20.04.2013 Views

S3-Guideline “Exocrine Pancreatic Carcinoma” 20071 ... - DGVS

S3-Guideline “Exocrine Pancreatic Carcinoma” 20071 ... - DGVS

S3-Guideline “Exocrine Pancreatic Carcinoma” 20071 ... - DGVS

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong> <strong>Carcinoma”</strong> 2007 1<br />

Results of an Evidence-Based Consensus Conference<br />

(13. – 14.10.2006)<br />

<strong>S3</strong>-Leitlinie „Exokrines Pankreaskarzinom“ 2007<br />

Authors G. Adler, T. Seufferlein, S. C. Bischoff, H.-J. Brambs, S. Feuerbach, G. Grabenbauer, S. Hahn, V. Heinemann,<br />

W. Hohenberger, J. M. Langrehr, M. P. Lutz, O. Micke, H. Neuhaus, P. Neuhaus, H. Oettle, P. M. Schlag, R. Schmid,<br />

W. Schmiegel, K. Schlottmann, J. Werner, B. Wiedenmann, I. Kopp<br />

Affiliation Department of Internal Medicine I, University Medical Center Ulm<br />

Bibliography<br />

DOI 10.1055/s-2008-1027420<br />

Z Gastroenterol 2008; 46:<br />

449 – 482 Georg Thieme<br />

Verlag KG Stuttgart · New York ·<br />

ISSN 0044-2771<br />

Correspondence<br />

Prof. Dr. Guido Adler<br />

Department of Internal<br />

Medicine I, University Medical<br />

Center<br />

Robert-Koch-St. 8<br />

89081 Ulm<br />

Germany<br />

Tel.: ++ 49/7 31/50 04 45 00<br />

Fax: ++ 49/7 31/50 04 45 02<br />

guido.adler@uniklinik-ulm.de<br />

Introduction: Scope and aim of the<br />

guideline<br />

!<br />

More than 95% of pancreatic carcinomas are<br />

adenocarcinomas. They develop by malignant<br />

degeneration of the exocrine part of the pancreas.<br />

According to current knowledge, the pancreatic<br />

carcinoma develops from a premalignant<br />

stage of the epithelium of the pancreatic<br />

duct system (PanIN: pancreatic intraepithelial<br />

neoplasia). Cystic tumors that also arise from<br />

duct cells and acinar cell carcinoma which develops<br />

from secretory pancreatic parenchyma<br />

cells are not as common. Even less common<br />

are endocrine tumors that arise from endocrine<br />

cells in the islets of Langerhans.<br />

About 12800 people develop pancreatic carcinoma<br />

per year in Germany. Men and women are affected<br />

with about the same frequency. <strong>Pancreatic</strong><br />

carcinoma in men takes 9th place and in women<br />

7th place in the statistic of newly developed cancer<br />

in Germany. Most of the people that are affected<br />

develop the disease at an older age: The mean<br />

age at which men and women are diagnosed is 68<br />

and 75 years, respectively. <strong>Pancreatic</strong> carcinoma<br />

with 12100 deaths was the 5th most common<br />

cause of death in the year 2000. Thus, it causes<br />

about 6% of all cancer deaths. The pancreatic carcinoma<br />

incidence is very close to its annual mortality<br />

rate. Long-term survival is the exception.<br />

1 Commissioned by the German Society for Digestive and<br />

Metabolic Diseases (<strong>DGVS</strong>) und the German Cancer Society<br />

(DKG). In cooperation with the Task Force Radiologic<br />

Oncology of the German Cancer Society (ARO),<br />

Pancreatectomy Team e. V., German Society for Surgery<br />

(DGCH), German Society for Hematology and Oncology<br />

(DGHO), German Society for Palliative MedicineGerman<br />

Society for Pathology (DGP), German Society for Radiooncology<br />

(DEGRO), German Society for Visceral Surgery<br />

(CAO-V)/Surgical Task Force Oncology (CAO-V), German<br />

Society for Radiology (DRG), German Joint Societies for<br />

Clinical Chemistry and Laboratory Medicine (DGKL)<br />

(Chairmen: G. Adler, T. Seufferlein, I. Kopp)<br />

Leitlinie 449<br />

Thus, the 5-year survival rate for pancreatic carcinoma<br />

of 4% is the lowest of all malignant diseases.<br />

The reasons are the late diagnosis, the resulting<br />

low curative resection rate, and the rapid and<br />

aggressive metastasis.<br />

In the last few years important progress has been<br />

made not only in the understanding of pancreatic<br />

carcinoma development but also in its diagnosis<br />

and therapy. Therefore, it was in the interest of<br />

both the <strong>DGVS</strong> and the German Cancer Society<br />

to have a high-quality guideline compiled that is<br />

based on the best available scientific evidence<br />

and existing clinical experience.<br />

The aim of the guideline “exocrine pancreatic<br />

carcinoma” is to ensure an evidence based, comprehensive,<br />

and optimal care for patients with<br />

pancreatic cancer. The guideline is meant to accomplish:<br />

E early diagnosis of pancreatic carcinoma,<br />

E thus, the possibilty of a higher rate of resections<br />

with a curative intent,<br />

E considerably prolonged survival with a good<br />

quality of life in the palliative situation,<br />

E prolonged survival with a good quality of life<br />

in the postoperative situation,<br />

E strongly improved pain and malnourishment<br />

reduction during follow-up.<br />

The guideline addresses anyone involved in the<br />

diagnosis, therapy, and follow-up of outpatients<br />

or inpatients. According to the definition of<br />

guidelines, it is meant to help doctors and patients<br />

to decide on diagnostic and therapeutic<br />

procedures. The guideline does not release the<br />

doctor from his responsibility to individually<br />

examine the adequate procedure for the overall<br />

situation of each patient. Reasons should be given<br />

in case of deviation from the guideline. The<br />

physician’s task is to continuously ensure the<br />

quality of curative and palliative treatment.<br />

This guideline also addresses persons indirectly<br />

involved e. g. health care providers or medical<br />

services of the health insurance companies.<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


450<br />

Leitlinie<br />

The methodologic recommendations of the AWMF on the preparation<br />

of guidelines (http://www.awmf-leitlinien.de), the<br />

guideline manual of the Medical Center of Quality in Medicine<br />

(http://www.aezq.de), and the German Instrument on Methodologic<br />

<strong>Guideline</strong> Evaluation (http://www.delbi.de) were the<br />

basis for the organization of the guideline procedure. As is demonstrated<br />

in the following chapter (guideline report), the<br />

guideline fulfills the criteria of an evidence and consensusbased<br />

guideline (<strong>S3</strong>).<br />

This guideline is estimated to be valid for 3 years. If during this<br />

time important changes in care become evident, the coordination<br />

groups will decide whether individual topics or the complete<br />

guideline must be updated ahead of time.<br />

The preparation of the guideline was financially supported by<br />

the <strong>DGVS</strong> and the DKG. The work of the participants taking<br />

part in the consensus process and the resulting recommendations<br />

were not influenced by this support.<br />

This manuscript is the English version of the German guidelines<br />

that were published in the Zeitschrift für Gastroenetrologie<br />

in June 2007 [1].<br />

Organizational procedure and methodologic basis of<br />

the consensus process (guideline report)<br />

!<br />

Following the commission by the <strong>DGVS</strong> and the DKG, the organizational<br />

procedure for the guideline preparation was discussed<br />

with the chairman of the guideline commission, Prof. H.-K.<br />

Selbmann. Subsequent methodologic supervision was done by<br />

the vice chairman Dr. Ina Kopp. The guideline project was registered<br />

at the AWMF on July 4, 2005 (AWMF-Register-No. 032/<br />

010).<br />

Recruitment of participants<br />

To recruit the members of the steering committee (coordinators)<br />

presidents of the societies involved in diagnostics, therapy,<br />

and follow-up of pancreatic carcinoma were asked to<br />

name two representatives. After voting on the topics, the naming<br />

and invitation of task force members was performed together<br />

with the coordinators (l " Table 1). Participants were doctors<br />

from various care levels who work in hospitals and private<br />

practices. They represented the key disciplines that are relevant<br />

for pancreatic carcinoma: internal medicine (gastroenterology,<br />

hematology-oncology), surgery (visceral surgery), radiooncology,<br />

pathology, diagnostic radiology, palliative medicine,<br />

pain therapy, dietary medicine, psychotherapy-psychosomatics,<br />

laboratory medicine, and epidemiology. The patients’ representative<br />

was appointed by the Task Force of the Pancreatectomized<br />

Patients e. V. Dormagen.<br />

Organizational procedure<br />

Following the voting on the topics with the coordinators, the<br />

list of questions was generated and the search terms for the<br />

literature search were defined (time course l " Table 2). Afterwards<br />

the list of questions and the search terms were revised<br />

and amended by the task forces. A systematic literature<br />

search according to defined search terms was done in<br />

PubMed/Medline for the period from 1995 to May 2006 (inclusive).<br />

The search terms that were used are listed in the<br />

appendix. A total of 42345 citations were viewed (l " Table 3)<br />

and appendix (l " Table 10). 1409 were chosen according to<br />

the following criteria: investigations in humans, original arti-<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Table 1 Task forces<br />

1. Risk factors/screening/risk groups<br />

chairmen<br />

S.C. Bischoff Universität Hohenheim, Lehrstuhl für Ernährungsmedizin<br />

und Prävention, Stuttgart<br />

S. Hahn Knappschaftskrankenhaus, Medizinische Universitätsklinik,<br />

Bochum<br />

R. Schmid Klinikum rechts der Isar der TU, II. Medizinische<br />

Klinik und Poliklinik, München<br />

members<br />

G. Adler Universitätsklinikum, Zentrum für Innere Medizin,<br />

Klinik für Innere Medizin I, Ulm<br />

A. Aschoff Universitätsklinikum, Klinik für Diagnostische und<br />

Interventionelle Radiologie, Ulm<br />

I. Bergheim Universität Hohenheim, Lehrstuhl für Ernährungsmedizin<br />

und Prävention, Stuttgart<br />

T. Gress Universitätsklinikum Giessen und Marburg GmbH,<br />

Klinik für Innere Medizin, SP Gastroenterologie,<br />

Endokrinologie und Stoffwechsel, Marburg<br />

V. Keim Universitätsklinikum, Medizinische Klinik und Poliklinik<br />

II, Leipzig<br />

M. Lerch Universitätsklinikum, Klinik und Poliklinik für Innere<br />

Medizin A, Greifswald<br />

J. Lüttges Klinikum Saarbrücken, Pathologisches Institut,<br />

Saarbrücken<br />

G. Nagel Universität Ulm, Institut für Epidemiologie, Ulm<br />

H. Rieder Universitätsklinikum, Institut für Humangenetik<br />

und Anthropologie, Düsseldorf<br />

2. Diagnostics<br />

chairmen<br />

H.-J. Brambs Universitätsklinikum, Klinik für Diagnostische und<br />

Interventionelle Radiologie, Ulm<br />

K. Schlottmann Katharinen-Hospital gGmbH, Innere Klinik I, Unna<br />

B. Wiedenmann CharitØ Universitätsmedizin, CharitØCentrum 13,<br />

Medizinische Klinik mit SP Hepatologie und Gastroenterologie,<br />

Campus Virchow, Berlin<br />

members<br />

M. Dobritz Klinikum rechts der Isar der TU, Institut für Rçntgendiagnostik,<br />

München<br />

P. Mçller Universitätsklinikum, Comprehensive Cancer<br />

Center, Institut für Pathologie, Ulm<br />

J. Mçssner Universitätsklinikum, Zentrum für Innere Medizin,<br />

Medizinische Klinik und Poliklinik II; Gastroenterologie<br />

und Hepatologie, Leipzig<br />

S.T. Post Universitätsklinikum gGmbH, Chirurgische Klinik,<br />

Mannheim<br />

S.N. Reske Universitätsklinikum, Klinik für Nuklearmedizin,<br />

Ulm<br />

J.F.Riemann Klinikum der Stadt Ludwigshafen am Rhein<br />

gGmbH, Medizinische Klinik C, Ludwigshafen<br />

T. Rçsch CharitØ Universitätsmedizin, CharitØCentrum 13,<br />

Medizinische Klinik mit SP Hepatologie und Gastroenterologie,<br />

Zentrale interdisziplinäre Endoskopie,<br />

Campus Virchow, Berlin<br />

A.-O. Schäfer Universitätsklinikum, Radiologische Universitätsklinik,<br />

Rçntgendiagnostik, Freiburg<br />

W. Uhl St. Josef-Hospital, Klinikum der Ruhr-Universität,<br />

Klinik für Chirurgie, Bochum<br />

C. Wagener Universitätsklinikum, Institut für Klinische Chemie/Zentrallaboratorien,<br />

Hamburg-Eppendorf<br />

3. Surgical therapy of pancreatic carcinoma (curative intention)<br />

chairmen<br />

J. Werner Universitätsklinikum, Chirurgische Klinik, Klinik<br />

für Allgemein-, Viszeral- und Transplantationschirurgie,<br />

Heidelberg<br />

W. Hohenberger Chirurgische Klinik mit Poliklinik der Friedrich-<br />

Alexander-Universität Erlangen-Nürnberg, Allgemeine,<br />

Bauch- und Thoraxchirurgie, Erlangen<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Table 1 (Fortsetzung)<br />

members<br />

G. Baretton Universitätsklinikum, Institut für Pathologie,<br />

Dresden<br />

R. Bittner Marienhospital, Klinik für Allgemein- und<br />

Viszeralchirurgie, Stuttgart<br />

D. Dittert Universitätsklinikum, Institut für Pathologie,<br />

Dresden<br />

P. Galle Klinikum der Johannes Gutenberg-Universität, I.<br />

Medizinische Klinik und Poliklinik, Mainz<br />

D. Henne-Bruns Universitätsklinikum, Zentrum für Chirurgie, Klinik<br />

für Allgemein-, Viszeral- und<br />

Transplantationschirurgie, Ulm<br />

U.T. Hopt Universitätsklinikum, Chirurgische Universitätsklinik,<br />

Allgemein- und Viszeralchirurgie, Freiburg<br />

J. Izbicki Universitätsklinikum, Klinik und Poliklinik für Allgemein-,<br />

Viszeral- und Thoraxchirurgie, Hamburg-<br />

Eppendorf<br />

G. Klçppel Universitätsklinikum Schleswig-Holstein, Institut<br />

für Pathologie, Campus Kiel<br />

K.H. Link Asklepios Paulinen Klinik, Chirurgisches Zentrum,<br />

Wiesbaden<br />

P. Neuhaus CharitØ Universitätsmedizin, CharitØCentrum 8<br />

für Chirurgische Medizin, Klinik für Allgemein-,<br />

Visceral- und Transplantationschirurgie (CVK),<br />

Berlin<br />

J. Pelz Chirurgische Klinik mit Poliklinik der Friedrich-<br />

Alexander-Universität Erlangen-Nürnberg, Allgemeine,<br />

Bauch- und Thoraxchirurgie, Erlangen<br />

B. Rau Universitätsklinikum des Saarlandes, Klinik für<br />

Allgemeine Chirurgie, Viszeral-, Gefäß und Kinderchirurgie,<br />

Homburg/Saar<br />

P. Schlag CharitØ Universitätsmedizin, Robert-Rçssle-Klinik,<br />

Klinik für Chirurgie und Chirurgische Onkologie,<br />

Campus Berlin-Buch, Berlin<br />

H.D. Saeger Universitätsklinikum, Klinik und Poliklinik für<br />

Viszeral-, Thorax- u. Gefäßchirurgie, Dresden<br />

4. Neoadjuvant and adjuvant non-surgical therapy of pancreatic carcinoma<br />

chairmen<br />

J.M. Langrehr CharitØ Universitätsmedizin, CharitØCentrum 8<br />

für Chirurgische Medizin, Klinik für Allgemein-,<br />

Visceral- und Transplantationschirurgie (CVK),<br />

Berlin<br />

H. Oettle CharitØ Universitätsmedizin, Medizinische Klinik<br />

m.S. Hämatologie und Onkologie, Campus<br />

Virchow, Berlin<br />

W. Schmiegel Knappschaftskrankenhaus, Medizinische Universitätsklinik,<br />

Bochum<br />

members<br />

C. Bokemeyer Universitätsklinikum, II. Medizinische Klinik und<br />

Poliklinik, Hamburg-Eppendorf<br />

Th. Brunner University of Oxford, Churchill Hospital, Radiation<br />

Oncology and Biology; Headington, Oxford<br />

W. Budach Universitätsklinikum, Klinik und Poliklinik für<br />

Strahlentherapie und radiologische Onkologie,<br />

Düsseldorf<br />

H. Friess Universitätsklinikum, Chirurgische Klinik, Allgemein-,<br />

Viszeral- und Transplantationschirurgie,<br />

Heidelberg<br />

M. Geissler Klinikum Esslingen, Klinik für Onkologie, Gastroenterologie<br />

und Allgemeine Innere Medizin,<br />

Esslingen<br />

T. Meyer Chirurgische Klinik mit Poliklinik der Friedrich-<br />

Alexander-Universität Erlangen-Nürnberg, Allgemeine,<br />

Bauch- und Thoraxchirurgie, Erlangen<br />

Table 1 (Fortsetzung)<br />

A. Reinacher-<br />

Schick<br />

Knappschaftskrankenhaus, Medizinische Universitätsklinik,<br />

Bochum<br />

H.J. Schmoll Universitätsklinikum, Zentrum für Innere Medizin,<br />

Universitätsklinik und Poliklinik für Innere Medizin<br />

IV, Halle (Saale)<br />

H. Witzigmann Krankenhaus Dresden-Friedrichstadt, Klinik für<br />

Allgemein- und Abdominalchirurgie, Dresden<br />

5. Palliative therapy<br />

chairmen<br />

G. Grabenbauer Radiologische Gemeinschaftspraxis am Klinikum<br />

Coburg<br />

V. Heinemann Universitätsklinikum Großhadern, Medizinische<br />

Klinik und Poliklinik III, München<br />

M.P. Lutz Caritasklinik St. Theresia, Medizinische Klinik,<br />

Saarbrücken<br />

members<br />

U.R. Fçlsch Universitätsklinikum, Klinik für Allgemeine Innere<br />

Medizin, Kiel<br />

J.T. Hartmann Universitätsklinikum, Medizinische Universitätsklinik,<br />

Innere Medizin II, Tübingen<br />

S. Hegewisch- Internistische Gemeinschaftspraxis Eppendorf<br />

Becker<br />

F. Kullmann Klinikum der Universität, Klinik und Poliklinik für<br />

Innere Medizin I, Regensburg<br />

M. Lçhr Universitätsklinikum, II. Medizinische Universitätsklinik,<br />

Mannheim<br />

R. Porschen Klinikum Bremen Ost, Zentrum für Innere Medizin,<br />

Medizinische Klinik, Bremen<br />

M. Schilling Universitätsklinikum des Saarlandes, Klinik für<br />

Allgemeine Chirurgie, Viszeral-,Gefäß- und Kinderchirurgie,<br />

Homburg/Saar<br />

T. Seufferlein Universitätsklinikum, Zentrum für Innere Medizin,<br />

Klinik für Innere Medizin I, Ulm<br />

6. Supportive therapy and follow-up<br />

chairmen<br />

S. Feuerbach Klinikum der Universität, Institut für Rçntgendiagnostik,<br />

Regensburg<br />

O. Micke Franziskus Hospital gGmbH, Klinik für Strahlenheilkunde<br />

und Radioonkologie, Bielefeld<br />

H. Neuhaus Evangelisches Krankenhaus, Medizinische Klinik,<br />

Düsseldorf<br />

members<br />

C.F. Dietrich Caritas-Krankenhaus, Innere Medizin II, Bad Mergentheim<br />

A. Feil Arbeitskreis der Pankreatektomierten e.V., Siegen<br />

U. Graeven Kliniken Mariahilf, Klinik für Hämatologie, Onkologie<br />

und Gastroenterologie, Mçnchengladbach<br />

G. Hege-Scheuing<br />

Universitätsklinikum, Klinik für Anästhesiologie,<br />

Ulm<br />

Leitlinie 451<br />

E. Klar Universitätsklinikum, Abteilung für Allgemeine,<br />

Thorax-, Gefäß- und Transplantationschirurgie,<br />

Rostock<br />

F. Lordick Klinikum rechts der Isar der TU, III. Medizinische<br />

Klinik, München<br />

S. Pauls Universitätsklinikum, Klinik für Diagnostische und<br />

Interventionelle Radiologie, Ulm<br />

W. Scheppach Universitätsklinikum, Medizinische Klinik und Poliklinik<br />

I, Würzburg<br />

J. Weitz Universitätsklinikum, Klinik für Allgemein-,<br />

Viszeral- und Transplantationschirurgie, Sektion<br />

Chirurgische Onkologie, Heidelberg<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


452<br />

Leitlinie<br />

Table 2 Time course of consensus process<br />

category aspect time period<br />

initiation determination of conference date 1/2006<br />

choice of conference participants 1/2006<br />

preparation, revision, and amendment of the questionnaire by the respective task forces (topic 1 – 6) 2 – 3/2006<br />

literature search determination of search terms for the literature search 4/2006<br />

systematic literature search 5/2006<br />

delphi-questionnaire dispatch of final questionnaire including publications from 1995 on mid 5/2006<br />

response to questionnaire by task forces 6 – 7/2006<br />

summary/evaluation of results of the questionnaire by coordinators and dispatch with recommendations<br />

to all consensus conference participants<br />

9 – 2006<br />

conference preliminary discussion of consensus conference<br />

consensus conference:<br />

12.9.2006<br />

task force meetings 13.10.2006<br />

plenary session 13.–14.10.2006<br />

preparation of manuscript by consensus conference coordinators and chairmen until 2/2007<br />

decisions on open issues by questioning all concensus conference participants via email 5.4.2007<br />

dispatch of final manuscript to all conference participants and incorporation of all comments 15.4.2007<br />

peer review of the manuscript 6.5.2007<br />

Table 3 Literature search<br />

topic references initial selected references<br />

topic 1 16584 258<br />

topic 2 12419 407<br />

topic 3 5 994 202<br />

topic 4 2 739 111<br />

topic 5 2 241 262<br />

topic 6 2 368 169<br />

total 42345 1 409<br />

Table 4 Basis for evidence level: Center of Evidence Based Medicine Oxford,<br />

Topics 1, 3, 4, 5, and 6<br />

recommendation<br />

grade<br />

evidence<br />

level<br />

A 1 systematic review (SR) with<br />

homogeneity (no heterogeneity<br />

with respect to the results of individual<br />

studies) of randomized<br />

controlled studies (RCT)<br />

B 2a<br />

systematic review with homogeneity<br />

of cohort studies<br />

2b<br />

Individual cohort studies plus<br />

RCTs of low quality (e. g. followup<br />

< 80%)<br />

C 3 systematic reviews with homogeneity<br />

of case-control-studies as<br />

well as individual case-controlstudies:<br />

EL 3<br />

C 4 case studies and cohort studies as<br />

well as case-control-studies of low<br />

quality (i. e. cohorts: no clearly<br />

defined group for comparison, no<br />

outcome/exposition assessment<br />

in experimental and control<br />

groups, insufficient follow-up;<br />

case-control-studies: no clearly<br />

defined group for comparison)<br />

D 5 expert opinion or inconsistent or<br />

inconclusive studies of every evidence<br />

level<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

cles, or systematic reviews, and the number of examined or<br />

treated patients (> 10), no case reports. The literature was<br />

made available as an Endnote-file. In May 2006 the final<br />

questionnaires and the abstracts of the citations were sent<br />

to all members of the guideline process (n = 79). The members<br />

were requested to evaluate them by the middle of July<br />

(Delphi-technique). Until the end of August, the coordinators<br />

of the individual topics summarized the results from the<br />

task forces and developed recommendations for consensus<br />

finding. Four weeks before the consensus conference, a preliminary<br />

discussion took place with the coordinators of the<br />

individual topics. The evaluation table of the list of questions<br />

and the methodology for consensus finding during the consensus<br />

conference were discussed. The documents were sent<br />

to all participants of the consensus conference by the middle<br />

of September.<br />

The consensus conference took place on October 13 th and 14 th ,<br />

2006 in Stuttgart-Hohenheim. On the first day, the suggestions<br />

for consensus finding for the individual topics were formulated<br />

within the task forces (nominal group process, n = 7 – 12).<br />

Dr. Kopp (AWMF) was available for support in methodologic<br />

questions. On the second day, the consensus suggestions were<br />

presented, discussed, and if necessary modified in the plenum<br />

(n = 57). The voting of the evidence level and recommendation<br />

grade was performed using a TED-system.<br />

Evidence level and recommendation grade classification<br />

The evidence level of the relevant studies was defined according<br />

to the recommendations of the Center for Evidence-Based<br />

Medicine, Oxford, UK (http://www.cebm.net/) (l " Table 4). During<br />

the consensus conference preparation, it was discovered<br />

that these criteria do not apply to individual studies of Topic<br />

2 (diagnostics). Therefore, a modified classification of the evidence<br />

level was employed for the evaluation of the literature<br />

on diagnostics (l " Table 5). For all topics within the group processes<br />

it was considered whether the results of the studies are<br />

applicable to the guideline target population in Germany.<br />

The recommendation grades were assigned based on the evidence<br />

level. The classification of the recommendation grade is<br />

shown in (l " Table 6). In most cases the evidence level determines<br />

the recommendation grade (l " Table 4, 6). In justified<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Table 5 Basis for evidence level: Centre for Evidence-Based Medicine Oxford,<br />

Topic 2<br />

recommendation<br />

grade<br />

evidence<br />

level<br />

A 1a<br />

Systematic review (SR) with homogeneity<br />

(no heterogeneity with respect<br />

to results of individual studies) of level<br />

1 diagnostic studies. Clinical decision<br />

rule with 1b studies also from different<br />

clinical centers?<br />

1b<br />

Validating cohort studies (test quality<br />

of specific tests based on existing evidence,<br />

no explorative data collection<br />

with regression analysis)<br />

1c<br />

Specificity of diagnostic results that is<br />

so high (“absolute”) that a positive result<br />

leads to diagnosis or sensitivity of<br />

diagnostic results that is so high (“absolute”)<br />

that a negative result leads to<br />

exclusion of the diagnosis.<br />

B 2a<br />

Systematic review with homogeneity<br />

of diagnostic studies > level 2<br />

2b<br />

Explorative cohort studies with good<br />

reference basis (i. e. independent of<br />

test, blinded, or objective for all included<br />

subjects).<br />

C 3 systematic overview with homogeneity<br />

of studies EL3b or better<br />

C 4 case-control-studies of poor or reference<br />

standard that is not independent<br />

D 5 expert opinion without critical discussion<br />

Table 6 Recommendation grade classification<br />

A<br />

B<br />

C<br />

D<br />

consistent studies with evidence level 1 available<br />

consistent studies with evidence level 2 or 3 or extrapolations of<br />

studies with evidence level 1<br />

studies with evidence level 4 or extrapolations of studies with<br />

evidence level 2 or 3<br />

expert opinion or inconsistent or inconclusive studies of every<br />

evidence level<br />

cases, a deviation from this rule within the consensus conference<br />

was allowed:<br />

E consistency and effect size of the studies,<br />

E consideration of benefits, risks, and side effects,<br />

E applicability on extended patient groups, on the German<br />

health care system, or on the availability of resources.<br />

Consensus degree classification<br />

The TED-system was used to vote in the plenum of the consensus<br />

conference. Thus, immediately after the vote on individual<br />

questions, the absolute or percent approval of the participants<br />

was shown electronically. This resulted in the classification of<br />

the consensus magnitude (l " Table 7).<br />

<strong>Guideline</strong> implementation<br />

The quality of the guideline cannot be assessed before it has<br />

been implemented in the daily health care routine. Aside from<br />

the present scientific publication, the guideline will be published<br />

electronically by the AWMF in its information system<br />

“AWMF online” (http://www.awmf.org/). The German Cancer<br />

Table 7 Consensus size classification<br />

strong consensus approval of > 95% of participants<br />

consensus approval of > 75 – 95% of participants<br />

majority approval approval of > 50 – 75% of participants<br />

no consensus approval of < 50% of participants<br />

Leitlinie 453<br />

Society and the German Society for Digestive and Metabolic<br />

Diseases (<strong>DGVS</strong>) will also make the guideline available electronically.<br />

Following the publication of the complete version of<br />

the guideline, a short version and a version for patients will<br />

be introduced.<br />

Topic 1:<br />

Risk factors/screening/risk groups<br />

!<br />

Risk factors<br />

Introduction<br />

For many years diet has been discussed as a possible risk factor<br />

for the development of exocrine pancreatic carcinoma. However,<br />

there are no unanimous recommendations or recommendations<br />

established in clinical practice as to whether dietary<br />

procedures are effective in the prevention of pancreatic cancer<br />

and if so, which dietary factors play a role. Aside from the dietary<br />

factors, life style and work-related exposure must be discussed<br />

as possible risk factors for the development of pancreatic<br />

carcinoma. Finally, it will be discussed whether drug prophylaxis<br />

can reduce the risk of developing pancreatic carcinoma.<br />

Diet<br />

Diet recommendations<br />

Currently, a specific diet recommendation for the risk reduction<br />

of pancreatic carcinoma cannot be given. The current diet recommendations<br />

of the German Society for Nutrition (DGE) should be<br />

followed to reduce the risk of pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 2b, strong consensus<br />

Comments<br />

Some connection between dietary factors and pancreatic carcinoma<br />

was considered likely in a comprehensive literature<br />

review of the World Cancer Research Fund in 1997. However,<br />

these associations were not rated as convincing [2]. In the last<br />

12 years, connections between dietary factors and the development<br />

of pancreatic carcinoma were repeatedly found in original<br />

publications. They were rated with evidence levels 2b to<br />

3b [3 – 7]. However, there is also one group of investigators<br />

who found no association between dietary factors and pancreatic<br />

carcinoma in their cohort study [8, 9]. Nonetheless, a<br />

dietary recommendation to reduce pancreatic carcinoma risk<br />

is indicated, particularly because an association seems plausible<br />

from a biologic point of view [10].<br />

There is no clear relationship between fiber intake and risk of<br />

pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 3, strong consensus<br />

Comments<br />

The studies on fiber intake are contradictory. A protective effect<br />

of an increased intake of fiber was described in two case control<br />

studies [4, 11]. However, this was not confirmed in another<br />

case control study in men [12]. Due to the overall insufficient<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


454<br />

Leitlinie<br />

studies with contradictory results, a positive recommendation<br />

cannot be given.<br />

Recommendation<br />

An increased intake of legumes cannot be recommended to reduce<br />

the risk of pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 2b, strong consensus<br />

Comments<br />

A protective effect of legumes on the incidence of pancreatic<br />

carcinoma was found in two cohort studies that were done on<br />

subpopulations (Adventists) [13, 14]. Since both publications<br />

included a very special study population a selection bias cannot<br />

be excluded. Thus, a general recommendation cannot be<br />

given.<br />

A benefit of an increased fruit and vegetable intake to reduce<br />

the risk of pancreatic cancer can currently not be proven.<br />

Recommendation grade: C, evidence level: 2b, strong consensus<br />

Comments<br />

Increased fruit and vegetable consumption reduced the pancreatic<br />

carcinoma risk in several case control studies [3, 4, 15,<br />

16]. However, in a cohort study, which is considered more relevant,<br />

no relationship was observed between fruit and vegetable<br />

consumption and pancreatic carcinoma risk [17]. Nonetheless,<br />

a recommendation that encourages eating fruits and<br />

vegetables is rated as desirable, because regular consumption<br />

decreases the risk of cancer in general [18].<br />

Recommendation<br />

Consuming vitamin C rich foods may help reduce the risk of pancreatic<br />

carcinoma.<br />

Recommendation grade: D, evidence level 3, majority approval<br />

Comments<br />

Two case control studies indicate a protective relationship between<br />

the consumption of vitamin C rich foods and pancreatic<br />

carcinoma [4, 19]. However, the recommendation grade is restricted<br />

by limitations of both studies. Parameters such as age<br />

and smoking are not sufficiently taken into account by Lin et al.<br />

[19] and the population size is too small. Ji et al. [4] only found<br />

a significant association between vitamin C rich food and the<br />

risk of pancreatic carcinoma in men.<br />

A low-fat diet does not help reduce pancreatic carcinoma risk.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

An increased consumption of saturated fats was associated with<br />

a higher pancreatic carcinoma risk in a cohort study with male<br />

smokers [20]. This result is consistent with an ecologic study<br />

[21]. However, in other cohort studies there was no relationship<br />

[7], and in a study with an Asian population an inverse association<br />

was even described [4].<br />

A low-cholesterol diet does not help reduce the risk of pancreatic<br />

carcinoma.<br />

Recommendation grade: B, evidence level 2b, strong consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Comments<br />

In a case control study an increased pancreatic carcinoma risk<br />

was seen in the group with a high cholesterol diet [19]. However,<br />

this observation could not be confirmed in a cohort study<br />

[8].<br />

The reduction of red meat consumption does not help reduce<br />

the risk of pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 2b, strong consensus<br />

Comments<br />

The studies on this topic are contradictory. The consumption of<br />

red meat was connected with an increased risk of pancreatic<br />

carcinoma in a cohort study [7]. However, it remained unclear<br />

whether the red meat itself or the way it was prepared was responsible<br />

for the risk association [22]. No relationship between<br />

the consumption of red meat and the risk of pancreatic carcinoma<br />

was found in two other studies of which one was a cohort<br />

study by Michaud et al. [4, 8]. Therefore, it must be assumed<br />

that the positive association described by Nothlings [7]<br />

is most likely caused by the way the meat was prepared rather<br />

than by the consumption of the meat itself. This leads to the<br />

overall evaluation that reducing red meat consumption is not<br />

associated with a reduced pancreatic carcinoma risk.<br />

A relationship between the preferred consumption of white<br />

meat and pancreatic carcinoma risk cannot be reported.<br />

Recommendation grade: B, evidence level 2b, strong consensus<br />

Comments<br />

There are two studies that deal with this topic. Both found no<br />

association between the consumption of white meat and pancreatic<br />

carcinoma risk [7, 22].<br />

The consumption of smoked/grilled food may be associated<br />

with an increased risk of pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 3, majority approval<br />

Comments<br />

Three studies show that the consumption of smoked/grilled<br />

foods is associated with a higher pancreatic carcinoma risk<br />

[22 – 24]. However, all three studies are merely case control<br />

studies so that an evidence level of 3 can be given.<br />

An increased consumption of fish should not be recommended<br />

to decrease the risk of pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

Two cohort studies [7, 8] showed no relationship between fish<br />

consumption and pancreatic carcinoma risk. Thus, two case control<br />

studies that showed a protective effect are less important [5,<br />

25].<br />

A general recommendation to reduce the consumption of<br />

sugar cannot be given.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

There is evidence that the consumption of sugar is associated<br />

with an increased risk of pancreatic carcinoma [24, 26, 27].<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


However, a possible statistically significant relationship was<br />

reported only in women [28].<br />

An increased consumption of milk and milk products does not<br />

lead to a risk reduction of pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

A connection between milk and cheese consumption and the<br />

risk of pancreatic carcinoma was ruled out [7]. However, a protective<br />

effect against pancreatic carcinoma was reported for<br />

fermented milk products [11]. Since these data pertain only to<br />

certain milk products and not to milk and all milk products, a<br />

general recommendation cannot be given for this food group.<br />

The protective effect of fermented milk products has so far<br />

only been proven in a single study.<br />

Giving up excessive alcohol consumption may be recommended<br />

to reduce the risk of pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 3, strong consensus<br />

Comments<br />

Numerous studies showed no connection between moderate alcohol<br />

consumption and pancreatic carcinoma [29 –32]. However,<br />

several studies indicate that very high alcohol consumption<br />

or binge-drinking may be associated with an increased pancreatic<br />

carcinoma risk [32 – 34]. Therefore, it is recommended that<br />

alcohol consumption should be limited to a moderate quantity.<br />

This is especially the case because increased alcohol consumption<br />

may be connected to the pathogenesis of other malignant<br />

diseases. It may also result in chronic pancreatic and hepatic diseases.<br />

A general renouncement of coffee cannot be recommended.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

High consumption of coffee (more than three cups per day) was<br />

associated with an increased risk of pancreatic carcinoma in several<br />

case control studies [35– 37]. This could not be confirmed in<br />

several other cohort studies [29, 30, 40] with the exception of<br />

two [38, 39].<br />

An increased consumption of tea to reduce the pancreatic<br />

carcinoma risk cannot be recommended.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

In general, there is no connection between tea consumption and<br />

pancreatic carcinoma risk [38, 41]. Green tea may possibly have<br />

a protective effect on the development of pancreatic carcinoma<br />

[4]. In contrast, a cohort study that was done in Japan did not<br />

show such a connection [42]. These data may only apply to Europe.<br />

Life style<br />

Recommendations<br />

Recommendations on life style are indicated to reduce the risk of<br />

pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Leitlinie 455<br />

Comments<br />

Results from family studies show that aside from a genetic component<br />

certain life styles are associated with a higher risk of developing<br />

pancreatic carcinoma [43, 44]. Especially smoking and<br />

obesity have been proven as risk factors for pancreatic carcinoma<br />

[45]. This will be described in the following in more detail.<br />

Obesity is associated with an increased risk of pancreatic<br />

carcinoma. Therefore, excess weight should be avoided.<br />

Recommendation grade: A, evidence level 2a, strong consensus<br />

Comments<br />

A meta-analysis [46], as well as five cohort studies [47 –51], and<br />

one case control study [52] found that various obese populations<br />

(BMI > 30) had an increased pancreatic carcinoma risk. Thus, a<br />

clear connection can be assumed.<br />

A general recommendation to encourage exercise to control<br />

weight can be given.<br />

Recommendation grade: C, evidence level 2b, consensus<br />

Comments<br />

The results from cohort studies [47, 51, 53] and a case control<br />

study [54] indicate a protective effect of exercise on the risk of<br />

pancreatic carcinoma. Particularly overweight persons seem to<br />

benefit from exercise [47]. However, a detailed recommendation<br />

cannot be given, because contradictory results were also<br />

found [48], and not all observations and associations were significant<br />

[51] or consistent [47].<br />

Avoiding tobacco consumption is recommended to reduce<br />

the risk of pancreatic carcinoma.<br />

Recommendation grade: A, evidence level 2b, strong consensus<br />

Comments<br />

Smoking tobacco doubles the risk of pancreatic carcinoma. This<br />

relationship is consistently proven by cohort studies [39, 50,<br />

55–57] and case control studies [58, 59]. Individual genetic<br />

factors seem to influence the degree of association [60 – 62]. A<br />

connection was found even between passive smoking and risk<br />

of pancreatic carcinoma [63].<br />

Work-related risk factors<br />

Recommendation<br />

Contact with pesticides, herbicides, and fungicides may increase<br />

the risk of pancreatic carcinoma. Other potential risk factors<br />

may be chlorinated hydrocarbons, chromium and chromium<br />

compounds, electromagnetic fields, and fuel fumes.<br />

Recommendation grade: C, evidence level 2b–3, consensus<br />

Comments<br />

Some occupational and work areas seem to be associated with<br />

a slightly increased disease risk [64 – 66]. The abovementioned<br />

chemicals may possibly represent particular risk factors [67–<br />

73].<br />

Drug prophylaxis<br />

Recommendation<br />

There is at present no drug prophylaxis to reduce the risk of pancreatic<br />

carcinoma.<br />

Recommendation grade: B, evidence level 2a, consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


456<br />

Leitlinie<br />

Comments<br />

Neither the supplementation with antioxidants [74, 75] nor the<br />

intake of non-steroidal antirheumatics [76–78] leads to a reduction<br />

of pancreatic carcinoma risk.<br />

Screening of asymptomatic population<br />

Recommendation<br />

A screening of asymptomatic persons with CA19–9 for early diagnosis<br />

should not be done.<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Comment<br />

Two studies with large study populations show that screening<br />

of asymptomatic persons is not sensible due to the low positive<br />

predictive value [79, 80].<br />

Recommendation<br />

Molecular biologic screening methods such as mutation analysis<br />

to screen the asymptomatic normal population are currently not<br />

recommended.<br />

Recommendation grade: A, evidence level 5, strong consensus<br />

Comment<br />

There is currently no scientific evidence which warrants the<br />

screening of the asymptomatic normal population with molecular<br />

biologic methods.<br />

Recommendation<br />

Imaging techniques to screen the asymptomatic normal population<br />

can currently not be recommended.<br />

Recommendation grade: C, evidence level 3b, strong consensus<br />

Comment<br />

Currently, there is no scientific evidence that mandates the<br />

screening of the asymptomatic normal population with imaging<br />

techniques [79].<br />

Risk group – identification and monitoring<br />

Sporadic pancreatic carcinoma in the family (sporadic<br />

pancreatic cancer kindred: SPC)<br />

Recommendation<br />

Compared to the normal population, first degree relatives of patients<br />

with pancreatic carcinoma have an increased risk of also<br />

developing pancreatic cancer.<br />

Evidence level 2b, consensus<br />

Comments<br />

The risk is twice as high for first degree relatives of patients<br />

with pancreatic carcinoma. If the patient is under 60 years of<br />

age, the risk is 3 times as high [81, 82]. Second and third degree<br />

relatives are not suitable as index patients to define an individually<br />

increased pancreatic carcinoma risk. If pancreatic<br />

carcinoma occurs in two or more first degree relatives in one<br />

family, this family fulfills the currently valid criteria of “familial<br />

pancreatic carcinoma”. Thus, it does not belong to the group<br />

of sporadic pancreatic carcinoma.<br />

Recommendation<br />

A recommendation for primary prevention for relatives of pancreatic<br />

carcinoma patients which differs from that for the normal<br />

population cannot be given.<br />

Recommendation grade: D, evidence level 5, strong consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Comment<br />

In general, the recommendation for the normal population can<br />

also be applied to relatives of pancreatic carcinoma patients.<br />

There is currently no scientific evidence that supports a benefit<br />

of deviating procedures.<br />

Familial pancreatic carcinoma<br />

(familial pancreatic cancer kindred: FPC)<br />

An increased pancreatic carcinoma risk was observed in several<br />

hereditary syndromes. In these cases, pancreatic carcinoma<br />

is not considered a leading clinical/phenotypic form (see below).<br />

Today, familial pancreatic carcinoma (FPC) is differentiated<br />

from these syndromes. FPC is always assumed if at least<br />

two first degree relatives (independent of age) in one family<br />

have pancreatic carcinoma and the family does not fulfill the<br />

clinical or familial anamnestic criteria of other hereditary syndromes<br />

(see below). FPC-tumors cannot be histologically differentiated<br />

from sporadic tumors. A prospective study from<br />

Germany showed that between 1 – 3% of all pancreatic carcinoma<br />

patients fulfill the FPC-criteria. Thus, FPC has a similar<br />

frequency to other hereditary tumor diseases [83]. The mean<br />

age of onset of FPC is not significantly different from sporadic<br />

cases (€ 62 years). However, preliminary data seem to indicate<br />

that offspring of FPC-patients may develop pancreatic carcinoma<br />

up to 10 years earlier (anticipation) [84]. As for all hereditary<br />

diseases, relatives from FPC-families should be referred to<br />

genetic consultation. A specific gene defect for FPC was found<br />

for only a small subgroup (about 10%). This is why predictive<br />

gene diagnostics outside of clinical studies cannot be recommended<br />

at this time.<br />

Compared to the normal population, family members with at<br />

least two first degree relatives who have pancreatic carcinoma<br />

(independent of the patient’s age) have a highly increased risk<br />

of also developing pancreatic carcinoma.<br />

Evidence level 2b, consensus<br />

Comments<br />

The risk of a first degree relative to develop pancreatic carcinoma<br />

is 18 times as high if two family members have this disease.<br />

It can increase up to 57 times if three or more persons are<br />

affected. However, these numbers are based only on one prospective<br />

study [85]. Therefore, they must be interpreted with<br />

caution.<br />

Recommendation<br />

A recommendation for primary prevention for relatives of FPC-families<br />

which differs from that for the normal population cannot be<br />

given.<br />

Recommendation grade: C, evidence level 3, consensus<br />

Comments<br />

In general, the recommendations for the normal population<br />

can also be applied to relatives of pancreatic carcinoma patients.<br />

There is currently no scientific evidence that supports<br />

a benefit of deviating procedures. Recently, data on preliminary<br />

screening tests using imaging techniques for families at<br />

risk were published [86 – 88]. However, they have not been<br />

confirmed by independent studies. Therefore, based on the<br />

current data a general recommendation of screening tests<br />

such as endosonography, ERCP, or MRCP for high risk groups<br />

are not recommended outside of controlled studies because<br />

of the high risk of false positive results.<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Other diseases/syndromes which are associated with an<br />

increased pancreatic carcinoma risk<br />

Patients with Peutz-Jeghers-Syndrome have a 36 to 42% lifetime<br />

risk of developing pancreatic carcinoma.<br />

Evidence level 2a, strong consensus<br />

Comments<br />

Two studies prove the much higher risk of developing pancreatic<br />

carcinoma for patients with Peutz-Jeghers-Syndrome [89, 90].<br />

Patients with B-K mole-syndrome (including pancreatic-carcinoma-melanoma-syndrome)<br />

have an up to 17% lifetime risk<br />

of developing pancreatic carcinoma.<br />

Evidence level 2b, strong consensus<br />

Comments<br />

Several studies in FAMMM or families with pancreatic-carcinoma-melanoma-syndrome<br />

prove a much higher risk of pancreatic<br />

carcinoma [83, 91–93].<br />

Patients and relatives of patients with hereditary breast or<br />

ovarian cancer have a higher risk of contracting pancreatic<br />

carcinoma.<br />

Evidence level 2a, consensus<br />

Comments<br />

There are only few systematic studies on the pancreatic carcinoma<br />

risk in families with hereditary breast or ovarian cancer.<br />

These are usually based on families with known germ line mutations<br />

in the BRCA 1 and 2 genes. The pancreatic carcinoma<br />

risk seems to be 2 to 6 times as high in these families [94 – 96].<br />

Patients and relatives of patients with HNPCC may have a<br />

higher risk of developing pancreatic carcinoma.<br />

Evidence level 3, consensus<br />

Comment<br />

The data available are very sparse and point towards a possible<br />

risk increase [97, 98].<br />

Patients and their relatives with ataxia teleangiectasia do not<br />

have an increased pancreatic carcinoma risk.<br />

Evidence level 3, consensus<br />

Comment<br />

It is difficult to evaluate the data on pancreatic carcinoma risk<br />

of ataxia teleangiectasia patients, because of the few numbers<br />

of cases. A higher risk of pancreatic carcinoma does not seem<br />

to be evident [99].<br />

Patients with FAP and their relatives have an increased risk of<br />

pancreatic carcinoma.<br />

Evidence level 3, consensus<br />

Comments<br />

One study reports an increased relative risk of 4.5 for pancreatic<br />

carcinoma [100].<br />

Patients with cystic fibrosis probably have no increased risk<br />

for pancreatic carcinoma.<br />

Evidence level 3, consensus<br />

Comment<br />

There are no definite statements in the literature on this subject.<br />

Patients with Li-Fraumeni-syndrome and their relatives<br />

may also have an increased risk of developing pancreatic<br />

carcinoma.<br />

Evidence level 4, consensus<br />

Leitlinie 457<br />

Comment<br />

Data are sparse on this subject and further studies are necessary<br />

[97].<br />

Other diseases that may be associated with a higher risk of<br />

pancreatic carcinoma<br />

Patients with von Hippel’s disease and Fanconi’s anemia may have<br />

an increased risk of pancreatic carcinoma while patients with neurofibromatosis<br />

do not.<br />

Evidence level 4<br />

Comments<br />

One study reported an association of pancreatic carcinoma with<br />

von Hippel’s disease [97]. Furthermore, gene mutations in the<br />

FANCC and FANCG genes were determined in patients with pancreatic<br />

carcinoma. A loss of heterozygosity was found in resected<br />

tumors. These data suggest that patients with mutations in the<br />

FANCC gene as is the case in Fanconi’s anemia may have a predisposition<br />

for pancreatic carcinoma. The relative and life time risks<br />

are not known [101].<br />

Recommendation<br />

In general, the recommendation for the normal population to reduce<br />

the risk of pancreatic carcinoma also applies to relatives of<br />

the abovementioned pancreatic carcinoma patients with hereditary<br />

disease. There is currently no scientific evidence that supports<br />

a benefit of deviating procedures (see also FPC). This recommendation<br />

applies only to the pancreatic carcinoma risk and not to deviating<br />

recommendations on screening/monitoring of the corresponding<br />

hereditary disease.<br />

Recommendation grade: D, evidence level 4, consensus<br />

Patients with hereditary pancreatitis<br />

Patients with hereditary pancreatitis have a highly increased risk<br />

of pancreatic carcinoma.<br />

Evidence level 2b, strong consensus<br />

Comment<br />

The cumulative risk of developing pancreatic carcinoma by the<br />

age of 70 is between 40 and 44% in this group [102, 103].<br />

Recommendation<br />

Imaging techniques can currently not be recommended to monitor<br />

patients with hereditary pancreatitis.<br />

Recommendation grade: C, evidence level 3b, consensus<br />

Comments<br />

Prospective screening of families at risk was performed at two<br />

institutions (Johns Hopkins University and University of Seattle).<br />

Johns Hopkins recommends an annual screening and the Seattle-program<br />

a screening every two to three years. Lesions that<br />

are typical for chronic pancreatitis and unexpectedly frequent<br />

IPMNs were found using endosonography and ERCP. The value<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


458<br />

Leitlinie<br />

of these results is not known. Therefore, this type of monitoring<br />

should only be done under study conditions [86 – 88, 104].<br />

Chronic pancreatitis<br />

Patients with many years of chronic pancreatitis have an increased<br />

risk of developing pancreatic carcinoma.<br />

Evidence level 2b, strong consensus<br />

Comments<br />

The data on the relative risk are heterogeneous and vary from<br />

2.3 to 18.5. The cumulative incidence is reported to be 1.1% after<br />

5 years, 1.8% after 10 years, and 4% after 20 years [105 – 109].<br />

Type 2 diabetes mellitus<br />

Patients with type 2 diabetes mellitus have an increased risk for<br />

pancreatic carcinoma.<br />

Evidence level 2b, consensus<br />

Comments<br />

About 1% of diabetics age 50 years and younger develop pancreatic<br />

carcinoma in the following 3 years. That is equal to an<br />

about 8-fold higher risk [110]. Further studies show a risk that<br />

is lower [50, 111 – 118]. It must be taken into account that diabetes<br />

can be caused by pancreatic carcinoma [119, 120].<br />

Recommendation on genetic consultation<br />

The genetic consultation of patients with genetic syndromes and<br />

their relatives is performed according to the directives of the German<br />

Medical Association.<br />

Recommendation grade: A, consensus<br />

Topic 2:<br />

Diagnostics<br />

!<br />

Diagnostics in case of newly occurring symptoms<br />

Recommendation<br />

Newly occurring pain in the epigastric region and back should<br />

result in tests for diagnosing pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 3, strong consensus<br />

Comments<br />

In general, newly occurring pain in the epigastric region or<br />

back which could be caused by pancreatitis or pancreatic carcinoma<br />

mandate further diagnostics. No literature is available<br />

that states for which symptoms alone or in combination and<br />

from which age pancreatic carcinoma should be considered.<br />

The diagnostics alone or in combination that are necessary to<br />

exclude pancreatic tumors have also not been reported. Newly<br />

occurring back pain alone which cannot be explained by alterations<br />

of the musculoskeletal system should not result in<br />

testing for pancreatic carcinoma.<br />

l " Table 8 suggests an age and suspicion level adapted procedure<br />

which is based on expert opinion.<br />

Recommendation<br />

A newly occurring or manifest type 2 diabetes mellitus with no further<br />

pancreatic carcinoma symptoms should not lead to pancreatic<br />

carcinoma diagnostic testing.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Table 8 Diagnostic procedures adapted to age and suspicion level for newly<br />

occurring pain of the epigastric region and back<br />

suspicion<br />

level<br />

age<br />

(years)<br />

symptoms procedure<br />

low < 50 just pain1 sonography in case of<br />

persisting symptoms<br />

intermediate<br />

50 justpain1 sonography, if necessary CT<br />

pain plus2 sonography, if necessary CT<br />

high > 50 pain plus sonography, if necessary CT/<br />

endosonography<br />

1 Newly occurring pain that radiates localized/belt-like to the back and that<br />

is perceivable during the night should be clarified further regardless of age.<br />

If the suspicion level is high or if the ultrasound is negative, complementary<br />

CT, or endosonography should be implemented.<br />

2 Pain plus other symptoms (inappetence, weight loss, weakness).<br />

Comment<br />

According to current studies, there is only a moderate connection<br />

between type 2 diabetes and pancreatic carcinoma [110,<br />

112, 115]. This and the few possibilities for early pancreatic carcinoma<br />

diagnosis currently available, make diagnostic testing<br />

for pancreatic cancer not useful for type 2 diabetes mellitus patients<br />

without further symptoms.<br />

Recommendation<br />

Newly occurring painless jaundice should result in diagnostic testing<br />

for pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 2b, strong consensus<br />

Comment<br />

<strong>Pancreatic</strong> and bile duct carcinoma are the most frequent cause<br />

(20%) for newly occurring jaundice in patients over 60 years of<br />

age [121 –123].<br />

Recommendation<br />

In certain cases (patients > 50 years with primary “idiopathic”<br />

pancreatitis), acute pancreatitis of unknown etiology should result<br />

in additional procedures for the diagnosis of pancreatic carcinoma.<br />

Recommendation grade: C, evidence level 4, consensus<br />

Comments<br />

The pancreatic carcinoma incidence seems to be higher in patients<br />

with acute pancreatitis and even more so in patients<br />

with an acute episode of chronic pancreatitis. However, they<br />

do not exceed 1 – 2% of all pancreatic carcinoma cases or a maximum<br />

of 5% of idiopathic forms. There are only few systematic<br />

studies on this topic, especially on the idiopathic forms. Data<br />

using endosonography in this context are well documented.<br />

Thus, the primary recommendation is endoscopic ultrasound,<br />

which should be done during the symptom-free interval following<br />

acute pancreatitis. MD-CT examinations are an alternative.<br />

If the endoscopic ultrasound is negative, a clarification of the<br />

ducts should be considered. However, only data on ERCP are<br />

available, which has the risk of post-ERCP-pancreatitis. By analogy,<br />

MRCP is recommended [124, 125].<br />

In general, CA 19–9-assessment should not be done to diagnose<br />

pancreatic carcinoma. Algorithms or diagnostic procedures are<br />

also not recommended to clarify an increased CA 19–9-value.<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Although pancreatic carcinoma may also develop before the<br />

age of 50, the age dependent incidence curve does not show<br />

an increase until after the age of 50 [122]. Therefore, diagnostic<br />

testing should be done in patients age 50 years and older.<br />

Imaging techniques for primary diagnostics<br />

Recommendation<br />

Different procedures such as sonography, endosonography, multidetector-CT,<br />

MRT with MRCP or ERCP are suitable to confirm suspected<br />

tumors.<br />

Recommendation grade: B, evidence level 2, consensus<br />

Comments<br />

The abovementioned procedures are not always available.<br />

Therefore, they cannot be implemented in an algorithm defined<br />

for every situation. It was agreed that initially an ultrasound<br />

examination of the epigastric region should be done.<br />

This would allow a tentative diagnosis of pancreatic carcinoma<br />

and perhaps also diagnosis of metastases. The diagnosis<br />

of pancreatic carcinoma is also possible with all other procedures.<br />

However, ERCP alone is not sufficient to diagnose ductal<br />

pancreatic carcinoma, because it depicts only the ductal<br />

changes and not the lesion itself [126, 127].<br />

Computed tomography (multi-detector-CT) should be done<br />

using a biphasic contrast medium protocol (pancreas parenchyma<br />

phase and portal venous phase). The section thickness<br />

should be £ 3 mm. The MRT/MRCP should be done with a field<br />

strength of at least 1.5 tesla and standard weighting (T1 and T2<br />

including MRCP). The section thickness should be 5 – 7 mm.<br />

The endoscopic ultrasound may be done with a mechanical radial<br />

scanner as well as an electronic endosonography device.<br />

However, the published data are based on mechanical radial<br />

scanners. Although the electronic endosonography most likely<br />

has a better local resolution, it has not been investigated whether<br />

carcinomas can be identified with a higher sensitivity with<br />

this device.<br />

Recommendation<br />

Sonography of the epigastric region, endosonography, multi-detector-CT,<br />

as well as MRT in combination with MRCP are first choice<br />

diagnostic procedures to detect pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comments<br />

According to the literature multi-detector-CT and MRT in combination<br />

with MRCP are the most sensitive procedures for the<br />

detection of pancreatic carcinoma. If used by specialists, endosonography<br />

may partly be even more sensitive. A clear recommendation<br />

for or against one of these procedures cannot be given.<br />

The procedure for which there is the most expertise in the<br />

facility should be used. Different complementary procedures<br />

may also have to be implemented.<br />

Cytologic and laboratory diagnostics<br />

Recommendation<br />

If pancreatic carcinoma is suspected, brush cytology from the bile<br />

ducts is not sensitive enough. It is also not recommended to use<br />

brush cytology from the pancreas duct to detect pancreatic carcinoma.<br />

Therefore, an ERCP for tissue diagnostics is not indicated.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Leitlinie 459<br />

Comments<br />

The literature does not allow for an adequate statement on the<br />

abovementioned question. Brush techniques in pancreas as well<br />

as in the bile duct system are risky and not useful.<br />

Laboratory tests<br />

Recommendation<br />

If there is a pancreatic lesion, CA 19–9-testing should be done.<br />

Recommendation grade: C, evidence level 2a, majority approval<br />

Comments<br />

CA 19–9 laboratory tests may be used in individual cases for<br />

differential diagnostics. Potential resectability detected with<br />

imaging techniques and a very high preoperative CA 19–9-value<br />

may make staging laparoscopy necessary. In such cases the tumor<br />

load is often greater than expected from the imaging technique<br />

e. g. a disseminated tumor. However, so far there are no<br />

cost-benefit analyses which support the use of this test [128–<br />

131].<br />

Tissue diagnostics: procedure in case of pancreatic lesion<br />

Recommendations<br />

Initially a resection should be done if there is a potentially resectable<br />

lesion in the pancreas that may be a tumor. An endosonographic<br />

guided biopsy may be done if differential diagnostic<br />

indications are present that would change the procedure e. g. if<br />

metastases are suspected because of an earlier malignancy.<br />

Recommendation grade: B, evidence level 2, consensus<br />

Comments<br />

The endosonographically guided fine needle puncture is sensitive<br />

and highly specific for histologic or cytologic diagnosis of<br />

pancreatic carcinoma [132–134]. However, in most cases it is<br />

unnecessary, because if resectable, lesions of unclear dignity<br />

should also be operated on. Therefore, for a potentially resectable<br />

lesion, a preoperative biopsy to confirm the diagnosis of<br />

pancreatic cancer is not necessary.<br />

Recommendation<br />

If nonetheless a biopsy is done for differential diagnostics, biopsies<br />

should be preferably taken from those lesions that have the<br />

lowest risk of complications.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Recommendation<br />

A biopsy to confirm a diagnosis is mandatory before specific palliative<br />

therapy is initiated independently of whether the pancreatic<br />

tumor is locally advanced, inoperable, or metastatic.<br />

Recommendation grade: B, evidence level 2a, strong consensus<br />

Comment<br />

The diagnosis must be cytologically or histologically confirmed<br />

before palliative chemotherapy is started to exclude false treatment<br />

due to other differential diagnoses [135].<br />

Recommendation<br />

The lesion that is easiest to reach and that has the lowest risk is<br />

biopsied independently of whether it is the primary tumor or a<br />

metastasis.<br />

Recommendation grade: D, evidence level 5, strong consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


460<br />

Leitlinie<br />

Preoperative staging<br />

Recommendation<br />

Preoperatively, multi-detector-CT and endosonography should be<br />

preferred to evaluate local tumor spread or resectability.<br />

Recommendation grade: A, evidence level 1a, consensus<br />

Comments<br />

The standard procedure to evaluate the size of primary tumors<br />

and the local tumor spread is a CT. A combination of transabdominal<br />

and endoscopic ultrasound can give similar diagnostic<br />

results. However, the image documentation of sonographic procedures<br />

is often insufficient from a surgeon’s point of view. The<br />

sonographic and endosonographic procedures are dependent on<br />

who performs them. If sufficient multi-detector-CTs and MRIs<br />

are available preoperatively, no other procedures are necessary.<br />

Because of the high costs, an MRI is not always done for primary<br />

evaluation of local tumor spreading [136].<br />

Recommendation<br />

Abdominal sonography is mandatory to assess systemic tumor<br />

spreading. If no systemic metastases were detected using abdominal<br />

sonography or if a study protocol requests imaging according<br />

to RECIST-criteria (Response-Evaluation-Criteria in Solid Tumors),<br />

then abdominal multi-detector-CT is mandatory.<br />

X-ray thorax examinations are part of the tumor staging.<br />

MRT as well as thoracic-CT are assessed as optional for the evaluation<br />

of systemic tumor spread. Endosonography, ERCP, MRCP,<br />

and bone scans are not used for tumor staging.<br />

Currently, FDG-PET examinations as well as micrometastasis diagnostics<br />

from whole blood do not play a role in preoperative diagnostics.<br />

Recommendation grade: D, evidence level 2b, consensus<br />

Comments<br />

A comparison of the literature on preoperative staging of pancreatic<br />

carcinoma demonstrated a slightly higher sensitivity of<br />

endoscopic ultrasound versus computed tomography. However,<br />

when the studies were compared they revealed a high heterogeneity<br />

with regard to design, quality, results, as well as methodologic<br />

weaknesses [137]. Literature on MRT showed a high<br />

sensitivity and specificity for the detection of biliary obstruction,<br />

but not a high sensitivity for the differentiation between<br />

benign or malignant causes of the obstruction [138]. However,<br />

this study only took publications into account that were published<br />

until the end of 2003. PET-studies are too complex and<br />

expensive even though their potency was shown to be similar<br />

to other methods. The significance of the new PET-CT-procedure<br />

must be evaluated in future studies [139].<br />

Recommendation<br />

Staging-laparoscopy is optional.<br />

Recommendation grade: D, evidence level 2b, consensus<br />

Comments<br />

Staging-laparoscopy is of particular interest if e. g. due to excessively<br />

high CA 19–9-values or ascites there is suspicion of peritoneal<br />

carcinosis which is not visible by imaging techniques.<br />

Findings in up to one third of those patients exclude curative<br />

resection [140, 141].<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Cystic processes<br />

Recommendation<br />

To differentiate cystic processes epigastric ultrasound, multi-detector-CT,<br />

MRT with MRCP, endosonography, and ERCP are principally<br />

implemented.<br />

Recommendation grade: D, evidence level 3, consensus<br />

Comments<br />

None of the methods that are mentioned allows a definite diagnosis.<br />

At most a robust conclusion can be made if they are combined<br />

with the below mentioned procedures and in connection<br />

with the individual clinical constellations.<br />

Recommendations<br />

An endosonographically guided biopsy should be performed if a<br />

cyst is considered to be suspicious.<br />

Recommendation grade: B, evidence level 3, consensus<br />

The tumor markers CA19–9 and CEA as well as cytology can be<br />

determined in aspirates.<br />

Recommendation grade: D, evidence level 2a, consensus<br />

Comments<br />

The abovementioned procedures are based on one original article.<br />

Further prospective multi-center trials are necessary to validate<br />

the recommendation [142].<br />

Therapy evaluation in the palliative situation<br />

Recommendation<br />

An epigastric ultrasound should always be done to examine the<br />

course of a tumor response during palliative chemotherapy. CT<br />

should only be done if the study conditions demand it (RECISTcriteria)<br />

or if the epigastric ultrasound does not give information<br />

on the course of the response.<br />

Recommendation grade: D, consensus<br />

Topic 3:<br />

Surgical therapy of pancreatic carcinoma<br />

(curative intention)<br />

!<br />

Introduction<br />

The following describes the surgical procedure for pancreatic<br />

tumors. In particular criteria are defined that make resectability<br />

of tumors with curative intent possible. Aside from the perioperative<br />

patient management, the topics that are dealt with<br />

are surgical techniques, surgical tactics for different tumor entities,<br />

disease stages, as well as preparation of surgical specimens<br />

that are relevant for prognosis. The data from the literature<br />

can be applied to the conditions in Germany. They are the<br />

basis for the evidence and recommendation grades of the individual<br />

topics.<br />

Criteria for irresectability from the patient’s perspective<br />

Recommendation<br />

Age should not be a criterion to exclude a patient from pancreatic<br />

carcinoma resection.<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Comorbidity may be a criterion against resection.<br />

Recommendation grade: C, evidence level 4, strong consensus<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Comment<br />

Due to the demographic development the population will become<br />

older in the next decades. Therefore, the number of elderly<br />

patients (> 75 years) with pancreatic carcinoma will rise.<br />

Already today, different reports show that chronologic age is<br />

not a contraindication for pancreatic surgery, because the operative<br />

results for younger patient groups are comparable<br />

[143 –145]. In contrast to chronologic age, major visceral surgery<br />

may be contraindicated if comorbidities are present. It is<br />

not possible to specify individual clinical scenarios based on<br />

the literature. However, it is known that up to 30% of all perioperative<br />

complications and up to 50% of all postoperative<br />

deaths have cardiac causes. In addition, patients with chronic<br />

respiratory diseases or liver cirrhosis have an increased risk of<br />

developing perioperative complications. The decision whether<br />

pancreatic surgery is beneficial and safe for these risk patients<br />

must be carefully considered. Furthermore, an interdisciplinary<br />

evaluation of the complete constellation must be performed<br />

before surgery. The treatment alternatives and the patient’s<br />

wishes should also be taken into consideration [146].<br />

Criteria for irresectability due to the tumor<br />

Extrapancreatic tumor manifestation<br />

Recommendation<br />

<strong>Pancreatic</strong> carcinoma in healthy patients may be resectable even<br />

if it has infiltrated neighboring organs.<br />

Recommendation grade: C, evidence level 3, consensus<br />

Infiltration of Haller’s tripod and the superior mesenteric<br />

artery<br />

Recommendation<br />

If Haller’s tripod and the superior mesenteric artery are infiltrated<br />

in healthy patients, a resection is rarely possible.<br />

Recommendation grade: C, evidence level 4, strong consensus<br />

Infiltration of the portal vein and the superior mesenteric<br />

vein<br />

Recommendation<br />

The infiltration of the portal vein should not be a contraindication<br />

for resection. If the superior mesenteric vein is infiltrated, the possiblity<br />

of resection is less likely than if the portal vein is infiltrated.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comments<br />

The aim of pancreatic surgery is an R 0 resection independently<br />

of the location of the tumor [147]. Locally advanced carcinomas<br />

should be resected if necessary en bloc with the neighboring<br />

organs, because the prognosis for extended R 0 resection is<br />

identical to standard resections [148 –152].<br />

Resection of the portal vein and the superior mesenteric vein<br />

should be performed during pancreatic surgery if it makes an<br />

R0 resection possible. A systematic review shows that in a high<br />

percentage of these patients the lymph nodes are already infiltrated<br />

[153]. Many series from pancreas centers demonstrate<br />

that the morbidity and the mortality of an en bloc resection of<br />

pancreas and portal vein (or superior mesenteric vein) and the<br />

patient’s long-term prognosis are comparable to that of patients<br />

without portal vein resection if their disease stages correspond<br />

[147, 149, 151, 154 –156].<br />

The infiltration of Haller’s tripod and the superior mesenteric<br />

artery rarely allows resections in healthy patients. A recommendation<br />

on the extent of the vessel infiltration is deliberate-<br />

Leitlinie 461<br />

ly not made. The resection of tumor infiltrated arteries during<br />

pancreas resection is technically possible, but should currently<br />

be viewed as experimental [150, 152, 157, 158].<br />

Preoperative cholestasis<br />

Recommendation<br />

Preoperative drainage of bile using stents should not be generally<br />

done. It should be performed only if cholangitis is present or if<br />

an operation cannot take place immediately.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comments<br />

In 73% of the cases an infection is seen after ERCP and stent<br />

placement [159]. Infections of the biliary system at the time<br />

of pancreas resection are associated with an increased morbidity.<br />

Therefore, a stent should not be placed if an operation<br />

can be done quickly [160 – 163]. If cholangitis is present or the<br />

operation cannot be done right away e.g. due to neoadjuvant<br />

therapy, a stent should be placed.<br />

Perioperative antibiotic prophylaxis<br />

Recommendation<br />

A perioperative antibiotic prophylaxis should always be done.<br />

Recommendation grade: A, evidence level 1c, strong consensus<br />

There should be no difference in the prophylaxis of patients with<br />

and without stents.<br />

Recommendation grade: C, consensus<br />

Comments<br />

Antibiotic prophylaxis should always be done during hepatobiliary<br />

surgery, because it significantly reduces postoperative infections<br />

including wound infections [164]. It should not be maintained<br />

postoperatively, because there is no advantage. However,<br />

there is a higher risk of complications including development of<br />

resistances and allergies [165]. If cholangitis is present, an antibiotic<br />

therapy should be done.<br />

Perioperative somatostatin prophylaxis<br />

Recommendation<br />

A reduction of pancreas-specific complications by perioperative<br />

somatostatin prophylaxis is supported by a meta-analysis of randomized<br />

studies. However, the prophylaxis with somatostatin cannot<br />

be recommended for all patients, because existing studies define<br />

pancreas fistula differently. This could lead to differences in<br />

the classification of postoperative complications.<br />

Recommendation grade: C, evidence level 1a, consensus<br />

Comments<br />

The studies are heterogeneous with respect to the benefit of<br />

somatostatin prophylaxis. None of the studies showed a difference<br />

in postoperative mortality. However, a meta-analysis<br />

of 1918 patients and 10 randomized studies shows that somatostatin<br />

significantly reduces the morbidity and pancreas-specific<br />

complications including the frequency of biochemically<br />

detectable fistula [166]. Further differentiation of the resection<br />

methods in subgroup analyses shows no benefit of somatostatin<br />

prophylaxis. Therefore, the patient group that benefits<br />

from a perioperative inhibition of secretion using somatostatin<br />

can currently not be identified.<br />

At present there is no uniform definition of a pancreatic fistula.<br />

This complicates the evaluation of the efficacy of somatostatin<br />

prophylaxis and may lead to differences in the classification of<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


462<br />

Leitlinie<br />

postoperative complications. Therefore, the comparison of published<br />

data is difficult. A new classification of pancreatic fistulas<br />

was developed at a consensus conference in 2005 [167]. This is<br />

an attempt to make an internationally accepted and comparable<br />

definition of fistulas in future publication possible. Therefore, a<br />

general recommendation on the perioperative application of somatostatin<br />

cannot be given.<br />

Objectives for intraoperative resection margins<br />

Recommendation<br />

There are no definite data on necessary safety margins when resecting<br />

pancreatic carcinoma. Macroscopically assessed resection<br />

margins for curative resection (R0) can be deduced from practical<br />

experience. They should be as follows:<br />

E in pancreas tissue: 10 mm,<br />

E for bile ducts: 10 mm,<br />

E at the stomach/pylorus: 10 mm.<br />

For anatomic reasons, no objectives can be given for R 0 resections<br />

in the area of the retroperitoneum.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Intraoperative sonography of the liver<br />

Recommendation<br />

An intraoperative sonography is not necessary if the preoperative<br />

abdominal CT is inconspicuous.<br />

Recommendation grade: B, evidence level 3, consensus<br />

If the results of abdominal sonography or CT are not clear, a clarification<br />

should be attempted prior to surgery. In unclear cases an<br />

intraoperative sonography can lead to further clarification.<br />

Recommendation grade: D, evidence level 3, consensus<br />

Comments<br />

The quality of the new generation of multi-detector-CT is so<br />

high that if metastases were not detected using preoperative<br />

CT diagnostics, an intraoperative sonography would give no<br />

further information. Therefore, if a preoperative CT is inconspicuous,<br />

an intraoperative sonography is not necessary [168].<br />

If the preoperative clarification of changes in the liver is still<br />

unclear, an intraoperative sonography may be useful to assess<br />

the operability of pancreatic tumors. If the resectability of the<br />

pancreatic tumor was assessed laparoscopically, an intraoperative<br />

sonography can replace the palpation and add to the mere<br />

inspection of the liver surface [169].<br />

Intraoperative peritoneal lavage/cytology<br />

Recommendation<br />

Intraoperative peritoneal lavages have no therapeutic consequences.<br />

Therefore, there is no indication for a peritoneal lavage with<br />

cytology.<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Comments<br />

Most patients have a relapse after curative resection of pancreatic<br />

carcinoma. It is speculated that this is due to micrometastases<br />

in the peritoneum which are not detectable at the<br />

time of surgery [170]. Even though peritoneal micrometastases<br />

could be detected in peritoneal lavages, a prognostic relevance<br />

of positive cytologic results was not found. Therefore,<br />

micrometastases are not an exclusion criterion for radical resection,<br />

and a peritoneal lavage is not necessary [171, 172].<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Extent of resection/surgical technique/preferred<br />

anastomosis technique<br />

Recommendation<br />

The aim of pancreatic surgery is an R0 resection independently of<br />

the location of the tumor.<br />

Pancreas head carcinoma<br />

Recommendation<br />

The resection of the pancreas head tumors usually includes the<br />

partial duodenopancreatectomy with or without pylorus preservation.<br />

In rare cases a total pancreatectomy may be necessary if<br />

the carcinoma has spread. If neighboring organs and other structures<br />

are infiltrated, the resection should be extended.<br />

Recommendation grade: A, evidence level 1c, consensus<br />

Classic Whipple versus pp-Whipple<br />

Recommendation<br />

Both procedures (pylorus preserving [pp] versus stomach resecting<br />

partial duodenopancreatectomy [classic]) are comparable with respect<br />

to postoperative complications and oncologic long-term results<br />

(1a).<br />

Recommendation grade: A, evidence level 1a, consensus<br />

Comment<br />

A meta-analysis of the literature on pylorus preserving and classic<br />

duodenopancreatectomy demonstrated no relevant differences<br />

between both procedures with respect to patient mortality,<br />

morbidity, and survival. However, the studies that were analyzed<br />

were very heterogeneous [173].<br />

Carcinoma of the pancreatic tail<br />

Recommendation<br />

The surgical procedure for pancreas tail carcinomas is left-sided<br />

pancreatic resection. The abovementioned criteria on the surgical<br />

expansion apply [174 –176].<br />

Recommendation grade: A, evidence level 3, consensus<br />

<strong>Pancreatic</strong> body cancer<br />

Recommendation<br />

In general a subtotal left-sided pancreatic resection or if necessary<br />

a total duodenopancreatectomy is indispensable for pancreas body<br />

cancer [174–176].<br />

Recommendation grade: B, evidence level 3, consensus<br />

Tumor adherence to surrounding organs<br />

Recommendation<br />

An R0 resection can be performed even if the neighboring organs<br />

are infiltrated (extrahepatic local manifestations). The goal of every<br />

resection must be an R0 resection [148–152].<br />

Recommendation grade: C, evidence level 3, consensus<br />

Radical extension of lymphadenectomy<br />

Recommendation<br />

So far, a benefit of extended lymph node resections was not observed.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

A standardized lymphadenectomy during pancreas head cancer<br />

surgery according to Whipple should be performed as follows:<br />

complete and circular dissection of the lymph nodes of the hepatoduodenal<br />

ligament as well as around the common hepatic<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


artery and the portal vein and the cranial part of the superior<br />

mesenteric artery. Furthermore, lymph nodes at the right side<br />

of the celiac trunk and the lymph nodes at the right hemicircumference<br />

of the superior mesenteric artery stem should be<br />

dissected.<br />

The term “radical extension of lymphadenectomy” includes different<br />

degrees of dissection. The number of removed lymph<br />

nodes and the additional time needed for an extended lymphadenectomy<br />

differ significantly between the 4 randomized studies<br />

on this topic [177–179]. Consistently all 4 studies from<br />

Europe, Japan, and the USA showed no additional benefit with<br />

respect to long-term survival after extended radical dissection<br />

of the lymph nodes compared to standard dissection (definition<br />

see above). Thus, so far no benefit was shown for the extended<br />

resection.<br />

Operative strategy for IPMT/cystic pancreatic tumors<br />

Recommendation<br />

All potentially malignant tumor entities of the pancreas are primarily<br />

resected with the same goal and to the same extent as the<br />

ductal pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comments<br />

Cystic pancreatic tumors have a wide spectrum of histologic<br />

characteristics including inflammatory (pseudocysts) and benign<br />

(serous) cysts as well as cysts of unknown or malignant<br />

origin (IPMT, mucinous) [180]. A definite differentiation between<br />

benign and malignant is not possible with modern CT or<br />

MRT examinations. Endosonographic and cytologic differentiation<br />

of cystic contents are not as good as differentiation using<br />

CEA values of cystic contents and, thus, not definite [181, 182].<br />

If the operative risk is justifiable, resection is recommended,<br />

because it is not possible to differentiate benign and malignant<br />

cystic lesions preoperatively [183–185]. However, for high risk<br />

patients and patients with small serous tumors without symptoms<br />

a resection can be sustained. In this case, a close monitoring<br />

of the pancreas is useful [184]. A limited resection (segmental<br />

resection, enucleation) for serous cystic adenoma and the<br />

early stages of IPMT of the collateral duct are sufficient [180,<br />

185]. All potentially malignant tumor entities should primarily<br />

be operated with the same goal and to the same extent of resection<br />

as the ductual pancreatic carcinoma, because the prognosis<br />

of advanced IPMT and mucinous cystic adenocarcinoma is identical<br />

to ductual adenocarcinoma.<br />

Therapeutic procedures for distant metastases<br />

Recommendation<br />

Resection of distant metastases of ductal pancreatic cancer (organ<br />

metastases, peritoneal carcinosis, lymph node metastases that are<br />

defined as distant metastases) does not improve the prognosis and,<br />

thus, should usually be avoided.<br />

Recommendation grade: A, evidence level 3, consensus<br />

Resection of intraoperatively detectable distant metastases<br />

Recommendation<br />

Even if the tumor is resectable, a resection should not be done if<br />

distant metastases are only detectable intraoperatively.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Leitlinie 463<br />

Comments<br />

In high volume centers, the morbidity of pancreatic surgery is<br />

markedly reduced and the mortality is under 5%. If metastases<br />

have been detected and the complication rate is acceptable,<br />

pancreas resection can lead to excellent pain control. In<br />

several publications it was already considered to be an alternative<br />

to palliative double bypass surgery. However, despite<br />

the progress in adjuvant therapy, so far no survival advantage<br />

has been shown for resection of metastatic pancreatic cancer<br />

[186–188].<br />

Laparoscopic surgical techniques<br />

Recommendation<br />

<strong>Pancreatic</strong> cancer is an indication for laparoscopic tumor staging.<br />

However, laparoscopic surgery of pancreatic carcinoma is currently<br />

not recommended.<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Comments<br />

In about 20% of the cases a curative resection is not possible<br />

despite standardized preoperative staging of pancreatic cancer<br />

using modern imaging techniques. This is due to local tumor<br />

spreading or peritoneal carcinosis or liver metastasis that is<br />

only detectable intraoperatively [189]. In this case patients<br />

can benefit from diagnostic laparoscopy, as a minimal invasive<br />

technique. It can complete the staging (see also Topic 2)<br />

[189, 190]. A diagnostic laparoscopy is especially useful for<br />

the staging of cancer in the pancreatic body and tail, because<br />

of the late development of symptoms compared to pancreatic<br />

head carcinomas. In contrast, laparoscopically performed oncologic<br />

pancreatic resections, although possible, are experimental<br />

[191].<br />

Intraoperative frozen section examination<br />

Recommendation<br />

Pancreas and if applicable bile duct resection margins should be<br />

tested by frozen section analysis independently of frozen section<br />

testing for the diagnosis of distant metastases [192]. In addition,<br />

putative liver metastases and possibly peritoneal carcinosis should<br />

be assessed. Lymph nodes and the retroperitoneal margin do not<br />

have to be evaluated.<br />

Recommendation grade: A, evidence level 1c, consensus<br />

Intraoperative needle or incision biopsies of the primary pancreatic<br />

tumor should not be done to clarify its dignity.<br />

Recommendation grade: B, evidence level 4, consensus<br />

Histologic evaluation of resected tumors<br />

Recommendation<br />

Criteria for the R0 resection are tumor-free margins at the hepatic<br />

duct, the pancreas resection area, as well as the circumferential<br />

inclusive retroperitoneal margin. An additional prerequisite is<br />

that no distant metastases (organs, peritoneum, lymph nodes) remain<br />

[193, 194].<br />

Recommendation grade: B, evidence level 2, consensus<br />

The specimen should be prepared using ink to mark the retroperitoneal<br />

margin [193, 194].<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Necessary information from the pathologist:<br />

a) pT-classification<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

b) pN-classification<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


464<br />

Leitlinie<br />

c) Number of lymph nodes examined<br />

Recommendation grade: C, evidence level 4, consensus<br />

d) Lymph node micrometastases<br />

Recommendation grade: B, evidence level 3, consensus<br />

e) R-classification<br />

Recommendation grade: B, evidence level 2a, strong consensus<br />

f) Status at the resection area of the remaining pancreas<br />

Recommendation grade: B, evidence level 2a, strong consensus<br />

g) Status at the retropancreatic resection area<br />

Recommendation grade: B, evidence level 2a, strong consensus<br />

h) Lymph vessel invasion<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

i) Blood vessel invasion<br />

Recommendation grade: B, evidence level 3; strong consensus<br />

j) Nerve sheath invasion<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Comments<br />

A recommendation by the UICC is only available on the least<br />

number of lymph nodes that should be removed. To determine<br />

pN0 at least 10 lymph nodes should be examined. This number<br />

is not supported by data in the literature. The number of examined<br />

lymph nodes should be documented in the histopathologic<br />

report.<br />

The UICC-definition (0.2 – 2 mm) should be applied for the identification<br />

of lymph node micrometastases. This does not include<br />

so-called disseminated tumor cells [195].<br />

Recommendation<br />

The histologic results are most important for the frozen section<br />

analysis. Histology and perhaps immunohistology are necessary<br />

for paraffin embedded tissue [193, 194].<br />

Recommendation grade: A, evidence level 1c, consensus<br />

Topic 4:<br />

Adjuvant and neoadjuvant non-surgical therapy of<br />

pancreatic carcinoma<br />

!<br />

The only curative therapy option for pancreatic carcinoma is surgical<br />

therapy. However, the long-term survival after resection is<br />

still under 20%. Local relapse as well as distant metastases play a<br />

role in recurrence.<br />

The curative intended pancreatic resection is a prerequisite for<br />

adjuvant therapy (R0/R1). Following surgery, the tumor should<br />

be histopathologically staged. It is especially important to pay<br />

attention to the resection margins, particularly the retroperitoneal<br />

margins. This should be documented (see also Topic 3 surgical<br />

therapy).<br />

Recommendation<br />

Following R0 resection of pancreatic carcinoma, adjuvant chemotherapy<br />

is indicated for UICC-stage I-III.<br />

Recommendation grade: A, evidence level 1b, consensus<br />

Comments<br />

Adjuvant chemotherapy should be done after R0 resection (primary<br />

tumor-M0). An advantage in the disease-free survival was<br />

shown in two randomized phase-III studies and a meta-analysis<br />

[196 –198]. However, this may not mean an improved overall<br />

survival [196]. Frequently, the patient’s quality of life rapidly<br />

worsens if pancreatic carcinoma recurs. Therefore, to delay re-<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

lapse an adjuvant therapy is recommended for all patients after<br />

R0 resection if no distant metastases are present. If possible,<br />

patients should be included in clinical trials to assess the optimal<br />

duration and type of adjuvant therapy. If a patient is treated<br />

outside of clinical studies, the patient’s disease course should<br />

be documented with respect to relapse, survival, and side effects<br />

as part of the tumor documentation/quality assurance.<br />

Recommendation<br />

Adjuvant/additive chemotherapy can be done after R 0 resection of<br />

metastases.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Comments<br />

Currently, the importance of additive chemotherapy after R 0<br />

resection of metastases cannot be conclusively evaluated. Similar<br />

to the role of metastasis resection, the role of additive<br />

chemotherapy also cannot be evaluated with the data available.<br />

As already shown in Chapter 3, outside of clinical trials<br />

pancreatic cancer should generally not be resected if distant<br />

metastases are detectable even if it were possible.<br />

Recommendation<br />

Tumor associated risk factors (e.g. G 3-, T 4-stage) are not important<br />

for the decision to initiate adjuvant chemotherapy.<br />

Recommendation grade: A, evidence level 1b, consensus<br />

Comments<br />

Currently, no differentiated procedures can be derived from the<br />

studies available for the adjuvant situation based on pancreatic<br />

carcinoma grading and staging [196, 197]. General contraindications<br />

for adjuvant therapy are summarized in l " Table 9.<br />

Table 9 Contraindications of adjuvant chemotherapy for pancreatic carcinoma<br />

general condition of health poorer than ECOG 2<br />

uncontrolled infections<br />

liver cirrhosis Child B and C<br />

severe coronary heart disease; heart failure (NYHA III and IV)<br />

preterminal and terminal kidney failure<br />

restricted bone marrow function<br />

inability to have regular follow-up examinations<br />

Recommendation<br />

There is no general restriction with respect to age for the implementation<br />

of adjuvant chemotherapy.<br />

Recommendation grade: A, evidence level 1b, consensus<br />

Comments<br />

Patients that were over 80 years of age were also included in<br />

the published trials. An unfavorable side effect profile was not<br />

observed for this age group [196, 197].<br />

Recommendation<br />

Adjuvant chemotherapy should be done for ECOG-status 0 – 2.<br />

Recommendation grade: A, evidence level 1b, consensus<br />

Comments<br />

This recommendation follows the general practical experience.<br />

Patients with a Karnofsky-performance-status of ‡ 50% (corresponding<br />

to maximum ECOG 3) were also included in the CON-<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


KO1-study [196]. The study by Neoptolemos et al. does not mention<br />

the performance status of the patients that were included.<br />

Therefore, the indication for adjuvant chemotherapy can be assessed<br />

individually for ECOG-stage 3.<br />

Recommendation<br />

The following adjuvant chemotherapy protocols can be implemented:<br />

a) 5-FU/folinic acid (Mayo-protocol)<br />

Recommendation grade: none, evidence level 2b, consensus<br />

Comments<br />

The study by Neoptolemos et al. was evaluated by the group<br />

as an RCT of low quality with 2b because of its methodologic<br />

weakness (method of randomization, quality control of surgical<br />

and radiation therapy) [198].<br />

b) 5-FU/folinic acid (AIO-scheme)<br />

Recommendation grade: D, evidence level 5, consensus<br />

Comment<br />

Although there are no studies available on infusional 5-FU-protocols<br />

for adjuvant pancreatic carcinoma therapy, if 5-FU is chosen,<br />

all 5-FU-protocols were considered possible.<br />

c) Gemcitabine<br />

Recommendation grade: A, evidence level 1b, consensus<br />

Comment<br />

The largest controlled, randomized study is presently available<br />

for gemcitabine [196].<br />

d) Loco-regional chemotherapy<br />

Recommendation grade: none, evidence level 5, consensus<br />

Comment<br />

A recommendation cannot be given, because sufficient data are<br />

not available.<br />

Recommendation<br />

Adjuvant chemotherapy should last 6 months.<br />

Recommendation grade: A, evidence level 3, consensus<br />

Comment<br />

This recommendation is based on the treatment duration that<br />

was selected in the randomized studies [196, 198]. For pancreatic<br />

carcinoma there are no studies that have compared the effect<br />

of long-term versus short-term adjuvant therapy.<br />

Recommendation<br />

If possible, adjuvant chemotherapy should be initiated within<br />

6 weeks after surgery.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comment<br />

There are no systematic investigations on the best time to begin<br />

adjuvant chemotherapy after pancreatic resection. The recommendation<br />

is based on the procedure used in the available<br />

studies (therapy start 10 – 42 days after surgery [196] or start<br />

of therapy median 46 days after surgery [198]).<br />

Leitlinie 465<br />

Recommendation<br />

An additive chemotherapy should be performed for R1 resected<br />

pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 3, majority consensus<br />

Comment<br />

This recommendation is based on a subgroup analysis of the<br />

CONKO1-study in which 61 patients with R1 resection were<br />

treated either with placebo (n = 27) or gemcitabine (n = 34). Patients<br />

treated with gemcitabine demonstrated a significantly<br />

better disease-free survival and a better overall survival than<br />

the placebo treated group [196].<br />

Recommendation<br />

The additive therapy with gemcitabine should be performed for 6<br />

months.<br />

Recommendation grade: A, evidence level 3, majority consensus<br />

Comment<br />

A similar therapy duration as for the adjuvant situation applies.<br />

Following an R0 resection for pancreatic carcinoma, there is<br />

no indication for an adjuvant radiochemotherapy outside of<br />

clinical trials.<br />

Recommendation grade: A, evidence level 1b, consensus<br />

Comment<br />

A current meta-analysis [197] as well as the ESPAC-1-study<br />

[198] show no benefit of adjuvant radiochemotherapy for pancreatic<br />

carcinoma.<br />

Recommendation<br />

Additive radiochemotherapy for pancreatic carcinoma cannot be<br />

generally recommended after R1 resection. For individual cases<br />

it may be an option that should be individually discussed. Ideally<br />

these patients should be treated as part of randomized, controlled<br />

study protocols.<br />

Recommendation grade: D, evidence level 3, consensus<br />

Comment<br />

Currently, only few data are available on the efficacy of radiochemotherapy<br />

following R1 resection. There is no direct comparison<br />

between the efficacy of chemotherapy and radiochemotherapy<br />

[199]. The distinct side effects of radiochemotherapy<br />

compared to chemotherapy have to be considered. A continuous<br />

infusion of 5-FU is recommended after R1 resection if additive<br />

radiochemotherapy is done outside of trials followed by treatment<br />

with gemcitabine as monotherapy. Preferably the treatment<br />

should be done as part of a clinical trial.<br />

Recommendation<br />

Surgery is the only curative therapeutic procedure for pancreatic<br />

cancer. Chemotherapy, radiochemotherapy, or radiation therapy<br />

without surgery are not indicated if the intention is curative.<br />

Recommendation grade: A, evidence level 4, consensus<br />

Recommendation<br />

Neoadjuvant radiation therapy, radiochemotherapy, or chemotherapy<br />

are presently not indicated for pancreatic carcinoma outside of<br />

clinical trials. This applies to patients with locally advanced, inoperable<br />

tumors as well as for patients with T4-tumors.<br />

Recommendation grade: D, evidence level 3, consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


466<br />

Leitlinie<br />

Comment<br />

Currently, the available data are not sufficient for this kind of<br />

treatment. It should only be done as part of a clinical trial.<br />

Recommendation<br />

There is no indication for intraoperative radiation therapy for pancreatic<br />

carcinoma.<br />

Recommendation grade: D, evidence level 4, strong consensus<br />

Recommendation<br />

Targeted therapies and immunotherapeutic approaches do not<br />

play a role in adjuvant or neoadjuvant therapies for pancreatic<br />

carcinoma.<br />

Recommendation grade: A, evidence level 5, strong consensus<br />

Comment<br />

Currently, a recommendation cannot be given, because the published<br />

data are insufficient.<br />

All the data from the literature on this topic are applicable to<br />

Germany.<br />

Topic 5:<br />

Palliative therapy<br />

!<br />

Introduction<br />

Since the studies of Mallinson [200], Palmer [201], and Glimelius<br />

[202], chemotherapy in the palliative situation has been<br />

shown to be better than the best supportive therapy with respect<br />

to survival and quality of life. This is confirmed by a recent<br />

Cochrane-analysis [203]. Gemcitabine was established as<br />

standard in palliative chemotherapy for pancreatic carcinoma<br />

by the Burris trial [204]. The role of new combination therapies<br />

was investigated in several phase-III studies, and the role<br />

of molecular therapies (“targeted therapies”) was and is being<br />

established in phase-III studies. Most of the completed trials<br />

have not been published in journals. They are only available<br />

as conference presentations or abstracts.<br />

Indication for chemotherapy<br />

Recommendation<br />

Metastatic pancreatic cancer is an indication for palliative chemotherapy.<br />

Recommendation grade: A, evidence level 2b, strong consensus<br />

Comment<br />

Palliative chemotherapy leads to longer survival, an improvement<br />

of quality of life, and “clinical benefit” i. e. especially use<br />

of less pain medication and less weight loss [200–202, 204].<br />

Recommendation<br />

Chemotherapy should be immediately initiated after metastases<br />

have been detected. It should not be postponed until tumor size<br />

progression, metastasis-induced symptoms, or other complications<br />

have developed.<br />

Recommendation grade: B, evidence level 5, strong consensus<br />

Recommendation<br />

Weight course, serum albumin, CA 19–9-value, hemoglobin value,<br />

and tumor differentiation grade at diagnosis do not influence the<br />

decision for or against chemotherapy.<br />

Recommendation grade: C, evidence level 3, strong consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Comment<br />

The benefit of chemotherapy is questionable for patients in poor<br />

general condition (KI < 70% ECOG > 2) [196, 205 –207]. This is<br />

confirmed by a subgroup analysis of a phase-III study that so<br />

far is only available in abstract form [208].<br />

Recommendation<br />

Patients with locally advanced, inoperable pancreatic carcinoma<br />

should also be treated from the time of diagnosis.<br />

Recommendation grade: B, evidence level 2b, strong consensus<br />

Comments<br />

In the subgroup of patients with locally advanced, inoperable<br />

pancreatic carcinoma current phase-III studies demonstrate a<br />

similar benefit of chemotherapy as for the metastatic situation<br />

[204, 206, 207, 209, 210].<br />

Drugs for palliative first line therapy<br />

Recommendation<br />

Data from several phase-III studies establish gemcitabine as a<br />

standard first line therapy for systemic palliative treatment.<br />

Recommendation grade: A, evidence level 1, consensus<br />

Comments<br />

This recommendation is based among others on several phase-<br />

III studies that show a consistent 1-year survival of 18 – 20%<br />

with gemcitabine therapy [204, 206, 207, 209, 210].<br />

Recommendation<br />

Gemcitabine should be given in conventional doses (1000 mg/m 2<br />

over 30 minutes).<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comments<br />

There is a pharmacokinetic rationale for protracted infusion of<br />

gemcitabine as a so-called fixed-dose-rate-infusion (10 mg/<br />

m 2 /min over 150 min; FDR). There is one published randomized<br />

phase-II study on this topic, which was negative in its<br />

primary endpoint (time to treatment failure) [211]. A phase-III<br />

study that compares the conventional gemcitabine therapy<br />

with FDR-infusion is only preliminary. However, it does not<br />

show a significant advantage of FDR-infusion [212].<br />

Recommendation<br />

5-FU/folinic acid is not recommended as a standard therapy.<br />

Recommendation grade: C, evidence level 4, consensus<br />

Comments<br />

Only phase-II data are available on protracted infusion of 5-FU<br />

for pancreatic carcinoma and these show inconsistent results.<br />

A direct comparison with gemcitabine in a phase-III study is<br />

missing [213–215].<br />

Recommendations<br />

Currently, combination chemotherapies with gemcitabine cannot<br />

be generally recommended as standard first line therapies for metastatic<br />

or locally advanced, inoperable pancreatic carcinoma.<br />

Recommendation grade: A, evidence level 1, consensus<br />

This is explained for individual combinations in the following in<br />

more detail.<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Gemcitabine/oxaliplatin or gemcitabine/cisplatin<br />

Recommendation<br />

This combination should not be used as standard.<br />

Recommendation grade: A, evidence level 1, consensus<br />

Comments<br />

There are 2 randomized, controlled studies on the combination<br />

of gemcitabine with oxaliplatin. Both show a benefit with respect<br />

to tumor response and a consistent trend towards longer survival<br />

for the combination therapy. However, the difference in survival<br />

did not reach significance in either study. The combination therapy,<br />

however, has a higher rate of side effects [206 –212].<br />

There are 2 randomized, controlled studies on the combination<br />

of gemcitabine with cisplatin. They demonstrate a consistent<br />

trend towards longer survival. However, these studies also<br />

show no statistically significant difference in survival. Clearly<br />

more side effects were seen than with the monotherapy [207,<br />

216].<br />

5-FU/oxaliplatin<br />

Recommendation<br />

This combination should not be used as standard treatment.<br />

Recommendation grade: A, evidence level 2a, strong consensus<br />

Comment<br />

There is only one small phase-II study with few numbers of patients<br />

that show an advantage of a combination of 5-FU and<br />

oxaliplatin compared to monotherapy with 5-FU or oxaliplatin<br />

[217].<br />

Gemcitabine/capecitabine or gemcitabine/5-FU<br />

Recommendation<br />

The combination gemcitabine and capecitabine or 5-FU is not recommended<br />

as standard therapy until definite publications are<br />

available.<br />

Recommendation grade: C, evidence level 2, consensus<br />

Comments<br />

The combination of gemictabine and oral fluoropyrimidin capecitabine<br />

was investigated in 2 randomized, controlled studies.<br />

A significantly better median and 1-year survival of the<br />

total population was shown for the combination in only one<br />

study [208, 218]. So far, these studies have merely been published<br />

as abstracts. Randomized, controlled studies on the combination<br />

of 5-FU and gemcitabine showed no significant difference<br />

in survival [209, 219, 220].<br />

Gemcitabine/erlotinib<br />

Recommendation<br />

A statistically significant difference in survival was demonstrated<br />

in favor of the combination of gemcitabine and the EGF-receptor<br />

tyrosine kinase inhibitor erlotinib [221]. The difference in median<br />

survival is about 2 weeks. The 1-year survival is 24% for the group<br />

receiving the combination therapy compared to 19% for the group<br />

receiving gemcitabine monotherapy.<br />

Recommendation grade: A, evidence level 1, consensus<br />

Comments<br />

An evaluation of the study that was published online by the<br />

CHMP of EMEA shows that there is a 22% increase in the median<br />

survival of patients with metastatic pancreatic carcinoma<br />

Leitlinie 467<br />

receiving gemcitabine plus erlotinib compared to gemcitabine<br />

plus placebo. However, the survival of patients with locally advanced,<br />

inoperable pancreatic carcinoma did not differ between<br />

both arms of the trial. Thus, the EMEA recommended the approval<br />

of a combination of gemcitabine and erlotinib for the indication<br />

“metastastic pancreatic carcinoma”. Particularly patients<br />

receiving a combination therapy with erlotinib who have<br />

a skin reaction (‡ grade 2) after 4 – 8 weeks seem to benefit. For<br />

patients who do not develop skin rash within the first 8 weeks<br />

of treatment, therapy with erlotinib should be reconsidered if<br />

there is no tumor response detectable with imaging techniques<br />

(i. e. mere stabilization of the disease, no partial or complete remission).<br />

Gemcitabine/bevacizumab<br />

Recommendation<br />

This combination is not recommended.<br />

Recommendation grade: B, evidence level 2, strong consensus<br />

Comments<br />

In a phase-II study it was shown that a combination of gemcitabine<br />

and the VEGF-antibody bevacizumab was beneficial.<br />

However, the toxicity was increased [222]. A current prematurely<br />

terminated phase-III study did not show a better survival<br />

of the combination treatment compared to monotherapy.<br />

So far the study is only available as an abstract [223].<br />

Capecitabine/oxaliplatin<br />

Recommendation<br />

The data available for the combination of capecitabine and oxaliplatin<br />

is insufficient.<br />

Recommendation grade: B, evidence level 2b, strong consensus.<br />

Recommendation<br />

Patients with a good Karnofsky-index (‡ 90% or ECOG 0 – 1) may<br />

benefit more from a therapy with gemcitabine/oxaliplatin, gemcitabine/cisplatin,<br />

or gemcitabine/capecitabine than from monotherapy.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comment<br />

Subgroup analyses of phase-III-studies demonstrate that patients<br />

with KPS ‡ 90% who receive combination therapy have<br />

a greater survival advantage [206 – 208].<br />

Other drugs<br />

Drugs such as mitomycin C (recommendation grade: A, evidence<br />

level 1) [213], a combination of cisplatin, epirubicine, 5-FU, and<br />

gemcitabine (PEFG) (recommendation grade: A, evidence level<br />

2) [224], and a combination of gemcitabine and docetaxel (recommendation<br />

grade: B, evidence level 2b) [225 –228] are not<br />

important in first line therapy of metastatic or locally advanced,<br />

inoperable pancreatic carcinoma.<br />

Second line treatment<br />

Recommendation<br />

If the first line therapy fails, a second line therapy can be done.<br />

This is especially true if the patient is in good general health, it<br />

is the patient’s wish, pretreatment was insufficient, or there is a<br />

good tumor response during first line therapy.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


468<br />

Leitlinie<br />

Comments<br />

Several studies, mostly phase-II, show the efficacy of second<br />

line therapy for metastatic pancreatic carcinoma with respect<br />

to tumor response. Treatments with 5-FU, capecitabine, or raltitrexed,<br />

also in combination with oxaliplatin, docetaxel, or irinotecan<br />

are possible [229 – 235].<br />

Palliative therapy algorithms<br />

Recommendation<br />

Palliative chemotherapy should be started with gemcitabine.<br />

Recommendation grade: B, evidence level 5, strong consensus<br />

Comment<br />

There are no studies that investigate the sequential palliative<br />

therapy of pancreatic carcinoma. It is recommended to start<br />

treatment with gemcitabine. An alternative for metastatic pancreatic<br />

carcinoma is a combination of gemcitabine and erlotinib.<br />

For patients with locally advanced, inoperable tumors and very<br />

good performance-status a combination of gemcitabine with a<br />

platinum analogue or capecitabine is possible (see above).<br />

Recommendation<br />

Standard practice is continuous treatment until progression.<br />

Recommendation grade: C, evidence level 5, strong consensus<br />

Comment<br />

So far, this problem has not been formally investigated. The current<br />

data available do not yet allow a change of the present practice<br />

of continuous therapy.<br />

Clinical efficacy of chemotherapy<br />

Recommendation<br />

Remission behavior, clinical benefit, or a combination of both are<br />

used to evaluate the clinical efficacy.<br />

Recommendation grade: A, evidence level 1b, strong consensus<br />

Comment<br />

These parameters are based on standard clinical practice which<br />

is supported by phase-II studies [204].<br />

Palliative radiation therapy<br />

Recommendation<br />

Indications for palliative radiation therapy are limited to symptomatic<br />

metastases (especially skeletal and cerebral metastases).<br />

Recommendation grade: A, evidence level 1, strong consensus<br />

Radiation therapy alone of a locally advanced, inoperable pancreatic<br />

tumor is not indicated.<br />

Recommendation grade: B, evidence level 2a, strong consensus<br />

[236]<br />

Radiochemotherapy for locally advanced, inoperable<br />

tumors<br />

Recommendation<br />

If radiation therapy is performed for locally advanced, inoperable<br />

pancreatic carcinoma, radiochemotherapy should be done.<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comment<br />

In several (mostly older) studies radiochemotherapy is better<br />

than radiation therapy alone for locally advanced, inoperable<br />

pancreatic carcinoma [237 –241]. However, the data are inconsistent.<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Recommendations<br />

Radiochemotherapy is currently not standard for locally advanced,<br />

inoperable pancreatic carcinoma. It may benefit patients with<br />

questionable resectable pancreatic carcinoma.<br />

Consensus<br />

In 5 randomized radiochemotherapy studies (randomized phase-<br />

II-studies) a median survival of 10.3 and 13.2 months was reported<br />

[242–246].<br />

Recommendation grade: C, evidence level 3<br />

Comparable survival times of 8.7 to 11.7 months were reported for<br />

locally advanced, inoperable pancreatic carcinoma in 4 randomized<br />

phase-III-studies on chemotherapy [206, 207, 210, 247].<br />

Recommendation grade: B, evidence level 2b, consensus<br />

Comment<br />

Older randomized phase-II-studies [239, 242], which directly<br />

compare radiochemotherapy and chemotherapy show no difference<br />

or a small advantage of radiochemotherapy. A recent abstract<br />

of a phase-III study shows a slight disadvantage of radiochemotherapy<br />

[248]. The current Cochrane-review reports that<br />

a better survival can be reached with radiochemotherapy compared<br />

to the best supportive therapy or radiation therapy alone.<br />

However, radiochemotherapy is associated with higher toxicity.<br />

The review concludes that currently there is not enough evidence<br />

to recommend radiochemotherapy as a better alternative<br />

to chemotherapy alone for patients with locally advanced, inoperable<br />

pancreatic carcinoma [203].<br />

Problematic for the radiochemotherapy concept is the fast development<br />

of metastases of some patients with pancreatic carcinoma.<br />

This is taken into consideration by a concept of the<br />

French GERCOR-group. A retrospective analysis showed that<br />

patients with locally advanced, inoperable pancreatic carcinoma<br />

who do not progress after 3 months of chemotherapy with<br />

respect to metastases benefit more from radiochemotherapy<br />

that is subsequently performed than from chemotherapy alone<br />

[249]. This concept should be tested in a prospective study.<br />

Recommendation<br />

Radiochemotherapy is not indicated for metastastic pancreatic<br />

carcinoma.<br />

Recommendation grade: B, evidence level 4, strong consensus<br />

If radiochemotherapy is performed for locally advanced, inoperable<br />

pancreatic carcinoma, it should be conventionally fractionated<br />

and three-dimensionally planned (single dose 1.8 – 2 Gy; total<br />

dose 50 – 54 Gy) so as not to increase the toxicity [243–246].<br />

Recommendation grade: C, evidence level 3, consensus<br />

Recommendation<br />

Surgery should be considered after pertinent staging (if necessary<br />

also by laparoscopy) if there is a clear response of the locally advanced<br />

pancreatic tumor to radiochemotherapy and if potential<br />

resectability is reached.<br />

Recommendation grade: C, evidence level 3, consensus<br />

Comment<br />

A secondary resectability can be reached in about 10% of patients<br />

who have undergone RCT [250].<br />

Recommendation<br />

If radiochemotherapy is performed outside of clinical trials, it<br />

should be done with infusional 5-FU.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Comment<br />

The largest amount of data are available for continuous infusion<br />

of the radiosensitizer 5-FU [243, 244, 246, 251, 252]. It has an acceptable<br />

side effect profile.<br />

Recommendation<br />

A combination of 5-FU/mitomycin C during radiochemotherapy<br />

should not be used outside of clinical trials because of increased<br />

hematologic toxicity [241, 253].<br />

Recommendation grade: A, evidence level 2b, consensus<br />

Gemcitabine or a combination of gemcitabine and cisplatin should<br />

also not be used outside of clinical trials because of increased toxicity<br />

[245, 246, 254–257].<br />

Recommendation grade: B, evidence level 3, consensus<br />

Importance of targeted-therapy approaches<br />

Recommendation<br />

Molecular therapy strategies such as marimastat (recommendation<br />

grade: A, evidence level 1), tanomastat (recommendation<br />

grade: A, evidence level 1), tipifarnib (recommendation grade:<br />

A, evidence level 1), and cetuximab (recommendation grade: C,<br />

evidence level 3) currently do not play a role in the palliative<br />

therapy of pancreatic cancer [210, 222, 258–260].<br />

Consensus<br />

Intra-arterial chemotherapy<br />

Recommendation<br />

Intra-arterial chemotherapy does not play a role in palliative<br />

treatment of pancreatic carcinomas [261, 262].<br />

Recommendation grade: B, evidence level 4, strong consensus<br />

Immunotherapeutic/gene therapeutic approaches<br />

Recommendation<br />

E Immunotherapeutic or gene therapeutic approaches<br />

Recommendation grade: C, evidence level 2b, consensus.<br />

E or hyperthermia € radiation therapy/radiochemo- or chemotherapy<br />

Recommendation grade: C, evidence level 4, consensus.<br />

currently do not play a role in palliative treatment of pancreatic<br />

carcinomas.<br />

Comment<br />

These therapeutic approaches are currently experimental and<br />

should not be used outside of clinical trials, because insufficient<br />

study data are available [263].<br />

Radiofrequency thermoablation<br />

Recommendation<br />

Radiofrequency thermoablation does not play a role in the palliative<br />

treatment of pancreatic carcinomas.<br />

Recommendation grade: B, evidence level 5, strong consensus<br />

Topic 6:<br />

Supportive therapy and follow-up<br />

!<br />

Pain therapy<br />

Recommendations<br />

The standard rules for tumor pain therapy apply for the treatment<br />

of pain in patients with pancreatic carcinoma. The WHO’s pain ladder<br />

is suitable for pain therapy of pancreatic carcinoma. Other procedures<br />

against pain are currently not available [264 – 266].<br />

Leitlinie 469<br />

It should be taken into account that the severity and the development<br />

of pain during pancreatic carcinoma can depend on the diet.<br />

The treatment procedure should be adapted accordingly (possible<br />

additional dose). Invasive neuroablative procedures (especially celiac<br />

blockade) also play a role in pancreatic carcinoma [267–269].<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Recommendation<br />

There are no specific criteria that influence the drug selection for<br />

pancreatic tumor pain therapy. This is true for non-opioids (NSAR,<br />

COXIBE, metamizol, paracetamol).<br />

Recommendation grade: D, evidence level: 4<br />

This recommendation also applies to the use and selection of<br />

opioids. There is no evidence of superiority of certain substances.<br />

Recommendation grade: A, evidence level 1b, strong consensus<br />

Comments<br />

Two reviews of studies on tumor pain treatment concluded<br />

that a meta-analysis is not possible for methodologic reasons.<br />

This is due to the heterogeneity of the methods and the completely<br />

different tools that were used to evaluate the results.<br />

Based on these restricted data, NSAR was superior to placebo.<br />

Only in a few studies the combination of NSAR and opioids<br />

was slightly better than each drug alone. Nonetheless, the difference<br />

was significant [270, 271].<br />

Recommendation<br />

A certain application route (oral or transdermal) for opioids should<br />

not be preferred for pancreatic carcinoma. Possible gastrointestinal<br />

problems, especially constipation, which can be caused by motility<br />

disorders must be taken into account [272, 273].<br />

Recommendation grade: D, evidence level 3, consensus<br />

Recommendation<br />

Adjuvants should be used according to the WHO-pain ladder. There<br />

are no specific recommendations for the use of adjuvants such as<br />

anti-emetica for pancreatic carcinoma [268, 274].<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Recommendation<br />

Invasive treatment methods (preferably subcutaneous or intravenous<br />

application of opioids, if necessary application near the spine)<br />

can be indicated if pain cannot be sufficiently controlled using the<br />

WHO-pain ladder [275].<br />

Recommendation grade: D, evidence level 3, strong consensus<br />

Recommendation<br />

Celiac blockade can principally be indicated for pancreatic carcinoma<br />

in some patients [276].<br />

Recommendation grade: C, evidence level 1b, strong consensus<br />

Comments<br />

There is a meta-analysis on the topic of celiac blockade which<br />

takes 59 publications into account [277]. However, only 24 report<br />

on 2 or more patients. Thus, more than half are case studies.<br />

21 have retrospective character. 63% use a celiac blockade<br />

for pancreatic carcinoma. The methods used were definitely not<br />

in line with present standards in all cases. In 32% no radiologic<br />

control of the alcohol application was done. Only 2 studies that<br />

were taken into account dealt with pain quality. The pain characteristics<br />

were also not considered. In summary, in 70 – 90% it<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


470<br />

Leitlinie<br />

was concluded that the method has a good long-term effect,<br />

side effects are transient and harmless, and severe complications<br />

are rare. Another publication [278] (evidence level 3) hypothesizes<br />

that the plexus blockade with concomitant administration<br />

of opioids improves pain therapy and survival. This is a<br />

double-blind, randomized study where patients receiving systemic<br />

therapy had a plexus blockade with alcohol or a “sham”plexus<br />

blockade. Furthermore, the quality of life (QoL) was taken<br />

into account. 100 patients with non-resectable pancreatic<br />

carcinoma and pain were included in the study. The plexus<br />

blockade significantly improved the pain reduction compared<br />

to systemic therapy alone. However, it did not affect the patients’<br />

survival and had no effect on the opioid dose.<br />

Recommendation<br />

The data available on the optimal time of celiac blockade are insufficient.<br />

Recommendation grade: D, evidence level 4, strong consensus<br />

Recommendation<br />

A certain method representing an optimal technical procedure for<br />

celiac blockade should not be preferred.<br />

Recommendation grade: C, evidence level 4, consensus<br />

Comments<br />

Radiologic procedures (fluoroscopy or CT) were not implemented<br />

in 32% in a meta-analysis by Eisenberg et al. [277]. There are<br />

no studies that compare the advantage and disadvantage of different<br />

methods (CT, fluoroscopy, US, EUS). There are no studies<br />

that compare various painkillers for the blockade. Also, different<br />

amounts of alcohol and local anesthetics were used.<br />

Recommendation<br />

The role of thoracoscopic splanchniectomy for pain therapy of pancreatic<br />

carcinoma cannot be definitely evaluated. This is a reserve<br />

procedure because of its invasiveness.<br />

Recommendation grade: D, evidence level 3, strong consensus<br />

Comments<br />

A retrospective analysis of 59 patients compares videothoracoscopic<br />

splanchniectomy and percutaneous celiac blockade in<br />

patients with inoperable pancreatic carcinoma with respect to<br />

pain reduction, quality of life, and opioid dose administered.<br />

Improvement of quality of life and a pronounced pain reduction<br />

was seen for both methods. Quality of life was slightly better for<br />

patients who received a plexus blockade. The amount of opioid<br />

that was necessary was reduced using both methods. Therefore,<br />

videothoracoscopic splanchniectomy is recommended as a reserve<br />

procedure, because the plexus blockade is less invasive<br />

and still as effective [279].<br />

Recommendation<br />

Radiation therapy with the sole aim of pain therapy is an exception<br />

for pancreatic carcinoma.<br />

Recommendation grade: D, consensus<br />

Comments<br />

The indication of radiation therapy for pain treatment is the exception,<br />

because of the immediate effect of e.g. a plexus blockade.<br />

Comparative studies are not available.<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Recommendation<br />

Pancreas enzymes are not suited for pain therapy of pancreatic<br />

carcinoma.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Recommendation<br />

Psycho-oncologic support may be useful to ease pain of patients<br />

with pancreatic carcinoma.<br />

Recommendation grade: B, evidence level 5, strong consensus<br />

Diet and tumor cachexia<br />

Enteral diet<br />

Recommendation<br />

There are no specific diet recommendations for patients with metastatic<br />

pancreatic carcinoma. In general, it should be kept in mind<br />

that patients with malignant tumors need a diet with sufficient<br />

calories because of progressive weight loss.<br />

A supportive or total enteral diet can help minimize weight loss<br />

if spontaneous food intake is insufficient [280].<br />

Recommendation grade: B, evidence level 3, strong consensus<br />

Recommendation<br />

There are no specific diet recommendations for patients with pancreatic<br />

carcinoma during radiation therapy. The guideline “enteral<br />

diet” of the DGEM 2003 states: “According to the data presently<br />

available a routine enteral diet is not indicated after radiation in<br />

the abdomen” [281].<br />

Recommendation grade: D, evidence level 5, strong consensus<br />

Vitamins, minerals and micronutrients<br />

Recommendation<br />

At physiologic concentrations, vitamins, minerals, and other micronutrients<br />

that are contained in food are important components<br />

of a balanced diet. There are no indications that taking<br />

vitamins, minerals, and other micronutrients in so-called pharmacologic<br />

doses is useful. Also, there is no proof of its harmlessness<br />

[281].<br />

Recommendation grade: B, evidence level 5, strong consensus<br />

Comment<br />

A recently published meta-analysis on the supplementation of<br />

anti-oxidants in primary and secondary prevention demonstrated<br />

that supplementation of the diet with beta-carotene, vitamin<br />

A, and vitamin E may increase the death rate instead of reducing<br />

it [282].<br />

Diet following pancreatectomy or during pancreatic duct<br />

obstruction<br />

Recommendation<br />

The consequences of exocrine and endocrine pancreas insufficiency<br />

have to be kept in mind when recommending a diet for<br />

patients after pancreatectomy or for patients with long-term<br />

pancreas duct obstruction. When treating exocrine pancreas insufficiency<br />

an adequate amount of pancreas enzymes must be<br />

given at meals. If pancreoprive diabetes is present, the patient<br />

should be treated with insulin according to standard principles.<br />

There are no further diet specific recommendations.<br />

Recommendation grade: B, evidence level 4, strong consensus<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Drugs to stimulate appetite against tumor cachexia<br />

Recommendation<br />

Although there are individual positive studies on several drugs, e. g.<br />

ibuprofen, megesterolacetate, steroids, thalidomide, and cannabinoids,<br />

the clinical role of appetite stimulating drug therapy for patients<br />

with metastatic pancreatic carcinoma with tumor cachexia<br />

cannot be definitely evaluated [283–286].<br />

Recommendation grade: D, evidence level 1b–3, strong consensus<br />

Comment<br />

The studies on individual substances are of different quality.<br />

Some are poor while others such as a controlled, randomized<br />

trial which compares treatment of dronabinol, megesterolacetate,<br />

and the combination of both for patients with tumor cachexia<br />

are fairly sound. This study shows that megesterolacetate<br />

monotherapy is superior. However, a placebo arm is missing,<br />

only about 1/3 of the patients had gastrointestinal tumors, and<br />

the number of patients with pancreatic carcinoma is not reported<br />

[285].<br />

Supportive therapy for further symptoms of advanced<br />

pancreatic carcinoma<br />

Recommendation<br />

An important goal of supportive therapy is the maintenance or<br />

improvement of the quality of life. The patient should be specifically<br />

asked about uncomfortable symptoms (e.g. fatigue, itching,<br />

diarrhea, constipation). These symptoms should be treated.<br />

Recommendation grade: B, evidence level 5, strong consensus<br />

Procedures for tumor-induced cholestasis<br />

Tumor-induced cholestasis is a frequent symptom which makes<br />

palliative therapy necessary for pancreatic carcinoma patients.<br />

Stents<br />

Recommendation<br />

Metal stents are the therapy of choice. If the survival time is expected<br />

to be < 3 months, plastic stents should be used.<br />

Recommendation grade: A, evidence level 1a, consensus<br />

Comments<br />

A meta-analysis based on 21 studies compares surgical procedures<br />

to stent-methods and metal stents to plastic ones. The studies<br />

included 1454 patients. Stent-techniques have a lower complication<br />

rate compared to surgery. However, they also have a<br />

higher relapse rate with respect to obstruction. There was a tendency<br />

for the group that had surgery to have a higher 30-day<br />

mortality. There was no difference in the technical and therapeutic<br />

success. Metal stents have a lower re-occlusion rate than<br />

plastic ones. There was no difference in the technical success, the<br />

primary therapeutic success, the complications, or the 30-day<br />

mortality. Overall, metal stents are recommended as first choice<br />

therapy [287].<br />

Recommendation<br />

If metal mesh stents are used, they do not have to be polyurethane<br />

coated.<br />

Recommendation grade: B, evidence level 2b, majority consensus<br />

Comments<br />

Two studies deal with polyurethane-coated stents [288, 289].<br />

The results of a one-arm study on polyurethane-coated stents<br />

including 30 patients correspond to those reported in the literature<br />

for non-coated metal stents [288]. Thus, there is no<br />

benefit of the coated stents. A second study with 112 patients<br />

compares the polyurethane-coated stents with self-expanding<br />

metal stents. It reports a significantly higher openness rate of<br />

distal bile duct stenoses for the former, because the coating<br />

prevents tumor infiltration. However, there was no difference<br />

in the survival between the groups. The group that received<br />

the coated stents demonstrated a higher rate of cholecystitis<br />

and pancreatitis [289].<br />

Recommendation<br />

Percutaneous transhepatic cholangiodrainage, PTCD, is sensible<br />

for palliative therapy of pancreatic carcinoma if endoscopic therapy<br />

(e. g. due to tumor-induced duodenal stenoses) is not possible.<br />

PTCD is also indicated if endoscopic therapy is not effective.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comment<br />

The literature on PTCD is “historic” [290]. There are no recent<br />

studies that compare the percutaneous application of metal<br />

stents with endoscopic access using the same stent design.<br />

Biliodigestive anastomosis<br />

Recommendation<br />

Surgery with the only goal of placing a biliodigestive anastomosis<br />

is surely the exception. However, the placement is indicated if the<br />

tumor turns out to be non-resectable during curative intended surgery<br />

and if cholestasis is present and survival time is expected to be<br />

longer. It must be differentiated between patients with peritoneal<br />

spreading or liver metastases.<br />

Recommendation grade: B, evidence level 5, consensus<br />

Recommendation<br />

Choledochojejunostomy should be preferred over other bypass procedures<br />

if a biliodigestive anastomosis is done in the palliative situation<br />

[291–293].<br />

Recommendation grade: B, evidence level 2, consensus<br />

Relapsing cholangitis after biliodigestive anastomosis<br />

Recommendation<br />

Before therapeutic intervention a drainage blockade must be excluded.<br />

If a mechanical hindrance is present necessary steps should<br />

be taken. Then long-term antibiotic treatment can be attempted.<br />

Recommendation grade: B, evidence level 5, consensus<br />

Comment<br />

There is no special literature on this topic.<br />

Leitlinie 471<br />

Tumor-induced stenoses of the upper gastrointestinal<br />

tract<br />

Recommendation<br />

A drug therapy attempt for tumor-induced functional pyloric stenosis<br />

is justified. The drugs metoclopramide and erythromycin<br />

should be preferred. Endoscopic stent placement and surgical intervention<br />

are not recommended.<br />

Recommendation grade: D, evidence level 5, strong consensus<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


472<br />

Leitlinie<br />

Comment<br />

There is no special literature on this topic. The recommendations<br />

are based on experience in standard clinical practice.<br />

Recommendation<br />

Principally there are two palliative therapeutic procedures available<br />

for obstruction of the duodenum: an endoscopic stent placement<br />

or a surgical gastroenterostomy. The literature that is available<br />

does not demonstrate a superiority of one of the methods.<br />

Recommendation grade: B, evidence level 3, consensus<br />

Comment<br />

The data published are mainly monocentric case study collections<br />

of patients with duodenal obstruction of different origins<br />

[294, 295]. There are no randomized prospective studies that<br />

compare endoscopic stenting and surgical gastroenterostomy.<br />

Prophylactic gastroenterostomy<br />

Recommendation<br />

According to the current data it seems sensible to perform a prophylactic<br />

gastroenterostomy if irresectability is diagnosed during<br />

surgery.<br />

Recommendation grade: C, evidence level 2b, consensus<br />

Comment<br />

The publication on this topic, a monocentric, prospective, randomized<br />

study shows that a prophylactic gastrojejunostomy<br />

for non-resectable periampullary carcinoma significantly reduces<br />

the future development of pyloric stenosis [145].<br />

Importance of hematopoetic growth factors for locally<br />

advanced pancreatic carcinoma<br />

Recommendation<br />

Granulocyte-stimulating growth factors (G-CSF or GM-CSFs) are<br />

not important for the supportive therapy of locally advanced pancreatic<br />

carcinoma.<br />

Recommendation grade: A, evidence level 1, strong consensus<br />

Comment<br />

Chemotherapy that is so aggressive that the administration of<br />

granulocyte-stimulating growth factors is necessary is not recommended<br />

for palliative treatment of locally advanced or metastatic<br />

pancreatic carcinoma. Refer to the current guidelines of<br />

ASCO on the use of growth factors [296].<br />

Recommendation<br />

Erythrocyte-stimulating growth factors (erythropoietin) can under<br />

certain circumstances play a role.<br />

Recommendation grade: A, evidence level 1, strong consensus<br />

Comment<br />

Also, refer to the current guidelines of ASCO [297] or EORTC<br />

[298].<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Role of cytoprotectives/radical scavengers during<br />

chemotherapy and/or radiation therapy of pancreatic<br />

carcinoma<br />

Recommendation<br />

Amifostin or other cytoprotectives do not play a role in chemotherapy<br />

and/or radiation therapy of pancreatic carcinoma.<br />

Recommendation grade: A, evidence level 1, strong consensus<br />

Comment<br />

This recommendation follows the meta-analysis of the chemotherapy<br />

and radiation therapy expert panel of the ASCO [299].<br />

Evaluation of pancreatic carcinoma patients’ quality of<br />

life<br />

Recommendation<br />

QLQ-C30 and the related specific pancreas module QLQ-PAN 26 offer<br />

suitable tools for the assessment of quality of life.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Comment<br />

The routine implementation of QLQ-C30 and QLQ-PAN 26 are<br />

only reasonable for clinical studies.<br />

Follow-up<br />

Follow-up program after curative resection<br />

Recommendation<br />

A structured follow-up program cannot be recommended independent<br />

of the pancreatic carcinoma stage.<br />

Recommendation grade: A, evidence level 5, consensus<br />

Comments<br />

There is no scientific proof that structured follow-up with regular<br />

staging examinations leads to an improvement of the survival<br />

of patients with pancreatic cancer.<br />

Medical history and a physical examination are necessary if exocrine<br />

or endocrine insufficiency is suspected. The examination<br />

can be performed regularly by a general practitioner.<br />

Rehabilitation following curative pancreatic carcinoma resection<br />

Recommendation<br />

For individual patients rehabilitation may be useful under certain<br />

conditions. This should be coordinated with the family.<br />

Recommendation grade: D, evidence level 5, consensus<br />

Comment<br />

There is no assessable literature on the role of rehabilitation<br />

after pancreatic carcinoma resection with curative intent.<br />

Acknowledgements<br />

!<br />

We would like to thank the staff of the Clinic for Internal Medicine<br />

I, University of Ulm, T. Locher, R. Lorenz, M. Porzner, I. Rueß,<br />

A. Stein, E. Thanner, and Christian Aslan for the support in preparing<br />

and implementing the consensus conference.<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Appendix<br />

!<br />

Table 10 Search terms for the literature search: search level 1: all task forces: pancreatic cancer | pancreatic neoplasm | pancreatic carcinoma | ductal adenocarcinoma<br />

of the pancreas; task force-specific search levels 2 – 5: task force<br />

search level 2<br />

topic 1<br />

risk factors<br />

search level 3 search level 4 search level 5<br />

risk factor nutrition * | dietary fibre | fiber |<br />

legume|fruit|vegetables<br />

vitaminc|fat|cholesterol|<br />

red meat | white meat | barbecued<br />

|<br />

grilled | nitrate | nitrite | fish<br />

sugar | saccharide | milk | milk<br />

produce | alcohol | coffee | tea<br />

lifestyle<br />

occupational risks<br />

| *exercise | sports | physical activity<br />

| weight | obesity | nicotine<br />

| weight | obesity | nicotine |<br />

prophylaxis |<br />

vitamins |<br />

prevention<br />

NSAID<br />

screening CA19–9 | genomic analysis | genetic<br />

analysis | DNA array tool |<br />

technique | method<br />

Cost-effectiveness<br />

risk group<br />

* | prevention | propyhlaxis tool | technique | method | genetic analysis<br />

sporadic<br />

screening | surveillance<br />

CA19–9 |<br />

pancreatic juice<br />

(k-ras/p16)<br />

risk group<br />

* | familial pancreatic | hereditary * | genetic consultant<br />

genetic analysis<br />

hereditary<br />

pancreatic cancer | prevention | tool | technique | method<br />

(k-ras/p16)<br />

prophylaxis |<br />

CA19–9 |<br />

genetic consultation<br />

surveillance<br />

pancreatic juice<br />

Peutz-Jeghers Syndrome | FAMMM<br />

Syndrome | pancreatic cancer<br />

melanoma syndrome | Hereditary<br />

breast/ovarian cancer (BRCA2) |<br />

HNPCC | Ataxia teleangiectasia |<br />

FAP<br />

Cystic fibrosis | Li-Fraumeni syndrome<br />

| Lindau’s disease | Neurofibromatosis<br />

| Fanconi’s anemia |<br />

Seattle family X | Familial fibrocystic<br />

pancreatic atrophy |<br />

Hereditary pancreatitis<br />

risk group chronic pancreatitis<br />

diabetes<br />

topic 2<br />

diagnostics<br />

diagnosis abdominal pain| back ache |back<br />

pain | hyperglycemia | diabetes |<br />

jaundice |<br />

acute pancreatitis<br />

ERCP | ultrasound | MRI |<br />

endoscopic ultrasound |<br />

computed tomography | MRCP<br />

therapy | pancreatectomy<br />

surveillance | CA 19–9<br />

age<br />

supposed malignancy |<br />

elevated CA19–9 |<br />

sensitivity<br />

cytology | aspirate<br />

laboratory test | serum marker<br />

bile duct | pancreatic duct<br />

pancreatic tumor | lesion biopsy | surgery solitary | curative |<br />

size | localisation |<br />

technique |<br />

palliative |<br />

metastatic<br />

cystic | pseudocyst |<br />

serous | mucinous<br />

resection | biopsy | aspirate<br />

Leitlinie 473<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


474<br />

Leitlinie<br />

Table 10 (Fortsetzung)<br />

search level 2 search level 3 search level 4 search level 5<br />

biopsy pathological finding |<br />

content | criteria |<br />

histological report<br />

pathological report<br />

pancreatectomy<br />

preoperative<br />

ultrasound | computer<br />

diagnostic |<br />

tomography | ct | mri |<br />

staging |<br />

bone scan | chest x-ray |<br />

topic 3<br />

response to chemotherapy<br />

endoscopic ultrasound |<br />

ercp | mrcp | pet |<br />

micrometastases |<br />

laparoscopy |<br />

surgical treatment of pancreatic carcinoma (curative intention)<br />

surgery inoperable | nonresectable age | elderly |<br />

Celiac artery |<br />

comorbidity |<br />

truncus coeliacus |<br />

infiltration<br />

Superior mesenteric artery |<br />

Superior mesenteric vein |<br />

Liver/hepatic vein<br />

cholestasis<br />

resection margins<br />

preoperative stent |<br />

ERCP<br />

inoperative ultrasonography |<br />

peritoneal lavage<br />

cytologic examination<br />

perioperative antibiotics |<br />

somastatin |<br />

octreotide<br />

stent<br />

tumor of the head | pylorus preserving<br />

| tumor of the tail | tumor<br />

ofthebody|locallyadvanced<br />

growth | extended lymphadenectomy<br />

| cystic tumors of the<br />

pancreas | intracuctal papillary<br />

mucinous tumor | metastatic |<br />

metastases | laparoscopic | frozen<br />

section | rapid section |<br />

R0 resection<br />

pancreatic fistula<br />

topic 4<br />

adjuvant and neoadjuvant non-surgical therapy of pancreatic carcinoma<br />

adjuvant<br />

chemotherapy<br />

adjuvant chemoradiation | chemoradiotherapy<br />

| radiochemotherapy<br />

radiotherapy |<br />

radiation<br />

chemoradiation |<br />

chemoradiotherapy | radiochemotherapy<br />

histology | pathology |criteria<br />

inflammation | neoplastic<br />

curative resection | R0 resection differentiation | locally<br />

advanced |mircometastases<br />

Age | elderly | ECOG | 5-FU/folinic<br />

acid (Mayo) | 5-FU/folinic acid<br />

(AIO) | Gemcitabine | CapecetabineRegional|Start|timeaftersurgery<br />

| postoperative begin | R 1<br />

resection<br />

R0 resection | UICC |<br />

risk factors<br />

5-fluorouracil continuous infusion<br />

| 5-fluorouracil | Mitomycin |<br />

Gemcitabine | age | elderly |<br />

ECOG | R 1 resection<br />

curative intention<br />

curative intention<br />

chemotherapy curative intention<br />

neoadjuvant<br />

radiotherapy |<br />

neoadjuvant<br />

radiation<br />

locally advanced | inoperable | T 4<br />

| protocol | duration | dosis |<br />

technique | chemotherapy<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

differentiation |<br />

locally advanced |<br />

micrometastases<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


Table 10 (Fortsetzung)<br />

search level 2 search level 3 search level 4 search level 5<br />

radiotherapy |<br />

downsizing |<br />

locally advanced | T4 |<br />

surgery<br />

chemoradiation |<br />

tumor mass reduction |<br />

inoperable<br />

radiochemotherapy |<br />

chemoradiotherapy<br />

stable disease<br />

neoadjuvant<br />

radiochemotherapy<br />

intraoperative radiation |<br />

intraoperative radiotherapy<br />

adjuvant therapy |<br />

neoadjuvant therapy<br />

locally advanced |<br />

inoperable<br />

surgery | radiochemotherapy<br />

UICC | risk factors differentiation | locally<br />

advanced | micrometastases<br />

targeted therapies<br />

topic 5<br />

palliative chemotherapy and radiationtherapy<br />

palliative chemotherapy * | metastases | metastatic | progressive<br />

| size | prognostic factors<br />

symptoms | locally advanced | inoperable<br />

| first line |<br />

gemcitabine<br />

5-fluorouracil/folinic acid (AIO) |<br />

gemcitabine/oxaliplatin |<br />

gemcitabine/cisplatin |<br />

oxaliplatin/5-fluorouracil/folinic<br />

acid |<br />

gemcitabine/capecitabine | gemcitabine/erlotinib<br />

|<br />

gemcitabine/bevacizumab|<br />

capecitabine/oxaliplatin<br />

polychemotherapy<br />

second line<br />

Karnofski | ECOG | albumin |<br />

CA19–9 | differentiation |<br />

hemoglobin<br />

fixed dose rate | infusion<br />

*|indication|<br />

5-FU/folinic acid (Mayo) |<br />

5-fluorouracil continuous<br />

infusion |<br />

oxaliplatin/5-fluorouracil/<br />

folinic acid |<br />

capectiabine mono |<br />

capecitabine/oxaliplatin<br />

third line * | inidcation |<br />

oxaliplatin/5-fluorouracil/<br />

folinic acid |<br />

5-fluorouracil/mitomycin |<br />

capecitabine/mitomycin<br />

sequence | order |<br />

duration | time |<br />

surveillance | efficacy<br />

palliative radiotherapy locally advanced | inoperable |<br />

metastatic | symptoms | protocoll<br />

|chemotherapy<br />

* | 5-fluorouracil continous<br />

infusion |<br />

5-fluorouracil/mitomycin |<br />

gemcitabine<br />

palliative therapies targeted therapy marimastat | tanomastat |<br />

tipifamib | bevacizumab |<br />

cetuximab<br />

topic 6<br />

supportive therapy<br />

intra-arterial chemotherapy<br />

immunotherapy | gene therapy |<br />

hyperthermia | radiofrequency<br />

thermoablation<br />

* | indication<br />

palliation pain therapy * | WHO | non-opioids |<br />

opioids<br />

nmda antagonists |<br />

celiac plexus block |<br />

pancreatic enzymes |<br />

thorascoscopic splanchnicectomy<br />

| radiotherapy | radiation<br />

application | antiemetics | constipation<br />

nutrition metastatic | cachexia nutrition | vitamins<br />

Leitlinie 475<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


476<br />

Leitlinie<br />

Table 10 (Fortsetzung)<br />

search level 2 search level 3 search level 4 search level 5<br />

radiotherapy | radiation |<br />

pancreatectomy<br />

supplements<br />

appetite corticosteroids | dronabinol | megestrol<br />

acetate | growth hormone<br />

| Eicosapentaenoic acid | probiotics<br />

| ibuprofen<br />

symptoms fatigue | pruritus | constipation |<br />

diarrhea | pleural effusion<br />

cholestatsis stent | stenting<br />

PTCD<br />

plastic | metal<br />

biliodigestive anastomosis cholangitis<br />

gastric outlet obstruciton drug therapy<br />

stent | stenting | surgery<br />

erythromycin |<br />

metoclopramide<br />

duodenal obstruction<br />

palliative surgery<br />

stent | stenting | surgery<br />

hematopoietic growth factors * | granulocytes | erythrocytes<br />

cytoprotective agents * | amifostine<br />

pancreatic enzymes pancreatectomy<br />

follow-up |<br />

after-care<br />

curative resection rehabilitation<br />

References<br />

1 Adler G, Seufferlein T, Bischoff SC et al. <strong>S3</strong>-<strong>Guideline</strong>s <strong>“Exocrine</strong> pancreatic<br />

cancer” 2007. Z Gastroenterol 2007; 45: 487–523<br />

2 Glade MJ. Food, nutrition, and the prevention of cancer: a global perspective.<br />

American Institute for Cancer Research/World Cancer Research<br />

Fund, American Institute for Cancer Research, 1997. Nutrition<br />

1999; 15: 523–526<br />

3 Fernandez E, La Vecchia C, Decarli A. Attributable risks for pancreatic<br />

cancer in northern Italy. Cancer Epidemiol Biomarkers Prev 1996; 5:<br />

23–27<br />

4 Ji BT, Chow WH, Gridley G et al. Dietary factors and the risk of pancreatic<br />

cancer: a case-control study in Shanghai China. Cancer Epidemiol<br />

Biomarkers Prev 1995; 4: 885–893<br />

5 Soler M, Chatenoud L, La Vecchia C et al. Diet, alcohol, coffee and pancreatic<br />

cancer: final results from an Italian study. Eur J Cancer Prev<br />

1998; 7: 455–460<br />

6 Nkondjock A, Krewski D, Johnson KC et al. Dietary patterns and risk of<br />

pancreatic cancer. Int J Cancer 2005; 114: 817–823<br />

7 Nothlings U, Wilkens LR, Murphy SP et al. Meat and fat intake as risk<br />

factors for pancreatic cancer: the multiethnic cohort study. J Natl<br />

Cancer Inst 2005; 97: 1458–1465<br />

8 Michaud DS, Giovannucci E, Willett WC et al. Dietary meat, dairy products,<br />

fat, and cholesterol and pancreatic cancer risk in a prospective<br />

study. Am J Epidemiol 2003; 157: 1115–1125<br />

9 Michaud DS, Skinner HG, Wu K et al. Dietary patterns and pancreatic<br />

cancer risk in men and women. J Natl Cancer Inst 2005; 97: 518–524<br />

10 Hine RJ, Srivastava S, Milner JA et al. Nutritional links to plausible mechanisms<br />

underlying pancreatic cancer: a conference report. Pancreas<br />

2003; 27: 356–366<br />

11 de Bueno Mesquita HB, Maisonneuve P, Runia S et al. Intake of foods<br />

and nutrients and cancer of the exocrine pancreas: a populationbased<br />

case-control study in The Netherlands. Int J Cancer 1991; 48:<br />

540–549<br />

12 Lyon JL, Slattery ML, Mahoney AW et al. Dietary intake as a risk factor<br />

for cancer of the exocrine pancreas. Cancer Epidemiol Biomarkers<br />

Prev 1993; 2: 513–518<br />

13 Fraser GE. Associations between diet and cancer, ischemic heart disease,<br />

and all-cause mortality in non-Hispanic white California Seventh-day<br />

Adventists. Am J Clin Nutr 1999; 70: 532S–538S<br />

14 Mills PK, Beeson WL, Abbey DE et al. Dietary habits and past medical<br />

history as related to fatal pancreas cancer risk among Adventists.<br />

Cancer 1988; 61: 2578–2585<br />

15 Chan JM, Wang F, Holly EA. Vegetable and fruit intake and pancreatic<br />

cancer in a population-based case-control study in the San Francisco<br />

bay area. Cancer Epidemiol Biomarkers Prev 2005; 14: 2093–2097<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

16 Negri E, La Vecchia C, Franceschi S et al. Vegetable and fruit consumption<br />

and cancer risk. Int J Cancer 1991; 48: 350–354<br />

17 Larsson SC, Hakansson N, Naslund I et al. Fruit and vegetable consumption<br />

in relation to pancreatic cancer risk: a prospective study.<br />

Cancer Epidemiol Biomarkers Prev 2006; 15: 301–305<br />

18 Vainio H, Weiderpass E. Fruit and vegetables in cancer prevention.<br />

Nutr Cancer 2006; 54: 111–142<br />

19 Lin Y, Tamakoshi A, Hayakawa T et al. Nutritional factors and risk of<br />

pancreatic cancer: a population-based case-control study based on<br />

direct interview in Japan. J Gastroenterol 2005; 40: 297–301<br />

20 Stolzenberg-Solomon RZ, Pietinen P, Taylor PR et al. Prospective study<br />

of diet and pancreatic cancer in male smokers. Am J Epidemiol 2002;<br />

155: 783–792<br />

21 Zhang J, Zhao Z, Berkel HJ. Animal fat consumption and pancreatic<br />

cancer incidence: evidence of interaction with cigarette smoking.<br />

Ann Epidemiol 2005; 15: 500–508<br />

22 Anderson KE, Sinha R, Kulldorff M et al. Meat intake and cooking techniques:<br />

associations with pancreatic cancer. Mutat Res 2002; 506 –<br />

507: 225–231<br />

23 Anderson KE, Kadlubar FF, Kulldorff M et al. Dietary intake of heterocyclic<br />

amines and benzo(a)pyrene: associations with pancreatic cancer.<br />

Cancer Epidemiol Biomarkers Prev 2005; 14: 2261–2265<br />

24 Ghadirian P, Baillargeon J, Simard A et al. Food habits and pancreatic<br />

cancer: a case-control study of the Francophone community in Montreal,<br />

Canada. Cancer Epidemiol Biomarkers Prev 1995; 4: 895–899<br />

25 Fernandez E, Chatenoud L, La Vecchia C et al. Fish consumption and<br />

cancer risk. Am J Clin Nutr 1999; 70: 85–90<br />

26 Michaud DS, Liu S, Giovannucci E et al. Dietary sugar, glycemic load,<br />

and pancreatic cancer risk in a prospective study. J Natl Cancer Inst<br />

2002; 94: 1293–1300<br />

27 Silvera SA, Rohan TE, Jain M et al. Glycemic index, glycemic load, and<br />

pancreatic cancer risk (Canada). Cancer Causes Control 2005; 16:<br />

431–436<br />

28 Schernhammer ES, Hu FB, Giovannucci E et al. Sugar-sweetened soft<br />

drink consumption and risk of pancreatic cancer in two prospective<br />

cohorts. Cancer Epidemiol Biomarkers Prev 2005; 14: 2098–2105<br />

29 Lin Y, Tamakoshi A, Kawamura T et al. Risk of pancreatic cancer in relation<br />

to alcohol drinking, coffee consumption and medical history:<br />

findings from the Japan collaborative cohort study for evaluation of<br />

cancer risk. Int J Cancer 2002; 99: 742–746<br />

30 Michaud DS, Giovannucci E, Willett WC et al. Coffee and alcohol consumption<br />

and the risk of pancreatic cancer in two prospective United<br />

States cohorts. Cancer Epidemiol Biomarkers Prev 2001; 10:<br />

429–437<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


31 Silverman DT. Risk factors for pancreatic cancer: a case-control study<br />

based on direct interviews. Teratog Carcinog Mutagen 2001; 21: 7–<br />

25<br />

32 Brown LM. Epidemiology of alcohol-associated cancers. Alcohol<br />

2005; 35: 161–168<br />

33 Ye W, Lagergren J, Weiderpass E et al. Alcohol abuse and the risk of<br />

pancreatic cancer. Gut 2002; 51: 236–239<br />

34 Talamini G, Bassi C, Falconi M et al. Alcohol and smoking as risk factors<br />

in chronic pancreatitis and pancreatic cancer. Dig Dis Sci 1999;<br />

44: 1303–1311<br />

35 MacMahon B, Yen S, Trichopoulos D et al. Coffee and cancer of the<br />

pancreas. N Engl J Med 1981; 304: 630–633<br />

36 Gullo L, Pezzilli R, Morselli-Labate AM. Coffee and cancer of the pancreas:<br />

an Italian multicenter study. The Italian <strong>Pancreatic</strong> Cancer<br />

Study Group. Pancreas 1995; 11: 223–229<br />

37 Lyon JL, Mahoney AW, French TK et al. Coffee consumption and the<br />

risk of cancer of the exocrine pancreas: a case-control study in a<br />

low-risk population. Epidemiology 1992; 3: 164–170<br />

38 Harnack LJ, Anderson KE, Zheng W et al. Smoking, alcohol, coffee, and<br />

tea intake and incidence of cancer of the exocrine pancreas: the Iowa<br />

Women’s Health Study. Cancer Epidemiol Biomarkers Prev 1997; 6:<br />

1081–1086<br />

39 Qiu D, Kurosawa M, Lin Y et al. Overview of the epidemiology of pancreatic<br />

cancer focusing on the JACC Study. J Epidemiol 2005; 15<br />

(Suppl 2): S157–167<br />

40 Tavani A, La Vecchia C. Coffee and cancer: a review of epidemiological<br />

studies, 1990 – 1999. Eur J Cancer Prev 2000; 9: 241–256<br />

41 La Vecchia C, Negri E, Franceschi S et al. Tea consumption and cancer<br />

risk. Nutr Cancer 1992; 17: 27–31<br />

42 Nagano J, Kono S, Preston DL et al. A prospective study of green tea<br />

consumption and cancer incidence, Hiroshima and Nagasaki (Japan).<br />

Cancer Causes Control 2001; 12: 501–508<br />

43 Hemminki K, Dong C, Vaittinen P. Cancer risks to spouses and offspring<br />

in the Family-Cancer Database. Genet Epidemiol 2001; 20:<br />

247–257<br />

44 Hemminki K, Jiang Y. Cancer risks among long-standing spouses. Br J<br />

Cancer 2002; 86: 1737–1740<br />

45 Nilsen TI, Vatten LJ. A prospective study of lifestyle factors and the<br />

risk of pancreatic cancer in Nord-Trondelag, Norway. Cancer Causes<br />

Control 2000; 11: 645–652<br />

46 Berrington de Gonzalez A, Sweetland S, Spencer E. A meta-analysis of<br />

obesity and the risk of pancreatic cancer. Br J Cancer 2003; 89: 519–<br />

523<br />

47 Michaud DS, Giovannucci E, Willett WC et al. Physical activity, obesity,<br />

height, and the risk of pancreatic cancer. Jama 2001; 286: 921–929<br />

48 Patel AV, Rodriguez C, Bernstein L et al. Obesity, recreational physical<br />

activity, and risk of pancreatic cancer in a large U. S. Cohort. Cancer<br />

Epidemiol Biomarkers Prev 2005; 14: 459–466<br />

49 Rapp K, Schroeder J, Klenk J et al. Obesity and incidence of cancer: a<br />

large cohort study of over 145000 adults in Austria. Br J Cancer<br />

2005; 93: 1062–1067<br />

50 Larsson SC, Permert J, Hakansson N et al. Overall obesity, abdominal<br />

adiposity, diabetes and cigarette smoking in relation to the risk of<br />

pancreatic cancer in two Swedish population-based cohorts. Br J<br />

Cancer 2005; 93: 1310–1315<br />

51 Berrington de Gonzalez A, Spencer EA, Bueno-de-Mesquita HB et al. Anthropometry,<br />

physical activity, and the risk of pancreatic cancer in<br />

the European prospective investigation into cancer and nutrition.<br />

Cancer Epidemiol Biomarkers Prev 2006; 15: 879–885<br />

52 Silverman DT, Swanson CA, Gridley G et al. Dietary and nutritional factors<br />

and pancreatic cancer: a case-control study based on direct interviews.<br />

J Natl Cancer Inst 1998; 90: 1710–1719<br />

53 Stolzenberg-Solomon RZ, Pietinen P, Taylor PR et al. A prospective<br />

study of medical conditions, anthropometry, physical activity, and<br />

pancreatic cancer in male smokers (Finland). Cancer Causes Control<br />

2002; 13: 417–426<br />

54 Hanley AJ, Johnson KC, Villeneuve PJ et al. Physical activity, anthropometric<br />

factors and risk of pancreatic cancer: results from the Canadian<br />

enhanced cancer surveillance system. Int J Cancer 2001; 94:<br />

140–147<br />

55 Coughlin SS, Calle EE, Patel AV et al. Predictors of pancreatic cancer<br />

mortality among a large cohort of United States adults. Cancer Causes<br />

Control 2000; 11: 915–923<br />

Leitlinie 477<br />

56 Lin Y, Tamakoshi A, Kawamura T et al. A prospective cohort study of<br />

cigarette smoking and pancreatic cancer in Japan. Cancer Causes<br />

Control 2002; 13: 249–254<br />

57 Yun YH, Jung KW, Bae JM et al. Cigarette smoking and cancer incidence<br />

risk in adult men: National Health Insurance Corporation<br />

Study. Cancer Detect Prev 2005; 29: 15–24<br />

58 Chiu BC, Lynch CF, Cerhan JR et al. Cigarette smoking and risk of bladder,<br />

pancreas, kidney, and colorectal cancers in Iowa. Ann Epidemiol<br />

2001; 11: 28–37<br />

59 Bonelli L, Aste H, Bovo P et al. Exocrine pancreatic cancer, cigarette<br />

smoking, and diabetes mellitus: a case-control study in northern Italy.<br />

Pancreas 2003; 27: 143–149<br />

60 Duell EJ, Holly EA, Bracci PM et al. A population-based, case-control<br />

study of polymorphisms in carcinogen-metabolizing genes, smoking,<br />

and pancreatic adenocarcinoma risk. J Natl Cancer Inst 2002;<br />

94: 297–306<br />

61 Miyasaka K, Kawanami T, Shimokata H et al. Inactive aldehyde dehydrogenase-2<br />

increased the risk of pancreatic cancer among smokers<br />

in a Japanese male population. Pancreas 2005; 30: 95–98<br />

62 Wang L, Miao X, Tan W et al. Genetic polymorphisms in methylenetetrahydrofolate<br />

reductase and thymidylate synthase and risk of<br />

pancreatic cancer. Clin Gastroenterol Hepatol 2005; 3: 743–751<br />

63 Villeneuve PJ, Johnson KC, Mao Y et al. Environmental tobacco smoke<br />

and the risk of pancreatic cancer: findings from a Canadian population-based<br />

case-control study. Can J Public Health 2004; 95: 32–37<br />

64 Alguacil J, Pollan M, Gustavsson P. Occupations with increased risk of<br />

pancreatic cancer in the Swedish population. Occup Environ Med<br />

2003; 60: 570–576<br />

65 Alguacil J, Porta M, Benavides FG et al. Occupation and pancreatic<br />

cancer in Spain: a case-control study based on job titles. PANKRAS<br />

II Study Group. Int J Epidemiol 2000; 29: 1004–1013<br />

66 Laakkonen A, Kauppinen T, Pukkala E. Cancer risk among Finnish food<br />

industry workers. Int J Cancer 2006; 118: 2567–2571<br />

67 Fryzek JP, Garabrant DH, Harlow SD et al. A case-control study of selfreported<br />

exposures to pesticides and pancreas cancer in southeastern<br />

Michigan. Int J Cancer 1997; 72: 62–67<br />

68 Ji BT, Silverman DT, Stewart PA et al. Occupational exposure to pesticides<br />

and pancreatic cancer. Am J Ind Med 2001; 39: 92–99<br />

69 Ojajarvi IA, Partanen TJ, Ahlbom A et al. Occupational exposures and<br />

pancreatic cancer: a meta-analysis. Occup Environ Med 2000; 57:<br />

316–324<br />

70 Ojajarvi A, Partanen T, Ahlbom A et al. Risk of pancreatic cancer in<br />

workers exposed to chlorinated hydrocarbon solvents and related<br />

compounds: a meta-analysis. Am J Epidemiol 2001; 153: 841–850<br />

71 Weiderpass E, Vainio H, Kauppinen T et al. Occupational exposures<br />

and gastrointestinal cancers among Finnish women. J Occup Environ<br />

Med 2003; 45: 305–315<br />

72 Yassi A, Tate RB, Routledge M. Cancer incidence and mortality in<br />

workers employed at a transformer manufacturing plant: update to<br />

a cohort study. Am J Ind Med 2003; 44: 58–62<br />

73 Ji J, Hemminki K. Socioeconomic and occupational risk factors for<br />

pancreatic cancer: a cohort study in Sweden. J Occup Environ Med<br />

2006; 48: 283–288<br />

74 Bjelakovic G, Nikolova D, Simonetti RG et al. Antioxidant supplements<br />

for prevention of gastrointestinal cancers: a systematic review and<br />

meta-analysis. Lancet 2004; 364: 1219–1228<br />

75 Bjelakovic G, Nikolova D, Simonetti RG et al. Antioxidant supplements<br />

for preventing gastrointestinal cancers. Cochrane Database Syst Rev<br />

CD 004183 2004<br />

76 Harris RE, Beebe-Donk J, Doss H et al. Aspirin, ibuprofen, and other<br />

non-steroidal anti-inflammatory drugs in cancer prevention: a critical<br />

review of non-selective COX-2 blockade (review). Oncol Rep<br />

2005; 13: 559–583<br />

77 Jacobs EJ, Connell CJ, Rodriguez C et al. Aspirin use and pancreatic cancer<br />

mortality in a large United States cohort. J Natl Cancer Inst 2004;<br />

96: 524–528<br />

78 Coogan PF, Rosenberg L, Palmer JR et al. Nonsteroidal anti-inflammatory<br />

drugs and risk of digestive cancers at sites other than the large<br />

bowel. Cancer Epidemiol Biomarkers Prev 2000; 9: 119–123<br />

79 Homma T, Tsuchiya R. The study of the mass screening of persons<br />

without symptoms and of the screening of outpatients with gastrointestinal<br />

complaints or icterus for pancreatic cancer in Japan, using<br />

CA 19–9 and elastase-1 or ultrasonography. Int J Pancreatol 1991; 9:<br />

119–124<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


478<br />

Leitlinie<br />

80 Kim JE, Lee KT, Lee JK et al. Clinical usefulness of carbohydrate antigen<br />

19–9 as a screening test for pancreatic cancer in an asymptomatic<br />

population. J Gastroenterol Hepatol 2004; 19: 182–186<br />

81 McWilliams RR, Rabe KG, Olswold C et al. Risk of malignancy in firstdegree<br />

relatives of patients with pancreatic carcinoma. Cancer 2005;<br />

104: 388–394<br />

82 Klein AP, Brune KA, Petersen GM et al. Prospective risk of pancreatic<br />

cancer in familial pancreatic cancer kindreds. Cancer Res 2004; 64:<br />

2634–2638<br />

83 Bartsch DK, Sina-Frey M, Lang S et al. CDKN2A germline mutations in<br />

familial pancreatic cancer. Ann Surg 2002; 236: 730–737<br />

84 McFaul CD, Greenhalf W, Earl J et al. Anticipation in familial pancreatic<br />

cancer. Gut 2006; 55: 252–258<br />

85 Tersmette AC, Petersen GM, Offerhaus GJ et al. Increased risk of incident<br />

pancreatic cancer among first-degree relatives of patients<br />

with familial pancreatic cancer. Clin Cancer Res 2001; 7: 738–744<br />

86 Brentnall TA. Management strategies for patients with hereditary<br />

pancreatic cancer. Curr Treat Options Oncol 2005; 6: 437–445<br />

87 Canto MI, Goggins M, Yeo CJ et al. Screening for pancreatic neoplasia<br />

in high-risk individuals: an EUS-based approach. Clin Gastroenterol<br />

Hepatol 2004; 2: 606–621<br />

88 Canto MI, Goggins M, Hruban RH et al. Screening for early pancreatic<br />

neoplasia in high-risk individuals: a prospective controlled study.<br />

Clin Gastroenterol Hepatol 2006; 4: 766–781; quiz 665<br />

89 Giardiello FM, Brensinger JD, Tersmette AC et al. Very high risk of cancer<br />

in familial Peutz-Jeghers syndrome. Gastroenterology 2000; 119:<br />

1447–1453<br />

90 Lim W, Olschwang S, Keller JJ et al. Relative frequency and morphology<br />

of cancers in STK11 mutation carriers. Gastroenterology 2004;<br />

126: 1788–1794<br />

91 Lynch HT, Brand RE, Hogg D et al. Phenotypic variation in eight extended<br />

CDKN2A germline mutation familial atypical multiple mole<br />

melanoma-pancreatic carcinoma-prone families: the familial atypical<br />

mole melanoma-pancreatic carcinoma syndrome. Cancer 2002;<br />

94: 84–96<br />

92 Rulyak SJ, Brentnall TA, Lynch HT et al. Characterization of the neoplastic<br />

phenotype in the familial atypical multiple-mole melanoma-pancreatic<br />

carcinoma syndrome. Cancer 2003; 98: 798–804<br />

93 Vasen HF, Gruis NA, Frants RR et al. Risk of developing pancreatic cancer<br />

in families with familial atypical multiple mole melanoma associated<br />

with a specific 19 deletion of p16 (p16-Leiden). Int J Cancer<br />

2000; 87: 809–811<br />

94 Friedenson B. BRCA1 and BRCA2 pathways and the risk of cancers<br />

other than breast or ovarian. MedGenMed 2005; 7: 60<br />

95 Asperen CJvan, Brohet RM, Meijers-Heijboer EJ et al. Cancer risks in<br />

BRCA2 families: estimates for sites other than breast and ovary.<br />

J Med Genet 2005; 42: 711–719<br />

96 Thompson D, Easton DF. Cancer Incidence in BRCA1 mutation carriers.<br />

J Natl Cancer Inst 2002; 94: 1358–1365<br />

97 Lilley M, Gilchrist D. The hereditary spectrum of pancreatic cancer:<br />

the Edmonton experience. Can J Gastroenterol 2004; 18: 17–21<br />

98 Aarnio M, Sankila R, Pukkala E et al. Cancer risk in mutation carriers<br />

of DNA-mismatch-repair genes. Int J Cancer 1999; 81: 214–218<br />

99 Geoffroy-Perez B, Janin N, Ossian K et al. Cancer risk in heterozygotes<br />

for ataxia-telangiectasia. Int J Cancer 2001; 93: 288–293<br />

100 Giardiello FM, Offerhaus GJ, Lee DH et al. Increased risk of thyroid and<br />

pancreatic carcinoma in familial adenomatous polyposis. Gut 1993;<br />

34: 1394–1396<br />

101 Couch FJ, Johnson MR, Rabe K et al. Germ line Fanconi anemia complementation<br />

group C mutations and pancreatic cancer. Cancer Res<br />

2005; 65: 383–386<br />

102 Howes N, Lerch MM, Greenhalf W et al. Clinical and genetic characteristics<br />

of hereditary pancreatitis in Europe. Clin Gastroenterol Hepatol<br />

2004; 2: 252–261<br />

103 Lowenfels AB, Maisonneuve P, DiMagno EP et al. Hereditary pancreatitis<br />

and the risk of pancreatic cancer. International Hereditary Pancreatitis<br />

Study Group. J Natl Cancer Inst 1997; 89: 442–446<br />

104 Kimmey MB, Bronner MP, Byrd DR et al. Screening and surveillance<br />

for hereditary pancreatic cancer. Gastrointest Endosc 2002; 56:<br />

S82–86<br />

105 Bansal P, Sonnenberg A. Pancreatitis is a risk factor for pancreatic<br />

cancer. Gastroenterology 1995; 109: 247–251<br />

106 Karlson BM, Ekbom A, Josefsson S et al. The risk of pancreatic cancer<br />

following pancreatitis: an association due to confounding? Gastroenterology<br />

1997; 113: 587–592<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

107 Lowenfels AB, Maisonneuve P, Cavallini G et al. Pancreatitis and the<br />

risk of pancreatic cancer. International Pancreatitis Study Group. N<br />

Engl J Med 1993; 328: 1433–1437<br />

108 Malka D, Hammel P, Maire F et al. Risk of pancreatic adenocarcinoma<br />

in chronic pancreatitis. Gut 2002; 51: 849–852<br />

109 Talamini G, Falconi M, Bassi C et al. Incidence of cancer in the course<br />

of chronic pancreatitis. Am J Gastroenterol 1999; 94: 1253–1260<br />

110 Chari ST, Leibson CL, Rabe KG et al. Probability of pancreatic cancer<br />

following diabetes: a population-based study. Gastroenterology<br />

2005; 129: 504–511<br />

111 Coughlin SS, Calle EE, Teras LR et al. Diabetes mellitus as a predictor of<br />

cancer mortality in a large cohort of US adults. Am J Epidemiol 2004;<br />

159: 1160–1167<br />

112 Huxley R, Ansary-Moghaddam A, Berrington de Gonzalez A et al. Type-<br />

II diabetes and pancreatic cancer. A meta-analysis of 36 studies. Br J<br />

Cancer 2005; 92: 2076–2083<br />

113 Stolzenberg-Solomon RZ, Graubard BI, Chari S et al. Insulin, glucose,<br />

insulin resistance, and pancreatic cancer in male smokers. Jama<br />

2005; 294: 2872–2878<br />

114 Wideroff L, Gridley G, Mellemkjaer L et al. Cancer incidence in a population-based<br />

cohort of patients hospitalized with diabetes mellitus<br />

in Denmark. J Natl Cancer Inst 1997; 89: 1360–1365<br />

115 Calle EE, Murphy TK, Rodriguez C et al. Diabetes mellitus and pancreatic<br />

cancer mortality in a prospective cohort of United States adults.<br />

Cancer Causes Control 1998; 9: 403–410<br />

116 Chow WH, Gridley G, Nyren O et al. Risk of pancreatic cancer following<br />

diabetes mellitus: a nationwide cohort study in Sweden. J Natl Cancer<br />

Inst 1995; 87: 930–931<br />

117 Silverman DT, Schiffman M, Everhart J et al. Diabetes mellitus, other<br />

medical conditions and familial history of cancer as risk factors for<br />

pancreatic cancer. Br J Cancer 1999; 80: 1830–1837<br />

118 Everhart J, Wright D. Diabetes mellitus as a risk factor for pancreatic<br />

cancer. A meta-analysis. Jama 1995; 273: 1605–1609<br />

119 Gullo L, Pezzilli R, Morselli-Labate AM. Diabetes and the risk of pancreatic<br />

cancer. Italian <strong>Pancreatic</strong> Cancer Study Group. N Engl J Med<br />

1994; 331: 81–84<br />

120 Rousseau MC, Parent ME, Pollak MN et al. Diabetes mellitus and cancer<br />

risk in a population-based case-control study among men from<br />

Montreal, Canada. Int J Cancer 2006; 118: 2105–2109<br />

121 Bjornsson E, Ismael S, Nejdet S et al. Severe jaundice in Sweden in the<br />

new millennium: causes, investigations, treatment and prognosis.<br />

Scand J Gastroenterol 2003; 38: 86–94<br />

122 Reisman Y, Gips CH, Lavelle SM et al. Clinical presentation of (subclinical)<br />

jaundice – the Euricterus project in The Netherlands. United<br />

Dutch Hospitals and Euricterus Project Management Group. Hepatogastroenterology<br />

1996; 43: 1190–1195<br />

123 Watanabe I, Sasaki S, Konishi M et al. Onset symptoms and tumor locations<br />

as prognostic factors of pancreatic cancer. Pancreas 2004;<br />

28: 160–165<br />

124 Balthazar EJ. Pancreatitis associated with pancreatic carcinoma. Preoperative<br />

diagnosis: role of CT imaging in detection and evaluation.<br />

Pancreatology 2005; 5: 330–344<br />

125 Mujica VR, Barkin JS, Go VL. Acute pancreatitis secondary to pancreatic<br />

carcinoma. Study Group Participants. Pancreas 2000; 21: 329–<br />

332<br />

126 Adamek HE, Albert J, Breer H et al. <strong>Pancreatic</strong> cancer detection with<br />

magnetic resonance cholangiopancreatography and endoscopic retrograde<br />

cholangiopancreatography: a prospective controlled study.<br />

Lancet 2000; 356: 190–193<br />

127 Hanninen EL, Ricke J, Amthauer H et al. Magnetic resonance cholangiopancreatography:<br />

image quality, ductal morphology, and value<br />

of additional T2- and T 1-weighted sequences for the assessment of<br />

suspected pancreatic cancer. Acta Radiol 2005; 46: 117–125<br />

128 Forsmark CE, Lambiase L, Vogel SB. Diagnosis of pancreatic cancer and<br />

prediction of unresectability using the tumor-associated antigen<br />

CA19–9. Pancreas 1994; 9: 731–734<br />

129 Nazli O, Bozdag AD, Tansug T et al. The diagnostic importance of CEA<br />

and CA19–9 for the early diagnosis of pancreatic carcinoma. Hepatogastroenterology<br />

2000; 47: 1750–1752<br />

130 Ritts Jr RE, Nagorney DM, Jacobsen DJ et al. Comparison of preoperative<br />

serum CA19–9 levels with results of diagnostic imaging modalities<br />

in patients undergoing laparotomy for suspected pancreatic or<br />

gallbladder disease. Pancreas 1994; 9: 707–716<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


131 Tessler DA, Catanzaro A, Velanovich V et al. Predictors of cancer in patients<br />

with suspected pancreatic malignancy without a tissue diagnosis.<br />

Am J Surg 2006; 191: 191–197<br />

132 Agarwal B, Abu-Hamda E, Molke KL et al. Endoscopic ultrasound-guided<br />

fine needle aspiration and multidetector spiral CT in the diagnosis<br />

of pancreatic cancer. Am J Gastroenterol 2004; 99: 844–850<br />

133 Klapman JB, Chang KJ, Lee JG et al. Negative predictive value of endoscopic<br />

ultrasound in a large series of patients with a clinical suspicion<br />

of pancreatic cancer. Am J Gastroenterol 2005; 100: 2658–2661<br />

134 Varadarajulu S, Wallace MB. Applications of endoscopic ultrasonography<br />

in pancreatic cancer. Cancer Control 2004; 11: 15–22<br />

135 David O, Green L, Reddy V et al. <strong>Pancreatic</strong> masses: a multi-institutional<br />

study of 364 fine-needle aspiration biopsies with histopathologic<br />

correlation. Diagn Cytopathol 1998; 19: 423–427<br />

136 Bipat S, Phoa SS, Delden OM et al. Ultrasonography, computed tomography<br />

and magnetic resonance imaging for diagnosis and determining<br />

resectability of pancreatic adenocarcinoma: a meta-analysis.<br />

J Comput Assist Tomogr 2005; 29: 438–445<br />

137 Dewitt J, Devereaux BM, Lehman GA et al. Comparison of endoscopic<br />

ultrasound and computed tomography for the preoperative evaluation<br />

of pancreatic cancer: a systematic review. Clin Gastroenterol<br />

Hepatol 2006; 4: 717–725; quiz 664<br />

138 Romagnuolo J, Bardou M, Rahme E et al. Magnetic resonance cholangiopancreatography:<br />

a meta-analysis of test performance in suspected<br />

biliary disease. Ann Intern Med 2003; 139: 547–557<br />

139 Pakzad F, Groves AM, Ell PJ. The role of positron emission tomography<br />

in the management of pancreatic cancer. Semin Nucl Med 2006; 36:<br />

248–256<br />

140 Schachter PP, Avni Y, Shimonov M et al. The impact of laparoscopy and<br />

laparoscopic ultrasonography on the management of pancreatic cancer.<br />

Arch Surg 2000; 135: 1303–1307<br />

141 Vollmer CM, Drebin JA, Middleton WD et al. Utility of staging laparoscopy<br />

in subsets of peripancreatic and biliary malignancies. Ann<br />

Surg 2002; 235: 1–7<br />

142 van der Waaij LA, Dullemen HM, Porte RJ. Cyst fluid analysis in the differential<br />

diagnosis of pancreatic cystic lesions. A pooled analysis.<br />

Gastrointest Endosc 2005; 62: 383–389<br />

143 Fong Y, Blumgart LH, Fortner JG et al. <strong>Pancreatic</strong> or liver resection for<br />

malignancy is safe and effective for the elderly. Ann Surg 1995; 222:<br />

426–434; discussion 434 – 437<br />

144 Lightner AM, Glasgow RE, Jordan TH et al. <strong>Pancreatic</strong> resection in the<br />

elderly. J Am Coll Surg 2004; 198: 697–706<br />

145 Lillemoe KD, Cameron JL, Hardacre JM et al. Is prophylactic gastrojejunostomy<br />

indicated for unresectable periampullary cancer? A prospective<br />

randomized trial. Ann Surg 1999; 230: 322–328; discussion<br />

328 – 330<br />

146 American College of Physicians. <strong>Guideline</strong>s for assessing and managing<br />

the perioperative risk from coronary artery disease associated<br />

with major noncardiac surgery. Ann Intern Med 1997; 127: 309–312<br />

147 Wagner M, Redaelli C, Lietz M et al. Curative resection is the single<br />

most important factor determining outcome in patients with pancreatic<br />

adenocarcinoma. Br J Surg 2004; 91: 586–594<br />

148 Fuhrman GM, Leach SD, Staley CA et al. Rationale for en bloc vein resection<br />

in the treatment of pancreatic adenocarcinoma adherent to<br />

the superior mesenteric-portal vein confluence. <strong>Pancreatic</strong> Tumor<br />

Study Group. Ann Surg 1996; 223: 154–162<br />

149 Leach SD, Lee JE, Charnsangavej C et al. Survival following pancreaticoduodenectomy<br />

with resection of the superior mesenteric-portal<br />

vein confluence for adenocarcinoma of the pancreatic head. Br J<br />

Surg 1998; 85: 611–617<br />

150 Sasson AR, Hoffman JP, Ross EA et al. En bloc resection for locally advanced<br />

cancer of the pancreas: is it worthwhile? J Gastrointest Surg<br />

2002; 6: 147–157; discussion 157 – 158<br />

151 Nakao A, Takeda S, Inoue S et al. Indications and techniques of extended<br />

resection for pancreatic cancer. World J Surg 2006; 30: 976–982;<br />

discussion 983 – 984<br />

152 Varadhachary GR, Tamm EP, Abbruzzese JL et al. Borderline resectable<br />

pancreatic cancer: definitions, management, and role of preoperative<br />

therapy. Ann Surg Oncol 2006; 13: 1035–1046<br />

153 Siriwardana HP, Siriwardena AK. Systematic review of outcome of<br />

synchronous portal-superior mesenteric vein resection during pancreatectomy<br />

for cancer. Br J Surg 2006; 93: 662–673<br />

154 Carrere N, Sauvanet A, Goere D et al. <strong>Pancreatic</strong>oduodenectomy with<br />

mesentericoportal vein resection for adenocarcinoma of the pancreatic<br />

head. World J Surg 2006; 30: 1526–1535<br />

Leitlinie 479<br />

155 Riediger H, Makowiec F, Fischer E et al. Postoperative morbidity and<br />

long-term survival after pancreaticoduodenectomy with superior<br />

mesenterico-portal vein resection. J Gastrointest Surg 2006; 10:<br />

1106–1115<br />

156 Bachellier P, Nakano H, Oussoultzoglou PD et al. Is pancreaticoduodenectomy<br />

with mesentericoportal venous resection safe and worthwhile?<br />

Am J Surg 2001; 182: 120–129<br />

157 Li B, Chen FZ, Ge XH et al. Pancreatoduodenectomy with vascular reconstruction<br />

in treating carcinoma of the pancreatic head. Hepatobiliary<br />

Pancreat Dis Int 2004; 3: 612–615<br />

158 Settmacher U, Langrehr JM, Husmann I et al. Reconstruction of visceral<br />

arteries with homografts in excision of the pancreas. Chirurg<br />

2004; 75: 1199–1206<br />

159 Schwarz RE. Technical considerations to maintain a low frequency of<br />

postoperative biliary stent-associated infections. J Hepatobiliary<br />

Pancreat Surg 2002; 9: 93–97<br />

160 Gerke H, White R, Byrne MF et al. Complications of pancreaticoduodenectomy<br />

after neoadjuvant chemoradiation in patients with and<br />

without preoperative biliary drainage. Dig Liver Dis 2004; 36: 412–<br />

418<br />

161 Jagannath P, Dhir V, Shrikhande S et al. Effect of preoperative biliary<br />

stenting on immediate outcome after pancreaticoduodenectomy. Br<br />

J Surg 2005; 92: 356–361<br />

162 Martignoni ME, Wagner M, Krahenbuhl L et al. Effect of preoperative<br />

biliary drainage on surgical outcome after pancreatoduodenectomy.<br />

Am J Surg 2001; 181: 52–59; discussion 87<br />

163 Sohn TA, Yeo CJ, Cameron JL et al. Do preoperative biliary stents increase<br />

postpancreaticoduodenectomy complications? J Gastrointest<br />

Surg 2000; 4: 258–267; discussion 267 – 268<br />

164 Targarona EM, Garau J, Munoz-Ramos C et al. Single-dose antibiotic<br />

prophylaxis in patients at high risk for infection in biliary surgery: a<br />

prospective and randomized study comparing cefonicid with mezlocillin.<br />

Surgery 1990; 107: 327–334<br />

165 Kujath P, Bouchard R, Scheele J et al. Current perioperative antibiotic<br />

prophylaxis. Chirurg 2006; 77: 490, 492–498<br />

166 Connor S, Alexakis N, Garden OJ et al. Meta-analysis of the value of somatostatin<br />

and its analogues in reducing complications associated<br />

with pancreatic surgery. Br J Surg 2005; 92: 1059–1067<br />

167 Bassi C, Dervenis C, Butturini G et al. Postoperative pancreatic fistula:<br />

an international study group (ISGPF) definition. Surgery 2005; 138:<br />

8–13<br />

168 Finlayson C, Hoffman J, Yeung R et al. Intraoperative ultrasound does<br />

not improve detection of liver metastases in resectable pancreatic<br />

cancer. Am J Surg 1998; 175: 99–101<br />

169 Kolecki R, Schirmer B. Intraoperative and laparoscopic ultrasound.<br />

Surg Clin North Am 1998; 78: 251–271<br />

170 Warshaw AL, Fernandez-del Castillo C. <strong>Pancreatic</strong> carcinoma. N Engl J<br />

Med 1992; 326: 455–465<br />

171 Konishi M, Kinoshita T, Nakagohri T et al. Prognostic value of cytologic<br />

examination of peritoneal washings in pancreatic cancer. Arch Surg<br />

2002; 137: 475–480<br />

172 Yachida S, Fukushima N, Sakamoto M et al. Implications of peritoneal<br />

washing cytology in patients with potentially resectable pancreatic<br />

cancer. Br J Surg 2002; 89: 573–578<br />

173 Diener MK, Knaebel HP, Heukaufer C et al. A Systematic Review and<br />

Meta-analysis of Pylorus-preserving Versus Classical <strong>Pancreatic</strong>oduodenectomy<br />

for Surgical Treatment of Periampullary and <strong>Pancreatic</strong><br />

Carcinoma. Ann Surg 2007; 245: 187–200<br />

174 Christein JD, Kendrick ML, Iqbal CW et al. Distal pancreatectomy for<br />

resectable adenocarcinoma of the body and tail of the pancreas.<br />

J Gastrointest Surg 2005; 9: 922–927<br />

175 Shimada K, Sakamoto Y, Sano T et al. Prognostic factors after distal<br />

pancreatectomy with extended lymphadenectomy for invasive pancreatic<br />

adenocarcinoma of the body and tail. Surgery 2006; 139:<br />

288–295<br />

176 Shoup M, Conlon KC, Klimstra D et al. Is extended resection for adenocarcinoma<br />

of the body or tail of the pancreas justified? J Gastrointest<br />

Surg 2003; 7: 946–952; discussion 952<br />

177 Pedrazzoli S, DiCarlo V, Dionigi R et al. Standard versus extended lymphadenectomy<br />

associated with pancreatoduodenectomy in the surgical<br />

treatment of adenocarcinoma of the head of the pancreas: a<br />

multicenter, prospective, randomized study. Lymphadenectomy<br />

Study Group. Ann Surg 1998; 228: 508–517<br />

178 Farnell MB, Pearson RK, Sarr MG et al. A prospective randomized trial<br />

comparing standard pancreatoduodenectomy with pancreatoduo-<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


480<br />

Leitlinie<br />

denectomy with extended lymphadenectomy in resectable pancreatic<br />

head adenocarcinoma. Surgery 2005; 138: 618–628; discussion<br />

628 – 630<br />

179 Yeo CJ, Cameron JL, Lillemoe KD et al. <strong>Pancreatic</strong>oduodenectomy with<br />

or without distal gastrectomy and extended retroperitoneal lymphadenectomy<br />

for periampullary adenocarcinoma, part 2: randomized<br />

controlled trial evaluating survival, morbidity, and mortality. Ann<br />

Surg 2002; 236: 355–366; discussion 366 – 368<br />

180 Brugge WR, Lauwers GY, Sahani D et al. Cystic neoplasms of the pancreas.<br />

N Engl J Med 2004; 351: 1218–1226<br />

181 Brugge WR, Lewandrowski K, Lee-Lewandrowski E et al. Diagnosis of<br />

pancreatic cystic neoplasms: a report of the cooperative pancreatic<br />

cyst study. Gastroenterology 2004; 126: 1330–1336<br />

182 Sedlack R, Affi A, Vazquez-Sequeiros E et al. Utility of EUS in the evaluation<br />

of cystic pancreatic lesions. Gastrointest Endosc 2002; 56:<br />

543–547<br />

183 Salvia R, Fernandez-del Castillo C, Bassi C et al. Main-duct intraductal<br />

papillary mucinous neoplasms of the pancreas: clinical predictors of<br />

malignancy and long-term survival following resection. Ann Surg<br />

2004; 239: 678–685; discussion 685 – 687<br />

184 Bassi C, Salvia R, Molinari E et al. Management of 100 consecutive<br />

cases of pancreatic serous cystadenoma: wait for symptoms and see<br />

at imaging or vice versa? World J Surg 2003; 27: 319–323<br />

185 Hirano S, Kondo S, Ambo Y et al. Outcome of duodenum-preserving<br />

resection of the head of the pancreas for intraductal papillary-mucinous<br />

neoplasm. Dig Surg 2004; 21: 242–245<br />

186 Takada T, Yasuda H, Amano H et al. Simultaneous hepatic resection<br />

with pancreato-duodenectomy for metastatic pancreatic head carcinoma:<br />

does it improve survival? Hepatogastroenterology 1997; 44:<br />

567–573<br />

187 Mann O, Strate T, Schneider C et al. Surgery for advanced and metastatic<br />

pancreatic cancer – current state and perspectives. Anticancer<br />

Res 2006; 26: 681–686<br />

188 Shrikhande SV, Kleeff J, Reiser C et al. <strong>Pancreatic</strong> resection for M1 pancreatic<br />

ductal adenocarcinoma. Ann Surg Oncol 2007; 14: 118–127<br />

189 Friess H, Kleeff J, Silva JC et al. The role of diagnostic laparoscopy in<br />

pancreatic and periampullary malignancies. J Am Coll Surg 1998;<br />

186: 675–682<br />

190 Urbach DR, Swanstrom LL, Hansen PD. The effect of laparoscopy on<br />

survival in pancreatic cancer. Arch Surg 2002; 137: 191–199<br />

191 Santoro E, Carlini M, Carboni F. Laparoscopic pancreatic surgery: indications,<br />

techniques and preliminary results. Hepatogastroenterology<br />

1999; 46: 1174–1180<br />

192 Cioc AM, Ellison EC, Proca DM et al. Frozen section diagnosis of pancreatic<br />

lesions. Arch Pathol Lab Med 2002; 126: 1169–1173<br />

193 Luttges J, Zamboni G, Kloppel G. Recommendation for the examination<br />

of pancreaticoduodenectomy specimens removed from patients<br />

with carcinoma of the exocrine pancreas. A proposal for a standardized<br />

pathological staging of pancreaticoduodenectomy specimens<br />

including a checklist. Dig Surg 1999; 16: 291–296<br />

194 Compton CC, Henson DE. Protocol for the examination of specimens<br />

removed from patients with carcinoma of the exocrine pancreas: a<br />

basis for checklists. Cancer Committee, College of American Pathologists.<br />

Arch Pathol Lab Med 1997; 121: 1129–1136<br />

195 Sakata E, Shirai Y, Yokoyama N et al. Clinical significance of lymph<br />

node micrometastasis in ampullary carcinoma. World J Surg 2006;<br />

30: 985–991<br />

196 Oettle H, Post S, Neuhaus P et al. Adjuvant chemotherapy with gemcitabine<br />

vs observation in patients undergoing curative-intent resection<br />

of pancreatic cancer: a randomized controlled trial. Jama 2007;<br />

297: 267–277<br />

197 Stocken DD, Buchler MW, Dervenis C et al. Meta-analysis of randomised<br />

adjuvant therapy trials for pancreatic cancer. Br J Cancer<br />

2005; 92: 1372–1381<br />

198 Neoptolemos JP, Stocken DD, Friess H et al. A randomized trial of chemoradiotherapy<br />

and chemotherapy after resection of pancreatic<br />

cancer. N Engl J Med 2004; 350: 1200–1210<br />

199 Penberthy DR, Rich TA, Shelton CH et al. A pilot study of chronomodulated<br />

infusional 5-fluorouracil chemoradiation for pancreatic cancer.<br />

Ann Oncol 2001; 12: 681–684<br />

200 Mallinson CN, Rake MO, Cocking JB et al. Chemotherapy in pancreatic<br />

cancer: results of a controlled, prospective, randomised, multicentre<br />

trial. Br Med J 1980; 281: 1589–1591<br />

201 Palmer KR, Kerr M, Knowles G et al. Chemotherapy prolongs survival<br />

in inoperable pancreatic carcinoma. Br J Surg 1994; 81: 882–885<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

202 Glimelius B, Hoffman K, Sjoden PO et al. Chemotherapy improves survival<br />

and quality of life in advanced pancreatic and biliary cancer.<br />

Ann Oncol 1996; 7: 593–600<br />

203 Yip D, Karapetis C, Strickland A et al. Chemotherapy and radiotherapy<br />

for inoperable advanced pancreatic cancer. Cochrane Database Syst<br />

Rev 3 CD002093 2006<br />

204 Burris HA3 rd, Moore MJ, Andersen J et al. Improvements in survival<br />

and clinical benefit with gemcitabine as first-line therapy for patients<br />

with advanced pancreas cancer: a randomized trial. J Clin Oncol<br />

1997; 15: 2403–2413<br />

205 Storniolo AM, Enas NH, Brown CA et al. An investigational new drug<br />

treatment program for patients with gemcitabine: results for over<br />

3000 patients with pancreatic carcinoma. Cancer 1999; 85: 1261–<br />

1268<br />

206 Louvet C, Labianca R, Hammel P et al. Gemcitabine in combination<br />

with oxaliplatin compared with gemcitabine alone in locally advanced<br />

or metastatic pancreatic cancer: results of a GERCOR and<br />

GISCAD phase III trial. J Clin Oncol 2005; 23: 3509–3516<br />

207 Heinemann V, Quietzsch D, Gieseler F et al. Randomized phase III trial<br />

of gemcitabine plus cisplatin compared with gemcitabine alone in<br />

advanced pancreatic cancer. J Clin Oncol 2006; 24: 3946–3952<br />

208 Herrmann R, Bodoky G, Rushstaller T et al. Gemcitabine (G) plus capecitabine<br />

(C) versus G alone in locally advanced or metastatic pancreatic<br />

cancer: a randomized phase III study of the Swiss Group for<br />

Clinical Cancer Research (SAKK) and the Central European Cooperative<br />

Oncology Group (CECOG). J Clin Oncol 2005; 23: A4010<br />

209 Berlin JD, Catalano P, Thomas JP et al. Phase III study of gemcitabine<br />

in combination with fluorouracil versus gemcitabine alone in patients<br />

with advanced pancreatic carcinoma: Eastern Cooperative<br />

Oncology Group Trial E 2297. J Clin Oncol 2002; 20: 3270–3275<br />

210 Van Cutsem E, Velde H, Karasek P et al. Phase III trial of gemcitabine<br />

plus tipifarnib compared with gemcitabine plus placebo in advanced<br />

pancreatic cancer. J Clin Oncol 2004; 22: 1430–1438<br />

211 Tempero M, Plunkett W, Van Ruiz Haperen V et al. Randomized phase<br />

II comparison of dose-intense gemcitabine: thirty-minute infusion<br />

and fixed dose rate infusion in patients with pancreatic adenocarcinoma.<br />

J Clin Oncol 2003; 21: 3402–3408<br />

212 Poplin E, Levy D, Berlin J et al. Phase III trial of gemcitabine (30 min<br />

infusion) versus gemcitabine (fixed-dose rate infusion (FDR) versus<br />

gemcitabine + oxaliplatin (GEMOX) in patients with advanced pancreatic<br />

cancer (E6201). J Clin Oncol 2006; 24: LBA4004<br />

213 Maisey N, Chau I, Cunningham D et al. Multicenter randomized phase<br />

III trial comparing protracted venous infusion (PVI) fluorouracil (5-<br />

FU) with PVI 5-FU plus mitomycin in inoperable pancreatic cancer.<br />

J Clin Oncol 2002; 20: 3130–3136<br />

214 Ducreux M, Rougier P, Pignon JP et al. A randomised trial comparing<br />

5-FU with 5-FU plus cisplatin in advanced pancreatic carcinoma.<br />

Ann Oncol 2002; 13: 1185–1191<br />

215 Lutz MP, Koniger M, Muche R et al. A phase II study of weekly 24-h infusion<br />

of high-dose 5-fluorouracil in advanced pancreatic cancer. Z<br />

Gastroenterol 1999; 37: 993–997<br />

216 Colucci G, Giuliani F, Gebbia V et al. Gemcitabine alone or with cisplatin<br />

for the treatment of patients with locally advanced and/or metastatic<br />

pancreatic carcinoma: a prospective, randomized phase III<br />

study of the Gruppo Oncologia dell’Italia Meridionale. Cancer 2002;<br />

94: 902–910<br />

217 Ducreux M, Mitry E, Ould-Kaci M et al. Randomized phase II study<br />

evaluating oxaliplatin alone, oxaliplatin combined with infusional<br />

5-FU, and infusional 5-FU alone in advanced pancreatic carcinoma<br />

patients. Ann Oncol 2004; 15: 467–473<br />

218 Cunningham D, Chau I, Stocken DD et al. Phase III randomized comparison<br />

of gemcitabine (GEM) vs. gemcitabine plus capecitabine<br />

(Gem-CAP) in patients with advanced pancreatic cancer. Eur J Cancer<br />

2005; 3: PS11<br />

219 Di Costanzo F, Carlini P, Doni L et al. Gemcitabine with or without<br />

continuous infusion 5-FU in advanced pancreatic cancer: a randomised<br />

phase II trial of the Italian oncology group for clinical research<br />

(GOIRC). Br J Cancer 2005; 93: 185–189<br />

220 Riess M, Helm A, Niedergethmann I et al. A randomised prospective,<br />

multicentre phase III trial of gemcitabine, 5-fluorouracil (5-FU), folinic<br />

acid vs. gemcitabine alone in patients with advanced pancreatic<br />

cancer. J Clin Oncol 2005; 23: A1092<br />

221 Moore MJ, Goldstein D, Hamm J et al. Erlotinib plus gemcitabine compared<br />

with gemcitabine alone in patients with advanced pancreatic<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


cancer: a phase III trial of the National Cancer Institute of Canada<br />

Clinical Trials Group. J Clin Oncol 2007; 15: 1960–6<br />

222 Kindler HL, Friberg G, Singh DA et al. Phase II Trial of Bevacizumab<br />

Plus Gemcitabine in Patients With Advanced <strong>Pancreatic</strong> Cancer.<br />

J Clin Oncol 2005; 23: 8033–8040<br />

223 Kindler H, Niedzwiecki D, Hollis D et al. A double-blind, placebo controlled,<br />

randomized phase III trial of gemcitabine (G) plus bevacizumab<br />

(B) versus gemcitabine plus placebo (P) in patients with advanced<br />

pancreatic cancer (PC): a preliminary analysis of Cancer and<br />

Leukemia Group B (CALGB) 80303. Proceedings of the ASCO Gastrointestinal<br />

Cancers Symposium 2007; A108<br />

224 Reni M, Passoni P, Bonetto E et al. Final results of a prospective trial of<br />

a PEFG (Cisplatin, Epirubicin, 5-Fluorouracil, Gemcitabine) regimen<br />

followed by radiotherapy after curative surgery for pancreatic adenocarcinoma.<br />

Oncology 2005; 68: 239–245<br />

225 Lutz MP, Van Cutsem E, Wagener T et al. Docetaxel plus gemcitabine<br />

or docetaxel plus cisplatin in advanced pancreatic carcinoma: randomized<br />

phase II study 40984 of the European Organisation for Research<br />

and Treatment of Cancer Gastrointestinal Group. J Clin Oncol<br />

2005; 23: 9250–9256<br />

226 Shepard RC, Levy DE, Berlin JD et al. Phase II study of gemcitabine in<br />

combination with docetaxel in patients with advanced pancreatic<br />

carcinoma (E1298). A trial of the eastern cooperative oncology<br />

group. Oncology 2004; 66: 303–309<br />

227 Ridwelski K, Fahlke J, Kuhn R et al. Multicenter phase-I/II study using<br />

a combination of gemcitabine and docetaxel in metastasized and<br />

unresectable, locally advanced pancreatic carcinoma. Eur J Surg Oncol<br />

2006; 32: 297–302<br />

228 Jacobs AD, Otero H, Picozzi Jr VJ et al. Gemcitabine combined with<br />

docetaxel for the treatment of unresectable pancreatic carcinoma.<br />

Cancer Invest 2004; 22: 505–514<br />

229 Ulrich-Pur H, Raderer M, Verena Kornek G et al. Irinotecan plus raltitrexed<br />

vs raltitrexed alone in patients with gemcitabine-pretreated<br />

advanced pancreatic adenocarcinoma. Br J Cancer 2003; 88: 1180–<br />

1184<br />

230 Tsavaris N, Kosmas C, Skopelitis H et al. Second-line treatment with<br />

oxaliplatin, leucovorin and 5-fluorouracil in gemcitabine-pretreated<br />

advanced pancreatic cancer: A phase II study. Invest New Drugs<br />

2005; 23: 369–375<br />

231 Reni M, Pasetto L, Aprile G et al. Raltitrexed-eloxatin salvage chemotherapy<br />

in gemcitabine-resistant metastatic pancreatic cancer. Br J<br />

Cancer 2006; 94: 785–791<br />

232 Demols A, Peeters M, Polus M et al. Gemcitabine and oxaliplatin (GE-<br />

MOX) in gemcitabine refractory advanced pancreatic adenocarcinoma:<br />

a phase II study. Br J Cancer 2006; 94: 481–485<br />

233 Oettle H, Arnold D, Esser M et al. Paclitaxel as weekly second-line<br />

therapy in patients with advanced pancreatic carcinoma. Anticancer<br />

Drugs 2000; 11: 635–638<br />

234 Burris 3 rd HA, Rivkin S, Reynolds R et al. Phase II trial of oral rubitecan<br />

in previously treated pancreatic cancer patients. Oncologist 2005;<br />

10: 183–190<br />

235 Mitry E, Ducreux M, Ould-Kaci M et al. Oxaliplatin combined with 5-<br />

FU in second line treatment of advanced pancreatic adenocarcinoma.<br />

Results of a phase II trial. Gastroenterol Clin Biol 2006; 30:<br />

357–363<br />

236 Willett CG, Czito BG, Bendell JC et al. Locally advanced pancreatic cancer.<br />

J Clin Oncol 2005; 23: 4538–4544<br />

237 Moertel CG, Childs Jr DS, Reitemeier RJ et al. Combined 5-fluorouracil<br />

and supervoltage radiation therapy of locally unresectable gastrointestinal<br />

cancer. Lancet 1969; 2: 865–867<br />

238 Moertel CG, Frytak S, Hahn RG et al. Therapy of locally unresectable<br />

pancreatic carcinoma: a randomized comparison of high dose<br />

(6000 rads) radiation alone, moderate dose radiation (4000 rads<br />

+ 5-fluorouracil), and high dose radiation + 5-fluorouracil: The Gastrointestinal<br />

Tumor Study Group. Cancer 1981; 48: 1705–1710<br />

239 Klaassen DJ, MacIntyre JM, Catton GE et al. Treatment of locally unresectable<br />

cancer of the stomach and pancreas: a randomized comparison<br />

of 5-fluorouracil alone with radiation plus concurrent and<br />

maintenance 5-fluorouracil – an Eastern Cooperative Oncology<br />

Group study. J Clin Oncol 1985; 3: 373–378<br />

240 Cohen L, Woodruff KH, Hendrickson FR et al. Response of pancreatic<br />

cancer to local irradiation with high-energy neutrons. Cancer 1985;<br />

56: 1235–1241<br />

241 Cohen SJ, Dobelbower Jr R, Lipsitz S et al. A randomized phase III study<br />

of radiotherapy alone or with 5-fluorouracil and mitomycin-C in pa-<br />

Leitlinie 481<br />

tients with locally advanced adenocarcinoma of the pancreas: Eastern<br />

Cooperative Oncology Group study E 8282. Int J Radiat Oncol<br />

Biol Phys 2005; 62: 1345–1350<br />

242 Gastrointestinal Tumor Study Group. Treatment of locally unresectable<br />

carcinoma of the pancreas: comparison of combined-modality<br />

therapy (chemotherapy plus radiotherapy) to chemotherapy alone.<br />

J Natl Cancer Inst 1988; 80: 751–755<br />

243 Shinchi H, Takao S, Noma H et al. Length and quality of survival after<br />

external-beam radiotherapy with concurrent continuous 5-fluorouracil<br />

infusion for locally unresectable pancreatic cancer. Int J Radiat<br />

Oncol Biol Phys 2002; 53: 146–150<br />

244 McGinn CJ, Zalupski MM, Shureiqi I et al. Phase I trial of radiation dose<br />

escalation with concurrent weekly full-dose gemcitabine in patients<br />

with advanced pancreatic cancer. J Clin Oncol 2001; 19: 4202–4208<br />

245 Kachnic LA, Shaw JE, Manning MA et al. Gemcitabine following radiotherapy<br />

with concurrent 5-fluorouracil for nonmetastatic adenocarcinoma<br />

of the pancreas. Int J Cancer 2001; 96: 132–139<br />

246 Crane CH, Abbruzzese JL, Evans DB et al. Is the therapeutic index better<br />

with gemcitabine-based chemoradiation than with 5-fluorouracil-based<br />

chemoradiation in locally advanced pancreatic cancer? Int<br />

J Radiat Oncol Biol Phys 2002; 52: 1293–1302<br />

247 Rocha Lima CM, Green MR, Rotche R et al. Irinotecan plus gemcitabine<br />

results in no survival advantage compared with gemcitabine monotherapy<br />

in patients with locally advanced or metastatic pancreatic<br />

cancer despite increased tumor response rate. J Clin Oncol 2004;<br />

22: 3776–3783<br />

248 Chauffert B, Mornex F, Bonnetain F et al. Phase III trial comparing initial<br />

chemoradiotherapy (intermittent cisplatin and infusional 5-FU)<br />

followed by gemcitabine vs. gemcitabine alone in patients with locally<br />

advanced non metastatic pancreatic cancer: A FFCD-SFRO<br />

study. J Clin Oncol 2006; 24: A4008<br />

249 Huguet F, Andre T, Hammel P et al. Impact of chemoradiotherapy after<br />

disease control with chemotherapy in locally advanced pancreatic<br />

adenocarcinoma in GERCOR Phase II and III studies. J Clin Oncol<br />

2007; 20: 326–331<br />

250 Sa Cunha A, Rault A, Laurent C et al. Surgical resection after radiochemotherapy<br />

in patients with unresectable adenocarcinoma of the<br />

pancreas. J Am Coll Surg 2005; 201: 359–365<br />

251 Fisher BJ, Perera FE, Kocha W et al. Analysis of the clinical benefit of 5fluorouracil<br />

and radiation treatment in locally advanced pancreatic<br />

cancer. Int J Radiat Oncol Biol Phys 1999; 45: 291–295<br />

252 Furuse J, Kinoshita T, Kawashima M et al. Intraoperative and conformal<br />

external-beam radiation therapy with protracted 5-fluorouracil<br />

infusion in patients with locally advanced pancreatic carcinoma.<br />

Cancer 2003; 97: 1346–1352<br />

253 Brunner TB, Grabenbauer GG, Kastl S et al. Preoperative Chemoradiation<br />

in Locally Advanced <strong>Pancreatic</strong> Carcinoma: A Phase II Study. Onkologie<br />

2000; 23: 436–442<br />

254 Ikeda M, Okada S, Tokuuye K et al. A phase I trial of weekly gemcitabine<br />

and concurrent radiotherapy in patients with locally advanced<br />

pancreatic cancer. Br J Cancer 2002; 86: 1551–1554<br />

255 Talamonti MS, Catalano PJ, Vaughn DJ et al. Eastern Cooperative Oncology<br />

Group Phase I trial of protracted venous infusion fluorouracil<br />

plus weekly gemcitabine with concurrent radiation therapy in patients<br />

with locally advanced pancreas cancer: a regimen with unexpected<br />

early toxicity. J Clin Oncol 2000; 18: 3384–3389<br />

256 Brunner TB, Grabenbauer GG, Klein P et al. Phase I trial of strictly<br />

time-scheduled gemcitabine and cisplatin with concurrent radiotherapy<br />

in patients with locally advanced pancreatic cancer. Int J Radiat<br />

Oncol Biol Phys 2003; 55: 144–153<br />

257 Micke O, Hesselmann S, Bruns F et al. Results and follow-up of locally<br />

advanced cancer of the exocrine pancreas treated with radiochemotherapy.<br />

Anticancer Res 2005; 25: 1523–1530<br />

258 Xiong HQ, Rosenberg A, LoBuglio A et al. Cetuximab, a monoclonal antibody<br />

targeting the epidermal growth factor receptor, in combination<br />

with gemcitabine for advanced pancreatic cancer: a multicenter<br />

phase II Trial. J Clin Oncol 2004; 22: 2610–2616<br />

259 Bramhall SR, Rosemurgy A, Brown PD et al. Marimastat as first-line<br />

therapy for patients with unresectable pancreatic cancer: a randomized<br />

trial. J Clin Oncol 2001; 19: 3447–3455<br />

260 Moore MJ, Hamm J, Dancey J et al. Comparison of gemcitabine versus<br />

the matrix metalloproteinase inhibitor BAY 12 – 9566 in patients<br />

with advanced or metastatic adenocarcinoma of the pancreas: a<br />

phase III trial of the National Cancer Institute of Canada Clinical<br />

Trials Group. J Clin Oncol 2003; 21: 3296–3302<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.


482<br />

Leitlinie<br />

261 Beger HG, Link KH, Gansauge F. Adjuvant regional chemotherapy in<br />

advanced pancreatic cancer: results of a prospective study. Hepatogastroenterology<br />

1998; 45: 638–643<br />

262 Ghaneh P, Neoptolemos JP. Conclusions from the European Study<br />

Group for <strong>Pancreatic</strong> Cancer adjuvant trial of chemoradiotherapy<br />

and chemotherapy for pancreatic cancer. Surg Oncol Clin N Am<br />

2004; 13: 567–587, vii-viii<br />

263 Lohr M, Hoffmeyer A, Kroger J et al. Microencapsulated cell-mediated<br />

treatment of inoperable pancreatic carcinoma. Lancet 2001; 357:<br />

1591–1592<br />

264 Mercadante S. Pain treatment and outcomes for patients with advanced<br />

cancer who receive follow-up care at home. Cancer 1999;<br />

85: 1849–1858<br />

265 Zech DF, Grond S, Lynch J et al. Validation of World Health Organization<br />

<strong>Guideline</strong>s for cancer pain relief: a 10-year prospective study.<br />

Pain 1995; 63: 65–76<br />

266 Marinangeli F, Ciccozzi A, Leonardis M et al. Use of strong opioids in<br />

advanced cancer pain: a randomized trial. J Pain Symptom Manage<br />

2004; 27: 409–416<br />

267 Grahm AL, Andren-Sandberg A. Prospective evaluation of pain in exocrine<br />

pancreatic cancer. Digestion 1997; 58: 542–549<br />

268 Caraceni A, Portenoy RK. Pain management in patients with pancreatic<br />

carcinoma. Cancer 1996; 78: 639–653<br />

269 Ross GJ, Kessler HB, Clair MR et al. Sonographically guided paracentesis<br />

for palliation of symptomatic malignant ascites. Am J Roentgenol<br />

1989; 153: 1309–1311<br />

270 McNicol E, Strassels S, Goudas L et al. Nonsteroidal anti-inflammatory<br />

drugs, alone or combined with opioids, for cancer pain: a systematic<br />

review. J Clin Oncol 2004; 22: 1975–1992<br />

271 Carr DB, Goudas LC, Balk EM et al. Evidence report on the treatment of<br />

pain in cancer patients. J Natl Cancer Inst Monogr 2004; 32: 23–31<br />

272 Payne R, Mathias SD, Pasta DJ et al. Quality of life and cancer pain:<br />

satisfaction and side effects with transdermal fentanyl versus oral<br />

morphine. J Clin Oncol 1998; 16: 1588–1593<br />

273 De Conno F, Ripamonti C, Saita L et al. Role of rectal route in treating<br />

cancer pain: a randomized crossover clinical trial of oral versus rectal<br />

morphine administration in opioid-naive cancer patients with<br />

pain. J Clin Oncol 1995; 13: 1004–1008<br />

274 Cherny NI. The management of cancer pain. CA Cancer J Clin 2000;<br />

50: 70–116; quiz 117 – 120<br />

275 Gilmer-Hill HS, Boggan JE, Smith KA et al. Intrathecal morphine delivered<br />

via subcutaneous pump for intractable pain in pancreatic cancer.<br />

Surg Neurol 1999; 51: 6–11<br />

276 Staats PS, Hekmat H, Sauter P et al. The effects of alcohol celiac plexus<br />

block, pain, and mood on longevity in patients with unresectable<br />

pancreatic cancer: a double-blind, randomized, placebo-controlled<br />

study. Pain Med 2001; 2: 28–34<br />

277 Eisenberg E, Carr DB, Chalmers TC. Neurolytic celiac plexus block for<br />

treatment of cancer pain: a meta-analysis. Anesth Analg 1995; 80:<br />

290–295<br />

278 Wong GY, Schroeder DR, Carns PE et al. Effect of neurolytic celiac<br />

plexus block on pain relief, quality of life, and survival in patients<br />

with unresectable pancreatic cancer: a randomized controlled trial.<br />

Jama 2004; 291: 1092–1099<br />

279 Stefaniak T, Basinski A, Vingerhoets A et al. A comparison of two invasive<br />

techniques in the management of intractable pain due to inoperable<br />

pancreatic cancer: neurolytic celiac plexus block and videothoracoscopic<br />

splanchnicectomy. Eur J Surg Oncol 2005; 31: 768–<br />

773<br />

280 Arends J, Bodoky G, Bozzetti F et al. ESPEN <strong>Guideline</strong>s on Enteral Nutrition:<br />

Non-surgical oncology. Clin Nutr 2006; 25: 245–259<br />

Adler G et al. <strong>S3</strong>-<strong>Guideline</strong> <strong>“Exocrine</strong> <strong>Pancreatic</strong>… Z Gastroenterol 2008; 46: 449–482<br />

281 Arends J, Zuercher G, Fietkau R et al. DGEM Leitlinie Enterale Ernährung:<br />

Onkologie. Akt Ernähr Med 2003; 28: 61–68<br />

282 Bjelakovic G, Nikolova D, Gluud LL et al. Mortality in randomized<br />

trials of antioxidant supplements for primary and secondary prevention:<br />

systematic review and meta-analysis. Jama 2007; 297: 842–<br />

857<br />

283 Wigmore SJ, Falconer JS, Plester CE et al. Ibuprofen reduces energy expenditure<br />

and acute-phase protein production compared with placebo<br />

in pancreatic cancer patients. Br J Cancer 1995; 72: 185–188<br />

284 Gordon JN, Trebble TM, Ellis RD et al. Thalidomide in the treatment of<br />

cancer cachexia: a randomised placebo controlled trial. Gut 2005;<br />

54: 540–545<br />

285 Jatoi A, Windschitl HE, Loprinzi CL et al. Dronabinol versus megestrol<br />

acetate versus combination therapy for cancer-associated anorexia:<br />

a North Central Cancer Treatment Group study. J Clin Oncol 2002;<br />

20: 567–573<br />

286 Loprinzi CL, Kugler JW, Sloan JA et al. Randomized comparison of megestrol<br />

acetate versus dexamethasone versus fluoxymesterone for<br />

the treatment of cancer anorexia/cachexia. J Clin Oncol 1999; 17:<br />

3299–3306<br />

287 Moss AC, Morris E, Mac Mathuna P. Palliative biliary stents for obstructing<br />

pancreatic carcinoma. Cochrane Database Syst Rev<br />

CD 004200 2006<br />

288 Hausegger KA, Thurnher S, Bodendorfer G et al. Treatment of malignant<br />

biliary obstruction with polyurethane-covered Wallstents. Am<br />

J Roentgenol 1998; 170: 403–408<br />

289 Isayama H, Komatsu Y, Tsujino T et al. A prospective randomised<br />

study of “covered” versus “uncovered” diamond stents for the management<br />

of distal malignant biliary obstruction. Gut 2004; 53: 729–<br />

734<br />

290 Speer AG, Cotton PB, Russell RC et al. Randomised trial of endoscopic<br />

versus percutaneous stent insertion in malignant obstructive jaundice.<br />

Lancet 1987; 2: 57–62<br />

291 Urbach DR, Bell CM, Swanstrom LL et al. Cohort study of surgical bypass<br />

to the gallbladder or bile duct for the palliation of jaundice due<br />

to pancreatic cancer. Ann Surg 2003; 237: 86–93<br />

292 DiFronzo LA, Egrari S, O’Connell TX. Choledochoduodenostomy for<br />

palliation in unresectable pancreatic cancer. Arch Surg 1998; 133:<br />

820–825<br />

293 Aranha GV, Prinz RA, Greenlee HB. Biliary enteric bypass for benign<br />

and malignant disease. Am Surg 1987; 53: 403–406<br />

294 Song HY, Shin JH, Yoon CJ et al. A dual expandable nitinol stent: experience<br />

in 102 patients with malignant gastroduodenal strictures.<br />

J Vasc Interv Radiol 2004; 15: 1443–1449<br />

295 Kaw M, Singh S, Gagneja H et al. Role of self-expandable metal stents<br />

in the palliation of malignant duodenal obstruction. Surg Endosc<br />

2003; 17: 646–650<br />

296 Smith TJ, Khatcheressian J, Lyman GH et al. 2006 update of recommendations<br />

for the use of white blood cell growth factors: an evidence-based<br />

clinical practice guideline. J Clin Oncol 2006; 24:<br />

3187–3205<br />

297 Rizzo JD, Lichtin AE, Woolf SH et al. Use of epoetin in patients with<br />

cancer: evidence-based clinical practice guidelines of the American<br />

Society of Clinical Oncology and the American Society of Hematology.<br />

J Clin Oncol 2002; 20: 4083–4107<br />

298 Bokemeyer C, Aapro MS, Courdi A et al. EORTC guidelines for the use<br />

of erythropoietic proteins in anaemic patients with cancer: 2006 update.<br />

Eur J Cancer 2007; 43: 258–270<br />

299 Schuchter LM, Hensley ML, Meropol NJ et al. 2002 update of recommendations<br />

for the use of chemotherapy and radiotherapy protectants:<br />

clinical practice guidelines of the American Society of Clinical<br />

Oncology. J Clin Oncol 2002; 20: 2895–2903<br />

Downloaded by: Thieme Verlagsgruppe. Copyrighted material.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!