Zentralbl Chir 2016; 141(04): 415-420
DOI: 10.1055/s-0033-1350857
Übersicht
Georg Thieme Verlag KG Stuttgart · New York

Chirurgische Therapie der peritonealen Metastasierung in Abhängigkeit von Tumorentität, -stadium und -charakteristik(a)

Surgical Management of Peritoneal Surface Malignancy with Respect to Tumour Type, Tumour Stage and Individual Tumour Biology
S. Beckert
1   Klinik für Allgemeine, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Deutschland
,
F. Struller
1   Klinik für Allgemeine, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Deutschland
,
E.-M. Grischke
2   Universitäts-Frauenklinik, Universitätsklinikum Tübingen, Deutschland
,
J. Glatzle
1   Klinik für Allgemeine, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Deutschland
,
D. Zieker
1   Klinik für Allgemeine, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Deutschland
,
A. Königsrainer
1   Klinik für Allgemeine, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Deutschland
,
I. Königsrainer
1   Klinik für Allgemeine, Viszeral- und Transplantationschirurgie, Universitätsklinikum Tübingen, Deutschland
› Author Affiliations
Further Information

Publication History

Publication Date:
15 November 2013 (online)

Zusammenfassung

Eine Metastasierung in das viszerale oder parietale Peritoneum (Peritonealkarzinose) wird nach wie vor als terminales Krankheitsstadium angesehen. Stimuliert durch die Pionierarbeit von Prof. Dr. Paul Sugarbaker wurde das Konzept der Peritonektomie in Form einer maximalen Zytoreduktion (ZRS) mit intraoperativer hyperthermer Chemotherapie (HIPEC) entwickelt, wodurch sich die Überlebenszeit, verglichen mit alleiniger systemischer Chemotherapie, bei selektionierten Patienten nahezu verdoppeln ließ. Diese Therapieform hat sich in spezialisierten Zentren mit vertretbarer Mortalität und Morbidität etabliert, wobei die größte Herausforderung in einer adäquaten Patientenselektion liegt. Grundsätzlich ist die Peritonektomie und HIPEC sowohl bei primären peritonealen Tumoren wie dem Mesotheliom und dem Pseudomyxom sowie bei einem metastatischen Befall des Peritoneums durch gastrointestinale Tumoren oder das Ovarialkarzinom angezeigt. Die Rationale für diese Therapie liegt in der fehlenden systemischen Metastasierung, sodass der Befall des „Kompartments Abdomen“ als lokale Metastasierung verstanden werden kann. Ein Überlebensvorteil ergibt sich jedoch nur, wenn intraoperativ eine komplette (CC-0 oder CC-1) Zytoreduktion zu erzielen ist. Als Selektionskriterien gelten histopathologische Parameter, der Peritonealkarzinoseindex nach Sugarbaker (PCI) und der Allgemeinzustand des Patienten. Während bei den primär vom Peritoneum ausgehenden Tumoren die individuelle Tumorbiologie entscheidendes Prognose- und damit auch Selektionskriterium für die Peritonektomie und HIPEC ist, steht bei den gastrointestinalen Tumoren das Ausmaß des intraabdominellen Tumorbefalls im Vordergrund. Beim Magenkarzinom ist aufgrund des aggressiveren Wachstumsverhaltens die Peritonektomie und HIPEC nur bei synchroner und lokal begrenzter Peritonealkarzinose sinnvoll. Bei positiver Spülzytologie ohne makroskopischen Nachweis einer Peritonealkarzinose bleibt nach Ansprechen auf eine „neoadjuvante“ Chemotherapie durch eine Gastrektomie und HIPEC eine gewisse Option auf Kuration bestehen. Bisher hat die Therapieform der Peritonektomie und HIPEC leider noch keinen generellen Eingang in die jeweiligen Leitlinien gefunden. Lediglich in die aktuelle S3-Leitlinie kolorektales Karzinom ist sie als “Kann-Option“ aufgenommen worden.

Abstract

Peritoneal tumour dissemination is still considered as a terminal disease. For the last two decades, cytoreductive surgery (CRS) combined with intraoperative hyperthermic chemotherapy (HIPEC) has been popularised by Paul Sugarbaker almost doubling survival in selected patients compared with systemic chemotherapy alone. Nowadays, this particular treatment protocol is available in comprehensive cancer centres with reasonable mortality and morbidity. However, patient selection is still challenging. In general, CRS and HIPEC is indicated in primary peritoneal tumours such as mesothelioma and pseudomyxoma peritonei as well as in peritoneal metastases derived from gastrointestinal malignancies and ovarian cancers. Since systemic tumour spread is uncommon in patients with peritoneal metastases, peritoneal tumour dissemination was defined as localised disease within the „compartment abdomen“. However, CRS and HIPEC are only beneficial as long as complete cytoreduction is achieved (CC-0 or CC-1). Histopathological parameters, the Sugarbaker peritoneal carcinomatosis index (PCI) and general condition of the patient have been established as patient selection criteria. In primary peritoneal cancers, individual tumour biology is the predominant criterium for patient selection as opposed to intraabdominal tumour load in peritoneal metastases derived from gastrointestinal cancers. In gastric cancer, CRS and HIPEC should be restricted to synchronous limited disease because of its biological aggressiveness. In patients with free floating cancer cells without macroscopic signs of peritoneal spread, however, CRS and HIPEC following preoperative „neoadjuvant“ chemotherapy preserves chances for cure. So far, there is no general recommendation for CRS and HIPEC by clinical practice guidelines. In the recent S3 guideline for treatment of colorectal cancer, however, CRS and HIPEC have been included as possible treatment options.

 
  • Literatur

  • 1 Sugarbaker PH. Parietal peritonectomy. Ann Surg Oncol 2012; 19: 1250
  • 2 Sugarbaker PH. Peritonectomy procedures. Ann Surg 1995; 221: 29-42
  • 3 Esquivel J. Current status of colorectal cancer with peritoneal carcinomatosis. Ann Surg Oncol 2010; 17: 1968-1969
  • 4 Leitlinienprogramm Onkologie (Deutsche Krebsgesellschaft, Deutsche Krebshilfe, AWMF). S3-Leitlinie Kolorektales Karzinom, Langversion 1.0, AWMF Registrierungsnummer: 021-007OL. Im Internet: http://leitlinienprogramm-onkologie.de/Leitlinien.7.0.html Stand: 14.6.2013
  • 5 Stephens AD, Alderman R, Chang D et al. Morbidity and mortality analysis of 200 treatments with cytoreductive surgery and hyperthermic intraoperative intraperitoneal chemotherapy using the coliseum technique. Ann Surg Oncol 1999; 6: 790-796
  • 6 Piso P, Glockzin G, von Breitenbuch P et al. Quality of life after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal surface malignancies. J Surg Oncol 2009; 100: 317-320
  • 7 Elias D, Gilly F, Boutitie F et al. Peritoneal colorectal carcinomatosis treated with surgery and perioperative intraperitoneal chemotherapy: retrospective analysis of 523 patients from a multicentric French study. J Clin Oncol 2010; 28: 63-68
  • 8 Carr NJ, Sobin LH. Tumors of the Appendix. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, eds. WHO Classification of Tumors of the digestive System. 4th ed. Lyon: IARC Press; 2010: 122-125
  • 9 Elias D, Honore C, Ciuchendea R et al. Peritoneal pseudomyxoma: results of a systematic policy of complete cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Br J Surg 2008; 95: 1164-1171
  • 10 Bradley RF, Stewart JH, Russell GB et al. Pseudomyxoma peritonei of appendiceal origin: a clinicopathologic analysis of 101 patients uniformly treated at a single institution, with literature review. Am J Surg Pathol 2006; 30: 551-559
  • 11 Sugarbaker PH. Epithelial appendiceal neoplasms. Cancer J 2009; 15: 225-235
  • 12 Smeenk RM, Verwaal VJ, Antonini N et al. Survival analysis of pseudomyxoma peritonei patients treated by cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg 2007; 245: 104-109
  • 13 Cerruto CA, Brun EA, Chang D et al. Prognostic significance of histomorphologic parameters in diffuse malignant peritoneal mesothelioma. Arch Pathol Lab Med 2006; 130: 1654-1661
  • 14 Sugarbaker PH, Acherman YI, Gonzalez-Moreno S et al. Diagnosis and treatment of peritoneal mesothelioma: The Washington Cancer Institute experience. Semin Oncol 2002; 29: 51-61
  • 15 Yan TD, Deraco M, Baratti D et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for malignant peritoneal mesothelioma: multi-institutional experience. J Clin Oncol 2009; 27: 6237-6242
  • 16 Prat J. Ovarian carcinomas: five distinct diseases with different origins, genetic alterations, and clinicopathological features. Virchows Arch 2012; 460: 237-249
  • 17 Königsrainer I, Beckert S, Becker S et al. Cytoreductive surgery and HIPEC in peritoneal recurrent ovarian cancer: experience and lessons learned. Langenbecks Arch Surg 2011; 396: 1077-1081
  • 18 Trimble EL, Christian MC. Intraperitoneal chemotherapy for women with advanced epithelial ovarian carcinoma. Gynecol Oncol 2006; 100: 3-4
  • 19 Scambia G. Surgery Plus Hyperthermic Intra-peritoneal Chemotherapy (HIPEC) Versus Surgery Alone in Patients With Platinum-sensitive First Recurrence of Ovarian Cancer: a Prospective Randomized Multicenter Trial. NCT01539785. Im Internet: http://clinicaltrials.gov/ Stand: 8.9.2013
  • 20 Park SY. The Phase II Study of Intraoperative Hyperthermic Intraperitoneal Chemotherapy Followed by Intravenous Chemotherapy in Patients With Ovarian Cancer. NCT01091636. Im Internet: http://clinicaltrials.gov/ Stand: 8.9.2013
  • 21 Fagotti A. Surgery and Intraperitoneal Hyperthermic Chemotherapy in Patients With Platinum Sensitive Recurrent Ovarian Cancer. NCT01588964. Im Internet: http://clinicaltrials.gov/ Stand: 8.9.2013
  • 22 Ansaloni L. Stage IIIC Unresectable Epithelial Ovarian/Tubal Cancer With Partial or Complete Response After 1st Line Neoadjuvant Chemotherapy (3 Cycles CBDCA+Paclitaxel): a Phase 3 Prospective Randomized Study Comparing Cytoreductive Surgery + Hyperthermic Intraperitoneal Chemotherapy (CDDP+Paclitaxel) + 3 Cycles CBDCA+Paclitaxel vs. Cytoreductive Surgery Alone + 3 Cycles CBDCA+Paclitaxel. NCT01628380. Im Internet: http://clinicaltrials.gov/ Stand: 8.9.2013
  • 23 van Driel W. Phase III Randomised Clinical Trial for Stage III Ovarian Carcinoma Randomising Between Secondary Debulking Surgery With or Without Hyperthermic Intraperitoneal Chemotherapy. NCT00426257. Im Internet: http://clinicaltrials.gov/ Stand: 8.9.2013
  • 24 Chi D. A Phase II Randomized Study: Outcomes After Secondary Cytoreductive Surgery With or Without Carboplatin Hyperthermic Intraperitoneal Chemotherapy (HIPEC) Followed by Systemic Combination Chemotherapy for Recurrent Platinum-Sensitive Ovarian, Fallopian Tube, or Primary Peritoneal Cancer. NCT01767675. Im Internet: http://clinicaltrials.gov/ Stand: 8.9.2013
  • 25 Sakata Y, Ohtsu A, Horikoshi N et al. Late phase II study of novel oral fluoropyrimidine anticancer drug S-1 (1 M tegafur-0.4 M gimestat-1 M otastat potassium) in advanced gastric cancer patients. Eur J Cancer 1998; 34: 1715-1720
  • 26 Koizumi W, Narahara H, Hara T et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol 2008; 9: 215-221
  • 27 Yonemura Y, Elnemr A, Endou Y et al. Multidisciplinary therapy for treatment of patients with peritoneal carcinomatosis from gastric cancer. World J Gastrointest Oncol 2010; 2: 85-97
  • 28 Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res 1996; 82: 359-374
  • 29 Pfannenberg C, Königsrainer I, Aschoff P et al. (18)F-FDG-PET/CT to select patients with peritoneal carcinomatosis for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg Oncol 2009; 16: 1295-1303
  • 30 Valle M, Federici O, Garofalo A. Patient selection for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy, and role of laparoscopy in diagnosis, staging, and treatment. Surg Oncol Clin N Am 2012; 21: 515-531
  • 31 Iversen LH, Rasmussen PC, Laurberg S. Value of laparoscopy before cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis. Br J Surg 2012; 100: 285-292
  • 32 da Silva RG, Sugarbaker PH. Analysis of prognostic factors in seventy patients having a complete cytoreduction plus perioperative intraperitoneal chemotherapy for carcinomatosis from colorectal cancer. J Am Coll Surg 2006; 203: 878-886
  • 33 Glehen O, Kwiatkowski F, Sugarbaker PH et al. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multi-institutional study. J Clin Oncol 2004; 22: 3284-3292
  • 34 Stubs P, Habermann P, Wex C et al. [Palliative chemotherapy for colorectal cancer–current state, significance, trends]. Zentralbl Chir 2010; 135: 535-540
  • 35 Glockzin G, Rochon J, Arnold D et al. A prospective multicenter phase II study evaluating multimodality treatment of patients with peritoneal carcinomatosis arising from appendiceal and colorectal cancer: the COMBATAC trial. BMC Cancer 2013; 13: 67
  • 36 Bristow RE, Tomacruz RS, Armstrong DK et al. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J Clin Oncol 2002; 20: 1248-1259
  • 37 Winter 3rd WE, Maxwell GL, Tian C et al. Tumor residual after surgical cytoreduction in prediction of clinical outcome in stage IV epithelial ovarian cancer: a Gynecologic Oncology Group Study. J Clin Oncol 2008; 26: 83-89
  • 38 Chi DS, Zivanovic O, Palayekar MJ et al. A contemporary analysis of the ability of preoperative serum CA-125 to predict primary cytoreductive outcome in patients with advanced ovarian, tubal and peritoneal carcinoma. Gynecol Oncol 2009; 112: 6-10
  • 39 Wimberger P, Wehling M, Lehmann N et al. Influence of residual tumor on outcome in ovarian cancer patients with FIGO stage IV disease: an exploratory analysis of the AGO-OVAR (Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group). Ann Surg Oncol 2010; 17: 1642-1648
  • 40 du Bois A, Reuss A, Pujade-Lauraine E et al. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: a combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe dʼInvestigateurs Nationaux Pour les Etudes des Cancers de lʼOvaire (GINECO). Cancer 2009; 115: 1234-1244
  • 41 Eisenkop SM, Spirtos NM. Procedures required to accomplish complete cytoreduction of ovarian cancer: is there a correlation with „biological aggressiveness“ and survival?. Gynecol Oncol 2001; 82: 435-441
  • 42 Crawford SC, Vasey PA, Paul J et al. Does aggressive surgery only benefit patients with less advanced ovarian cancer? Results from an international comparison within the SCOTROC-1 Trial. J Clin Oncol 2005; 23: 8802-8811
  • 43 Yamamura Y, Ito S, Mochizuki Y et al. Distribution of free cancer cells in the abdominal cavity suggests limitations of bursectomy as an essential component of radical surgery for gastric carcinoma. Gastric Cancer 2007; 10: 24-28
  • 44 Yonemura Y, Endou Y, Sasaki T et al. Surgical treatment for peritoneal carcinomatosis from gastric cancer. Eur J Surg Oncol 2010; 36: 1131-1138
  • 45 Verwaal VJ, Boot H, Aleman BM et al. Recurrences after peritoneal carcinomatosis of colorectal origin treated by cytoreduction and hyperthermic intraperitoneal chemotherapy: location, treatment, and outcome. Ann Surg Oncol 2004; 11: 375-379
  • 46 Königsrainer I, Horvath P, Struller F et al. Risk factors for recurrence following complete cytoreductive surgery and HIPEC in colorectal cancer-derived peritoneal surface malignancies. Langenbecks Arch Surg 2013; 398: 745-749
  • 47 Weber T, Link KH. [Multimodal therapy for colon cancer: state of the art]. Zentralbl Chir 2011; 136: 325-333
  • 48 Pestieau SR, Sugarbaker PH. Treatment of primary colon cancer with peritoneal carcinomatosis: comparison of concomitant vs. delayed management. Dis Colon Rectum 2000; 43: 1341-1346 discussion 1347–1348
  • 49 Gaedcke J, Liersch T, Hess C et al. [Rectal cancer: current status of multimodal therapy–when and how?]. Zentralbl Chir 2011; 136: 334-342
  • 50 Meyer HJ, Wilke H. [Gastric cancer: current status of multimodality treatment]. Zentralbl Chir 2011; 136: 317-324
  • 51 Yano M, Shiozaki H, Inoue M et al. Neoadjuvant chemotherapy followed by salvage surgery: effect on survival of patients with primary noncurative gastric cancer. World J Surg 2002; 26: 1155-1159
  • 52 Kochi M, Fujii M, Kanamori N et al. Neoadjuvant chemotherapy with S-1 and CDDP in advanced gastric cancer. J Cancer Res Clin Oncol 2006; 132: 781-785
  • 53 Yonemura Y, Shinbo M, Hagiwara A et al. Treatment for potentially curable gastric cancer patients with intraperitoneal free cancer cells. Gastroenterological Surgery 2008; 31: 802-812
  • 54 Bakrin N, Cotte E, Golfier F et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) for persistent and recurrent advanced ovarian carcinoma: a multicenter, prospective study of 246 patients. Ann Surg Oncol 2012; 19: 4052-4058
  • 55 Yang XJ, Huang CQ, Suo T et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves survival of patients with peritoneal carcinomatosis from gastric cancer: final results of a phase III randomized clinical trial. Ann Surg Oncol 2011; 18: 1575-1581
  • 56 Chua TC, Esquivel J, Pelz JO et al. Summary of current therapeutic options for peritoneal metastases from colorectal cancer. J Surg Oncol 2013; 107: 566-573
  • 57 Chua TC, Moran BJ, Sugarbaker PH et al. Early- and long-term outcome data of patients with pseudomyxoma peritonei from appendiceal origin treated by a strategy of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. J Clin Oncol 2012; 30: 2449-2456