83
É VOLUTION DE LA PRISE EN CHARGE PALLIATIVE DU CANCER DU PANCREAS AVANCE EN RELATION AVEC LARRIVEE DE NOUVEAUX MEDICAMENTS I MPLICATIONS SUR LA SURVIE ET CONSIDERATIONS MEDICO - ECONOMIQUES Thèse présentée à la Faculté de Médecine de l’Université de Genève pour obtenir le grade de Docteur en médecine par Sébastien MAZZURI de Lausanne (VD) Thèse n o 10452 Genève 2005 UNIVERSITÉ DE GENÈVE FACULTÉ DE MÉDECINE Section de médecine Clinique Département de Chirurgie Oncochirurgie Thèse préparée sous la direction du Dr. Arnaud D. ROTH , Privat-Docent et du Professeur Philippe MOREL

Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

Embed Size (px)

Citation preview

Page 1: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

ÉVOLUTION DE LA PRISE EN CHARGE PALLIATIVE DU CANCER DU PANCREAS

AVANCE EN RELATION AVEC L’ARRIVEE DE NOUVEAUX MEDICAMENTS

IMPLICATIONS SUR LA SURVIE ET

CONSIDERATIONS MEDICO -ECONOMIQUES

Thèse

présentée à la Faculté de Médecine de l’Université de Genève

pour obtenir le grade de Docteur en médecine par

Sébastien MAZZURI de

Lausanne (VD)

Thèse no 10452

Genève

2005

UNIVERSITÉ DE GENÈVE FACULTÉ DE MÉDECINE

Section de médecine Clinique

Département de Chirurgie

Oncochirurgie

Thèse préparée sous la direction du Dr. Arnaud D. ROTH , Privat-Docent et du Professeur Philippe MOREL

Page 2: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

i

Le présent travail a fait l’objet d’une publication :

Mazzuri, S., Roth, A. (2005). Modern systemic therapy of advanced pancreatic cancer: impact

on overall survival and medico economical aspects. Rev Med Suisse 1(24): 1616-20. French.

Page 3: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

ii

Page 4: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

iii

SYNOPSIS

Ce travail présente les résultats médico-économiques d’une étude rétrospective conduite sur la

cohorte des cas d’adénocarcinomes pancréatiques avancés suivis par la clinique d’Oncochirurgie

des Hôpitaux Universitaires de Genève ces dix dernières années. Le protocole comparait les

périodes 1994-98 et 1999-2003 (distinguées sur la base de la disponibilité de molécules modernes

telles que la gemcitabine).

Survie : les patients de la seconde période ont survécu significativement plus longtemps que

ceux de la période précédente (médiane de 8.7 versus 7.1 mois, probabilité de survie à 1 an de

40% versus 14% ; log-rank p=0.012), sans augmentation significative des épisodes

d’hospitalisation ;

Coûts : ce bénéfice de survie s’est accompagné d’une augmentation significative des dépenses

ambulatoires (médiane de 8'537 versus 5'005 CHF ; p=0.006).

Ces résultats sont à interpréter avec réserve au vu des limitations méthodologiques de l’étude.

Ils sont cependant corroborés dans la littérature actuelle par les données récentes de plusieurs

essais cliniques randomisés.

Page 5: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

iv

REMERCIEMENTS

Un très grand merci aux personnes suivantes, qui m’ont toutes été d’une aide inestimable dans

l’élaboration de ce travail et m’ont accordé beaucoup de leur temps malgré un emploi du temps

chargé :

- Merci en particulier à Arnaud Roth, de m’avoir proposé un sujet de thèse aussi intéres-

sant et comportant d’aussi multiples facettes, qui m’ont permis d’appréhender des

aspects pratiques et notamment statistiques à peine effleurés dans mes études. Merci

encore davantage pour sa sympathie, sa bonne humeur, son accessibilité et sa disponi-

bilité durant toute l’élaboration de ce travail ;

- Merci également à Nadine K’bourch pour son accueil, son dévouement et son aide

dans les méandres organisationnels du service d’Oncochirurgie ;

- Merci à toute l’équipe d’Oncochirurgie des H.U.G. pour leur accueil, leur sympathie,

leur cohésion et l’espoir qu’ils savent insuffler chaque jour à leurs patients par leur sou-

tien et leur attachement ;

- Merci à Bernadette Mermillod, de m’avoir permis d’éviter certains écueils statistiques

et d’aboutir à une présentation plus claire et solide de mes données ;

- Merci à Delphine Jaquier et Patrice Maricot pour leur maîtrise du système de

comptabilité analytique des H.U.G., rendant possible l’analyse médico-économique qui

donne une dimension plus originale car moins strictement médicale à ce travail ;

- Merci à Mariette Lapalud pour avoir su trouver et me faire parvenir aussi rapidement

les références nécessaires à la documentation de ce travail ;

- Merci à Isabelle Neyroud-Caspar de m’avoir fourni les informations épidémiologi-

ques relatives au Registre Genevois des Tumeurs ;

Ce travail, qui compte beaucoup pour moi, n’aurait sans doute pas pu aboutir sans votre aide.

Je vous souhaite à toutes et tous un parcours des plus enrichissant dans l’avenir !

Sébastien

Page 6: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 1 -

TABLE DES MATIERES

Table des matières __________________________________________________________ - 1 -

Résumé ___________________________________________________________________ - 3 -

Méthodologie ____________________________________________________________________- 5 -

Résultats ________________________________________________________________________- 7 -

Discussion_______________________________________________________________________- 9 -

Plan du travail __________________________________________________________________- 10 -

Introduction to pancreatic cancer _____________________________________________ - 11 -

Epidemiological considerations ____________________________________________________- 11 -

Pathology ______________________________________________________________________- 12 -

Development and genetics of pancreatic cancer_______________________________________- 13 -

Diagnosis and Staging____________________________________________________________- 14 -

Treatment overview and Survival __________________________________________________- 16 - Resectable pancreatic cancer _____________________________________________________________ - 16 - Locally advanced and metastatic pancreatic cancer ___________________________________________ - 18 -

Chemotherapy in advanced pancreatic carcinoma: literature review _________________ - 20 -

Methodological considerations_____________________________________________________- 20 - Performance status_____________________________________________________________________ - 20 - Evaluation of response in pancreatic cancer _________________________________________________ - 20 -

5-fluorouracil ___________________________________________________________________- 22 -

Other single-agent modalities [55]__________________________________________________- 23 -

Gemcitabine____________________________________________________________________- 23 - Initial phase I-II single-agent studies_______________________________________________________ - 23 - Phase III single-agent studies ____________________________________________________________ - 24 - Association regimens___________________________________________________________________ - 24 -

• Gemcitabine and 5-FU ___________________________________________________________ - 24 - • Gemcitabine and cisplatin ________________________________________________________ - 24 - • Gemcitabine and oxaliplatin (GemOx regimen) _______________________________________ - 25 - • Gemcitabine and irinotecan (IrinoGem regimen)_______________________________________ - 25 - • Gemcitabine and docetaxel________________________________________________________ - 25 - • Gemcitabine and epirubicin _______________________________________________________ - 25 -

Gemcitabine in radio-chemotherapeutic modalities ___________________________________________ - 25 -

Summary ______________________________________________________________________- 26 -

Retrospective study on a Geneva cohort ________________________________________ - 29 -

Introduction____________________________________________________________________- 29 -

Methodology ___________________________________________________________________- 30 - Patients______________________________________________________________________________ - 30 - Data mining __________________________________________________________________________ - 30 - Data analysis _________________________________________________________________________ - 32 -

• Cost analysis system in Geneva Cantonal Hospital _____________________________________ - 34 -

Page 7: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 2 -

• Conversion into standard units _____________________________________________________ - 35 - • Extra-hospital costs______________________________________________________________ - 36 -

Results ________________________________________________________________________- 37 - Epidemiological data ___________________________________________________________________ - 37 - Patients and Groups ____________________________________________________________________ - 38 - Treatment____________________________________________________________________________ - 45 - Survival analyses ______________________________________________________________________ - 47 - Hospitalization days____________________________________________________________________ - 49 - Economic analyses_____________________________________________________________________ - 53 -

Discussion______________________________________________________________________- 58 - Methodology _________________________________________________________________________ - 58 - Clinical Results _______________________________________________________________________ - 60 - Medico-economic Results _______________________________________________________________ - 63 -

Conclusion _______________________________________________________________ - 65 -

References________________________________________________________________ - 67 -

Page 8: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 3 -

RESUME

L’adénocarcinome pancréatique représente environ 90% des cas d’atteinte maligne du pan-

créas. Avec une incidence moyenne de 11.7 cas pour 100'000 de 1995 à 1998 à Genève, il repré-

sente environ 2 à 3% des diagnostics de néoplasies. Tous stades confondus, les taux de survie à 1

an et à 5 ans sont respectivement de 20% et de moins de 5%, tandis que la survie médiane après

résection n’est que de 13 à 20 mois. Pire encore, la survie médiane des cas métastatiques ainsi

que des cas loco régionalement avancés au diagnostic ne dépasse pas 3 à 6 mois et 6 à 10 mois,

respectivement. Ce pronostic catastrophique explique que la mortalité annuelle du carcinome du

pancréas se rapproche fortement de son incidence, puisque moins de 20% des patients se présen-

tent avec une maladie macroscopiquement limitée à l’organe lors du diagnostic initial : approxi-

mativement 40% souffrent d’une maladie loco régionalement dépassée, 40% présentent des

métastases viscérales –principalement hépatiques–, et 35% des implants tumoraux péritonéaux

(certains patients appartenant à deux, voire à ces trois catégories mutuellement non exclusives).

D’un point de vue thérapeutique, le seul traitement curatif repose sur une pancréatectomie, la

plus souvent partielle, qui ne peut être raisonnablement proposée qu’aux patients présentant une

maladie macroscopiquement limitée (stades I et II). Une radiothérapie adjuvante est parfois

offerte à distance de la chirurgie dans l’espoir d’un meilleur contrôle locorégional. Malheureuse-

ment, et en raison d’une symptomatologie frustre et souvent tardive, la majorité des patients sont

diagnostiqués à des stades avancés. Les possibilités thérapeutiques se limitent alors à des inter-

ventions palliatives de dérivation biliaire et/ou digestive si nécessaire, et à des traitements de

chimiothérapie/radiothérapie. Concernant la chimiothérapie, le 5-fluorouracile était considéré

comme le traitement de choix jusqu’au milieu des années 90, même si quelques études randomi-

sées contrôlées n’avaient pu mettre en évidence de bénéfice significatif par rapport à un traite-

ment de soutien seul en termes de survie.

L’introduction de la gemcitabine dans cette indication, en Suisse au deuxième semestre de

1998, puis de l’oxaliplatine, apporta un second souffle à la prise en charge palliative de ces

patients, comme peut en témoigner l’augmentation exponentielle des études cliniques de phase I

et II ces dernières années, cherchant à définir les régimes de drogues modernes les plus efficaces

et surtout les moins toxiques. D’une manière générale, ces innovations thérapeutiques ne se sont

pas traduites de manière fulgurante en termes d’indicateurs classiques que sont la survie médiane

et la survie à 1 an, n’améliorant la situation que de manière marginale quoique cependant statisti-

Page 9: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 4 -

quement significative. En réalité, la véritable amélioration a plutôt concerné la symptomatologie

de ces patients et l’impact de la maladie sur leur qualité de vie en termes d’index de performance

et de soulagement des douleurs. En effet, les essais randomisés contrôlés comparant des

protocoles modernes à base de gemcitabine au précédent traitement de référence à base de 5-fluo-

rouracile ont mis en évidence la supériorité significative des premiers sur le second en termes de

bénéfice clinique (Clinical Benefit Response) sans augmentation significative des effets secondai-

res indésirables sévères. Ainsi, l’attitude de la majorité des oncologues a passablement évolué

depuis l’introduction de la gemcitabine, passant d’une tendance à l’abstention thérapeutique ou à

la prescription peu convaincue de 5-fluorouracile, à la prescription de molécules modernes plus

fréquente car reconnue comme efficace en termes de bénéfice clinique. D’un point de vue plus

classique et quantitatif, et selon les données de la littérature actuelle que nous résumons en pre-

mière partie de notre travail, les patients atteints d’un carcinome du pancréas dépassé pouvaient

espérer une survie médiane d’environ 5 à 7 mois sous traitement de 5-fluorouracile seul. Cette

espérance est actuellement d’environ 7 à 10 mois sous traitement de gemcitabine. Concernant les

associations de gemcitabine à d’autres agents chimiothérapeutiques classiques tels que le cispla-

tine ou l’oxaliplatine, les espoirs fondés sur les résultats de nombreuses études de phase II ont été

très récemment mis à mal par la présentation au congrès de l’ASCO 2004 des conclusions déce-

vantes de plusieurs essais cliniques randomisés de phase III, ne parvenant pas à démontrer de

supériorité significative des régimes d’association sur les protocoles à base de gemcitabine seule

en termes de survie. Les résultats définitifs de deux études de phase III comparant une association

de gemcitabine et de Xeloda (formulation orale du 5-fluorouracile se rapprochant d’une infusion

continue) à la gemcitabine seule devraient être disponibles dans un avenir proche (protocoles

SAKK et MRC).

Dans ce contexte, il nous a paru intéressant de mener une étude rétrospective1 sur le sujet en

analysant l’ensemble des cas de carcinomes du pancréas ayant été suivis par le service

d’oncochirurgie des Hôpitaux Universitaires de Genève depuis octobre 1993 jusqu’en octobre

2003, soit sur la dernière décennie. Notre intention était davantage de comparer deux périodes

que deux traitements précis, c’est-à-dire une période de 1993 à 1998 avant l’introduction de la

gemcitabine, et la période suivante de 1999 à 2003, en posant l’hypothèse initiale selon laquelle

le changement d’attitude des oncologues relatif à la prescription de chimiothérapie dans cette

1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

Page 10: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 5 -

indication se traduirait par une différence significative en termes de survie et d’indicateurs gros-

siers de qualité de vie tels que la durée d’hospitalisation. De plus, au vu des considérations politi-

ques, assécurologiques et économiques actuelles qui remettent en question les pratiques

médicales modernes de la plupart des pays développés, nous avons voulu étendre l’analyse à une

dimension médico-économique et comparer les deux périodes en termes de dépenses pour

l’hôpital liées au cancer du pancréas avancé.

METHODOLOGIE

Pour ce faire, nous avons tout d’abord obtenu du Registre Genevois des Tumeurs les informa-

tions épidémiologiques relatives à la décennie analysée. Puis nous avons répertorié l’ensemble

des cas de carcinome du pancréas ayant bénéficié d’une consultation par le service

d’oncochirurgie, et inclus les patients suivis auprès de ce dernier durant une proportion significa-

tive de la maladie. Nous avons ensuite constitué une base de données comprenant des informa-

tions démographiques ainsi que les caractéristiques et jalons de la maladie : date de diagnostic

(définie comme la date d’entrée du séjour hospitalier ayant mené à ou confirmé le diagnostic) ;

facteurs pronostiques (âge, stade, taille de la tumeur, site primitif et index de performance au dia-

gnostic) ; nombre et durée d’hospitalisations (catégorisées en traitement, complication du traite-

ment, complication de la maladie et autres) ; détails du traitement (opérations, radiothérapie,

nombre de lignes, de cycles et doses de chimiothérapie). En ce qui concerne la définition des

groupes, nous avons classé chronologiquement les patients retenus en fonction de la date du

diagnostic de la maladie oncologique, puis considéré la première ligne de chimiothérapie reçue,

en excluant les modalités combinées de radio- et chimiothérapie. Nous avons alors retenu comme

date charnière pour la définition des deux périodes la date du premier cycle de gemcitabine pour

le premier patient recevant ce médicament en première intention. En outre, avec l’aide du l’Unité

d’Information Médico-Economique de l’hôpital, nous avons calculé sur la base du système de

comptabilité analytique des H.U.G. le coût de tous les séjours hospitaliers (H.U.G., Beau-Séjour

et CESCO) et des visites ambulatoires en oncochirurgie pour chacun des patients inclus dans

l’étude. De plus, nous avons calculé un coût moyen par jour d’hospitalisation pour les séjours

dans des établissements de convalescence (Joli-Mont, Montana, Loëx, Génolier etc.) afin de les

inclure dans notre estimation du total des coûts. En raison de la période de temps étendue recou-

verte par notre étude, l’ensemble des coûts a été annualisé en unités 2002 au moyen de facteurs

de pondération spécifiques afin de prendre en compte l’augmentation des coûts de la santé et de

permettre une comparaison entre les deux groupes. La quasi-totalité des patients considérés ayant

Page 11: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 6 -

été suivis intégralement par le service d’oncochirurgie de l’Hôpital Cantonal en ambulatoire, il

est peu probable que des dépenses importantes aient été encourues en dehors de cette

consultation. En conséquence, et étant donné que leur détermination aurait été largement aléatoire

compte tenu du design rétrospectif du protocole, les coûts extrahospitaliers (à ne pas confondre

avec les coûts ambulatoires) n’ont pas été considérés dans l’analyse.

Des analyses statistiques classiques ont alors été conduites sur la base des données récoltées,

avec un seuil de signification fixé à 0.05 : l’objectif primaire consistait en une analyse de survie,

effectuée selon la méthode de Kaplan et Meier afin de tenir compte des patients perdus de vue et

des patients toujours vivants au moment de l’analyse. Les objectifs secondaires comprenaient une

analyse de durée d’hospitalisation, ainsi qu’une comparaison économique des coûts pour l’hôpital

au cours des deux périodes considérées.

A noter que si notre intention initiale était de considérer les cas de carcinome pancréatique

tous stades confondus, nous avons finalement décidé de restreindre l’analyse aux stades III et IV,

respectivement loco régionalement avancés et métastatiques, pour des raisons d’homogénéité : les

deux groupes issus de la cohorte globale se sont en effet révélés lors de l’analyse des facteurs

pronostiques significativement différents en termes de proportion de stades I et II versus III et IV,

introduisant un important biais potentiel pour l’analyse de survie : il est bien démontré que les

patients bénéficiant d’une résection survivent significativement plus longtemps que les patients

diagnostiqués directement à un stade palliatif, et que ces deux catégories correspondent à deux

profils différents et incomparables de biologie tumorale. Une solution potentielle eût été de défi-

nir pour les patients réséqués une date de rechute, et de ne considérer que leur survie en situation

palliative. Cependant, la définition d’une telle date s’avère extrêmement hasardeuse étant donné

l’absence de protocole de suivi précis et l’absence de corrélation entre les marqueurs biologiques

et la charge tumorale en termes pronostiques. D’une manière générale, l’établissement d’un stade

pronostique en situation de récurrence étant un non-sens en oncologie, il aurait été difficile

d’apprécier l’homogénéité de groupes ainsi définis. Dans ce contexte, nous avons en définitive

conduit les analyses de survie ainsi que les calculs médico-économiques sur une population res-

treinte aux stades III et IV uniquement.

Page 12: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 7 -

RESULTATS

Avec un total de 84 malades, notre échantillon représentait environ 16% de l’ensemble des cas

de carcinome du pancréas ayant été déclarés dans le canton durant la période 1994-2001 (données

épidémiologiques non disponibles pour 2002 et 2003). La date pivot retenue correspondant au 22

décembre 1998, nous avons initialement déterminé deux groupes comprenant 39 patients pour la

première période (Groupe A) et 45 patients pour la seconde période (Groupe B). L’analyse de ces

patients en termes de facteurs démographiques et pronostiques n’a pas révélé de différences

significatives pour le sexe, la taille de la tumeur, sa localisation et l’index de performance au dia-

gnostic initial. L’âge au diagnostic était par contre significativement plus jeune dans le groupe A.

Mais surtout les deux groupes se sont avérés significativement différents en termes de stade au

diagnostic, les proportions respectives de stades I et II (patients résécables) et de stades III et IV

(maladies dépassées) se montant à 44%/56% versus 22%/78%, respectivement pour les groupes

A et B (p=0.037). Pour les raisons citées précédemment, nous avons alors décidé de conduire les

analyses ultérieures sur une cohorte restreinte aux cas loco régionalement avancés ou métastati-

ques au diagnostic, dont l’évolution clinique peut être considérée comme homogène. Les groupes

finaux comprenaient 22 patients pour le groupe A et 35 pour le groupe B. Une comparaison sta-

tistique en termes de facteurs démographiques et pronostiques n’a pas révélé de différence statis-

tiquement significative excepté la persistance d’un écart d’âge au diagnostic initial (médiane de

59 ans pour le groupe A contre 66 pour le groupe B, p=0.005).

En ce qui concerne l’objectif primaire de l’étude, une analyse de survie a révélé une différence

significative entre les deux groupes, avec une médiane de survie de 7.1 et 8.7 mois et une proba-

bilité de survie à un an de 14% et 40% pour le groupe A et le groupe B, respectivement (log-rank

p=0.012, hazard ratio=0.52, intervalle de confiance à 95% 0.23-0.84). Les patients ayant survécu

plus d’une année, soit les « survivants à long terme », sont majoritairement responsables de cette

observation, comme en témoigne l’écartement des courbes au-delà de 300 jours. Cependant, un

recouvrement des intervalles de confiance à 95%, expliqué par la petite taille des effectifs, nous

pousse à interpréter ces résultats plutôt comme une tendance qu’une différence formellement

significative (intervalles de confiance à 95% pour la probabilité de survie à un an : 3-35% contre

24-58%, respectivement). Ces valeurs sont compatibles avec les chiffres moyens publiés dans la

littérature. Une analyse de la quantité de chimiothérapie administrée aux patients des deux grou-

pes a également révélé une différence significative, avec une médiane de 1 dose administrée par

patient pour le groupe A contre 14 pour le groupe B (p=0.0002).

Page 13: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 8 -

Une analyse du nombre total de journées d’hospitalisation n’a pas révélé de différences signi-

ficatives entre les deux groupes. Dès lors, une analyse comparant les deux groupes en termes de

survie ambulatoire (durée des séjours en hôpital retranchée de la survie totale) a logiquement

confirmé le bénéfice de survie initialement observé pour le groupe B (médiane de 144 jours hors

hospitalisation pour le groupe A contre 199 pour le groupe B, P=0.013, hazard ratio=0.52, inter-

valle de confiance à 95% 0.24-0.84). Si l’on accepte ce paramètre comme une appréciation gros-

sière de la qualité de vie, nous concluons prudemment que les gains observés en termes de survie

avec les traitements actuels ne semblent pas être grevés d’une morbidité excessive : en effet, la

majorité des séjours hospitaliers pour les patients inclus dans l’étude correspondaient à des situa-

tions de complications sévères liées à la maladie, telles que des obstructions biliaire ou digestive

sanctionnées chirurgicalement, ou à des situations de soins de confort en fin de vie.

L’analyse du nombre de consultations en ambulatoire au service d’oncochirurgie pour traite-

ment ou suivi a elle aussi révélé une différence significative entre les deux groupes, les patients

du groupe B affichant une médiane de 28 visites contre 21 pour le groupe A (p=0.007). Ces

résultats sont à mettre en relation avec les observations médico-économiques correspondantes :

une comparaison globale des deux groupes n’a pas révélé de différence significative en termes de

total des coûts hospitaliers. Par contre, une stratification plus fine en coûts reliés aux hospitalisa-

tions et coûts reliés aux visites ambulatoires a corroboré les résultats précédemment exposés : les

deux groupes n’ont pas présenté de différence significative en termes de coûts reliés aux hospita-

lisations, mais les patients du groupe B ont coûté significativement plus cher en visites

ambulatoires (médiane de 5'005 CHF pour le groupe A versus 8'537 pour le groupe B, p=0.006).

La faible proportion du total des coûts représentée par ces dernières (6% pour le groupe A versus

15% pour le groupe B) peut expliquer que cette différence ne se reflète pas dans l’analyse glo-

bale. De même, l’augmentation de cette proportion entre les deux périodes est à mettre en relation

avec l’augmentation de la durée de survie et du nombre de chimiothérapies administrées, de

même qu’avec l’augmentation du coût moyen des drogues correspondantes.

Page 14: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 9 -

DISCUSSION

Si nos résultats sont indéniablement sujets aux biais classiques inhérents aux analyses rétros-

pectives et à l’importance de la période de temps considérée, ils n’en suggèrent pas moins une

conclusion intéressante, dont la portée locale et exploratoire mériterait d’être confirmée prospec-

tivement sur une échelle plus large : considérées dans leur globalité, nos observations suggèrent

qu’il existe une faible proportion de patients atteints de carcinome pancréatique avancé dont la

biologie tumorale plus favorable pourrait se traduire par une meilleure susceptibilité à la chimio-

thérapie. Ce sont ces patients qui semblent bénéficier majoritairement des nouvelles molécules, et

il n’est désormais plus rare de suivre des survivants à long terme pendant parfois 2 à 3 ans, voire

même davantage. Étant donné que la différence de survie observée est une différence de survie

ambulatoire, on peut supposer à première vue que ce bénéfice s’accompagne d’un certain main-

tien de la qualité de vie. De plus, et par voie de conséquence, à cette prolongation correspond

également une multiplication des consultations de suivi et de traitement loco régionalement,

entraînant un surcoût ambulatoire certain quoique modeste. Si dans le cas précis du carcinome

pancréatique avancé ce surcoût n’est pas très impressionnant en raison de la survie médiane très

limitée des patients concernés (quoique certains survivants à long terme inclus dans notre étude

aient atteint une fourchette de coûts ambulatoires allant de 20'000 à 50'000 CHF), une extrapola-

tion à la population suisse dans son ensemble, et surtout à d’autres pathologies malignes bien plus

prévalentes, permet de replacer cette problématique au centre des préoccupations économiques et

politiques actuelles. A cet égard, le cas du carcinome colorectal métastatique est édifiant, puisque

la survie médiane observée a quasiment doublé sur les dix dernières années : estimée entre 10 et

12 mois au début des années 90, elle dépasse actuellement les 20 mois avec les associations chi-

miothérapeutiques les plus modernes. A ce doublement de l’espérance de vie médiane des

patients s’est associée une augmentation de près de 340 fois du prix moyen des médicaments,

passant d’environ 100-300 dollars pour le 5-fluorouracile à environ 20’000-30'000 dollars pour

les nouvelles thérapies ciblées (bevacizumab, cetuximab), et ce en considérant uniquement les

huit premières semaines de traitement !

L’évolution de l’oncologie moderne illustre donc particulièrement bien le prix à payer pour

repousser davantage les limites de la maladie : si le progrès médical n’est certainement pas à lui

tout seul responsable de l’augmentation des coûts de la santé, il en participe indéniablement, tant

par la mise à disposition de molécules toujours plus ciblées, plus complexes à développer et pres-

que invariablement plus chères que par l’augmentation de survie qu’elle rendent possible pour

Page 15: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 10 -

des patients nécessitant des soins –soit une consommation de ressources médicales– réguliers.

Compte tenu de l’impact possible qu’auront les mesures politiques actuellement en chantier en

Suisse sur la disponibilité de ce progrès au plus grand nombre à moyen terme, il est de notre point

de vue nécessaire d’impliquer au plus vite tous les acteurs du secteur –législateur, fournisseurs de

soins, payeurs, et avant tout, patients– dans une réflexion plus transparente et consensuelle.

PLAN DU TRAVAIL

Après une introduction épidémiologique et clinique générale à la pathologie maligne du pan-

créas, nous mènerons une revue systématique de la littérature concernant la prise en charge loco

régionalement du cancer pancréatique avancé et présenterons les associations médicamenteuses

les plus prometteuses évaluées ces dernières années dans le cadre du traitement de cette patholo-

gie. Ensuite, nous introduirons la problématique de notre étude, présenterons notre méthodologie

ainsi que nos résultats, que nous commenterons enfin sur les plans méthodologique, clinique et

médico-économique.

Page 16: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 11 -

INTRODUCTION TO PANCREATIC CANCER

EPIDEMIOLOGICAL CONSIDERATIONS

Pancreatic ductal adenocarcinoma is the most common malignant disease of the exocrine pan-

creas and accounts for approximately 90% of pancreatic cancer cases. There are about 30,000

newly diagnosed cases and approximately the same number of deaths from this disease annually

in the United States. Pancreas cancer accounts for 2% to 3% of all malignant neoplasms diag-

nosed worldwide [1]. All stages combined, 1-year and 5-year survival rates are 20% and less than

5%, respectively. Median survival time is 13 to 20 months after pancreatic resection with less

than 10% to 20% of patients being long-term survivors (i.e. surviving more than one year); it is 3

to 6 months and 6 to 10 months for patients with metastatic and locally advanced disease, respec-

tively. Unfortunately, at the time of presentation, less than 20% of patients have a disease macro-

scopically confined to the organ: approximately 40% have locally advanced disease, 40% show

visceral metastases and 35% show peritoneal implants [2] (the latter categories being not mutu-

ally exclusive). This explains why, whilst being the ninth cancer in men and the eighth in women

in terms of frequency, pancreatic cancer is the fourth cause of cancer deaths in both sex (US data)

[1], causing around 200,000 deaths yearly in the world. Data from the National Cancer Database

report a peak age of occurrence of 70 to 79 years [3].

The two biggest risk factors for pancreatic cancer are ageing and smoking. Other predisposing

factors include occupational exposure to leather tanning and machine shops [4]. Patients with

hereditary chronic pancreatitis are at increased risk. The estimated risk for this specific

population to age 70 may be as high as 40%, several decades after onset of the initial pancreatitis

[5]. This familial syndrome of autosomal dominant inheritance and of 80% penetrance rate is

characterized by recurrent episodes of pancreatitis occurring early in childhood and recurring

throughout life. Its incidence is uncertain, but as many as 5% of patients experiencing an episode

of pancreatitis may have the hereditary type of the disease. In hereditary pancreatitis, uninhibited

mutated trypsin [6] initiates a proteolytic cascade within the pancreas itself, leading to

autodigestion and acute pancreatitis. In patients with chronic pancreatitis from other causes, the

risk of pancreatic adenocarcinoma has been estimated between 2 and 15 times greater than in

control people [7, 8].

Page 17: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 12 -

Familial pancreatic cancer is another autosomal dominant condition with an unidentified

causative mutation. Diagnostic criteria are the following: two or more first degree relatives with

pancreatic ductal adenocarcinoma, one first degree relative with early-onset disease (less than 50

years at the time of diagnosis) or two or more second degree relatives with the disease, one of

whom having experienced early-onset cancer. Other genetic causes of pancreatic cancer, associ-

ated with an increased risk of invasive skin melanoma, include mutations of the CDKN2 [9],

p16INK4 [10] and BRCA1/2 [11, 12] tumor suppressor genes.

PATHOLOGY

The TNM classification system for pancreatic tumors was modified in 2002 [13]:

TNM Classification and

Staging System Characteristics

Tx Primary tumor not assessable T0 No evidence of primary tumor Tis Carcinoma in situ T1 Tumor limited to the pancreas, <2cm in diameter T2 Tumor limited to the pancreas, >2cm in diameter

T3 Tumor extending beyond the pancreas but without involving the celiac axis or the superior mesenteric artery

T4 Tumor involving the celiac axis or the superior mesenteric artery (unresectable primary tumor)

Nx Regional lymph nodes not assessable N0 Regional lymph nodes not involved N1 Regional lymph nodes involved Mx Distant metastasis not assessable M0 No distant metastasis M1 Distant metastasis/es

Stage 0 Tis, N0, M0 Stage I T1-2, N0, M0 Stage II T3, N0-1, M0 Stage III T4, any N, M0 Stage IV Any T, any N, M1

This revised classification is clinically more coherent in terms of prognostic: contrasting with

previous iterations, stages I and II now correspond to resectable tumors and stages III and IV to

unresectable ones, respectively locally advanced and metastatic.

Based on National Cancer Database registry data, 78% of pancreatic adenocarcinomas arise in

the head region, while 11% arise in the body and 11% in the tail [3]. Because of their proximity

to the common bile duct, patients with a tumor arising in the pancreatic head commonly present

Page 18: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 13 -

at earlier stages (longest axis usually 2 to 3.5 cm) with painless jaundice caused by bile duct

obstruction. Conversely, patients with a tumor arising in the body and/or tail region present at a

later stage (longest axis usually 5 to 7 cm), typically with pain caused by malignant infiltration of

neural and retroperitoneal structures. Accordingly, the ratio of limited to advanced disease (stage

I/stage IV) is 0.7 for tumors arising in the head region, but only 0.24 for body tumors and 0.1 for

tail tumors [3].

From a practical viewpoint, locally advanced pancreatic cancer is defined as an unresectable

tumor without evidence of distant metastases. Generally, one of the following features accounts

for surgical unresectability [14]:

- Extensive peripancreatic lymph node involvement; - Encasement or occlusion of the superior mesenteric vein or superior mesenteric vein/portal

vein confluence; - Direct involvement of the superior mesenteric artery, inferior vena cava, aorta or celiac

axis.

DEVELOPMENT AND GENETICS OF PANCREATIC CANCER

Regarding the pancreatic carcinoma precursor lesion, some clinical studies have demonstrated

a chronological sequence between pancreatic duct lesions and invasive carcinomas and thus

established that the former were truly precursors to the latter [15, 16]. The Pancreatic Intraepithe-

lial Neoplasia (PanIN) nomenclature for early lesions was standardized in 1999 based on this

sequence. It grades lesions on a 1 to 3 scale, grade 1 showing the least architectural and cytologi-

cal atypia and grade 3, equivalent to carcinoma in situ, showing the most [17].

From a molecular genetic perspective, activating point mutations in proto-oncogene K-ras

(seen in 90% of pancreatic adenocarcinoma and in roughly 30% of all cancers, including colon,

lung and bladder carcinoma) and over expression of proto-oncogene Her2/neu (a member of the

epidermal growth factor receptor family) are seen in the earliest precursor lesions and are thought

to be early genetic events in the development of pancreatic carcinoma [8]. K-ras protein normally

goes through a cycle of GTP-GDP exchange and is involved in signal transduction of extra cel-

lular mitogenic stimuli, thus promoting cell growth and proliferation. Oncogenic K-ras is unable

to hydrolyze bound GTP, resulting in constant K-ras signaling that cannot be switched off [18].

This is followed by papillary hyperplasic lesions. Inactivation of the p16 tumor suppressor gene,

which stops cellular proliferation, seems to occur at this stage (PanIN 2 and 3), and the p53 (seen

in 50-70% of pancreatic tumors [19]), DPC4 (or SMAD4) and BRCA2 genes are apparently lost

in even later stages of the neoplasic process (almost exclusively in PanIN 3) [15, 16, 20]. The

Page 19: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 14 -

timing of other tumorigenic activities (DNA methylation, telomerase and matrix metalloprotein-

ase activation) and a number of low-frequency genetic alterations (inactivation of the MKK4,

STK11, ALK5 and TGFBR2 tumor suppressor genes) has not been described yet.

DIAGNOSIS AND STAGING

Initial symptoms of pancreatic carcinoma include pain, jaundice, anorexia, early satiety,

weight loss, depression and new-onset diabetes. Among them, painless jaundice offers the best

prognosis, with more than 50% of patients being eligible for resection based on preoperative

assessment. Conversely, 80% to 90% of patients presenting with pain are declared unresectable,

since this almost invariably results from celiac plexus invasion. Weight loss is due on the one

hand to interruption of gastrointestinal flow of bile and pancreatic juice, and on the other hand to

the typical catabolic state accompanying aggressive malignancies. Jaundice is most commonly

obstructive as a result of a tumor arising in the head of pancreas and obstructing the intra pancre-

atic bile duct. Other clinical signs of the disease include hepatomegaly, a palpable gallbladder

(Courvoisier’s sign, seen in 40% of cases), cachexia, Troisier’s sign (involved Virchow’s node)

and ascites [18].

Currently, no serum marker offers enough sensitivity or specificity to be employed as an

effective early diagnosis or screening tool. CA 19-9 serum level is the most sensitive and specific

one, but it is almost never positive in small, i.e. curable tumors [21]. Furthermore, benign causes

of bile duct obstruction and other gastrointestinal cancers may also raise serum CA 19-9 levels.

On the other hand, CA 19-9 dosages are commonly used in the follow-up of patients.

As already mentioned, a number of genetic lesions are associated with pancreatic adenocarci-

noma, including mutations of the K-ras (90%), p16 (80%), p53 (50%) and DPC4 (50%) genes

[22]. Among them, however, none has demonstrated enough sensitivity nor specificity to serve as

a discriminating diagnostic tool, even the most extensively studied K-ras mutation [17]. Potential

for gene therapy is being currently intensively explored, for instance by replacement of tumor

suppressor genes, use of antisense oligonucleotides, immunotherapy or gene-directed enzyme

prodrug therapies [23]. So far, results have been rather disappointing: for example, a combination

regimen of gemcitabine plus tipifarnib (farnesyltransferase inhibitor) has not been shown superior

to gemcitabine alone in terms of overall survival time in a recent randomized phase III trial [24].

The role of diagnostic imaging in the assessment of a suspected pancreatic carcinoma is, first,

to establish the diagnosis of a malignant lesion, and second to assess resectability. CT scanning is

Page 20: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 15 -

the first-choice imaging technique, since it assesses pancreatic parenchymal lesions and provides

information about the ductal system, local extension, the presence or absence of biliary dilation,

regional lymph nodes status and the status of other target organs of distant metastases [4]. Small

tumors, i.e. curable lesions, may fall below the detection level of this standard imaging technique.

However, the sensitivity of dual-phase spiral CT scanning for identifying unresectable tumors

may be as high as 90% [25], although less optimistic results have been published in a recent

review [26] (i.e. about 75%). Thus, in patients with radiological absence of the above mentioned

unresectability criteria, surgeons may still be confronted to advanced tumors at the time of

laparotomy [27, 28].

Magnetic resonance imaging does not seem to be more sensitive than CT scanning for the

diagnosis of pancreatic cancer, but its specificity may be superior, allowing for the distinction of

a clear mass in patients whose CT assessment can only evidence indeterminate enlargement of

the pancreatic head. However, its ability to discriminate resectable lesions from unresectable

tumors has not been proven superior to CT scan imaging [29].

ERCP adds generally little to diagnostic staging, but it remains indicated for patients present-

ing with biliary obstruction when an ampullary lesion is suspected. On the other hand, endoscopic

ultrasound may be the most accurate imaging test for the diagnosis of pancreatic cancer. This

technique seems especially effective in identifying tumors less than 3 cm in diameter, which may

be missed on CT scans [30]. It is also a highly effective modality to obtain a tissue diagnosis with

a very low complication rate, minimizing the risk of tumor dissemination (which is occasionally

attributed to percutaneous needle biopsy).

Finally, laparoscopic staging is performed in some centers, since it may prevent unnecessary

laparotomy by detecting occult micro metastasis in 30% of patients [26, 31]. However, when

laparoscopy is performed on patients thought to have resectable disease according to high-quality

CT criteria, the proportion of unresectability findings is in the range of 4% to 13% only, as

reviewed by Pisters et al. [26]. This observation does not support the use of laparoscopy as a rou-

tine staging procedure.

Page 21: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 16 -

Nevertheless, the condition for defining an adequate treatment strategy relies on an accurate

tissue diagnosis, which is unfortunately often difficult to obtain, despite clinical or radiological

evidence of pancreatic malignancy. Indeed, paucity of cancer cells in the diagnostic sample as

well as intense local fibrosis associated with the disease often challenge histological diagnosis

[17]. Hence, many patients enter treatment without a formal tissue diagnosis (depending on cen-

ters). Also, presentation and radiological findings in chronic pancreatitis and pancreatic cancer

overlap in a significant proportion of cases. Thus, as much as 5 % to 10% of patients with sus-

pected pancreatic cancer may turn out to suffer from chronic pancreatitis upon examination of the

resection specimen. Conversely, around 5% of patients with presumed chronic pancreatitis who

undergo resection end up with a final histological diagnosis of pancreatic cancer. This diagnostic

dilemma places a significant degree of stress on surgical decision-making, since pancreatic resec-

tion may have a mortality rate of as much as a 5% and a morbidity rate of about 40% [18].

TREATMENT OVERVIEW AND SURVIVAL

RESECTABLE PANCREATIC CANCER

The only curative treatment of pancreatic adenocarcinoma is surgical resection, i.e. either a

pancreaticoduodenectomy for lesions arising in the head region or a left splenopancreatectomy or

sometimes a total pancreatectomy for lesions arising in the body and tail regions. Surgery is not

proposed if liver or peritoneal metastases are present at the time of diagnosis or if metastases are

identified in lymph nodes that are not routinely removed as part of the procedure. In the absence

of distant metastases, resectability depends on major vascular invasion by the tumor. However,

advances in surgical technique now allow for en bloc resection of the superior mesenteric

vein/portal vein confluence for isolated tumor extension although it is not clear whether such a

procedure shall have a positive influence on the overall survival time [32]. Data support a current

mortality rate for pancreaticoduodenectomy in the range of 1% to 5% [33, 34], strongly contrast-

ing with the 30-day mortality rate of this procedure in the 1960s and 1970s, which approached

25%. Unfortunately, despite evident progresses in diagnostic and staging modalities and

improved surgical management, there has not been any dramatic improvement in overall survival

time for patients diagnosed with pancreatic carcinoma over the past two decades [35].

Page 22: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 17 -

Adjuvant therapy

Patterns of failure after surgical resection are well established: loco regional recurrence

develop from persistent intra abdominal microscopic disease as well as hepatic or peritoneal

metastases from persistent micro metastatic disease [36]. The principles of adjuvant therapy sug-

gest trying and eradicating this persistent microscopic disease before additional proliferation into

clinically evident recurrence.

In a 2003 review, Abrams et al. point out recent evidence regarding adjuvant therapy in

resectable pancreatic cancer [37]: in 1985, an important although controversial study by the

Gastrointestinal Tumor Study Group prospectively randomized post-pancreatectomy patients to

either observation (control arm) or to split course radiotherapy (40 Gy in 2 20-Gy sequences, 20

fractions and 6 weeks) with bolus 5-FU (500 mg/m2 intravenously daily on each of the first three

radiation days of each 20 Gy sequence), followed by bolus 5-FU once weekly for 2 years after

completion of radiotherapy [38]. This study evidenced a positive effect this adjuvant protocol,

since median and long-term survival time were both significantly improved in the latter group of

patients (20 months versus 11 months, P=0.03, and 19% versus 5% 5-year survival probability,

respectively). Yet, the results of this study were contested since less than fifty patients entered the

trial. A follow-up GITSG study registered an additional 30 patients to receive adjuvant 5-FU and

radiation therapy [39]. This group of patients showed a median survival time of 18 months and a

5-year survival probability of 17%.

The European Organization for Research and Treatment of Cancer (EORTC) recently com-

pleted a randomized phase III trial evaluating adjuvant chemo radiation versus observation in 218

patients having undergone curative-intent resection of the pancreatic head and carcinomas of the

periampullar region [40]. The chemo radiation regimen was similar to the one given in the

GITSG trial, but without maintenance therapy. Survival data showed improvements in the chemo

radiation arm, with a median survival time of 24.5 months versus 19 months and a 5-year survival

probability of 28% versus 22%, however not reaching statistical significance. Although the

authors concluded that routine use of adjuvant chemo radiotherapy should not become a standard

treatment in resectable pancreatic cancer, their radiation schedule (40 Gy in 2 20-Gy sequences)

was not the most effective one. Overall however, chemo radiotherapy as an adjuvant treatment

after resection of pancreatic cancer still remains controversial in terms of survival time benefit

[41]. A randomized trial by the European Study Group for Pancreatic Cancer even evidenced

potential deleterious effects for chemo radiotherapy versus chemotherapy [42].

Page 23: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 18 -

Neoadjuvant therapy

As much as 26% of patients with a tumor considered as resectable suffer from metastatic evo-

lution 3 months after surgery [43]. In this context, the rationale for proposing preoperative chemo

radiation regimens in potentially resectable pancreatic cancer is three-fold [44]: first, neoadjuvant

treatment would eliminate the delay of adjuvant treatment due to postoperative recovery; second,

neoadjuvant treatment might downstage tumors, thus increasing the likelihood of negative surgi-

cal margins; and third, neoadjuvant treatment could help prevent peritoneal tumor cell implanta-

tion and dissemination caused by manipulation during surgery. Preoperative chemo radiation for

pancreatic cancer was pioneered by Evans et al. in a phase II trial administering preoperative

radiation (50.4 Gy) and concurrent continuous infusion 5-FU 300 mg/m2/day in 28 patients with

cytological or histological proof of pancreatic head adenocarcinoma [45]. The ECOG then con-

ducted a trial testing preoperative continuous 5-FU 1000 mg/m2/day on days 2 to 5 and 29 to 32

and mitomycin-C 10 mg/m2 on day 2 of a 50.4 Gy radiation schedule [46]. Of 53 patients, 41

underwent surgery; 17 did not benefit from resection because of local extension or metastases,

while 24 underwent resection and showed a median survival time of 15.7 months compared with

9.7 for the entire group.

More controversial is the ultimate role of downstaging neoadjuvant chemo radiation followed

by surgery for patients with initially unresectable tumors. Indeed, interpretation of the literature is

complicated by differences in respectability criteria. Moreover, it is argued that surgical compli-

cations may occur more frequently secondary to inflammation and fibrosis following chemo

radiation [44].

LOCALLY ADVANCED AND METASTATIC PANCREATIC CANCER

Being the main focus of the present report, this section will be further developed with a review

of the use of recent chemotherapy modalities in this largely predominant subgroup of pancreatic

cancer patients. General considerations about their management are however introduced first.

Advanced pancreatic carcinoma is a major challenge to oncologists because of its chemo

resistance. However, observation has long suggested that chemotherapy could improve survival

time in inoperable pancreatic carcinoma [47, 48]. Especially the first trial, by Mallinson et al., led

clinicians to test many chemotherapeutic drugs as single agents or in combination therapies in

advanced pancreatic tumors. Response rates were highly variable and often not reproducible.

Among chemotherapeutic agents, 5-fluorouracil (5-FU) has certainly been the most studied.

Observed response rates were variable and did not show any consistent benefit on either survival

Page 24: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 19 -

time or symptoms relief. Despite this lack of evidence in advanced pancreatic carcinoma man-

agement, 5-FU was the first chemotherapeutic agent considered as the treatment of choice for this

affection, since other molecules or combinations had failed to demonstrate any improvement in

response rates or survival time. Over the last decade however, gemcitabine, a pyrimidine antime-

tabolite, was shown to somewhat improve disease stabilization, clinical benefit response and sur-

vival time. After approval for use in the late 1990s, this agent is now considered as first choice

for treatment of advanced pancreatic cancer. These observations tended to boost clinical research

on combinations with other chemotherapeutic agents as well as radiation therapy. It is obvious

now that the introduction of gemcitabine did not fundamentally change the prognosis of advanced

pancreatic cancer in terms of classical endpoints such as survival time, and that its quantitative

benefit is only minor. However, before the approval of gemcitabine in this indication, chemother-

apy was considered as not very efficient and was therefore not systematically offered to patients.

Pioneer studies with gemcitabine, demonstrating significant benefits in terms of symptoms

improvements, changed the attitude of oncologists, as reflected by the exponential increase in the

literature about palliative management of advanced pancreatic cancer over the last decade. Che-

motherapy is now considered as an important therapeutic modality in this management, and an

intense clinical research activity is underway to determine the most efficient regimen.

In addition to chemotherapy, palliation by surgical or medical means is of primary importance:

when advanced disease is diagnosed. For example, biliary bypass may be indicated in patients

with obstructive jaundice, and duodenal bypass when gastric outlet obstruction is pending. For

the numerous patients whose predominant symptom is pain, use of long-acting opiate analgesics

may be indicated. When ineffective or not tolerated, intra operative, percutaneous or EUS-

directed celiac block may provide adequate and efficient pain relief [49]. Also, to compensate for

pancreatic exocrine failure, patients can be supplemented with enteric-coated pancreatic enzymes.

Page 25: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 20 -

CHEMOTHERAPY IN ADVANCED PANCREATIC CARCINOMA: LITERATURE REVIEW

This section systematically reviews the results of recently published studies regarding man-

agement of advanced pancreatic carcinoma. The respective references were obtained on Medline

or by consulting review articles [50-58]. General considerations about clinical endpoints are pre-

sented first.

METHODOLOGICAL CONSIDERATIONS

PERFORMANCE STATUS

The Karnofsky index [59] rates performance on a 0 to 100 scale in steps of 10 depending on

patient’s ability to perform normal activities and to do active work as well as on need for

assistance:

- 100: Normal, no complaints, no evidence of disease - 90: Able to carry on normal activities, minor signs of symptoms of disease - 80: Normal activity with effort, some signs or symptoms of disease - 70: Cares for self, unable to carry on normal activity or to do active work - 60: Requires occasional assistance, but able to care for most of his needs - 50: Requires considerable assistance and frequent medical care - 40: Disabled, requires special care and assistance - 30: Severely disabled, hospitalization indicated though death not imminent - 20: Very sick, hospitalization necessary, active supportive treatment necessary - 10: Moribund, fatal processes progressing rapidly - 0: Dead

An alternate categorization of performance status was then developed by the ECOG [60], pro-

viding a five-point scale easier to use and more precise and narrow in its message:

- 0: Normal activity - 1: Symptoms, but nearly ambulatory - 2: Some bed time, but needs to be in bed less than 50% of normal daytime - 3: Needs to be in bed more than 50% of normal daytime - 4: Unable to get out of bed

Intra- and inter observer correlations for the two scales were found to be very high [61] and

some authors even developed an equivalency table between them [62].

EVALUATION OF RESPONSE IN PANCREATIC CANCER

Response to chemotherapy is highly dependent on performance status and disease stage at ini-

tial diagnosis [63]. Thus, comparison between studies is difficult due to the variability in such

prognostic factors at the time of inclusion. Moreover, despite modern imaging techniques, it is

frequently impossible to get accurate localization and measurement of tumor extension within

Page 26: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 21 -

adjacent tissues because of the desmoplastic and local inflammatory reactions induced by the dis-

ease itself, making it particularly difficult to differentiate between tumor and normal surrounding

tissue [64]. Consequently, tumor response may be underestimated because reactive tissue would

not readily shrink during successful chemotherapy, or by contrast overestimated in case of faster

decrease of inflammatory reactions. Endoscopic ultrasound has been shown to be more sensitive

than CT scan for the assessment of the initial tumor and regional lymph nodes (i.e. staging), but

its performance is not better when it comes to the follow-up assessment of pancreatic parenchyma

[65, 66], a situation that might change with recent developments in three-dimensional linear

endoscopic ultrasound [67]. Hence, clinicians are facing two problems of tumor size definition at

the time of diagnosis and of imprecise estimation of tumor response after chemotherapy. There-

fore, conventional endpoints such as tumor response rates may well inadequately evaluate

response to treatment [64]. Unfortunately, the vast majority of studies considered stabilization of

disease as the primary goal of palliative treatment and focused on such outcomes as complete

response (disappearance of all known disease), partial response (50% or more decrease in tumor

size) and stable disease (between 50% decrease and 25% increase in tumor size), according to the

reduction, stabilization or increase of the primary tumor and/or its metastases as evidenced by

radiological findings (World Health Organization reporting principles for results of cancer treat-

ment [68]). From that perspective, it might be worth paying more attention to survival data,

namely median survival time and 1-year survival probability, the latter characterizing patients

who showed a prolonged treatment benefit and providing superior information about the

responder minority.

Importantly, specific attention was given in recent years to pain relief and improvement of

other disease-related symptoms, leading to the development of the Clinical Benefit Response

(CBR) notion. CBR is a measure of clinical improvement in terms of pain intensity and analgesic

consumption, performance status and weight change: a patient is considered as a responder if

rated positive for either of the primary measures of pain (i.e. reduction of more than 50% in pain

intensity or analgesics consumption) or performance status (i.e. improvement of KPS by more

than 20 points) for at least 4 weeks without a negative rating in the other parameters within 12

weeks, or if there is a weight gain of 7% above pretreatment baseline while the two primary

parameters remain stable [69]. CBR is now considered as a better indicator of therapeutic

efficacy in palliative settings in pancreatic cancer, as clinical experience has shown that small

reductions of tumor size, corresponding to stable disease from radiological criteria, may often

Page 27: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 22 -

lead to spectacular improvements of pain and abdominal discomfort symptoms. Indeed, because

of its proximity to the celiac ganglion and its propensity for early invasion of this structure, small

changes in size of a pancreatic adenocarcinoma can lead to significant relief of symptoms. Evi-

dence of the impact of gemcitabine on CBR is the main rationale, which supports its current

position as a first-choice treatment modality in advanced pancreatic cancer.

Regarding serum markers, the prognostic importance of CA 19-9 has been demonstrated for

adjuvant treatment of operable pancreatic cancer [70]. Stemmler et al. [71] evaluated patients

with advanced pancreatic carcinoma and initially elevated CA 19-9 levels, who were subse-

quently treated with a gemcitabine-cisplatin combination regimen. Interestingly, independent of

CT response evaluation, CA 19-9 responders survived significantly longer than CA 19-9 non-

responders (9.8 versus 5.8 months, P=0.022), suggesting that CA 19-9 serum kinetics may serve

as an early and reliable indicator of response and warranting further evaluation.

In the future, Positron Emission Tomography (PET) may be more sensitive than traditional

imaging as a technique to measure treatment response in pancreatic cancer [50]. Indeed, uptake

and phosphorylation of glucose is higher in tumor cells, making it possible to tell apart malignant

and nonmalignant tissue in the tumoral region.

5-FLUOROURACIL

Although many oncologists considered the thymidylate synthase inhibitor 5-FU as a standard

treatment prior to the introduction of gemcitabine, there was no consistent evidence to support its

benefit in patients with advanced pancreatic cancer in terms of disease response, survival time or

quality of life. Indeed, drug efficacy (bolus 5-FU alone or biomodulated by folinic acid or inter-

feron) measured by objective response rate was highly variable, as well as median survival time,

reported to range between 3.5 and 6.2 months in a recent review [55]. However, recent assess-

ment of high dose 24-hour infusion 5-FU 2600 mg/m2 and of capecitabine 2500 mg/m2 (oral

equivalent to 5-FU, which simulates a continuous infusion) showed median survival times of 8.3

months and 6.1 months, respectively, with low toxicity, suggesting some efficacy of 5-FU

administered intravenously at continuous high doses [72, 73].

Page 28: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 23 -

OTHER SINGLE-AGENT MODALITIES [55]

Topoisomerase I inhibitors: Irinotecan (or CPT-11) was associated with response rates of 9%

and a median survival time of 5.2 months [74]. Rubitecan or 9NC showed activity in previously

untreated patients with a median survival time of 7.3 months [75]. Exatecan showed an encourag-

ing activity with a median survival time of 10.6 months, and warrants further clinical evaluation

alone or in combination. Myelosuppression is the principal side effect of these drugs.

Taxanes: Both docetaxel (Taxotere®) and paclitaxel showed very modest activity in advanced

pancreatic cancer with low response rates and a median survival time of approximately 5 months

[76-79]. Jaundice and liver dysfunction is a clear limit in treatment with taxanes.

Platins: Cisplatin and oxaliplatin used alone are only modestly active, with published median

survival times of 3.4 to 4 months and response rates of 21% [80]. Combination with 5-FU yields

better results, with a median survival time between 4 and 11 months according to the regimen.

GEMCITABINE

INITIAL PHASE I-II SINGLE-AGENT STUDIES

Several initial phase I-II studies established gemcitabine as a promising molecule in the che-

motherapy of advanced pancreatic cancer, encouraging its subsequent evaluation in methodologi-

cally more robust trials. These protocols used 800-1500mg/m2 of gemcitabine on a weekly basis,

in either the conventional 4-week schedule (treatment on days 1, 8 and 15) or the initial 7-week

regimen (treatment continuously for 7 weeks with one week of rest, followed by the regular 4-

week schedule). Table 1 at the end of this section summarizes the details of these studies.

Although observed objective response rates were relatively low (4.3% to 18.2%), there were a

substantial number of patients who exhibited stable disease (18.8% to 50%). Excluding the trial

of Rothenberg et al. [81], which enrolled patients who were experiencing disease progression

under 5-FU therapy, the median overall survival time was 4.8 to 9.8 months, i.e. higher than the

typical 3-6 months survival time without treatment. 1-year survival ranged between 14.3% and

39%. Importantly, CBR was achieved in 26.6% to 48% of the patients, and toxicities were gener-

ally mild and manageable, with grade 3-4 mostly reversible hematological toxicities ranging from

6.1% to 27.3% (leucopenia), 6.6% to 36.4% (neutropenia), 0% to 18% (thrombocytopenia) and

3% to 22% (anemia). Other toxicities were mild, with nausea and vomiting, fever, rash and

transaminase elevation most commonly reported.

Page 29: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 24 -

Following these encouraging results, other administration modalities of gemcitabine were

evaluated, for instance a high-dose infusion (2200 mg/m2) [82] and a fixed dose-rate infusion

regimen (10 mg/min/m2) [83]. These alternatives to the conventional gemcitabine administration

modalities are still undergoing clinical validation and are not considered as standard yet.

PHASE III SINGLE-AGENT STUDIES

Subsequently, single-agent gemcitabine regimens were evaluated in randomized phase III trials,

six of which are detailed in Table 2 at the end of this section, as reviewed by Fung et al. [51].

The pioneer trial by Burris et al. [84] demonstrated superiority of gemcitabine over 5-FU as a

first-line therapy in patients with locally advanced or metastatic pancreatic carcinoma: 23.8% of

patients in the gemcitabine arm achieved CBR compared to 4.8% in the 5-FU arm (P=0.002).

Median survival time was 5.65 months versus 4.41 (P=0.003), and 1-year survival probability

18% versus 2%. The rather low values for median survival time have to be considered in the

context of unfavorable eligibility criteria.

Several other randomized trials later compared gemcitabine to newer therapeutic classes in

pancreatic cancer treatment: the matrix metallo proteinase inhibitor marimastat [85] and Bay 12-

9566 [86]; ZD9331, a novel antifolate inhibitor of thymidylate synthase [87]; more recently the

farnesyl transferase inhibitor tipifarnib [88]; and exatecan [89]. Single-agent gemcitabine

systematically demonstrated its superiority to the other molecules.

ASSOCIATION REGIMENS

Considering that bi-therapies are often more effective than single-agent regimens in oncology,

gemcitabine was also evaluated in combination with other chemotherapeutic molecules:

• Gemcitabine and 5-FU

After in vitro demonstration of synergistic cytotoxicity [90] and encouraging initial evaluation

in phase I-II trials [91, 92] (reviewed by Oettle et al. [93]), a randomized phase III study by the

ECOG recently failed to show any significant advantage of regimens combining gemcitabine and

5-FU over gemcitabine alone [94, 95].

• Gemcitabine and cisplatin

After in vitro observation of synergy in several other solid tumors [96] and encouraging early-

phase trials [97, 98], a recent randomized phase III study failed to show any significant difference

in terms of survival time between gemcitabine alone and in combination with cisplatin [99], how-

ever being of small statistical power (97 patients).

Page 30: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 25 -

• Gemcitabine and oxaliplatin (GemOx regimen)

After in vitro observation of a sequence-dependent synergy with gemcitabine [100] and

encouraging initial clinical trials [101, 102], a recent GERCOR/GISCAD study presented at the

2004 ASCO meeting failed to reach statistical significance for the GemOx arm in comparison to

single-agent gemcitabine in terms of overall survival time [103]. However, the GemOx arm evi-

denced significantly better response rate, clinical benefit and progression-free survival time, thus

warranting further evaluation in trials designed with a higher statistical power.

• Gemcitabine and irinotecan (IrinoGem regimen)

Early-phase evaluation of this regimen evidenced only poor results [104, 105].

• Gemcitabine and docetaxel

Early-phase evaluation of this regimen evidenced only poor results [106-110].

• Gemcitabine and epirubicin

Early-phase evaluation of this regimen evidenced only poor results [111, 112].

During the last annual meeting of the American Society of Clinical Oncology, disappointing

results were also presented for gemcitabine in combination with exatecan [113] or pemetrexed

[114]. Hence, none of the above-mentioned association regimens demonstrated its superior-

ity to single-agent gemcitabine in terms of overall survival time so far. The results of two

randomized phase III trials enrolling more than 300 patients and evaluating gemcitabine in com-

bination with capecitabine versus gemcitabine alone are expected in the early future (SAKK and

MRC protocols).

GEMCITABINE IN RADIO-CHEMOTHERAPEUTIC MODALITIES

Gemcitabine demonstrated very potent radio sensitizing properties in preclinical studies [115,

116], and radiation enhancement is achieved by using much lower doses than those resulting in

cell kill. Gemcitabine-mediated radio sensitization is probably related to a concurrent disruption

of deoxynucleotide pools (inducing an inhibition of radio-induced DNA damage repair) and a

subsequent redistribution of cells into the radiosensitive S phase of the cell cycle [117, 118]. A

few disparate trials have been reported, with varying objectives from palliative to curative,

neoadjuvant or adjuvant use [51]. Among the variety of regimens that were evaluated, response

rates varied from 16.7% to 57.1%, while toxicities were generally manageable with appropriately

adapted doses of gemcitabine.

Page 31: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 26 -

Several other trials evaluated more aggressive association regimens with radiation, for exam-

ple gemcitabine and 5-FU in protracted intravenous infusion [119, 120], gemcitabine and cis-

platin or gemcitabine and paclitaxel [121, 122]. Given the contrasting efficacy results of those

early-phase trials, larger phase III studies will be necessary to determine the precise role of

chemo radiation therapy in patients with locally advanced pancreatic cancer.

SUMMARY

Overall, these data show that the introduction of gemcitabine has only moderately benefited

patients in terms of survival time, but much more significantly in terms of CBR. Although

numerous combination regimens have undergone clinical testing, none has demonstrated its supe-

riority to gemcitabine alone in any randomized phase III study so far. Available evidence indi-

cates that remission rates of 10% to 30%, CBR of 40% to 60%, overall survival times of 7 to 10

months and 1-year survival probabilities of 20% to 30% may be currently expected from chemo-

therapy in advanced pancreatic carcinoma with regimens based on single-agent gemcitabine [53].

This chemotherapeutic modality should currently be considered as the standard treatment outside

any experimental protocol, until future randomized phase III trials establish the superiority of

another drug or combination regimen.

Page 32: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 27 -

Table 1 – Single-agent studies of gemcitabine in advanced pancreatic cancer

Toxicity

Hematological (G3/4) (%)

First author [Ref]

Design of study

(Phase)

Number of patients

Regimen

(Drug dose)

(mg/m

2)

Partial response (%)

Stable disease (%)

Disease stabilization

(PR + SD)

Clinical Benefit

Response (%)

1-year survival (%)

Time to progression

(months)

Median survival time

(months)

Leucocytes

Neutrophils

Thrombocytes

Anemia

Others

Karasek 2003 [123] Prospective (observational)

100 1000 x q3 of 4 13 48 61 26 NA 3.2 7.5 NA 8 3 5 G3/4 not observed

Crino 2001 [124] Prospective (II) 33 1000 x 7 wk, then q3 of 4 wk 12.1 45.5 57.6 NA NA NA 7.7 9 NA 12 12

(G2/G3)

Nausea/vomiting G1/2 (24%/9%); G1/2 skin rash (9%); Peripheral edema (24%); Asthenia (39%); G1 alopecia

(3%)

Meyer 2001 [125] Prospective (II) 28 1000 x 7 wk, then q3 of 4 wk 7.2 39.2 46.4 NA 14.3 NA 9.8 11 NA 18 8 G3: Diarrhea (7%); Nausea (7%); Edema (4%); Alopecia

(20%)

Okada 2001 [126] Prospective (I) 11 (a) 1000 x q3 of 4 wk

(b) 1000 x 7 wk, then q3 of 4 wk

18.2 36.4 54.6 28.6 18.2 NA 6.4 27.3 36.4 0 9.1 G3/4 anorexia (27.3%); G3: Nausea/vomiting (9.1%); Fatigue (18.2%); GOT/GPT (9.1%); Weight loss (9.1%)

Aykan 2000 [127] Prospective (II) 14 1000 x 7 wk, then q3 of 4 wk 14.3 50 64.3 NA 14.3 6 6 NA NA NA NA No G3/4 toxicity

Kurtz 1999 [128] Prospective

(compassionate) 74 1000 x 7 wk, then q3 of 4 wk 4.3 39.1 43.4 48 NA NA 5 NA 27 14 22

G3/4: Asthenia (11.6%); Nausea/vomiting (9.3%); Anorexia (7%); (Partial) occlusion (7%); Edema (2.3%); Cardiac insufficiency (2.3%); Embolization of catheter (2.3%); Arterial hypotension (2.3%); Jaundice (2.3%);

Dehydration (2.3%); Abdominal pain (2.3%)

Storniolo 1999 [63] Prospective

(Investigational New Drug)

3023 1000 x 7 wk, then q3 of 4 wk 10.6 18.4 15 2.7 4.8 NA NA NA NA

Fever (7.3%); Pain (6.8&); Asthenia (6%); Abdominal pain (5.5%); Dyspnea (5%); Dehydration (4.5%); Nausea/vomiting (3.9%); Anorexia (3.6%); Deep thrombophlebitis (3.2%); Jaundice (3.2%); Ascites

(2.9%); Edema (2.9%); Sepsis (2.4%) Manzano 1998

[129] Prospective (II) 15 1000 x 7 wk, then q3 of 4 wk NA NA NA 26.6 39 NA NA NA 6.6 NA NA G4 asthenia (6.6%)

Petrovic 1998 [130] Prospective (II) 11 1000 x 7 wk, then q3 of 4 wk NA NA NA 33 26 NA 6.65 NA NA NA NA Well tolerated

Carmichael 1996 [131]

Prospective (II) 34 800 x q3 of 4 wk (max 1000) 6.3 18.8 25.1 NA NA NA 6.3 6.1 24.5 9.1 3

G3: Nausea/vomiting (26.7%); Diarrhea (3.3%); Pain (3.3%); Consciousness alteration (3.3%)

G2: Cutaneous (10%); Fever (20%); Infection (6.7%); Nausea/vomiting (20%); Consciousness alteration

(13.3%)

Rothenberg 1996 [81]

Prospective (II) 74 1000 x 7 wk, then q3 of 4 wk

(max 1250) 10.5 29.8 40.3 27 4 2.53 3.85 9.8 26.2 4.9 11.4

G4: Hemorrhage (1.6%); Vomiting (1.6%) G2/3: Constipation (4.8%/1.6%); Cutaneous (9.5%/0%); Fever (14.3%/1.6%); Alopecia (1.6%/0%); Infection (0%/3.2%); Nausea/vomiting (28.6%/6.3%); Oral mucositis (1.6%/0%); Pain (3.2%/1.6%); Peripheral neuropathy (1.6%/0%); Pulmonary (1.6%/0%);

Consciousness alteration (1.6%/3.2%)

Casper 1994 [132] Prospective (II) 44 800 x q3 of 4 wk (max 1500) 11 31.8 42.8 NA 23 3.7 5.6 NA NA NA NA Mild to moderate flu-like syndrome: 100% Mild hemolytic-uremic syndrome: 1pt

Page 33: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 28 -

Table 2 – Randomized gemcitabine studies in advanced pancreatic cancer (underlined results correspond to a statistically significant difference between arms)

Toxicity

Hematological (G3/4) (%)

First author [Ref]

Design of study

(Phase)

Number of patients

Regimen

(Drug dose)

(mg/m

2)

Partial response (%)

(incl. complete resp.)

Stable disease (%)

Disease stabilization

(PR + SD)

Clinical Benefit

Response (%)

1-year survival (%)

Time to progression

(months)

Median survival time

(months)

Leucocytes

Neutrophils

Thrombocytes

Anemia

Others

139 Gemcitabine 1000 x 7 wk, then q3 of 4

wk 4.3 44.6 48.9 NA 25 3.5 6.6 NA 34.5 6.5 10.1 G3/4: Nausea (3.6%); Vomiting (5%); Lethargy (5.8%)

Moore 2003 [86]

Randomized (III)

138 Bay-12-9566 800 mg p.o. b.i.d. 0.7 22.5 23.2 NA 10 1.7 3.7 NA 2.2 0 10.1 G3/4: Nausea (8%); Vomiting (2.9%); Lethargy (6.5%)

163 Gemcitabine 1000 x q3 of 4 wk 5.6 NA NA NA NA 2.2 5.4 16 5 11 10 G3/4: Diarrhea (4%); Nausea/vomiting (19%)

Gemcitabine 1000 x q3 of 4 wk

Berlin 2002 [95]

Randomized (III)

164 + 5-FU 600 x q3 of 4 wk

6.9 NA NA NA NA 3.4 6.7 29 7 19 10 G3/4: Diarrhea (10%); Nausea/vomiting (15%)

44 Gemcitabine 1000 x 7 wk, then q3 of 4

wk 9.2 33.4 42.6 49 NA 1.9 4.7 4 5 2 4

G3/4: Mucositis (2%); Diarrhea (0%); Nausea/vomiting (2%)

Gemcitabine 1000 x 7 wk, then q3 of 4 wk

Colucci 2002 [99]

Randomized (III)

53 + Cisplatin 25 day 1 1h before gem.

26.4 30.2 56.6 52.6 NA 4.7 7 4 9 2 6 G3/4: Mucositis (0%); Diarrhea (4%); Nausea/vomiting

(2%)

103 Gemcitabine 1000 x 7 wk, then q3 of 4

wk 26 NA NA NA 19 3.8 5.6 2 7 5 2

G3/4: Cardiac dysfunction (1%); Constipation (1%); Fever without infection (1%); Infection (2%); Local (1%); Nausea (4%); Cortical (5%); Motor (4%);

Pulmonary (9%); Skin (3%)

104 Marimastat 5 mg b.i.d. 2.8 NA NA NA 14 1.8 3.7 0 1 3 4 G3/4: Cardiac dysrhythmias (1%); Diarrhea (1%);

Musculoskeletal (7%); Nausea (2%); Cerebellar (2%); Vision (1%); pulmonary (3%); weight gain or loss (1%)

105 Marimastat 10 mg b.i.d. 2.8 NA NA NA 14 2 3.5 0 0 0 3 G3/4: Musculoskeletal (7%); Nausea (1%); Motor (1%);

Vomiting (1%)

Bramhall 2001 [85]

Randomized (III)

102 Marimastat 25 mg b.i.d. 2.8 NA NA NA 20 1.9 4.2 0 0 2 2 G3/4: Musculoskeletal (12%); Nausea (1%); Motor

(1%); Pulmonary (1%)

54 Gemcitabine 1000 x q3 of 4 wk NA NA NA NA NA NA NA 5.5 NA 7.4 9.2 G3/4: Mucositis (3.7%); Diarrhea (7.4%)

G3: Nausea/vomiting (3.7%) Gemcitabine 1000 x q3 of 4 wk

Heinemann 2001 [133]

Randomized (III)

46 + Cisplatin 50 (day 1, 15) q4 wk

NA NA NA NA NA NA NA 13 NA 8.6 13 G3/4: Mucositis (4.3%); Diarrhea (10.8%)

G3: Nausea/vomiting (33%)

63 Gemcitabine 1000 x 7 wk, then q3 of 4

wk (max 1250) 5.4 39 44.4 23.8 18 2.1 5.65 9.7 25.9 9.7 9.7

G3/4: Nausea/vomiting (12-7%) G3: Diarrhea (1.6%); constipation (3.2%); consciousness

alteration (1.6%); Pain (1.6%)

Burris 1997 [84]

Randomized (III)

63 5-FU 600 weekly 0 19 19 4.8 2 0.9 4.41 1.6 4.9 1.6 0 G3: Nausea/vomiting (4.8%); Diarrhea (4.8%);

Constipation (1.6%); Consciousness alteration (1.6%)

Page 34: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 29 -

RETROSPECTIVE STUDY ON A GENEVA COHORT

INTRODUCTION

Although not having revolutionarily improved classical therapeutic outcomes such as overall

survival time in patients suffering from advanced pancreatic cancer, gemcitabine significantly

improved more qualitative outcomes such as clinical benefit response, which is often physicians’

predominant concern in a palliative context. In parallel, it also markedly stimulated clinical

research.

Numerous combination regimens based on gemcitabine have been evaluated so far in early

clinical trials, and promising ones are now undergoing further assessment in randomized phase III

studies. However, for obvious methodological reasons, the experimental situation of those trials

is often rather different from daily clinical practice, both in terms of patient homogeneity and of

therapeutic algorithm rigidity. Therefore, and because introduction of gemcitabine in the mid-90s

was a cornerstone in chemotherapeutic management of advanced pancreatic cancer, it seemed to

us interesting to retrospectively analyze a Geneva cohort of patients suffering from this

malignancy who were followed in the cantonal hospital over the last decade, considering out-

comes such as overall survival time and 1-year survival probability. Our baseline hypothesis was

that the more organized and systematic delivery of chemotherapy after the above-mentioned

introduction of gemcitabine would have improved those outcomes in comparison to the pre-

ceding period, when chemotherapy prescription for advanced pancreatic cancer was poorly stan-

dardized. Besides, other efficient medications for the management of advanced pancreatic cancer,

such as oxaliplatin, were introduced following gemcitabine, and may also have contributed to

further improve those outcomes (e.g., in second or third-line therapies).

Economic concerns have a growing importance in healthcare-related discussions and decisions

in most developed countries as the sector’s share of their gross national product is rising:

clinicians are nowadays under increasing pressure to adopt more cost-effective treatment

practices as a result of initiatives taken by major third-party payers, government and business. For

example, in the Swiss hospital sector and following other European countries like Germany, there

has recently been a movement towards adopting prospective payment systems based on

diagnosis-related groups (DRGs) for the financial retribution of health care providers.

Page 35: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 30 -

Although representing less than 1% of the population, physicians determine, through their

decisions about their patients’ care, how nearly 10% of gross national product is being spent.

Considerations of justice in macro allocation of scarce resources raise the question whether pro-

longing survival time by only a few months (e.g., in some oncology situations) can be considered

as ethically justified with regard to society as a whole when incurred costs to achieve such results

are enormously high. But because of methodological as well as practical hurdles, precise deter-

mination of incurred costs for a given diagnostic or therapeutic option in a given clinical setting

has historically proven to be considerably difficult. Accordingly, cost-effectiveness and cost-

benefit studies are still rare in comparison to the wealth of publications considering clinical

aspects of medicine. In particular, these studies were sometimes criticized for their low practical

pertinence, given their often strongly rigorous experimental context. Therefore, it also seemed

interesting to us to retrospectively conduct basic economical analyses in the same cohort of

patients, i.e. mainly an assessment of costs.

METHODOLOGY

PATIENTS

References of all patients eligible for this retrospective study were retrieved by data mining

the local electronic Oncosurgery database in the Geneva Cantonal Hospital. All pancreatic ade-

nocarcinoma cases that were evaluated at least once by a practitioner belonging to this service

from October 1st, 1993 to October 31st, 2003 were considered as being eligible. The final cohort

was determined after exclusion of those patients who did not undergo at least one antitumoral

therapy under the responsibility of the service, i.e. of all patients who were evaluated only and

who underwent therapy outside of the cantonal hospital.

DATA MINING

First, we gathered epidemiological data from the Geneva Cancer Registry, which systemati-

cally records every cancer case among residents and non-residents, whether it be diagnosed in the

public or the private setting, from hospital databases, state laboratories and independent practitio-

ner declarations as well as death certificates, among others. The total number of pancreatic carci-

nomas declared in the Geneva canton from 1993 to 2003 was thus determined, so as to appreciate

the representativeness of the studied sample.

Page 36: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 31 -

For each patient included in the study, the following information –when available– were then

collected from paper and electronic medical records:

- Sex;

- Birth date;

- Death date or censoring date;

- Initial diagnosis date, defined so as to minimize as much as possible potential biases in

survival time estimates and to allow for the maximal homogeneity of included patients:

the date retained was the first day of the hospital stay leading to the diagnosis or its

confirmation;

- Known prognostic factors, including ECOG PS at the time of diagnosis, defined as the

score mentioned in the record of the first oncosurgery consultation; tumor stage (TNM

classification) at the time of diagnosis, re-interpreted according to the new 2002 classi-

fication system from initial radiological, surgical and pathological records; primary site

at the time of diagnosis (head versus body and/or tail); and tumor size at the time of

diagnosis, defined as the size in centimeters of the tumoral maximal axis mentioned on

CT-Scan reports;

- Treatment, including the first modality (curative surgery or chemotherapy), as well as

subsequent radiotherapy and/or chemotherapy modalities. For chemotherapeutic proce-

dures, the number of effectively administered lines and cycles of each drug or drug

association as well as the doses were collected;

- Hospitalizations, including dates of entry and discharge, department and nature of stay

(categorized as treatment, complication of treatment, complication of disease and

others);

- Economic data available from the hospital’s Medico-Economic Department and corre-

sponding to the costs for the institution of every hospitalization and ambulatory visit to

the cantonal hospital for every included patient from the diagnosis of the disease to

death or censoring (cf. further explanations about the detailed cost accounting proce-

dures of the Geneva Cantonal Hospital after the following section).

Page 37: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 32 -

DATA ANALYSIS

Group definition

For each and every patient included, the following parameters were then calculated when the

underlying information was available:

- Age at diagnosis, calculated by subtracting the birth date from the retained diagnosis

date;

- Survival time, calculated by subtracting the retained diagnosis date from the death or

censoring date. The date of October 31, 2003 was chosen as censoring date for patients

still alive at the time of analysis.

Then, two groups were defined on a chronological basis: patients included in the study were

ranked according to the date of their initial diagnosis and their first-intent chemotherapeutic

treatment was then considered (generally following diagnosis for stage III-IV patients and fol-

lowing recurrence for stage I-II resected patients), namely gemcitabine-containing regimen ver-

sus any other regimen. The diagnosis date for the first patient who received gemcitabine as a

first-line therapy to manage his disease was used to distinguish between the two groups. Thus,

group A comprised patients who did not receive gemcitabine as a first-line therapy, whereas all

patients in group B received this drug first. In this process, we did not consider any chemotherapy

regimen administered in combination with radiotherapy. We expected this date to occur some-

where in late 1998 according to the introduction of gemcitabine in Switzerland for this indication

and its official reimbursement by health insurances. Although a few patients categorized in group

A would inevitably prove to have received gemcitabine at some point of their later therapeutic

management, by definition it was not as first-intent therapy.

Following this step, statistic analyses for various parameters and outcomes were conducted.

Chi-square tests were used for binomial comparisons (a Fisher’s exact test was employed when

the number of patients under a given condition was inferior to five), as well as nonparametric

Mann-Whitney tests for ordered numerical data such as age. Two-sided p-values were systemati-

cally calculated, and a level of 0.05 was considered as statistically significant for their interpreta-

tion. Concerning survival analyses as well as cost comparisons, graphs were constructed accord-

ing to the method developed by Kaplan and Meier [134], and log-rank tests were used to compare

groups. SPSS 11.0 and GraphPad Prism 3 were used to compute those calculations.

Page 38: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 33 -

Medical analyses

The following analyses were conducted first:

- Comparisons for demographic factors, mainly sex;

- Comparisons for known prognostic factors, i.e. stage, tumor size and age at initial

diagnosis;

- Comparisons of treatment modalities and especially of the total number of chemother-

apy doses received.

At this point, our first intention was to compare the two groups in terms of total survival time

(i.e. from initial diagnosis to death or censoring), including patients diagnosed at early stages and

resected as well as patients diagnosed at later stages and directly treated in a palliative setting.

However, as will be presented in the results section, it turned out that groups A and B were sig-

nificantly inhomogeneous with respect to the proportion of stage I-II versus stage III-IV patients,

group A including significantly more early stages. Therefore, any global survival comparison

would have been inevitably biased by this difference in favor of group A, since it is well known

that resected patients have a longer survival time than patients diagnosed at later stages and have

different tumor biology. A potential solution to this issue would have been to compute survival

time in a palliative setting for resected patients, i.e. from the time of their recurrence. However,

precise definition of the date of recurrence is problematic: since survival time is so short for

patients suffering from advanced pancreatic cancer, it is crucial to be as systematic as possible in

such a definition to avoid important biases and analyze meaningful data. Since there was no sys-

tematic and standardized follow-up algorithm for these patients after resection in terms of radio-

logical examinations and biologic analyses in Geneva during the considered period, retaining as

the date of recurrence the date of a CT-Scan evidencing loco regional recurrence or metastases,

the date of a serum CA 19-9 elevation or the date of a consultation whose record concludes to the

recurrence would all have jeopardized our results, being far too imprecise. Therefore, we decided

to restrict the main analyses to stage III-IV patients only, i.e. to patients initially diagnosed at

advanced stages and directly treated in a palliative setting. From a clinical viewpoint, this restric-

tion makes much more sense in that we ended up comparing homogeneous populations, with

diagnosis dates as precisely defined as could be in a retrospective design. For indicative purposes,

demographic figures regarding the whole cohort will be provided in the results section.

Page 39: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 34 -

The following analyses were then conducted in the cohort restricted to stage III-IV patients:

- Primary endpoint: survival analysis;

- Secondary endpoints: comparison of the two groups in terms of hospitalization days,

distinguishing hospitalizations for treatment (e.g. permanent venous access implanta-

tion, chemotherapy or radiotherapy), complications of treatment (e.g. post-chemother-

apy fever with or without infection, severe vomiting or other treatment toxicity), com-

plications of disease (e.g. obstructive jaundice or ileus requiring surgical or interven-

tional radiological derivation procedures, cholangitis episodes and ultimately terminal

care and death) and other causes. The first hospitalization, leading to or confirming the

diagnosis, as well as the subsequent convalescence, were systematically considered as

treatment. Finally, total incurred economic costs for the hospital were compared using

log-rank tests to account for censored patients.

Economical analyses

• Cost analysis system in Geneva Cantonal Hospital

As far as economic analyses and cost comparisons are concerned, some explanations about the

method employed for costs computations using the cost accounting system used in the Geneva

Cantonal Hospital are detailed below. The fundamental problem in cost accounting is to link

costs actually incurred to a given unit of measurement –generally a product or service–, such as a

particular stay for a particular patient due to a particular pathology. This process, known as allo-

cation, is far more complex than it seems at first glance, since costs such as administrative,

maintenance, electricity or research and teaching expenditures –also known as indirect costs–

need to be allocated to each unit of output according to specific criteria since they are not directly

attributable to any given particular product or service. Indeed, although less critical than the

direct costs of medications, medical consultations or radiological procedures, for example, which

can all be related quite easily to a particular service, those indirect costs are at least as important

to consider when the true consumption of resources driven by a particular output are to be deter-

mined. Thus, the problem is to choose the least bad allocation method, knowing that it will

inevitably be wrong. Actually, true costs can never be known, although every organization needs

to understand as precisely as possible the mechanisms of consumption of resources used to

deliver its products/services. The process in the Geneva Cantonal Hospital can be summarized as

follows:

Page 40: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 35 -

1. Financial data is processed to obtain information about true expenses incurred by the

consumption of individual resources (e.g. salary costs, electricity expenses, medica-

tions etc.) during a particular period;

2. Those expenses are then related to specific basic activities of the hospital to yield spe-

cific costs (e.g. the specific overall cost of radiological procedures or of medical care).

This step involves building a kind of virtual hospital, whose structure corresponds to

its basic activities;

3. Direct costs are linked directly to each type of service provided, while indirect costs

need to be allocated to different activities to yield a complete unitary cost for each

service (e.g. laboratory services, radiological exams, nurse and medical care). The

complete unitary cost includes all direct costs for the service provided (e.g. a medical

consultation, a particular radiological investigation or medications) as well as a given

proportion of all support services, whose costs are indirect (e.g. maintenance, research

and teaching expenses, electricity etc.);

4. From the previous step, detailed unitary costs for various units of output are computed,

such as the cost of one hour of hospitalization, the cost of one ambulatory visit to a

particular department or the cost of one unit of laboratory or radiological procedure.

Some of those data (such as the cost of one hospitalization day in a particular depart-

ment) are used for benchmarking and financing purposes;

5. More importantly for our discussion, the detailed costs for each particular case can be

further inferred from the detailed services provided in each care situation (listed as a

result of the coding process at the end of each stay for each patient) as well as from

previously calculated unitary costs. For example, if a patient has undergone the

implantation of a permanent venous access device as well as the administration of two

cycles of chemotherapy during a given stay, those services can be linked to their corre-

sponding unitary costs to yield the total cost of the stay.

• Conversion into standard units

Our intention was to compare costs incurred for patients who were hospitalized at different

periods in time, spanning a decade, which is considerable from an economic perspective. Costs

incurred for one hour of medical consultation in 1995, for example, are in no way directly com-

parable to the costs incurred for the same service in 2003: this is partly due to general inflation,

Page 41: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 36 -

but more importantly to increases in general health care costs over the last decade. To overcome

this problem, a weighting factor was calculated to express each cost in 2002 units. 2002 was

chosen because most of the data for 2003 were not yet available at the time of analysis. To com-

pute those multiplication factors, the average cost of one typical hospitalization day in a general

care unit of the Geneva Cantonal Hospital was considered, in each year previous to 2002, with

respect to this reference period and based on the same database available from the Medico-

Economic Department. The average cost was obtained by dividing the total expenses incurred for

general care units by the total number of hospitalization days in the same units. Comparison to

2002 yielded several weighting factors, by which every cost incurred previous to this reference

year was multiplied to obtain standardized values. This approach was deemed reasonable, since

most hospitalizations of patients suffering from a palliative-stage pancreatic cancer occur in

general care units.

• Extra-hospital costs

Costs incurred during rehabilitation stays outside of the cantonal hospital, i.e. mainly conva-

lescence stays, as well as costs of terminal care stays, were also included in the analysis. Consid-

ering that every convalescence stay is more or less comparable to another, in that it rarely

includes specific and expensive surgical or radiological procedures, but only basic clinical man-

agement, a typical cost was calculated for one day of hospitalization in a convalescence hospital

whose financial statements and activity report were available for 2002 (Clinique Genevoise de

Montana). This value was then extrapolated to stays in other hospitals after correction for the

year with respect to 2002. Total estimated costs for each stay were calculated simply by multi-

plying this unitary per diem value by the length of the stay.

Last, the majority of patients included were followed almost exclusively by the oncology

service during most of their disease. Hence, it is unlikely that major expenses were incurred out-

side this consultation. Consequently, and due to obvious methodological issues, extra-hospital

costs were not considered in our medico-economic analyses.

Page 42: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 37 -

RESULTS

EPIDEMIOLOGICAL DATA

The following pathologic variants of pancreatic cancer were considered when retrieving

information from the Geneva Cancer Registry (reference [135] was used until 1994 and reference

[136] from 1995 to 2002):

- Neoplasm, malignant

- Carcinoma, NOS

- Carcinoma, anaplastic type, NOS

- Adenocarcinoma, NOS

- Tubular adenocarcinoma

- Papillary adenocarcinoma, NOS

- Mucoepidermoid carcinoma

- Cystadenocarcinoma, NOS

- Mucinous adenocarcinoma

- Mucin-producing adenocarcinoma

- Infiltrating duct carcinoma

- Acinar cell carcinoma

- Adenosquamous carcinoma

- No microscopic confirmation; clinically malignant tumor

According to information available from the Registry, the incidence of pancreatic carcinoma

in the canton of Geneva was 13.3/100,000 newly diagnosed cases per year by men and

12.3/100,000 by women (standardized rates, Geneva population), or 12.7/100,000 by men and

8.3/100,000 by women (standardized rates, European population) from 1993 to 2001. 529 cases

of pancreatic carcinoma were diagnosed between January 1st, 1993 and December 31st, 2001 in

the canton of Geneva (about 400’000 inhabitants). 462 cases were residents in the canton at the

time of diagnosis and 67 cases were non-residents. Data for 2002 and 2003 were not available at

the time of analysis. Among these 529 cases, 51.8% were classified as adenocarcinoma, not oth-

erwise specified, and 31.9% as clinically malignant tumors without microscopic confirmation.

The breakdown by year is shown in the following table:

Page 43: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 38 -

Year Residents Non-Residents

1993 56 7 1994 62 12 1995 40 5 1996 47 4 1997 43 4 1998 57 5 1999 45 12 2000 56 9 2001 56 9 Total 462 67

Table 3: Number of pancreatic carcinoma cases diagnosed in Geneva from 1993 to 2001

PATIENTS AND GROUPS

Our research on the Oncosurgery database in the cantonal hospital yielded a total of 86

patients who were followed by the service for a pancreatic adenocarcinoma during at least one

treatment cycle following the diagnosis of their disease from October 1st, 1993 to October 31st,

2003. Of these, two patients had to be excluded, one because his pancreatic tumor never recurred

after initial curative-intent surgery (and we intended to consider treatment at a palliative stage),

and one because his tumor was actually not of pancreatic origin, as evidenced by pathology

reports (it was an ampulla of Vater carcinoma). Thus, the final cohort consisted of 84 patients,

who were subsequently ranked according to the date of diagnosis. The pivotal date retained to

distinguish between group A and B was December 22, 1998, corresponding approximately to the

generalization of gemcitabine use in Switzerland (second half of 1998) in this indication.

Accordingly, a Group A, consisting of 39 patients, and a Group B, consisting of 45 patients,

were defined. The following table details the percentage of newly diagnosed cases of pancreatic

carcinoma in Geneva followed in Oncosurgery in the cantonal hospital, and included in the pre-

sent study. 1993 was not included in the total percentage calculation, since the starting point for

inclusion was only October of this year:

Page 44: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 39 -

Year

Number of cases diagnosed

and followed-up in

Oncosurgery

Corresponding percentage

of total diagnosed cases in

Geneva

1993 (October onwards) 1 2% 1994 11 15% 1995 6 13% 1996 5 10% 1997 7 15% 1998 9 15% 1999 8 14% 2000 12 18% 2001 16 25% 2002 6 NA 2003 2 NA

Total (1994-2001) 74 16% Table 4: Percentage of pancreatic cancer patients diagnosed in Geneva and followed-up in Oncosurgery in

the Cantonal Hospital

Page 45: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 40 -

Demographic and prognostic characteristics of the two groups are shown below:

Parameter Value Group A (n=39) Group B (n=45) Statistical Test p-value

F 16 (41%) 15 (33%) Sex M 23 (59%) 30 (67%)

Chi-Square 0.47

I 3 (8%) - (0%) II 14 (36%) 9 (21%) III 7 (18%) 13 (30%) IV 15 (39%) 22 (50%)

Stage at

diagnosis

Missing - 1

Mann-Whitney 0.06

Head 25 (68%) 29 (64%) Body/Tail 12 (32%) 16 (36%) Primary Site

at diagnosis Missing 2 - Chi-Square 0.77

Range 1.5-6 1.5-7 Median 4 4 Tumor Size at

diagnosis (cm) Missing 12 7 Mann-Whitney 0.93

Range 41-78 44-79 Median 60 66 25th

Percentile 50 60

Age at

diagnosis 75th

Percentile 68 72

Mann-Whitney 0.03

0 10 (39%) 7 (27%) 1 11 (42%) 13 (50%) 2 5 (19%) 5 (19%) 3 - (0%) 1 (4%)

ECOG PS at

diagnosis

Missing 13 19

Mann-Whitney 0.41

Curative Surgery (Stages I-II)

17 (44%) 10 (22%) Initial

Treatment Palliative (Stages III-IV)

22 (56%) 35 (78%)

Chi-Square 0.04

Table 5: Patients characteristics

As evidenced in Table 5, the two groups were roughly comparable in terms of demographic

factors such as sex (p=0.47), but not in terms of prognostic factors: indeed, while primary site,

tumor size and ECOG PS at initial diagnosis were quite homogeneous between the two popula-

tions (p=0.78, 0.93 and 0.41, respectively), age as well as stage at initial diagnosis differed sig-

nificantly (p=0.03 and 0.06, respectively, this latter result being at the significance limit). Ageing

is usually considered as a general prognostic factor for oncologic diseases, in part according to

the assumption that co morbidities generally increase with advancing age. Histogram graphs with

a theoretical normal curve as well as box plots for age at diagnosis are reproduced below for the

two groups:

Page 46: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 41 -

Figure 1: Histogram graph for age at diagnosis (Group A)

Figure 2: Histogram graph for age at diagnosis (Group B)

Page 47: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 42 -

Figure 3: Box plot for age at diagnosis (total cohort)

More importantly, and as mentioned in the methodology section, it appeared that the propor-

tion of patients diagnosed at early stages, when a curative first-intent therapy could still be

offered (stages I and II), was higher in group A than in group B. Indeed, considering together

stages I-II versus stages III-IV, proportions were respectively 44%/56% for group A versus

22%/78% for group B (p=0.04). As evidenced in the preceding literature review, it is well known

that patients who can be offered a curative-intent pancreatic resection have a longer survival time

than those whose tumor is diagnosed at a palliative stage, and who can be offered chemothera-

peutic management only. Tumor biology, in terms of clinical evolution and response to treatment,

is clearly different between the two situations. Thus, the two groups initially defined were not

comparable because of this significant difference in proportions between early and late stages,

which would have introduced a strong bias in favor of group A in subsequent global survival

comparisons. Therefore, we excluded all patients who had benefited from a surgical resection, i.e.

all patients who were diagnosed with a stage I or II tumor, and considered stage III-IV patients

only, i.e. patients who were directly treated in a palliative setting. This exclusion yielded a

smaller cohort, consisting of 22 patients for group A and 35 for group B. Subsequent analyses

were conducted on this restricted cohort. Demographic and prognostic factors comparisons are

presented below:

Page 48: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 43 -

Parameter Value Group A

(n=22)

Group B

(n=35)

Statistical

Test

p-

value

F 9 (41%) 12 (34%) Sex M 13 (59%) 23 (66%)

Chi-Square 0.61

III (locally advanced disease)

7 (32%) 13 (37%) Stage at diagnosis IV (metastatic

disease) 15 (68%) 22 (63%)

Chi-Square 0.68

Head 11 (55%) 19 (54%) Body/Tail 9 (45%) 16 (46%) Primary Site at

diagnosis Missing 2 - Chi-Square 0.96

Range 2-6 1.5-7 Median 4 4 Tumor Size at

diagnosis (cm) Missing 9 6

Mann-Whitney

0.65

Range 41-78 50-79 Median 59 66

25th Percentile 50 61 Age at diagnosis

75th Percentile 65 73

Mann-Whitney

0.005

0 6 (38%) 6 (30%) 1 6 (38%) 9 (45%) 2 4 (24%) 4 (20%) 3 - 1 (5%)

ECOG PS at

diagnosis

Missing 6 15

Mann-Whitney

0.74

Table 6: Patients characteristics (Stage III-IV at initial diagnosis)

The two groups were still comparable for sex (p=0.61), primary site, tumor size and ECOG PS

at initial diagnosis (p=0.96, 0.65 and 0.74, respectively). In contrast to the preceding comparison,

the two populations were also comparable in terms of stage at diagnosis, with roughly two thirds

of patients initially diagnosed with stage IV (i.e. metastatic) disease, and one third with stage III

(i.e. locally advanced) disease (p=0.68). However, age at diagnosis was still inhomogeneous

between the two populations (p=0.005), as illustrated by the following histograms with a theo-

retical normal curve as well as by the corresponding box plot:

Page 49: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 44 -

Figure 4: Histogram graph for age at palliative diagnosis (Group A)

Figure 5: Histogram graph for age at palliative diagnosis (Group B)

Page 50: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 45 -

Figure 6: Box plot for age at palliative diagnosis (stage III-IV patients only)

TREATMENT

Details of the surgical, radio therapeutic and chemotherapeutic management for the two

groups of patients are shown below. Figures for chemotherapeutic modalities represent the num-

ber of doses administered. For indicative purposes, data for the total cohort are also provided:

Page 51: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 46 -

Total Cohort Stage III-IV patients Treatment Modality

Group A (n=39) Group B (n=45) Group A (n=22) Group B (n=35)

Curative-intent

Duodenopancreatectomy 15 10 - -

Curative-intent left

Splenopancreatectomy 2 - - -

Palliative-intent surgical

intestinal derivation 11 7 10 7

Palliative-intent biliary

derivation 8 7 8 7

Palliative-intent biliary

stent 3 9 3 9

Palliative-intent

intestinal stent - 1 - 1

Adjuvant Chemo

radiotherapy (45-60 Gy)

14 (6 with 5-FU 500 mg/m2 and 6 with Cisplatin 6 mg/m2)

9 (7 with 5-FU 500 mg/m2 and 2 with Cisplatin 6 mg/m2)

- -

Palliative-intent

Radiotherapy (30-60

Gy)

8 10 8 10

Gemcitabine 1000

mg/m2 11 (92 doses) 11 (107 doses) 6 (54 doses) 8 (71 doses)

Gemcitabine 1000

mg/m2 + Capecitabine

1300 mg/m2

2 (12 doses) 31 (350 doses) - 25 (331 doses)

Oxaliplatin 50-80 mg/m2

+ Irinotecan 50-80

mg/m2

2 (3 doses) 14 (56 doses) - 12 (52 doses)

Leucovorin 30 mg/m2 +

5-FU 2300 mg/m2/24h 14 (206 doses) 13 (122 doses) 7 (28 doses) 12 (118 doses)

Leucovorin 30 mg/m2 +

5-FU 500 mg/m2 iv 7 (93 doses) 1 (2 doses) 3 (14 doses) 1 (2 doses)

5-FU 1700-2000

mg/m2/wk 1 (1 dose) 2 (17 doses) - 2 (17 doses)

Docetaxel 25-30 mg/m2

+ CPT-11 50-90 mg/m2 1 (1 dose) 1 (6 doses) - 1 (6 doses)

5-FU 200 mg/m2 +

Cisplatin 60 mg/m2 +

Epirubicin 50 mg/m2

2 (4 doses) - 2 (4 doses) -

Epirubicin 120 mg/m2 1 (2 doses) - 1 (2 doses) - Total number of

effectively administered

doses

414 660 102 597

Range of per-patient

administered doses 0-124 0-80 0-19 0-80

Median per-patient

administered doses 2 11 1 14

Log-rank p-value 0.07 0.0002 Table 7: Treatment characteristics for the total cohort and for stage III-IV patients

5-FU and Cisplatin doses given as part of a chemo radio therapeutic regimen are not mentioned under chemotherapeutic regimen, and were not included in total cycles calculations.

Median number of total per-patient administered doses and p-value were computed using a log-rank test to account for censored patients

Page 52: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 47 -

The following Kaplan-Meier graph illustrates the significant difference in per-patient admin-

istered doses for the two groups of stage III-IV patients:

Figure 7: Kaplan-Meier graph of total per-patient chemotherapy doses for stage III-IV patients. Figures indicated in parentheses correspond to (total patients; censored). A tick indicates censored patients.

Log-rank p=0.0002

This graph provides the proportion of patients having received a given amount of chemother-

apy doses from initial diagnosis to death or censoring.

SURVIVAL ANALYSES

While none of stage III-IV patients included in group A was still alive and none had been cen-

sored, four stage III-IV patients included in group B were still alive and undergoing therapy at the

time of analysis (October 31st, 2003). The total survival time Kaplan-Meier graph is shown

below:

Page 53: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 48 -

Figure 8: Kaplan-Meier graph of palliative survival time for stage III-IV patients. Figures indicated in

parentheses correspond to (total patients; censored). A tick indicates censored patients. Median survival time was 214 days for group A and 262 days for group B.

Log-rank p=0.012. Hazard ratio=0.52 (95% CI=0.23 to 0.84)

This graph plots survival time in palliative stage along the x-axis and the proportion of patients

still alive at any point in time along the y-axis.

Statistics Group A (n=22) Group B (n=35)

Median Survival time 214 days (7.1 months) 262 days (8.7 months)

95% confidence interval 131-296 days 116-408 days

25th Percentile 265 days (8.8 months) 785 days (26.2 months)

75th Percentile 107 days (3.6 months) 112 days (3.7 months)

1-year Survival probability 14% 40%

95% percentage confidence

interval 3-35% 24-58%

Log-rank p-value 0.012

Table 8: Log-rank test results for survival comparison in stage III-IV patients

Thus, this analysis revealed a statistically significant difference in survival time between the

two groups: median survival time was 7.1 months and 1-year survival probability was 14% for

group A versus 8.7 months and 40% for group B, respectively (p=0.012, hazard ratio=0.52, 95%

Page 54: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 49 -

CI 0.23-0.84). As evidenced by an increasing gap between the two curves after 300 days, patients

having survived more than one year mainly accounted for this difference, i.e. group B long-term

survivors survived significantly longer than group A ones.

Building on the previously observed significant difference between the two groups in terms of

total per-patient administered chemotherapy doses, this latter variable was plotted against

survival time:

Survival

16001400120010008006004002000

100

80

60

40

20

0

Group B

Dead

Group B

Alive

Group A

Dead

Figure 9: Total number of per-patient doses versus survival time graph for stage III-IV patients.

Non-parametric Spearman’s correlation coefficient ρρρρ=0.76

As expected, the longer patients survived, the more doses of chemotherapy they received.

Non-parametric analysis of this association yielded a Spearman’s correlation coefficient ρ of

0.76.

HOSPITALIZATION DAYS

Four basic categories of hospitalization were distinguished: hospitalization for treatment pur-

poses, hospitalization due to treatment complications, disease complications, and finally other

causes not related to treatment or disease. Detailed results corresponding to these categories are

provided below, without consideration of patients group:

Page 55: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 50 -

Treatment Treatment

complications

Disease

complications Others

Category Number of

stays Category

Number of

stays Category

Number of

stays Category

Number of

stays

Diagnosis 51 (including 10 explorative laparotomies)

Fever following

chemotherapy 4 Terminal care 41 Unknown 11

Port-a-Cath implantation

38 Treatment toxicity

6 (including 1 allergy, 1 diarrhea, 2 nausea & vomiting)

Jaundice � Biliary stent

12 Unrelated digestive hemorrhage

1

Digestive derivation

16 Port-a-Cath change

2 Fever 14 (including 3 pneumonia)

Biliary derivation

8 Hypoglycemia 1 General decline

10

Chemotherapy 9

Anemia following anastomotic ulcer

1 Ileus 8 (including 6 derivations)

Convalescence 6 Thrombosis 5 (including 2 Trousseau’s syndromes)

Radiotherapy 1 Ascites 3 Pain 3 Hernia 3

Digestive hemorrhage

3

Bone

metastasis 2

CNS vascular

event 2

Convalescence 2

Inaugural diabetes

2

Table 9: Number of stays for different hospitalization categories Categories are not mutually exclusive, i.e. a single stay may have been justified by several categories.

Derivative procedures were considered as treatment when occurring during the same stay as the diagnosis

In terms of total hospitalization days, a log-rank test was conducted to account for censored

patients. The corresponding Kaplan-Meier graph is shown below:

Page 56: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 51 -

Figure 10: Kaplan-Meier graph of total days of hospitalization for stage III-IV patients. Figures indicated

in parentheses correspond to (total patients; censored). A tick indicates censored patients. Log-rank p=0.50

This graph indicates the proportion of patients having reached a particular number of hospi-

talization days from diagnosis to death or censoring. With a median of 55 days in group A and 60

days in group B, the two populations were not significantly different (log-rank p=0.50).

Considering this result and the significant difference in terms of total survival, it was thus

expected that the ambulatory survival time (calculated as total survival time minus hospitalization

days) be significantly different between the two groups. The corresponding Kaplan-Meier graph

is shown below:

Page 57: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 52 -

Figure 11: Kaplan-Meier graph of palliative ambulatory survival time (total survival minus hospitalization days) for stage III-IV patients. Figures indicated in parentheses correspond to (total patients; censored). A

tick indicates censored patients. Median ambulatory survival time was 144 days for group A and 199 days for group B.

Log-rank p=0.013. Hazard ratio=0.52 (95% CI=0.24 to 0.84)

Indeed, the two groups were shown to differ significantly in terms of ambulatory survival

time, with a median of 144 days for group A patients and 199 days for group B (p=0.013, hazard

ratio=0.52, 95% CI 0.24-0.84).

The two groups were then compared in terms of ambulatory visits to Oncosurgery in the can-

tonal hospital: assuming that patients who survived longer were also followed and treated for a

longer period of time, one could expect a higher number of consultations for group B patients.

The corresponding Kaplan-Meier graph is shown below:

Page 58: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 53 -

Figure 12: Number of ambulatory visits in Oncosurgery for stage III-IV patients. Figures indicated in

parentheses correspond to (total patients; censored). A tick indicates censored patients. Median=21 (Group A) and 28 (Group B). Log-rank p=0.007

Thus, with a median of 28 visits, patients from group B were indeed seen significantly more in

ambulatory consultations than patients from group A (median of 21 visits; p=0.007).

ECONOMIC ANALYSES

Cost data were not available for two Group B patients. Also, reliable economic information

was only available from January 1st, 1995. Hence, all patients whose records showed a hospitali-

zation prior to this date were considered in economic analyses, which corresponds to 8 stage III-

IV patients in group A. Thus, economic analyses were finally conducted on 33 group B patients

and 14 group A patients only. Remaining patients were handled as missing values in statistical

tests.

The following table shows the weighting factors that were computed to express all costs in

2002-units:

Year

Weighting

Factor

2003 1 2002 1 2001 1.0893 2000 1.1148 1999 1.0810 1998 1.1964 1997 1.1964 1996 1.1964 1995 1.1964

Table 10: Weighting factors for costs conversion into 2002 units

Page 59: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 54 -

A per diem cost for convalescence and terminal care stays was computed by dividing total

expenses incurred by a clinic whose financial data were available (CHF 10,661,562.5) by its total

hospitalization days over the corresponding period (23,700). This yielded an average per diem

cost of CHF 449.85, which was rounded to CHF 450 to compute the costs of all convalescence or

terminal care stays after conversion into 2002 units.

Economic data for one patient is given below, as an example of the level of detail available:

Table 11: Example of economic data for one patient Values in Swiss Francs (CHF), annualized to 2002 levels (cf. Methodology).

Reference numbers censored for confidentiality purposes

As evidenced in Table 11, it was possible to break down total costs into five subgroups of

expenses, namely anesthesia, surgery, clinical care, imaging, laboratory and intensive care costs.

The sum corresponds to the total in the fourth column. Medication costs were a further break-

down available; they are however already spread over the five above-mentioned categories.

Finally, it was possible to distinguish between costs incurred for hospitalization stays and costs

incurred for ambulatory visits to the hospital (i.e. mainly for chemotherapeutic treatment and

follow-up in the context of the present analysis, since treatment was mainly provided on an out-

patient basis). Several visits are usually included under a unique reference number because of the

hospital’s administrative process (i.e. the same reference number is used for every ambulatory

visit until the patient is hospitalized). Similarly, convalescence and terminal care stays in other

clinics were taken into account as explained by extrapolating a global cost from a theoretical per

diem.

Patient Reference Number

Stay Reference Number

Entry Date for the considered stay

Total Costs annualized to 2002

Anaesthesia Costs annualized to 2002

Surgery Costs annualized to 2002

Clin

ical Care costs annualized to 2002

Imaging Costs annualized to 2002

Laboratory Costs annualized to 2002

Intensive Care Costs annualized to 2002

Included Cost of Medications annualized

to 2002

Hospital Stay

Ambulatory Visit

XXX XXX XXX XXX 30/01/2003 12,479 999 428 9,848 - 1,205 - 200 x

XXX XXX XXX XXX 13/02/2003 2,934 - - 1,632 395 907 - 57 x

XXX XXX XXX XXX 13/03/2003 18,799 1,539 2,869 13,727 - 663 - 305 x

XXX XXX XXX XXX 23/04/2003 72 - - 72 - - - 1 x

XXX XXX XXX XXX 10/05/2003 13,526 - - 12,066 588 873 - 225 x

47,811 2,538 3,297 37,345 983 3,647 - 788 Total

Page 60: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 55 -

The following Kaplan-Meier graph shows total incurred costs for the two stage III-IV patients

groups, i.e. the costs of all hospitalizations, all ambulatory visits to the cantonal hospital and all

convalescence and terminal care stays for each patient:

Figure 13: Kaplan-Meier graph of total costs incurred by the hospital for stage III-IV patients. Figures

indicated in parentheses correspond to (total patients; censored). A tick indicates censored patients. Cost data were not available for 8 group A patients and 2 group B, who were treated as missing values.

Monetary unit: CHF Log-rank p=0.76

This graph provides the proportion of patients having reached a given cost from diagnosis to

death or censoring. With a median of CHF 82,380 for group A and of CHF 76,750 for group B,

the two populations were not significantly different (p=0.76) with respect to total costs incurred

by the cantonal hospital, which include all care services provided both on an in-patient and out-

patient basis.

Since the cost accounting system of the cantonal hospital allowed to distinguish between hos-

pitalization-related costs and costs related to ambulatory visits, statistics for the two categories

were computed for indicative purposes using data available for deceased patients. Results are

provided in the following table:

Page 61: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 56 -

Subgroup A (n=14) Subgroup B (n=29)

Statistic Cost

Category Total

patients Mean Median

Total

patients Mean Median

Hospitalization 1,054,067 75,291 78,960 2,071,276 62,766 50,120

Ambulatory 67,622 4,830 5,005 379,230 11,491 8,537 Total Costs

Total 1,121,689 80,121 83,964 2,450,506 74,257 57,609

Ratio Hosp:Amb 15.6:1 15.8:1 5.5:1 5.7:1

Hospitalization 94% 85% Proportion of

total Ambulatory 6% 15%

Hospitalization 21,933 – 168,873

3,864 – 173,732

Range

Ambulatory 418 – 11,513

510 – 42,877

Table 12: Hospitalization-related and ambulatory costs for stage III-IV deceased patients Monetary unit: CHF

Additional analyses were then conducted to further assess possible differences between the

two groups along this dimension: given the absence of any significant difference in terms of hos-

pitalization days, one could expect that hospitalization-related costs would be comparable. How-

ever, considering that patients who survived longer were also followed and treated for a longer

period of time, as evidenced by the number of ambulatory visits analysis, one could expect

ambulatory costs in group B to exceed ambulatory costs in group A. Given the low proportion of

total costs represented by ambulatory costs (approximately 6% in group A and 15% in group B),

it was possible that this difference was not significant enough in absolute figures to influence

previously exposed total costs results. The corresponding graphs are shown below:

Page 62: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 57 -

Figure 14: Hospitalization-related total costs for stage III-IV patients. Figures indicated in parentheses

correspond to (total patients; censored). A tick indicates censored patients. Cost data were not available for 8 group A patients and 2 group B, who were treated as missing values.

Monetary unit: CHF Median=78,960 (Group A) and 50,120 (Group B). Log-rank p=0.93

Figure 15: Ambulatory total costs for stage III-IV patients. Figures indicated in parentheses correspond to (total patients; censored). A tick indicates censored patients. Cost data were not available for 8 group A

patients and 2 group B, who were treated as missing values. Monetary unit: CHF

Median=5,005 (Group A) and 8,537 (Group B). Log-rank p=0.006

Thus, corroborating the ambulatory visits analysis, the two groups were shown to differ sig-

nificantly in terms of ambulatory costs.

Page 63: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 58 -

DISCUSSION

Two groups of patients suffering from advanced pancreatic cancer were chronologically

defined, according to the first chemotherapy regimen with respect to the introduction of gemcit-

abine in Switzerland. The two groups were shown to differ significantly in terms of total survival

time (median of 7.1 in group A versus 8.7 months in group B, p=0.012), ambulatory survival time

(median of 4.8 in group A versus 6.6 months in group B, P=0.013), median per-patient chemo-

therapy doses received (1 in group A versus 14 in group B, p=0.0002), ambulatory visits in

Oncosurgery (median of 21 in group A versus 28 in group B, p=0.007) and ambulatory costs

(median of CHF 5,005 in group A versus 8,537 in group B, p=0.006). Patients having survived

more than one year, i.e. “long-term survivors”, mainly accounted for the observed differences.

There were no significant differences in terms of total hospitalization days as well as total and

hospitalization-related costs. These results are discussed below in more details.

METHODOLOGY

The method employed to define a cut-off date and distinguish the two groups, according to the

date of the first-line gemcitabine treatment for the first patient, was based on the following

rationale: the availability of this drug and of subsequently introduced medications such as

oxaliplatin, together with the significant improvements in clinical benefit responses that those

new treatments made possible, was supposed to have changed the attitude of many oncologists

regarding advanced pancreatic cancer management. As it is well corroborated by an exponential

increase in the relevant medical literature since 1996 (which corresponds to the introduction of

gemcitabine in the U.S.), chemotherapy was, from then on, considered as an efficient modality in

advanced pancreatic cancer management, and was therefore more systematically prescribed. Our

intention was to assess this change on a local cohort over the last decade along clinical dimen-

sions –i.e. mainly survival time– as well as medico-economic dimensions –i.e. costs incurred by

the hospital.

As far as diagnosis dates are concerned, the first day of the hospitalization stay leading to or

confirming the diagnosis was systematically retained. This systematic approach was meant to

allow for maximal precision in the definition of this crucial milestone. Given the rather large time

span of the study, i.e. a decade, it can however not be totally excluded that evolution of diagnos-

tic procedures introduced some bias towards an artificially longer observed survival time for

group B patients, an effect known as Will Rogers’ phenomenon [137]. By demonstrating metasta-

ses that would have formerly been silent and unidentified, more sensitive diagnostic technology

Page 64: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 59 -

can result in stage migration: patients originally classified in less advanced stages could be

assigned to a more advanced one. As a consequence, the prognosis of each stage cohort shall

improve artificially because the less advanced stage cohort looses some of its worst cases (with

silent more advanced disease) while the more advanced cohort shall be enriched by better prog-

nostic cases. The following figure illustrates this phenomenon:

Figure 17: Will Rogers phenomenon; during the period corresponding to group B, patients with

metastases that would have gone undiagnosed in group A could have been assigned to a stage III or IV due to progresses in diagnostic procedures, hereby increasing the overall prognosis in both stage I-II and stage III-IV

subgroups

Given the retrospective design of the present study, it is impossible to assess the importance of

this effect in survival time estimations. However, apart from strictly technological evolutions in

ultrasonography and CT-Scan techniques, there has not been any fundamental change of diag-

nostic strategy for pancreatic cancer in Geneva over the last decade, for example towards a more

aggressive diagnostic behavior such as a screening process: patients were typically referred for

jaundice or general alteration and subsequently underwent radiological procedures (typically

ultrasound followed by CT-Scan) that led to the suspicion of a malignant process affecting pan-

creas. Diagnosis was then formally confirmed by histological examination of a tissue sample.

Therefore, we believe the occurrence of a significant Will Rogers’ phenomenon is improbable in

this particular setting.

With respect to medico-economic analyses, several methodological limitations deserve special

comments. First, the hospitalization cost calculation process relied mainly on the accuracy of

medical coding at the end of each stay or ambulatory visit. Since coding was not yet assumed by

professionals in Geneva but by care providers themselves, it may have sometimes been incom-

Page 65: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 60 -

plete or imprecise. Consequently, incurred costs may have been underestimated. However, eco-

nomic calculations can be considered as homogenous between the two groups, since they rely on

the same cost accounting system, whose procedures have not fundamentally changed over the

considered time period (1995-2003). Second, the method employed to express all cost in 2002

units was correct, financially speaking. However, 2003 costs were expressed using the same

weighting as 2002 ones, since economic data to compute the corresponding conversion factor

were not available at the time of analysis. Similarly, information to compute weighting factors

was not available prior to 1998, and a single factor was therefore used for years 1995 to 1997.

Furthermore, weighting factors were corrected between 1999 and 2000, and the apparent reduc-

tion between those two years (weighting factor 1.08 versus 1.11) does not truly represent the real

evolution of health care costs in Switzerland, which were rather steadily increasing from year to

year. Although those limitations could have been circumvented by computing a compound annual

growth rate between 1995 and 2002, we chose to use official factors, which reflect the apprecia-

tion by the hospital itself of its incurred costs. Last, ambulatory costs outside Oncosurgery were

not included at all in cost analyses because corresponding data were not available retrospectively.

However, Oncosurgery followed all included patients during their entire disease and it is unlikely

that any significant expense occurred outside the hospital. Given those limitations though, med-

ico-economic results should more be considered more reflecting local experience of a particular

hospital service than pretend to any generalization.

CLINICAL RESULTS

First, epidemiological information provided by the Geneva Cancer Registry showed that our

study cohort was including approximately 16% of the total number of cases of pancreatic carci-

noma having occurred in Geneva from 1994 to 2001. Since such percentages are generally not

published in the literature, it is difficult to comment on the representativeness of our sample

compared to other studies. Interestingly, 32% of cases were considered as clinically malignant

tumors without histological confirmation, which might reflect the difficulty in getting appropriate

tissue samples in this pathology.

Second, in terms of basic characteristics, the significant initial stage difference between the

two populations at the time of diagnosis has already been mentioned in the methodology section.

It was decided to restrict all subsequent analyses to stage III-IV patients to avoid biased survival

time and medico-economic results: indeed, from a survival time perspective, resected patients

have a clearly different biological evolution from those suffering from a more advanced disease

Page 66: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 61 -

that precludes any surgical intervention. Also, from a medico-economic perspective, the two

populations present a different cost structure, with resected patients incurring, for example, major

expenses related to the initial surgery, while patients suffering from an advanced disease incur

major expenses related, for instance, to mechanical intestinal or biliary complications. As already

mentioned, a potential solution would have been to consider only the palliative phase of the dis-

ease for resected patients. However, starting point definition of this period was highly hazardous

given the absence of any rigorously standardized follow-up procedures, both radiological and

biological. Furthermore, on the biological side, it is well known that CA 19-9 level at the time of

recurrence gives a much poorer indication than the initial TNM staging in terms of tumor load

and prognosis. More generally, it does not make any clinical sense to stage patients at relapse for

prognosis assessment purposes, making it almost impossible to assess patient homogeneity along

this dimension.

As far as stages are concerned, the proportion of locally advanced and metastatic pancreatic

cancers between the two groups was comparable, and corresponds roughly to published results on

larger series (proportion of about 1/3 to 2/3, respectively). With respect to other prognostic fac-

tors, the two populations were also homogeneous, with values corroborating previously published

results (ECOG PS, median size of the primary tumor), except for a slightly higher proportion of

body and tail locations for the primary tumor (30% in the present cohort versus around 20% in

the literature).

The subsisting significant difference between the two groups of stage III-IV patients in terms

of age at initial diagnosis (median of 59 for group A and 66 for group B, p=0.005) was expected

to favor group A in survival analyses. Given the limited influence of this factor in comparison to

other prognostic variables and the survival results actually in favor of group B, stratified analyses

were not conducted. Interestingly, this difference can be interpreted as reflecting behavioral

changes among medical oncologists who, realizing the clinical benefit of newer chemotherapeutic

molecules in this indication, might have proposed such treatments to a greater range of patients.

Third and as expected from the retrospective design of the present study and from therapeutic

management of included patients (usually outside any protocol), treatment modalities were rather

inhomogeneous. For instance, due to the method employed for defining the two groups, it was

unavoidable that some patients belonging to group A had actually received a few cycles of gem-

citabine at some point of their management, since they sometimes survived several months after

1998, i.e. after this drug had been made available. However and by definition, patients in group A

Page 67: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 62 -

never received gemcitabine as a first-intent therapy, while it was the case for every group B

patient. Overall, the evolution of chemotherapy prescribing practice in Geneva over the last dec-

ade can be summarized as follows: in group A, the most often administered treatment consisted

of 5-FU and leucovorin-based regimens (total of 42 doses). In group B, contrastingly, the most

often administered fist-line regimen consisted of an association of gemcitabine and capecitabine

(which simulates a continuous infusion of 5-FU; total of 331 doses administered), with second-

line regimens generally associating 5-FU and leucovorin (total of 120 doses administered) or

oxaliplatin and irinotecan (total of 52 doses administered). The total number of per-patient doses

was significantly different between the two groups, probably related to the corresponding differ-

ence in survival time (patients in group B enjoyed a prolonged survival time compared to patients

in group A, which allowed them to receive more cycles) and to behavioral changes among medi-

cal oncologists towards prescribing chemotherapy more systematically in this indication.

Fourth, in terms of survival analysis, we observed a statistically significant difference in favor

of group B patients (median survival time of 214 days or 7.1 months in group A versus 262 days

or 8.7 months in group B, p=0.012, hazard ratio=0.52, 95% CI 0.23-0.84), despite the age profile

discrepancy, which was favorable to group A. Large and overlapping 95% confidence intervals

reflect the small size of the samples. Therefore, the observed significant difference should more

be considered as a positive trend. However, recalling previously exposed data from several

reviews [50-58], this trend is in line with average values published so far in the literature, respec-

tively for 5-FU-based regimens (which were the main treatment in group A patients) and for

gemcitabine-5-FU combinations (which were the first-intent treatment for a vast majority of

patients in group B). Interestingly, the difference was mainly accounted for by the long-term sur-

vivor fraction of patients in group B, which can be better appreciated by considering 1-year sur-

vival probabilities (14% in group A versus 40% in group B; overlapping 95% confidence inter-

vals: [3-35%] in group A versus [24-58%] in group B). As it is well corroborated by the litera-

ture, gemcitabine as well as some other new molecules available for advanced pancreatic cancer

management seem to particularly benefit those patients whose tumor biology tends to respond to

chemotherapy. Patients belonging to this specific category now survive longer, sometimes even

much longer than the average 9 months mentioned in medical textbooks. It is however possible,

given the small groups in the context of our study, that a difference in proportion of different

tumor biology profiles included in groups A and B introduced an unidentified bias in survival

analyses and partly accounted for the observed difference. It is also possible, as is suggested by

Page 68: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 63 -

the older age of patients from group B at the time of diagnosis, that the overall epidemiology of

pancreas cancer (i.e. its biological characteristics) changed over the past few years and accounted

for the observed difference. In absence of supporting evidence in today’s medical literature, this

would require further investigations.

Fifth, in terms of total hospitalization days, the two groups were comparable, indicating that

patients in group B, although having survived longer, did not experience significantly more hos-

pital stays than in group A. This was further evidenced by comparing the two groups in terms of

ambulatory survival (calculated as total survival time minus hospitalization days for each

patient), which was shown to be significantly different (median of 144 days or 4.8 months in

group A versus 199 days or 6.6 months in group B, p=0.013). According to the hospitalization

category analysis (table 11), patients suffering from advanced pancreatic cancer were mainly

hospitalized for serious disease-related complications as well as for terminal palliative care. In

this context, the absence of difference in hospitalization days between the two groups suggests

that the increase in total survival time was accompanied by a relative preservation in terms of

quality of life, patients having been able to spend more time in their usual environment, followed

on an outpatient basis. Consequently, on the ambulatory side, the significant difference between

the two groups in terms of visits to the outpatient clinic for treatment or check-up is probably

related to the difference in survival time: patients in group B, who survived longer, were also

followed periodically, and they received more chemotherapy cycles than in group A, as previ-

ously exposed. Intuitively, this pattern had to be similar in terms of medico-economic analyses.

MEDICO-ECONOMIC RESULTS

The two groups were not significantly different in terms of total costs incurred by the hospital.

However, when costs were broken down into hospitalization-related and ambulatory-related cate-

gories, a significant difference was evidenced for ambulatory costs, which corresponded for pan-

creatic cancer mainly to outpatient visits for treatment, check-up and general management. Those

expenditures were significantly higher for patients in group B than in group A (median of CHF

8,537 versus 5,005; p=0.006). Considering absolute figures, the low proportion of total costs rep-

resented by the ambulatory category probably explains why, given the small size of the samples,

this difference was not reflected in global analyses (this proportion was 6% in group A and

increased to 15% in group B). In line with the previously observed difference in per-patient

administered chemotherapy doses as well as in the number of ambulatory visits to Oncosurgery,

Page 69: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 64 -

these results are probably related to the increased survival time of group B patients, who were

followed and treated longer than in group A, without being hospitalized more often.

We are aware that the low number of patients for whom economic data were available (par-

ticularly in group A, n=14) somewhat jeopardizes our results from a statistical standpoint, and

hence that the latter can at best be considered as indicating a tendency. They are however worth

mentioning because the trend they suggest could be further assessed prospectively, and because

they rely on a methodology that draws strongly upon the Geneva teaching Hospital’s cost

accounting system, which could be more extensively used for such medico-economic analyses in

the future.

Page 70: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 65 -

CONCLUSION

Several biases related to the retrospective design of the present analyses might limit their

scope and preclude them to be considered as general. Therefore, this study should be interpreted

in its local and exploratory context. Results exposed, and particularly medico-economic observa-

tions, need to be validated in prospective studies of larger scope. But taken all together, our

results suggest the following interpretation: since late 1998, a more standardized and systematic

prescription of new chemotherapeutic drugs in advanced pancreatic cancer benefited a small per-

centage of responders in terms of survival time, although the quantitative absolute increase was

minor. Furthermore, if the number of hospitalization days is accepted as a rough proxy for the

quality of life, those responders seem not to have experienced significantly more debilitating

complications in absolute terms. On the contrary, they were managed for a relatively long period

on an ambulatory basis, periodically consulting the outpatient clinic for treatment and general

check-ups. These observations suggest that in the future, along with the development of more

effective molecules and their systematic assessment in combination regimens, some patients suf-

fering from advanced pancreatic cancer may experience a prolonged relief of symptoms.

From an economic perspective, however, every progress has its cost. In that sense, present

medico-economic results should not be underestimated: prolonging survival time in patients suf-

fering from an advanced malignant disease that cannot be cured inevitably leads to prolonged

treatment and follow-up, and repeated biologic and radiological investigations. In the case of

advanced pancreatic cancer, given the dismal prognosis, ambulatory costs may not sound impres-

sive in absolute figures, although some individuals reached a range of CHF 20,000 to 50,000 of

ambulatory costs in the present cohort. However, extrapolating those figures to the entire Swiss

population gives a much clearer idea of the financial implications of medical progresses that may

seem minor at passing glance. Also, considering other much more prevalent advanced malignant

diseases helps understand this dilemma from the centrality it currently occupies in politicians’

and economic stakeholders’ minds. A particularly good example is metastatic colorectal cancer,

where chemotherapy evolved significantly over the last decade, from 5-FU modulated by leuco-

vorin to poly-chemotherapy regimens (fluoropyrimidines with oxaliplatin or irinotecan) inte-

grated into multi-disciplinary therapeutic strategies. As a result, median survival time of patients

with metastatic colorectal cancer currently ranges between 17 and 22 months [138], during which

they have to be treated with expensive association regimens, together with check-ups and peri-

Page 71: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 66 -

odic reassessments. Benefits to those patients are beyond contest compared to the situation ten

years ago, when median survival time figures were approximately half the current ones (between

10 and 12 months). But those benefits have a cost. It was recently estimated that the doubling of

median survival time achieved over the past decade has been accompanied by a staggering 340-

fold increase in drug costs –just for the initial eight weeks of treatment (approximately $100-300

for fluorouracil-based regimens to $20,000-30,000 for bevacizumab or cetuximab via $10,000 for

irinotecan or oxaliplatin) [139]. Thus, medication costs for the initial eight weeks of treatment for

the estimated 56,000 colorectal cancer patients diagnosed at stage IV or developing metastases in

2004 in the United States were estimated in the range of $666 million to $1.2 billion (with the

addition of monoclonal-antibody therapy).

Health care is fascinating in that it incorporates two crucial dimensions of quality of medical

services and economic efficiency, which are often considered as antagonist or incompatible. This

multidimensionality is probably the reason why it cannot obey a single liberal economic model

and why it is currently characterized by a complex and country-specific mixture of regulations

and market-openness, whose ideal proportions have yet to be defined. That health care-related

costs are steadily increasing in developed countries is a fact, as is the necessity of addressing this

situation. However, it is also a fact that people overall are more and more expecting and

demanding medical excellence. Ultimately, trade-off between quality and economic burden is a

choice each society as a whole has to make for itself. This choice implies communication and

concord between all involved stakeholders –regulators, payers, providers and above all patients.

Page 72: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 67 -

REFERENCES

1. Jemal, A., Murray, T., Samuels, A., Ghafoor, A., Ward, E., and Thun, M. J. (2003). Cancer statistics, 2003. CA Cancer J Clin 53, 5-26.

2. Evans, A., Abbruzzese, J., and Willett, C. G. (2001). Cancer of the pancreas. In "Cancer: Principles and practice of oncology" (V. T. DeVita, S. Hellman, and S. A. Rosenberg, Eds.), Lippincott, Williams & Wilkins, Philadelphia.

3. Sener, S. F., Fremgen, A., Menck, H. R., and Winchester, D. P. (1999). Pancreatic cancer: a report of treatment and survival trends for 100,313 patients diagnosed from 1985-1995, using the National Cancer Database. J Am Coll Surg 189, 1-7.

4. Ginsberg, G. G. (2000). New developments in pancreatic cancer. Semin Gastrointest Dis 11, 162-7.

5. Lowenfels, A. B., Maisonneuve, P., DiMagno, E. P., Elitsur, Y., Gates, L. K., Jr., Perrault, J., and Whitcomb, D. C. (1997). Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J Natl Cancer Inst 89, 442-6.

6. Whitcomb, D. C., Preston, R. A., Aston, C. E., Sossenheimer, M. J., Barua, P. S., Zhang, Y., Wong-Chong, A., White, G. J., Wood, P. G., Gates, L. K., Jr., Ulrich, C., Martin, S. P., Post, J. C., and Ehrlich, G. D. (1996). A gene for hereditary pancreatitis maps to chromosome 7q35. Gastroenterology 110, 1975-80.

7. Lowenfels, A. B., Maisonneuve, P., Cavallini, G., Ammann, R. W., Lankisch, P. G., Andersen, J. R., Dimagno, E. P., Andren-Sandberg, A., and Domellof, L. (1993). Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med 328, 1433-7.

8. Konner, J., and O'Reilly, E. (2002). Pancreatic cancer: epidemiology, genetics, and approaches to screening. Oncology (Williston Park) 16, 1615-22, 1631-2; discussion 1632-3, 1637-8.

9. Whelan, A. J., Bartsch, D., and Goodfellow, P. J. (1995). Brief report: a familial syndrome of pancreatic cancer and melanoma with a mutation in the CDKN2 tumor-suppressor gene. N Engl J Med 333, 975-7.

10. Goldstein, A. M., Fraser, M. C., Struewing, J. P., Hussussian, C. J., Ranade, K., Zametkin, D. P., Fontaine, L. S., Organic, S. M., Dracopoli, N. C., Clark, W. H., Jr., and et al. (1995). Increased risk of pancreatic cancer in melanoma-prone kindreds with p16INK4 mutations. N Engl J Med 333, 970-4.

11. Lorenzo Bermejo, J., and Hemminki, K. (2004). Risk of cancer at sites other than the breast in Swedish families eligible for BRCA1 or BRCA2 mutation testing. Ann Oncol 15, 1834-41.

Page 73: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 68 -

12. Lynch, H. T., Deters, C. A., Snyder, C. L., Lynch, J. F., Villeneuve, P., Silberstein, J., Martin, H., Narod, S. A., and Brand, R. E. (2005). BRCA1 and pancreatic cancer: pedigree findings and their causal relationships. Cancer Genet Cytogenet 158, 119-25.

13. (2002). Definition of TNM (pancreatic cancer section). In "AJCC Cancer Staging Handbook" (F. L. Greene, D. L. Page, I. D. Fleming, A. G. Fritz, C. M. Balch, D. G. Haller, and M. Morrow, Eds.), Springer, Chicago.

14. Ryan, D. P., and Willett, C. G. (2002). Management of locally advanced adenocarcinoma of the pancreas. Hematol Oncol Clin North Am 16, 95-103.

15. Hruban, R. H., Goggins, M., Parsons, J., and Kern, S. E. (2000). Progression model for pancreatic cancer. Clin Cancer Res 6, 2969-72.

16. Hruban, R. H., Wilentz, R. E., and Kern, S. E. (2000). Genetic progression in the pancreatic ducts. Am J Pathol 156, 1821-5.

17. Rocha Lima, C. M., and Centeno, B. (2002). Update on pancreatic cancer. Curr Opin Oncol 14, 424-30.

18. Magee, C. J., Shekouh, A., Ghaneh, P., and Neoptolemos, J. P. (2002). Update on pancreatic cancer. Hosp Med 63, 200-6.

19. Li, D., and Jiao, L. (2003). Molecular epidemiology of pancreatic cancer. Int J Gastrointest Cancer 33, 3-14.

20. Sohn, T. A., and Yeo, C. J. (2000). The molecular genetics of pancreatic ductal carcinoma: a review. Surg Oncol 9, 95-101.

21. Kardamakis, D. (1996). Tumour serum markers: clinical and economical aspects. Anticancer Res 16, 2285-8.

22. Hahn, S. A., and Kern, S. E. (1995). Molecular genetics of exocrine pancreatic neoplasms. Surg Clin North Am 75, 857-69.

23. Humphreys, M. J., Greenhalf, W., Neoptolemos, J. P., and Ghaneh, P. (1999). The potential for gene therapy in pancreatic cancer. Int J Pancreatol 26, 5-21.

24. Van Cutsem, E., van de Velde, H., Karasek, P., Oettle, H., Vervenne, W. L., Szawlowski, A., Schoffski, P., Post, S., Verslype, C., Neumann, H., Safran, H., Humblet, Y., Perez Ruixo, J., Ma, Y., and Von Hoff, D. (2004). Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol 22, 1430-8.

25. Diehl, S. J., Lehmann, K. J., Sadick, M., Lachmann, R., and Georgi, M. (1998). Pancreatic cancer: value of dual-phase helical CT in assessing resectability. Radiology 206, 373-8.

26. Pisters, P. W., Lee, J. E., Vauthey, J. N., Charnsangavej, C., and Evans, D. B. (2001). Laparoscopy in the staging of pancreatic cancer. Br J Surg 88, 325-37.

Page 74: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 69 -

27. Bluemke, D. A., Cameron, J. L., Hruban, R. H., Pitt, H. A., Siegelman, S. S., Soyer, P., and Fishman, E. K. (1995). Potentially resectable pancreatic adenocarcinoma: spiral CT assessment with surgical and pathologic correlation. Radiology 197, 381-5.

28. Fuhrman, G. M., Charnsangavej, C., Abbruzzese, J. L., Cleary, K. R., Martin, R. G., Fenoglio, C. J., and Evans, D. B. (1994). Thin-section contrast-enhanced computed tomography accurately predicts the resectability of malignant pancreatic neoplasms. Am J Surg 167, 104-11; discussion 111-3.

29. Ahmad, N. A., Lewis, J. D., Siegelman, E. S., Rosato, E. F., Ginsberg, G. G., and Kochman, M. L. (2000). Role of endoscopic ultrasound and magnetic resonance imaging in the preoperative staging of pancreatic adenocarcinoma. Am J Gastroenterol 95, 1926-31.

30. Rosch, T., Lorenz, R., Braig, C., Feuerbach, S., Siewert, J. R., Schusdziarra, V., and Classen, M. (1991). Endoscopic ultrasound in pancreatic tumor diagnosis. Gastrointest Endosc 37, 347-52.

31. Jimenez, R. E., Warshaw, A. L., Rattner, D. W., Willett, C. G., McGrath, D., and Fernandez-del Castillo, C. (2000). Impact of laparoscopic staging in the treatment of pancreatic cancer. Arch Surg 135, 409-14; discussion 414-5.

32. Leach, S. D., Lee, J. E., Charnsangavej, C., Cleary, K. R., Lowy, A. M., Fenoglio, C. J., Pisters, P. W., and Evans, D. B. (1998). Survival following pancreaticoduodenectomy with resection of the superior mesenteric-portal vein confluence for adenocarcinoma of the pancreatic head. Br J Surg 85, 611-7.

33. Yeo, C. J., Cameron, J. L., Lillemoe, K. D., Sitzmann, J. V., Hruban, R. H., Goodman, S. N., Dooley, W. C., Coleman, J., and Pitt, H. A. (1995). Pancreaticoduodenectomy for cancer of the head of the pancreas. 201 patients. Ann Surg 221, 721-31; discussion 731-3.

34. Yeo, C. J., Cameron, J. L., Sohn, T. A., Lillemoe, K. D., Pitt, H. A., Talamini, M. A., Hruban, R. H., Ord, S. E., Sauter, P. K., Coleman, J., Zahurak, M. L., Grochow, L. B., and Abrams, R. A. (1997). Six hundred fifty consecutive pancreaticoduodenectomies in the 1990s: pathology, complications, and outcomes. Ann Surg 226, 248-57; discussion 257-60.

35. Tsiotos, G. G., Farnell, M. B., and Sarr, M. G. (1999). Are the results of pancreatectomy for pancreatic cancer improving? World J Surg 23, 913-9.

36. Griffin, J. F., Smalley, S. R., Jewell, W., Paradelo, J. C., Reymond, R. D., Hassanein, R. E., and Evans, R. G. (1990). Patterns of failure after curative resection of pancreatic carcinoma. Cancer 66, 56-61.

37. Abrams, R. A. (2003). Adjuvant therapy for pancreatic adenocarcinoma: what have we learned since 1985? Int J Radiat Oncol Biol Phys 56, 3-9.

38. Kalser, M. H., and Ellenberg, S. S. (1985). Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 120, 899-903.

Page 75: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 70 -

39. (1987). Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Gastrointestinal Tumor Study Group. Cancer 59, 2006-10.

40. Klinkenbijl, J. H., Jeekel, J., Sahmoud, T., van Pel, R., Couvreur, M. L., Veenhof, C. H., Arnaud, J. P., Gonzalez, D. G., de Wit, L. T., Hennipman, A., and Wils, J. (1999). Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg 230, 776-82; discussion 782-4.

41. Choti, M. A. (2004). Adjuvant therapy for pancreatic cancer--the debate continues. N Engl J Med 350, 1249-51.

42. Neoptolemos, J. P., Stocken, D. D., Friess, H., Bassi, C., Dunn, J. A., Hickey, H., Beger, H., Fernandez-Cruz, L., Dervenis, C., Lacaine, F., Falconi, M., Pederzoli, P., Pap, A., Spooner, D., Kerr, D. J., and Buchler, M. W. (2004). A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 350, 1200-10.

43. Spitz, F. R., Abbruzzese, J. L., Lee, J. E., Pisters, P. W., Lowy, A. M., Fenoglio, C. J., Cleary, K. R., Janjan, N. A., Goswitz, M. S., Rich, T. A., and Evans, D. B. (1997). Preoperative and postoperative chemoradiation strategies in patients treated with pancreaticoduodenectomy for adenocarcinoma of the pancreas. J Clin Oncol 15, 928-37.

44. Tsai, J. Y., Iannitti, D. A., and Safran, H. (2003). Combined modality therapy for pancreatic cancer. Semin Oncol 30, 71-9.

45. Evans, D. B., Rich, T. A., Byrd, D. R., Cleary, K. R., Connelly, J. H., Levin, B., Charnsangavej, C., Fenoglio, C. J., and Ames, F. C. (1992). Preoperative chemoradiation and pancreaticoduodenectomy for adenocarcinoma of the pancreas. Arch Surg 127, 1335-9.

46. Hoffman, J. P., Lipsitz, S., Pisansky, T., Weese, J. L., Solin, L., and Benson, A. B., 3rd (1998). Phase II trial of preoperative radiation therapy and chemotherapy for patients with localized, resectable adenocarcinoma of the pancreas: an Eastern Cooperative Oncology Group Study. J Clin Oncol 16, 317-23.

47. Mallinson, C. N., Rake, M. O., Cocking, J. B., Fox, C. A., Cwynarski, M. T., Diffey, B. L., Jackson, G. A., Hanley, J., and Wass, V. J. (1980). Chemotherapy in pancreatic cancer: results of a controlled, prospective, randomised, multicentre trial. Br Med J 281, 1589-91.

48. Palmer, K. R., Kerr, M., Knowles, G., Cull, A., Carter, D. C., and Leonard, R. C. (1994). Chemotherapy prolongs survival in inoperable pancreatic carcinoma. Br J Surg 81, 882-5.

49. Polati, E., Finco, G., Gottin, L., Bassi, C., Pederzoli, P., and Ischia, S. (1998). Prospective randomized double-blind trial of neurolytic coeliac plexus block in patients with pancreatic cancer. Br J Surg 85, 199-201.

50. el-Rayes, B. F., Shields, A. F., Vaitkevicius, V., and Philip, P. A. (2003). Developments in the systemic therapy of pancreatic cancer. Cancer Invest 21, 73-86.

Page 76: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 71 -

51. Fung, M. C., and Sakata, T. (2002). What's new in pancreatic cancer treatment? J Hepatobiliary Pancreat Surg 9, 61-75.

52. Oettle, H., Arnold, D., Hempel, C., and Riess, H. (2000). The role of gemcitabine alone and in combination in the treatment of pancreatic cancer. Anticancer Drugs 11, 771-86.

53. Heinemann, V. (2002). Gemcitabine-based combination treatment of pancreatic cancer. Semin Oncol 29, 25-35.

54. Heinemann, V. (2002). Gemcitabine in the treatment of advanced pancreatic cancer: a comparative analysis of randomized trials. Semin Oncol 29, 9-16.

55. Kozuch, P., Petryk, M., and Bruckner, H. W. (2002). A comprehensive update on the use of chemotherapy for metastatic pancreatic adenocarcinoma. Hematol Oncol Clin North Am 16, 123-38.

56. Haller, D. G. (2003). Chemotherapy for advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 56, 16-23.

57. Louvet, C., Andre, T., and Artru, P. (2002). [Gemcitabine-based combinations in inoperable pancreatic cancers]. Bull Cancer 89 Spec No, S96-101.

58. Claude, L., and Mornex, F. (2003). [Chemoradiation in pancreatic carcinoma]. Cancer Radiother 7, 254-65.

59. Karnofsky, D. A., and Burchenal, J. H. (1949). The clinical evaluation of chemotherapeutic agents in cancer. In "Evaluation of Chemotherapeutic Agents" (M. CM, Ed.), Columbia University Press, New York.

60. Zubrod, C. G., Schneiderman, M. A., and Frei, A. (1960). Appraisal of methods for the study of chemotherapy in man: comparative therapeutics trial of nitrogen mustard and triethylene thiophosphoramide. J Chron Dis 11, 7-33.

61. Roila, F., Lupattelli, M., Sassi, M., Basurto, C., Bracarda, S., Picciafuoco, M., Boschetti, E., Milella, G., Ballatori, E., Tonato, M., and et al. (1991). Intra and interobserver variability in cancer patients' performance status assessed according to Karnofsky and ECOG scales. Ann Oncol 2, 437-9.

62. Buccheri, G., Ferrigno, D., and Tamburini, M. (1996). Karnofsky and ECOG performance status scoring in lung cancer: a prospective, longitudinal study of 536 patients from a single institution. Eur J Cancer 32A, 1135-41.

63. Storniolo, A. M., Enas, N. H., Brown, C. A., Voi, M., Rothenberg, M. L., and Schilsky, R. (1999). An investigational new drug treatment program for patients with gemcitabine: results for over 3000 patients with pancreatic carcinoma. Cancer 85, 1261-8.

64. Rothenberg, M. L., Abbruzzese, J. L., Moore, M., Portenoy, R. K., Robertson, J. M., and Wanebo, H. J. (1996). A rationale for expanding the endpoints for clinical trials in advanced pancreatic carcinoma. Cancer 78, 627-32.

Page 77: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 72 -

65. Wiersema, M. J. (2001). Accuracy of endoscopic ultrasound in diagnosing and staging pancreatic carcinoma. Pancreatology 1, 625-32.

66. Howard, T. J., Chin, A. C., Streib, E. W., Kopecky, K. K., and Wiebke, E. A. (1997). Value of helical computed tomography, angiography, and endoscopic ultrasound in determining resectability of periampullary carcinoma. Am J Surg 174, 237-41.

67. Fritscher-Ravens, A., Knoefel, W. T., Krause, C., Swain, C. P., Brandt, L., and Patel, K. (2005). Three-dimensional linear endoscopic ultrasound-feasibility of a novel technique applied for the detection of vessel involvement of pancreatic masses. Am J Gastroenterol 100, 1296-302.

68. Miller, A. B., Hoogstraten, B., Staquet, M., and Winkler, A. (1981). Reporting results of cancer treatment. Cancer 47, 207-14.

69. Burris, H., and Storniolo, A. M. (1997). Assessing clinical benefit in the treatment of pancreas cancer: gemcitabine compared to 5-fluorouracil. Eur J Cancer 33 Suppl 1, S18-22.

70. Gattani, A. M., Mandeli, J., and Bruckner, H. W. (1996). Tumor markers in patients with pancreatic carcinoma. Cancer 78, 57-62.

71. Stemmler, J., Stieber, P., Szymala, A. M., Schalhorn, A., Schermuly, M. M., Wilkowski, R., Helmberger, T., Lamerz, R., Stoffregen, C., Niebler, K., Garbrecht, M., and Heinemann, V. (2003). Are Serial CA 19-9 Kinetics Helpful in Predicting Survival in Patients with Advanced or Metastatic Pancreatic Cancer Treated with Gemcitabine and Cisplatin? Onkologie 26, 462-7.

72. Lutz, M. P., Koniger, M., Muche, R., Ellenrieder, V., Steinkamp, M., Adler, G., and Gress, T. M. (1999). A phase II study of weekly 24-h infusion of high-dose 5-fluorouracil in advanced pancreatic cancer. Z Gastroenterol 37, 993-7.

73. Cartwright, T. H., Cohn, A., Varkey, J. A., Chen, Y. M., Szatrowski, T. P., Cox, J. V., and Schulz, J. J. (2002). Phase II study of oral capecitabine in patients with advanced or metastatic pancreatic cancer. J Clin Oncol 20, 160-4.

74. Wagener, D. J., Verdonk, H. E., Dirix, L. Y., Catimel, G., Siegenthaler, P., Buitenhuis, M., Mathieu-Boue, A., and Verweij, J. (1995). Phase II trial of CPT-11 in patients with advanced pancreatic cancer, an EORTC early clinical trials group study. Ann Oncol 6, 129-32.

75. Stehlin, J. S., Giovanella, B. C., Natelson, E. A., De Ipolyi, P. D., Coil, D., Davis, B., Wolk, D., Wallace, P., and Trojacek, A. (1999). A study of 9-nitrocamptothecin (RFS-2000) in patients with advanced pancreatic cancer. Int J Oncol 14, 821-31.

76. Okada, S., Sakata, Y., Matsuno, S., Kurihara, M., Sasaki, Y., Ohashi, Y., and Taguchi, T. (1999). Phase II study of docetaxel in patients with metastatic pancreatic cancer: a Japanese cooperative study. Cooperative Group of Docetaxel for Pancreatic Cancer in Japan. Br J Cancer 80, 438-43.

Page 78: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 73 -

77. Androulakis, N., Kourousis, C., Dimopoulos, M. A., Samelis, G., Kakolyris, S., Tsavaris, N., Genatas, K., Aravantinos, G., Papadimitriou, C., Karabekios, S., Stathopoulos, G. P., and Georgoulias, V. (1999). Treatment of pancreatic cancer with docetaxel and granulocyte colony-stimulating factor: a multicenter phase II study. J Clin Oncol 17, 1779-85.

78. Whitehead, R. P., Jacobson, J., Brown, T. D., Taylor, S. A., Weiss, G. R., and Macdonald, J. S. (1997). Phase II trial of paclitaxel and granulocyte colony-stimulating factor in patients with pancreatic carcinoma: a Southwest Oncology Group study. J Clin Oncol 15, 2414-9.

79. Rougier, P., Adenis, A., Ducreux, M., de Forni, M., Bonneterre, J., Dembak, M., Clouet, P., Lebecq, A., Baille, P., Lefresne-Soulas, F., Blanc, C., and Armand, J. P. (2000). A phase II study: docetaxel as first-line chemotherapy for advanced pancreatic adenocarcinoma. Eur J Cancer 36, 1016-25.

80. Wils, J. A., Kok, T., Wagener, D. J., Selleslags, J., and Duez, N. (1993). Activity of cisplatin in adenocarcinoma of the pancreas. Eur J Cancer 29A, 203-4.

81. Rothenberg, M. L., Moore, M. J., Cripps, M. C., Andersen, J. S., Portenoy, R. K., Burris, H. A., 3rd, Green, M. R., Tarassoff, P. G., Brown, T. D., Casper, E. S., Storniolo, A. M., and Von Hoff, D. D. (1996). A phase II trial of gemcitabine in patients with 5-FU-refractory pancreas cancer. Ann Oncol 7, 347-53.

82. Ulrich-Pur, H., Kornek, G. V., Raderer, M., Haider, K., Kwasny, W., Depisch, D., Greul, R., Schneeweiss, B., Krauss, G., Funovics, J., and Scheithauer, W. (2000). A phase II trial of biweekly high dose gemcitabine for patients with metastatic pancreatic adenocarcinoma. Cancer 88, 2505-11.

83. Tempero, M., Plunkett, W., Ruiz Van Haperen, V., Hainsworth, J., Hochster, H., Lenzi, R., and Abbruzzese, J. (2003). Randomized phase II comparison of dose-intense gemcitabine: thirty-minute infusion and fixed dose rate infusion in patients with pancreatic adenocarcinoma. J Clin Oncol 21, 3402-8.

84. Burris, H. A., 3rd, Moore, M. J., Andersen, J., Green, M. R., Rothenberg, M. L., Modiano, M. R., Cripps, M. C., Portenoy, R. K., Storniolo, A. M., Tarassoff, P., Nelson, R., Dorr, F. A., Stephens, C. D., and Von Hoff, D. D. (1997). Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15, 2403-13.

85. Bramhall, S. R., Rosemurgy, A., Brown, P. D., Bowry, C., and Buckels, J. A. (2001). Marimastat as first-line therapy for patients with unresectable pancreatic cancer: a randomized trial. J Clin Oncol 19, 3447-55.

86. Moore, M. J., Hamm, J., Dancey, J., Eisenberg, P. D., Dagenais, M., Fields, A., Hagan, K., Greenberg, B., Colwell, B., Zee, B., Tu, D., Ottaway, J., Humphrey, R., and Seymour, L. (2003). Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12-9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 21, 3296-302.

Page 79: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 74 -

87. Smith, D., and Gallagher, N. (2003). A phase II/III study comparing intravenous ZD9331 with gemcitabine in patients with pancreatic cancer. Eur J Cancer 39, 1377-83.

88. Van Custem, E., Karasek, P., and Oettle, H. (2002). Phase III trial comparing gemcitabine+R115777 (Zarnestra) versus gemcitabine+placebo in advanced pancreatic cancer (abstract). Proc Am Soc Clin Oncol 21, 517.

89. Cheverton, P., Friess, H., Salek, Geddes, C., Bodoky, G., Valle, J., and Humblet, Y. (2004). Phase III results of exatecan (DX-8951f) versus gemcitabine (Gem) in chemotherapy of patients with advanced pancreatic cancer (abstract). In "ASCO Fortieth Annual Meeting, New Orleans LA" (G. SM, Ed.), American Society of Clincal Oncology 2004.

90. Schulz, L., Schalhorn, A., and Wilmanns, W. (1998). Synergistic interaction of gemcitabine and 5-fluorouracil in colon cancer cells (abstract). Proc Am Soc Clin Oncol 17, A965.

91. Castellano, D., Paz-Ares, L., and Pronk, L. (2000). A phase I/II clinical and pharmacologic study of dose-escalating and dose-sequencing administration of gemcitabine and folinic acid/fluorouracil in advanced unresectable pancreatic cancer (abstract). Ann Oncol 11, 61.

92. Correale, P., Cerretani, D., Marsili, S., Pozzessere, D., Petrioli, R., Messinese, S., Sabatino, M., Roviello, F., Pinto, E., Francini, G., and Giorgi, G. (2003). Gemcitabine increases systemic 5-fluorouracil exposure in advanced cancer patients. Eur J Cancer 39, 1547-51.

93. Oettle, H., and Riess, H. (2002). Gemcitabine in combination with 5-fluorouracil with or without folinic acid in the treatment of pancreatic cancer. Cancer 95, 912-22.

94. Di Costanzo, F., Sdrobolini, A., Carlini, P., Massidda, B., Mattioli, R., Lop, A., Barletta, E., Moscetti, L., Recchia, F., Tralongo, P., and Ospedaliera, A. (2001). Gemcitabine alone or in combination with 5-FU continuous infusion in the treatment of advanced pancreatic cancer: a GOIRC reandomized phase II trial (abstract). Proc Am Soc Clin Oncol 20, 612.

95. Berlin, J. D., Catalano, P., Thomas, J. P., Kugler, J. W., Haller, D. G., and Benson, A. B., 3rd (2002). Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol 20, 3270-5.

96. Peters, G. J., Ruiz van Haperen, V. W., Bergman, A. M., Veerman, G., Smitskamp-Wilms, E., van Moorsel, C. J., Kuiper, C. M., and Braakhuis, B. J. (1996). Preclinical combination therapy with gemcitabine and mechanisms of resistance. Semin Oncol 23, 16-24.

97. Philip, P. A., Zalupski, M. M., Vaitkevicius, V. K., Arlauskas, P., Chaplen, R., Heilbrun, L. K., Adsay, V., Weaver, D., and Shields, A. F. (2001). Phase II study of gemcitabine and cisplatin in the treatment of patients with advanced pancreatic carcinoma. Cancer 92, 569-77.

98. Heinemann, V., Wilke, H., Mergenthaler, H. G., Clemens, M., Konig, H., Illiger, H. J., Arning, M., Schalhorn, A., Possinger, K., and Fink, U. (2000). Gemcitabine and cisplatin in the treatment of advanced or metastatic pancreatic cancer. Ann Oncol 11, 1399-403.

Page 80: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 75 -

99. Colucci, G., Giuliani, F., Gebbia, V., Biglietto, M., Rabitti, P., Uomo, G., Cigolari, S., Testa, A., Maiello, E., and Lopez, M. (2002). Gemcitabine alone or with cisplatin for the treatment of patients with locally advanced and/or metastatic pancreatic carcinoma: a prospective, randomized phase III study of the Gruppo Oncologia dell'Italia Meridionale. Cancer 94, 902-10.

100. Faivre, S., Raymond, E., Woynarowski, J. M., and Cvitkovic, E. (1999). Supraadditive effect of 2',2'-difluorodeoxycytidine (gemcitabine) in combination with oxaliplatin in human cancer cell lines. Cancer Chemother Pharmacol 44, 117-23.

101. Louvet, C., Andre, T., Lledo, G., Hammel, P., Bleiberg, H., Bouleuc, C., Gamelin, E., Flesch, M., Cvitkovic, E., and de Gramont, A. (2002). Gemcitabine combined with oxaliplatin in advanced pancreatic adenocarcinoma: final results of a GERCOR multicenter phase II study. J Clin Oncol 20, 1512-8.

102. Alberts, S. R., Townley, P. M., Goldberg, R. M., Cha, S. S., Sargent, D. J., Moore, D. F., Krook, J. E., Pitot, H. C., Fitch, T. R., Wiesenfeld, M., and Mailliard, J. A. (2003). Gemcitabine and oxaliplatin for metastatic pancreatic adenocarcinoma: a North Central Cancer Treatment Group phase II study. Ann Oncol 14, 580-5.

103. Louvet, C., Labianca, R., Hammel, P., Lledo, G., de Braud, F., Andre, T., Cantore, M., Ducreux, M., Zaniboni, A., and de Gramont, A. (2004). GemOx (Gemcitabine + Oxaliplatin) versus Gem (Gemcitabine) in non resectable pancreatic adenocarcinoma: final resutls of the GERCOR/GISCAD Intergroup Phase III. In "ASCO Fortieth Annual Meeting, New Orleans LA" (G. SM, Ed.), American Society of Clincal Oncology 2004.

104. Rocha Lima, C. M., Savarese, D., Bruckner, H., Dudek, A., Eckardt, J., Hainsworth, J., Yunus, F., Lester, E., Miller, W., Saville, W., Elfring, G. L., Locker, P. K., Compton, L. D., Miller, L. L., and Green, M. R. (2002). Irinotecan plus gemcitabine induces both radiographic and CA 19-9 tumor marker responses in patients with previously untreated advanced pancreatic cancer. J Clin Oncol 20, 1182-91.

105. Stathopoulos, G. P., Rigatos, S. K., Dimopoulos, M. A., Giannakakis, T., Foutzilas, G., Kouroussis, C., Janninis, D., Aravantinos, G., Androulakis, N., Agelaki, S., Stathopoulos, J. G., and Georgoulias, V. (2003). Treatment of pancreatic cancer with a combination of irinotecan (CPT-11) and gemcitabine: a multicenter phase II study by the Greek Cooperative Group for Pancreatic Cancer. Ann Oncol 14, 388-94.

106. Jacobs, A. D., Otero, H., and Picozzi, V. J. (2000). A phase I/II study of gemcitabine and docetaxel in patients with unresectable pancreatic cancer (abstract). Proc Am Soc Clin Oncol 19, 265.

107. Cascinu, S., Gasparini, G., Catalano, V., Silva, R. R., Pancera, G., Morabito, A., Giordani, P., Gattuso, D., and Catalano, G. (1999). A phase I-II study of gemcitabine and docetaxel in advanced pancreatic cancer: a report from the Italian Group for the Study of Digestive Tract Cancer (GISCAD). Ann Oncol 10, 1377-9.

108. Lueck, A., Ridwelski, K., and Kettner, E. (2000). Final results of a phase I study of weekly gemcitabine and docetaxel in pancreatic carcinoma and preliminary results of a phase II study (abstract). Proc Am Soc Clin Oncol 19, 318.

Page 81: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 76 -

109. Stathopoulos, G. P., Mavroudis, D., Tsavaris, N., Kouroussis, C., Aravantinos, G., Agelaki, S., Kakolyris, S., Rigatos, S. K., Karabekios, S., and Georgoulias, V. (2001). Treatment of pancreatic cancer with a combination of docetaxel, gemcitabine and granulocyte colony-stimulating factor: a phase II study of the Greek Cooperative Group for Pancreatic Cancer. Ann Oncol 12, 101-3.

110. Ryan, D. P., Kulke, M. H., Fuchs, C. S., Grossbard, M. L., Grossman, S. R., Morgan, J. A., Earle, C. C., Shivdasani, R., Kim, H., Mayer, R. J., and Clark, J. W. (2002). A Phase II study of gemcitabine and docetaxel in patients with metastatic pancreatic carcinoma. Cancer 94, 97-103.

111. Scheithauer, W., Kornek, G. V., Raderer, M., Hejna, M., Valencak, J., Miholic, J., Kovats, E., Lang, F., Funovics, J., Bareck, E., and Depisch, D. (1999). Phase II trial of gemcitabine, epirubicin and granulocyte colony-stimulating factor in patients with advanced pancreatic adenocarcinoma. Br J Cancer 80, 1797-802.

112. Neri, B., Cini, G., Doni, L., Fulignati, C., Turrini, M., Pantalone, D., Mini, E., De Luca Cardillo, C., Fioretto, L. M., Ribecco, A. S., Moretti, R., Scatizzi, M., Zocchi, G., and Quattrone, A. (2002). Weekly gemcitabine plus Epirubicin as effective chemotherapy for advanced pancreatic cancer: a multicenter phase II study. Br J Cancer 87, 497-501.

113. O'Reilly, E. M., Abou-Alfa, G. K., Letourneau, R., Harker, W. G., Modiano, M. R., Hurwitz, H., Tchekmedyian, N. S., Ackerman, J., De Jager, R. L., and Eckhardt, S. G. (2004). A randomized phase III trial of DX-8951f (Exatecan Mesylate; DX) and Gemcitabine (GEM) vs. Gemcitabine alone in advanced pancreatic cancer (abstract). In "ASCO Fortieth Annual Meeting, New Orleans LA" (G. SM, Ed.), American Society of Clincal Oncology 2004.

114. Richards, D. A., Kindler, H. L., Oettle, H., Ramanathan, R. K., Van Laethem, J. L., Peeters, M., Fuchs, M., John, W., Arning, M., and Von Hoff, D. (2004). A randomized phase III study comparing gemcitabine + pemetrexed versus gemcitabine in patients with locally advanced and metastatic pancreas cancer (abstract). In "ASCO Fortieth Annual Meeting, New Orleans LA" (G. SM, Ed.), American Society of Clincal Oncology 2004.

115. Doyle, T. H., Mornex, F., and McKenna, W. G. (2001). The clinical implications of gemcitabine radiosensitization. Clin Cancer Res 7, 226-8.

116. Lawrence, T. S., Eisbruch, A., McGinn, C. J., Fields, M. T., and Shewach, D. S. (1999). Radiosensitization by gemcitabine. Oncology (Huntingt) 13, 55-60.

117. McGinn, C. J., Lawrence, T. S., and Zalupski, M. M. (2002). On the development of gemcitabine-based chemoradiotherapy regimens in pancreatic cancer. Cancer 95, 933-40.

118. Ostruszka, L. J., and Shewach, D. S. (2000). The role of cell cycle progression in radiosensitization by 2',2'-difluoro-2'-deoxycytidine. Cancer Res 60, 6080-8.

119. Talamonti, M. S., Catalano, P. J., Vaughn, D. J., Whittington, R., Beauchamp, R. D., Berlin, J., and Benson, A. B., 3rd (2000). Eastern Cooperative Oncology Group Phase I trial of protracted venous infusion fluorouracil plus weekly gemcitabine with concurrent

Page 82: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 77 -

radiation therapy in patients with locally advanced pancreas cancer: a regimen with unexpected early toxicity. J Clin Oncol 18, 3384-9.

120. Osteen, R. T., Zinner, M. J., and Fuchs, C. S. (1999). Phase I trial of concurrent gemcitabine, infusional 5 fluorouracil and radiation therapy in patients with localized, unresectable pancreatic adenocarcinoma (abstract). Proc Am Soc Clin Oncol 18, 284.

121. Martenson, J. A., Vigliotti, A. P., Pitot, H. C., Geeraerts, L. H., Sargent, D. J., Haddock, M. G., Ghosh, C., Keppen, M. D., Fitch, T. R., and Goldberg, R. M. (2003). A phase I study of radiation therapy and twice-weekly gemcitabine and cisplatin in patients with locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys 55, 1305-10.

122. Safran, H., Dipetrillo, T., Iannitti, D., Quirk, D., Akerman, P., Cruff, D., Cioffi, W., Shah, S., Ramdin, N., and Rich, T. (2002). Gemcitabine, paclitaxel, and radiation for locally advanced pancreatic cancer: a Phase I trial. Int J Radiat Oncol Biol Phys 54, 137-41.

123. Karasek, P., Skacel, T., Kocakova, I., Bednarik, O., Petruzelka, L., Melichar, B., Bustova, I., Spurny, V., and Trason, T. (2003). Gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer: a prospective observational study. Expert Opin Pharmacother 4, 581-6.

124. Crino, L., Mosconi, A. M., Calandri, C., Corgna, E., Porrozzi, S., Chiara, S., Nobili, M. T., and Tonato, M. (2001). Gemcitabine in advanced pancreatic cancer: a phase II trial. Am J Clin Oncol 24, 296-8.

125. Meyer, F., Hirbaschek, A., Kuhn, R., Eichelmann, K., Luck, A., Lippert, H., and Ridwelski, K. (2001). Antineoplastic effect and quality of life in patients with pancreatic carcinoma who received gemcitabine monotherapy. Viszeralchirurgie 36, 85-90.

126. Okada, S., Ueno, H., Okusaka, T., Ikeda, M., Furuse, J., and Maru, Y. (2001). Phase I trial of gemcitabine in patients with advanced pancreatic cancer. Jpn J Clin Oncol 31, 7-12.

127. Aykan, F., Argon, A., Alici, S., and Bulutlar, G. (2000). A phase II trial of gemcitabine in patients with advanced pancreatic cancer (APC) (abstract). Proc Am Soc Clin Oncol 19, 1241.

128. Kurtz, J. E., Trillet-Lenoir, V., Bugat, R., Negrier, S., Adenis, A., Kayitalire, L., Ripoche, V., and Dufour, P. (1999). Compassionate use of gemcitabine in advanced pancreatic cancer: a French multicentric study. Bull Cancer 86, 202-206.

129. Manzano, H., Esquerdo, G., Rita, J., Agutar, D., Arrivi, A., Pazo, R., Martin, J., and Terrasa, J. (1998). Clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreatic cancer (abstract). Ann Oncol 9, 256.

130. Petrovic, Z., Jovic, J., Perisic, N., and Brocic-Pecelj, T. (1998). Monotherapy with gemcitabine in patients with advanced pancreatic carcinoma (abstract). Ann Oncol 9, 641.

131. Carmichael, J., Fink, U., Russell, R. C., Spittle, M. F., Harris, A. L., Spiessi, G., and Blatter, J. (1996). Phase II study of gemcitabine in patients with advanced pancreatic cancer. Br J Cancer 73, 101-5.

Page 83: Evolution de la prise en charge palliative du cancer du ... · 1 Protocole de recherche formellement approuvé par le Comité d’éthique des Hôpitaux Universitaires de Genève

- 78 -

132. Casper, E. S., Green, M. R., Kelsen, D. P., Heelan, R. T., Brown, T. D., Flombaum, C. D., Trochanowski, B., and Tarassoff, P. G. (1994). Phase II trial of gemcitabine (2,2'-difluorodeoxycytidine) in patients with adenocarcinoma of the pancreas. Invest New Drugs 12, 29-34.

133. Heinemann, V., Quietzsch, D., Gieseler, F., Schonekas, H., Rost, A., Neuhaus, H., Clemens, M., Szymala, M., Hiddemann, W., Schalhorn, A., and Grosshadern, K. (2001). Gemcitabine plus cisplatin versus gemcitabine in advanced pancreatic cancer : preliminary results of a randomized phase III trial (abstract). Proc Am Soc Clin Oncol 20, 625.

134. Kaplan, E. L., and Meier, P. (1958). Nonparametric estimation of incomplete observations. J Am Stat Assoc 53, 457-481.

135. (1976). "ICD-O International classification of diseases for oncology," 1st. ed., Geneva.

136. (1990). "ICD-O International classification of diseases for oncology," 2nd. ed., Geneva.

137. Feinstein, A. R., Sosin, D. M., and Wells, C. K. (1985). The Will Rogers phenomenon. Stage migration and new diagnostic techniques as a source of misleading statistics for survival in cancer. N Engl J Med 312, 1604-8.

138. Andre, T., Louvet, C., and de Gramont, A. (2004). Colon cancer: what is new in 2004? Bull Cancer 91, 75-80.

139. Schrag, D. (2004). The price tag on progress--chemotherapy for colorectal cancer. N Engl J Med 351, 317-9.