented group compared with

Originally 65 randomized, but 5 withdrew consent No information on blinding

RCT Cost study

Moderate

Page 131: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 115

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

33,171 for the brachytherapy group (P = 0.005). This difference was due to higher costs for the initial treatment (4,615 versus 23,857, P < 0.0001). Sensitivity analyses showed that the charges for a brachytherapy session had to be reduced from 6,092 to 4,222 (31%) to make this therapeutic concept cost-competitive.

Shim CS 2005 [272] NA Patients with cancer at the oesophagogastric junction (n = 36)

Newly designed antireflux stent (n = 12), a Dostent (n = 12), or a standard open stent (n = 12)

After 1 week, dysphagia had improved in all patient groups (P < 0.05), but the degree of improvement did not differ between the three groups. The DeMeester score was significantly lower in the group with the newly designed antireflux stent than in the other groups. The fraction of the total recording time during which oesophageal pH was below 4 was 3.14 +/- 5.78 % using the newly designed antireflux stent, in comparison with 29.25 +/- 15.41 % in the Dostent group and 15.01 +/- 11.72 % in the standard open stent group (P < 0.001). Fewer reflux episodes occurred with the newly designed antireflux stent than with the Dostent or standard open stent. There were no complications with any of the three stents.

No information on randomization procedure or blinding

RCT Moderate

Shenfine J 2005 [273] NA Patients with oesophageal cancer deemed unsuitable for surgery (n = 217)

SEMS 18 mm (n = 54) SEMS 24 mm (n = 54) Rigid intubation (n = 57) All other non-SEMS treatments (n = 52)

There was no difference in cost or effectiveness between SEMS and non-SEMS therapies, and 18-mm SEMS had equal effectiveness to, but less associated pain than, 24-mm SEMS. Rigid intubation was associated with a worse quality of swallowing and increased late morbidity. Bipolar electrocoagulation and ethanol tumour necrosis were poor in primary palliation. A survival advantage was found for non-stent therapies, but there was a significant

Secondary randomization possible after treatment failure following non-SEMS treatment Correct randomization protocol

RCT Cost study

High

Page 132: Guideline pour la prise en charge du cancer oesophagien et ...

116 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

delay to treatment. The length of stay accounts for the majority of the cost to the NHS. Patients were found to have distinct individual treatment preferences.

Polinder S 2004 (SIREC trial)

[113] NA Patients with inoperable oesophagogastric cancer

Ultraflex stent (n=108) or single-dose brachytherapy (12 Gy, n=101)

The initial costs of stent placement were higher than the costs of brachytherapy (1500 euro vs 570 euro; P<0.001). Total medical costs were, however, similar (stent 11 195 euro vs brachytherapy 10 078 euro, P>0.20). Total hospital stay during follow-up was 11.5 days after stent placement vs 12.4 days after brachytherapy, which was responsible for the high intramural costs in both treatment groups (stent 6512 euro vs brachytherapy 7982 euro, P>0.20). Costs for medical procedures during follow-up were higher after stent placement (stent 249 euro vs brachytherapy 168 euro, P=0.002), while the costs of extramural care were similar (1278 euro vs 1046 euro, P>0.20).

See also Homs MY 2004, Lancet No information on blinding

RCT Cost study

Moderate

Dilatation

No additional studies found

Surgery

No additional studies found

Chemotherapy

Homs MY 2006 [114] February 2006

Patients with metastatic carcinoma of the oesophagus or GE-junction Siewert type I

RCTs comparing chemotherapy with best supportive care for patients with metastatic carcinoma of the oesophagus and GE-junction, as well as RCTs comparing different chemotherapeutic therapies. Treatments with single chemotherapeutic agents as

Only two RCTs with a total of 42 participants compared chemotherapy with best supportive care for metastatic oesophageal cancer. No survival benefit was shown for chemotherapy treatment in these RCTs. Five RCTs with a total of 1242 participants compared different chemotherapy regimes. Due to variation in patient population and chemotherapy regimes, it was not possible to perform

7 RCTs included SR High

Page 133: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 117

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

well as combination regimes in all doses and schedules were included. Studies in which patients received additional treatment for dysphagia such as stent placement or brachytherapy were not excluded.

a formal pooled analysis. There was no consistent benefit of any specific chemotherapy regimen.

Ajani J 2006 [269] October 2005

Patients with oesophageal cancer

Oral capecitabine 4 trials identified that included patients with oesophageal cancer

Medline search only No information on selection, quality appraisal, etc

SR Low

Trumper M 2006

[274] NA Patients with gastro-oesophageal cancer, older than 70 years (n = 257)

Platinum-containing regimen (ECF, MCF), PVI 5-FU (protracted venous infusion of 5-fluorouracil) ± mitomycin C (MMC), or FAMTX

There were no significant differences in the incidence of grades 3/4 toxicity between the two cohorts. Objective and symptomatic response rates, failure-free and overall survival were not significantly different. In a multivariate analysis, independent prognostic factors for survival were performance status and locally advanced disease, not age. Patients >70 years with OGC obtained similar benefits from palliative chemotherapy with respect to symptomatic response, tumour regression and survival, without increased toxicities.

Pooled analysis of 3 RCTs (retrospective subanalysis): which trials??

MA Low

Duffour J 2006 [275] NA 232 patients with histologically proven carcinoma of the oesophagus (n = 38), stomach or pancreas, and metastatic disease

Cisplatin with continuous 5FU (n = 19) vs. bolus 5FU and Leucovorin (n = 19)

All tumours: Safety remained acceptable and comparable in the two arms except for the severe grade 3-4 mucositis, which was lower in arm B (4.5 vs. 16.4%, p < 0.009). Efficacy in terms of tumour response and survival was similar in the two arms, showing an objective response rate (after external review) of 18.6% (95% confidence interval (CI) 11.4-25.8%) in arm A vs. 15% (95% CI 8.5-21.6%) in arm B, an overall median survival of 24 weeks in arm A vs. 24.7 in

Mixed patient population Blinded assessors?

RCT Moderate

Page 134: Guideline pour la prise en charge du cancer oesophagien et ...

118 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

arm B (p = 0.83) and a progression-free median survival of 12.4 weeks vs. 12.1 in arms A and B, respectively (p = 0.91).

Sumpter K 2005 [270] NA Patients with inoperable, histologically verified locally advanced or metastatic adenocarcinoma, squamous cell or undifferentiated carcinoma of the oespohagus, oesophagogastric junction (OGJ) or stomach

ECF (n = 53) ECX (n = 48) EOF (n = 55) EOX (n = 48) (E = epirubucin, C = cisplatin, F = 5FU, O = oxaliplatin, X = capecitabin)

First interim analysis was performed when 80 pts had been randomised. Doselimiting fluoropyrimidine toxicities were stomatitis, palmar plantar erythema (PPE) and diarrhoea; 5.1% of X-treated pts experienced grade 3/4 toxicity. Protocol planned dose escalation of X to 625 mg/m² b.i.d. was instituted and a second interim analysis has been performed. A total of 204 pts were randomised at the time of the protocol planned 2nd interim analysis. Grade 3/4 fluoropyrimidine-related toxicity was seen in 13.7% pts receiving F, 8.4% pts receiving X 500 mg/m² b.i.d. and 14.7% pts receiving X 625 mg/m² b.i.d. Combined complete and partial response rates were ECF 31% (95% CI 18.7–46.3), EOF 39% (95% CI 25.9–53.1), ECX 35% (95% CI 21.4–50.3), EOX 48% (95% CI 33.3–62.8). Grade 3/4 fluoropyrimidine toxicity affected 14.7% of pts treated with X 625 mgm�2 b.i.d.�1, which is similar to that observed with F, confirming this to be the optimal dose. The replacement of C by O and F by X does not appear to impair efficacy. The trial continues to total accrual of 1000 pts.

Interim analysis 6 originally randomized pts not included in results Noninferiority study No information on blinding of randomization

RCT Moderate

Radiotherapy

Wenger U 2005 Bergquist H 2005

[112] NA Patients with incurable cancer of the oesophagus or gastro-oesophageal junction

Self-expandable metallic stent (n = 30) or 3 x 7 Gy brachytherapy (n = 30)

The group of patients treated with stent reported significantly better HRQL scores for dysphagia (P < 0.05) at the 1-month follow-up, but most other HRQL scores, including functioning and symptom scales, deteriorated.

Originally 65 randomized, but 5 withdrew consent No information on blinding

RCT Cost study

Moderate

Page 135: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 119

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

There was no difference in survival or complication rates between the two treatment strategies. There was a significant difference in the change of dysphagia scores between the time of inclusion and the 1-month follow-up visit, in favour of the stented group (P = 0.03). This difference had disappeared at 3 months. Median total lifetime costs were 17,690 for the stented group compared with 33 171 for the brachytherapy group (P = 0.005). This difference was due to higher costs for the initial treatment (4615 versus 23 857, P < 0.0001). Sensitivity analyses showed that the charges for a brachytherapy session had to be reduced from 6092 to 4222 (31%) to make this therapeutic concept cost-competitive.

Polinder S 2004 (SIREC trial)

[113] NA Patients with inoperable oesophagogastric cancer

Ultraflex stent (n=108) or single-dose brachytherapy (12 Gy, n=101)

The initial costs of stent placement were higher than the costs of brachytherapy (1500 euro vs 570 euro; P<0.001). Total medical costs were, however, similar (stent 11 195 euro vs brachytherapy 10 078 euro, P>0.20). Total hospital stay during follow-up was 11.5 days after stent placement vs 12.4 days after brachytherapy, which was responsible for the high intramural costs in both treatment groups (stent 6512 euro vs brachytherapy 7982 euro, P>0.20). Costs for medical procedures during follow-up were higher after stent placement (stent 249 euro vs brachytherapy 168 euro, P=0.002), while the costs of extramural care were similar (1278 euro vs 1046 euro, P>0.20).

See also Homs MY 2004, Lancet No information on blinding

RCT Cost study

Moderate

Page 136: Guideline pour la prise en charge du cancer oesophagien et ...

120 Upper GI Cancer KCE reports 75

Follow up.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

CBO [56] July 2003 In het eerste jaar kan bij patiënten na behandeling van een oesofaguscarcinoom worden gekozen voor frequente follow-upintervals (drie en eventueel zes weken en daarna driemaandelijks), waarna in de daarop volgende jaren deze frequentie teruggaat naar elke zes maanden in het tweede jaar en daarna jaarlijks. Na vijf jaar is er geen indicatie meer voor routinematige follow-up. Er is slechts plaats voor aanvullend onderzoek op geleide van klachten.

Virgo KS 1999 Goodnight J 1996 Johnson FE 1997 Marsh JC 1997

Very low

SIGN [47] 2004 Follow up of patients with oesophageal cancer should monitor symptoms, signs and nutritional status. Allum WH 2002 Very low

Study ID Ref Search date Population Intervention Results Comments Study type Level of

evidence No additional studies found

Quality of life

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 The possibility of reduction in quality of life after surgery should be considered when discussing treatment options, particularly when preoperative staging suggests that surgery would be unlikely to be curative.

Blazeby 2000 Low

Study ID Ref Search

date Population Intervention Results Comments Study type Level of

evidence Early lesions

Moraca R 2006 [276] 36 patients with high-grade dysplasia (HGD) or intramucosal cancer (IMC)

Esophagectomy 28 survivors, significant differences in postesophagectomy gastrointestinal symptoms (decrease on heartburn and slower speed of eating)

Not specific scale : Medical Outcomes Study 36-Item Short-Form Health Survey

Prospective cohort study

Low

Surgery

Cense HA 2006 [277] 109 patients undergoing a transthoracic resection

Esophagectomy For survivors, no difference in long-term (at 2 years) quality of life or survival between those submitted to the intensive care unit for a short period vs. a long period.

Scale : Medical Outcome Studies Short Form-20 and the Rotterdam Symptom Check List

Prospective study

Low

Lagergren 2007

[278] 90 patients who underwent surgery for oesophageal cancer. 52% survivors >/=3 years.

Esophagectomy Recovery except that scores for physical function, breathlessness, diarrhea, and reflux significantly worse than at baseline (P < .01)

It’s necessary to inform patients of the long-term consequences of surgery

Prospective study

Low

Page 137: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 121

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Neoadjuvant treatment

Blazeby 2005 [279] 34 patients undergoing chemoradiotherapy + surgery 28 patiens undergoing chemotherapy + surgery 34 patients undergoing surgery alone

Quality of life assessment with QLQ-C30 and QLQ-OES24 (dysphagia scale from the EORTC)

After neoadjuvant treatment, but before surgery, HRQL returned to baseline levels. Recovery of HRQL was not hampered by preoperative treatment, and fewer problems with postoperative nausea, emesis, and dysphagia were reported by patients who had undergone neoadjuvant treatment compared with patients who had undergone surgery alone.

Recovery of HRQL after esophagectomy was not impaired by neoadjuvant treatment

Prospective study

Low

Palliative treatment

Homs 2004 [280] Patients with dysphagia from inoperable oesophageal carcinoma

Ultraflex stent (n = 108) or single dose (12 Gy) brachytherapy (n = 101).

Significant difference in favour of brachytherapy on four out of five functional scales of the EORTC QLQ-C30 (role, emotional, cognitive and social) (P < 0.05). Major improvements were seen on the dysphagia and eating scales of the EORTC OES-23.

The effects of single dose brachytherapy on HRQoL compared favourably to those of stent placement for the palliation of oesophageal cancer. Inlcuded in SIGN guideline.

RCT High

Psychological support

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Kuchler T [115] Patients (N = 271) with a preliminary diagnosis of cancer of the oesophagus (n = 31), stomach, liver/gallbladder, pancreas, or colon/rectum

Standard care as provided on the surgical wards vs. formal psychotherapeutic support in addition to routine care during the hospital stay

Kaplan-Meier survival curves demonstrated better survival for the experimental group than the control group. The unadjusted significance level for group differences was P = .0006 for survival to 10 years. Cox regression models that took TNM staging or the residual tumor classification and tumor site into account also found significant differences at 10 years. Secondary analyses found that differences in favor of the experimental group occurred in patients with stomach, pancreatic, primary liver, or colorectal cancer.

Not specifically for oesophageal cancer.

RCT Moderate

Page 138: Guideline pour la prise en charge du cancer oesophagien et ...

122 Upper GI Cancer KCE reports 75

Nutritional support CPG ID Ref Searc

h date

Recommendation Supporting evidence Level of evidence

CBO [56] July 2003

Het verdient aanbeveling gedurende de behandeling met gecombineerde chemoradiotherapie bij een patiënt met oesofaguscarcinoom de voedselinname te evalueren en zo nodig te starten met dieetmaatregelen.

Jeremic B 1998 Safran H 2001 Sikora SS 1998

Low

Het verdient aanbeveling om reeds in een vroeg stadium bij patiënten met een oesofaguscarcinoom het lichaamsgewicht te controleren, de voedselinname te evalueren en zo nodig te starten met voedingsinterventie.

Het verdient aanbeveling bij electieve gastro-intestinale chirurgie terughoudend te zijn met het stimuleren van orale drinkvoeding gezien het gebrek aan aangetoonde effectiviteit.

Op grond van de beschikbare literatuur is er mogelijk plaats voor het gebruik van immunonutritie in de perioperatieve fase bij stabiele patiënten met een oesofaguscarcinoom. Bij ernstig zieke patiënten is terughoudendheid geboden met argininerijke immunonutritie.

In de postoperatieve fase kan men overwegen om gedurende metabole stress te streven naar een inname van 1,5-1,7 gram eiwitten per kilogram actueel lichaamsgewicht per dag en 25-30 kcal voor mannen en 20-25 kcal voor vrouwen per kilogram actueel lichaamsgewicht per dag. Hyperalimentatie dient te worden voorkomen, omdat dit samen kan gaan met metabole ontregelingen.

Het verdient aanbeveling in de postoperatieve fase langdurig het lichaamsgewicht, de voedselinname en de aanwezigheid van aan voeding gerelateerde klachten te evalueren. Bij gewichtsverlies, onvoldoende inname en bij klachten, kan door middel van dieetadviezen zo nodig met orale drinkvoeding dan wel (nachtelijke) enterale voeding worden geprobeerd gewichtsverlies te voorkomen en de voedingstoestand te verbeteren.

Het verdient aanbeveling na een oesofagusresectie gebruik te maken van enterale voeding (sondevoeding) wanneer orale voeding onvoldoende mogelijk is.

Het verdient aanbeveling om in de perioperatieve fase bij patiënten met een oesofaguscarcinoom slechts gebruik te maken van parenterale voeding indien enterale voeding niet mogelijk is.

Dieetmaatregelen bij patiënten met een inoperabel oesofaguscarcinoom kunnen het beste zijn gericht op het welbevinden van de patiënt, dat wil zeggen dat de kwaliteit van leven belangrijker is dan de effecten van voeding op lange termijn.

SIGN [47] 2004 Patients undergoing surgery for oesophageal cancer who are identified as being at high nutritional risk should be considered for preoperative nutritional support.

Gianotti L 2002 Bozzetti F 2000 Braga M 1999 & 2002

High

All patients undergoing surgery for oesophageal cancer should be considered for early postoperative nutritional support preferably by the enteral route.

Several RCTs High

Corticosteroids or megestrol acetate should be considered for patients with advanced oesophageal cancer who are anorexic.

Observational studies Low

Page 139: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 123

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

Lobo DN 2006 [281] 120 patients undergoing resection for cancers of the pancreas, oesophagus and stomach

Jejunostomy feeding with an immune modulating diet (Group A, n = 60) or an isonitrogenous, isocaloric feed (Group B, n = 60) for 10-15 days

Feed delivery, although less than targeted, was similar in both groups. There were 6 (11%) deaths in each group. Median (IQR) postoperative hospital stay was 14.5 (12-23) days in Group A and 17.5 (13-23) days in Group B (P=0.48). A total of 24 (44%) patients in each group had infective complications (P=1.0). A total of 21 (39%) patients in Group A and 28 (52%) in Group B had non-infective complications (P=0.18). Jejunostomy-related complications occurred in 26 (48%) patients in Group A and 30 (56%) in Group B (P=0.3).

12 excluded from analysis (3 protocol violoations, 9 unresectable disease)

RCT High

Ryan AM 2006 [282] 205 consecutive patients who underwent oesophagectomy for malignancy

Early enteral nutrition via a needle catheter jejunostomy

Ninety-two per cent of patients were successfully fed exclusively by NCJ post-oesophagectomy, and 94% of patients were tolerating a maintenance regimen of 2000 ml feed over 20 h by day 2 post-operatively. Patients spent a median of 15 days on jejunostomy feeding post-surgery (range 2-112 days); however, 26% required prolonged jejunostomy feeding (>20 days). Minor gastrointestinal complications were effectively managed by slowing the rate of infusion, or administering medication. Three (1.4%) serious complications of jejunostomy feeding occurred, all requiring re-laparotomy, one resulting in death. NCJ feeding was extremely effective in preventing severe post-operative weight loss in the majority of oesophagectomy patients post-op. However, oral intake was generally poor at discharge with only 65% of requirements being met orally. Sixteen patients (8%) patients required home jejunostomy feeding. By the first post-operative month, a further 6% (12) patients were recommenced on jejunostomy feeding.

Prospective study

Low

Gabor S 2005 [283] 44 consecutive patients (38 males and 6 females; mean age 62, range 30-82) with oesophageal

Early enteral feeding compared with parenteral nutrition

There was a significant difference in the interval until the first bowel movements. No difference in overall 30 d mortality was identified. A poor nutritional status was a

Observational study

Low

Page 140: Guideline pour la prise en charge du cancer oesophagien et ...

124 Upper GI Cancer KCE reports 75

Study ID Ref Search date Population Intervention Results Comments Study type Level of evidence

carcinoma (stages I-III), who had undergone radical resection and reconstruction. Historical control of 44 patients

significant prognostic factor for an increased mortality. Early enteral feeding significantly reduces the duration of ICU treatment and total hospital stay in patients who undergo oesophagectomy or oesophagogastrectomy for oesophageal carcinoma. The mortality rate is not affected.

Treatment of recurrent disease

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

CBO [56] July 2003 Bij patiënten met … een recidief na eerdere oesofagusresectie in verband met een oesofaguscarcinoom heeft stentplaatsing de voorkeur.

Only observational studies were found, but no prospective trials.

Page 141: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 125

APPENDIX 5

EVIDENCE TABLES OF GASTRIC CANCER BY CLINICAL QUESTION Diagnosis of gastric cancer.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Alarm symptoms

SIGN [47] 2004 Patients presenting with any of the following alarm symptoms should be referred for early endoscopy: dysphagia, recurrent vomiting, anorexia, weight loss, gastrointestinal blood loss.

Meineche-Schmidt V 2002 Numans ME 2001 Wallace MB 2001 Kapoor N 2005

Low

FNCLCC [129] January 2002

Le bilan diagnostique repose sur la recherche des signes cliniques d’appel (douleur abdominale, amaigrissement, dyspepsie), … (standard).

Potet 1993 Faivre 1997

Very low

Oesophagogastroscopy

SIGN [47] 2004 Flexible upper GI endoscopy is recommended as the diagnostic procedure of choice in patients with suspected gastric cancer.

Graham DY 1982 Abbas SZ 2004

Low

Routine use of chromoendoscopy during upper GI endoscopy is not recommended, but may be of value in selected patients at high risk of gastric malignancy.

Mitooka H 1995 Very low

FNCLCC [129] January 2002

Le bilan diagnostique repose sur … la gastroscopie … (standard). No evidence provided

Barium X-ray

SIGN [47] 2004 No recommendation, only discussion

Biopsy

FNCLCC [129] January 2002

Le bilan diagnostique … doit être confirmé par des biopsies multiples. Ces biopsies (nombre minimum 5 à 8) doivent être réalisées sur toutes les anomalies du relief muqueux permettant un examen d’anatomie et cytologie pathologique (standard).

No evidence provided

Study ID Ref Search

date Population Intervention Results Comments Study type Level of

evidence Alarm symptoms

Bowrey DJ 2006 [49] NA Patients referred for open-access gastroscopy

Application of referral guidelines using alarm symptoms

Gastroscopy identified oesophagogastric carcinoma in 123 (3%) of the 4,018 subjects. Of these 123 patients, 104 (85%) with oesophagogastric cancer had ‘‘alarm’’ symptoms (anemia, mass, dysphagia, weight loss, vomiting) and

Prospective study

Low

Page 142: Guideline pour la prise en charge du cancer oesophagien et ...

126 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

would have satisfied the referral criteria. The remaining 15% would not have been referred for initial endoscopic assessment because their symptoms were those of uncomplicated ‘‘benign’’ dyspepsia. The patients with ‘‘alarm’’ symptoms had a significantly more advanced tumor stage (metastatic disease in 47% vs 11%; p < 0.001), were less likely to undergo surgical resection (50% vs 95%; p<0.001), and had a poorer survival (median, 11 vs 39 months; p = 0.01) than their counterparts without such symptoms.

Marmo R 2005 [233] NA Patients with uncomplicated dyspepsia

Endoscopy Identification of risk factors

A total of 5,224 patients with uncomplicated dyspepsia were considered (training sample). Twenty-two (16 males) had malignancy at endoscopy. These patients were about 20 yr older than patients with no malignancy (p < 0.001). The mean age of females with cancer was almost 10 yr higher compared to males (p= 0.08). Such differences in age were confirmed in a split sample of 3,684 patients (p < 0.001 and p < 0.05, respectively). The age cut-offs identified were 35 yr for males and 56 yr for females.

Prospective study

Low

Oesophagogastroscopy

Lam EC 2007 [284] Patients with thickening of the gastric wall on CT (n = 71)

Upper endoscopy and EUS by one endosonographer, blinded to the results of previous investigations

Normal upper endoscopy was strongly associated with normal EUS. Abnormal upper endoscopy was associated with abnormal EUS in 70% of cases.

Prospective study

Low

Barium X-ray

No additional studies found

Biopsy

No additional studies found

Page 143: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 127

Staging of patients with gastric cancer.

CPG ID Ref Search date Recommendation Supporting evidence

Level of evidence

CT scan

SIGN [47] 2004 In patients with gastric cancer, CT scan of the chest and abdomen with IV contrast and gastric distention with oral contrast or water should be performed routinely. The liver should be imaged in the portal venous phase.

Kamel IR 2004 Low

FNCLCC [129] January 2002 Le bilan d’extension doit inclure au minimum un scanner abdominopelvien et une radiographie du thorax (standard).

Lee 2001 Delia 2000 Mani 2001 Dux 1999 Davies 1997

Low

Le scanner thoracique peut être proposé pour le bilan d’extension (option).

Endoscopic ultrasonography

FNCLCC [129] January 2002 Une écho-endoscopie peut être proposée pour affiner le diagnostic en cas d’hypertrophie des plis gastriques sans histologie, et pour le diagnostic des tumeurs classées T4 et pour mieux apprécier l’envahissement pariétal et ganglionnaire si un traitement néoadjuvant est envisagé (option).

Kelly 2001 Mancino 2000 Willis 2000 Tseng 2000 Matsumoto 2000

Low

Laparoscopy

SIGN [47] 2004 Laparoscopy should be considered in patients with oesophageal tumours with a gastric component, and in patients with gastric tumours being considered for surgery where full thickness gastric wall involvement is suspected.

Fujimara T 2002 Clements DM 2004 Molloy RG 1995 Wilkiemeyer MB 2004

Low

FNCLCC [129] January 2002 Une laparoscopie peut être proposée pour améliorer l’établissement du stade préthérapeutique dans le cas où un risque d’extension péritonéale existe, notamment dans les tumeurs suspectées comme T3 et T4 (option).

Numerous observational studies

Low

Magnetic resonance imaging

SIGN [47] 2004 MRI should be reserved for those patients who cannot undergo CT or used for additional investigation following CT/EUS.

Sohn KM 2000 Wu LF 2003 Kersjes 1997 Lauenstein TC 2004 Wong R 2000

Low

PET scan

SIGN [47] 2004 PET is not routinely indicated in the staging of gastric cancers. Mochiki E 2004 Yeung HW 1998

Low

FNCLCC [66] October 2002

L’impact de la TEP-FDG sur la prise en charge thérapeutique des patients atteints d’un cancer de l’estomac reste à déterminer par des études prospectives (recommendation).

McAteer 1999 Hoffmann 1999 Couper 1998 Depotter 2002 Kinkel 2002

Page 144: Guideline pour la prise en charge du cancer oesophagien et ...

128 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

CT scan

Kwee RM 2007 [130] August 2006

Patients with gastric carcinoma without neoadjuvant treatment

MDCT Pathologic examination after surgery as standard

6 studies on MDCT identified Diagnostic accuracy of overall T staging: 77% - 89% Sensitivity for assessing serosal involvement: 83% - 100% Specificity for assessing serosal involvement: 80% - 97%

English articles only Also retrospective studies included Overall moderate quality of included studies

SR Low

Chen CY 2007 [131] Patients with gastric cancer (M0) (n = 69)

MDCT Pathologic examination after surgery as standard (n = 55)

Overall accuracy of T staging: 89% (with MPR images), or 49/55 correctly staged Overall accuracy of N staging: 78% (with MPR images), or 43/55 correctly staged

Prospective study

Low

Kikuchi S 2006 Patients with gastric adenocarcinoma (n = 74)

MDCT Standard: histopathological diagnosis (n = 70); endoscopic biopsies and clinical results (n = 4)

Tumor detected in only 47% of patients. Insufficient data to calculate diagnostic measures.

Standard not identical for all patients

Prospective study

Very low

Endoscopic ultrasonography

Kwee RM 2007 [130] August 2006

Patients with gastric carcinoma without neoadjuvant treatment

EUS Pathologic examination after surgery as standard

23 studies on EUS identified Diagnostic accuracy of overall T staging: 65% - 92% Sensitivity for assessing serosal involvement: 78% - 100% Specificity for assessing serosal involvement: 68% - 100%

English articles only Also retrospective studies included Overall moderate quality of included studies

SR Low

Ichikawa T 2007 [285] Patients with early gastric cancer scheduled for endoscopic submucosal dissection

EUS, three different miniature probes

30-MHz probes most accurate of invasion depth (92%)

Prospective study

Low

Gines A 2006 [286] Patients with large gastric folds at endoscopy and biopsies negative for malignancy (n = 61)

EUS Standard: histology or clinical follow-up of at least 2 years

Enlargement of deep layers was only independent predictive factor for malignancy.

Prospective study

Low

Matthes K 2006 [287] TA-EUS vs. L-EUS Excluded: only 4 patients with gastric cancer

RCT

Page 145: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 129

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Chu KM 2004 [288] Patients with histopathologically confirmed adenocarcinoma of the stomach

EUS for detection of ascites Standard: staging laparoscopy

Sensitivity 61%, specificity 99% Prospective study

Low

Laparoscopy

Mortensen MB 2006 [79] 411 patients with upper gastrointestinal tract cancer, of which 134 had biopsy-proven gastric cancer

Combined endoscopic ultrasonography (EUS) and laparoscopic ultrasonography (LUS) ‘Standard’: treatment actually undertaken

The combination of EUS and LUS correctly predicted R0 resection in 90.6%, R1–R2 in 91% and irresectability in 91.4% of patients. Ten patients (2.4%) had explorative laparotomy only. There were no complications associated with the EUS and LUS procedures.

To avoid false-positive findings with regard to local irresectability ultrasonographic criteria were adjusted so that only a combination of several ultrasonographic criteria with a 100 per cent predictive value for local irresectability were used Standard is not identical for all patients

Prospective study

Very low

Deogracias ML 2006 [289] Patients operated on for gastric carcinoma (n = 41)

Staging laparoscopy Open gastrectomy in 33 cases

No port-site metastases or port-site recurrence

Selection bias: study sample is taken from a prospective database of surgically treated patients

Prospective study

Very low

Magnetic resonance imaging

Kwee RM 2007 [130] August 2006

Patients with gastric carcinoma without neoadjuvant treatment

MRI Pathologic examination after surgery as standard

3 studies on MRI identified Diagnostic accuracy of overall T staging: 71% - 83% Sensitivity for assessing serosal involvement: 90% - 93% Specificity for assessing serosal involvement: 91% - 100%

English articles only Also retrospective studies included Overall moderate quality of included studies

SR Low

PET scan

No additional studies found

Sentinel lymph node mapping

Lee JH 2006 [136] Patients with cT1N0 gastric adenocarcinoma (T < 5 cm) (n = 64)

Dye and isotope double tracers to improve SLNB Histology as standard

Overall sensitivity 71%; pT1 subset: 88% Prospective study

Low

Mochiki E 2006 [138] Patients with cN0M0 SLN mapping with Tc-99m Overall sensitivity 83% and specificity Prospective Low

Page 146: Guideline pour la prise en charge du cancer oesophagien et ...

130 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

gastric adenocarcinoma (n = 59)

colloidal rhenium sulphide Histology as standard

100%; pT1 subset 100% sensitivity study

Gretschel S 2005 [135] Patients with stage I-IV gastric cancer

SLN mapping with 99mTc-colloid (n = 15) or dye (n = 19) Maruyama computer model Histology as standard

Difficult to interpret Only 33 patients included in calculation of accuracy

Prospective study

Very low

Miwa K 2003 [137] Patients with early gastric carcinoma treated with surgery (n = 211)

Sentinel node biopsy after mapping with vital dye

The assay was successful in mapping the lymphatic basins in 203 (96%) of 211 patients. The dye stained one or more metastatic nodes in 31 patients, but failed to indicate a metastatic node in four patients with a large involved node. Meticulous postoperative examination of all resected nodes in the standard paraffin slices revealed no new metastases. Sensitivity 89%, specificity 100%.

Prospective study

Low

Simsa J 2003 [139] Patients with gastric cancer treated with surgery (D2 lymphadenectomy) (n = 22)

Sentinel node biopsy after mapping with vital dye

Success rate of sentinel node detection with a good relation between metastatic involvement of the sentinel node and other nodes was 56% (13 out of 22 patients). Two false-negatives. Insufficient data to calculate diagnostic measures.

Prospective study

Very low

Page 147: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 131

Diagnosis and treatment of early lesions.

CPG ID Ref Search date

Recommendation Supporting evidence

Level of evidence

SIGN [47] 2004 Routine use of chromoendoscopy during upper GI endoscopy is not recommended, but may be of value in selected patients at high risk of gastric malignancy.

Mitooka H 1995 Very low

Pathologists should follow the revised Vienna classification for reporting dysplasia. Schlemper RJ 2000 Where radical intervention is contemplated on the basis of high-grade dysplasia or early adenocarcinoma the

diagnosis should be validated by a second pathologist experienced in this area and further biopsies should be taken if there is uncertainty.

Patients diagnosed with high-grade dysplasia should have careful assessment (CT, EUS, rigorous biopsy protocol +/- EMR) to exclude coexisting cancer.

In the absence of invasive cancer, patients with high-grade dysplasia should be offered endoscopic treatment. Takekoshi T 1994 Very Low The assessment and management of patients with high-grade dysplasia should be centralised to units with the

appropriate endoscopic facilities and expertise.

Early gastric cancer limited to the superficial submucosa should be treated by EMR. Wang YP 2006 Low Mucosal ablative techniques, such as PDT, APC, or laser should be reserved for the management of residual

disease after EMR and not for initial management if invasive disease is present in patients fit for surgery. No evidence provide for gastric cancer

FNCLCC [129] January 2002

La mucosectomie n’est pas une attitude standard (standard).

La mucosectomie endoscopique peut être proposée pour les patients à risques opératoires présentant une lésion usT1N0 limitée à la muqueuse sans envahissement de la muscularis mucosae (option).

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Lee SH 2006 [290] Patients with early gastric cancer (n = 72)

EMR with submucosal injection of normal saline solution (n = 36) vs. fibrinogen mixture solution (n = 36)

No differences in hard outcomes. Shorter mean procedure time in FM group.

Randomization with sealed, opaque envelopes Not clear if blinded assessors

RCT Moderate

Page 148: Guideline pour la prise en charge du cancer oesophagien et ...

132 Upper GI Cancer KCE reports 75

Neoadjuvant treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Chemotherapy

SIGN [47] 2004 The neoadjuvant use of either chemotherapy or radiotherapy for patients with gastric cancer is not recommended outside clinical trials.

Janunger KG 2002 Earle CC 2003

High

CCO [150] April 2003

There is insufficient evidence from randomized trials to recommend neoadjuvant chemotherapy outside of a clinical trial.

Songun I 1999 Fujii M 1999 (abstract) Kang YK 1999 (abstract)

High

FNCLCC [129] January 2002

La chimiothérapie néoadjuvante n’est pas indiquée dans les cancers de l’estomac résécables en dehors d’essais thérapeutiques.

Songun I 1999 Fujii M 1999 (abstract) Kang YK 1999 (abstract)

High

Radiotherapy

SIGN [47] 2004 The neoadjuvant use of either chemotherapy or radiotherapy for patients with gastric cancer is not recommended outside clinical trials.

Janunger KG 2002 Earle CC 2003

High

CCO [150] April 2003

There is insufficient evidence from randomized trials to recommend neoadjuvant radiation therapy outside of a clinical trial.

Zang ZX 1998 Skoropad V 1999a Skoropad V 1999b Skoropad V 2002

High

FNCLCC [129] January 2002

No recommendations, only discussion

Immunotherapy

CCO [150] April 2003

There is insufficient evidence from randomized trials to recommend neoadjuvant immunotherapy outside of a clinical trial.

Gouchi A 1997 Peters KM 1990 Terashima M 1998

High

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Chemotherapy

Wu AN 2007 [147] June 2005

Patients with resectable gastric cancer

Neoadjuvant chemotherapy followed by surgery vs. surgery alone

Of the four clinical trials enrolled, there were 250 and 332 cases in total, with 106 and 126 deaths at the end of follow-up in the NAC and control group, respectively. The OR (odds ratio) was 1.05 (95%CI: 0.73-1.50), which was not statistically significant. Of the evaluable 129 patients receiving NAC, 28.7% demonstrated either a complete or a partial response. Two studies of NAC in resectable gastric cancer had resection rate data available for analysis The R0 resection rate in the NAC group was

SR High

Page 149: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 133

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

comparable to that in the control (OR: 0.96 (95%CI: 0.51-1.83)). The morbidity and mortality of NAC varied with the regimens used preoperatively. Of the 129 patients included in the analyzed studies, some acceptable toxicity was observed.

Cunningham D 2006

[148] Patients with histologically proven adenocarcinoma of the stomach or lower third of the oesophagus that was considered to be stage II (through the submucosa) or higher, with no evidence of distant metastases, or locally advanced inoperable disease, as evaluated by computed tomography, chest radiography, ultrasonography, or laparoscopy.

Perioperative chemotherapy and surgery (n = 250) vs. surgery alone (n = 253)

As compared with the surgery group, the perioperative-chemotherapy group had a higher likelihood of overall survival (HR for death, 0.75; 95%CI 0.60 to 0.93; P=0.009; five-year survival rate, 36 percent vs. 23 percent) and of progression-free survival (HR for progression, 0.66; 95%CI 0.53 to 0.81; P<0.001). No separate data for gastric cancer, but no clear evidence of heterogeneity of treatment effect according to the site of the primary tumour

Randomization procedure? No information of blinding

RCT Moderate

Radiotherapy

No additional studies found

Immunotherapy

Ates E 2004 [291] Excluded: only 11 patients with gastric cancer; no separate analysis of these patients

RCT

Romano F 2004a [292] Patients with histologically proven gastric adenocarcinoma treated with radical surgery (n = 69)

Preoperative IL-2 (n = 34) vs. surgery alone (n = 35)

IL-2 group showed lower postoperative complications (2/34 vs. 11/35; P < 0.05). Median overall and disease-free survivals were longer, even if not significantly, in the IL-2 group than in the control arm (P = 0.07 and P = 0.06 respectively).

No information on randomization procedure or blinding

RCT Moderate

Romano F 2004b

[151] Patients with histologically proven gastric adenocarcinoma treated with radical surgery (n = 39)

Preoperative IL-2 (n = 19) vs. surgery alone (n = 20)

Excluded: same patients

RCT

Page 150: Guideline pour la prise en charge du cancer oesophagien et ...

134 Upper GI Cancer KCE reports 75

Restaging after neoadjuvant treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 Patients with locally advanced disease having chemotherapy/chemoradiotherapy should have their response assessed for an impact on the potential to operate ; following a good response the patient should be restaged and the role restaged and the role of surgery re-evaluated by the multidisciplinary team.

Ross P 1996 Wilke H 1995

Low

Surgical treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 Gastric cancer resectional surgery should be carried out in high volume specialist surgical units by frequent operators.

Bachmann MO 2002 Low

Surgery for gastric cancer should be aimed at achieving an R0 resection. Marubini E 2002 Bozzetti F 1997

High

Consideration should be given to pouch formation after total gastrectomy. Lehnert T 2004 High? D2 lymphadenectomy, with a minimum of 25 lymph nodes removed, should be considered for patients

undergoing gastrectomy. Routine resection of additional organs (spleen and pancreas) during gastrectomy is not recommended.

FNCLCC [129] January 2002

Une chirurgie d’exérèse à visée curative doit être réalisée (standard). Maruyama 1987 Jakl 1995 Allum 1989

Pour les tumeurs distales (1/3 inférieur, antre), il est indiqué de pratiquer une gastrectomie des 4/5 et curage ganglionnaire monobloc avec élargissement monobloc de nécessité aux organes de voisinage atteints (standard).

Gouzi 1989 Bozzetti 1999

High

Pour les tumeurs proximales (2/3 supérieurs), il est indiqué de pratiquer une gastrectomie totale et curage ganglionnaire monobloc avec élargissement monobloc de nécessité aux organes de voisinage atteints (standard).

No evidence provided

Pour les cancers superficiels et pour des petites tumeurs limitées, les techniques de chirurgie laparoscopique sont applicables dans le cadre d’études prospectives (option).

Observational studies Low

Un curage ganglionnaire de type D1 (ganglions périgastriques, groupes 1 à 6 soit ≥ 15 ganglions) doit être réalisé selon la topographie de la tumeur (standard).

Dent 1988 Robertson 1994 Cischieri 1996 Bonenkamp 1999 Wu 2004

High

Un curage de type D2 (D1 + ganglions pédiculaires (groupes 1 à 11 soit ≥ 25 ganglions)) sans splénectomie ni pancréatectomie peut être envisagé (option).

Dent 1988 Robertson 1994 Cischieri 1996 Bonenkamp 1999 Wu 2004

Page 151: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 135

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Csendes 2002 Des essais randomisés évaluant le curage D2 sans splénectomie sont recommandés. Une splénectomie

est indiquée en cas d'adénopathies de l'artère splénique ou de cancer de la grosse tubérosité atteignant la séreuse (recommandation).

Csendes 2002 Subanalyses of RCTs

Moderate

Il n’y a pas de modalités standards de reconstruction après gastrectomie distale subtotale (standard). Chareton 1996 High La reconstruction peut être effectuée par rétablissement de continuité par anastomose gastrojéjunale ou

par établissement de continuité par anastomose gastroduodénale (option).

Il est recommandé de privilégier le rétablissement de continuité par la technique habituelle du chirurgien (recommandation).

No evidence provided

Après gastrectomie totale, le rétablissement de la continuité digestive peut se faire par anse jéjunale selon la technique en Y de Roux, avec ou sans reconstitution d’un réservoir (J Pouch), ou par constitution d’un réservoir avec interposition entre oesophage et duodénum d’une anse jéjunale en S ou du côlon transverse gauche (option).

5 RCTs 4 prospective trials

Low

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Volume-outcome relation

Enzinger P 2007 [293] Patients with stage IB through IV (M0) gastric and gastroesophageal junction adenocarcinoma, previously randomized to adjuvant chemoradiation after surgery or surgery alone (n = 448)

Effect of hospital surgical volume, as assessed by Medicare claims data, on overall survival and gastric cancer recurrence

Hospital surgical volume was not predictive of overall survival (P = 0.46) or disease-free survival (P = 0.43) at a median follow-up of 8.9 years. However, patients who underwent either a D1 or D2 dissection at a high- or moderate-volume center experienced an adjusted hazard ratio of 0.80 (95% CI, 0.53-1.20) for overall survival and 0.78 (95% CI, 0.53-1.14) for disease-free survival compared with those patients resected at a low-volume hospital; these results were not statistically significant. When a D0 resection was performed, hospital procedure volume showed no impact on survival.

Retrospective selection of patients from a RCT (n = 556)

Observational study

Very low

Type of surgery

Hosono S 2006 [164] August 2006

Patients with gastric cancer

Laparoscopy assisted distal gastrectomy (LADG) vs. conventional open distal gastrectomy (CODG)

Compared to CODG, LADG was a longer procedure (weighted mean difference 54.3; 95%CI 38.8 to 69.8; P < 0.001), but was associated with a lower associated morbidity (OR 0.54; 95%CI 0.37 to 0.77; P < 0.001); this was most significant for postoperative ileus (OR

Inclusion of 4 RCTs (only 1 Western study) and 12 retrospective studies English only No information on how quality appraisal was done

SR Moderate

Page 152: Guideline pour la prise en charge du cancer oesophagien et ...

136 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

0.27; 95%CI 0.09 to 0.84; P = 0.02). There was no significant difference between the two groups in anastomotic, pulmonary, and wound complications and mortality.

Hayashi H 2005 [163] Patients with early gastric cancers in the lower half of the stomach (n = 28)

Open distal gastrectomy (n = 14) vs. laparoscopy-assisted distal gastectomy (n = 14)

The LADG group required a significantly shorter period of postoperative epidural anesthesia and had no major postsurgery complications. Pathologic examinations showed that surgery was equally radical in the two groups.

Randomization with sealed envelope Blinding???

RCT Moderate

Huscher C 2005 [165] Patients with distal gastric cancer

Laparoscopic-assisted (n = 30) vs. open radical subtotal gastrectomy (n = 29)

Operative mortality rates were 6.7% (2 patients) in the OG and 3.3% (1 patient) in the LG (P = not significant); morbidity rates were 27.6% and 26.7%, respectively (P = not significant). Five-year overall and disease-free survival rates were 55.7% and 54.8% and 58.9% and 57.3% in the OG and the LG, respectively (P = not significant).

Not clear if randomization was blinded Blinded assessment??

RCT Moderate

Inaba T 2004 [294] Patients with gastric cancer undergoing distal gastrectomy or total gastrectomy

Upper midline incision vs. transverse incision

Times for both opening and closing the abdominal cavity were longer with a transverse incision, in both the distal gastrectomy group and total gastrectomy group. In the patients in whom continuous epidural analgesia was used postoperatively, the number of additional doses of analgesics was smaller in the transverse-incision group after distal gastrectomy. The incidence of postoperative pneumonia was lower in the transverse-incision group after distal gastrectomy. The number of patients with postoperative intestinal obstruction was smaller in the transverse-incision group than in the midline-incision group after distal gastrectomy. In contrast to distal gastrectomy, there was no significant difference in the number of doses of postoperative analgesics, incidence of postoperative pneumonia, or incidence of postoperative intestinal obstruction between the two study groups after total gastrectomy.

No information about randomization procedure Blinding???

RCT Moderate

Lee JH 2005 [166] Patients with early gastric cancer

Laparoscopy-assisted distal gastrectomy (LADG) including lymphadenectomy vs.

The mean postoperative hospital stay and the duration of analgesic administration were shorter for group L but not significantly. Postoperative pulmonary complications occurred more

Blinding?? RCT Moderate

Page 153: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 137

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

open distal gastrectomy frequently in the O group (p = 0.043). At a median follow-up of 14 months, there has been no recurrence of disease in either group.

Lee WJ 2003 [295] Patients with gastric cancer treated with extended lymph node dissection (D2) during gastrectomy

D2 gastric resection performed with the LigasureTM system (n = 40) vs. resection using conventional haemostatic methods (n = 40)

Ligasure was associated with less intraoperative blood loss (mean (s.d.) 142(73) versus 239(124) ml; P = 0.001) and a shorter operating time (mean(s.d.) 169(25) versus 222(28) min; P = 0.001) than conventional operation. Postoperative drainage fluid volumes were greater in the Ligasure group (mean (s.d.) 1577(940) versus 886(542) ml; P = 0.020). There were no differences in postoperative complications or hospital stay.

No information on randomization procedure

RCT Moderate

Sasako M 2006 [296] Patients with gastric cancer of the cardia or subcardia

Left thoracoabdominal approach (LTA) (n = 85) vs. abdominal-transhiatal approach (TH) (n = 82)

5-year overall survival was 52.3% (95% CI 40.4-64.1) in the TH group and 37.9% (26.1-49.6) in the LTA group. The hazard ratio of death for LTA compared with TH was 1.36 (0.89-2.08, p=0.92). Three patients died in hospital after LTA but none after TH. Morbidity was worse after LTA than after TH.

No blinding RCT Moderate

Yu W 2006 [297] Patients with gastric adenocarcinoma who underwent total gastrectomy (n = 216)

Total gastrectomy either with (n = 104) or without (n = 103) splenectomy

Gastrectomy combined with splenectomy tended to be associated with slightly higher morbidity and mortality rates, a slightly greater incidence of lymph node metastasis at the splenic hilum and along the splenic artery, and marginally better survival, but there were no statistically significant differences between the groups. Splenectomy had no impact on survival in patients with metastatic lymph nodes at the hilum of the spleen or in those with metastatic lymph nodes along the splenic artery.

9 patients excluded from analysis No information on blinding

RCT Moderate

Lymphadenectomy

McCulloch 2004 [152] April 2001

Patients with gastric cancer

Limited vs. extended lymph node removal during gastrectomy

Meta-analysis of randomised trials did not reveal any survival benefit for extended lymph node dissection (Risk ratio = 0.95 (95%CI 0.83 - 1.09), but showed increased postoperative mortality (RR 2.23, 95% CI 1.45 - 3.45). Pre-specified subgroup analysis suggested a possible benefit in stage T3+ tumours (RR = 0.68, 95% CI 0.42-

Two randomised and two non-randomised comparisons of limited (D1) versus extended (D2) node dissection and 11 cohort studies of either D1 or D2 resection were

SR High

Page 154: Guideline pour la prise en charge du cancer oesophagien et ...

138 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

1.10). Non-randomised comparisons showed no significant survival benefit for extended dissection (RR 0.92, 95% CI 0.83 -1.02), but decreased mortality (RR 0.65, 95% CI 0.45-0.93). Subgroup analysis showed apparent benefit in UICC stage II and IIIa. Observational studies of D2 resection reported much better mortality and survival than those of D1 surgery, but the settings were strikingly different.

analysed.

Degiuli M 2004 [154] Patients with potentially curable gastric cancer

D1 (n = 76) and D2 gastrectomy (n = 86)

Complications developed in 10.5% of patients after D1 and in 16.3% of patients after D2 gastrectomy (p<0.29). Reoperation rate was 3.4% after D2 and 2.6% after D1 resection. Post-operative mortality rate was 1.3% after D1 and 0% after D2 gastrectomy.

No information on blinding RCT Moderate

Hartgrink H 2004 [156] Patients with gastric adenocarcinoma treated with curative intent

Limited (D1) (n = 380) and extended (D2) lymph node dissection (n = 331)

Morbidity (25% v 43%; P <.001) and mortality (4% v 10%; P =.004) were significantly higher in the D2 dissection group. After 11 years there is no overall difference in survival (30% v 35%; P =.53). Of all subgroups analyzed, only patients with N2 disease may benefit of a D2 dissection. The relative risk ratio for morbidity and mortality is significantly higher than one for D2 dissections, splenectomy, pancreatectomy, and age older than 70 years.

No information on randomization or blinding

RCT Moderate

Kulig J 2007 [157] Patients with gastric adenocarcinoma treated with curative intent

Standard D2 (n = 141) or extended D2+ (n = 134) lymphadenectomy

The overall morbidity rates were comparable in D2 (27.7%; 95% confidence interval [CI], 20.3-35.1) and D2+ (21.6%; 95% CI, 13.7-29.5) groups (P = .248). Pre-existing cardiac disease, splenectomy, and excessive blood loss were identified as risk factors for overall and nonsurgical complications. Postoperative mortality rates were 4.9% (95% CI, 1.4-8.5) and 2.2% (95% CI, 0-4.7), respectively (P = .376).

Interim analysis Separate randomization for all participating centres No information on blinding

RCT Moderate

Sano T 2004 [158] Patients with potentially curable gastric adenocarcinoma (T2-subserosa, T3, or T4) (n = 523)

Japanese standard D2 vs. D2 + para-aortic nodal dissection

Although the morbidity for the extended surgery group (28.1%) was slightly higher than the standard group (20.9%), there was no difference in the incidence of four major complications (anastomotic leak, pancreatic fistula, abdominal abscess, pneumonia) between the two groups.

No information on blinding RCT Moderate

Page 155: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 139

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Hospital mortality was reported at 0.80%: one patient in each group died of operative complications, while one from each group died of rapid progressive cancer while inpatient.

Wu C 2004 & 2006

[298] Patients with gastric adenocarcinoma treated with curative intent

D1 (ie, level 1) lymphadenectomy (n = 110) vs. D3 (ie, levels 1, 2, and 3) dissection (n = 111)

Overall 5-year survival was significantly higher in patients assigned D3 surgery than in those assigned D1 surgery (59.5% [95% CI 50.3-68.7] vs 53.6% [44.2-63.0]; difference beteween groups 5.9% [-7.3 to 19.1], log-rank p=0.041). 215 patients who had R0 resection (ie, no microscopic evidence of residual disease) had recurrence at 5 years of 50.6% [41.1-60.2] for D1 surgery and 40.3% [30.9-49.7] for D3 surgery (difference between groups 10.3% [-3.2 to 23.7], log-rank p=0.197).

No blinding RCT Moderate

Yonemura Y 2006

[159] Patients with advanced gastric adenocarcinoma

D2 (level 1 and 2 lymphadenectomy) (n = 128) vs. D4 (D2 plus lymphadenectomy of para-aortic lymph nodes) dissection (n = 128)

Five (4%) and two (2%) medical complications developed in the D2 and D4 groups, respectively. Surgical complications developed in 28 (22%) and 48 patients (38%) after D2 and D4 gastrectomy. The most common complications were anastomotic leakage, pancreatic fistula, and abdominal abscess. Pancreatic fistula developed in 6 (19%) of 32 patients after D4 plus pancreatosplenectomy, but the incidence of pancreatic fistula after D2 gastrectomy plus pancreatosplenectomy was low (6%, 1/16). Two patients died within 30 days of operation (0.8%, 2/256), and each patient belonged to the D2 and D4 group.

No information on randomization procedure. No blinded assessment

RCT Moderate

Reconstruction

Adachi S 2003 [299] Patients with gastric cancer (n = 30)

Roux-en-Y esophagojejunostomy (R-Y), R-Y with pouch (P-Y), and jejunal interposition with pouch (P-I) after total gastrectomy

No information on hard endpoints. Reconstruction should be performed with pouch formation after total gastrectomy with curative intent.

Randomization with sealed envelope In case or recurrence or impossible follow-up within 2 years after surgery: replacement by other patient Blinding???

RCT Moderate

Ishikawa M 2005 [300] Patients with gastric Roux-en-Y 5 of 24 patients with RY reconstruction Randomization with sealed RCT Moderate

Page 156: Guideline pour la prise en charge du cancer oesophagien et ...

140 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

carcinoma who underwent distal gastrectomy (n = 50)

reconstruction vs. conventional Billroth I reconstruction

developed gastrojejunal stasis in the early postoperative period, which led to a longer postoperative hospital stay as compared with the B-I group (mean +/- S.D; B-I; 19.0 +/- 6.2, RY; 31.8 +/- 21.7 days) (P < 0.05). Endoscopic examination revealed that the frequency of bile reflux (P < 0.01) and degree of inflammation in the remnant stomach (P < 0.05) were less in the RY group than in the B-I group. However, inflammatory findings in the lower esophagus were observed in 7 (27%) of B-I, and 8 (35%) of the RY group, suggesting that late phase esophagitis was not improved in the RY group.

envelopes No information on blinding

Iwata T 2006 [160] Patients with gastric cancer in the upper third of the stomach who underwent proximal gastrectomy (n = 9)

Reconstruction by jejunal pouch interposition (JPI group) (n = 4) vs. reconstruction by jejunal interposition (JI group) (n = 5)

The JPI group showed a significant dietary advantage. Three months after surgery, JPI patients could eat more than 80% of the volume of their preoperative meals, whereas JI patients ate less than 50%. The percentage of postoperative body weight loss was higher in the JI group than in the JPI group because the volume of the remnant stomach was more adequate in the latter. Moreover, it was easier to enter the remnant stomach and duodenum for endoscopic fiberscopy in the JPI group for the treatment of hepato-biliary pancreatic disease.

Very small sample size No information on randomization procedure or blinding

RCT Moderate

Kono K 2003 [162] Patients receiving total gastrectomy for early gastric cancer (n = 50)

Roux-en-Y reconstruction group without pouch (RY group) or the jejunal pouch reconstruction group (pouch group)

Jejunal pouch reconstruction provided the better QOL than Roux-en-Y reconstruction without pouch both at short-term and long-term periods. Pouch reconstruction provided less bile reflux into the esophagus compared with Roux-en-Y reconstruction.

No information on randomization procedure or blinding

RCT Moderate

Mochiki E 2004 [301] Patients with gastric cancer treated by total gastrectomy (n = 12)

Jejunal interposition without a pouch (n = 12) vs. jejunal interposition with a pouch (n = 14)

The interposed jejunum with a pouch shows marked disturbances from the motor pattern of a normal jejunum during the fasting and fed states. These motor abnormalities may be responsible for insufficient food intake of the J-P group.

No information on randomization procedure or blinding

RCT Moderate

Shibata C 2004 [302] Patients with early gastric cancer (n = 81)

Pylorus-preserving gastrectomy (PPG) (n = 36) vs. conventional distal gastrectomy (CDG) with

There were no differences in early results between PPG and CDG. The incidence of early dumping syndrome was lower in PPG (8%) than in CDG (33%). Other late results including the

Randomization with sealed envelope No information on blinding 7 patients excluded from

RCT Moderate

Page 157: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 141

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Billroth I anastomosis (n = 38)

incidence of gallstones were not different between the 2 groups.

analysis

Yoo C 2005 [161] Patients with upper third gastric cancer

Proximal gastrectomy with jejunal pouch interposition (PGJP) (n = 25) vs. Roux-en-Y esophagojejunostomy (TGRY) (n = 26)

No significant differences in operating time, hospital stay, and postoperative complications. Blood loss was significantly less in the PGJP group (P = 0.036). Nineteen patients (73%) in the TGRY group had one or more postgastrectomy symptoms, which was significantly more frequent than in the PGJP group (32%; P = 0.012). There were also significant differences between the two groups with regard to food intake, weight recovery, hemoglobin, and serum vitamin B12 levels in favour of PGJP.

RCT High

Adjuvant treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Chemotherapy

SIGN [47] 2004 Postoperative chemotherapy for patients with gastric cancer is not recommended outside a clinical trial. Janunger 2002 Earle 2003

High

CCO [150] April 2003

For patients unable to undergo radiation, adjuvant chemotherapy alone may be of benefit, particularly for patients with lymph node metastases. The optimal regimen remains to be defined.

Numerous MA and RCTs High

FNCLCC [129] January 2002

La chimiothérapie adjuvante n’est pas indiquée dans le traitement des cancers de l’estomac en dehors d’essais thérapeutiques (standard).

Numerous MA and RCTs High

Il est recommandé de poursuivre les essais thérapeutiques, en améliorant les protocoles de chimiothérapie, comparant la chimiothérapie adjuvante à la chirurgie seule ou à la chirurgie + radiochimiothérapie postopératoire (recommandation).

Radiotherapy

CCO [150] April 2003

There is insufficient evidence from randomized trials to recommend adjuvant radiation therapy outside of a clinical trial.

Hallissey 1994 Kramling 1996

High

FNCLCC [129] January 2002

La radiothérapie externe adjuvante n'est pas une attitude thérapeutique standard dans les cancers de l'estomac (standard).

Hallissey 1994 High

Chemoradiotherapy

SIGN [47] 2004 Postoperative chemoradiation for patients with gastric cancer is not recommended outside a clinical trial.

McDonald JS 2001 Moderate

CCO [150] April 2003

Following surgical resection, patients whose tumours penetrated the muscularis propria or involved regional lymph nodes should be considered for adjuvant combined chemoradiotherapy. The current standard protocol consists of one cycle of 5-FU (425 mg/m2/day) and leucovorin (20 mg/m2/day) in a

Dent 1979 Moertel 1984 McDonald 2001

High

Page 158: Guideline pour la prise en charge du cancer oesophagien et ...

142 Upper GI Cancer KCE reports 75

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

daily regimen for five days, followed one month later by 4,500 cGy (180 cGy/day) of radiation given with 5-FU (400 mg/m2/day) and leucovorin (20 mg/m2/day) on days 1 through 4 and the last three days of radiation. One month after completion of radiation, two cycles of 5-FU (425 mg/m2/day) and leucovorin (20 mg/m2/day) in a daily regimen for five days are given at monthly intervals.

FNCLCC [129] January 2002

Chez tous les patients opérés d’un cancer de l’estomac R0 avec un curage D1 ou D2 et de stade II ou III, la radiochimiothérapie adjuvante n’est pas considérée comme un traitement standard. En cas de curage < D1 (moins de 15 ganglions examinés) et chez des patients T3 et/ou N+, la radiochimiothérapie peut être proposée chez les patients non dénutris selon les modalités de l’essai de Macdonald (standard).

Bleiberg 1989 Dent 1979 Moertel 1984 McDonald 2001

High

En cas de chirurgie avec curage D1 ou D2 et avec un envahissement ganglionnaire majeur (N2 ou N3), une radiochimiothérapie adjuvante peut être proposée dans l’attente d’autres résultats, chez des patients non dénutris (option).

Il est recommandé de réaliser des essais thérapeutiques évaluant la place de la radiochimiothérapie adjuvante dans le traitement des cancers de l’estomac (accord d’experts). Le choix entre ces différentes options thérapeutiques doit prendre en compte l'avis ou le souhait des patients (recommandation).

Intraperitoneal chemotherapy

SIGN [47] 2004 Intraperitoneal chemotherapy and immunotherapy for patients with gastric cancer are not recommended outside a clinical trial.

Yonemura Y 2001 Moderate

Immunotherapy

SIGN [47] 2004 Intraperitoneal chemotherapy and immunotherapy for patients with gastric cancer are not recommended outside a clinical trial.

Earle 2003 High

CCO [150] April 2003

There is insufficient evidence from randomized trials to recommend adjuvant immunotherapy outside of a clinical trial.

Ochiai 1983 Yamamura 1986 Bonfanti 1988 Jakesz 1988 Kim 1992 Popeila 1982 Imaizumi 1990 Kim 1997

High

FNCLCC [129] January 2002

L’immunochimiothérapie adjuvante n’est pas un standard dans les cancers de l’estomac en occident (standard).

Jakesz 1988 IGTSG 1988 Primrose 1998 Tonnesen 1988 Langman 1999

High

Il est recommandé de poursuivre les essais thérapeutiques comparant l’immunochimiothérapie adjuvante à la chirurgie seule (recommandation).

Page 159: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 143

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Chemotherapy

Ajani J 2006 [269] October 2005

Patients with gastric cancer

Oral capecitabine Narrative presentation of results, no conclusions to be drawn.

Medline search only No information on selection, quality appraisal, etc

SR Low

Berrardi R 2004 [303] Not stated

Patients with gastric cancer

Adjuvant chemotherapy 3 RCTs in favour, 3 RCTs against. Narrative presentation of results.

No information on search at all.

SR? Low

Cascinu S 2007 [304] Gastric cancer patients at high risk for recurrence including patients with serosal invasion (stage pT3 N0) and/or lymph node metastasis (stage pT2 or pT3 N1, N2, or N3)

PELFw regimen (consisting of eight weekly administrations of cisplatin (40 mg/m2), LV (250 mg/m2), epidoxorubicin (35 mg/m2), 5-FU (500 mg/m2), and glutathione (1.5 g/m2) with the support of filgrastim) (n = 201) vs. 5-FU/LV regimen (consisting of six monthly administrations of a 5-day course of 5-FU (375 mg/m2 daily) and LV (20 mg/m2 daily, 5-FU/LV)) (n = 196)

5-year survival rates were 52% in the PELFw arm and 50% in the 5-FU/LV arm. Compared with the 5-FU/LV regimen, the PELFw regimen did not reduce the risk of death (HR = 0.95, 95% confidence interval [CI] = 0.70 to 1.29) or relapse (HR = 0.98, 95% CI = 0.75 to 1.29). Less than 10% of patients in either arm experienced a grade 3 or 4 toxic episode. Neutropenia (occurring more often in the PELFw arm) and diarrhea and mucositis (more prevalent in the 5-FU/LV arm) were the most common serious side effects. Nevertheless, only 19 patients (9.4%) completed the treatment in the PELFw arm and 85 (43%) patients completed the treatment in the 5-FU/LV arm.

No information on blinding

RCT Moderate

Nishikawa T 2007

[305] Excluded: re-analysis of previous RCT (2001)

RCT

Nitti D 2006 [172] NA Patients with adenocarcinoma of the stomach or esophago-gastric junction treated with curative resection

FAMTX or FEMTX vs. control In a planned combined analysis of the two trials, no significant differences were found between the treatment and control arms for either DFS (hazards ratio: 0.98, P=0.87) or OS (hazards ratio: 0.98, P=0.86). The 5-year OS was 43% in the treatment arm and 44% in the control arm and the 5-year DFS was 41% and 42%, respectively.

Prospective meta-analysis of 2 RCTs Blinding??

MA Moderate

Oba K 2006 [168] March 2005

Patients with gastric cancer

Adjuvant oral fluorinated pyrimidines vs. surgery alone

For the 4 trials that met the eligibility criteria, the estimated hazard ratio was 0.73 (95%CI=0.60-0.89) in favour of oral

Meta-analysis of 4 Japanese centrally randomized controlled

MA High

Page 160: Guideline pour la prise en charge du cancer oesophagien et ...

144 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

fluorinated pyrimidines. clinical trials conducted between 1980 and 2005

Tentes A 2006 [306] Patients with locally advanced gastric cancer after potentially curative gastrectomy

Adjuvant intraarterial chemotherapy (IARC) vs. surgery alone

Five-year survival rate for the study and the control group was 52 and 54%, respectively (p>0.05). Mean survival for the study and the control group was 50+/-8 and 62+/-10 months, respectively (p>0.05). The number of recurrences and the failure sites were comparable (p>0.05).

No information on randomization procedure or blinding

RCT Moderate

Ueda Y 2006 [307] Excluded: protocol Bouche O 2005 [169] Stage II-III-IVM0 gastric

cancer patients treated with curative resection (n = 260)

Postoperative chemotherapy (n = 127) vs. surgery alone (n = 133). 5-Fluorouracil (5-FU) 800 mg/m(2) daily (5-day continuous infusion) was initiated before day 14 after resection. One month later, four 5-day cycles of 5-FU (1 g/m² per day) plus cisplatin (100 mg/m(2) on day 2) were administered every 4 weeks.

5- and 7-year overall survival were 41.9% and 34.9% in the control group versus 46.6% and 44.6% in the chemotherapy group (P=0.22). Cox model hazard ratios were 0.74 [95%CI 0.54-1.02; P=0.063] for death and 0.70 (95%CI 0.51-0.97; P=0.032) for recurrence.

Blinding??? RCT Moderate

Tsuburaya A 2005

[308] Excluded: protocol

Chipponi J 2004 [170] Patients with gastric cancer and lymph node or serosal involvement or both

Surgery alone (n = 104) vs. surgery and adjuvant chemotherapy (daily administration of 200 mg/m² of folinic acid, 5-fluorouracil (375 mg/m² during the first session increasing 25 mg by session until reaching 500 mg/m²) and CDDP 15 mg/m² (n = 101)

Because of toxicity, 54% of the patients stopped the protocol before the end of the nine courses, and 46% of the patients received the nine courses including 32% with a decreased dose and 14% with a full dose. The 5-year survival rate was 39% in the control group and 39% in the chemotherapy group.

No information on blinding

RCT Moderate

Nashimoto A 2003

[171] Serosa-negative gastric cancer patients (excluding patients who were T1N0) (n = 252)

Adjuvant chemotherapy (intravenous mitomycin 1.33 mg/m², fluorouracil (FU) 166.7 mg/m², and cytarabine 13.3

There was no significant difference in relapse-free and overall survival between the arms (5-year relapse-free survival 88.8% chemotherapy v 83.7%

No information on blinding

RCT Moderate

Page 161: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 145

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

mg/m² twice weekly for the first 3 weeks after surgery, and oral FU 134 mg/m² daily for the next 18 months for a total dose of 67 g/m²) vs. surgery alone

surgery alone; P =.14 and 5-year survival 91.2% chemotherapy v 86.1% surgery alone; P =.13, respectively). Nine patients (7.1%) in the chemotherapy arm and 17 patients (13.8%) in the surgery-alone arm had cancer recurrence.

Radiotherapy

No additional studies found

Chemoradiotherapy

No additional studies found

Intraperitoneal chemotherapy

Yan TD 2007 [175] December 2006

Patients with locally advanced resectable gastric cancer

Adjuvant intraperitoneal chemotherapy

A significant improvement in survival was associated with hyperthermic intraoperative intraperitoneal chemotherapy (HIIC) alone (HR = 0.60; 95% CI = 0.43 to 0.83; p = 0.002) or combined with early postoperative intraperitoneal chemotherapy (EPIC) (HR = 0.45; 95% CI = 0.29 to 0.68; p = 0.0002). Survival improvement was marginally significant (p = 0.06) with normothermic intraoperative intraperitoneal chemotherapy, but not significant with EPIC alone or delayed postoperative intraperitoneal chemotherapy. Intraperitoneal chemotherapy was also found to be associated with higher risks for intra-abdominal abscess (RR = 2.37; 95% CI = 1.32 to 4.26; p = 0.003) and neutropenia (RR = 4.33; 95% CI = 1.49 to 12.61; p = 0.007).

13 RCTs of which 10 were of good quality Very good SR

SR High

Yu W 2006 [309] Excluded narrative review Xu DZ 2004 [174] January

2003 Patients undergoing curative resection for gastric cancer

Adjuvant intraperitoneal chemotherapy

Intraperitoneal chemotherapy may benefit the patients after curative resection for locally advanced gastric cancer, and the combination of intraperitoneal chemotherapy with

Also identified by Yan et al. Chinese and English only 11 RCTs identified, of which 2 were not

SR High

Page 162: Guideline pour la prise en charge du cancer oesophagien et ...

146 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

hyperthermia or activated carbon particles may provide more benefits to patients due to the enhanced antitumor activity of drugs. Sensitivity analysis and fail-safe number suggested that the result was comparatively reliable. However, of 11 trials, only 3 studies were of high quality.

identified by Yan et al. (Chen ZX 1996, Tan CQ 2000)

Immunotherapy

Chen DW 2005 [310] Patients with gastric carcinoma who had undergone major surgery

Immunonutrition (n = 20) vs. standard nutrition (n = 20)

No serious adverse effects were recorded with the two formulas. Postoperative procedures were smooth in both groups.

No full-text RCT

Oba K 2007 [176] Not stated

Patients with curative resections of gastric cancers

Immunochemotherapy with polysaccharide K (PSK)

The overall hazard ratio for eligible patients was 0.88 (95% confidence interval, 0.79-0.98; P = 0.018) with no significant heterogeneity [chi (2)(8) for heterogeneity = 11.7; P = 0.16]. The results of this meta-analysis suggest that adjuvant immunochemotherapy with PSK improves the survival of patients after curative gastric cancer resection.

8 RCTs included SR High

Farreras N 2005 [311] Patients with gastric cancer (n = 66)

Early postoperative enteral immunonutrition (formula supplemented with arginine, omega-3 fatty acids and ribonucleic acid (RNA)) vs. an isocaloric-isonitrogenous control

Patients fed with immunonutrition (n=30) showed significantly lower episodes of surgical wound healing complications (0 vs. 8 (26.7%) P=0.005) when compared to patients fed with the control formula (n=30).

No ITT anlysis (6 patients excluded, 3 in both arms)

RCT Moderate

Popiela T 2004 [177] Patients with stage III or IV gastric cancer who had undergone curative resection (n = 156)

BCG + FAM (immunochemotherapy) vs. FAM (chemotherapy) vs. control (surgery only)

Overall 10-year survival was 47.1% for the immunochemotherapy group (P < 0.037 vs FAM and P < 0.0006 vs control), 30% for the chemotherapy group (vs control, NS), and 15.2% for the control group. In patients with pT2/T3 primary tumors, 10-year survival was 55.3% for BCG + FAM vs 28.2% for FAM (P < 0.01) and 14.6% for the control group (P < 0.00018). BCG +

Randomization with envelope No information on blinding

RCT Moderate

Page 163: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 147

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

FAM significantly improved the survival of patients with intestinal-type but not diffuse-type cancer. Immunochemotherapy was well tolerated.

Sato Y 2004 [312] Patients who underwent curative resection of gastric cancer

800 mg/d 5'-DFUR (Furtulon) for 2 years from 2 weeks after the operation (n = 143) vs. OK-432 plus 5'-DFUR by the same regimen (n = 144)

The 5-year survival rates for groups A and B were 62.9% and 63.8%, respectively, showing no significant difference (P = 0.7996).

No full-text RCT

Palliative treatment.

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

Surgery

SIGN [47] 2004 Palliative gastrectomy should be avoided in patients with gastric cancer who have disseminated peritoneal disease.

Green D 2002 Low

D2 lymphadenectomy should be avoided in patients with gastric cancer in the palliative setting. Hanazaki K 1999 Low Health professionals should take the following factors into account when considering palliative gastric

resection: - peritoneal disease - tumour diameter - histological type - degree of differentiation

Green D 2002 Kunisaki C 2003

Low

Laparoscopic bypass or gastric outlet stenting are alternatives to palliative gastric bypass. Brune IB 1997 Low Palliative gastric bypass should be avoided when malignant ascites or small bowel obstruction are

present. Blair S 2001 Low

Exploratory laparotomy alone should be avoided. Ouchi K 1998 Low FNCLCC [129] January

2002 La chirurgie palliative est indiquée pour les sténoses symptomatiques, les tumeurs qui saignent et parfois en urgence pour perforation. Il n’y a pas d’indication à la chirurgie s’il existe une atteinte péritonéale, une ascite néoplasique ou des métastases hépatiques et si la tumeur est peu symptomatique (standard).

Low

L’exérèse ou la dérivation peuvent être proposes (option). Low L’indication de chirurgie palliative dépend de l’intensité des signes fonctionnels, de l’état général et

nutritionnel, de l’âge, des ressources thérapeutiques complémentaires utilisables et surtout de la résécabilité et d’une espérance de vie supérieure à 6 mois. L’exérèse est préférable à la derivation (recommendation).

Low

Chemotherapy

Page 164: Guideline pour la prise en charge du cancer oesophagien et ...

148 Upper GI Cancer KCE reports 75

CPG ID Ref Search date

Recommendation Supporting evidence Level of evidence

SIGN [47] 2004 In patients with locally advanced or metastatic cancer of the stomach with good performance status combination chemotherapy including cisplatin and infusional 5FU (such as ECF or MCF) should be considered.

Janunger K 2002 High

FNCLCC [129] January 2002

La chimiothérapie doit être proposée aux patients en bon état général dans le but d’améliorer leur survie (standard).

Murad 1993 Pyrhonen 1995 Glimelius 1997

High

Modalités (option): - monochimiothérapie par 5FU - acide folinique - polychimiothérapie par des combinaisons à base de 5FU et de cisplatine

Cullinan 1995 Kim 1993 Cullinan 1994 Many other RCTs

High

Il est recommandé de poursuivre les essais thérapeutiques pour préciser l’intérêt de nouveaux médicaments en première ligne et en seconde ligne. Le choix de la chimiothérapie doit être fait en fonction de l’état général et de l’âge. Une polychimiothérapie est recommandée pour les patients en bon état général. Le choix entre ces différentes options thérapeutiques doit prendre en compte l'avis ou le souhait des patients (recommendation).

No evidence provided

Supportive care

SIGN [47] 2004 Patients with gastric cancer should have access to a specialist palliative care team. Smeenk FW 1998 Miccinesi G 2003 Costantini M 2003 Costantini M 1993

Low

Page 165: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 149

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Chemotherapy

Ajani J 2006 [269] October 2005

Patients with gastric cancer

Oral capecitabine Narrative presentation of results, no conclusions to be drawn.

Medline search only No information on selection, quality appraisal, etc

SR Low

Trumper M 2006

[274] Excluded Retrospective subanalysis of 3 RCTs

Di Cosimo S 2003

[313] February 2003

Patients with advanced gastric cancer

Docetaxel Eight phase II trials focused on docetaxel as a single agent. Considering collectively the 262 evaluable patients enrolled in these studies, the mean response rate (RR) was 19% (CI 95% 14-24%). Docetaxel was well tolerated with a dose-limiting myelosuppression (grade 3-4 neutropenia in 36-95% of cases). Adding fluorouracil, an RR ranging from 22% to 86% was registered, due to differences in populations studied (young vs elderly) and modalities of drug administration (continuous vs. bolus infusion). RRs for docetaxel-cisplatin combination were 56%, 37% and 36% in three phase II trials and 35% in a phase III trial. The addition of both cisplatin and fluorouracil to docetaxel did not increase toxicity. Randomized trials comparing docetaxel-cisplatin-fluorouracil with ciplatin-fluorouoracil or epirubicin-cisplatin-fluorouracil, the most commonly used regimens, are ongoing.

No information on selection process or quality appraisal

SR Low

Wagner AD 2005 & 2006

[181] February 2005

Patients with advanced gastric cancer

Chemotherapy Analysis of chemotherapy versus best supportive care (HR = 0.39; 95% CI, 0.28 to 0.52) and combination versus single agent, mainly fluorouracil (FU) -based chemotherapy (HR = 0.83; 95%

27 RCTs included SR High

Page 166: Guideline pour la prise en charge du cancer oesophagien et ...

150 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

CI = 0.74 to 0.93) showed significant overall survival benefits in favor of chemotherapy and combination chemotherapy, respectively. In addition, comparisons of FU/cisplatin-containing regimens with versus without anthracyclines (HR = 0.77; 95% CI, 0.62 to 0.95) and FU/anthracycline-containing combinations with versus without cisplatin (HR = 0.83; 95% CI, 0.76 to 0.91) both demonstrated a significant survival benefit for the three-drug combination. Comparing irinotecan-containing versus nonirinotecan-containing combinations (mainly FU/cisplatin) resulted in a nonsignificant survival benefit in favor of the irinotecan-containing regimens (HR = 0.88; 95% CI, 0.73 to 1.06), but they have never been compared against a three-drug combination.

Wohrer SS 2004 [314] Patients with advanced gastric cancer

Chemotherapy Analysis of results shows chemotherapy to be superior to best supportive care alone. Combination chemotherapy compared with monochemotherapy is associated with significantly higher overall (complete plus partial) response rates but nevertheless results in similar survival. ECF (epirubicin, cisplatin and 5-fluorouracil) currently represents one of the most effective regimens for advanced gastric cancer, whereas among the newer combinations, irinotecan- or taxane-based regimens have also given promising results. In patients with a poor performance status, consideration could be given to leucovorin-modulated 5-fluorouracil alone.

Medline and English only No information on selection process or quality appraisal

SR Low

Ajani J 2005 [315] Untreated patients with confirmed advanced

DCF (docetaxel 75 mg/m2, cisplatin 75 mg/m² on day 1,

Of 158 randomly assigned patients, 155 (DCF, n = 79; DC, n = 76) received

RCT High

Page 167: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 151

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

gastric or gastroesophageal adenocarcinoma

and fluorouracil 750 mg/m²/d as continuous infusion on days 1 to 5) (n = 79) vs. DC (docetaxel 85 mg/m² and cisplatin 75 mg/m² on day 1) every 3 weeks (n = 76)

treatment. The confirmed ORR was 43% for DCF (n = 79) and 26% for DC (n = 76). Median time to progression was 5.9 months for DCF and 5.0 months for DC. Median overall survival time was 9.6 months for DCF and 10.5 months for DC. The most frequent grade 3 and 4 events per patient included neutropenia (DCF = 86%; DC = 87%) and GI (DCF = 56%; DC = 30%).

Bouche O 2004 [316] Patients with histologically proven metastatic gastric or cardial adenocarcinoma without linitis

LV 200 mg/m² (2-hour infusion) followed by FU 400 mg/m² (bolus) and FU 600 mg/m² (22-hour continuous infusion) on days 1 and 2 every 14 days (LV5FU2; arm A) (n = 45) vs. LV5FU2 plus cisplatin 50 mg/m² (1-hour infusion) on day 1 or 2 (arm B) (n = 44), vs. LV5FU2 plus irinotecan 180 mg/m² (2-hour infusion) on day 1 (arm C) (n = 45)

The overall response rates, which were confirmed by an independent expert panel, were 13% (95%CI, 3.4% to 23.3%), 27% (95%CI, 14.1% to 40.4%), and 40% (95%CI, 25.7% to 54.3%) for arms A, B, and C, respectively. Median progression-free survival and overall survival times were 3.2 months (95%CI, 1.8 to 4.6 months) and 6.8 months (95%CI, 2.6 to 11.1 months) with LV5FU2, respectively; 4.9 months (95%CI, 3.5 to 6.3 months) and 9.5 months (95%CI, 6.9 to 12.2 months) with LV5FU2-cisplatin, respectively; and 6.9 months (95%CI, 5.5 to 8.3 months) and 11.3 months (95%CI, 9.3 to 13.3 months) with LV5FU2-irinotecan, respectively.

No information on exact randomization procedure or blinding Included in Wagner 2006

RCT Moderate

Cocconi G 2003 [317] Patients with untreated advanced gastric carcinoma

5-Fluorouracil (5-FU), doxorubicin and methotrexate (FAMTX) (n = 97) vs. cisplatin, epirubicin, leucovorin and 5-FU (PELF) (n = 98)

The complete response (CR) rates to PELF and FAMTX were, respectively, 13% [95% confidence intervals (CI) 6% to 20%] and 2% (95% CI 0% to 5%; P = 0.003), and the objective response rates [CR plus partial response (PR) rates] 39% (95% CI 29% to 49%) and 22% (95% CI 13% to 30%; P = 0.009), thus significantly favouring the PELF combination. The survival rates after 12 months (30.8% versus 22.4%) and 24 months (15.7% versus 9.5%) were also

No information on blinding Included in Wagner 2006

RCT Moderate

Page 168: Guideline pour la prise en charge du cancer oesophagien et ...

152 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

higher among patients receiving PELF, but these differences were not statistically significant. The toxicities were qualitatively different but quantitatively similar. Both regimens seem to be feasible provided that careful patient monitoring is assured.

Koizumi W 2004

[318] Patients with advanced unresectable gastric cancer

Cisplatin + mitomycin + oral 5’-DFUR (n = 32) vs. Cisplatin + oral 5’-DFUR (n = 29)

No significant differences in response rate and median survival time.

No information on exact randomization procedure Blinding??

RCT Moderate

Lutz M 2007 [319] Patients with advanced gastric cancer

Weekly FU 3000 mg/m²/24 hours (HD-FU) (n = 37), FU 2600 mg/m²/24 hours plus dl-FA 500 mg/m² or l-FA 250 mg/m² (HD-FU/FA) (n = 53), vs. FU 2000 mg/m²/24 hours plus FA plus biweekly Cis 50 mg/m² (n = 51), each administered for 6 weeks with a 1-week rest

Confirmed responses were observed in 6.1% (two of 33) of the eligible patients treated with HD-FU, in 25% (12 of 48, including one complete remission [CR]) with HD-FU/FA, and in 45.7% (21 of 46, including four CRs) with HD-FU/FA/Cis. The HD-FU arm was closed after stage 1 because the required minimum number of responses was not met. The median progression-free survival of all patients in the HD-FU, HD-FU/FA, and HD-FU/FA/Cis arm was 1.9, 4.0, and 6.1 months, respectively. The median overall survival was 7.1, 8.9, and 9.7 months, and the survival rate at 1 year was 24.3%, 30.3%, and 45.3%, respectively. Grade 4 toxicities were rare. The most relevant grade 3/4 toxicities were neutropenia in 1.9%, 5.4%, and 19.6%, and diarrhea in 2.7%, 1.9%, and 3.9% of the cycles in the HD-FU, HD-FU/FA, and HD-/FU/Cis arms, respectively.

No information on exact randomization procedure

RCT Moderate

Moehler M 2005 [320] Patients with untreated metastatic or locally advanced gastric cancer

Irinotecan plus high-dose 5-fluorouracil (5-FU) and leucovorin (LV) (ILF) (n = 56) vs. etoposide plus 5-FU/LV (ELF) (n = 58)

The objective clinical response rates after 14 weeks treatment were 30% for ILF and 17% for ELF (RR 0.57, 95%CI 0.29–1.13, P=0.077). Overall response rates over the entire treatment period for ILF and ELF were 43 and 24%, respectively (RR 0.56, 95%CI 0.33–0.97;

No information on exact randomization procedure Blinding??? Included in Wagner 2006

RCT Moderate

Page 169: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 153

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

P=0.047). For ILF and ELF, respectively, median progression-free survival was 4.5 vs 2.3 months, time to treatment failure was 3.6 vs 2.2 months (P=0.45), and overall survival was 10.8 vs 8.3 months (P=0.28). Both regimens were well tolerated, the main grade 3/4 toxicities being diarrhoea (18%, ILF) and neutropenia (57%, ELF).

Ohtsu A 2003 [321] Patients with advanced gastric cancer

Fluorouracil (FU) alone vs. FU plus cisplatin (FP) vs. uracil and tegafur plus mitomycin (UFTM)

At the interim analysis, the UFTM arm showed a significantly inferior survival with higher incidences of hematologic toxic effects than did control arm FU alone, and the registration to UFTM was terminated. Both investigational regimens, FP and UFTM, had a significantly higher incidence of hematologic toxic effects than FU alone, although the effects were manageable. The overall response rates of the FU-alone, FP, and UFTM arms were 11%, 34%, and 9%, respectively. The median progression-free survival was 1.9 months with FU alone, 3.9 months with FP, and 2.4 months with UFTM, respectively. Although FP demonstrated a higher response rate (P < .001) and longer progression free survival than did FU alone (P < .001), no differences in overall survival were observed between the arms. The median survival times and 1-year survival rates were 7.1 months and 28% with FU, 7.3 months and 29% with FP, and 6.0 months and 16% with UFTM, respectively.

No information on blinding Included in Wagner 2006

RCT Moderate

Park SH 2006 [322] Patients with measurable metastatic gastric cancer

Paclitaxel + 5FU (n = 38) vs. docetaxel + 5FU (n = 38)

No significant differences No information on randomization procedure

RCT Moderate

Pozzo C 2004 [323] Patients with advanced gastric or oesophageal-gastric junction

Irinotecan [80 mg/m2² IV], FA (500 mg/m² IV) and a 22-h infusion of 5-FU (2000 mg/m²

The overall response rate in the irinotecan/5-FU/FA arm was 42.4%, with a complete response rate of 5.1%.

No ITT RCT Moderate

Page 170: Guideline pour la prise en charge du cancer oesophagien et ...

154 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

adenocarcinoma IV), weekly for 6 weeks with a 1-week rest (n = 59), vs. irinotecan (200 mg/m² IV) and cisplatin (60 mg/m² IV), on day 1 for 3 weeks (n = 58)

Corresponding figures for the irinotecan/cisplatin arm were 32.1% and 1.8%, respectively. The median time to progression was 6.5 months (irinotecan/5-FU/FA) and 4.2 months (irinotecan/ cisplatin) (P < 0.0001), with median survival times of 10.7 and 6.9 months, respectively (P = 0.0018). The major toxicity was grade 3/4 neutropenia, which was more pronounced with irinotecan/cisplatin than with irinotecan/5-FU/FA (65.7% versus 27%). Diarrhea was the main grade 3/4 non-hematological toxicity with both irinotecan/5-FU/FA (27.0%) and irinotecan/cisplatin (18.1%).

Sadighi S 2006 [324] Patients with advanced gastric cancer

Docetaxel, cisplatin, 5-FU (TCF) (n = 44) vs. epirubicin, cisplatin, 5-FU (ECF) (n = 42)

Only the TCF group showed statistically and clinically meaningful improvement in global QOL (P = 0.001). Surgical and pathologic response were better with TCF, but there was no difference in survival rate between two groups.

No information on randomization procedure or blinding

RCT Moderate

Sumpter K 2005 [270] Patients with inoperable, histologically verified locally advanced or metastatic adenocarcinoma, squamous cell or undifferentiated carcinoma of the oespohagus, oesophagogastric junction (OGJ) or stomach

ECF (n = 53) ECX (n = 48) EOF (n = 55) EOX (n = 48) (E = epirubucin, C = cisplatin, F = 5FU, O = oxaliplatin, X = capecitabin)

First interim analysis was performed when 80 pts had been randomised. Doselimiting fluoropyrimidine toxicities were stomatitis, palmar plantar erythema (PPE) and diarrhoea; 5.1% of X-treated pts experienced grade 3/4 toxicity. Protocol planned dose escalation of X to 625 mg/m² b.i.d. was instituted and a second interim analysis has been performed. A total of 204 pts were randomised at the time of the protocol planned 2nd interim analysis. Grade 3/4 fluoropyrimidine-related toxicity was seen in 13.7% pts receiving F, 8.4% pts receiving X 500 mg/m² b.i.d. and 14.7% pts receiving X 625 mg/m² b.i.d. Combined complete and partial

Interim analysis 6 originally randomized pts not included in results Noninferiority study No information on blinding of randomization

RCT Moderate

Page 171: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 155

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

response rates were ECF 31% (95% CI 18.7–46.3), EOF 39% (95% CI 25.9–53.1), ECX 35% (95% CI 21.4–50.3), EOX 48% (95% CI 33.3–62.8). Grade 3/4 fluoropyrimidine toxicity affected 14.7% of pts treated with X 625 mgm�2 b.i.d.�1, which is similar to that observed with F, confirming this to be the optimal dose. The replacement of C by O and F by X does not appear to impair efficacy. The trial continues to total accrual of 1000 pts.

Takahashi Y 2004

[325] Patients with advanced or recurrent gastric carcinoma

Tailored CPT-11 + S-1 vs S-1 Excluded: protocol RCT

Thuss-Patience P 2005

[326] Patients with metastatic or locally advanced gastric adenocarcinoma without prior chemotherapy (n = 94)

ECF (epirubicin 50 mg/m² day 1, cisplatin 60 mg/m² day 1, and fluorouracil 200 mg/m² days 1 through 21, every 3 weeks) (n = 45) or DF (docetaxel 75 mg/m² day 1, and fluorouracil 200 mg/m² days 1 through 21, every 3 weeks) (n = 45)

In the DF arm, two patients (4.4%, intent to treat) experienced a confirmed complete tumor remission as best response, and 15 patients (33.3%) experienced a confirmed partial remission (overall response rate [ORR], 37.8%; 95% CI, 25.9% to 51.9%). Two patients (4.4%) in the ECF arm showed confirmed complete remission, and 14 (31.1%) showed confirmed partial remission (ORR, 35.6%; 95% CI, 24.8% to 48.7%). For the DF and ECF arms, the median survival was 9.5 and 9.7 months, and the median time to tumor progression 5.5 and 5.3 months, respectively.

No information on randomization procedure Four patients excluded because of violation of inclusion criteria

RCT Moderate

Tsuburaya A 2005

[308] Excluded: protocol RCT

Berardi R 2004 [303] Not stated

Patients with gastric cancer

No information on search at all: excluded

SR? Low

Casaretto L 2006

[180] Not stated

Patients diagnosed with gastric adenocarcinoma

Chemotherapy versus support cancer treatment

The 1-year survival rate was 8% for support care and 20% for chemotherapy (RR = 2.14, 95%CI = 1.00-4.57, P = 0.05); 30% of the patients in the chemotherapy group and 12% in the

5 RCTs included SR High

Page 172: Guideline pour la prise en charge du cancer oesophagien et ...

156 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

support care group attained a 6-month symptom-free period (RR = 2.33, 95%CI = 1.41-3.87, P < 0.01). Quality of life evaluated after 4 months was significantly better for the chemotherapy patients (34%; RR = 2.07, 95%CI = 1.31-3.28, P < 0.01) with tumor mass reduction (RR = 3.32, 95%CI = 0.77-14.24, P = 0.1).

Bonnetain F 2005

[327] Patients with metastatic gastric adenocarcinoma

LV5FU2-irinotecan vs. LV5FU2 alone vs. LV5FU2-cisplatin

Patients with a stable or improved global health ranged from 11% in the LV5FU2-cisplatin arm to 18% in the LV5FU2-irinotecan arm. The irinotecan-based-therapy presented 14 to 15 scores with a better QoL.

No information on randomization procedure or blinding

RCT Moderate

Van Cutsem E 2006

[328] Patients with advanced gastric cancer

Docetaxel 75 mg/m² and cisplatin 75 mg/m² (day 1) plus fluorouracil 750 mg/m²/d (days 1 to 5) every 3 weeks (n = 221) vs. cisplatin 100 mg/m² (day 1) plus fluorouracil 1,000 mg/m²/d (days 1 to 5) every 4 weeks (n = 224)

TTP was longer with DCFvs. CF (32% risk reduction; log-rank P < .001). Overall survival was longer with DCF versus CF (23% risk reduction; log-rank P = .02). Two-year survival rate was 18% with DCF and 9% with CF. Overall response rate was higher with DCF (P = .01). Grade 3 to 4 treatment-related adverse events occurred in 69% (DCF) v 59% (CF) of patients. Frequent grade 3 to 4 toxicities for DCF vs. CF were: neutropenia (82% vs. 57%), stomatitis (21% vs. 27%), diarrhea (19% vs. 8%), lethargy (19% vs. 14%). Complicated neutropenia was more frequent with DCF than CF (29% vs. 12%).

No information on exact randomization procedure

RCT Moderate

Stenting

Dormann A 2004

[329] September 2003

Patients with malignant symptomatic gastroduodenal obstruction

Self-expanding metal stents Clinical success was achieved in 526 patients in the group in which technical success was reported (89 %; 87 % of the entire group undergoing stenting). Disease-related factors accounted for the majority of clinical failures. Oral

32 case series included (only 10 prospective) No information on quality appraisal or selection process (two reviewers?)

SR Very low

Page 173: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 157

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

intake became possible in all of the patients in whom a successful procedure was carried out, with 87 % taking soft solids or a full diet, with final resolution of symptoms occurring after a mean of 4 days. There was no procedure-related mortality. Severe complications (bleeding and perforation) were observed in seven patients (1.2 %). Stent migration was reported in 31 patients (5 %). Stent obstruction occurred in 104 cases (18 %), mainly due to tumor infiltration. The mean survival period was 12.1 weeks.

Hosono S 2007 [178] September 2006

Patients with malignant gastroduodenal obstruction

Endoscopic stenting vs. surgical gastroenterostomy

Endoscopic stenting was found to be associated with higher clinical success (P = 0.007), a shorter time from the procedure to starting oral intake (P < 0.001), less morbidity (P = 0.02), lower incidence of delayed gastric emptying (P = 0.002), and a shorter hospital stay (P < 0.001) than surgical gastro-enterostomy. There was no significant difference between the two groups in the analysis of 30-day mortality.

English only 1 RCT and 8 non-randomized retrospective studies included

SR Low

Jeurnink S 2007 [179] December 2005

Patients with gastric outlet obstruction

Stent vs. gastrojejunostomy No differences between stent placement and gastrojejunostomy were found in technical success (96% vs. 100%), early and late major complications 7% vs. 6% and 18% vs. 17%, respectively) and persisting symptoms (8% vs. 9%). Initial clinical success was higher after stent placement (89% vs. 72%). Minor complications were less frequently seen after stent placement in the patient series (9% vs. 33%), however the pooled analysis showed no differences (OR: 0.75, p = 0.8). Recurrent obstructive symptoms were more common after stent placement (18% vs. 1%). Hospital

Medline only No information on study selection Inclusion of 44 studies (2 RCTs)

SR Low

Page 174: Guideline pour la prise en charge du cancer oesophagien et ...

158 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

stay was prolonged after GJJ compared to stent placement (13 days vs. 7 days). The mean survival was 105 days after stent placement and 164 days after GJJ.

Fiori E 2004 [330] Included in Jeurnink S RCT Mehta S 2006 [331] Included in Jeurnink S RCT Wenger U 2006 [271] Patients with incurable

cancer of the distal oesophagus or gastric cardia (n = 41)

Antireflux stent (n = 19) vs. standard stent (n = 22)

Fewer patients with complications were observed in the antireflux stent group (n = 3) than in the standard group (n = 8), but no statistically significant difference was shown (p = 0.14). The survival rates were similar in the two groups (p = 0.99; hazard ratio, 1.0; 95% confidence interval, 0.5-2.0). The groups did not differ significantly in terms of studied oesophageal or respiratory symptoms or quality of life. Clinically relevant improvement in dysphagia occurred in both groups. Dyspnea decreased after antireflux stent insertion (mean score change, -11), and increased after insertion of standard stent (mean score change, +21).

Interim report Central randomization, but no information on procedure Patients were blinded, but no information on blinding of clinicians

RCT Moderate

Follow up.

No evidence identified.

Page 175: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 159

APPENDIX 6

EVIDENCE TABLES OF GASTRIC LYMPHOMA BY CLINICAL QUESTION Diagnosis and staging of gastric lymphoma.

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Molecular diagnosis Weston 1998 [332] Mix of 50 patients of which

15 had gastric lymphoma PCR monoclonality Standard: histology (endoscopy)

Sensitivity 73%, specificity 46% However, correct interpretation of these values is impossible

Selection of patients?? Prospective study

Very low

Endoscopy Palazzo 1993 [333] Patients with primary

gastric lymphoma (n = 50) EUS vs. endoscopy Standard: histology (surgery) (n = 24)

Endoscopy correctly assessed tumour extent in 25% of cases

Decision for surgery not based on EUS Selection of patients?? Blinding?

Prospective study

Very low

Ultrasonography Hoepffner 2003 [203] Patients with gastric NHL

(n = 44) EUS (n = 44) vs. CT (n = 24) vs. US (n = 34) Standard: histology (surgery)

Sensitivity for diagnosis: 27% (specificity unknown)

CT not performed in all patients Selection of patients?? Blinding?

Prospective study

Very low

CT scan Ferreri 1998 [334] Patients with gastric MALT

lymphoma (n = 20) Low-grade: n = 11; high-grade: n = 9

CT for detection of perigastric adenopathy Standard: histology (surgery)

Sensitivity to distinguish II1 from I: 27% Specificity 100%

No information on how patients were selected Small study sample Blinding?

Retrospective (?) study

Very low

Vorbeck 2002 [202] Patients with gastric MALT lymphoma (n = 14)

EUS vs. CT Standard: histology (endoscopy)

Sensitivity for diagnosis: 57% (specificity unknown)

Selection of patients?? Small study sample

Prospective study

Very low

Hoepffner 2003 [203] Patients with gastric NHL (n = 44)

EUS (n = 44) vs. CT (n = 24) vs. US (n = 34) Standard: histology (surgery)

Sensitivity for diagnosis: 42% (specificity unknown)

CT not performed in all patients Selection of patients?? Blinding?

Prospective study

Very low

Endoscopic ultrasonography Caletti 1993 [199] Patients with endoscopic

gross appearance suspicious for gastric cancer of lymphoma

EUS Standard: histology (surgery) (n = 86)

42 gastric cancers, 44 primary lymphomas, 142 patients with benign gastric lesions or suspected pancreatic diseases. Sensitivity (lymphoma): 89% Specificity: 97% PPV: 87%; NPV: 97%

No information on blinding during surgery Standard not applied to all patients, no information on follow-up of 142 patients without gastric malignancy

Prospective study

Low

Page 176: Guideline pour la prise en charge du cancer oesophagien et ...

160 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Palazzo 1993 [333] Patients with primary gastric lymphoma (n = 50)

EUS vs. endoscopy Standard: histology (surgery) (n = 24)

Diagnosis of LN metastasis: sensitivity 100%, specificity 80%

Decision for surgery not based on EUS Selection of patients?? Blinding?

Prospective study

Very low

Fischbach 2002 [200] Patients with primary gastric B-cell lymphoma stage I and II (n = 80)

EUS Standard: histology (surgery)

Sensitivity to distinguish II1 from I: 59% Specificity 71%

10 patients excluded from statistical analysis, because EUS was considered inappropriate for evaluation of non-regional lymph nodes

Prospective study

Low

Fusaroli 2002 [335] Patients with gastric MALT lymphoma (n = 54; n = 42, six months after treatment)

EUS Blinded assessment of photographs by 9 other endosonographers

Inter-observer agreement T-stage: kappa 0.38 before and 0.37 after treatment Inter-observer agreement N-stage: kappa 0.63 before and 0.34 after treatment

Prospective study

Very low

Vorbeck 2002 [202] Patients with gastric MALT lymphoma (n = 14)

EUS vs. CT Standard: histology (endoscopy)

Sensitivity for diagnosis: 93% (specificity unknown)

Selection of patients?? Small study sample

Prospective study

Very low

Hoepffner 2003 [203] Patients with gastric NHL (n = 44)

EUS (n = 44) vs. CT (n = 24) vs. US (n = 34) Standard: histology (surgery)

Sensitivity for diagnosis: 95% (specificity unknown)

CT not performed in all patients Selection of patients?? Blinding?

Prospective study

Very low

PET scan Rodriguez 1997 [201] Mix of 15 patients, of which

8 had primary gastric NHL and 7 others served as control

PET vs. CT or MRI Standard: endoscopic biopsies

Sensitivity 88% Patient selection?? Comparator not applied to all patients

Prospective study

Very low

Gallium 67 imaging Kataoka 1990 [336] Patients with NHL of the GI

tract (n = 24, of which 20 had gastric NHL), selected out of 189 consecutive patients

Gallium 67 (n = 24) vs. barium study (n = 24) vs. CT (n = 16)

Sensitivity for diagnosis: 95% (specificity unknown)

Not only patients with gastric NHL Blinding?

Retrospective study

Very low

Page 177: Guideline pour la prise en charge du cancer oesophagien et ...

KCE Reports 75 Upper GI Cancer 161

Treatment of low-grade (Ie and IIE) MALT lymphoma

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Helicobacter eradication Montalban 2001 [208] Patients with stage I low-

grade gastric MALT lymphoma

Eradication treatment 91% achieved disappearance of the lymphoma

Prospective study

Low

Fischbach 2004 [207] Patients with low grade gastric MALT lymphoma

Exclusive H pylori eradication

Complete regression of lymphoma in 56 patients (62%), minimal residual disease in 17 patients (18%), partial remission in 11 patients (12%), no change in four patients (4%), and progressive disease in two patients (2%)

Prospective study

Low

Chemotherapy Aviles 2005 [209] 241 patients with low-grade

MALT lymphoma in early stage (IE and IIE)

surgery (80 cases), radiotherapy (78 cases), and chemotherapy (83 cases) (CHOP)

Event-free survival at follow-up (median follow-up was 7.5 y) : Surgery : 52 % Radiotherapy : 52% Chemotherapy : 87% ( p<0.01) Overall survival: Surgery : 80 % Radiotherapy : 75% Chemotherapy : 87% ( p= 0.4)

RCT High

Treatment of high-grade gastric lymphoma IE and IIE

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

Haim N 1995 [337] 26 patients , 15 stage I, 7 stage II, 4 stage IV

Chemotherapy : CHOP : 18 patients, Pro MACE/MOPP: 8 patients

75% endoscopically-confirmed response follow up : 22 months : no recurrence

Data support non-surgical approach

Prospective study

Low

Takaneka T 1997 [338] 25 patients suffering from primary gastric lymphomas (stage II)

21 patients treated by total gastrectomy 4 patients treated by partial gastrectomy all received chemotherapy

Disease free survival at 10 years : 81.6 and 92% respectively

Prospective study

Low

Kochi M 2007 [339] 10 patients suffering from gastric lymphoma

10 patients treated by preoperative chemotherapy and extensive surgery

Disease free survival at 86 months (range 40 to 102 m) : 100 %

After chemotherapy, microscopic evaluation did not reveal residual lymphoma cells

Prospective study

Low

Willich NA 2000 [340] 190 patients suffering from gastric lymphoma

58 patients underwent surgery 83 patients underwent chemotherapy (CHOP)

Overall survival at follow-up (max 68 months (median : 29) : was 88% (surgery) and 86%

Prospective comparative study

Low

Page 178: Guideline pour la prise en charge du cancer oesophagien et ...

162 Upper GI Cancer KCE reports 75

Study ID Ref Search date

Population Intervention Results Comments Study type Level of evidence

(chemotherapy) after 5 years for all stages and histologies.

Chen LT 2001 [341] patients suffering from high-grade gastric lymphomas (DLBCL)

16 patients received a brief antibiotic therapy as first-line treatment

Eradication of H pylori: 15 patients Rapid gross tumor regression: 10 patients Complete remission rate: 62.5% (CI 35.8% - 89.1%) Median duration of complete response was 31.2 months (range, 14.4 to 49.1 months)

Prospective study

Low

Binn M 2003 [193] patients suffering from localized gastric lymphomas (DLBCL)

58 patients underwent chemotherapy 48 patients underwent surgery +chemotherapy

Overall survival at follow-up (59 months (range 3–128) : Best prognosis : 90.5%(chemotherapy alone) versus 91.1% (surgery +chemotherapy) (P = 0.303) Mild Prognosis : 85.9%(chemotherapy alone) versus 91.6% (surgery +chemotherapy) (P = 0.187)

a similar 5-year survival rate (>90%) is to be expected with either surgery plus chemotherapy or chemotherapy alone

Prospective comparative study

Low

Koch P 2005 [342] 393 patients with aggressive localized PGL were treated between December 1996 and December 2003

332 patients underwent “conservative treatment” (radiotherapy +/-chemotherapy) 61 patients received surgery and conservative treatment.

Overall survival at 42 months : For surgery was 86% versus 91.0% for patients without surgery

Treatment decision was left to each participating centre

Prospective study

Low

Ishikura S 2005 [343] 52 patients with aggressive localized PGL were treated between December 1999 and December 2003

Chemotherapy (CHOP) and radiotherapy (40.5Gy)

Complete remission rate: 48 on 52 (CI 82-98%%) Overall survival at follow-up (median follow-up was 2y) : 94%

Prospective study

Low

Aviles A 2006 [344] 101 patients with high grade lymphoma were treated between December 1999 and December 2003

52 patients underwent surgery +chemotherapy (CEOP-Bleo) 49 patients underwent chemotherapy alone Total : 101

Overall survival at follow-up (median follow-up was 5 y) : Surgery + chemotherapy: 78% (CI 70-88%) Chemotherapy: 76% (CI 70-87%) P=0.8

RCT High

Page 179: Guideline pour la prise en charge du cancer oesophagien et ...

KCE reports 75 Upper GI Cancer 163

APPENDIX 7 TNM CLASSIFICATION

(source: Digestive System Tumours. In: Sobin LH, Witteking Ch, editors. TNM Classification of Malignant Tumours. 6th ed. New York: Wiley-Liss; 2002. p. 57-96.)

Oesophageal cancer

PRIMARY TUMOUR (T)

TX Primary tumour cannot be assessed

T0 No evidence of primary tumour

Tis Carcinoma in situ

T1 Tumour invades lamina propria or submucosa

T2 Tumour invades muscularis propria

T3 Tumour invades adventitia

T4 Tumour invades adjacent structures

REGIONAL LYMPH NODES (N)

NX Regional lymph nodes cannot be assessed

N0 No regional lymph node metastasis

N1 Regional lymph node metastasis

DISTANT METASTASIS (M)

MX Distant metastasis cannot be assessed

M0 No distant metastasis

M1 Distant metastasis

Tumours of the lower thoracic oesophagus:

M1a Metastasis in celiac lymph nodes

M1b Other distant metastasis

Tumours of the midthoracic oesophagus:

M1a Not applicable

M1b Nonregional lymph nodes and/or other distant metastasis

Tumours of the upper thoracic oesophagus:

M1a Metastasis in cervical nodes

M1b Other distant metastasis

STAGE GROUPING Stage 0 Tis N0 M0 Stage I T1 N0 M0 Stage IIA T2 N0 M0 T3 N0 M0 Stage IIB T1 N1 M0 T2 N1 M0 Stage III T3 N1 M0 T4 Any N M0 Stage IV Any T Any N M1 Stage IVA Any T Any N M1a Stage IVB Any T Any N M1b

Page 180: Guideline pour la prise en charge du cancer oesophagien et ...

164 Upper GI Cancer KCE reports 75

Gastric cancer

PRIMARY TUMOUR (T)

TX Primary tumour cannot be assessed

T0 No evidence of primary tumour

Tis Carcinoma in situ : intraepithelial tumour without invasion of the lamina propria

T1 Tumour invades lamina propria or submucosa

T2 Tumour invades muscularis propria or subserosa

T2a Tumour invades muscularis propria

T2b Tumour invades subserosa

T3 Tumour penetrates serosa (visceral peritoneum) without invasion of adjacent structures

T4 Tumour invades adjacent structures

REGIONAL LYMPH NODES (N)

NX Regional lymph nodes cannot be assessed

N0 No regional lymph node metastasis

N1 Metastasis in 1 to 6 regional lymph nodes

N2 Metastasis in 7 to 15 regional lymph nodes

N3 Metastasis in more than 15 regional lymph nodes

DISTANT METASTASIS (M)

MX Distant metastasis cannot be assessed

M0 No distant metastasis

M1 Distant metastasis

STAGE GROUPING Stage 0 Tis N0 M0 Stage IA T1 N0 M0 Stage IB T1 N1 M0 T2a/b N0 M0 Stage II T1 N2 M0 T2a/b N1 M0 T3 N0 M0 Stage IIIA T2a/b N2 M0 T3 N1 M0 T4 N0 M0 Stage IIIB T3 N2 M0 Stage IV T4 N1-3 M0 T1-3 N3 M0 Any T Any N M1

Page 181: Guideline pour la prise en charge du cancer oesophagien et ...

KCE reports 75 Upper GI Cancer 165

APPENDIX 8

USED MESH AND SEARCH TERMS FOR ADDITIONAL EVIDENCE SEARCH IN MEDLINE (OVID)

Oeophageal cancer: 1 exp esophageal neoplasms/ 2 (esophag$ adj5 neoplas$).tw. 3 (oesophag$ adj5 neoplas$).tw. 4 (esophag$ adj5 cancer$).tw. 5 (oesophag$ adj5 cancer$).tw. 6 (esophag$ adj5 carcin$).tw. 7 (oesophag$ adj5 carcin$).tw. 8 (esophag$ adj5 tumo$).tw. 9 (oesophag$ adj5 tumo$).tw. 10 (esophag$ adj5 metasta$).tw. 11 (oesophag$ adj5 metasta$).tw. 12 (esophag$ adj5 malig$).tw. 13 (oesophag$ adj5 malig$).tw. 14 or/1-13

Gastric cancer:

1 exp stomach neoplasms/ 2 (stomach adj5 neoplas$).tw. 3 (stomach adj5 cancer$).tw. 4 (stomach adj5 carcin$).tw. 5 (stomach adj5 tumo$).tw. 6 (stomach adj5 metasta$).tw. 7 (stomach adj5 malig$).tw. 8 (gastric adj5 neoplas$).tw. 9 (gastric adj5 cancer$).tw. 10 (gastric adj5 carcin$).tw. 11 (gastric adj5 tumo$).tw. 12 (gastric adj5 metasta$).tw. 13 (gastric adj5 malig$).tw. 14 or/1-13

Page 182: Guideline pour la prise en charge du cancer oesophagien et ...

166 Upper GI Cancer KCE reports 75

Systematic reviews / meta-analyses: 1 meta-analysis.pt,ti,ab,sh. 2 1 or (meta anal$ or metaanal$).ti,ab,sh. 3 (methodol$ or systematic$ or quantitativ$).ti,ab,sh.

4 ((methodol$ or systematic$ or quantitativ$) adj (review$ or overview$ or survey$)).ti,ab,sh.

5 (medline or embase or index medicus).ti,ab.

6 ((pool$ or combined or combining) adj (data or trials or studies or results)).ti,ab.

7 3 or 4 or 5 or 6 8 7 and review.pt,sh. 9 2 or 8

Randomized controlled trials:

1 Randomized controlled trials/ 2 Randomized controlled trial.pt. 3 Random allocation/ 4 Double blind method/ 5 Single blind method/ 6 Clinical trial.pt. 7 exp clinical trials/ 8 or/1-7 9 (clinic$ adj trial$1).tw. 10 ((singl$ or doubl$ or treb$ or tripl$) adj (blind$3 or mask$3)).tw. 11 Placebos/ 12 Placebo$.tw. 13 Randomly allocated.tw. 14 (allocated adj2 random).tw. 15 or/9-14 16 8 or 15 17 Case report.tw. 18 Letter.pt. 19 Historical article.pt. 20 Review of reported cases.pt. 21 Review, multicase.pt. 22 or/17-21 23 16 not 22

Page 183: Guideline pour la prise en charge du cancer oesophagien et ...

KCE reports 75 Upper GI Cancer 167

Chemotherapy: 1 exp drug therapy/ 2 exp chemotherapy adjuvant/ 3 exp drug therapy combination/ 4 exp antineoplastic agents combined/ 5 chemothera$.tw. 6 exp chemotherapy/ 7 or/1-6

Endoscopy: 1 endoscopy/ or endoscopy, digestive system/ or endoscopy, gastrointestinal/ 2 Gastroscopy/ 3 Esophagoscopy/ 4 or/1-3

Staging (e.g. gastric cancer): 1 exp stomach neoplasms/ 2 (stomach adj5 neoplas$).tw. 3 (stomach adj5 cancer$).tw. 4 (stomach adj5 carcin$).tw. 5 (stomach adj5 tumo$).tw. 6 (stomach adj5 metasta$).tw. 7 (stomach adj5 malig$).tw. 8 (gastric adj5 neoplas$).tw. 9 (gastric adj5 cancer$).tw. 10 (gastric adj5 carcin$).tw. 11 (gastric adj5 tumo$).tw. 12 (gastric adj5 metasta$).tw. 13 (gastric adj5 malig$).tw. 14 or/1-13

15 tomography scanners, x-ray computed/ or tomography, x-ray computed/ or magnetic resonance imaging/ or positron-emission tomography/ or tomography, spiral computed/ or tomography/

16 Endosonography/ 17 Laparoscopy/ 18 Bronchoscopy/ 19 Thoracoscopy/ 20 Ultrasonography/ 21 15 or 16 or 17 or 18 or 19 or 20 22 Neoplasm Staging/ 23 "Tumor Markers, Biological"/ 24 "Immunoenzyme Techniques"/ 25 23 or 24 26 Stomach Neoplasms/an, pa, di, ra, ri, us [Analysis, Pathology, Diagnosis,

Page 184: Guideline pour la prise en charge du cancer oesophagien et ...

168 Upper GI Cancer KCE reports 75

Radiography, Radionuclide Imaging, Ultrasonography] 27 21 or 22 or 25 28 14 and 27 29 26 or 28 30 Prospective Studies/ 31 29 and 30

Recurrence (e.g. oesophageal cancer): 1 Salvage Therapy/ 2 neoplasm recurrence, local/ or recurrence/ 3 exp esophageal neoplasms/ 4 (esophag$ adj5 neoplas$).tw. 5 (oesophag$ adj5 neoplas$).tw. 6 (esophag$ adj5 cancer$).tw. 7 (oesophag$ adj5 cancer$).tw. 8 (esophag$ adj5 carcin$).tw. 9 (oesophag$ adj5 carcin$).tw. 10 (esophag$ adj5 tumo$).tw. 11 (oesophag$ adj5 tumo$).tw. 12 (esophag$ adj5 metasta$).tw. 13 (oesophag$ adj5 metasta$).tw. 14 (esophag$ adj5 malig$).tw. 15 (oesophag$ adj5 malig$).tw. 16 or/3-15 17 1 or 2 18 16 and 17

Page 185: Guideline pour la prise en charge du cancer oesophagien et ...

This page is left intentionally blank.

Page 186: Guideline pour la prise en charge du cancer oesophagien et ...

Dépôt légal : D/2008/10.273/17

Page 187: Guideline pour la prise en charge du cancer oesophagien et ...

KCE reports

1. Efficacité et rentabilité des thérapies de sevrage tabagique. D/2004/10.273/2. 2. Etude relative aux coûts potentiels liés à une éventuelle modification des règles du droit de la

responsabilité médicale (Phase 1). D/2004/10.273/4. 3. Utilisation des antibiotiques en milieu hospitalier dans le cas de la pyélonéphrite aiguë.

D/2004/10.273/6. 4. Leucoréduction. Une mesure envisageable dans le cadre de la politique nationale de sécurité des

transfusions sanguines. D/2004/10.273/8. 5. Evaluation des risques préopératoires. D/2004/10.273/10. 6. Validation du rapport de la Commission d’examen du sous financement des hôpitaux.

D/2004/10.273/12. 7. Recommandation nationale relative aux soins prénatals: Une base pour un itinéraire clinique de

suivi de grossesses. D/2004/10.273/14. 8. Systèmes de financement des médicaments hospitaliers: étude descriptive de certains pays

européens et du Canada. D/2004/10.273/16. 9. Feedback: évaluation de l'impact et des barrières à l'implémentation – Rapport de recherche:

partie 1. D/2005/10.273/02. 10. Le coût des prothèses dentaires. D/2005/10.273/04. 11. Dépistage du cancer du sein. D/2005/10.273/06. 12. Etude d’une méthode de financement alternative pour le sang et les dérivés sanguins labiles dans

les hôpitaux. D/2005/10.273/08. 13. Traitement endovasculaire de la sténose carotidienne. D/2005/10.273/10. 14. Variations des pratiques médicales hospitalières en cas d’infarctus aigu du myocarde en Belgique.

D/2005/10.273/12 15. Evolution des dépenses de santé. D/2005/10.273/14. 16. Etude relative aux coûts potentiels liés à une éventuelle modification des règles du droit de la

responsabilité médicale. Phase II : développement d'un modèle actuariel et premières estimations. D/2005/10.273/16.

17. Evaluation des montants de référence. D/2005/10.273/18. 18. Utilisation des itinéraires cliniques et guides de bonne pratique afin de déterminer de manière

prospective les honoraires des médecins hospitaliers: plus facile à dire qu'à faire.. D/2005/10.273/20

19. Evaluation de l'impact d'une contribution personnelle forfaitaire sur le recours au service d'urgences. D/2005/10.273/22.

20. HTA Diagnostic Moléculaire en Belgique. D/2005/10.273/24, D/2005/10.273/26. 21. HTA Matériel de Stomie en Belgique. D/2005/10.273.28. 22. HTA Tomographie par Emission de Positrons en Belgique. D/2005/10.273/30. 23. HTA Le traitement électif endovasculaire de l’anévrysme de l’aorte abdominale (AAA).

D/2005/10.273.33. 24. L'emploi des peptides natriurétiques dans l'approche diagnostique des patients présentant une

suspicion de décompensation cardiaque. D/2005/10.273.35 25. Endoscopie par capsule. D2006/10.273.02. 26. Aspects médico-légaux des recommandations de bonne pratique médicale. D2006/10.273/06. 27. Qualité et organisation des soins du diabète de type 2. D2006/10.273/08. 28. Recommandations provisoires pour les évaluations pharmacoéconomiques en Belgique.

D2006/10.273/11. 29. Recommandations nationales Collège d’oncologie : A. cadre général pour un manuel d’oncologie

B. base scientifique pour itinéraires cliniques de diagnostic et traitement, cancer colorectal et cancer du testicule. D2006/10.273/13.

30. Inventaire des bases de données de soins de santé. D2006/10.273/15. 31. Health Technology Assessment : l’antigène prostatique spécifique (PSA) dans le dépistage du

cancer de la prostate. D2006/10.273/18. 32. Feedback: évaluation de l'impact et des barrières à l'implémentation - Rapport de recherche:

partie II. D2006/10.273/20. 33. Effets et coûts de la vaccination des enfants Belges au moyen du vaccin conjugué

antipneumococcique. D2006/10.273/22. 34. Trastuzumab pour les stades précoces du cancer du sein. D2006/10.273/24.

Page 188: Guideline pour la prise en charge du cancer oesophagien et ...

35. Etude relative aux coûts potentiels liés à une éventuelle modification des règles du droit de la responsabilité médicale – Phase III : affinement des estimations. D2006/10.273/27.

36. Traitement pharmacologique et chirurgical de l'obésité. Prise en charge résidentielle des enfants sévèrement obèses en Belgique. D/2006/10.273/29.

37. Health Technology Assessment Imagerie par Résonance Magnétique. D/2006/10.273/33. 38. Dépistage du cancer du col de l’utérus et recherche du Papillomavirus humain (HPV).

D/2006/10.273/36 39. Evaluation rapide de technologies émergentes s'appliquant à la colonne vertébrale : remplacement

de disque intervertébral et vertébro/cyphoplastie par ballonnet. D/2006/10.273/39. 40. Etat fonctionnel du patient: un instrument potentiel pour le remboursement de la kinésithérapie

en Belgique? D/2006/10.273/41. 41. Indicateurs de qualité cliniques. D/2006/10.273/44. 42. Etude des disparités de la chirurgie élective en Belgique. D/2006/10.273/46. 43. Mise à jour de recommandations de bonne pratique existantes. D/2006/10.273/49. 44. Procédure d'évaluation des dispositifs médicaux émergeants. D/2006/10.273/51. 45. HTA Dépistage du Cancer Colorectal : état des lieux scientifique et impact budgétaire pour la

Belgique. D/2006/10.273/54. 46. Health Technology Assessment. Polysomnographie et monitoring à domicile des nourrissons en

prévention de la mort subite. D/2006/10.273/60. 47. L'utilisation des médicaments dans les maisons de repos et les maisons de repos et de soins

Belges. D/2006/10.273/62 48. Lombalgie chronique. D/2006/10.273/64. 49. Médicaments antiviraux en cas de grippe saisonnière et pandémique. Revue de littérature et

recommandations de bonne pratique. D/2006/10.273/66. 50. Contributions personnelles en matière de soins de santé en Belgique. L'impact des suppléments.

D/2006/10.273/69. 51. Besoin de soins chroniques des personnes âgées de 18 à 65 ans et atteintes de lésions cérébrales

acquises. D/2007/10.273/02. 52. Rapid Assessment: Prévention cardiovasculaire primaire dans la pratique du médecin généraliste

en Belgique. D/2007/10.273/04. 53. Financement des soins Infirmiers Hospitaliers. D/2007/10 273/06 54. Vaccination des nourrissons contre le rotavirus en Belgique. Analyse coût-efficacité 55. Valeur en termes de données probantes des informations écrites de l’industrie pharmaceutique

destinées aux médecins généralistes. D/2007/10.273/13 56. Matériel orthopédique en Belgique: Health Technology Assessment. D/2007/10.273/15. 57. Organisation et Financement de la Réadaptation Locomotrice et Neurologique en Belgique

D/2007/10.273/19 58. Le Défibrillateur Cardiaque Implantable.: un rapport d’évaluation de technologie de santé

D/2007/10.273/22 59. Analyse de biologie clinique en médecine général. D/2007/10.273/25 60. Tests de la fonction pulmonaire chez l'adulte. D/2007/10.273/28 61. Traitement de plaies par pression négative: une évaluation rapide. D/2007/10.273/31 62. Radiothérapie Conformationelle avec Modulation d’intensité (IMRT). D/2007/10.273/33. 63. Support scientifique du Collège d’Oncologie: un guideline pour la prise en charge du cancer du

sein. D/2007/10.273/36. 64. Vaccination HPV pour la prévention du cancer du col de l’utérus en Belgique: Health Technology

Assessment. D/2007/10.273/42. 65. Organisation et financement du diagnostic génétique en Belgique. D/2007/10.273/45. 66. Drug Eluting Stents en Belgique: Health Technology Assessment. D/2007/10.273/48. 67. Hadronthérapie. D/2007/10.273/51. 68. Indemnisation des dommages résultant de soins de santé - Phase IV : Clé de répartition entre le

Fonds et les assureurs. D/2007/10.273/53. 69. Assurance de Qualité pour le cancer du rectum – Phase 1: Recommandation de bonne pratique

pour la prise en charge du cancer rectal D/2007/10.273/55 70. Etude comparative des programmes d’accréditation hospitalière en Europe. D/2008/10.273/02 71. Recommandation de bonne pratique clinique pour cinq tests ophtalmiques. D/2008/10.273/05 72. L’offre de médecins en Belgique. Situation actuelle et défis. D/2008/10.273/08 73. Financement du programme de soins pour le patient gériatrique dans l’hôpital

classique : Définition et évaluation du patient gériatrique, fonction de liaison et évaluation d’un instrument pour un financement approprié. D/2008/10.273/12

Page 189: Guideline pour la prise en charge du cancer oesophagien et ...

74. Oxygénothérapie Hyperbare: Rapid Assessment. D/2008/10.273/14. 75. Guideline pour la prise en charge du cancer oesophagien et gastrique : éléments scientifiques à

destination du Collège d’Oncologie D/2008/10.273/17

Page 190: Guideline pour la prise en charge du cancer oesophagien et ...