uva-dare (digital academic repository) advanced colorectal ... · in patients with peritoneal...

287
UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl) UvA-DARE (Digital Academic Repository) Advanced colorectal cancer: Exploring treatment boundaries Hompes, D.N.M. Link to publication Citation for published version (APA): Hompes, D. N. M. (2013). Advanced colorectal cancer: Exploring treatment boundaries. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. Download date: 26 Jun 2020

Upload: others

Post on 18-Jun-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Advanced colorectal cancer: Exploring treatment boundaries

Hompes, D.N.M.

Link to publication

Citation for published version (APA):Hompes, D. N. M. (2013). Advanced colorectal cancer: Exploring treatment boundaries.

General rightsIt is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s),other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, statingyour reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Askthe Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam,The Netherlands. You will be contacted as soon as possible.

Download date: 26 Jun 2020

Page 2: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

Advanced

colorectal

cancer

Exploring treatment

boundaries

D. Hompes

Advanced colorectal cancer

Exploring treatment boundaries

D. H

ompes | 2013

Page 3: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne
Page 4: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

Advanced colorectal cancer: Exploring treatment boundaries

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad van doctor aan de Universiteit van Amsterdam op gezag van de Rector Magnificus

prof.dr. D.C. van den Boom ten overstaan van een door het college van promoties ingestelde

commissie, in het openbaar te verdedigen in de Agnietenkapel op dinsdag 28 mei 2013, te 14:00 uur

door

DAPHNE NELLY MARCELLA HOMPES geboren te Leuven, België

Page 5: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

Promotiecommissie: Promotor: prof.dr. E.J.T. Rutgers Co-promotores: dr. V.J. Verwaal prof.dr. A. D’Hoore Overige leden: prof.dr. E. van Cutsem prof.dr. W.A. Bemelman prof.dr. C.J.A. Punt

dr. P.J. Tanis dr. J. Verheij

dr. M.L. van Velthuysen Faculteit der Geneeskunde

Page 6: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

Contents:

I. General introduction and outline of the thesis

II. Neo-adjuvant treatment for metastatic disease of

colorectal origin

1. Unresectable peritoneal carcinomatosis from colorectal cancer: a single center experience.

Hompes D, Boot H, van Tinteren H, Verwaal V. Unresectable peritoneal carcinomatosis from colorectal cancer: a single center experience. J Surg Oncol 2011; 104: 269-273. 2. Neo-adjuvant chemotherapy for unresectable

peritoneal carcinomatosis from colorectal cancer: a prospective pilot study.

Hompes D, Aalbers A, Boot H, van Velthuysen ML, Vogel W, Verwaal V. Neo-adjuvant chemotherapy for unresectable peritoneal carcinomatosis from colorectal cancer: a prospective pilot study. Submitted to Ann Surg Oncol. 3. Review: Incidence and clinical significance of

Bevacizumab-related non-surgical and surgical serious adverse events.

Hompes D, Ruers T. Review: Incidence and clinical significance of Bevacizumab-related non-surgical and surgical serious adverse events. Eur J Surg Oncol 2011; 37: 737-746.

Page 7: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

III. Cytoreductive surgery and HIPEC for peritoneal carcinomatosis of colorectal origin 1. The Treatment of peritoneal carcinomatosis of

colorectal cancer with complete cytoreductive surgery and hyperthermic intra-peritoneal peroperative chemotherapy (HIPEC) with Oxaliplatin: a Belgian multicentre prospective phase II clinical study.

Hompes D, D'Hoore A, Van Cutsem E, Fieuws S, Ceelen W, Peeters M, Van der Speeten K, Bertrand C, Kerger J, Legendre H. The treatment of peritoneal carcinomatosis of colorectal cancer with complete cytoreductive surgery and hyperthermic intra-peritoneal peroperative chemotherapy (HIPEC) with Oxaliplatin: a Belgian multicentre prospective observational clinical study. Ann Surg Oncol 2012; 19: 2186-2194. 2. The use of Oxaliplatin or Mitomycin C in HIPEC

treatment for peritoneal carcinomatosis from colorectal cancer: a comparative study.

Hompes D D’Hoore A, Mirck B, Fieuws S, Bruijn S, Verwaal V. The use of Oxaliplatin or Mitomycin C in HIPEC treatment for peritoneal carcinomatosis from colorectal cancer: a comparative study. Submitted to Eur J Surg Oncol. 3. HIPEC in T4a colon cancer: a defendable

treatment to improve oncologic outcome? Hompes D, Tiek J, Wolthuis A, Fieuws S, Penninckx F, Van Cutsem E, D’Hoore A. HIPEC in T4a colon cancer: a defendable treatment to improve oncologic outcome? Ann Oncol 2012; 23: 3123-3129.

Page 8: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

IV. RFA in colorectal liver metastases treatment

1. Radiofrequency ablation as a treatment tool for liver metastases of colorectal origin.

Hompes D, Prevoo W, Ruers T. Radiofrequency ablation as a treatment tool for liver metastases of colorectal origin. Cancer Imaging 2011; 24: 23-30. Review. 2. Morbidity and mortality of laparoscopic vs.

open radiofrequency ablation for hepatic malignancies.

Topal B, Hompes D, Aerts R, Fieuws S, Thijs M, Penninckx F. Morbidity and mortality of laparoscopic vs. open radiofrequency ablation for hepatic malignancies. Eur J Surg Oncol 2007; 33: 603-607.

3. Laparoscopic liver resection using radiofrequency coagulation.

Hompes D, Aerts R, Penninckx F, Topal B. Laparoscopic liver resection using radiofrequency coagulation. Surg Endosc 2007; 21: 175-180.

V. General Discussion

VI. Conclusions

Acknowledgments

Curriculum vitae

Page 9: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne
Page 10: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

1

I

General introduction and outline of the thesis

Page 11: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

2

Page 12: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

3

Colorectal cancer (CRC) is the third most common cancer in men and the second most common cancer in women worldwide, with more than 1.200.000 new diagnoses every year and resulting in 600.000 deaths/year1-

3. In Europe alone, CRC is responsible for 200.000 deaths/year4. The International Agency for Research on Cancer published European cancer figures in 2006: the estimated age-standardized incidence rate per 100.000 was 55.4 for men and 34.6 for women, with estimated age-standardized mortality rates per 100.000 of 27.3 in men and 16.6 in women, respectively5. As with the majority of cancers, long-term survival is dependent on the stage of the disease at diagnosis. The average 5-year overall survival (OS) ranges from approximately 90% for stage I disease to 5% or less for stage IV (metastatic) CRC6.

The scope of this thesis mainly focuses on advanced and metastatic CRC. About 20-30% of CRC patients present with stage IV disease at diagnosis and about 50-60% of CRC patients will eventually develop metastases at some point in the course of their disease4,7,. Peritoneal carcinomatosis (PC) is reported to occur in about 12-13% of patients with CRC8-10. Often, it presents in combination with other sites of disease, such as liver and/or lung metastases10,11. In general, about 50% of CRC patients will develop colorectal liver metastases (CRLM)4,12.

Two decades ago, (palliative) systemic chemotherapy was the only available treatment option for metastatic CRC, resulting in a very poor outcome. But over the last 15 to 20 years, there has been a significant evolution in systemic treatment modalities for CRC, with more efficient systemic chemotherapy (e.g. oxaliplatin- or irinotecan-based chemotherapy) and the addition of biologicals (e.g. bevacizumab, cetuximab) to the treatment regimens13-31. Furthermore, there has been a major surgical effort to expand the indications of surgical resection for metastatic

Page 13: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

4

disease, resulting in longevity in a significant subset of patients. Also, the concept of neo-adjuvant chemotherapy has significantly increased the number of patients who are suitable for metastasectomy4,32-47. This clinical thesis aims to explore the boundaries of treatment in advanced CRC. It is subdivided in 3 parts:

In the first part, we focused on unresectable PC from colorectal origin. We retrospectively studied all patients that were diagnosed with unresectable PC from a single center (the Netherlands Cancer Institute/Antoni van Leeuwenhoek-hospital in Amsterdam [the Netherlands]) over the last 5 years. We analyzed survival data and risk factors potentially influencing prognosis. Subsequently, we performed a prospective pilot study to evaluate whether neo-adjuvant chemotherapy can increase resectability in a subgroup of patients with unresectable PC, as this is already a validated strategy for CRLM. We also reviewed literature to evaluate the incidence and clinical significance of Bevacizumab-related non-surgical and surgical serious adverse events in metastatic CRC: we explored at what morbidity and mortality cost bevacizumab can be added to the neo-adjuvant chemotherapy regimens for patients with stage IV CRC? In the second part, we focused on resectable PC from colorectal origin. Complete cytoreductive surgery (CCRS) followed by Hyperthermic Intra-Peritoneal Chemotherapy (HIPEC) has progressively become an accepted combined treatment modality for this subgroup of patients. A prospective, multicenter, observational study was performed in Belgium to evaluate the outcome of CCRS and HIPEC with Oxaliplatin in patients with resectable PC from CRC. Is this very invasive combined treatment modality worthwhile? And what is the clinical cost in morbidity and mortality that comes with it’s survival benefits?

Page 14: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

5

We then performed a retrospective analysis of prospectively gathered data from 2 HIPEC centers: the Netherlands Cancer Institute/Antoni van Leeuwenhoek-hospital in Amsterdam [the Netherlands] and the University Hospitals Gasthuisberg in Leuven [Belgium]. Morbidty and mortality, as well as survival data were compared after CCRS and HIPEC with Mitomycin C and Oxaliplatin, respectively. Which intraperitoneal agent should be prefered for HIPEC? And finally, a reflection was made on the potential role for ‘prophylactic’ HIPEC in patients with T4a colon cancer, who are at high risk of developing PC. The burden of PC in T4a tumors was assessed, as well as the risk for local intraperitoneal recurrence as the only site of metastatic disease. This is crucial to adequately estimate the potential benefits of HIPEC as an adjuvant treatment strategy in T4a colon cancer patients. In the third part, we focused on the additive value of RFA in the treatment of CRLM. Does RFA push treatment boundaries for CRLM? A review of literature assessed RFA with and without chemotherapy, RFA with and without liver resection, RFA for solitary unresectable CRLM, surgical and percutaneous imaging-guided RFA and RFA compared with chemotherapy. The reported survival and recurrence data, as well as morbidity and mortality data in these different settings were analyzed. Furthermore, the review reflects on a possible role for RFA in resectable CRLM. A retrospective analysis of RFA for liver lesions compared morbidity and mortality data after laparoscopic versus open RFA. And finally, RFA is evaluated as a coagulative tool to prepare liver resection planes: is this a useful strategy to reduce intra-operative blood loss?

Page 15: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

6

References 1. Jemal A, Bray F, Center M. Global cancer statistics. CA

Cancer J Clin 2011; 61: 69-90. 2. Ferlay J, Shin H, Forman D. Estimates of worldwide

burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010; 127: 2893-2917.

3. Jemal A, Center M, DeSantis C. Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev 2010; 19: 1893-1907.

4. Van Cutsem E, Nordlinger B, Adam R. Towards a pan-European consensus on the treatment of patients with colorectal liver metastases. Eur J Cancer 2006; 42: 2212-2221.

5. Ferlay J, Autier M, Boniol M. Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol 2007; 18: 581-592.

6. Mayer R. Gastrointestinal Tract Cancer. Harrison’s Principles of Internal Medicine 16th ed. 2005; pt.5 §77: 527-531.

7. Mentha G, Majno P, Terraz S. Treatment strategies for the management of advanced colorectal liver metastases detected synchronously with the primary tumor. Eur J Surg Oncol 2007; 33: S76-S83.

8. Folprecht G, Köhne C, Lutz M. Systemic chemotherapy in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440.

9. Jayne D, Fook S, Seow-Choen: Peritoneal carcinomatosis from colorectal cancer. Br J Surg 2002; 89: 1545-1550.

10. Chu D, Lang N, Thompson C: Peritoneal carcinomatosis in nongynecologic malignancy. A prospective study of prognostic factors. Cancer 1989; 63: 364 – 367.

11. Köhne C, Vanhoefer U, Hartung G. Clinical predictive factors. Eur J Cancer 2009; 45 suppl 1: 43-49.

12. Nordlinger B, Sorbye H, Glimelius B. Perioperative chemotherapy with FOLFOX4 and surgery versus surgery

Page 16: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

7

alone for resectable liver metastases from colorectal cancer (EORTC Intergroup trial 40983): a randomized controlled trial. Lancet 2008; 371(9617): 1007-1016.

13. Schmoll H, Van Cutsem E, Stein A. ESMO Consensus Guidelines for management of patients with colon and rectal cancer. a personalized approach to clinical decision making. Ann Oncol 2012; 23: 2479-2516.

14. Prenen H, van Cutsem E. Oncological Management of unresectable liver metastases. Dig Dis 2012; 30 Suppl 2: 137-142.

15. Malvezzi M, Arfé A, Bertuccio P. European cancer morality predictions for the year 2011. Ann Oncol 2011; 22: 947-956.

16. Wagner A, Arnold D, Grothey A. Anti-angiogenic therapy for metastatic colorectal cancer. Cochrane Database Syst Rev 2009; 8(3): CD005392.

17. Klinger M, Eipeldauer S, Gruenberger T. Bevacizumab protects against sinusoidal obstruction syndrome and does not increase response rate in neoadjuvant xelox/folfox therapy of colorectal cancer liver metastases. Eur J Surg Oncol 2009; 35: 515-520.

18. Ribero D, Wang H, Vauthey J. Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer 2007; 110: 2761-2767.

19. Saltz L, Clarke S, Diaz-Rubio E. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2008; 26: 2013-2019.

20. Kabbinavar F, Hurwitz H, Rosen O. Addition of bevacizumab to fluorouracil-based first-line treatment of metastatic colorectal cancer: pooled analysis of cohorts of older patients from two randomized clinical trials. J Clin Oncol 2009; 27: 199-205.

Page 17: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

8

21. Hochster H, Hart L, Hedrick E. Safety and efficacy of oxaliplatin and fluoropyrimidine regimens with or without bevacizumab as first-line treatment of metastatic colorectal cancer: results of the TREE Study. J Clin Oncol 2008; 26: 3523-3529.

22. Giantonio B, Catalano P, Meropol N. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol 2007; 25: 1539-1544.

23. Kabbinavar F, Schulz J, Novotny W. Addition of bevacizumab to bolus fluorouracil and leucovorin in first-line metastatic colorectal cancer: results of a randomized phase II trial. J Clin Oncol 2005; 23: 3697-3705.

24. Kabbinavar F, Hambleton J, Hurwitz H. Combined analysis of efficacy: the addition of bevacizumab to fluorouracil/leucovorin improves survival for patients with metastatic colorectal cancer. J Clin Oncol 2005; 23: 3706-3712.

25. Hurwitz H, Fehrenbacher L, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350: 2335-2342.

26. Gruenberger B, Tamandl D, Gruenberger T. Bevacizumab, capecitabine and oxaliplatin as neoadjuvant therapy for patients with potentially curable metastatic colorectal cancer. J Clin Oncol 2008; 26: 1830-1835.

27. Van Cutsem E, Rivera F, Cunningham D. Safety and efficacy of first-line bevacizumab with folfox, xelox, folfiri and fluoropyrimidines in metastatic colorectal cancer: the BEAT study. Ann Oncol 2009; 20: 1842-1847.

28. Kozloff M, Yood M, Grothey A. Clinical outcomes associated with bevacizumab-containing treatment of

Page 18: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

9

metastatic colorectal cancer: the BRiTE observational cohort study. The Oncologist 2009; 14: 862-870.

29. Blazer D, Kishi Y, Vauthey N. Pathologic response to preoperative chemotherapy: a new outcome end point after resection of hepatic colorectal metastases. J Clin Oncol 2008; 25: 5344-5351.

30. Poultsides G, Servais E, Saltz L. Outcome of primary tumor in patients with synchronous stage IV colorectal cancer receiving chemotherapy without surgery as initial treatment. J Clin Oncol 2009; 27: 3379-3384.

31. Kabbinavar F, Hurwitz H, Fehrenbacher L. Phase II

randomized trial comparing bevacizumab plus

fluorouracil (FU)/leucovorin(LV) with 5FU/LV alone in

patients with metastatic colorectal cancer. J Clin Oncol

2003; 21: 60-65.

32. Yan T, Black D, Savady R. Systematic review on the

efficacy of cytoreductive surgery combined with

perioperative intraperitoneal chemotherapy for

peritoneal carcinomatosis fran colorectal cancer. J Clin

Oncol 2006; 24: 4011-4019.

33. Verwaal V, Bruin S, Boot H. 8-year follow-up of

randomized trial: cytoreduction and hyperthermic

intraperitoneal chemotherapy versus systemic

chemotherapy in patients with peritoneal

carcinomatosis of colorectal cancer. Ann Surg Oncol

2008; 15: 2426-2432.

34. Franko J, Ibrahim Z, Gusani N. Cytoreductive surgery and

hyperthermic intraperitoneal chemoperfusion versus

systemic chemotherapy alone for colorectal peritoneal

carcinomatosis. Cancer 2010; 116: 3756-3762.

35. Elias D, Glehen O, Pocard M. A comparative study of

complete cytoreductive surgery plus intraperitoneal

chemotherapy to treat peritoneal dissemination from

Page 19: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

10

colon, rectum, small bowel and nonpseudomyxoma

appendix. Ann Surg 2010; 251: 896-901.

36. Elias D, Gilly F, Boutitie F. 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.

37. Verwaal V, van Ruth S, Witkamp A. Long-term survival of

peritoneal carcinomatosis of colorectal origin. Ann Surg

Oncol 2005; 12: 65-71.

38. Elias D, Pocard M. Treatment and prevention of

peritoneal carcinomatosis from colorectal cancer. Surg

Oncol Clin N Am 2003; 12: 543-559.

39. van Leeuwen B, Graf W, Pahlman L. Swedish experience

with peritonectomy and HIPEC in peritoneal

carcinomatosis. Ann Surg Oncol 2008; 15: 745-753.

40. Gomes da Silva R, Sugarbaker P. 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.

41. Malik H, Farid S, Al-Mukthar A. A critical appraisal of the

role of neoadjuvant chemotherapy for colorectal liver

metastases: a case-controlled study. Ann Surg Oncol

2007; 14: 3519-3526.

42. Adam R, Vibert E, Pitombo M. Induction chemotherapy

and surgery for colorectal liver metastases. Bull Cancer

2006; 93 Suppl 1: S45-49.

43. Fusai G, Davidson B. Strategies to increase the

respectability of liver metastases from colorectal cancer.

Dig Surg 2003; 20: 481-496.

Page 20: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

11

44. Ruers T, Bleichrodt R. Treatment of liver metastases, an

update on the possibilities and results. Eur J Cancer

2002; 38: 1023-1033.

45. Alberts S, Poston G. Treatment advances in liver-limited

metastatic colorectal cancer. Clin Colorectal Cancer

2011; 10: 258-265.

46. Bismuth H, Adam R, Levi F. Resection of nonresectable

liver metastases from colorectal cancer after

neoadjuvant chemotherapy. Ann Surg 1996: 224: 509-

520.

47. Adam R, Delvart V, Pascal G. Rescue surgery for

unresectable colorectal liver metastases downstaged by

chemotherapy: a model to predict long-term survival.

Ann Surg 2004; 240: 644-657.

48. Pfannenschmidt J, Egerer G, Bischof M. Surgical

intervention for pulmonary metastases. Dtsch Arztebl

Int 2012;109:645-51.

49. Petre E, Jia X, Thornton R. Treatment of Pulmonary

Colorectal Metastases by Radiofrequency Ablation. Clin

Colorectal Cancer 2012 Sep 28 [Epub ahead of print].

50. Goonerante D, Gray C, Lim M. Survival outcome in New

Zealand after resection of colorectal lung metastases.

ANZ J Surg 2012 Nov 27 [Epub ahead of print].

51. Nozawa H, Sunami E, Nakajima J. Synchronous and

metachronous lung metastases in patients with

colorectal cancer: a 20-year monocentric experience.

Exp Ther Med 2012; 3: 449-45.

Page 21: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

12

Page 22: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

13

II Neo-adjuvant systemic

treatment for metastatic disease of

colorectal origin

Page 23: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

14

Page 24: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

15

1. Unresectable peritoneal carcinomatosis from colorectal

cancer: a single center experience.

Hompes D, Boot H, van Tinteren H, Verwaal V

J Surg Oncol 2011; 104: 269-273.

II

Page 25: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

16

Page 26: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

17

Unresectable Peritoneal Carcinomatosis from Colorectal Cancer: a single center experience

D.Hompes1, H.Boot2, H. van Tinteren3, V.Verwaal1

1 Department of Gastro-intestinal Surgery, 2 Department of Digestive Oncology, 3 Department of Biostatistics, The Netherlands Cancer Institute / Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands

Introduction Peritoneal carcinomatosis (PC) is reported to occur in about 12% of patients with colorectal cancer (CRC)1. Often, it presents in combination with other sites of disease, such as liver and/or lung metastases2,3. Unresectable PC has a very poor prognosis, with a median overall survival of 5-6 months2,4-

7. Although modern chemotherapy regimens have gained effectiveness in treating systemic metastases in CRC, with a median survival of up to 23.9 months and a 5-year overall survival of 13% being reported8, no real long-term survival for advanced CRC can be reached by systemic treatment9,10. Because PC is considered locally contained disease, the concept of macroscopically Complete Cytoreductive Surgery (CCRS), followed by Hyperthermic Intra-Peritoneal Chemotherapy (HIPEC) to eradicate residual microscopic disease was developed5,11-14. In a recent report by Elias et al, this multi-modality treatment achieved a median survival of 62.7 months and a 5-year survival of 51% in a well-selected patient group8. Nevertheless, the completeness of cytoreduction remains the main factor influencing survival after this treatment modality. Therefore, patients with PC tumor loads of 6-7 abdominal regions are not eligible for this approach6,15.

The aim of this paper is to perform a revision of all patients with PC from CRC who presented at our institute over

Page 27: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

18

the last five years and were found to be uneligible for CCRS and HIPEC. It describes all patient- and tumor-related factors possibly affecting survival of inoperable PC from CRC.

Patients and methods A retrospective review of the Netherlands Cancer Institute’s database from January 2005 to December 2009 was performed. This analysis includes all patients with PC from CRC, who underwent surgical exploration (laparoscopy if possible or limited laparotomy when necessary) to assess the extent of PC and were, at that moment, judged not to be eligible for CCRS and HIPEC. The quantitative assessment of the extent of PC was performed as described by Verwaal et al16. In our institution patients with PC in 5 or less abdominal regions, with no or very limited small bowel involvement and without systemic metastases are considered eligible for CCRS + HIPEC. Thus, the criteria for “unresectability” are defined as follows: 6-7 abdominal regions affected by PC, involvement of mesentery or small bowel in the PC, presence of liver metastases, retroperitoneal lymph nodes, vascular invasion and/or neural invasion. Statistical analysis was performed using the SPSS Statistics 17.0 program. An extensive univariate analysis for overall survival (OS) was performed. Due to results of the univariate analysis and due to the small number of patients combined with the retrospective nature of this trial the performance of a multivariate analysis was not considered useful. Patient population In a time span of 5 years (January 2005 to December 2009) 315 patients with PC from CRC were screened to undergo CCRS + HIPEC. Forty-three patients were surgically evaluated and judged to be uneligible for CCRS and HIPEC based on the criteria mentioned above. The male/female ratio was 29/14, with a median age at diagnosis of the primary CRC of 56.1 years

Page 28: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

19

[range 33.0-76.0 years] and a median age at surgical exploration for PC of 57.1 years [range 34.8-76.8 years]. Thirteen patients had relevant comorbidity, mainly consisting of cardiovascular and respiratory pathology and 37.2% of patients had an ASA score of 2 or higher. Tumor characteristics [Table 1] All primary tumors were histologically proven colorectal adenocarcinomas. Over 58% of primary tumors could be resected and in 74.4% of PC cases occurred synchronously to the primary tumor. The median time span between diagnosis of the primary tumor and the surgical exploration for the assessment of the PC was 7.2 months [range 0.0-102.3 months]. TNM-stage of the primary tumor is described in Table 1. More than half of the primary tumors were located on the right side of the colon. In 37.2% of patients ascites was present at diagnosis of the primary tumor. In 25 patients tumor differentiation and cell type were clearly defined: the majority of these patients had moderately to poorly differentiatied tumors and more than half of them showed a signet and/or mucinous cell type. Unresectability [Table 2] In almost 70% of patients more 6-7 regions of the abdomen were affected and in 58.1% of cases the small bowel and/or mesentery were involved in the PC. Furthermore, systemic disease was found in 16.3% of patients. In 6 patients colorectal liver metastases were found: in 5 patients this was a peroperative finding, not known from preoperative imaging and in 1 patient preoperative imaging showed a solitary liver metastasis, but this patient needed surgery, because the construction of an ostomy was indicated. Thus, no unnecessary laparotomies were performed. In 18.6% of patients a palliative diversion or ostomy was performed at surgical exploration, in order to resolve or prevent obstruction due to PC.

Page 29: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

20

Results Median OS in this series was 6.3 months, ranging from 0.4 to 33.1 months. At the time of analysis only 6 patients were still alive. After being found ineligible for CCRS and HIPEC, 72.1% of patients received some form of palliative systemic chemotherapy [Table 3]. In the majority of cases this consisted of fluoropyrimidine-based chemotherapy combined with oxaliplatin or irinotecan, either with or without bevacizumab. For the 31 patients receiving chemotherapy median OS was 9.3 months [range 0.9-33.1 months], whereas the 12 patients who didn’t receive chemotherapy had a median overall survival of 3.1 months [range 0.4-6.5 months]. However, patient characteristics, such as ASA score, age and the occurrence of postoperative complications after surgical assessment of PC, as well as tumor characteristics (advanced tumor stage T3-4N+), seemed to be less favorable in the patient group not receiving palliative systemic treatment [Table 3]. Although this apparent difference in patient characteristics did not reach statistical significance, there was a weak but significant negative Pearson correlation between patients with an ASA score ≥ 2 and the administration of palliative chemotherapy (correlation -0.379; p=0.012). Furthermore, a strong negative Pearson correlation could be found between the occurrence of postoperative complications after surgical exploration for PC and whether or not palliative chemotherapy was administered (correlation -0.572; p<0.001) [Table 4]. Patients receiving palliative chemotherapy seemed to do statistically better in the univariate analysis for overall survival (p<0.001) [Table 5]. No other intrinsic patient and tumor characteristics involved in the univariate analysis seemed to have a clear influence on OS. Given these results, performing a multivariate analysis would not have been useful.

Page 30: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

21

Discussion Literature on the outcome of unresectable PC from CRC is fairly scarce and results differ among trials. In the era before CCRS and HIPEC a prospective study of prognostic factors in PC of non-gynecologic malignancy was performed by Chu et al2. Forty-five of the 100 patients included in the study had PC of CRC. In their analysis disease-free survival (DFS) of less than 1 year, the presence of ascites and lung metastases adversely affected survival. Tumor burden, examined by size, number and distribution of tumor nodules in the peritoneal cavity, did not affect survival2. In 2000 Sadeghi et al published the results of the EVOCAPE 1 trial, in which 370 patients with PC from non-gynecologic malignancies were followed prospectively4. Patients treated by CCRS and HIPEC were excluded from this trial, leaving only those patients undergoing palliative treatment to be analyzed. In general, patients with PC of digestive origin were found to have a very poor prognosis. One hundred-eighteen patients in this trial had PC of colorectal origin. Median OS for this group was 5.2 months [range 0.6-44.8 months]. Only the extent of PC was significantly correlated to survival. Synchronous PC, T-stage, lymph node involvement, tumor differentiation, the presence of ascites and the presence of liver metastases didn’t have a statistically significant influence on prognosis. A retrospective analysis by Jayne et al included 3019 patients with CRC: 349 (13%) had PC, of whom 214 patients (61%) had synchronous PC and 135 patients (39%) developed metachronous PC17. In the synchronous PC group 72% of patients underwent a resection of their primary tumor and a debulking of the PC, whereas only in 2% of patients in the metachronous PC group an actual debulking could be performed. In the PC group a significantly higher proportion of primary tumors had advanced disease stage at diagnosis. Furthermore, extent of PC disease and T-stage were identified as predictors of survival for synchronous PC17.

Page 31: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

22

In our retrospective analysis all of the above mentioned adverse predictive factors didn’t seem to have a statistically significant influence on outcome [Table 5]. Possibly, this is due to the small number of patients analyzed in this study. Only patients receiving palliative systemic chemotherapy seemed to have a statistically better outcome than those who didn’t. For patients with unresectable PC who didn’t undergo palliative chemotherapy a median OS of 3.1 months [range 0.4-6.5 months] was registered [Table 3], which concurs with current literature2,4-7. Those patients who could undergo palliative chemotherapy reached a median OS of 9.3 months [range 0.9-33.1 months] [Table 3]. This is comparable to the results of the randomized trial performed by Verwaal et al18, which reported a median disease-specific survival of 12.6 moths for patients with resectable and unresectable PC who were treated with systemic chemotherapy only (i.e. 5-FU/leucovorin). Under modern chemotherapy regimens median OS of 16.8 to 23.9 months have been reported for patients with PC of CRC8,19. Nevertheless, although modern 5-FU-based systemic therapy combined with oxaliplatin or irinotecan either with or without bevacizumab was administered in most patients undergoing palliative chemotherapy in our study, a survival of almost 2 years was only rarely reached. On the other hand, only patients with unresectable PC were included in this study, which implies an up front selection of more extensive disease.

To our opinion, intact bowel passage is essential for a patient to be able to receive chemotherapy. Therefore, construction of a bowel diversion or ostomy should be performed whenever indicated to resolve or prevent obstruction due to PC. Furthermore, the negative Pearson correlation between ASA score ≥ 2 as well as the occurrence of postoperative complications after surgical exploration on the one hand and the administration of palliative chemotherapy on the other hand suggests that a better general condition and performance status (PS) facilitates patients to undergo and tolerate chemotherapy and it’s side effects [Table 4]. The

Page 32: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

23

importance of PS was already stressed by Folprecht et al by retrospective analysis of a large database with 3823 patients with advanced CRC1. Also, peritoneal involvement was identified as an independent negative predictive factor. A review of literature by Köhne et al, describing clinical predictive factors for the outcome of patients with metastatic CRC under chemotherapy, came to the same conclusions3. Finally, in our series lymph node involvement was not identified as a predictive factor adversely affecting survival. This concurs with the results of the prospective EVOCAPE 1 trial4. Possibly, this suggests that the local phenomenon of PC might have a stronger adverse influence on outcome than the systemic spread of advanced CRC by nodal involvement.

Conclusions This analysis shows the influence of the administration of systemic chemotherapy on survival in patients with unresectable PC of colorectal origin. No other predictive factors could be clearly identified. Nevertheless, even in the era of modern chemotherapy regimens, the median survival of patients with unresectable PC of CRC undergoing chemotherapy is only 9.3 months. Furthermore, the fact that patients with unresectable PC from CRC undergoing palliative chemotherapy seem to have a better median OS, is probably largely attributable to the fact that this patient group has a better general condition, thus enabling them to tolerate systemic treatment.

Page 33: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

24

Table 1: primary tumor characteristics

N = 43 adenocarcinomas

Primary tumor resected

Yes

No

25

18

Chemotherapy

for primary tumor

Yes

No

22

21

PC Synchronous

Metachronous

34

9

T-stage cis

1

2

3

4

x

1

0

1

11

21

9

N-stage 0

1

2

x

6

6

15

16

Localization Right Appendix

Caecum / colon ascendens

4

22

Left Transverse colon

Colon descendens

Sigmoid

Rectum

1

1

9

3

unknown 3

Pathology Differentiation Good

Moderate

Poor

Not specified

2

13

10

18

Cell type Signet

Mucinous

Not specified

13

16

18

Preoperative CEA (µg/l)

Elevated (>6 µg/l) / not elevated

12.0 [1.0-867.0]

22/21

Page 34: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

25

Table 2: Operative findings at surgical exploration for PC Reasons for unresectability 6-7 regions affected (a)

Mesentery / small bowel involvement (b)

Both (a) + (b)

Liver metastases

Retroperitoneal N+

Vascular invasion

Neural invasion

other

30

25

19

6

1

0

1

2

Peroperative Ascites 10

Table 3: postoperative treatment + outcome after surgical exploration for PC

Median OS [range] 6.3 months [0.4-33.1 months]

OS with Chemotherapy [range] N=31

9.3 months [0.9-33.1 months]

Chemotherapy-naive

ASA ≥ 2

Median age at surgery

Postoperative complications

T3-4N+

15/31

8/31

55.3 years [38.4-71.7]

2/31

12/31

OS without chemotherapy [range] N=12

3.1 [0.4-6.5 months]

Chemotherapy-naive

ASA ≥ 2

Median age at surgery

Postoperative complications

T3-4N+

5/12

8/12

60.0 years [34.8-76.8]

7/12

7/12

OS=overall survival

Page 35: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

26

Table 4: Pearson correlations

OS (m)

palliative chemotherapy:

0=no; 1=yes

ASA ≥2:

0=no; 1=yes

Age S

<60 years

T3-4N+ relevant postoperative complications

OS (months) Pearson Correlation

1 ,484**

-,235 -,024 ,041 -,361*

Sig. (2-tailed)

,001 ,129 ,879 ,817 ,017

N 43 43 43 43 34 43

palliative chemotherapy: 0=no; 1=yes

Pearson Correlation

,484**

1 -,379

*

,197 -,265 -,572**

Sig. (2-tailed)

,001 ,012 ,205 ,130 ,000

N 43 43 43 43 34 43

ASA ≥2: 0=no; 1=yes

Pearson Correlation

-,235 -,379* 1 -,143 ,312 ,314

*

Sig. (2-tailed)

,129 ,012 ,360 ,072 ,041

N 43 43 43 43 34 43

Age S <60 years

Pearson Correlation

-,024 ,197 -,143 1 -,160 ,017

Sig. (2-tailed)

,879 ,205 ,360 ,365 ,915

N 43 43 43 43 34 43

T3-4N+ Pearson Correlation

,041 -,265 ,312 -,160 1 ,256

Sig. (2-tailed)

,817 ,130 ,072 ,365 ,144

N 34 34 34 34 34 34

relevant postoperative complications

Pearson Correlation

-,361* -,572

** ,314

* ,017 ,256 1

Sig. (2-tailed)

,017 ,000 ,041 ,915 ,144

N 43 43 43 43 34 43

**:Correlation is significant at the 0.01 level (2-tailed)

*: Correlation is significant at the 0.05 level (2-talied)

Age S = age at surgical exploration for PC

Page 36: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

27

Table 5: Univariate analysis for Overall Survival

Intrinsic patient and tumor characteristics p-value

Patient Male/Female Age at diagnosis primary tumor < or > 50 Age at surgery for PC < or > 60 Time interval primary diagnosis to surgery

< or > 24 months ASA

ASA < or ≥2

0.731

0.146 0.933

0.306

0.139 0.135

Tumor Resection primary tumor Synchronous / metachronous PC T-stage

T4 or <T4 N-stage

N+ or N0 Advanced disease: stage III-IV

0.708 0.576

0.447

0.561 0.556

Localization primary tumor Right colon

Left colon

0.990 0.996

Pathology: differentiation Good/moderate or Poor

Signet cell or non-signet cell

0.369 0.942

Surgery Chemotherapy before surgery for PC Elevated CEA (>6µg/L)

Yes or no Operative findings:

< or > 6-7 regions involved Mesentery / small bowel involved or not

Peroperative ascites present or not

0.715

0.755

0.634 0.487 0.171

Postop. chemotherapy

Yes or no <0.001

Page 37: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

28

References 1. Folprecht G, Köhne C, Lutz M. Systemic chemotherapy

in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440.

2. Chu D, Lang N, Thompson C. Peritoneal carcinomatosis in nongynecologic malignancy. A prospective study of prognostic factors. Cancer 1989; 63: 364-367.

3. Köhne C, Vanhoefer U, Hartung G. Clinical predictive factors. Eur J Cancer 2009; 45 suppl 1: 43-49.

4. Sadeghi B, Arvieux C, Gilly F. Peritoneal carcinomatosis from non-gynecologic malignancies. Results of the EVOCAPE I multicentric prospective study. Cancer 2000; 88: 358-363.

5. Elias D, Gilly F, Glehen O. Peritoneal 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.

6. Davies J, O’Neil B. Peritoneal carcinomatosis of gastrointestinal origin: natural history and treatment options. Exp Opin Invest Drugs 2009; 18: 913-919.

7. Bloemendaal A, Verwaal V, van Ruth S. Conventional surgery and systemic chemotherapy for peritoneal carcinomatosis of colorectal origin: a prospective study. Eur J Surg Oncol 2005; 31: 1145-1151.

8. Elias D, Lefevre J, Chevalier J. Complete Cytoreductive Surgery plus Intraperitoneal Chemohyperthermia with Oxaliplatin for peritoneal Carcinomatosis of Colorectal Origin. J Clin Oncol 2009; 27: 681-685.

9. Cao C, Yan T, Morris D. A systematic review and meta-analysis of cytoreductive surgery with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 2009; 16: 2152-2165.

10. De Gramont A, Figer A, Seymour M. Leucovorin and fluorouracil with or without oxaliplatin as first-line

Page 38: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

29

treatment in advanced colorectal cancer. J Clin Oncol 2000; 18: 2938-2947.

11. Glehen O, Elias D, Deraco M. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multiinstitutional study; J Clin Oncol 2004; 22: 3284-3292.

12. Witkamp A, de Bree E, Van Goethem R. Rationale and techniques of intra-operative hyperthermic intraperitoneal chemotherapy. Cancer Treat Rev 2001; 27: 365-374.

13. Sugarbaker P. A curative approach to peritoneal carcinomatosis from colorectal cancer. Semin Oncol 2005; 32: S68-73.

14. Sugarbaker P. Surgical management of carcinomatosis from colorectal cancer. Clin Colon Rectal Surg 2005; 18: 190-203.

15. Verwaal V, Bruin S, Boot H. 8-year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol 2008; 15: 2426-2432.

16. Swellengrebel H, Zoetmulder F, Verwaal V. Quantitative intra-operative assessment of peritoneal carcinomatosis - a comparison of three prognostic tools. Eur J Surg Oncol 2009; 35:1078-1084.

17. Jayne D, Fook S, Seow-Choen F. Peritoneal carcinomatosis from colorectal cancer. Br J Surg 2002; 89: 1545-1550.

18. Verwaal V, Bruin S, Boot H. 8-Year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal

Page 39: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

30

carcinomatosis of colorectal cancer. Ann Surg Oncol 2008; 15: 2426-2432.

19. Franko J, Ibrahim Z, Gusani N. Cytoreductive surgery and hyperthermic intraperitoneal chemoperfusion versus systemic chemotherapy alone for peritoneal carcinomatosis. Cancer 2010; 116: 3756-3762.

Page 40: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

31

2. Neo-adjuvant chemotherapy for unresectable peritoneal

carcinomatosis from colorectal cancer: a prospective pilot study

Hompes D, Aalbers A, Boot H, van Velthuysen ML,

Vogel W, Verwaal V Submitted to Ann Surg Oncol.

II

Page 41: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

32

Page 42: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

33

A prospective pilot study to assess neo-adjuvant chemotherapy for unresectable peritoneal

carcinomatosis from colorectal cancer

D.Hompes1, A.Aalbers1, H.Boot2, M.-L.van Velthuysen3, W.Prevoo4, W.Vogel5, V.Verwaal1

1 Department of Surgical Oncology, 2 Department of Digestive Oncology, 3 Department of Pathology, 4 Department of Radiology, 5 Department of Nuclear Medicine, Antoni van Leeuwenhoek-Hospital/the Netherlands Cancer Institute, the Netherlands

Introduction Peritoneal carcinomatosis(PC) occurs in 12-13% of patients with colorectal cancer(CRC)1-3. The majority of them present with unresectable PC, unfit for complete cytoreductive surgery(CCRS) and hyperthermic intraperitoneal chemotherapy(HIPEC).

Unresectable PC from CRC has a poor prognosis, with a median OS of 5-6 months4-9. Currently these patients are treated with systemic chemotherapy. For unresectable metastatic CRC a median overall survival(OS) of almost 2 years has been reported under modern chemotherapy regimens, with a 5-year OS of about 13%10, but without a real chance of cure11,12. Literature data on systemic therapy for PC from CRC are scarce1,4,5,13-15[Table 1]. After 5-FU/leucovorin-based chemotherapy a median OS of 5.2-12.6 months was reported13,15. It is suggested that patients with PC have a longer survival with systemic treatment, but the available data are difficult to interpret, as studies often include patients with resectable as well as unresectable PC1,4,5,13-15 and PC often occurs with other metastatic sites (e.g. liver or lung metastases)1,4,5,16. Also, the patient’s performance status and

Page 43: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

34

comorbidity at diagnosis probably influence treatment choice and outcome2,9,16. This prospective observational pilot study aims to document the actual response rate and response characteristics of neo-adjuvant modern systemic chemotherapy in patients with initially unresectable PC from CRC, by laparoscopy before and after chemotherapy.

Patients & Methods Assessment of the extent of PC This trial aims to evaluate the efficacy of neo-adjuvant chemotherapy with capecitabine, oxaliplatin and bevacizumab in patients with unresectable PC from CRC. Response to this neo-adjuvant therapy will be assessed by laparoscopic evaluation of the intra-abdominal extent of disease by the 7 region count as described by Verwaal et al.17 and, when a peritoneal biopsy is considered safe, by histopathological examination of a PC-affected region of peritoneum. Unresectable PC is defined as a tumor load of more than 5 of the 7 regions17,18 and/or extensive involvement of the small bowel by PC. Inclusion and exclusion criteria[Table S1] Patients who have initially been excluded from CCRS+HIPEC due to their extent of disease, are eligible for this trial. Patients should be fit to undergo the entire course of treatment. Patients with systemic metastasis (lung, liver, bone or other extra-abdominal sites) are excluded from this trial. Trial flowchart[Fig.S1] At Step I of the trial a blood sample is taken, with CEA and CA19.9. Then the first laparoscopy is performed, with documentation of all 7 abdominal regions with digital pictures and a biopsy of a PC-affected peritoneum [1.5cmx1.5cm] is taken and a similar peritoneum area is marked with clips. For safety reasons, regions that are inaccessible due of tumor or

Page 44: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

35

extensive adhesions will not be documented, but registered as inaccessible. These same safety considerations apply for the biopsy procedure. Before the start of the systemic chemotherapy a PET/CT is performed to search for systemic metastases. In Step II patients are administered 4 courses of systemic chemotherapy. Finally, in step III a re-evaluation of the extent of PC is performed with PET/CT or CT and a second laparoscopy, for which the same safety considerations apply as mentioned for Step I.

Chemotherapeutic agents Every 3 weeks patients receive

Capecitabine(1000mg/m² twice daily on day 1-14) and Oxaliplatin(130mg/m2 on day 1) with or without Bevacizumab(7.5mg/kg on day 1). In case of problematic oral intake 5-Fluorouracil(400mg/m2 in bolus + 600mg/m2 in infusion over 22 hours on day 1 and 2) and Oxaliplatin(85mg/m2 on day 1) with or without Bevacizumab(5mg/kg on day 1) are administered every 2 weeks.

Medical Ethics Committee[MEC] The trial protocol was revised and approved by the local MEC of the Netherlands Cancer Institute/Antoni van Leeuwenhoek-Hospital and by the National Research Authorities. All patients gave written informed consent.

Statistical considerations Primary objective is to assess the efficacy of neo-

adjuvant chemotherapy in reducing the number of regions affected from >5 regions to ≤5 regions. Because experience with the proposed chemotherapeutic regimen already exists and this regimen will also be given in case the patient is not

Page 45: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

36

operable, a one-stage design (rather than a two-stage design) is considered to be most efficient and practical. The highest response proportion which, if true, would imply that the treatment does not warrant further investigation, is set at 20%. The smallest proportion that would imply that the treatment has potential and will be investigated further is set at 40%. The hypothesis will be tested at α=0.05 and a 90% power. Applying the method of A’Hern, a sample size of 47 (evaluable) patients is needed with 15 being the minimum number of responses required for a conclusion of ‘efficacy’19.

Results Patient and tumor characteristics[Table 2] Eventually, 10 patients were included into the trial: 7

male and 3 female patients, with a median age of 60.3years[45.6-72.8years]. Median ASA score was 2[range1-3], 7 patients had an ECOG performance status(PS)20 of 0 and for 3 patients the PS was 1. Eight patients previously underwent laparotomy. In 50% of the patients the primary tumor was localized in the left colon. The pGTNM-data of the primary tumor are listed in Table 3. Forty percent of patients had a mucinous cell type. For all patients PC was already present at diagnosis of the primary tumor. In one patient both the primary tumor and the PC had been resected 28 months ago and this patients now presented with recurrent PC. The median time interval between the diagnosis of the primary tumor and inclusion into this trial was 2.1months[range 0.0-28.0months]. In half of the patients the primary tumor was still in situ at trial inclusion.

Peroperative findings before and after neo-adjuvant chemotherapy[Table 3]

All patients included had histopathological proof of PC from CRC. The median CEA-value at inclusion was 11.5µg/L[range1.0-256.0 µg/L].

Page 46: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

37

Trial Step I In 9 patients PET/CT showed PC without systemic metastases. In 7 of them laparoscopy showed 6-7 abdominal regions as well as the small bowel were affected by PC. In 2 patients the pelvis was inaccessible for assessment. For 2 patients extensive invasion of the small bowel was the main reason for unresectability. One of them had an inaccessible right hemiabdomen. In 8 patients a peritoneal biopsy could be taken, showing adenocarcinoma in 7 patients and mucus without cells in 1 patient. However, in this latter patient, the primary tumor was proven to be a moderately differentiated adenocarcinoma of the transverse colon. For logistic reasons, PET/CT could only be performed after laparoscopy in patient G. Unfortunately, liver metastases were found. Hence, this patient was excluded from the trial and started on palliative chemotherapy. Patient I developed a small bowel perforation during laparoscopy, resulting in a postoperative enterocutaneous fistula. She was also excluded from the protocol and, after recovery from this complication, started on palliative chemotherapy[5-FU/leucovorin/oxaliplatin].

Trial Step II Of the 8 patients that went on to neo-adjuvant chemotherapy, 4 received capecitabine/oxaliplatin with bevacizumab, whereas 4 received only capecitabine/oxaliplatin. Seven patients received 4 courses of chemotherapy, with dose reduction due to chemotoxicity in 6 patients. In patient H the systemic therapy was stopped after 2 courses, because of clinical disease progression with bowel obstruction. An early CT scan showed liver metastasis, enlarged mediastinal lymph nodes and progression of the PC. This patient died shortly after trial discontinuation.

Trial Step III After neo-adjuvant chemotherapy the median CEA-value was 13.0µg/L[range 2.0-156.0µg/L]. For patient B the second CT scan showed evident progression of the PC. Therefore, no

Page 47: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

38

second laparoscopy was performed. In the other 6 patients, who completed the neo-adjuvant chemotherapy, second PET/CT or CT showed stable disease in 3 and suggested slight regression of PC in the other 3. At second laparoscopy all 6 patients had PC in 6-7 abdominal regions as well as extensive small bowel involvement: PC had remained stable in 2 patients and progressed in 4 patients. The subhepatic space was inaccessible in 2 patients and the pelvis in 1 patient. For safety considerations peritoneal biopsies were only taken in 3 patients: 2 showed adenocarcinoma and 1 only showed mucus. It should be mentioned that patient I was found to have progressive PC disease at laparotomy outside of protocol, in spite of the palliative chemotherapy she had received. As an example, Fig.1 shows intra-operative laparoscopic views of the seven abdominal regions in 2 different patients before and after chemotherapy. The histopathological image of the peritoneal biopsies taken in patient A show a mucinous tumor in which cellularity almost disappeared after chemo, whereas in patient C chemotherapy barely changed the tumor’s cellularity[Fig.2].

Trial closure Because no response to chemotherapy could be demonstrated in either of the included patients, further inclusion of patients into this trial seemed unethical.

Discussion

About 12-13% of patients with CRC develop PC in the course of their disease1,4. Resectable PC can be treated with CCRS+HIPEC, resulting in 5-year OS rates of ≤50%21-29. But the majority of patients with PC present with unresectable disease4,16, which has a median OS of only 5-6 months4-9[Table 1].

Currently, systemic chemotherapy is considered the only available treatment option for these patients. Under modern chemotherapy regimens, literature reports a median OS of 23.9 months and a 5-year OS of 13% for metastatic CRC

Page 48: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

39

11,12. It is difficult to determine whether this finding for metastatic CRC in general also applies to the local phenomenon of PC.

Neo-adjuvant chemotherapy is accepted as a down-sizing technique for

initially unresectable systemic metastases from CRC, especially liver metastases30-37, with response rates as high as 50%32. Conversion to resectability occurs in 10-20%31,32,34-36, resulting in 5-year OS rates of 30-40%31. At present, literature data on the efficacy of (neo-adjuvant) systemic chemotherapy on PC are scarce and difficult to interpret. Three important remarks should be made:

First, Elias et al. performed a multicentre retrospective study on selected patients with resectable colorectal PC treated with palliative chemotherapy in order to compare their outcome with that of patients with a comparable extent of PC undergoing CCRS+HIPEC. Median survival was 23.9months in the chemotherapy-group versus 62.7months in the HIPEC-group10. Yet, this is a highly selected patient group with limited PC. Literature on systemic chemotherapy is far less optimistic, when patients with resectable and unresectable PC are combined in one trial: The randomized controlled trial by Verwaal et al. reports a disease-specific survival of 12.6months for this patient population14 and an analysis by Koppe et al. reports a median survival of 5.2-12.6months after 5-FU/LV-based chemotherapy13. For unresectable PC results are even worse. A retrospective analysis by Hompes et al. showed a median OS of 6.3months [range 0.4-33.1months] for these patients, with a median OS of 9.3months[0.9-33.1months] after chemotherapy versus 3.1months[range 0.4-6.5months] without chemotherapy9. A retrospective analysis by Pelz et al15 reported comparable findings, with median OS of 5.0months without chemotherapy versus 11.0months after 5-FU/LV and 12.0months under modern chemotherapy regimens.

Second, the patient’s general condition as well as the extent of PC at diagnosis have a significant influence on the

Page 49: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

40

choice of treatment and it’s outcome. The importance of the patient’s PS as a clinical predictive factor was shown by Folprecht et al.17 as well as Köhne et al.16. A systematic review by Stillwell et al.38 showed PS, age and ASA-score, as well as postoperative morbidity and mortality after palliative surgery are important prognostic factors for OS in patients with stage IV CRC and unresectable metastases. This coincides with the analysis by Hompes et al., which shows a significant correlation between survival and the patient’s age, ASA-score and complications after palliative surgery9. In Pelz’s study treatment differed significantly for different extents of PC15: patient’s who didn’t receive chemotherapy had a high tumor load more often. Also, patients with a low tumor load initially showed a tendency toward benefit from chemotherapy, but after 3 years there was practically no difference in survival for different extents of disease. The best results with chemotherapy were reached in patients with low tumor load and in patients with PC only (i.e. without systemic metastases) 15. There are several Indications that PC as a local phenomenon has a higher influence on OS than systemic dissemination5,9,15. The EVOCAPE1-trial showed that PC is a foremost cause of disease-specific mortality in patients with metastatic colorectal cancer5 and Pelz et al. stated that the biologically aggressive nature of PC impairs the functional status of patients to an extent that makes them only eligible for best supportive care15.

Third, the difficulty of PC assessment lies in the inability to image sub-centimetric lesions and assess tumor response on the RECIST-criteria. This leaves this subgroup of patients with stage IV CRC without any appreciable evidence of disease and treatment response cannot be accurately documented or monitored15. Thus, retrospective evaluation of PCI is difficult. Therefore, Pelz et al. used the terms “low”, “moderate” and “extensive” to describe the tumor burden, analog to the PCI15,39. Still, this remains an estimation of the extent of PC. The only reliable tool that is currently available to assess extent of PC is laparoscopy.

Page 50: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

41

This observational pilot study provides the first prospective assessment of the evolution of unresectable PC from CRC under modern systemic chemotherapy. Laparoscopy was performed to assess the extent of PC before and after combined chemotherapy with capecitabine/oxaliplatin with or without bevacizumab. All included patients had a good PS and ASA-score at PC diagnosis. One patient had to be excluded because of systemic metastases. Of the 9 patients that proceeded within the trial protocol, 2 developed early progressive disease, whereas 2 had macroscopically stable disease, 4 had progressive disease at laparoscopy and 1 patient had proven progressive disease at laparotomy after palliative chemotherapy outside of protocol. In other words, 78% of patients had progressive disease under chemotherapy and 22% had stable disease. No clear macroscopic response to chemotherapy could be demonstrated[Table 3]. Of course, the small number of patients is an important shortcoming of this trial, but we felt further inclusion of patients into the trial would be unethical, given the complete absence of response. Furthermore, despite the good PS of all patients at the start of chemotherapy, the majority of them completed their systemic chemotherapy with dose reduction, which indicates that PC has a substantial influence on the patient’s functional status. Finally, for safety reasons, 2 important concessions were done on the trial protocol: One is that all 7 abdominal regions were not always fully accessible for macroscopic assessment. Nevertheless, PC unresectability criteria were met in all cases, so this fact does not affect outcome. The other concession is that peritoneal biopsies could often not be taken. Biopsies before and after chemotherapy could only be compared in patient A and C: Patient C showed no obvious microscopic response to the chemotherapy. In patient A, who had a mucinous adenocarcinoma, cellularity almost disappeared after chemotherapy, but at laparoscopy there was even a slight macroscopic progression of PC and this patient died of his disease[Fig.2]. Whether patients with this

Page 51: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

42

cell type might benefit more from chemotherapy cannot be determined from this small trial: 4 patients had a mucinous type of PC, 3 of whom died, with a survival that ranges from 6.3 tot 16.8months. In conclusion, PC, which is a local phenomenon, does not seem to respond well to systemically administered chemotherapy. Trial inclusion was discontinued, because none of the included patients showed macroscopic response to the chemotherapy. None of the patients was the PC rendered resectable under chemotherapy.

Page 52: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

43

Table 1: median OS after systemic therapy for PC of CRC

LITERATURE PATIENT POPULATION TREATMENT OUTCOME

Author Journal

year

Type of study

N Total

N PC

from CRC

Patients with

synchronous systemic

Metastases also

included?

Surgery Chemotherapy Median OS (months) Resection Explorative

surgery + biopsy

or Palliative surgery

N patients regimen

Chu4

Cancer 1989

Prospective 100 45 yes 44* 56* “majority” 15

5-FU/LV15

6

Sadeghi5

Cancer 2000

Prospective 370 118 yes 144* 226* 97 5-FU/LV or

5-FU/oxaliplatin

5.2

Jayne1

BJS 2002

Retrospective 3019 392 yes 157 50 ns ns 7

Verwaal14

JCO 2003

Prospective 105 103 no 49 (debulking + HIPEC)

ns 77 5-FU/LV (or irinotecan if 5-FU/LV was given

within 1 year before inclusion)

12.6**

N = number, PC = peritoneal carcinomatosis, CRC = colorectal cancer, ns = not specified *not specified how many of these patients had PC from CRC ** survival in the standard arm (systemic therapy +/- palliative surgical procedure

Page 53: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

44

Table 2: patient and tumor characteristics

N=10 patients

Male/Female 7/3

Median age at inclusion 60.3 years [range 45.6-72.8 years]

Patient’s general condition ASA-score 1 2 3

4 3 3

ECOG PS 0 1

7 3

Primary tumor Localization Appendix Right colon

Left colon Rectosigmoid

Rectum

4 1 1 3 1

G-stage G2 Gx

2 8

T-stage T3 T4 Tx

1 4 5

N-stage N1 Nx

3 7

Cell type Mucinous type Signet cells

4 1

In situ at inclusion 5

Peritoneal Carcinomatosis (PC) Synchronous 10

Previous PC?* 1

Interval between diagnosis of PC and inclusion 2.1 months [range 0-28.0 months]

*one patient had a medical history of primary CRC and PC, which was resected earlier: this patient was now included into this trial with recurrent PC.

Page 54: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

45

Table 3: peroperative findings before and after neo-adjuvant chemotherapy

Patient Age (years)

Male (M) Or

Female (F)

Reason for unresectability (a=PC in 6-7/7 abdominal regions, b=extensive small

bowel invasion)

Neo-adj. chemo

Response on

PET/CT or CT?

Response at Laparoscopy?

Stable disease (SD) or

progressive disease

(PD)

Trial STOP

Cancer-related Death

A 68.7 M a,b Capox/BV 1# 0 PD No 1

B 52.9 M a,b Capox 0 NA*** PD No 1

C 45.6 M a,b Capox/BV 1 0 SD No 0

D 62.5 F a,b Capox/BV 0 0 SD No 0

E 58.2 M b Capox/BV 1 0 PD No 1

F 72.8 M a,b Capox 0 0 PD No 1

G 63.5 F a,b Palliative chemo

NA NA NA Yes* 0

H 52.7 M b Capox 0 NA*** PD No 1

I 49.8 F a,b Folfox NA NA PD Yes** 0

J 65.2 M a,b Capox 1 0 PD No 0 Capox=capecitabine/oxaliplatin, Fofox=5-FU/leucovorin/oxaliplatin, BV=bevacizumab NA=not applicable *: due to circumstances PET/CT was performed after laparoscopy: trial stop because of liver metastases **: trial stop because of small bowel perforation at laparoscopy: extensive PC ***: laparoscopy not performed because of clinical progressive disease (ileus) and systemic metastases/progressive disease on CT #: slight response on PET, but slight progression CT

Page 55: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

46

Table S1: Inclusion and Exclusion criteria

Inclusion criteria Exclusion criteria

* Primary tumor: histologically proven adenocarcinoma in appendix, colon or rectum, which can be considered the primary tumor related to the PC, which is still present at inclusion or has been previously resected. * Intraperitoneal disease: unresectable PC (i.e. PC in more than 5 of the 7 abdominal regions and/or extensive involvement of the small bowel by PC) of colorecal origin, proven by biopsy. * patients: - 18 years or older. - WHO performance status 0 or 1. - fit to undergo major surgery as well as chemotherapy. - life expectancy > 4 months without therapy. - written informed consent, obtained before inclusion into th trial. - normal bone marrow function (i.e. leucocytes> 2000/l (neutrophyl granulocytes >1.8) and thrombocytes> 80,000/l) and Hemoglobin≥ 5 mmol/l. - bilirubin < 2.5 times the normal upper limit. - ASAT and ALAT < 2.5 times the normal upper limit. - normal creatinine-values and a negative urine dipstick-test for proteins.

- Symptoms of bowel obstruction (in case of bowel obstruction symptoms bypass surgery or construction of an ostomy stoma is needed before inclusion into the study).

- Extra-peritoneal, systemic disease on CT thorax/abdomen. Liver, lung and bone metastases are considered sytemic disease.

- Bleeding diatheses or coagulopathy. - Medical history of CVA or TIA, uncontrolled hypertension, instable angina

pectoris, myocardial infarctian within 6 months before inclusion, congestif heart failure NYHA class II or higher, arhythmia.

- Instable or uncompensated respiratory disease. - Neuropathy in the medical history. - Pregnancy or lactation. Fertile patients who do not use birth control medication. - Previous or concomitant malignancies in the past 5 years, other than non-

melanoma skin cancer or carcinoma in situ. - Active infection. - Chemotherapy with oxaliplatin of fluorouracil within 1 year before inclusion,

progressive disease under oxaliplatin or fluorouracil at any point in time before inclusion, any previous major toxicity under one or more chemotherapy agents used in this protocol.

- Short bowel syndrome. - Surgery within 3 months before inclusiobn, which lead to an abdominal sepsis or

fistulisation (in case of fistulisation patients must have a stable situation of at least 3 months, without signs of active infection or abscess).

- Previous complete cytoreductive surgery and HIPEC (CCRS+HIPEC) and/or surgery leading to an anatomical situation in which CCRS+HIPEC are impossible.

- Current therapy with another study agent.

Page 56: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

47

Fig.1: digital pictures of PC in the 7 abdominal regions before and after neo-adjuvant chemotherapy

(Laparoscopic views from patient A and J before and after systemic chemotherapy)

Page 57: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

48

Fig.2: histopathology of PC from CRC before and after chemotherapy (patient A and patient C)

Page 58: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

49

Fig.S1: trial flowchart

Page 59: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

50

References

1. Jayne D, Fook S, Seow-Choen: Peritoneal carcinomatosis from colorectal cancer. Br J Surg 2002; 89: 1545-1550.

2. Folprecht G, Köhne C, Lutz M: Systemic chemotherapy in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425 – 440.

3. Hompes D, Tiek J, D’Hoore A: HIPEC in T4a colon cancer: a defendable treatment to improve oncologic outcome? Ann Oncol 2012 Jul 25 [Epub ahead of print]

4. Chu D, Lang N, Thompson C: Peritoneal carcinomatosis in nongynecologic malignancy. A prospective study of prognostic factors. Cancer 1989; 63: 364 – 367.

5. Sadeghi B, Arvieux C, Gilly F: Peritoneal carcinomatosis from non-gynecologic malignancies. Results of the EVOCAPE I multicentric prospective study. Cancer 2000; 88: 358 – 363.

6. Elias D, Gilly F, Glehen O: Peritoneal 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.

7. Davies J, O’Neil B:Peritoneal carcinomatosis of gastrointestinal origin: Natural history and treatment options. Exp Opin Invest Drugs 2009; 18: 913 – 919.

8. Bloemendaal AL, Verwaal VJ, van Ruth S: Conventional surgery and systemic chemotherapy for peritoneal carcinomatosis of colorectal origin: A prospective study. Eur J Surg Oncol 2005; 31: 1145 – 1151.

9. Hompes D, Boot H, van Tinteren H, Verwaal V: Unresectable peritoneal carcinomatosis from colorectal cancer: a single center experience. J Surg Oncol 2011; 104: 269 – 273.

10. Elias D, Lefevre J, Chevalier J: Complete cytoreductive surgery plus intraperitoneal chemohyperthermia with

Page 60: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

51

oxaliplatin for peritoneal carcinomatosis of colorectal origin. J Clin Oncol 2009; 27: 681 – 685.

11. Cao C, Yan T, Morris D: A systematic review and meta-analysis of cytoreductive surgery with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 2009; 16: 2152 – 2165.

12. De Gramont A, Figer A, Seymour M: Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 2000; 18: 2938 – 2947.

13. Koppe M, Boerman O, Oyen W: Peritoneal carcinomatosis of colorectal origin: incidence and strategies. Ann Surg 2006; 243: 212 – 222.

14. Verwaal V, van Ruth S, de Bree E: Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 2003; 21: 3737 – 3743.

15. Pelz J, Chua T, Esquivel J. Evaluation of best supportive care and systemic chemotherapy as treatment stratified according to the retrospective Peritoneal Surface Disease Severity Score (PSDSS) for peritoneal carcinomatosis of colorectal origin. BMC Cancer 2010; 10: 689.

16. Köhne C, Vanhoefer U, Hartung G: Clinical predictive factors. Eur J Cancer 2009: 45: 43 – 49.

17. Swellengrebel H, Zoetmulder F, Verwaal V: Quantitative intra-operative assessment of peritoneal carcinomatosis - a comparison of three prognostic tools. Eur J Surg Oncol 2009; 35: 1078 – 1084.

18. Verwaal V, van Tinteren H, van Ruth S: Predicting the survival of patients with peritoneal carcinomatosis of colorectal origin treated by aggressive cytoreduction

Page 61: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

52

and hyperthermic intraperitoneal chemotherapy. Br J Surg 2004; 91: 739 – 746.

19. A’Hern RP: Sample size tables for exact single stage phase II designs. Statistics in Medicine 2001; 20: 859 – 866.

20. Oken M, Creech R, Tormey D: Toxicity And Response Criteria Of The Eastern Cooperative Oncology Group. Am J Clin Oncol 1982; 5: 649 – 655.

21. Yan T, Black D, Savady R: Systematic review on the efficacy of cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis fran colorectal cancer. J Clin Oncol 2006; 24: 4011 – 4019.

22. Verwaal V, Bruin S, Boot H: 8-year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol 2008; 15: 2426 – 2432.

23. Franko J, Ibrahim Z, Gusani N: Cytoreductive surgery and hyperthermic intraperitoneal chemoperfusion versus systemic chemotherapy alone for colorectal peritoneal carcinomatosis. Cancer 2010; 116: 3756 – 3762.

24. Elias D, Glehen O, Pocard M: A comparative study of complete cytoreductive surgery plus intraperitoneal chemotherapy to treat peritoneal dissemination from colon, rectum, small bowel and nonpseudomyxoma appendix. Ann Surg 2010; 251: 896 – 901.

25. Elias D, Gilly F, Boutitie F: 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.

26. Verwaal V, van Ruth S, Witkamp A: Long-term survival of peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 2005; 12: 65 – 71.

Page 62: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

53

27. Elias D, Pocard M: Treatment and prevention of peritoneal carcinomatosis from colorectal cancer. Surg Oncol Clin N Am 2003; 12: 543 – 559.

28. van Leeuwen B, Graf W, Pahlman L: Swedish experience with peritonectomy and HIPEC in peritoneal carcinomatosis. Ann Surg Oncol 2008; 15: 745 – 753.

29. Gomes da Silva R, Sugarbaker P: 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.

30. Malik H, Farid S, Al-Mukthar A: A critical appraisal of the role of neoadjuvant chemotherapy for colorectal liver metastases: a case-controlled study. Ann Surg Oncol 2007; 14: 3519 – 3526.

31. Adam R, Vibert E, Pitombo M: Induction chemotherapy and surgery for colorectal liver metastases. Bull Cancer 2006; 93 Suppl 1: S45 – 49.

32. Fusai G, Davidson B. Strategies to increase the respectability of liver metastases from colorectal cancer. Dig Surg 2003; 20: 481 – 496.

33. Ruers T, Bleichrodt R: Treatment of liver metastases, an update on the possibilities and results. Eur J Cancer 2002; 38: 1023 – 1033.

34. Alberts S, Poston G: Treatment advances in liver-limited metastatic colorectal cancer. Clin Colorectal Cancer 2011; 10: 258 – 265.

35. Bismuth H, Adam R, Levi F: Resection of nonresectable liver metastases from colorectal cancer after neoadjuvant chemotherapy. Ann Surg 1996; 224: 509 – 520.

36. Adam R, Delvart V, Pascal G: Rescue surgery for unresectable colorectal liver metastases downstaged by chemotherapy: a model to predict long-term survival. Ann Surg 2004; 240: 644 – 657.

Page 63: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

54

37. Van Cutsem E, Nordlinger B, Adam R:Towards a pan-European consensus on the treatment of patients with colorectal liver metastases. Eur J Cancer 2006; 42: 2212 – 2221.

38. Stillwell A, Ho Y, Veitch C: Systemic review of prognostic factors related to overall survival in patients with stage IV colorectal cancer and unresectable metastases. World J Surg 2011; 35: 684 – 692.

39. Pelz J, Stojadinovic A, Nissan A: Evaluation of peritoneal disease severity score in patients with colon cancer with peritoneal carcinomatosis. J Surg Oncol 2009; 99: 9 – 15.

Page 64: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

55

II

3. Review: Incidence and clinical significance of Bevacizumab-related

non-surgical and surgical serious adverse events

Hompes D, Ruers T

Eur J Surg Oncol 2011; 37: 737-746.

Page 65: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

56

Page 66: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

57

Review: Incidence and clinical significance of Bevacizumab-related non-surgical and surgical serious adverse

events in metastatic colorectal cancer.

D.Hompes, T.Ruers

The Netherlands Cancer Institute, Amsterdam, The Netherlands

Introduction Angiogenesis is the formation of new blood vessels by

remodeling and expansion of primary vessels, which is crucial for tumor growth beyond 1-2mm and for metastasis. Tumoral angiogenesis is the result of complex molecular mechanisms and appears to be highly related to oxygen deprivation. Vascular endothelial growth factor (VEGF) is a glycoprotein that induces neovascularization and has an increased expression in tumor cells. Angiogenesis of tumor vasculature is fenestrated, chaotic and abnormal, which creates abnormal blood vessels and impairs effective delivery of chemotherapeutic agents to the targeted cancer cells1-3.

Bevacizumab (BV) is a humanized monoclonal antibody that neutralizes the ability of all active VEGF-isoforms to bind to the VEGF receptor (VEGFR) on the surface of endothelial cells. It has 2 potentially cytostatic effects: prevention of neovascularization and normalization of immature and abnormal blood vessels1,4,5. This is assumed to retard the shedding of metastatic cells in the circulation and improve the delivery of therapeutic agents in tumors6. Pharmacodynamic data on BV suggest that combination with cytotoxic chemotherapy enhances its anti-tumor activity1.

On the other hand, VEGF has critical role in wound healing, liver regeneration and endothelial integrity. Therefore, one can expect an anti-VEGF agent, such as BV, to cause mechanism-related serious adverse events (SAE) due to the

Page 67: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

58

reduction of VEGF availability5. The most frequently described side effects are arterial hypertension (AHT), gastrointestinal (GI) perforations, wound healing complications (WHC), serious bleeding, arterial and venous thromboembolic events (ATE and VTE), renal toxicity and influences on liver parenchyma5,7-13. Pharmacokinetic studies show that BV has a half-life of 20 days [range 11-50 days]. Consequently, BV-effects may persist despite discontinuation before surgery, but the time for dissipation of BV’s pharmacologic effects after the last dose has not been established. A dose of 5mg/kg every 2 weeks is accepted to be the most effective dose of BV. Waiting for two half-lives (i.e. ±6 weeks) would leave the equivalence of a dose of 1.25mg/kg in the circulation. This is far above the BV level that removes free VEGF from the circulation14-16. Hence, the time to surgery (TTS) that should be respected after discontinuation of BV has not been clearly defined yet. Recommendations vary from a TTS of 5-6 weeks1,7,17 to a more careful 6-8 week interval16. For postoperative initiation of BV a period of 28 days after surgery and a fully healed surgical incision are recommended, because of possible impairment of wound healing under BV16-18.

The aim of this paper is to review currently available literature, describing the actual extent, frequency and clinical importance of BV-related SAE after minor and major surgery, during or after systemic chemotherapy with BV, in patients with metastatic colorectal cancer (mCRC).

Methods A detailed PubMed search was performed in september 2010, using the following keywords: angiogenesis, angiogenesis-inhibitors, VEGF, anti-VEGF, bevacizumab, Avastin, surgery, complications, adverse events, metastatic colorectal cancer, wound healing, bleeding, hemorrhage, gastrointestinal perforation, thrombo-embolism. The search

Page 68: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

59

was limited to articles published in the English language and the eldest publication extracted dates from February 2001.

Results Overall Survival, Progression-Free Survival and Response Rate [Table 1]

Table 1 gives an overview of data on overall survival (OS), progression-free survival (PFS) and response rate (RR) reported by 7 randomized controlled trials (RCTs)19-25,31, 4 prospective8,26-28 and 3 retrospective studies11,29,30. A recent Cochrane Database systematic review by Wagner et al., including 5 randomized trials (>3000 patients)19,22,23,25,31, concluded BV prolongs both PFS and OS in patients with mCRC. The effect on PFS shows significant heterogeneity, which is probably attributable to differences in the treatment effect of BV in combinations with different “chemotherapy backbones” (i.e. different combinations of 5FU/LV, oxaliplatin, irinotecan). Furthermore, an absolute increase in tumor response of approximately 3% was found in favor of the patients treated with BV2.

Non-surgical BV-related Serious Adverse Events Prospective data (observational trials and RCTs)

Table 2 summarizes the serious adverse events (SAE, i.e. grade 3-5 adverse events) assumed to be related to BV-based chemotherapy, as reported by 6 RCT, 2 prospective observational trials and one pooled analysis of 2 RCT19-28. The authors agree that most BV-related adverse events (AE) are mild to moderate in severity and manageable using standard therapies. The First BEAT study reported grade 5 toxicity in 2% of patients, including hemorrhage (<0.5%), cardiac disorders (<0.5%), respiratory disorders (<0.5%), venous embolism (1%) and GI perforation (0.4%). An intention-to-treat-based analysis showed a 3% 60-day mortality27. The review by Wagner et al. showed no significant differences for treatment-related deaths

Page 69: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

60

and 60-day mortality with or without BV2. However, the number of treatment interruptions due to AE was significantly higher under chemotherapy with BV than without (21% versus 15%)2. The majority of trials attributed the most significant increase in toxicity to a significantly higher incidence of arterial hypertension (AHT) [Table 2]. Many trials also reported an increase in ATE [Table 2]. According to the analysis by Wagner et al. the increase in grade 3-4 AHT and ATE should be rated as statistically significant2. Bleeding as a SAE was reported from 0 to 5% in patients under BV-based treatment [Table 2]. In the ECOG study E3200 the incidence of hemorrhage was statistically higher for patients under second-line chemotherapy with BV (p=0.011)22. This was confirmed by the review of Wagner et al., whereas a non-significant increased incidence of bleeding was found under BV in first-line chemotherapy2. GI perforation was defined by Kabbinavar et al. as all events reported as GI abscess, perforation and fistula of any grade unrelated to surgery20. A rate of “spontaneous” GI perforations under BV of up to 3% was recorded [Table 2]. An extensive meta-analysis by Hapani et al. reported an incidence of GI perforation under BV treatment of <1%, resulting in a mortality of 22%32. Patients under BV-treatment had a significantly increased risk of GI perforation, with a positive correlation with higher doses of BV (5mg/kg versus 2.5mg/kg per week) and colorectal cancer (CRC)32. In general, most authors agreed the addition of BV to chemotherapy caused no clinically relevant aggravation of chemotherapy-related SAE. Patients aged ≥65 years didn’t appear to have greater risk with BV-treatment than younger patients20,28, except for a relatively higher proportion of ATE in patients ≥75 years2,28. Furthermore, despite more ECOG PS score >1 and more comorbidities in the BRiTE study, the SAE rate was not substantially different from other BV-treated RCT cohorts28.

Page 70: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

61

Risk for emergency surgery under BV-treatment? Few papers specify the risk for emergency surgery that is due to SAE in patients under BV-treatment. Poultsides et al. retrospectively described that out of 233 patients who received up-front triple drug chemotherapy (with oxaliplatin or irinotecan) with or without BV 16 patients (7%) underwent emergent surgery under chemotherapy: 5 resections for perforations and 3 resections plus 8 diversions for primary tumor obstruction30. Only 2 of the 5 tumor perforations occurred under BV-treatment. For the obstructive events the number of patients under BV was not clearly specified. No intractable bleedings, necessitating surgical intervention, were reported. Two patients with primary tumor complications (<1%) died within 30 days of surgery. The authors didn’t specify whether these patients were on BV-treatment, but they concluded that the risk of emergent intervention was not associated with the use of BV30. Of 529 patients under BV-based treatment in the ECOG study E3200 only 1 developed a grade 4 bleeding that required a hemostatic intervention and 6 events of bowel perforation were reported, 2 of which needed surgery and 2 cases were fatal22. In a multicenter trial of Kabbinavar et al. 2 of the 104 patients under BV-based treatment developed a bowel perforation (each associated with a colonic diverticulum). One patient died due to this complication. Nevertheless, SAE leading to death or study discontinuation were found to be similar with or without BV23, which concurs with the analysis of Wagner et al2.

Risks of BV-treatment if the primary tumor is still in situ? In case of asymptomatic synchronous stage IV CRC, the rationale for immediate resection of the primary tumor would be the prevention of primary-related AE that might make urgent surgery under chemotherapy necessary, thus possibly increasing mortality. On the other hand, immediate resection of an asymptomatic primary might cause an important delay or even exclude some patients from chemotherapy30. Few trials

Page 71: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

62

report specifically on the incidence of major complications related to the primary tumor in synchronous stage IV CRC under BV-based therapy. The First BEAT study reported a GI perforation in 8 of 223 patients (4%) with unresected primary tumors: only 3 of them occurred at the primary tumor site27. In the BRiTE study GI perforation rate was 3% in patients with an intact primary versus almost 2% in case of a resected primary tumor28. Multivariate analysis rated this as an independent risk factor for GI perforation, but event numbers were low: only 9 out of 305 patients (3%) with an unresected primary tumor developed a GI perforation. Whether these perforations occurred at the primary tumor site, was not specified. Based on a literature search, Poultsides et al. concluded that virtually all BV-related perforations were observed in the first 3 months of treatment (mostly within the first month) and occurred throughout the entire GI tract, hardly ever involving the site of the primary tumor22,25,30.

BV-related complications after surgery RCT data [Table 3]

The pooled analysis by Scappaticci et al.33 includes 1132 patients, who underwent 5FU/leucovorin- or Folfiri-based chemotherapy with or without BV in a phase II23 and phase III RCT25. This population was analyzed in two groups: In group 1 chemotherapy (with or without BV) was started 28-60 days after surgery and complication rates were similarly low in both groups (around 1%). In group 2 patients underwent major or emergent surgery during chemotherapy with or without BV. The higher number of surgical procedures in BV-treated patients was not explained, but apparently there were more elective, non-cancer related procedures in this group. The difference in SAE incidence with or without BV (13% versus 3%) didn’t reach statistical significance. Nevertheless, it might have clinical relevance, so careful monitoring of patients who need to undergo major surgery during BV-based chemotherapy is

Page 72: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

63

recommended. The “major” surgical procedures in the pooled analysis by Scappaticci et al. are listed in Table 5B. The small number of events in this analysis made it impossible to draw definite conclusions on the appropriate timing of surgery following the last BV dose as well as the influence of comorbidity (e.g. diabetes, obesity, smoking) on healing processes33. The NSABP C-08 trial (National Surgical Adjuvant Breast and Bowel Project) compared Folfox with or without BV in the adjuvant setting for stage II and III CRC. In this study BV increased the risk of wound healing complications (WHC), even if it was initiated 6 weeks from the time of surgery. Furthermore, it was suggested that extended use of BV can increase the long-term risk of WHC for up to 6 months after its cessation18,34.

Prospective observational data [Table 4] In the First BEAT study almost 12% of patients underwent surgery with curative intent “during trial participation”, of whom 64% (145 patients) underwent hepatectomy [Table 5A]. Bleeding was reported as an SAE in almost 3% and WHC in 2.0%27. In the BRiTE study almost 27% patients underwent surgery within 90 days of the last BV dose, of whom 30% (175 patients) underwent major abdominal surgery and 17% (88 patients) underwent hepatectomy. The incidence of serious WHC was 4.4%. The absolute number of severe WHC appeared higher after major than after minor surgery: 6% of abdominal procedures and 6% of hepatectomies developed severe WHC versus 3% for minor surgical procedures28 [Table 5B]. A subanalysis of the time to surgery (TTS) was performed [Table 5B], but no multivariate analysis was performed for WHC because event numbers were low. Concerning severe bleeding, a multivariate analysis couldn’t identify significant risk factors, not even antiplatelet therapy or anticoagulation28. This concurs with a trial of Saltz et al., who found bleeding events to be similar for BV-based chemotherapy with and without concurrent anticoagulation therapy19.

Page 73: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

64

Unfortunately, the BRiTE study and the First BEAT trial didn’t clearly specify the number of severe bleeding events that occurred postoperatively. Gruenberger et al. prospectively reported on 52 liver resections 5 weeks after BV35: Thirty-six percent of patients underwent a major hepatectomy (i.e. resection of ≥3 liver segments) [Table 5A]. Eleven postoperative SAE were recorded: 1 bowel perforation (for which reoperation was needed), 1 anastomotic leak, 1 wound hematoma, 1 wound infection, 3 cases of sepsis and 1 bile leak. There were no severe bleeding events. Twenty-one percent of patients underwent synchronous resection of liver and primary tumor [Table 5A], resulting in similar peri- and postoperative complication rates as in patients undergoing only hepatectomy. Concerning liver function and regeneration, Gruenberger et al. reported normal findings in 51 patients (98%)26. This concurs with observations from MD Anderson11 and a retrospective analysis by Klinger et al8, which suggests a reduced incidence and severity of oxaliplatin-related sinusoidal dilatation in patients under chemotherapy with BV. Finally, 2 phase I studies by Willett et al. included a very small number of patients who all underwent major surgery after BV-based chemotherapy. Reported complications were low6,36 [Table 5B].

Retrospective data Zawacki et al. performed the only study focusing purely on minor surgery under BV-based chemotherapy [Table 5C]. They analyzed WHC as a SAE under BV-treatment with variable doses before or after placement of a venous access port. All 6 dehiscences occured under BV-treatment. Despite the very small number of events statistical analysis was performed and the authors concluded that patients receiving BV within 10 days of port placement had a higher incidence of wound dehiscence37. A study from MD Anderson focused on hepatic surgery after chemotherapy with or without BV: 30% of patients underwent major hepatectomy38 [Table 5A]. Median time between discontinuation of BV and surgery was 58 days

Page 74: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

65

[range 31-117]. No significant association was found between use of BV and postoperative complication rate (49% with vs. 43% without BV). The time interval from discontinuation of BV to surgery was not associated with an increased likelihood of developing complications38. Reddy et al. analyzed morbidity of hepatectomy after irinotecan- or oxaliplatin-based chemotherapy with or without BV: 24% of patients underwent major hepatectomy and in 16% of patients synchronous extrahepatic procedures were performed39 [Table 5A]. The differences in complications after chemotherapy with or without BV were all rated statistically insignificant. An analysis for TTS (≤ or >8 weeks) showed no significant differences for overall, severe and hepatic complications39. D’Angelica et al. studied the influence of BV-based chemotherapy within 12 weeks from hepatectomy: 27% of patients underwent major hepatectomy40 [Table 5A]. Only 2 SAE (grade 3) were reported under BV: 1 subphrenic abscess and 1 groin abscess. Because of the heterogenous timing of BV administration (before, after or before and after surgery) and the low complication rate definitive conclusions are difficult, but the perioperative complication profile was within the limits expected in any group of patients undergoing hepatectomy40. Finally, Bose et al. suggested that, although a period of 28 days is recommended for postoperative initiation of BV, the time point for the start of BV should be individualized in case of comorbidity (e.g. diabetes, peripheral vascular disease) or wound-healing issues, thus allowing more time for wound healing16.

Discussion The BV-related SAE reported in literature are mild to moderate in severity and manageable using standard therapies. Furthermore, the incidence of SAE under BV-based chemotherapy is low: even very large, multicenter prospective and randomized controlled trials reported only very small numbers of SAE. In most trials the incidence of hemorrhage

Page 75: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

66

(2.0-3.0%), GI perforation (<1.0-2.0%) and ATE (1.0-2.0%) was higher under BV-based treatment, but overall the absolute number of patients affected remained very low. Also, BV-related grade 5 toxicity was rare. Thus, most authors agreed the addition of BV to chemotherapy caused no clinically relevant aggravation of chemotherapy-related SAE. Only few trials report specifically on the incidence of major complications related to the primary tumor in synchronous stage IV CRC under BV-based therapy. With the primary tumor still in situ during chemotherapy, BV-related GI perforations occur in about 3% of patients, but virtually all of these were observed in the first 3 months of treatment and occurred throughout the entire GI tract, hardly ever involving the primary tumor site. Thus, leaving the primary tumor in place during BV-treatment appears safe. Finally, the risk emergency surgery might be needed due to BV-related SAE, such as bleeding or perforation, is very low (estimated 2.0%).

The relation between postoperative SAE and BV-based chemotherapy can be divided in 3 groups: First, for patients undergoing surgery after BV discontinuation trials showed very low rates of SAE if a TTS of 5-6 weeks is respected. The majority of SAE reported are WHC. Bleeding and GI perforation occur infrequently. In most of these trials 30-40% of patients undergo major surgery, which suggests the low rate of SAE reported should be reliable. For minor surgery, WHC are the main issue, but rates are low if BV is discontinued within a 10 day period around the procedure. Second, patients undergoing major or emergency surgery during BV-treatment seemed at higher risk (SAE 1.3-2.7% vs. 0.0%), but this was rated statistically insignificant. The reported numbers of SAE remained relatively small in this setting, but they could still be clinically relevant. Thus, close monitoring of patients undergoing surgery under BV is advisable. Third, for the start of BV-treatment after major surgery few data are available. Small rates of SAE were found, especially if a period of minimally 28 days after surgery is respected before starting BV. However, in case of comorbidity

Page 76: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

67

or wound-healing issues after surgery, the time point for starting BV should be individualized to allow more time for wound healing.

Conclusion

This review shows that even trials consisting of large patient populations report small numbers of SAE of BV-related SAE in relationship to surgery. This has 2 consequences: Small numbers of events make statistical analysis and definitive conclusions difficult, if not impossible. On the other hand, the fact that even large populations show low absolute numbers of BV-related SAE in any of the settings examined above, suggest that BV-based treatment causes few clinically significant problems even when surgical procedures are involved.

Page 77: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

68

Table 1: Therapy with BV: OS / PFS / RR / CR

Page 78: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

69

Table 2: RCTs / Prospective trials: non-surgical BV-related SAE Ref. Author

Trial

Journal

N

+BV / total

Regimen Non-surgical SAE (%)

Hemorrhage GI

perforation

ATE VTE AHT Diarrhea Leucopenia

/

neutropenia

Death

(within

60d.)

RCTs [19] Saltz

Amended

NO16966 trial

JCO 2008

699/1400 Folfox / Capox +BV 2.0 <1.0 2.0 8.0 4.0 - - 2.0

vs vs vs vs vs vs

Folfox / Capox -BV 1.0 2.0 1.0 5.0 1.0 - - 1.6

[21] Hochster

Amended TREE

study

JCO 2008

213/360 ivFolfox / bolusFolfox / Capox +BV 1.4 2.3 - 2.7 3.6 18.6 26.0 1.9

vs vs vs vs vs

ivFolfox / bolusFolfox / Capox -BV 0.0 - - 1.3 - 29.3 28.7 3.4

[22] Giantonio

ECOG study

E3200

JCO 2007

529/820 Folfox +BV 3.4 0.6 0.9 3.4 6.2 - - 5.0

vs vs vs vs vs vs

Folfox -BV 0.4 0.0 0.4 2.5 1.8 - - 4.0

vs vs vs vs vs vs

BV alone 2.1 0.8 0.4 0.4 7.3 - - 6.0

(p=0.011) (p=0.62) (p=0.62) (p=0.008)

[23] Kabbinavar*

JCO 2005

104/209 5FU/LV +BV 5.0 2.0 10.0 9.0 16.0 39.0 5.0 5.0

vs vs vs vs vs vs vs vs

5FU/LV -BV 3.0 0.0 5.0 11.0 3.0 40.0 14.0 13.5

[24] Kabbinavar

JCO 2005

249/490 IFL +BV 5.0 1.0 5.0 10.0 16.0 37.0 5.0 -

vs vs vs vs vs vs vs

IFL -BV 2.0 0.0 3.0 9.0 3.0 34.0 19.0 -

[25] Hurwitz**

N Engl J Med

2004

402/813 IFL +BV 3.1 1.5 19.4 11.0 32.4 37.0 4.9

vs vs vs vs vs vs vs

IFL -BV 2.5 0.0 16.2 2.3 24.7 31.1 3.0

(p=0.26)

[20] Kabbinavar

JCO 2009

218/439

Patients ≥

65years

(pooled

analysis */**)

5FU/LV or IFL +BV 4.8 2.9 7.6 14.9 13.8 38.6 30.0 6.7

vs vs vs vs vs vs vs vs

5FU/LV or IFL -BV 3.7 0.0 2.8 17.5 1.8 33.2 23.5 8.8

Prospective

trials

[27] Van Cutsem

First BEAT study

Ann Oncol 2009

1914/1914 5FU-based chemotherapy +BV 3.0 2.0 1.0 - 5.0 4.0 - 3.0

[28] Kozloff

BRiTE study

The Oncologist

2009

1953/1953 “BV-containing therapy” (mainly 5FU-

based)

2.2 1.9 2.0 - 22.0 - - 2.1

[26] Gruenberger

EJSO 2009

56/56 Capox +BV 0.0 2.0 7.0 3.0 33.0 10.0 0.0

Page 79: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

70

Tables 1 & 2: p-values are mentioned if available Abbreviations: ng: not given; p=NS: statistically not significant; N +BV/total = number of patients undergoing surgery during or after BV-based treatment over the total population

Page 80: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

71

Table 3: RCT: post-surgery BV-related SAE

Ref. Author

Journal

N

surgery

+BV /

total

Regimen Post-surgery SAE (%)

[33] Scappaticci

J Surg

Oncol 2005

305/1132

(pooled

analysis */**)

GI

perforation

Abd.

fistula

Anastomotic

dehiscence

Intra-

abd.

bleeding

Other

Hemorrhages

Wound

Healing

Infection death

Group 1: Start BV 28-60d.

AFTER surgery

5FU/LV or IFL

+BV

0.9 - - 0.4 - - - -

vs vs

5FU/LV or IFL -

BV

0.0 - - 0.5 - - - -

Group 2: Major /

emergency

surgery DURING BV-treatment

5FU/LV or IFL

+BV

2.7 2.7 1.3 1.3 1.3 1.3 2.7 2.7

vs vs vs vs vs vs vs vs

5FU/LV or IFL -

BV

0.0 0.0 3.4 0.0 0.0 0.0 0.0 3.4

Page 81: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

72

Table 4: Prospective observational trials: post-surgery SAE Ref. Author

Trial

Journal

N

surgery

+BV /

total

TTS

Regimen Post-surgery SAE (%)

GI

perforation

Abd.

fistula

Anastomotic

dehiscence

Hemorrhage Wound

healing

Wound

infection

other

[27] Van Cutsem

First BEAT

study Ann Oncol

2009

225/1914 “during trial

participation”

5FU-based

chemotherapy +BV

- - - 2.7 2.0 - -

[28] Kozloff

BRiTE The

Oncologist

2009

521/1953 “within 90

days”

0-2 weeks

2-4 weeks

4-6 weeks

6-8 weeks

>8 weeks

“BV-containing

therapy” (mainly 5FU-based)

- - - - 4.4 1.5 -

9.7

3.2

3.0

5.9 2.2

[26] Gruenberger

JCO 2008

56/56 5 weeks Capox +BV 2.0 - 2.0 0.0 4.0 2.0 Bile

leak

2.0 Sepsis

6.0

[35] Gruenberger

JCO 2006

9 5 weeks Capox +BV - - - 0.0 0.0 11.1 0.0

[36] Willett

JCO 2005

5 7-9 weeks 5FU +BV +

radiotherapy

20.0 0.0 0.0 0.0 0.0 0.0 20.0

[6] Willett

Nat Med

2004

6 7 weeks 5FU +BV +

radiotherapy

0.0 0.0 0.0 0.0 0.0 0.0 0.0

Page 82: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

73

Table 5: Types of surgery vs postop.complications

Page 83: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

74

Tables 3, 4 & 5: p-values: mentioned if available ng: not given; p=NS: statistically not significant; TTS: time to surgery, i.e. the time interval between discontinuation of BV and surgery; TpS: time post surgery, i.e. the time interval between surgery en (re)start of BV; N surgery +BV / total= number of patients undergoing surgery during or after BV-based treatment over the total population

Abbreviations throughout the manuscript:

Vascular endothelial growth factor (VEGF), Bevacizumab (BV), VEGF receptor (VEGFR), adverse events (AE), serious adverse events (SAE),arterial hypertension (AHT), gastrointestinal (GI), wound healing complications (WHC), arterial thromboembolic events (ATE), venous thromboembolic events (VTE), time to surgery (TTS), colorectal cancer (CRC), metastatic CRC (mCRC), overall survival (OS), progression-free survival (PFS), response rate (RR), randomized controlled trial (RCT), 5-fluorouracil/leucovorin (5FU/LV), Memorial Sloan Kettering Cancer Center (MSKCC)

Page 84: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

75

References 1. Shih T, Lindley C. Bevacizumab: an angiogenesis

inhibitor for the treatment of solid malignancies. Clin Therap 2006; 28: 1779-1802.

2. Wagner A, Arnold D, Grothey A. Anti-angiogenic therapy for metastatic colorectal cancer. Cochrane Database Syst Rev 2009; 8(3): CD005392.

3. McDonald D, Baluk P. Significance of blood vessel leakiness in cancer. Cancer Res 2002; 62: 5381-5385.

4. Cao Y, Zhong W, Sun Y. Improvement of angiogenic cancer therapy by understanding the mechanisms of angiogenic factor interplay and drug resistance. Semin Cancer Biol 2009 may 27 [Epub ahead of print]

5. Los M, Roodhart J, Voest E. Target practice: lessons from phase III trials with bevacizumab and vatalanib in the treatment of advanced colorectal cancer. The Oncologist 2007; 12: 443-450.

6. Willett CG, Boucher Y, di Tomaso E. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 2004; 10: 145-147.

7. Nordlinger B, Van Cutsem E, Gruenberger T. Combination of surgery and chemotherapy and the role of targeted agents in the treatment of patients with colorectal liver metastases: recommendations from an expert panel. Ann Oncol 2009; 20: 985-992.

8. Klinger M, Eipeldauer S, Gruenberger T. Bevacizumab protects against sinusoidal obstruction syndrome and does not increase response rate in neoadjuvant xelox/folfox therapy of colorectal cancer liver metastases. Eur J Surg Oncol 2009; 35: 515-520.

9. Schiff D, Wen P, van den Brent M. Neurological adverse effects caused by cytotoxic and targeted therapies. Nat Rev Clin Oncol 2009; 6: 596-603.

10. Kelly R, Billemont B, Rixe O. Renal toxicity of targeted therapies. Target Oncol 2009; 4: 121-33.

Page 85: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

76

11. Ribero D, Wang H, Vauthey J. Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer 2007; 110: 2761-2767.

12. Fakih M, Lombardo J. Bevacizumab-induced nasal septum perforation. The Oncologist 2006; 11: 85-86.

13. Nalluri S, Chu D, Keresztes R. Risk of venous thromboembolism with the angiogenesis inhibitor bevacizumab in cancer patients: a meta-analysis. JAMA 2008; 300: 2277-2285.

14. Ellis L, Curley S, Grothey A. Surgical resection after downsizing of colorectal liver metastasis in the era of bevacizumab. J Clin Oncol 2005; 23: 4853-4855.

15. Gordon M, Margolin K, Talpaz M. Phase I safety and pharmacokinetic study of recombinant human anti-Vascular Endothelial Growth Factor in patients with advanced cancer. J Clin Oncol 2001; 19: 843-850.

16. Bose D, Meric-Bernstam F, Hofstetter W. Vascular endothelial growth factor targeted therapy in the perioperative setting: implications for patient care. Lancet Oncol 2010; 11: 373-382.

17. Information brochure Roche: Avastin. 18. Allegra C, Yothers G, O’Connell M. Initial safety report of

NSABP C-08: a randomized phase III study of modified FOLFOX6 with or without bevacizumab for the adjuvant treatment of patients with stage II or III colon cancer. J Clin Oncol 2009; 27: 3385-3390.

19. Saltz L, Clarke S, Diaz-Rubio E. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2008; 26: 2013-2019.

20. Kabbinavar F, Hurwitz H, Rosen O. Addition of bevacizumab to fluorouracil-based first-line treatment of metastatic colorectal cancer: pooled analysis of

Page 86: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

77

cohorts of older patients from two randomized clinical trials. J Clin Oncol 2009; 27: 199-205.

21. Hochster H, Hart L, Hedrick E. Safety and efficacy of oxaliplatin and fluoropyrimidine regimens with or without bevacizumab as first-line treatment of metastatic colorectal cancer: results of the TREE Study. J Clin Oncol 2008; 26: 3523-3529.

22. Giantonio B, Catalano P, Meropol N. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol 2007; 25: 1539-1544.

23. Kabbinavar F, Schulz J, Novotny W. Addition of bevacizumab to bolus fluorouracil and leucovorin in first-line metastatic colorectal cancer: results of a randomized phase II trial. J Clin Oncol 2005; 23: 3697-3705.

24. Kabbinavar F, Hambleton J, Hurwitz H. Combined analysis of efficacy: the addition of bevacizumab to fluorouracil/leucovorin improves survival for patients with metastatic colorectal cancer. J Clin Oncol 2005; 23: 3706-3712.

25. Hurwitz H, Fehrenbacher L, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350: 2335-2342.

26. Gruenberger B, Tamandl D, Gruenberger T. Bevacizumab, capecitabine and oxaliplatin as neoadjuvant therapy for patients with potentially curable metastatic colorectal cancer. J Clin Oncol 2008; 26: 1830-1835.

27. Van Cutsem E, Rivera F, Cunningham D. Safety and efficacy of first-line bevacizumab with folfox, xelox, folfiri and fluoropyrimidines in metastatic colorectal cancer: the BEAT study. Ann Oncol 2009; 20: 1842-1847.

Page 87: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

78

28. Kozloff M, Yood M, Grothey A. Clinical outcomes associated with bevacizumab-containing treatment of metastatic colorectal cancer: the BRiTE observational cohort study. The Oncologist 2009; 14: 862-870.

29. Blazer D, Kishi Y, Vauthey N. Pathologic response to preoperative chemotherapy: a new outcome end point after resection of hepatic colorectal metastases. J Clin Oncol 2008; 25: 5344-5351.

30. Poultsides G, Servais E, Saltz L. Outcome of primary tumor in patients with synchronous stage IV colorectal cancer receiving chemotherapy without surgery as initial treatment. J Clin Oncol 2009; 27: 3379-3384.

31. Kabbinavar F, Hurwitz H, Fehrenbacher L. Phase II randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin(LV) with 5FU/LV alone in patients with metastatic colorectal cancer. J Clin Oncol 2003; 21: 60-65.

32. Hapani S, Chu D, Wu S. Risk of gastrointestinal perforation in perforation in patients with cancer treated with bevacizumab: a meta-analysis. Lancet Oncol 2009; 10: 559-568.

33. Scappaticci F, Fehrenbacher L, Hurwitz H. Surgical wound healing complications in metastatic colorectal cancer patients treated with bevacizumab. J Surg Oncol 2005; 91: 173-180.

34. Wolmark N, Yothers G, O’Connell M. A phase III trialcomparing mFolfox6 to mFolfox plus bevacizumab in stage II and III carcinoma of the colon: results of NSABP protocol C-08. Proc Am Soc Clin Oncol 2009; 27: abstr LBA4.

35. Gruenberger T, Gruenberger B, Scheithauer W. Neoadjuvant therapy with bevacizumab. J Clin Oncol 2006; 24: 2592-2593.

36. Willett C, Boucher Y, Duda D. Surrogate markers for antiangiogenic therapy and dose-limiting toxicities for bevacizumab with radiation and chemotherapy:

Page 88: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

79

continued experience of a phase I trial in rectal cancer patients. J Clin Oncol 2005; 23: 8136-8139.

37. Zawacki W, Walker G, DeVasher E. Wound dehiscence or failure to heal following venous access port placement in patients receiving bevacizumab therapy. J Vasc Interv Radiol 2009; 20: 624-627.

38. Kesmodel S, Ellis L, Lin E. Preoperative bevacizumab does not significantly increase postoperative complication rates in patients undergoing hepatic surgery for colorectal cancer liver metastases. J Clin Oncol 2008; 26: 5254-5260.

39. Reddy S, Morse M, Hurwitz H. Addition of bevacizumab to irinotecan- and oxaliplatin-based preoperative chemotherapy regimens does not increase morbidity after resection of colorectal liver metastases. J Am Coll Surg 2008; 206: 96-106.

40. D’Angelica M, Kornprat P, Saltz L. Lack of evidence for increased morbidity after hepatectomy with perioperative use of bevacizumab: a matched case-control study. Ann Surg Oncol 2007; 14: 759-765.

Page 89: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

80

Page 90: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

81

III Complete cytoreductive

surgery and HIPEC for peritoneal carcinomatosis

of colorectal origin

Page 91: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

82

Page 92: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

83

1. The treatment of peritoneal

carcinomatosis of colorectal cancer with complete cytoreductive surgery

and hyperthermic intra-peritoneal peroperative chemotherapy (HIPEC)

with Oxaliplatin: a Belgian multicenter prospective

phase II clinical study

Hompes D, D’Hoore A, Van Cutsem E, Fieuws S,

Ceelen W, Peeters M, Van der Speeten K, Bertrand C, Kerger J, Legendre H

Ann Surg Oncol 2012; 19: 2186-2194.

III

Page 93: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

84

Page 94: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

85

The Treatment of Peritoneal Carcinomatosis of Colorectal Cancer with Complete Cytoreductive

Surgery and Hyperthermic Intra-peritoneal Peroperative Chemotherapy (HIPEC) with Oxaliplatin:

A Belgian Multicentre Prospective Observational Clinical Study.

D.Hompes1, A.D'Hoore1, E.Van Cutsem2, S.Fieuws3, W.Ceelen4, M.Peeters5, K.Van der Speeten6, C.Bertrand7, J.Kerger8,

H.Legendre9

1 Department of Abdominal Surgery, University Hospitals Gasthuisberg, Leuven, Belgium; 2 Department of Digestive Oncology, University Hospitals Gasthuisberg, Leuven, Belgium; 3 I-Biostat, Katholieke Universiteit Leuven and Universiteit Hasselt, Belgium; 4 Department of Abdominal Surgery, University Hospital Ghent. Ghent, Belgium; 5 Department of Digestive Oncology, University Hospital Ghent. Ghent, Belgium; 6 Department of Abdominal Surgery, Ziekenhuis Oost-Limburg, Genk, Belgium; 7 Department of Abdominal Surgery, Hopital de Jolimont, Jolimont. Belgium; 8 Department of Abdominal Surgery, UCL Mont-Godinne, Yvoir, Belgium; 9 Department of Digestive Oncology, UCL Mont-Godinne, Yvoir, Belgium.

Introduction About 10% of Colorectal Cancer (CRC) patients present with Peritoneal Carcinomatosis (PC) at the time of diagnosis and 25% of patients develop PC at recurrence1-4. Until recently,

Page 95: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

86

PC was considered a terminal condition, only to be palliated with systemic chemotherapy. Most frequently, PC is part of generalized metastatic disease (liver, lung, …), but in about 25-35% of cases PC is the only site of recurrent disease1,5-7. Therefore, it may be the first step in dissemination and should not necessarily be interpreted as generalized disease5,8. Such a locoregional tumor extension warrants a locoregional treatment approach. Thus, the background for the concept of complete cytoreductive surgery (CCRS) combined with hyperthermia and intraperitoneal chemotherapy (HIPEC) was developed to obtain locoregional disease control and long-term survival for PC [Table 1].

As Oxaliplatin proved to be effective for treating advanced colorectal cancer9, Pestieau et al. performed phamacokinetic studies on the intraperitoneal use of oxaliplatin10. Their experimental studies showed the exposure of peritoneal surfaces to Oxaliplatin was significantly increased with intraperitoneal administration, compared to intravenous administration (p<0.0001). The area under the curve (AUC) ratio (AUC peritoneal fluid/AUC plasma) was 16(±5):1 for intraperitoneal delivery as opposed to 1:5 (±2) for intravenous delivery (p=0.0059)10.Matheme et al showed a low systemic exposure of Oxaliplatin and a half-life of 29.5 minutes in the perfusate11. Elias et al. found high peritoneal and tumor Oxaliplatin concentrations and, based upon dose-escalation studies, recommended a dose of 460mg/m² Oxaliplatin in 2l/m² of 5% dextrose for HIPEC, at a temperature of 42-44°COover 30 minutes12.

The aim of this multicenter prospective observational clinical study was to assess the safety and efficacy of complete cytoreductive surgery (CCRS) followed by HIPEC with Oxaliplatin for patients with PC of CRC in different Belgian centers.

Materials and methods A Belgian prospective multicenter protocol and registry

was started in January 2004 and in august 2008 an analysis was

Page 96: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

87

performed for perioperative morbidity and mortality, as well as disease-free survival (DFS) and overall survival (OS). Nine centers, involved in the management of peritoneal disease by CCRS + HIPEC, collaborated for the development of the study protocol. The principal investigator, all cooperating centers and their respective medical ethics committees approved the final version of the protocol. Eventually, 6 surgical centers included consecutive patients with PC from CRC. Patients with intra-abdominal mesotheliomas or pseudomyxomas were excluded from the trial, as well as patients in whom there was evidence of extra-abdominal disease or liver metastases. Furthermore, patients who received chemotherapy or radiotherapy in the course of 4 weeks prior to surgery were excluded from the trial. The diagnosis of PC of CRC and the assessment of its resectability were achieved through CT, PET-CT and/or laparoscopic evaluation. Extent of disease at surgery

The extent of disease was described by the Peritoneal Cancer Index (PCI) according to Jacquet and Sugarbaker13, assessing the (number of) abdominal regions involved and scoring their maximum lesion size. As prescribed by protocol only patients with a PCI score less than 25 were allowed to be included in the trial.

Complete Cytoreductive Surgery (CCRS) Extensive debulking with peritonectomy and, when

needed, multi-organ resections were performed as described by Sugarbaker14. The aim was to obtain a macroscopically CCRS (R0/1), i.e. no macroscopically visual residual tumor was left at the end of the surgical resection. Only patients in whom a complete debulking (CC-0) could be reached, were to go on to the HIPEC-procedure.

Page 97: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

88

Hyperthermic Intraperitoneal Chemotherapy (HIPEC) protocol At the end of the surgical debulking procedure, about 1

hour before starting the actual HIPEC procedure, systemic folinic acid (20mg/m²) and 5-FU (400mg/m²) were administered. In the mean time, after complete cytoreduction was achieved, arrangements were made for the actual HIPEC-procedure. First, the abdominal cavity was rinced with saline and the colloseum for the open HIPEC technique was built up. Hyperthermic circulation was started with 2 l/m² of a glucose 5% solution until a steady state of about 41 to 42°C was reached. Oxaliplatin with a dose of 460mg/m² was then added and circulated for 30 minutes. Before construction of the necessary anastomoses and closure of the abdomen, the abdominal cavity was extensively rinced with 3 liters of saline solution. Statistical analysis

Cox regressions and log-rank tests were used to evaluate the relation between a set of variables and OS and DFS respectively. Hazard ratios (HR) and 95% confidence intervals (CI) are reported. DFS is defined as the time until recurrence. Note that there are no deceased patients without recurrence. Considered variables are patient-related (age and sex), primary tumor-related (CEA, grade of differentiation, mucinous type, synchronous occurrence of PC, node involvement of the primary tumor), extent of disease-related (number of abdominal regions involved in the PC, maximal diameter of the PC lesions, PCI score), surgical procedure-related (blood loss, need of transfusion, duration of the surgical procedure, small bowel resection, occurrence of a diaphragmatic tear) and per- and postoperative intra-abdominal and extra-abdominal complications). Restricted cubic splines15 are used in the Cox model to allow a non-linear relation between PCI and the (log) hazard ratio. Due to the low number of events (5 deaths, 21 recurrences), no stratified tests, nor multivariable Cox models are considered. For OS, an exact log-rank test (conditional on

Page 98: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

89

risk set) is used to verify the robustness of the result16. An optimal cutpoint for PCI is defined as the dichotomization maximizing the likelihood in the Cox regression model. A 95% CI for the cutpoint is constructed based on the likelihood function. Fisher exact tests, Mann-Whitney U tests and Spearman correlations are used to explore relations with the occurrence of intra-abdominal complications and with length of hospital stay.

All analyses have been performed using SAS software, version 9.2 of the SAS System for Windows. Copyright © 2002 SAS Institute Inc. SAS and all other SAS Institute Inc. product or service names are registered trademarks or trademarks of SAS Institute Inc., Cary, NC, USA.

Results Patient and Tumor Characteristics Between January 2004 and August 2008 48 consecutive

patients with PC from CRC were included in the trial, 17 of which were male and 31 female [Table 2]. Median age at surgery was 60 years [range 24-76 years]. In this series 72.9% of patients the primary tumor had already been previously resected. In 75% of patients PC was already present primary tumor presentation. The majority of primary tumors was localized in the ascending and rectosigmoid colon and had a moderate to poor differentiation. 39.6% of tumors had a mucinous cell type. Extent of disease

Median PCI was 11 [range 1-22], with a median of 6 abdominal regions [range 1 - 11] involved and a median lesion size score of 3 [range 1-3].

CCRS and HIPEC To obtain a macropscopically CCRS (CC-0) a median of 2

organs [range 2-6] needed to be resected, with anterior

Page 99: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

90

resection in 45.8%, total colectomy in 8.3% and small bowel resection in 12.5% of cases. A median of 1 anastomosis [range 0-6] was performed per patient. Almost one third were low anastomoses and 82.1% were performed after HIPEC. In 16 patients construction of a diversion was needed [Table 3]. Median operation time was 460 minutes [range 125-840 minutes], with a median blood loss of 475 ml [range 2-6000ml]. HIPEC posed few procedural problems. The 30-day mortality was 0%. Complication rate (any grade) was 52.1%, with 18 intra- and 17 extra-abdominal complications. All extra-abdominal complications were WHO grade 1 or 2, except for 1 WHO grade 4 pneumonia. For the intra-abdominal complications anastomotic leakage occurred in 10.4% of patients (all NCI-CTC grade 4) and bleeding in 6.3% (all National Cancer Institute-Common Toxicity Criteria (NCI-CTC) grade 4). One bowel perforation (2.1%) was reported (NCI-CTC grade 4), one abscess (2.1%) (NCI-CTC grade 4) and 1 rectovaginal fistula (NCI-CTC grade 2). Thus, reoperation was needed in 10 patients (20.8%). In 22.9% of patients prolonged ileus was registered, for which no reintervention was needed. Median hospital stay was 20 [range 5-65] days. [Table 4] Univariate analysis showed that the occurrence of intra-abdominal complications significantly affected hospital stay (p=0.002), but no risk factors for occurrence of postoperative complications could be found. Finally, it should be mentioned that 30 patients (62.5%) were started on 5-FU/leucovorin-based adjuvant chemotherapy combined with oxaliplatin or irinotecan within 8 to 12 weeks after CCRS + HIPEC. Overall and Disease-free Survival

At a median follow-up of 22.7 months [range 3.2-55.7months], overall survival (OS) was 97.9% (CI: 86.1-99.7) at 1 year and 88.7% (CI:73.6-95.4) at 2 years [Fig.1A]. Disease-free survival (DFS) at 1 year was 65.8% (CI: 52.3-76.2) and 45.5% (CI: 34.3-55.9) at 2 years [Fig.1B]. The median time until recurrence equals 19.8 months (95%CI: 12, upper limit not defined).

Page 100: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

91

There is no evidence for a relation between PCI and OS (p=0.14 assuming the relation to be linear, p=0.051 when allowing a nonlinear relation). Only after dichotomizing PCI, a significant difference in OS is found between patients with a low and patients with a high PCI. The optimal cutpoint equals 18, which is estimated with a high degree of uncertainty (95%CI: 10;21). Note that this result is still in correspondence with the dichotomization proposed by Elias et al.17. They made a distinction between patients with PCI≤15 and patients with PCI>15, which yields a significant difference (p=0.013) in the current study [Fig.2A]. Besides the dichotomized PCI, resection of small bowel (p=0.004), the occurrence of postoperative extra-abdominal complications (EAC) (p=0.0005) and a lesion size>5cm (p=0.043) are significantly correlated with OS [Table 5]. The same results are reached with the exact tests (results not shown). There is no evidence for a relation between PCI and disease-free survival (p=0.16 assuming the relation to be linear, p=0.12 when allowing a nonlinear relation). The optimal cutpoint coincides with the cutpoint proposed by Elias et al.17 (i.e. PCI≤15 versus PCI>15) and yields a significant difference between both groups (p=0.0009) [Fig.2B]. The 95%CI for the cutpoint equals (15;17). Besides the dichotomized PCI, transfusion (p=0.018) is significantly correlated with DFS [Table 5]. For the resection of small bowel at debulking a trend has been observed (p=0.067).

Discussion

The primary aim of this prospective observational multicentre trial was to assess the safety and efficacy of HIPEC with Oxaliplatin. Many of the larger series in literature report on overall safety and efficacy of CCRS + HIPEC, including patients who underwent HIPEC with Oxaliplatin as well as patients who received HIPEC with Mitomycin C (MMC) in one and the same publication18-21. This might complicate the interpretation and comparison of results. The number of papers

Page 101: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

92

reporting only on CCRS followed by HIPEC with Oxaliplatin is limited and quite often these papers include relatively small numbers of patients12,22,23.

In this Belgian multicenter trial there was no 30-day mortality. The overall complication rate was 52.1%, with anastomotic leakage in 10.4% of patients. bleeding in 6.3% and prolonged ileus in 22.9%. Surgical reintervention was needed in 20.8% of cases. The median hospital stay was 20 [range 5-65] days. These results concur with literature. Elias et al reported grade 3-5 serious adverse events (SAE) in up to 50% of patients who underwent CCRS, resulting in CC-0 resection, followed by HIPEC with Oxaliplatin12,23. Larger series on CCRS + HIPEC with Oxaliplatin or MMC reported mortality rates of <1% to 6%, grade 3-5 SAE rates of 31-66% and reintervention in 11-30% of cases1,18-20. The rate of anastomotic leakages is often not clearly specified. Elias et al reported a gastrointestinal tract complications/fistula rate of 9-10%12,19,20. It goes without saying that these high morbidity rates after CCRS+HIPEC imply that patients undergoing such extensive surgery should be well-selected regarding their general performance status, as well as their extent of disease.

The overall survival (OS) found in this multicenter Belgian registry was 97.9% at 1 year and 88.7% at 2 years, at a median follow-up of 22.7 months [range 3.2-55.7months] [Fig.1A]. This is similar to a retrospective comparative trial of Elias et al, comparing modern systemic therapy for PC to CCRS + HIPEC with Oxaliplatin, reporting a 2-year overall survival of 81% and a 5-year overall survival of 51%22. A systematic review by Morris et al rated the results of this study as level 2b evidence24. Disease-free survival (DFS) at 1 year was 65.7% and 45.4% at 2 years [Fig.1B], which was also concurrent with a phase II trial from Elias et al. showing a disease-free survival of almost 50% at 2 years25. Before the combination treatment of CCRS + HIPEC became available, no long-term survivors were reported for PC of CRC. Table 1 gives an overview of survival data reached with CCRS + HIPEC. If an R0/1 resection can be reached by CCRS, overall survival rates for CCRS + HIPEC are

Page 102: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

93

comparable to the 5-year overall survival rates of 35-60% reported by several large single- and multi-institutional experiences after curative liver resection for colorectal liver metastases26-31. This could be considered as level 1c evidence for this treatment strategy.

In this Belgian registry, a significant difference is found in OS as well as DFS when PCI is dichotomized between patients with PCI≤15 and patients with PCI>15 [Fig.2A, 2B]. This concurs with earlier papers published by Elias and Glehen et al. showing that the PCI, with an arbitrary cut-off at 15, had a significant impact on overall survival (p=0.019) and completeness of cytoreduction was found to be the principal independent prognostic indicator (p<0.001) 17. Besides the dichotomized PCI, resection small bowel (p=0.004), post-operative EAC (p=0.0005) and a lesion size>5cm (p=0.043) are significantly correlated with OS. Due to the low number of deceased patients and the low number of patients for some risk factors, results for OS need to be interpreted with care. For DFS, there is a significant correlation with transfusion (p=0.018) and a trend was observed for the resection of small bowel at debulking. In the largest HIPEC series reported by Elias et al. positive independent prognostic factors identified by multivariable analysis were complete debulking (R0/1 or CC-0), a low PCI, no invaded lymph nodes and the use of adjuvant chemotherapy20.

The conference on Peritoneal Surface Malignancies in Milan in 2006 attempted to reach a methodological consensus on the drugs to be used intraperitoneally for HIPEC, but did not succeed32. This illustrates the complexity of this topic. At present Mitomycin C (MMC) is still the most frequently used drug intraperitoneally24. Verwaal et al. performed the only prospective randomized phase III study comparing CCRS + HIPEC with palliative surgery + systemic chemotherapy, which provided compelling evidence that CCRS + HIPEC improves the survival in patients with PC of colorectal origin33,34. In this randomized trial MMC was used as intra-peritoneal drug, providing a high level of evidence (level 1b) for the efficacy of MMC24. A frequent criticism on this trial is the fact that patients

Page 103: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

94

in the control arm received 5FU/Leucovorin. At present, median OS of 16.8-23.9months are reported with modern chemotherapy regimens22,35. On the other hand, in a retrospective analysis by Hompes et al patients with unresectable PC of CRC, who received modern 5-FU-based systemic therapy combined with oxaliplatin or irinotecan either with or without bevacizumab, only rarely reached a survival of 2 years. Of course, these patients all had unresectable and thus extensive PC36. Nevertheless, a study by Franko et al recently confirmed the results of the randomize trial by Verwaal et al: their analysis stated that even if contemporary chemotherapy was used in the control arm the benefit in survival with CCRS + HIPEC is maintained 35. The characteristics of Oxaliplatin and MMC are summarized in Table 6. Both drugs have a large molecular weight32, resulting in high intraperitoneal drug concentrations during HIPEC, but with limited systemic absorption and toxicity37. They have a comparable tissue penetration depth and are both potentiated by hyperthermia32. Both drugs are alkylating agents, thus interfering with DNA and DNA-synthesis and their function is not cell cycle dependant10,38,39. For MMC the advised intraperitoneal dose is 35mg/m² with a perfusion duration of 90 minutes, resulting in an AUC-ratio [perfusate/plasma] of 10.1 +/- 4.6, whereas for Oxaliplatin the advised dose is 460mg/m² during only 30 minutes, resulting in an AUC-ratio [perfusate/plasma] of 12.8 +/-2.912,25,32,37. The intraperitoneal half-life for MMC is 49 minutes and 29.5 minutes for Oxaliplatin11,38. Elias et al. objectified the high uptake of Oxaliplatin in local tissues after HIPEC: 339ng/mg in tumoral tissue and 392ng/mg in the peritoneum12. Interestingly, besides this excellent regional exposure with intraperitoneal administration of Oxaliplatin, an early experimental pharmacokinetic study by Pestieau et al. showed the highest Oxaliplatin concentrations were found in colonic tissues10. MMC is accepted to have a pharmacokinetic profile resulting in rapid tissue concentration in residual tumor deposits and the peritoneum over prolonged periods of time39,

Page 104: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

95

but it causes severe neutropenia in 28% of patients12,25,38,40, which is not the case for Oxaliplatin. So, although the systemic absorption of intraperitoneal MMC is limited, the severe neutropenia points out to a significant systemic accumulation. This is probably due to the fact that toxicity is not only the result of the systemic absorption, but also of the metabolization of the chemotherapeutic agent. MMC is predominantly metabolized in the liver38, whereas oxaliplatin is not subjected to CYP450-mediated metabolism41. Urinary excretion is the predominant route of platinum elimination and tissue binding and renal elimination contribute equally to the clearance of ultrafilterable platinum from plasma41. Therefore, maintaining an adequate urinary output is crucial to prevent renal insufficiency. Finally, Oxaliplatin has a proven systemic efficacy in CRC and a synergistic activity when used in combination with 5-FU42-45. This is, of course, the reason why in many centers HIPEC with intraperitoneal Oxaliplatin is combined with the intravenous administration of 5FU/Leucovorin.

Conclusion

CCRS followed by HIPEC with Oxaliplatin for PC of colorectal origin can be implemented with acceptable morbidity. Long-term DFS and OS should allow to improve selection of patients who will benefit from this extensive surgical approach in view of prolonged survival with modern palliative chemo- and biological therapy.

Acknowledgements This trial was performed with a research grant from Sanofi-Aventis.

Page 105: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

96

Table 1: OS after CCRS + HIPEC

Author Journal ip chemo

N total

N syst.M+

R0/1 R2a (≤2.5mm)

R2b (>2.5mm)

Median OS (months)

Overall Survival

1y. 3y. 5y.

Verwaal1 Ann Surg Oncol 2005

MMC 117 0% 50.4% 37.6% 12% 21.8 75% 28% 19%

Verwaal34 Ann Surg Oncol 2008

MMC 54 0% 41% 41% 18% 22.2 / / R1: 45% R2: 5%

Franko35 Cancer 2010

MMC 67 Yes (%?) 91% 9.0% 34.7 / / /

Elias22 J Clin Oncol 2009

Oxali 48 0% 100% 0% 0% 62.7 / / 51%

Elias19 Ann Surg 2010

MMC or Oxali

440 15.9% 100% 0% 0% / / / 33%

Elias20 J Clin Oncol 2010

MMC or Oxali

523 15% 85% 10% 5% / 81% 41% 27%

Page 106: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

97

Table 2: Patient Characteristics

N = 48 patients

M/F 17/31

Primary tumor Previously resected 35 72.9%

PC present at primary tumor 36 75%

Localization: - Appendix

- Caeco-ascending colon

- Transverse colon

- Descending colon

- Rectosigmoid

9

12

2

5

20

Differentiation: - Well

- Moderately

- Poor

- Not specified

7

26

12

3

Mucinous cell type 19 39.6%

Page 107: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

98

Table 3: Surgical procedures performed during CCRS + HIPEC in 48 patients with PC from CRC

Median N organs resected 2 [2-6]

- Anterior resection

- Total colectomy

- Segmentary Small Bowel Resection

22/48 (45.8%)

4/48 (8.3%)

6/48 (12.5%)

Anastomoses - Median N

- Low localization

- Timing: after HIPEC

1 [0-6]

13/48 (27.1%)

32/48 (82.1%)

Diversion

- ileostomy

- colostomy

16 (33.3%) 11 (22.9%)

5 (10.4%)

Page 108: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

99

Table 4: Per- and postoperative data of CCRS + HIPEC in 48 patients with CRC

IAC = intra-abdominal complications; EAC = extra-abdominal complications

Median OP-time 460 min. [125-840]

Median Blood Loss

475 cc [2-6000]

Postoperative mortality

0

Postop. Complications

52.1%

N patients

%

IAC (N=18)

- Prolonged ileus

(food intollerance 9 days[2-56])

- Anastomotic leakage

- Bleeding

- Bowel perforation

- RV-fistula

- Abscess

11

5

3

1

1

1

22.9%

10.4%

6.3%

2.1%

2.1%

2.1%

EAC (N=16)

- Pulmonary

- Cardiac

- Renal/Urologic

- Hematologic

- other

6

1

6

1

6

12.5%

2.1%

12.5%

2.1%

12.5%

N reoperations 10 20.8%

Median hospital stay

20 days [5-65]

Page 109: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

100

Table 5: univariable results for OS and DFS

Page 110: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

101

Table 6: MMC and oxaliplatin as intraperitoneal drugs for HIPEC

MMC ip Oxaliplatin ip refs.

Molecular Weight (Dalton) 334.3 397.3 [32]

Tissue penetration depth 2000µ 1-2mm [32]

Mechanism

(Note: lysis of colon cancer cell lines = dose-related in CRC)

Large MW !

Antitumoral AB:

Alkylating agent (tetrazine)+ production free radicals

NOT cell cycle dependant

Large MW !

Biotranformation, followed by interaction with DNA (alkylating), thus disturbing DNA-

synthesis

NOT cell cycle dependant

[10,37,38 + pharma-ceutical

compendium]

AUCratio (perfusate/plasma) 10.1 +/-4.6 12.8 +/-2.9 [12,25,36]

Advised intraperitoneal dose 35mg/m² 460mg/m² [32]

Duration of perfusion 90min.

First 50% of dose, followed by 25% of dose at 30 and 60 minutes

30min [32]

Perfusate solution Isotonic salt solution Dextrose 5% [25,32,37]

t1/2 in perfusate 49min.* 29.5min.* [11,37]

Potentiation

by hyperthermia

Yes Yes [32]

Tissue concentration NS

“Rapid tissue concentration over prolonged time period”

High uptake in local tissues

[Cmax perit.=25x Cmax blood]

- Tumor 339ng/mg

- Peritoneum 392 ng/mg

[12,38]

Toxicity Grade 2-3 toxicity: 65%

Severe neutropenia: 28%

Fistulae: 17.6%

Grade 2-3 morbidity: 40%

No neutropenia

Fistulae: 10%

[12,25,37,39]

Page 111: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

102

Fig.1A: Overall Survival after CCRS + HIPEC (in months). Dashed lines represent the pointwise 95%CI.

Page 112: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

103

Fig.1B: Disease-Free Survival after CCRS + HIPEC (in months). Dashed lines represent the pointwise

95%CI.

Page 113: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

104

Fig.2A: Influence of the PCI on Overall Survival

Page 114: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

105

Fig.2B: Influence of the PCI on Disease-Free Survival

Page 115: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

106

References

1. Verwaal V, Zoetmulder F. Long-Term Survival of Peritoneal carcinomatosis of Colorectal Origin; Ann Surg Oncol 2005; 12: 65-71.

2. Cararo P, Segala M, Cesana D. Obstructing colonic cancer: failure and survival patterns over ten-year follow-up after one-stage curative surgery. Dis Colon Rectum 2001; 44: 243-250.

3. Jayne D, Fook S, Loi C. Peritoneal carcinomatosis from colorectal cancer. Br J Surg 2002; 89: 1545-1550.

4. Russell A, Pelton J, Reheis C. Adenocarcinoma of the colon: an autopsy study with implications for new therapeutic strategies. Cancer 1985; 56: 1446-1451.

5. Koppe M, Bleichrodt R. Peritoneal Carcinomatosis of Colorectal Origin: Incidence and Treatment Strategies; Ann Surg 2006; 243: 212-222.

6. Sadeghi B, Arvieux C, Glehen O. Peritoneal carcinomatosis from non-gynecological malignancies: results from the EVOCAPE 1 multicentric prospective study. Cancer 2000; 88: 358-63.

7. Shen P, Levine E, Hall J. Factors predicting survival after intraperitoneal hyperthermic chemotherapy with mitomycin C after cytoreductive surgery for patients with peritoneal carcinomatosis. Arch Surg 2003; 138: 26-33.

8. Sugarbaker P. Management of peritoneal-surface malignancy: the surgeon’s role; Langenbecks Arch Surg 1999; 384: 576-587.

9. Ragnhammar P, Hafstrom L. SBU-group. Swedish Council of technology assessment in health care. A systematic overview of chemotherapy effects in colorectal cancer; Acta Oncol 2001; 40: 282-308.

10. Pestieau S, Sugarbaker P. Pharmacokinetics of intraperitoneal Oxaliplatin: experimental studies; J Surg Oncol 2001; 76: 106-114.

11. Mahteme H, Pahlman L. Systemic exposure of the parent drug oxaliplatin during hyperthermic

Page 116: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

107

intraperitoneal perfusion; Eur J Clin Pharmacol 2008; 64: 907-911.

12. Elias D, Bonnay M. Heated intra-operative intraperitoneal oxaliplatin after complete resection of peritoneal carcinomatosis: pharmacokinetics and tissue distribution; Ann Oncol 2002; 13: 267-272.

13. Jacquet P, Sugarbaker P. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. In: Sugarbaker PH, editor. Peritoneal carcinomatosis: Principles of management. Editors. Boston: Kluwer Academic Publishers 1996: 359–37.

14. Sugarbaker PH. Peritonectomy procedures. Surg Oncol Clin N Am 2003; 12: 703-727; XIII.

15. Harrell F. Regression modeling strategies. New York: Springer, 2001

16. Heinze G, Gnant M, Schemper M. Exact logrank test for unequal follow-up. Biometrics 2003; 59: 1151-1157.

17. Elias D, Blot F. Curative treatment of peritoneal Carcinomatosis arising from colorectal cancer by complete resection and intraperitoneal chemotherapy; Cancer 2001; 92: 71-76.

18. van Leeuwen B, Graf W, Pahlman L. Swedish experience with peritonectomy and HIPEC. HIPEC in peritoneal carcinomatosis. Ann Surg Oncol 2007; 15: 745-753.

19. Elias D, Glehen O, Pocard M. A comparative study of complete cytoreductive surgery plus intraperitoneal chemotherapy to treat peritoneal dissemination from colon, rectum, small bowel and nonpseudomyxoma appendix. Ann Surg 2010; 251: 896-901.

20. Elias D, Gilly F, Boutitie F. Peritoneal 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.

Page 117: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

108

21. Glehen O, Elias D, Deraco M. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multiinstitutional study; J Clin Oncol 2004; 22: 3284-3292.

22. Elias D, Lefevre JH, Chevalier J. Complete Cytoreductive Surgery plus Intraperitoneal Chemohyperthermia with Oxaliplatin for peritoneal Carcinomatosis of Colorectal Origin. J Clin Oncol 2009; 27: 681-685.

23. Elias D, Pocard M. Treatment and prevention of peritoneal carcinomatosis from colorectal cancer. Surg Oncol Clin N Am 2003; 12: 543-559.

24. Cao C, Yan T, Black D, Morris D. A systematic review and meta-analysis of cytoreductive surgery with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 2009; 16: 2152-2165.

25. Elias D, Pocard M, Goere D. HIPEC with oxaliplatin in the treatment of peritoneal carcinomatosis of colorectal origin. Cancer Treat Res 2007; 134: 303-318.

26. Andres A, Majno PE, Morel P. Improved long-term outcome of surgery for advanced colorectal liver metastases: reasons and implications for management on the basis of a severity score. Ann Surg Oncol 2008; 15: 134–143.

27. Arru M, Aldrighetti L, Castoldi R. Analysis of prognostic factors influencing long-term survival after hepatic resection for metastatic colorectal cancer. World J Surg 2008; 32: 93–103.

28. Choti MA, Sitzmann JV, Tiburi MF. Trends in long-term survival following liver resection for hepatic colorectal metastases. Ann Surg 2002; 235: 759–766.

29. Zakaria S, Donohue JH, Que FG. Hepatic resection for colorectal metastases: value for risk scoring systems? Ann Surg 2007; 246: 183–191.

Page 118: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

109

30. House M, Ito H, Fong Y. Survival after hepatic resection for metastatic colorectal cancer: trends in outcomes for 1,600 Patients during two decades at a single institution. J Am Coll Surg 2010; 210: 744-752.

31. Maggiori L, Elias D. Curative treatment of colorectal peritoneal carcinomatosis: current status and future trends. Eur J Surg Oncol 2010; 36: 599-603.

32. Kusamura S, Elias D, Baratti D. Drugs, carrier solutions and temperature in hyperthermic intraperitoneal chemotherapy. J Surg Oncol 2008; 98: 247-252.

33. Verwaal V, van Ruth S, De Bree E. Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 2003; 21: 3737-3747.

34. Verwaal V, Bruijn S, Boot H. 8-Year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol 2008; 15: 2426-2432.

35. Franko J, Ibrahim Z, Gusani N. Cytoreductive surgery and hyperthermic intraperitoneal chemoperfusion versus systemic chemotherapy alone for colorectal peritoneal carcinomatosis. Cancer 2010; 116: 3756-3762.

36. Hompes D, Boot H, Verwaal V. Unresectable peritoneal carcinomatosis from colorectal cancer: a single center experience. J Surg Oncol 2011; 104: 269-273.

37. van Ruth S, Mathôt R, Sapridans R. Population pharmacokinetics and pharmacodynamics of mitomycin C during intra-operative hyperthermic intraperitoneal chemotherapy. Clin Pharmacokinet 2004; 43:131-143.

38. van Ruth S, Verwaal V, Zoetmulder F. Pharmacokinetics of intraperitoneal mitomycin C. Surg Oncol Clin N Am 2003; 12: 771-780.

Page 119: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

110

39. Lambert L, Armstrong T, Mansfield F. Incidence, risk factors and impact of severe neutropenia after hyperthermic intraperitoneal mitomycin C. Ann Surg Oncol 2009; 16: 2181-2187.

40. Verwaal V, van Tinteren H, Ruth S. Toxicity of cytoreductive surgery and hyperthermic intra-peritoneal chemotherapy. J Surg Oncol 2004; 85: 61-67.

41. Graham M, Lockwood G, Greenslade D. Clinical pharmacokinetics of oxaliplatin: a critical review. Clin Cancer Res 2000; 6: 1205-1218.

42. Soulié P, Raymond E, Brienza S. Oxaliplatin: the first DACH platinum in clinical practice. Bull Cancer 1997; 84: 665-673.

43. de Gramont A, Figer A, Seymour M. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 2000; 18: 2938-2947.

44. Makatsoris T, Kalafonos H, Aravantinos G. A phase II study of capecitabine plus oxaliplatin (XELOX): a new first-line option in metastatic colorectal cancer. Int J Gastrointest Cancer 2005; 35: 103-109.

45. Scheithauer W, Kornek G, Raderer M. Randomized multicenter phase II trial of two different schedules of capecitabine plus oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 2003; 21: 1307-1312.

Page 120: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

111

III

2. The use of Oxaliplatin or

Mitomycin C in HIPEC treatment

for peritoneal carcinomatosis from colorectal cancer: a comparative study

Hompes D, D’Hoore A, Mirck B, Fieuws S,

Bruin S, Verwaal V submitted to Eur J Surg Oncol.

Page 121: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

112

Page 122: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

113

The use of Oxaliplatin or Mitomycin C in HIPEC treatment for peritoneal carcinomatosis from

colorectal cancer: a comparative study.

D.Hompes1,2, A.D’Hoore2, B.Mirck1, A.Wolthuis2, S.Fieuws3, S.Bruin1, V.Verwaal1

1 Department of Surgical Oncology, the Netherlands Cancer Institute/ Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands 2 Department of Abdominal Surgery, University Hospitals Gasthuisberg, Leuven, Belgium. 3 I -Biostat, Katholieke Universiteit Leuven and Universiteit Hasselt, Belgium

Introduction Patients with limited peritoneal carcinomatosis (PC) as the only metastatic location of colorectal cancer (CRC) can benefit from radical surgery and HIPEC. This combined treatment strategy can result in long-term survival for these patients1-6. Completeness of cytoreductive surgery is a major determinant for long-term outcome. If a macroscopic complete cytoreduction (CC-0 or R0/1) can be achieved a 5-year overall survival of 40-50% has been documented.

At present, both Oxaliplatin and Mitomycin C (MMC) are used as intraperitoneal chemotherapy agents in HIPEC for PC of CRC. These agents are suitable for intraperitoneal use as they have a large molecular weight, resulting in high intraperitoneal drug concentrations during HIPEC, with limited systemic absorption and toxicity7,8. Oxaliplatin has become standard systemic treatment in CRC9-12, whereas MMC is only used as salvage treatment in advanced progressive CRC13-16. In literature, results on survival in HIPEC series for CRC are comparable for both agents [Table 1].

There are no prospective trials available comparing peri-operative complications and long-term outcome after HIPEC

Page 123: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

114

with Oxaliplatin versus MMC. Therefore, the choice for Oxaliplatin or MMC is mainly based on hospital tradition, rather than on comparison of efficacy and toxicity. This trial compares two cohorts of patients treated in 2 different centers: in one center HIPEC has been consistently performed with MMC, whereas the other center uses Oxaliplatin as the intraperitoneal agent. The aim is to assess differences in toxicity profile and long-term outcome.

Patients and Methods Patient cohorts

In this study 2 cohorts of patients treated with complete cytoreductive surgery (CCRS) and HIPEC for PC of CRC are compared. One patient cohort consists of patients treated at the Netherlands Cancer Institute / Antoni van Leeuwenhoek-hospital (the Netherlands) [NKI-AVL] between February 2004 and December 2006. The other patient cohort was treated at the University Hospital Gasthuisberg in Leuven (Belgium) [UZL] between June 2006 and September 2010. Both hospitals are HIPEC centers. NKI-AVL is a comprehensive cancer center with only specialist training facilities, whereas UZL is a large university hospital with basic as well as advanced medical training facilities. Both centers function as tertiary referral centers. The HIPEC protocols from both centers were approved by the respective Medical Ethics Committees and all patients had a written informed consent. Traditionally, UZL only offers HIPEC to patients in whom complete cytoreduction (R0/1) can be achieved, whereas NKI-AVL sometimes performes HIPEC in a selected group of patients in whom only incomplete cytoreduction (R2a/R2b) can be reached. To enable adequate comparison between both cohorts, this trial only includes patients in whom a macroscopically complete cytoreduction could be reached.

Page 124: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

115

Procedure After a full exploration of the abdominal cavity complete

cytoreductive surgery (CCRS) is performed. When macroscopically complete cytoreduction (CC-0) has been achieved the HIPEC procedure is started. When the hyperthermic perfusion reaches a steady state of 41-42°C the intraperitoneal drug is added to the perfusion. At the NKI-AVL HIPEC is performed with MMC as the intraperitoneal drug at a dose of a dose of 35mg/m² during 90 minutes. At the UZL CCRS is followed by HIPEC with Oxaliplatin as the intraperitoneal drug at a dose of 460mg/m² during 30 minutes. One hour before starting the HIPEC procedure with Oxaliplatin, Folinic Acid 20mg/m² and 5-Fluorouracil 400mg/m² (in 250ml saline) are administered intravenously to enhance the effect of the Oxaliplatin. Anastomoses are performed after the HIPEC procedure was finalized.

Follow-up

All patients were followed up intensely with comparable clinical and radiological assessment protocols. They were seen at the outpatient clinics every 3 months in the first 2 years after surgery, every 6 months in the following 2 years and once a year from the fifth postoperative year on. At each visit a blood sample was taken to determine the CEA level and a chest X-ray and liver ultrasound were performed. One to two CT thorax/abdomen scans were performed per year. Colonoscopy to detect local recurrence and/or metachronous primary colonic cancer was repeated every 2 to 3 years. Data processing and statistics

Data on patients and tumor characteristics, surgical procedure data, intra- and postoperative mortality and morbidity data and oncologic follow-up data on disease recurrence and survival data were prospectively registered in the respective institutional electronic databases of NKI-AVL and UZL. Morbidity data were scored according to National Cancer Institute-Common Terminology Criteria for Adverse Events

Page 125: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

116

guidelines (NCI-CTC AE version 4). To assess the extent of disease, defined as the extensiveness of PC in the abdominal cavity, the NKI-AVL applied the 7 region count as described by Verwaal et al.17, whereas UZL applied the Peritoneal Carcinomatosis Index (PCI) according to Sugarbaker et al.18. To enable adequate comparison between both study groups the UZL data on extent of disease were transformed into the 7 region count system using the standardized transcription17. Mann-Whitney U and (Fisher’s) exact tests are used to compare patient and tumor characteristics, peroperative data and morbidity data between both groups. A multivariable logistic regression model is used to compare the complication rate between both groups after correction for extent of disease (7 region count as a continuous predictor). Kaplan-Meier estimates are used to construct the overall and recurrence-free survival curve, yielding the median and interquartile range (IQR) time. A multivariable Cox regression model is used to compare the overall survival (OS) and recurrence-free survival (RFS) between both groups after correction for extent of disease. Deaths and recurrences are considered as events in the definition of RFS. A survival curve derived from this model is constructed comparing both groups at a mean 7 region count. From the logistic and Cox regression model, odds ratios (OR) and hazard ratios (HR) are reported, respectively, with a 95% confidence interval (95%CI). To quantify the median time to last follow-up19 a Kaplan-Meier estimate is used, censoring patients at moment of death. All analyses have been performed using SAS software, version 9.2 of the SAS System for Windows. Copyright © 2002 SAS Institute Inc. SAS and all other SAS Institute Inc. product or service names are registered trademarks or trademarks of SAS Institute Inc., Cary, NC, USA.

Results Patient and Tumor Characteristics [Table 2]

Fifty-six NKI-AVL patients were included in the MMC-group and 39 UZL patients were included in the Oxaliplatin-

Page 126: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

117

group. Table 2 summarizes patient and primary tumor characteristics. No statistically significant differences in patient characteristics were found across groups. Median age at surgery was 62.2 years [range 24.1-74.5] in the Oxaliplatin-group and 58.4 years [range 32.5-77.4] in the MMC-group. In the Oxaliplatin-group primary tumors have a more advanced T-stage (p=0.027) and a higher incidence of node-positivity (p=0.003). Mucinous tumor cell type was found in 46.2% of Oxaliplatin-patients and 48.2% of MMC-patients. PC occurred synchronously in 61.5% and 57.1% of patients in the Oxaliplatin-group and the MMC-group, respectively. It should be noted that for 1 patient of the Oxaliplatin-group the indication for CCRS+HIPEC was tumor positive cytology in ascites. Finally, 3 patients in the Oxaliplatin-group had liver metastases in their previous history. All 3 patients received chemotherapy in a neo-adjuvant setting and underwent curative liver resection. Furthermore, 1 patient had a very small liver metastasis (4mm in diameter), which was discovered and excised at CCRS+HIPEC.

Intra-operative Procedure Data [Table 3] The median 7 region count according to Verwaal et al.17 was scored 4 [range 0-7] for Oxaliplatin-patients versus 2.5 [range 1-6] for MMC- patients, which is a significant difference (p=0.004). Median duration of the total surgical procedure was comparable across groups: 380 minutes [range 177-660 minutes] in the Oxaliplatin-group and 360 minutes [range 150-600 minutes] in the MMC-group. On the other hand, median intra-operative blood loss was significantly different (p<0.001), i.e. 650ml [0-6000ml] in Oxaliplatin-patients versus 1230ml [range 0-5300ml] in MMC-patients. Postoperative complications [Table 3] Postoperative complications are listed in Table 3. The length of hospital stay is significantly longer for Oxaliplatin-patients (p=0.003). There is no postoperative 60-day mortality. In the Oxaliplatin-group 59% of patients have postoperative

Page 127: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

118

morbidity, with grade 3-4 serious adverse events in 48.7%, whereas this is 75% and 35.7% respectively for the MMC-group. Intra-abdominal complications (IAC) occur in 30.8% of Oxaliplatin-patients and 23.2% of MMC-patients. Subsequent surgical reinterventions are needed in 20.5% and 8.9%, respectively. Those differences are not statistically significant (p=0.64 and p=0.13). Extra-abdominal complications (EAC) are registered in 48.7% of Oxaliplatin-patients and 64.3% of MMC-patients (p=0.13). No Oxaliplatin-associated neurotoxicity is registered and in only 2 Oxaliplatin-patients a temporary 2- to 3-fold increase in creatinine occurs (grade 2 serious adverse event (SAE)). In 1 MMC-patient (1.8%) a grade 2 increase in liver function values is registered. Oxaliplatin-patients develop no hematologic complications, whereas neutropenia or leucopenia is registered in 15 of 56 MMC-patients (26.8%). This is statistically significant (p<0.001), but only 1 grade 3 leucopenia occurred, without clinical consequences. On univariate analysis, overall postoperative complications are not significantly different for Oxaliplatin and MMC. However, after statistical correction for the extent of disease (which is the only tumor-related parameter that is significantly different between both groups), the overall postoperative complication rate is significantly higher in the MMC-group (OR=2.68 (95%CI: 1.04-6.91), p=0.04), with a comparable IAC rate (OR=0.78 (95%CI: 0.30-2.03), p=0.61), but a tendency towards more EAC in the MMC-group (OR=2.23 (95%CI: 0.91-5.43), p=0.079).

Overall and Recurrence-Free Survival [Fig.1 & 2] Median follow-up time is significantly shorter for Oxaliplatin patients than for MMC-patients: 2.8 years compared to 5.1 years (derived from statistical analysis, censoring patients at moment of death). Median RFS is 12.2 months [IQR: 7.2-undefined] in the Oxaliplatin-group and 13.8 months [IQR: 7.0-25.8] in the MMC-group (p=0.87). Median OS is 37.1 months [IQR: 22.4-52.8] for Oxaliplatin-patients and 26.5 months [IQR: 16.9-64.8] for MMC-patients (p=0.45). Because

Page 128: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

119

the 7 region count is the only tumor-related parameter that is statistically different across both study groups (p=0.002), the survival analysis was corrected for extent of disease. Logistic regression analysis shows OS (HR=1.37 (95%CI: 0.74-2.54), p=0.32) [Fig.1] and RFS (HR=1.24 (95%CI: 0.75-2.05), p=0.39) [Fig.2] are not significantly different.

Discussion The debate on the treatment of resectable PC from CRC

with CCRS+HIPEC is slowly narrowing down to the details of the treatment modality. Oxaliplatin and MMC are alkylating chemotherapeutic agents, interfering with DNA and DNA-synthesis without being cell cycle dependant20-22. Because Oxaliplatin and MMC have a large molecular weight (397.3 Dalton and 334.3 Dalton, respectively), they can reach high intraperitoneal drug concentrations during HIPEC, with limited systemic absorption7,21. Furthermore, they have enhanced cytotoxicity under hyperthermia and a maximal tissue penetration depth of 2mm7. These characteristics make Oxaliplatin and MMC suitable as intraperitoneal agents for HIPEC. The recommended intraperitoneal dose for Oxaliplatin is 460mg/m² and perfusion time is limited to 30 minutes, whereas the advised dose for MMC is 35mg/m² with a perfusion duration of 90 minutes7,8,23,24. The intraperitoneal half-life for Oxaliplatin is 29.5 minutes and 49 minutes for MMC22,25 [Table 4]. In order to potentiate the Oxaliplatin activity, patients in the Oxaliplatin-group receive intravenous 5-Fluorouracil and Folinic Acid approximately 1 hour before starting the HIPEC procedure, to bathe the tumor and healthy tissue before HIPEC26. The 5- fluorouracil is administered intravenously, because it cannot be mixed with oxaliplatin inside the peritoneal cavity (because of pH incompatibility)27. In contrast to MMC, Oxaliplatin it is not stable in chloride-containing solutions: it can only be administered in a 5% dextrose perfusion solution28. This results in biochemical disturbances, which are manageable with a compensation of

Page 129: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

120

the sodium loss into the perfusate and a good control of hyperglycemia28,29. Oxaliplatin is easily detectable and therefore thouroughly studied for its distribution20,23. Besides an excellent regional exposure, high drug concentrations were found in colonic tissues20. This was later objectified by Elias et al. who reported a high uptake of Oxaliplatin in local tissues after HIPEC: 339ng/mg in tumoral tissue and 392ng/mg in the peritoneum23.

In this comparative cohort study a 3-year OS of 54.0% is observed for Oxaliplatin-patients and 3- and 5-year OS for MMC-patients is 41.1% and 25.6%. Median OS is 37.1 months [IQR: 22.4-52.8] for Oxaliplatin-patients and 26.5 months [IQR: 16.9-64.8] for MMC-patients (p=0.45). The survival analysis (corrected for extent of disease) shows curves that appear slightly in favour of the Oxaliplatin-group, but statistical analysis shows no difference in OS (HR=1.37 (95%CI: 0.74-2.54), p=0.32) [Fig.1] and RFS (HR=1.24 (95%CI: 0.75-2.05), p=0.39) [Fig.2] for the Oxaliplatin- and MMC-group. After CCRS+HIPEC for patients with PC from CRC the 5-year OS reported in literature can vary between 20 and 50%, with median survivals ranging from 22 to almost 5 years 1-6,30,31 [Table 1]. This wide variation in survival data is due to multiple variations in factors such as HIPEC indication, patient selection, PC tumor load (extent of disease), completeness of cytoreduction1-6,30,32-35, administration of neo-adjuvant systemic chemotherapy2-4,6 and presence of systemic metastases2-4. Obviously, the highest survival rates were reported in patients who had a complete cytoreducion and had few intra-operative complications. The extent of PC correlates well with the probability of achieving a complete cytoreduction17. It is acknowledged to be the next important factor to significantly influence outcome4,33,35. In our comparative study the extent of disease is the only tumor-related parameter that is significantly different between the Oxaliplatin- and MMC-group (p<0.004). Therefore, it was taken into account in the statistical analysis, to enable

Page 130: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

121

adequate comparison between the HIPEC results in both patient cohorts.

This study shows comparable overall morbidity as well as grade 3-5 toxicity for CCRS+HIPEC with Oxaliplatin and MMC. Univariate analysis shows no statistical differences across both groups for intra-abdominal complications (IAC) (p=0.64), extra-abdominal complications (EAC) (p=0.15) or the need for surgical reintervention (p=0.51) [Table 3]. But, after correction of the toxicity data for extent of disease with a logistic regression model, there is a higher overall complication rate in the MMC-group (OR=2.68 (95%CI: 1.04-6.91), p=0.04), with a tendency towards more EAC (OR=2.23 (95%CI: 0.91-5.43), p=0.079). The IAC rate remains comparable across groups (OR=0.78 (95%CI: 0.30-2.03), p=0.61). Table 5 presents literature data on morbidity and mortality after CCRS + HIPEC with Oxaliplatin and MMC intraperitoneal drug. For the interpretation of these data, the above mentioned remarks on variation in patient selection, tumor burden and completeness of cytoreduction should also be taken into account. Larger studies on Oxaliplatin and MMC as an intraperitoneal agent report a mortality of up to 6% and grade 3-5 serious adverse events in 31-66% of patients. A need for surgical reintervention is reported in 11-30% of patients4,5,23,30,32. Gastro-intestinal tract complications are reported to occur in 9-23% of patients3,4,23,36,37, but unfortunately the rate of anastomotic leakage after CCRS+HIPEC is often not clearly specified. Overall, the occurrence of fistulae after CCRS+HIPEC is reported to be 17.6% and 10% for MMC and Oxaliplatin, respectively22-24,36. MMC is known to cause severe neutropenia in 28% of patients22-24,36. In this paper 26.8% of patients were reported to have neutropenia or leucopenia. Despite the very limited systemic absorption of intraperitoneal MMC, there is a systemic effect which is probably due to systemic accumulation. This accumulation results not only from systemic absorption, but also the hepatic metabolization of MMC22. For Oxaliplatin the

Page 131: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

122

predominant route for the elimination is by urinary excretion. Furthermore, tissue binding and renal elimination contribute equally to the clearance of platinum from plasma38. When administered systemically, Oxaliplatin is known to cause severe neurotoxicity39,40, but our study reports no neurotoxicity.

Finally, as the intraperitoneal chemotherapy agents are

administered during the HIPEC-procedure in the operating room, safety for operating room personnel and nursing staff is evidently an important issue. Safety studies have demonstrated that MMC41,42 and Oxaliplatin43 do not represent a detectable safety hazard to the surgeon or other personnel in the operating room, when the recommended protective garment is used and the safety considerations are followed. In conclusion, based on the available data on safety and efficacy of CCRS+HIPEC in patient with PC from CRC, no strong plea can be made for the use of either Oxaliplatin or MMC. Although Oxaliplatin has become standard systemic treatment in CRC9-12, no clear benefit in OS and RFS can be demonstrated for HIPEC in this trial or in literature. MMC often results in neutropenia, whereas Oxaliplatin does not, but the clinical significance of this finding is minimal. Oxaliplatin results in equally high morbidity rates as MMC. Indirect arguments for which one might prefer Oxaliplatin over MMC as an intraperitoneal drug are the proven efficacy of (systemic) Oxaliplatin in CRC, the high Oxaliplatin concentrations in the targeted colonic tissues after HIPEC and the fact that Oxaliplatin is not metabolized hepaticly. Also, in a time of cost and facility restrictions, the shorter HIPEC perfusion time (only 30 minutes) could be an important argument to use Oxaliplatin.

Page 132: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

123

Table 1: Survival after CCRS and HIPEC with Oxaliplatin or MMC

Page 133: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

124

Table 2: Patient & tumor characteristics and operative data

Page 134: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

125

Table 3: Intra- and postoperative Serious Adverse Events (SAE)

Page 135: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

126

Table 4: Pharmacokinetics of CCRS and HIPEC with Oxaliplatin or MMC

Author/Journal/Year Mahteme H25

Eur J Clin

Pharmacol 2008

Elias D23

Ann Oncol 2002

van Ruth S8

Clin Pharmacokinet 2004

van Ruth S22

Surg Oncol Clin N Am

2003

Type of study Prospective Prospective Prospective Prospective

N patients 8 20 47 118

N CRC 4 6 22 -

Median PCI [range] 21 [6-37] 21 [4-31] - -

CC-0 4 20 - -

CC-1 (<2.5mm) 3 0 - -

CC-2 (<2.5cm) 1 0 - -

CC-3 (>2.5cm) 0 0 - -

R0/1 4 20 17 -

R2a (≤2.5mm) 3 0 24 -

R2b (>2.5mm) 1 0 6 -

Chemotherapeutic agent Oxaliplatin Oxaliplatin MMC MMC

Dose (mg/m²) 460 260 460* 35 15 40*

Duration HIPEC (min.) 30 30 90 90

Perfusate t1/2 (min.) 29.5 30 - 49

Plasma t1/2 (min.) 24.7 - - 76

AUC ratio 12.8+/-2.9 - 10.1+/-4.6 10.9-13.2

Cmax (peritoneum/plasma) 28.4 25 - 22.7 [at 35mg/m²]

% Absorbed 48.4+/-7.6 - - -

* dose escalation trials

Page 136: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

127

Table 5: Complications after CCRS and HIPEC with Oxaliplatin or MMC

Page 137: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

128

Fig.1: OS after CCRS + HIPEC with Oxali versus MMC (after correction for extent of disease)

Page 138: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

129

Fig.2: RFS after CCRS + HIPEC with Oxali versus MMC (after correction for extent of disease)

Page 139: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

130

References 1. Verwaal V, Bruin S, Boot H. 8-year follow-up of

randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol 2008; 15: 2426-2432.

2. Franko J, Ibrahim Z, Gusani N. Cytoreductive surgery and hyperthermic intraperitoneal chemoperfusion versus systemic chemotherapy alone for colorectal peritoneal carcinomatosis. Cancer 2010; 116: 3756-3762.

3. Elias D, Glehen O, Pocard M. A comparative study of complete cytoreductive surgery plus intraperitoneal chemotherapy to treat peritoneal dissemination from colon, rectum, small bowel and nonpseudomyxoma appendix. Ann Surg 2010; 251: 896-901.

4. Elias D, Gilly F, Boutitie F. 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.

5. Verwaal V, van Ruth S, Witkamp A. Long-term survival of peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol 2005; 12: 65-71.

6. Elias D, Lefevre J, Chevalier J. Complete cytoreductive surgery plus intraperitoneal chemohyperthermia with oxaliplatin for peritoneal carcinomatosis of colorectal origin. J Clin Oncol 2009; 27: 681-685.

7. Kusamura S, Elias D, Baratti D. Drugs, carrier solutions and temperature in hyperthermic intraperitoneal chemotherapy. J Surg Oncol 2008; 98: 247-252.

8. van Ruth S, Mathôt R, Sparidans R. Population pharmacokinetics and pharmacodynamics of mitomycin during intraoperative hyperthermic intraperitoneal chemotherapy. Clin Pharmacokinet 2004; 43: 131-143.

Page 140: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

131

9. Soulié P, Raymond E, Brienza S. Oxaliplatin: the first DACH platinum in clinical practice. Bull Cancer 1997; 84: 665-673.

10. de Gramont A, Figer A, Seymour M. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 2000; 18: 2938-2947.

11. Makatsoris T, Kalafonos HP, Aravantinos G. A phase II study of capecitabine plus oxaliplatin (XELOX): a new first-line option in metastatic colorectal cancer. Int J Gastrointest Cancer 2005; 35: 103-109.

12. Scheithauer W, Kornek G, Raderer M. Randomized multicenter phase II trial of two different schedules of capecitabine plus oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 2003; 21: 1307-1312.

13. Alkis N, Demerci U, Benekli M. Mitomycin-C in combination with fluoropyrimidines in the treatment of metastatic colorectal cancer after oxaliplatin and irinotecan failure. J BUON 2011; 16: 80-83.

14. Kang E, Choi Y, Kim J. Mitomycin-C, 5-fluorouracil, and leucovorin as a salvage therapy in patients with metastatic colorectal adenocarcinoma. Asia Pac J Clin Oncol 2010; 6: 286-91.

15. Michalaki V, Gennatas S, Gennatas C. Mitomycin C and UFT/leucovorin as salvage treatment in patients with advanced colorectal cancer. J BUON 2010; 15: 270-273.

16. Vormittag L, Kornek G, Gruhsmann B. UFT/leucovorin and mitomicyn C as salvage treatment in patients with advanced colorectal cancer – a retrospective analysis. Anticancer Drugs 2007; 18: 709-712.

17. Swellengrebel H, Zoetmulder F, Verwaal V. Quantitative intra-operative assessment of peritoneal carcinomatosis - a comparison of three prognostic tools. Eur J Surg Oncol 2009; 35: 1078-1084.

18. Jacquet P, Sugarbaker P. Clinical research methodologies in diagnosis and staging of patients with peritoneal

Page 141: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

132

carcinomatosis. In: Sugarbaker PH, editor. Peritoneal carcinomatosis: Principles of management. Editors. Boston: Kluwer Academic Publishers 1996: 359–37.

19. Schemper M, Smith T. A note on quantifying follow-up in studies of failure time. Control Clin Trials 1996; 17: 343-346.

20. Pestieau S, Belliveau J, Griffin H. Pharmacokinetics of intraperitoneal oxaliplatin: experimental studies. J Surg Oncol 2001; 76: 106-114.

21. Lambert L, Armstrong T, Lee J. Incidence, risk factors, and impact of severe neutropenia after hyperthermic intraperitoneal mitomycin C. Ann Surg Oncol 2009; 16: 2181-2187.

22. van Ruth S, Verwaal V, Zoetmulder F. Pharmacokinetics of intraperitoneal mitomycin C. Surg Oncol Clin N Am 2003; 12: 771-780.

23. Elias D, Bonnay J, Puizillou M. Heated intra-operative intraperitoneal oxaliplatin after complete resection of peritoneal carcinomatosis: pharmacokinetics and tissue distribution. Ann Oncol 2002; 13: 267-272.

24. Elias D, Pocard M, Goere D. HIPEC with oxaliplatin in the treatment of peritoneal carcinomatosis of colorectal origin. Cancer Treat Res 2007; 134: 303-318.

25. Mahteme H, Wallin I, Glimelius B. Systemic exposure of the parent drug oxaliplatin during hyperthermic intraperitoneal perfusion. Eur J Clin Pharmacol 2008; 64: 907-911.

26. Bécouarn Y, Ychou M, Ducreux M. Phase II trial of oxaliplatin as first-line chemotherapy in metastatic colorectal cancer patients: Digestive Group of French Federations of Cancer Centers. J Clin Oncol 1998; 16: 2739-2744.

27. Elias D, Benizri E, Di Pietrantonio D. Comparison of two kinds of intraperitoneal chemotherapy following complete cytoreductive surgery of colorectal peritoneal carcinomatosis. Ann Surg Oncol 2007; 14: 509-514.

Page 142: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

133

28. De Somer F, Ceelen W, Delanghe J. Severe hyponatremia, hyperglycemia, and hyperlactatemia are associated with intraoperative hyperthermic intraperitoneal chemoperfusion with oxaliplatin. Perit Dial Int 2008; 28: 61-66.

29. Ceelen W, Peeters M, Houtmeyers P. Safety and efficacy of hyperthermic intraperitoneal chemoperfusion with high-dose oxaliplatin in patients with peritoneal carcinomatosis. Ann Surg Oncol 2008; 15: 535-541.

30. Elias D, Pocard M. Treatment and prevention of peritoneal carcinomatosis from colorectal cancer. Surg Oncol Clin N Am 2003; 12: 543-559.

31. Maggiori L, Elias D. Curative treatment of colorectal peritoneal carcinomatosis: currents status and future trends. Eur J Surg Oncol 2010; 36: 599-603.

32. van Leeuwen B, Graf W, Pahlman L. Swedish experience with peritonectomy and HIPEC in peritoneal carcinomatosis. Ann Surg Oncol 2008; 15: 745-753.

33. Gomes da Silva R, Sugarbaker P. 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.

34. Verwaal V, van Ruth S, de Bree E. Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 2003; 21: 3737-3743.

35. Verwaal V, van Tinteren H, van Ruth S. Predicting the survival of patients with peritoneal carcinomatosis of colorectal origing treated by aggressive cytoreduction and hyperthermic intraperitoneal chemotherapy. Br J Surg 2004; 91: 739-746.

36. Verwaal V, van Tinteren H, van Ruth S. Toxicity of cytoreductive surgery and hyperthermic intra-peritoneal chemotherapy. J Surg Oncol 2004; 85: 61-67.

Page 143: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

134

37. Elias D, Goere D, Blot F. Optimization of hyperthermic intraperitoneal chemotherapy with oxaliplatin plus irinotecan at 43°C after complete cytoreductive surgery: mortality and morbidity in 106 consecutive patients. Ann Surg Oncol 2007; 14: 1818-1824.

38. Graham M, Lockwood G, Greenslade D. Clinical pharmacokinetics of oxaliplatin: a critical review. Clin Cancer Res 2000; 6: 1205-1218.

39. Kautio A, Haanpää M, Kautiainen H. Oxaliplatin Scale and National Cancer Institute-Common Toxicity Criteria in the Assessment of Chemotherapy-induced Peripheral Neuropathy. Anticancer Res 2011; 31: 3493-3496.

40. Grothey A. Oxaliplatin-safety: neurotoxicity. Semin Oncol 2003; 30: 5-13.

41. Stuart O, Stephens A, Welch L. Safety monitoring of the coliseum technique for heated intraoperative intraperitoneal chemotherapy with mitomycin C. Ann Surg Oncol 2002; 9: 186-191.

42. Schmid K, Boettcher M, Pelz J. Investigations on safety of hyperthermic intraoperative intraperitoneal chemotherapy (HIPEC) with mitomycin C. Eur J Surg Oncol 2006; 32: 1222-1225.

43. Näslund Andréasson S, Anundi H, Thorén S. Is platinum present in blood and urine from treatment givers during hyoerthermic intraperitoneal chemotherapy? J Oncol 2010; 2010:649719.

Page 144: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

135

III

3. HIPEC in T4a colon cancer: a defendable treatment

to improve oncologic outcome?

Hompes D, Tiek J, Wolthuis A, Fieuws S, Penninckx F, Van Cutsem E, D’Hoore A

Ann Oncol 2012; 23: 3123-3129.

Page 145: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

136

Page 146: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

137

HIPEC in T4a colon cancer: a defendable treatment to improve oncologic

outcome?

D.Hompes1, J.Tiek1, A.Wolthuis1, S.Fieuws2, F.Penninckx1, E.Van Cutsem3, A.D’Hoore1

1 Department of Abdominal Surgery, University Hospitals Gasthuisberg, Leuven, Belgium 2 I -Biostat, Katholieke Universiteit Leuven and Universiteit Hasselt,Belgium 3 Department of Digestive Oncology, University Hospitals Gasthuisberg, Leuven, Belgium

Introduction Colon tumors with transserosal invasion (T4a) have an

increased risk for the development of peritoneal carcinomatosis (PC), secondary to the intra-abdominal exfoliation of cancer cells that invaded the full-thickness colonic wall and its investing serosa. Shepherd et al. pointed at the significance of transserosal invasion, with local peritoneal involvement (LPI) in colorectal cancer (CRC) as a significant risk factor for intraperitoneal disease recurrence in colorectal cancer1-4. This is in agreement with the findings of Newland et al (Australian clinico-pathological staging (ACPS) group), who demonstrated a poor prognosis in rectal cancer with free serosal surface involvement5. Also, Park et al showed that the depth of bowel wall invasion is a significant risk factor for prognosis in colon and rectal cancer6. In a critical reflection on Shepherd’s findings Williams et al proposed to consider the use of intraperitoneal therapy for patients in this setting 4,7. As such, “adjuvant” Hyperthermia and Intraperitoneal Chemotherapy (HIPEC) could be a strategy to reduce the risk of PC and increase survival in stage II and III T4a patients. This clinical study aims to assess the burden of PC in T4 tumors and the risk for local intraperitoneal recurrence as the only site of metastatic disease. This is crucial to adequately

Page 147: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

138

estimate the potential benefits of HIPEC as an adjuvant treatment strategy in T4 patients. Current literature on this issue is scarce.

Patients and Methods A retrospective analysis was performed of prospectively

collected data on 379 consecutive patients who underwent elective or emergency surgery for colon cancer between January 2004 and January 2007 at the department of abdominal Surgery of the University Hospitals Leuven.

As a primary end point this study focused on the metachronous occurrence of PC as the sole location of tumor recurrence in colon cancer. The secondary end point was the calculation of a power analysis if one would aim to start a prospective randomized trial to prove the impact of adjuvant HIPEC in T4a colon cancer.

The trial protocol was approved by the local medical ethics committee.

The pathology report of every single resection specimens was revised by one of the authors (DH). Pathologic staging was performed according to the American Joint Committee on Cancer (AJCC) Colon and Rectum Cancer Staging [7th Edition]8.

According to a strict standard in-hospital protocol, patients were seen postoperatively at the outpatient clinic for oncological follow-up: every 3 months in the first 2 years after surgery, every 6 months in the following 2 years and once a year from the fifth postoperative year on. At each visit a blood sample was taken to determine the CEA level and imaging in the form of a chest X-ray and liver ultrasound and/or CT thorax/abdomen was performed to screen the patient for recurrent disease. At least one CT thorax/abdomen per year was performed. All patients with suspicion of peritoneal disease underwent a PET/CT-scan to rule out systemic recurrence in and outside of the peritoneal cavity. Colonoscopy was repeated every 2 to 3 years to assess local recurrence and/or metachronous primary colonic cancer. At closing date of the study (16-09-2010), no

Page 148: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

139

patients were discharged from follow-up, but 46 patients (12%) were lost to follow-up.

Statistical methodology Kaplan-Meier (KM) estimates are used to create a curve for the percentage overall survival (OS) as a function of the time since HIPEC surgery. A log-rank test is used to compare stage II and stage III patients. A multivariable Cox regression is used to evaluate this difference after correction for age, sex and post-operative chemo. A hazard ratio (HR) and 95% confidence interval (CI) is reported. To construct the curves for percentage relapse and for PC, cumulative incidence estimates are used instead of KM-estimates9. Censoring deceased patients without respectively relapse and PC in the KM-curve inappropriately assumes that those patients are still at risk after their death. To evaluate the relation between PC and OS, an extended Cox regression model is used, with PC as a binary non-reversible time-dependent covariate10. The length of follow-up is calculated using a KM estimate censoring the deceased patients11. P-values are considered significant if smaller than 0.05. Analyses have been performed using SAS software, version 9.2 of the SAS System for Windows. Copyright © 2002 SAS Institute Inc. SAS and all other SAS Institute Inc. product or service names are registered trademarks or trademarks of SAS Institute Inc., Cary, NC, USA.

Results Between January 2004 and January 2007, 379 patients

(204 male and 175 female patients) underwent surgery for colon cancer. There were no rectal cancers in this patient cohort. Pathologic staging revealed 39 stage I, 126 stage II, 89 stage III and 116 stage IV disease. For 9 tumors pTNM-staging was unknown. This study will only focus on the 126 patients with stage II (T3N0M0, T4N0M0) and 89 patients with stage III (T2N+M0, T3N+M0, T4N+M0) colon cancer. Table 1 summarizes patient and tumor characteristics of this patient group. The male/female ratio was 1.13/1, with a median age at surgery of

Page 149: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

140

72.3 years [range 35.9-95.0 years]. The location of the colon tumor was right-sided in 45.6% and left-sided in 54.4% of patients. Differentiation was good to moderate in 67.9% of patients, poor in 30.7% and differentiation data were unavailable in 1.4%.

Final common closing date of the study was 16-09-2010. At a median FU of 52.9 months [IQR: 35.8-64.8] 3- and 5-year overall survival (OS) for stage II and III disease was 78.0% (95%CI: 72.0-82.8) and 72.2%(95%CI: 65.6-77.7), respectively. There was a significant difference in OS between stage III and stage II patients (p=0.03), which remained significant after correction for age, sex and adjuvant chemotherapy (yes/no) in a multivariable Cox regression analysis [HR 4.1 (95%CI 2.1-8.2, p<0.001)] [Fig.1]. Relapse was defined as the development of local recurrence and/or metastatic disease, i.e. PC as well as systemic metastases. As already mentioned, the relapse analysis was restricted to patients with stage II-III colon cancer for whom the data on relapse were completely available (N=178 patients). Based on the cumulative incidence estimates, the relapse at 5 years equals 11.4% (95%CI: 5.8-17.0) for stage II and 40.0% (95%CI: 29.4-50.6) for stage III colon cancer (p<0.001) [Fig.2A]. For 94 T3 and 7 T4 tumors (80.2%) in Stage II disease and 60 T3 and 12 T4 tumors (80.9%) in stage III disease follow-up data were completely available [Table 2A]. For T3 tumors the relapse at 5 years equals 20.2% (95%CI: 14.3-26.2) versus 40.0% (95%CI: 20.8-59.2) T4 tumors (p=0.064) [Fig.2B]. Overall, 23 patients with T3 and T4 tumors (13.2%) without metastases (stage II and III) developed PC. Using Cox regression analysis (with PC as time-dependent predictor), PC has a significant detrimental effect on OS [HR is 6.3 (95%CI: 3.0-13.0, p<0.0001)] and there was no evidence that this effect was different for stage II and III disease (p=0.76) [Fig.S1]. Out of 14 T4a tumors 7 developed PC (50%), whereas only 1 out of 5 T4b tumors (20%) developed PC [Table 2B]. The percentage PC at 1 and 3 years was significantly lower for T3 tumors (4.5% and 9.3%) compared to T4 tumors (15.6% and 36.7%) (p=0.008) [Fig.3]. Furthermore, a higher proportion of patients with a T4 tumor (especially T4a) developed PC as the

Page 150: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

141

only location of disease recurrence: only in 3 of the 15 T3 tumors with PC (proportion 0.20, CI [0.043-0.481]) and 5 of the 8 T4 tumors with PC (proportion 0.625, CI [0.245-0.915]) did PC occur as the only metastatic site, i.e. without synchronous systemic metastatic disease (p=0.071). These 5 tumors all had a T4a staging [Table 2B].

Sample size calculation Assuming that about 25% of stage II and III T4 tumors will develop PC without synchronous systemic metastases, a total of 352 patients need to be included in a randomized controlled trial (176 patients in both treatment arms) in order to demonstrate with 80% power that adjuvant HIPEC would lead to a 50% reduction in the risk of PC without systemic metastases compared to those stage II and III T4 patients who would not receive HIPEC. At our institute, 26 out of 255 stage II-III colonic tumors were staged T4 over a time period of 3 years. Practically, this would mean that a cohort of 3443 patients with a stage II or III colonic tumor is needed, in order to enable inclusion of 352 T4 tumors. In case of monocentric inclusion this would take 40 years of inclusion!

Discussion In our study 26/215 patients with stage II-III CRC (12.1%) developed peritoneal metastases [Table 2A], which is in agreement with published data, as PC is described to occur in about 12-13% of patients with colorectal cancer (CRC)12,13. The development of peritoneal disease recurrence will compromise survival. For unresectable PC median survival is expected to be 3.1 to 9.3 months, depending on whether or not these patients are able to undergo systemic chemotherapy14-16. PC appears to be the only site of recurrent disease in about 25-35% of patients17,18. The understanding that peritoneal disease can occur without other metastatic location led to the concept of a radical approach of CCRS and HIPEC15,17-20. This combined treatment modality significantly increased survival, reaching a 5-

Page 151: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

142

year overall survival (OS) of 33-51%, provided an R0/1 resection can be reached by CCRS21-24. Shepherd et al highlighted the prognostic importance of deep serosal invasion and local peritoneal involvement (LPI) in CRC1-4. Transserosal invasion of colorectal tumors (T4) was shown to increase the risk for PC and/or local recurrence1-4. In our retrospective analysis a clear and significant (p=0.008) correlation was shown between transserosal invasion of colonic cancer (T4) and peritoneal recurrence [Fig.3]. The majority of these T4 tumors had a T4a staging [Table 2B]. In 5 of 8 T4 tumors that developed PC (proportion 0.625, CI [0.245-0.915]), PC was the only metastatic site, without synchronous systemic metastases. This could implicate that 5 out of the 19 T4 patients might eventually benefit from adjuvant HIPEC in order to prevent PC. This is 1.3% (5/379) of the total patient population presenting with colon cancer. At present, there is no adequate diagnostic tool available to peroperatively identify tumors with transserosal invasion. A number of studies addressed the prognostic importance of detection of free intraperitoneal tumor cells (IPTC) in colorectal cancer18,25-35. A review by Bleichrodt et al.18 as well as a meta-analysis by Rekhraj et al.25 examined several trials with various methodology for the detection of free IPTC. They concluded that the detection of free IPTC is associated with higher local and overall recurrence and poorer prognosis25. Nevertheless, it should be noted that there is a tremendous variation among trials in the IPTC detection methodology and sensitivity, as well as the definition of outcome measures. Furthermore, there is often no clear information on the T-stage of the included tumors and some of the trials also included patients with stage IV disease. A large prospective trial performed by Noura et al. aimed to evaluate the value of peritoneal lavage cytology in patients undergoing curative resection of stage I, II or III CRC26. Over a time span of 13 years 697 patients were included, 15 of whom (2.2%) had a positive cytology. Thus, apparently it takes a very large patient cohort to enable the identification of a very small number of patients at higher risk. Furthermore, there are

Page 152: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

143

more sensitive IPTC detection methods, such as immunocytochemistry33 or RT-PCR34,35, with detection rates of 12 to 47%. Nevertheless, despite this higher sensitivity, the significance of IPTC for survival remains unclear. Additionally, the ability to estimate the amount of free IPTC might have a higher clinical value than the mere detection of their presence26. Assuming one would be able to peroperatively identify T4a colonic tumors, one could consider the administration of HIPEC as “adjuvant” treatment in order to reduce the risk of peritoneal recurrence in this particular group of tumors. To our knowledge, only one study, performed by Noura et al., explored the effects of intraperitoneal chemotherapy (IPC) with Mytomycin C (MMC) on the prevention of peritoneal recurrence in CRC in patients with positive peritoneal lavage cytology27. Over a time span of 20 years 52 patients with stage II, III or IV CRC (all T3 and T4 tumors) and positive lavage cytology were included in this trial. Thirty-one of them received an IPC perfusate with MMC, at body temperature, through a closed-abdomen technique. It is not clearly described if and how patients were randomized into both trial arms. The authors concluded that this procedure of IPC appears effective in preventing peritoneal recurrence and prolonging cancer-specific survival. Multivariate analysis showed IPC was not a significant risk factor for cancer-specific survival. For the recurrence analysis stage IV patients were omitted. The recurrence rate was 3/24 (12.5%) in the IPC(+) group versus 6/12 (50%) in the IPC(-) group. This difference was rated significant, despite the small total number of recurrences27. Based on our own retrospective analysis a sample size calculation showed that a randomized controlled trial to demonstrate the benefit of adjuvant HIPEC in stage II and III T4 colon cancers, defined as a 50% risk reduction for the development of PC without systemic metastases, with a power of 80%, would require a sample size of 352 patients. As already mentioned above, this is an impossible task for a single institute. Even with a multicentric approach inclusion of 176 stage II-III T4 colonic tumors in each trial arm would be an ambitious undertaking.

Page 153: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

144

It should also be noted that there was no mortality in the trial of Noura et al. and only one patient (1.9%) was reported to have a serious adverse event related to the intraperitoneal chemotherapy27, whereas the combination of cytoreductive surgery and HIPEC has been extensively described to come with the price of very high morbidity17,22,24,36-43.

Finally, another important logistic remark should be added: In the first 24 to 72h after surgery fibrin tissue will inevitably be formed in the resection plane and adhesions will start to develop, thus entrapping any free IPTC that might be present in the abdomen and making them unattainable for HIPEC. Therefore, pathologic diagnosis of deep transserosal invasion should be made within 24h after the resection.

Elias et al. agreed that, at present, the risk of developing PC has not clearly been established for patients with pT4 lesions of the colon and that the avalaible data in literature are discordant24. CCRS + HIPEC is acknowledged to result in less morbidity and mortality, as well as better survival for patients with low PCI. However, early PC is currently not detectable clinically. Thus, Elias et al. proposed to perform a second look laparotomy plus HIPEC 1 year after resection of the primary tumor for asymptomatic patients at high risk of developing colorectal PC. This high-risk population was defined based on 3 primary tumor-associated criteria: synchronous macroscopic PC, synchronous ovarian metastases and perforation24,44,45. A prospective trial, including 41 patients, concluded these selection criteria appear to be accurate. PC was found and treated with CCRS+HIPEC in 23 patients (56%), whereas the other 18 patients underwent exploration and HIPEC. This approach yielded a 5-year OS and DFS of 90% and 44%, respectively45. Grade 3-4 morbidity was 9.7% and 1 patient died at day 69 postoperatively45. Based on these results a randomized controlled trial was designed to compare this second look + HIPEC strategy to standard follow-up alone. In conclusion, T4a colon tumors are indeed at significantly higher risk of developing PC. Furthermore, 25% (5/19) of stage II and III T4 tumors developed PC as the only site of metastases.

Page 154: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

145

This defines the window of opportunity for adjuvant HIPEC to prevent peritoneal recurrence. There is an urgent need to evaluate and validate diagnostic methods to allow peroperative detection of IPTC and/or transserosal invasion (pT4a). This should allow to implement HIPEC as an adjuvant treatment in this high risk group of patients. For the time being a strategy as proposed by Elias et al. could be the better approach.

Page 155: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

146

Table 1: Patient and tumor characteristics of patients with stage II and III colon cancer

N = 215 colon cancers

Male/Female 114/101

Median age at surgery

72.3 years [range 35.9-95.0 years]

Primary tumor Stage II

III

126

89

IIA IIB IIC IIIA IIIB IIIC

118 4 4 4 56 29

Differentiation Good

Moderate Poor

Unknown

22

124 66 3

Localization Right colon

Left colon

98

117

N= number Note: right colon = caeco-ascending colon / left colon = transverse, descending, sigmoid colon

Page 156: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

147

Table 2: Relapse A. in stage II & III disease

T-stage ALL Relapse

Systemic M+

Local PC N

Stage II

N=126

T3 No Yes Yes Yes Yes Yes

unknown

No Yes Yes No Yes Yes

unknown

No No No Yes Yes Yes

unknown

No No Yes Yes No Yes

unknown

82 5 2 2 1 2

24

T4 No Yes Yes Yes

unknown

No No Yes Yes

unknown

No No No Yes

unknown

No Yes No Yes

unknown

3 2 1 1 1

Stage III

N=89

T2 No Yes yes

No Yes yes

No No

unknown

No Yes Yes

2 1 1

T3 No Yes Yes Yes Yes Yes

unknown

No No Yes Yes Yes Yes

unknown

No No No No Yes

unknown unknown

No Yes No Yes Yes Yes

unknown

35 1

16 1 7 1 7

T4 No Yes Yes Yes

unknown

No Yes No Yes

unknown

No No Yes Yes

unknown

No Yes Yes No

unknown

6 2 3 1 5

N= number; systemic M+=systemic metastases; PC=peritoneal carcinomatosis

Page 157: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

148

B. for T4a and T4b colon tumors

T4 (N=19)

Recurrent disease

Local recurrence

N=5

Peritoneal Carcinomatosis

N=8

Systemic Metastases

N=5

N

T4a no no no 6

no yes no 2

yes yes no 3

no yes yes 2

T4b no no no 2

no no yes 1

yes yes yes 1

T4a,b yes no no 1

T4x no no no 1

N= number

Page 158: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

149

Fig.1: OS for stage II and III colon cancer (Survival curves are obtained from a Cox regression correcting for age, sex and postoperative chemotherapy and these curves reflect groups of patients with an average age and equal distribution fro sex and postoperative chemotherapy)

Page 159: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

150

Fig.2: Percentage relapse (cumulative incidence estimates) A: in stage II and stage III colon cancer

Page 160: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

151

B: for T3 and T4 tumors in patients with stage II-III colon cancer

Note: only stage II and III patients for whom data on relapse were complete, were included in this analysis.

Page 161: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

152

Fig.3: Percentage Peritoneal Carcinomatosis (PC) as a function of T-stage for stage II & III disease. (the percentage is based on cumulative incidence estimates)

Note: only T3 and T4 tumors for which data on relapse were complete, were included in this analysis.

Page 162: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

153

Fig.S1: Overall Survival as a function of TNM stage and PC Survival curves are obtained from a Cox regression with PC as a time-dependent factor, TNM staging (III vs II) as a baseline factor, and their interaction. The solid lines represent TNM stage II and III patients who do not develop PC. The dashed lines represent hypothetical samples of patients who develop PC after 12 months.

Page 163: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

154

Legend for all figures: TNM=II: stage II colon cancer TNM=III: stage III colon cancer PC=peritoneal carcinomatosis no PC=patients who do not develop PC; PC after 12 months=patients who develop PC after 12 months

Page 164: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

155

References

1. Shepherd N, Baxter K, Love S. The prognostic importance of peritoneal involvement in colonic cancer: a prospective evaluation. Gastroenterology 1997; 112: 1096-1102.

2. Ludeman L, Shepherd N. Serosal involvement in gastrointestinal cancer: its assessment and significance. Histopathology 2005; 47: 123-131.

3. Shepherd N, Baxter K, Love S. Influence of local peritoneal involvement on pelvic recurrence and prognosis in rectal cancer. J Clin Pathol 1995; 48: 849-855.

4. Petersen V, Baxter K, Shepherd N. Identification of objective pathological prognostic determinants and models of prognosis in Dukes’ B colon cancer. Gut 2002; 51: 65-69.

5. Newland RC, Chapuis PH. Peritoneal involvement by rectal cancer. J Clin Pathol 1996: 49: 694-695.

6. Park Y, Park K, Park J. Prognostic factors in 2230 Korean colorectal cancer patients: analysis of consecutively operated cases. World J Surg 1999; 23: 721-726.

7. Williams GT. A long hard look at Dukes’ B: identification of prognostic pathological factors in Dukes’ B colon cancer. Gut 2002; 51: 6-7.

8. Edge S, Byrd D, Compton C. AJCC Cancer Staging Manual, 7th Edition. (Eds.) 2010, ISBN 978-0-387-88440-0

9. Pintilie M. Competing risks: a practical perspective. 2006, Wiley, Chilchester.

10. Therneau T, Grambsch P. Modeling survival data: extending the Cox model. 2000, Springer-verlag, New York, NY.

11. Schemper M, Smith T. A note on quantifying follow-up in studies of failure time. Controlled Clinical Trials 1996; 17: 343-346.

12. Jayne D, Fook S, Seow-Choen F. Peritoneal carcinomatosis from colorectal cancer. Br J Surg 2002; 89: 1545-1550.

13. Folprecht G, Köhne C, Lutz M. Systemic chemotherapy in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440.

Page 165: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

156

14. Chu D, Lang N, Thompson C. Peritoneal carcinomatosis in nongynecologic malignancy. A prospective study of prognostic factors. Cancer 1989; 63: 364-367.

15. Sadeghi B, Arvieux C, Gilly F. Peritoneal carcinomatosis from non-gynecologic malignancies. Results of the EVOCAPE I multicentric prospective study. Cancer 2000; 88: 358-363.

16. Hompes D, Boot H, Verwaal V. Unresectable peritoneal carcinomatosis from colorectal cancer: a single center experience. J Surg Oncol 2011; 104: 269-273.

17. Verwaal V, Zoetmulder F. Long-Term Survival of Peritoneal carcinomatosis of Colorectal Origin; Ann Surg Oncol 2005; 12: 65-71.

18. Koppe M, Bleichrodt R. Peritoneal Carcinomatosis of Colorectal Origin: Incidence and Treatment Strategies; Ann Surg 2006; 243: 212-222.

19. Shen P, Levine E, Hall J. Factors predicting survival after intraperitoneal hyperthermic chemotherapy with mitomycin C after cytoreductive surgery for patients with peritoneal carcinomatosis. Arch Surg 2003; 138: 26-33.

20. Sugarbaker P. Management of peritoneal-surface malignancy: the surgeon’s role; Langenbecks Arch Surg 1999; 384: 576-587.

21. Elias D, Lefevre J, Chevalier J. Complete Cytoreductive Surgery plus Intraperitoneal Chemohyperthermia with Oxaliplatin for peritoneal Carcinomatosis of Colorectal Origin. J Clin Oncol 2009: 27: 681-685.

22. Elias D, Glehen O, Pocard M. A comparative study of complete cytoreductive surgery plus intraperitoneal chemotherapy to treat peritoneal dissemination from colon, rectum, small bowel and nonpseudomyxoma appendix. Ann Surg 2010; 251: 896-901.

23. Verwaal V, Bruijn S, Boot H. 8-Year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis

Page 166: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

157

of colorectal cancer. Ann Surg Oncol 2008; 15: 2426-2432.

24. Maggiori L, Elias D. Curative treatment of colorectal peritoneal carcinomatosis: current status and future trends. Eur J Surg Oncol 2010; 36: 599-603.

25. Rekhraj S, Aziz O, Prabhudesai S. Can intra-operative intraperitoneal free cancer cell detection techniques identify patients at higher recurrence risk following curative colorectal cancer: a meta-analysis. Ann Surg Oncol 2007; 15: 60-68.

26. Noura S, Ohue M, Seki Y. Long-term prognostic value of conventional peritoneal lavage cytology in patients undergoing curative colorectal cancer resection. Dis Colon Rectum 2009; 52: 1312-1320.

27. Noura S, Ohue M, Shingai T. Effects of intraperitoneal chemotherapy with Mitomycin C on the prevention of peritoneal recurrence in colorectal cancer patients with positive peritoneal lavage cytology findings. Ann Surg Oncol 2010; 18: 396-404.

28. Nishikawa T, Watanabe T, Sunami E. Prognostic value of peritoneal cytology and the combination of peritoneal cytology and peritoneal dissemination in colorectal cancer. Dis Colon Rectum 2009; 52: 2016-2021.

29. Fujii S, Shimada H, Yamagishi S. Evaluation of intraperitoneal lavage cytology before colorectal cancer resection. Int J Colorectal Dis 2009; 24: 907-914.

30. Katoh H, Yamashita K, Sato T. Prognostic significance of peritoneal tumour cells identified at surgery for colorectal cancer. Br J Surg 2009; 96: 769-777.

31. Baskaranathan S, Philips J, McCredden P. Free colorectal cancer cells on the peritoneal surface: correlation with pathologic variables and survival. Dis Colon Rectum 2004; 47: 2076-2079.

32. Hase K, Ueno H, Kuranaga N. Intraperitoneal exfoliated cancer cells in patients with colorectal cancer. Dis Colon Rectum 1998; 41: 1134-1140.

Page 167: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

158

33. Bosch B, Guller U, Schnider A. Perioperative detection of disseminated cells is an independent prognostic risk factor in patients with colorectal cancer. Br J Surg 2003; 90: 882-888.

34. Guller U, Zajac P, Schnider A. Disseminated single tumor cells as detected by real-time quantitative polymerase chain reaction represent a prognostic factor in patients undergoing surgery for colorectal cancer. Ann Surg 2002; 236: 768-75; discussion 775-776.

35. Hara M, Nakanishi H, Jun Q. Comparative analysis of intraperitoneal minimal free cancer cells between colorectal and gastric patients: using quantitative RT-PCR: possible reason for rare peritoneal recurrence in colorectal cancer. Clin Exp Metastasis 2007; 24: 179-189.

36. Elias D, Bonnay M, Puizillou J. Heated intra-operative intraperitoneal oxaliplatin after complete resection of peritoneal carcinomatosis: pharmacokinetics and tissue distribution. Ann Oncol 2002; 13: 267-272.

37. Verwaal V, van Tinteren H, van Ruth S. Toxicity of cytoreductive surgery and hyperthermic intra-peritoneal chemotherapy. J Surg Oncol 2004; 85: 61-67.

38. Van Ruth S, Mathôt R, Verwaal V. Population pharmacokinetics and pharmacodynamics of mitomycin during intraoperative hyperthemic intraperitoneal chemotherapy. Clin Pharmacokinet 2004; 43: 131-143.

39. Lambert L, Armstrong T, Lee J. Incidence, risk factors and impact of severe neutropenia after hyperthermic intraperitoneal mitomycin C. Ann Surg Oncol 2009; 16: 2181-2187.

40. Elias D, Goere D, Blot F. Optimization of hyperthermic intraperitoneal chemotherapy with oxaliplatin plus irinotecan at 43 degrees C after complete cytoreductive surgery: mortality and morbidity in 106 consecutive patients. Ann Surg Oncol 2007; 14: 1818-1824.

41. van Leeuwen B, Graf W, Pahlman L. Swedish experience with peritonectomy and HIPEC. HIPEC in peritoneal carcinomatosis. Ann Surg Oncol 2008; 15: 745-753.

Page 168: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

159

42. Elias D, Gilly F, Boutitie F. 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.

43. Elias D, Sideris L. Pharmacokinetics of heated intraoperative intraperitoneal oxaliplatin after complete resection of peritoneal carcinomatosis. Surg Oncol Clin N Am 2003; 12: 755-69.

44. Elias D, Goere D, Di Pietrantonio D. Results of systematic second-look surgery in patients at high risk of developing colorectal peritoneal carcinomatosis. Ann Surg 2008; 247: 445-450.

45. Elias D, Honoré C, Dumont F. Results of systematic second-look surgery plus HIPEC in asymptomatic patients presenting a high risk of developing colorectal peritoneal carcinomatosis. Ann Surg 2011; 254: 289-293.

Page 169: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

160

Page 170: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

161

Radiofrequency ablation (RFA)

for colorectal liver metastases treatment

IV

Page 171: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

162

Page 172: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

163

IV

1. Radiofrequency ablation as a treatment tool for liver metastases

of colorectal origin

Hompes D, Prevoo W, Ruers T

Cancer Imaging 2011; 24: 23-30.

Review

Page 173: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

164

Page 174: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

165

Reflections on RFA as a treatment tool for liver metastases of colorectal origin

D.Hompes1, W.Prevoo2, T.Ruers1

1 Department of surgery, 2 Department of radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands

Introduction Up to 50% of patients with colorectal cancer (CRC) develop colorectal liver metastases (CRLM) at some point in the course of their disease. Only 10-25% of these CRLM will present at diagnosis as resectable disease. Curative liver resection is currently considered to be the gold standard treatment for resectable CRLM. Hepatectomy has a morbidity of 17-37% and a mortality below 5%1,2. Recent reviews showed 5-year overall survival rates after hepatic resection of 22-58% and a 10-year survival of up to 28%3,4. Local recurrence rates after resection vary from 1.2-10.4%3. For unresectable CRLM recent improvements in systemic chemotherapy have lead to an overall survival of almost 2 years, but 5-year survival after chemotherapy is very rare3. In the past decade there have been several papers looking at the possible contribution of radiofrequency ablation to improve overall survival (OS) and progression-free survival (PFS) in patients with unresectable colorectal liver metastases. The aim of this paper is to look into the results of trials performed thusfar in the setting of unresectable liver metastases and to carefully reflect on a possible role of RFA in resectable disease.

General guidelines The following guidelines were described and are

generally accepted for RFA of CRLM4,5:

Page 175: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

166

- The number of lesions should not be considered an absolute contra-indication to RFA, but most centers preferentially treat patients with ≤5 lesions.

- Best rates of complete ablation are achieved in lesions with a maximum diameter of ≤3cm.

- Tumor location can be a problem in case of:

o lesions on the liver surface because of themal injury of adjacent structures (although this risk can be mitigated through pre-ablation percutaneous infusion of dextrose fluid into the space between the liver and the adjacent bowel, abdominal wall or diafragm)

o lesions adjacent to the hepatic hilum (risk of thermal injury to the biliary tract)

o lesions adjacent to large hepatic vessels (because of the “heat sink” phenomenon).

- Intrahepatic bile duct dilatation and untreatable/unmanageable coagulopathy are generally considered a contraindication for RFA. The same applies to the presence of bilioenteric anastomoses, which increases the incidence of hepatic abscess, although there are papers suggesting the combined use of RFA with extended antibiotic prophylaxis in these cases6. Tumors located <1cm from the main biliary duct are at risk for delayed stenosis of the main biliary duct.

RFA for unresectable CRLM in comparison to chemotherapy

Overall Survival and Recurrence after RFA In patients with unresectable CRLM current systemic chemotherapy can result in a median survival of up to 20.6

Page 176: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

167

months7. With regard to the use of RFA in patients with unresectable CRLM, literature reports on various treatment combinations with RFA in different therapy settings. Results are highly variable with 5-year survival rates from 3.0% to 30.5% and local recurrence rates varying from 5.0% to more than 60% [Table 1-3]. With RFA performed with an intention to cure in patients with only CRLM, survival and local failure are directly related to the size and number of the lesions and their location, as well as the number of ablations, the chosen approach (open, laparoscopically or percutaneously) and of course the physician’s experience4. A literature study by Crocetti and Lencioni et al showed that nonsurgical patients with ≤5 CRLM, each ≤5 cm in diameter, have a 5-year survival rate of 24-44% after RFA5,8. Best results are obtained with RFA for small (<3-4cm), solitary lesions, which can result in a 40% 5-year survival4,5. A meta-analysis by Mulier et al (3760 patients) found less local recurrence in small size lesions (<3cm)9. It should be mentioned that the literature search for this analysis focused on the period 1990-2004, thus also including early experiences with early RFA-technology and some results from low volume centers9. Sørensen et al found a 3- and 5-year survival of 64% and 44% respectively after RFA for unresectable CRLM10. This concurs with the results of a prospective study by Abitabile et al, reporting on 47 patients with CRLM (80% of which were unresectable), in which a 3-year OS rate of 57% was reached. Overall local recurrence rate was 8.8%. All CRLM ≤3cm (80.2% of lesions) were completely ablated, resulting in a local recurrence rate of 1.6%11. Gillams et al reported a median 3- and 5-year survival of 84% and 40% respectively with solitary CRLM of ≤4cm in diameter in 40 patients who were not candidates for resection12. Furthermore, these authors reported on 309 patients, who were treated with percutaneous RFA13. All patients were deemed inoperable for CRLM and were accepted for RFA with five or fewer lesions of ≤5cm, or as many as nine lesions of ≤4-4.5cm, or a solitary tumor of ≤7cm in diameter. Extrahepatic disease was not a contra-indication,

Page 177: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

168

provided it was stable on treatment. Presence of extrahepatic disease and liver tumor volume (defined by number and size of CRLM) were identified as significant survival factors. For patients with ≤5 CRLM of ≤5cm the 5-year survival from ablation was 24%, whereas this decreased significantly with higher lesion size or number or if extrahepatic disease was involved13. Siperstein et al prospectively reported on their 10-year experience with RFA for CRLM to assess the factors affecting long-term survival14. The inclusion criteria of this study allowed patients who failed chemotherapy, had presence of extrahepatic spread (23.5%), had up to 12 CRLM and a maximal lesion size of 10cm. A total of 292 RFA procedures were performed in 234 patients with CRLM. Actual 3- and 5-year overall survival were 20.2% and 18.4%, respectively14. Median OS was improved for ≤3 lesions versus >3 lesions. The presence of extrahepatic disease is known to predict poor DFS and OS after hepatic resection4. Currently available literature on RFA for CRLM in patients with extrahepatic varies in opinion4. In the study of Siperstein et al the presence of extrahepatic disease didn’t adversely affect OS in this series14. So, from retrospective studies as well as from non-randomized prospective studies, results of RFA for unresectable CRLM seem promising, with 5-year survival rates varying from 3-30%, depending on lesion size and number, physician’s RFA experience, etc . However, these results should be interpreted with caution, because of the non-randomized nature of these studies. On the other hand, for unresectable CRLM 5-year survival is rare after treatment with only systemic chemotherapy7. Whether RFA indeed results in superior survival compared to chemotherapy cannot be concluded from these data. This question can only be answered in randomized studies. In an attempt to create more clarity concerning the possible benefit of the combination RFA + systemic chemotherapy over chemotherapy-only in patients with a limited number of CRLM the EORTC designed the randomized

Page 178: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

169

controlled CLOCC-trial, in which patients were randomized between chemotherapy alone and chemotherapy plus RFA (+/- surgical resection). This study may for the first time provide solid evidence for the benefit of RFA in patients with unresectable CRLM, but definite results have to be awaited.

Resection of resectable CRLM versus RFA for (un)resectable CRLM

Combined treatment modalities with RFA In general, new modalities of treatment are reserved for patients not eligible to surgery. This implies a bias in patient selection and makes a fair comparison between techniques impossible15,16. There are concerns that RFA results in higher recurrence rates than liver resection15, but comparing results of different treatment modalities for CRLM is very difficult, as resectable CRLM are treated by liver resection and RFA is reserved for a “wider spectrum” of patients, in whom resection is contraindicated because of extrahepatic disease, vessel contiguity, comorbidity or an estimated insufficient residual volume of the liver remnant after resection. In other words, patients fulfilling the criteria for liver resection probably have a better prognosis17. Multiple papers compare liver resection for resectable lesions to RFA, with or without resection, for unresectable liver lesions [Table 1]. With 258 patients, Gleisner et al retrospectively analyzed a large patient cohort, including only patients with CRLM who were operated on with curative intent. Also, only RFA treatments performed at the time of open laparotomy were included in the study16. Overall survival (OS) rates of 72% at 3 years and 57.4% at 5-years were reported for the liver resection-only group, compared to 51.2% and 28.3% respectively in the RFA +/- resection group. Disease-free survival (DFS) at 3 and 5 years was 41.3% for the resection-only group compared to 8.9% and 0% respectively in the RFA+/-resection group16. These numbers concur with the other trials reported in Table 118-22. In the paper of Gleisner et al the

Page 179: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

170

combination of resection and RFA was associated with a significantly increased risk of extrahepatic failure at 1 year compared to patients who were undergoing only resection (p<0.05), but propensity score methods revealed differences in baseline tumor and treatment-related factors, resulting in a lack of comparability between resection-only resection+RFA groups. Furthermore, it should be mentioned that 11 patients underwent RFA alone (14 CRLM), of whom 5 patients had a solitary lesion ablated that abutted the confluence of the hepatic veins and was deemed unamenable to resection. This could make for a substantial bias 16. The prospective trial of Ruers et al and retrospective analysis Abdalla et al also included patients with liver-only disease in whom, at laparotomy, resection or local ablation were not feasible for technical reasons and who were subsequently treated systemic chemotherapy only18,19. In the study of Ruers et al patients treated with chemotherapy (N=39) only reached a median OS of 26 months with a 2- and 5-year OS of 51% and 15%, respectively18. The patient group undergoing local ablation (+/- resection) reached a 2- and 5-year OS survival of 56% and 27%, respectively18. Patient groups were too small to show any statistically significant difference between groups. On the other hand, Abdalla et al found a statistically significant difference in survival for patients treated with RFA as a component of therapy versus chemotherapy only, whether compared as a group (p=0.002) or when each subgroup was compared (p=0.005)19. Machi et al reported on 507 unresectable CRLM in 100 patients, treated with RFA, either after prior liver resection or before or after systemic chemotherapy. They found an OS at 3 and 5 years of 42% and 30.5%, respectively. The authors concluded RFA can contribute to encouraging long-term survival and appears to confer a survival benefit over systemic chemotherapy alone, particularly when performed as part of the first-line therapy20. A prospective study of Elias et al, treating unresectable CRLM with liver resection, RFA and

Page 180: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

171

chemotherapy as a combination, came to the same conclusion21. Concerning local failure of treatment, defined as recurrence of tumor at the initial treatment sites, resection seems to result in better results than RFA+/- resection [Table 1]. Still, given the bias in patient selection for the different treatment groups, this remains an impression. All authors agreed RCTs remain a necessity to assess the possible benefit of these combined-modality treatments.

RFA for unresectable solitary CRLM versus resection for solitary CRLM Table 2 summarizes the trials comparing RFA as a treatment tool for unresectable solitary CRLM to liver resection for resectable solitary CRLM. All of these studies had a retrospective design17,23-25. Oshowo et al reported a comparable 3-year OS for CT- or MRI-guided percutaneous RFA and liver resection (52.6% vs 55.4%, respectively)17, whereas the trials of White, Aloia and Hur reported a difference in OS and DFS in the advantage of liver resection23-25. After a separate analysis for patients with small (≤3cm) solitary CRLM, Aloia et al still reported a significant difference in local failure (i.e. 3% after liver resections versus 31% after RFA (p=0.001)) and 5-year OS (72% versus 18%, respectively (p=0.006))23. Hur et al, on the other hand, found similar 5-year survival rates for RFA and liver resection in solitary CRLM of ≤3cm in diameter (55.4% vs 56.1%) and similar local recurrence-free survival rates after 5 years (85.6% vs 95.7%)25. Abdalla wrote comments on this paper of Hur et al, stating that the relatively comparable outcome after treatment of small lesions should be viewed with optimism but caution, given the small number of patients in this study26. In the prospective study on multimodality treatment by Abdalla et al a sub-analysis showed that survival after treatment of a solitary CRLM by RFA was not comparable to survival after resection of a solitary lesion (p=0.025)19. However, in these non-randomized studies on RFA for solitary

Page 181: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

172

lesions there is a significant selection bias that reserves RFA for more difficultly situated lesions or high risk patients. Again this makes any firm conclusion impossible.

RFA as an alternative for resectable CRLM? At present, no evidence from RCTs is available to support the use of RFA as an alternative for the treatment for resectable CRLM. Nevertheless, some surgeons suggest RFA may replace resection, especially in certain circumstances, such as new hepatic metastases after previous liver resection or limited central disease that would require extended hemihepatectomy27. Elias et al described percutaneous RFA as an alternative to surgery for tumor recurrence after liver resection28. Incomplete local RFA treatment was observed in six of 47 patients (12.8%). Reported 1- and 2-year survival were 88% and 55%, respectively. The authors concluded percutaneous RFA increases the percentage of curative local treatments for patients with liver recurrence after hepatectomy28. Despite the current lack of evidence, a German survey by Birth et al showed RFA was already used as a tool for treatment of potentially curative resectable tumors in 25.9% of hospitals29.

RFA Approach For the sake of safety and minimizing local failure, it is mandatory that the lesions should be clearly visualized during RFA treatment. For surgical (open or laparoscopic) procedures intraoperative ultrasound is used. For percutaneous procedures ultrasound(US), as well as CT- or MRI-guidance are an option4. Intra-procedural US only provides a rough estimate of the size of ablation, since the bounderies of the hyperechoic area that arises during RFA do not automatically correlate with the actual coagulative damage. In a study by Cha et al an animal model was used to compare the monitoring of RFA by unenhanced CT

Page 182: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

173

versus US. CT indeed proved to be an effective way to monitor RFA, because of increased lesion discrimination, reproducible decreased attenuation during ablation, and improved correlation to pathologic size30. Both CT and MRI imaging are considered to be more reliable in this regard, although to date no RCTs have been performed to assess the preferable imaging modality9,31-34. A review of literature showed that local tumor recurrence rates varied from 6-40% and were related to the size, number and location of the lesions4. The indications for a percutaneous approach are a limited number of small tumors, preferably not adjacent to hollow viscera (but this can be overcome by pre-ablation percutaneous infusion of dextrose fluid into the space between the liver and the adjacent bowel), small recurrences after prior liver resection, and patients uneligible for a surgical approach for anatomic and/or clinical reasons. Multiple previous laparotomies or liver resections can be an indication for a percutaneous approach, because of a high probability of extensive adhesions. Furthermore, optimal visualization is mandatory4. Recently, Lencioni et al wrote an extensive review on percutaneous image-guided RFA for liver tumors8. They concluded that the currently available data in literature suggest that RFA can result in complete tumor eradication in properly selected candidates and provide indirect evidence that this treatment improves survival in nonsurgical patients with limited CRLM. It needs no further explanation that patients should be properly staged before treatment, in order to make an adequate patient selection for RFA. The number of lesions shouldn’t be considered an absolute contraindication for successful percutaneous RFA if adequate treatment of all lesions can be accomplished. On the other hand, tumor size is a very important predictive factor, but it should be realized that imaging studies tend to underestimate the real size of CRLM. In general, tumor size should not exceed a maximum diameter of 3-4cm27. Subsequent to advances in RFA technique and probes,

Page 183: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

174

eradication of up to 97% were described for lesions up to 4cm35,36. Table 3 summarizes the outcome of percutaneous RFA in CRLM. Although it may generally be felt that CT-guided procedures are more reliable, results for US- and CT-guided percutaneous approach are fairly similar20,37-39. This is probably partly due to the experience of the performing physician within the studies mentioned: from literature review Mulier et al concluded that authors who treat large numbers of patients had significantly fewer local recurrences. Significant improvement occurs after 40-50 cases, although the plateau phase in the learning curve is reached only at 100 procedures9,27. Nevertheless, as could be expected, larger lesions (> 5 cm) still result in worse outcomes9,13,31. Finally, a remark should be made on hospital stay and resulting costs: percutaneous RFA can be performed as a one-night hospital stay or even a day case, which substantially reduces costs when compared to laparoscopic or open RFA.

Morbidity & Mortality An ASCO review of recent literature shows RFA has a mortality of 0-2% and most commonly reported morbidity rate is 6-9%, with a low major complication rate4. A review by Stang et al showed a mortality of 0-3.7% and 13-27% major complications for open RFA, whereas this was 0% and 1.8-13% respectively for percutaneous RFA procedures40. In other words, RFA in general is considered to have a lower morbidity than major liver resection15 and for percutaneous RFA these numbers are even better4,8,15,40. Furthermore, literature study shows blood transfusion is rare after RFA and generally hospital stay is shorter27. Because of its low morbidity profile, percutaneous RFA can be repeated quite easily in case of local recurrence, with only very limited clinical consequences for the patient.

Page 184: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

175

Reflections and Conclusions The efficacy and reliability of RFA depends mainly on the diameter of the targeted lesions, the applied approach (surgical or percutaneous RFA) and the distance between the targeted lesion and large vessels (because of the ‘heat sink’ phenomenon). Obviously, larger CRLM require overlapping treatment zones, i.e. multiple RFA sessions, which makes the result less reliable9. RFA , for lesions larger > 5cm in diameter is even questionable9,31. The RFA-approach as such might progressively become a less interesting discussion point: Evidently, intra-operative ultrasound can be best performed in a surgical approach (and in particular open RFA) and the liver can be fully mobilized, which provides the optimal conditions for adequate placement of the RFA probe31, but basically the indication for each approach is a matter of patient selection. In case of grossly resectable disease with a few unresectable lesions there’s a preference for open RFA. On the other hand, patients with unresectable CRLM, who are not fit for major surgery due to extensive comorbidity, will be approached as minimally invasive as possible. Furthermore, the experience of the physician performing RFA appears to be an important factor influencing outcome9,27, the use of CT- or MRI-guidance improves the accuracy of the treatment31 and newer generation probes seem to result in lower local recurrence27. Therefore, in some highly specialized centers a percutaneous approach does seem to produce equivalent results to those achieved by liver resection in well-selected patients9.

For unresectable CRLM, currently available prospective and retrospective data18,19 strongly suggest a benefit of any combined modality treatment with RFA over a chemotherapy-only approach. The final results of the CLOCC-trial are awaited.

In current literature, there’s a lack of evidence for the use of RFA as an alternative for surgery in patients with resectable CRLM. Available series on RFA for small, solitary

Page 185: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

176

lesions have a contradictive outcome [Table 2]26. Nevertheless, one can assume that RFA for small, solitary lesions would result in a different (better) outcome if patient selection is shifted from unresectable to resectable CRLM, implying more favorable tumor biology, better localization etcetera. Furthermore, this would result in a more parenchymal sparing policy, in which extended liver resections could be avoided for small, but centrally located lesions, thus leaving in place a far larger liver remnant3,27. This would leave more room for retreatment of future local recurrences or newly developed CRLM at other sites of the liver.

Because the majority of papers currently available in literature “compare” RFA treatment for unresectable CRLM with liver resection for resectable CRLM, interpretation of the results in any direction remains very difficult and dangerous. The only real solution to this persistent deadlock is the design of a RCT for carefully chosen, resectable, small (≤3cm), solitary CRLM to be treated with RFA versus resection. Several authors expressed their sincere concerns about the dangers of conducting such a trial, as it might encourage inappropriate use of RFA by the occasional practicioner of thermal tumor ablation41. But one could also turn this argument around in favor of a RCT: the reality of clinical practice is indeed already catching up, as can be concluded from the survey of Birth et al29. Physicians are already autonomously interpreting the available data and, despite the lack of real evidence, deciding for themselves to treat selected resectable patients with RFA. If a RCT is not performed soon, determining what is really “good clinical practice”, this will become an impossible adventure.

Page 186: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

177

Table 1: Comparison of treatment modalities with/without RFA

Author Journal

N Patients

Therapy modality N OS (%) DFS (%) Local Recurrence at treatment sites (% patients) 3 years 5 years 3 years 5 years

Gleisner16

Arch Surg 2008

258 Resection alone 192 72.0 57.4 41.3 41.3 14.8**

RFA +/- resection 66 51.2 28.3 8.9 0.0 50.9-62.5**

Ruers18

Ann Surg Oncol 2007

201 Resection 117 65.0 51.0 35.0 32.0 0.9

Local ablation 45 40.0 27.0 15.0 11.0 11.0

Chemotherapy 39 37.5 15.0 - - -

Machi20

Cancer J 2006

100 RFA after previous liver resection 8 42.0 30.5 23.2 21.7 20.5** First-line RFA (before chemo) 55

Second-line RFA (after chemo) 45

Elias21

J Surg Oncol 2005

63 Liver resection +

RFA

55 47.0 - 27.0 - 7.2 at RFA sites***

154 7.2-9.0 at resection sites***

Abdalla19

Ann Surg 2004

418 Resection only 190 73.0 58.0 41.1 30.0 2.0

RFA + resection 101 43.0 - 16.7 - 5.0

RFA only 57 37.0 - 4.4 - 9.0

Chemotherapy only 70 13.2 7.7 - - -

Leblanc22

EJSO 2008

99 RFA alone 34 75.0* - - - 12.0

RFA + resection 28 68.0* - - - 8.0

Resection alone 37 83.0* - - - 6.0

*: OS at 2 years **: recurrence anywhere in the liver ***: % procedures

Page 187: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

178

Table 2: RFA for unresectable Solitary CRLM versus Resection for resectable CRLM

Author Journal

N patients Lesion Size (cm) [range]

Treatment modality OS (%) Local failure (% patients) 3 years 5 years

Oshowo17

BJS 2003

25 3 [1-10]

CT- / MRI-guided percutaneaous RFA 52.6 42.3 ns

20 4 [2-7]

Resection 55.4 34.2 15.0

White24

J Gastrointest Surg

2007

22 2 [1-5]

CT-guided percutaneous RFA 24.3 - 45.5

30 2.5 [1-5]

Resection 81.4 58.6 3.3

Aloia23

Arch Surg 2006

30 ≤3cm: 53% >3cm: 47%

Percutaneous/open RFA 57.0 27.0 37.0

150 ≤3cm: 42% >3cm: 58%

Resection 79.0 71.0 5.0

Hur25

EJSO 2009

25 ≤3cm: 60% Percutaneous/open RFA 77.9 55.4 28.0

>3cm: 40% 42.0

42 ≤3cm: 54.8% Resection 81.0 56.1 9.5

>3cm: 45.2% 30.0

ns = not specified

Page 188: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

179

Table 3: Percutaneous Imaging-guided RFA for unresectable CRLM

Author Journal

N patients

N procedures

N lesions

Approach Imaging guidance

Lesion size OS (%) Local recurrence (% lesions)

3 years 5 years

Veltri A37 Cardiovasc Intervent

Radiol 2008

122 166 199 Percutaneous: N=177 Surgical: N=22

US-guided (sometimes

contrast-enhanced)

All sizes 38.0 22.0 26.3

≤3 cm 50.0 27.5 33.3

>3 cm 32.5 12.5 66.7

(p=0.006) (p<0.0001)

Solbiati L38 Radiology

2001

109 162 172 Percutaneous: all CT- or US-guided All sizes 33.0 - 29.6

≤3 cm - - 16.5

>3 cm - - 56.1

(p=sign.)

Machi J20 Cancer J 2006

100 146 507 Percutaneous: N=61 Surgical: N=85

US-guided All sizes 42.0 30.5 6.7

≤1 cm Med.OS 40.0 months

-

>1 cm Med.OS 22.0 months

-

(p=0.0026)

Gillams A13 Eur Radiol

2009

309 617 - Percutaneous: all

CT- or US-guided <5 lesions of ≤5 cm, no extrahepatic disease 49.0 24.0 -

<5 lesions of ≤5 cm 40.0 18.0 -

>5 lesions and/or >5 cm 13.0 3.0 -

(p=0.000) -

Jakobs39 Anticancer Res

2006

68 - 183 Percutaneous: all CT-guided - 68.0 - 18.0

Page 189: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

180

References 1. House M, Ito H, Gönen M. Survival after hepatic

resection for metastatic colorectal cancer: trends in outcome for 1,600 patients during 2 decades at a single institution. J Am Coll Surg 2010; 210: 744-752.

2. Nordlinger B, Sorbye H, Glimelius B. Perioperative chemotherapy with FOLFOX4 and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC Intergroup trial 40983): a randomized controlled trial. Lancet 2008; 371(9617): 1007-1016.

3. Mulier S, Ruers T, Jamart J. Radiorequency ablation versus resection for resectable colorectal liver metastases: time for a randomized trial? An update. Dig Surg 2008; 25(6): 445-460.

4. Wong S, Mangu P, Choti M. American Society of Clinical Oncology 2009 clinical evidence review on radiofrequency ablation of hepatic metastases from colorectal cancer. J Clin Oncol 2010; 28(3): 493-508.

5. Crocetti L, de Baere T, Lencioni R. Quality improvement guidelines for RFA of liver tumours. Cardiovasc Intervent Radiol 2010; 33(1): 11-17.

6. Thomas K, Bream P, Berlin J. Use of percutaneous drainage to treat hepatic abscess after radiofrequency ablation of metastatic pancreatic adenocarcinoma. Am Surg 2004; 70(6): 496-9.

7. Wolpin B, Mayer. Systemic treatment of colorectal cancer. Gastroenterology 2008; 134(5): 1296-1310.

8. Lencioni R, Crocetti L, Pina MC. Percutaneous image-guided radiofrequency ablation of liver tumors. Abdom Imaging 2009; 34(5): 547-556.

9. Mulier S, Ni Y, Jamart J. Local recurrence after hepatic radiofrequency coagulation: multivariate meta-analysis and review of contributing factors. Ann Surg 2005; 242(2): 158-171.

Page 190: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

181

10. Sørensen S, Mortensen F, Nielsen D. Radiofrequency ablation of colorectal liver metastases: long-term survival. Acta Radiol 2007; 48(3): 253-258.

11. Abitabile P, Hartl U, Lange J. Radiofrequency ablation permits an effective treatment fro colorectal liver metastases. Eur J Surg Oncol 2007; 33(1): 67-71.

12. Gillams A, Lees W. Five-year survival following radiofrequency ablation of small, solitary, hepatic colorectal metastases. J Vasc Interv rediol 2008; 19(5): 712-717.

13. Gillams A, Lees W. Five-year survival in 309 patients with colorectal liver metastases treated with radiofrequency ablation. Eur Radiol 2009; 19(5): 1206-1213.

14. Siperstein A, Berber E, Ballem N. Survival after radiofrequency ablation of colorectal liver metastases: 10 years experience. Ann Surg 2007; 246(4): 559-565.

15. Gurusamy K, Ramamoorthy R, Imber C. Surgical resection versus non-surgical treatment for hepatic node positive patients with colorectal liver metastases. Cochrane Database Systematic Review 2010 Jan 20; (1): CD006797.

16. Gleisner A, Choti M, Pawlik T. Colorectal liver metastases: recurrence and survival following hepatic resection radiofrequency ablation and combined resection-radiofrequency ablation. Arch Surg 2008; 143(12): 1204-1212.

17. Oshowo A, Gillams A, Harisson E. Comparison of resection and radiofrequency ablation for treatment of solitary colorectal liver metastases. Br J Surg 2003; 90(10): 1240-1243.

18. Ruers T, Joosten J, Wiering B. Comparison between local ablative therapy and chemotherapy for non-resectable colorectal liver metastases: a prospective study. Ann Surg Oncol 2007; 14(3): 1161-1169.

Page 191: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

182

19. Abdalla E, Vauthey J, Ellis L. Recurrence and outcomes following hepatic resection, radiofrequency ablation and combined resection/ablation for colorectal liver metastases. Ann Surg 2004; 239(6): 818-827.

20. Machi J, Oishi AJ, Sumida K. Long-term outcome of radiofrequency ablation for unresectable liver metastases from colorectal cancer: evaluation of prognostic factors and effectiveness in first- and second-line management. Cancer J 2006; 12(4): 318-326.

21. Elias D, Baton O, Sideris L. Hepatectomy plus intraoperative radiofrequency ablation and chemotherapy to treat technically unresectable multiple colorectal liver metastases. J Surg Oncol 2005; 90(1): 36-42.

22. Leblanc F, Fonck M, Evrard S. Comparison of hepatic recurrences after resection or intraoperative radiofrequency ablation indicated by size and topographical characteristics of the metastases. Eur J Surg Oncol 2008; 34(2): 185-190.

23. Aloia T, Vauthey J, Loyer E. Solitary colorectal liver metastases: resection determines outcome. Arch Surg 2006; 141(5): 460-466; discussion 466-467.

24. White R, Avital I, D’Angelica M. Rates and patterns of recurrence for percutaneous radiofrequency ablation and open wedge resection for solitary colorectal liver metastases. J Gastrointest Surg 2007; 11(3): 256-263.

25. Hur H, Ko Y, Min B. Comparative study of resection and radiofrequency ablation in the treatment of solitary colorectal liver metastases. Am J Surg 2009; 197(6): 728-736.

26. Abdalla E. Commentary: Radiofrequency ablation for colorectal liver metastases: do not blame the biology when it is the technology. Am J Surg 2009; 197(6): 737-739.

27. Mulier S, Ni Y, Ruers T. Radiofrequency ablation versus resection for resectable colorectal liver metastases:

Page 192: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

183

time for a randomized trial? Ann Surg Oncol 2008: 15(1): 144-157.

28. Elias D, de Baere T, Smayra T. Percutaneous radiofrequency thermoablation as an alternative to surgery for treatment of liver tumor recurrence after hepatectomy. Br J Surg 2002; 89(6): 752-756.

29. Birth M, Hildebrand P, Dahmen G. Aktueller Stand der Radiofrequenzablation von lebertumoren: eine deutschlandweite Umfrage. Chirurg 2004; 75(4): 417-423.

30. Cha C, Lee F, Gurney J. CT versus sonography for monitoring radiofrequency ablation in a porcine liver. AJR AJR Am J Roentgenol 2000; 175(3):705-11.

31. Joosten J, Ruers T. Local radiofrequency ablation techniques for liver metastases of colorectal cancer. Crit Rev Oncol Hematol 2007: 62(2): 153-163.

32. Meloni M, Goldberg N, Livraghi T. Hepatocellular carcinoma treated with radiofrequency ablation: comparison of pulse inversion contrast-enhanced harmonic sonography, contrast-enhanced power Doppler sonography, and helical CT. AJR Am J Roentgenol 2001; 177(2): 375-380.

33. Mahnken A, Buecker A, Spuentrup E. MR-guided radiofrequecy ablation of hepatic malignancies at 1.5T: initial results. J Magn Reson Imaging 2004; 19(3): 342-348.

34. Lencioni R, Crocetti L, Cioni D. Percutaneous radiofrequency ablation of hepatic colorectal metastases: technique, indications, results and new promises. Invest Radiol 2004; 39(11): 689-697.

35. de Baere T, Elias D, Dromain C. Radiofrequency ablation of 100 hepatic metastases with a mean follow-up of more than 1 year. AJR Am J Roentgenol 2000: 175(6): 1619-1625.

Page 193: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

184

36. Helmberger T, Holzknecht N, Schöpf U. Radiofrequency ablation of liver metastases. Technique and initial results. Radiologe 2001; 41(1): 69-76.

37. Veltri A, Sacchetto P, Tosetti I. Radiofrequency ablation of colorectal liver metastases: small size favourably predicts technique effectiveness and survival. Cardiovasc Intervent Radiol 2008; 31(5): 948-956.

38. Solbiati L, Ierace T, Tonolini M. Radiofrequency thermal ablation of hepatic metastases. Eur J Ultrasound 2001; 13(2): 149-158.

39. Jakobs T, Hoffmann R, Helmberger T. Radiofrequency ablation of colorectal liver metastases: mid-term results in 68 patients. Anticancer Res 2006; 26(1B): 671-680.

40. Stang A, Fischbach R, Teichman W. A systematic review on the clinical benefit and role of radiofrequency ablation as treatment of colorectal liver metastases. Eur J Cancer 2009; 45(10): 1748-1756.

41. Curley A. Radiofrequency ablation versus resection for resectable colorectal liver metastases: time for a randomized trial? Ann Surg Oncol 2007; 15(1): 11-13.

Page 194: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

185

IV

2. Morbidity and mortality of laparoscopic vs. open radiofrequency ablation for hepatic malignancies

Topal B, Hompes D, Aerts R, Fieuws S,

Thijs M, Penninckx F Eur J Surg Oncol 2007; 33: 603-607.

Page 195: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

186

Page 196: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

187

Morbidity and mortality of laparoscopic vs. open radiofrequency

ablation for hepatic malignancies

B.Topal1, D.Hompes1, R. Aerts1, S. Fieuws2, M. Thijs3, F. Penninckx1

1 Department of Abdominal Surgery, 2 Department of Biostatistics, 3 Department of Radiology, University Hospital Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium

Introduction Hepatic resection offers the only chance of long-term

survival for selected patients with primary or metastatic liver cancer. Presence of extra-hepatic disease and extensive hepatic tumour burden is most common contraindication for resection. The vast majority of patients with liver malignancies therefore are not the candidates for surgical treatment. Several other therapeutic modalities are available and are considered palliative. Over the past decade, radiofrequency ablation (RFA) of liver tumours has gained widespread use. At this point, the role of RFA is considered complementary to surgical resection, but it may also represent a good alternative in selected patients who are at high risk for extra-hepatic cancer recurrence or who are poor candidates for resection1.

Radiofrequency ablation can be performed percutaneous, by laparotomy, or laparoscopy. Most patients are treated percutaneous, while only a few centres report the laparoscopic approach1-6. Early complications following RFA are more likely to occur in patients treated with open RFA (7.1%) compared with percutaneous RFA (4.4%)7. In contrast, RFA by laparoscopy or laparotomy is able to achieve superior local tumour control compared to percutaneous RFA, which is associated with local recurrence rates of up to 60%. Therefore, the short-term clinical benefits of percutaneous RFA do not

Page 197: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

188

overweigh the longerterm oncological outcome, indicating that percutaneous approach should be reserved for patients unfit for surgery8. The purpose of the present cohort study was to compare morbidity and mortality of laparoscopic (LRFA) vs. open (ORFA) radiofrequency ablation of liver cancer, and to define variables that can predict the occurrence of complications after RFA.

Methods Patients and tumours From October 1999 until November 2006, 154

consecutive patients with liver cancer were enrolled in a prospective non-randomized study to undergo RFA. Percutaneous approach was used in 12 patients (not candidates for laparotomy or laparoscopy) to treat 14 tumours. These patients were excluded from the present study. The male/female ratio was 93/49 and the median (range) age 62 (35-84) years. Patient co-morbidity was assessed using the American Society of Anaesthesiology (ASA) score, i.e. ASA II, 81 patients; ASA III, 41 patients, and ASA IV, 2 patients. Cirrhosis was present in 54 patients: Childe Pugh A 36, B 15 and C 3. Systemic chemotherapy or transarterial hepatic chemo-embolization (TACE) within 3 months prior to RFA was given in 60 patients.

All patients had histologically proven primary (n 56) or metastatic (n 86) liver cancer. A total of 277 liver tumours were treated with RFA: 76 hepatocellular carcinomas (HCC), 153 colorectal liver metastases (CRLM), and 48 other hepatic malignancies.

Surgical procedure Two hepatobiliary surgeons performed the RFA procedures. Tumour ablation was accomplished under ultrasound guidance, using a monopolar RF generator and a

Page 198: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

189

single or cluster cool-tip electrode (Tyco Healthcare, Radionics Europe NV), as described earlier9. Radiofrequency ablation was performed by laparotomy in 49 patients for 110 liver tumours, and by laparoscopy in 93 patients for 167 tumours. Pringle’s manoeuvre was performed in 6 patients during 4-12 min. Seventy-four patients underwent additional surgery simultaneously with the RFA procedure: colorectal resection in 22, hepatic resection in 22, and minor surgery in 44 patients.

Assessment of clinical outcome Postoperative complications were classified based on

the therapy-oriented severity grading system (TOSGS; grade 1: no need for specific intervention; grade 2: need for drug therapy; grade 3a: need for invasive therapy without general anaesthesia; grade 3b: invasive therapy under general anaesthesia; grade 4a: single organ dysfunction (including dialysis) with ICU stay; grade 4b: multiorgan dysfunction requiring ICU management; grade 5: death)10, and allocated to surgical site (SSC) vs. non-surgical site complications (NSSC). Surgical site complications were subdivided into hepatic (HSC) and non-hepatic site (NHSC) complications.

Statistical analysis The relation of a set of predictors (categorical and

continuous) with the presence of a postoperative complication was explored using Fisher’s exact, Cochran-Armitage trend and Mann-Whitney U tests. Due to the low rate of events (postoperative complications) and high number of variables, no multivariate analysis was performed. The following variables that could influence clinical outcome were taken into account: year of surgery, age, gender, ASA score, primary/metastatic liver cancer, type of liver tumour (HCC/CRLM/other), number of hepatic tumours, maximum tumour diameter (mm), hepatic tumour location (right/left liver), liver segment involvement (segments 1-8; yes/no), number of liver segments involved, cirrhosis (yes/no),Child-Pugh class A/B/C, pre-operative

Page 199: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

190

chemotherapy or TACE (yes/no), approach (percutaneous/laparotomy/laparoscopy; yes/no), duration of RFA (min), duration of surgery (min), intra-operative blood loss (ml), Pringle’s manoeuvre (yes/no), additional surgical procedure (yes/no), type of additional surgery (colorectal resection/hepatic resection/other; yes/no).

A p-value of ≤ 0.05 was considered statistically significant. Due to the exploratory character of the study, no corrections have been made for multiple testing. All analyses were performed using the statistical software SAS (version 9.1).

Results Morbidity and mortality

Postoperative complications were observed in 25 patients, with subsequent mortality in 2 patients. One patient with Child-Pugh C cirrhosis died because of progressive liver failure 5 weeks after LRFA for HCC. The other patient died because of myocardial infarction on the day after ORFA for CRLM.

Complication rate in patients who underwent RFA combined with another surgical procedure was 16/74. Complications after simultaneous hepatic resection were observed in 3/22 patients, and after simultaneous colorectal resection in 10/22 patients.

According to the TOSGS of complications, 10 patients were classified grade 2, 3 grade 3a, 8 grade 3b and 2 grade 4a. Surgical site complications occurred in 17 patients (HSC 6, NHSC 12), and NSSC in 9 patients. Median length of hospital stay (LOS) was 6 days (range 1-127) for all patients.

Laparoscopy vs. laparotomy Patient and tumour characteristics with respect to ORFA

vs. LRFA are presented in Table 1. The majority of patients with HCC and cirrhosis were treated by laparoscopy (p < 0.01). Laparotomy was performed more frequently in

Page 200: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

191

patients with metastatic liver cancer and in patients who underwent simultaneous colorectal and/or hepatic resection (p < 0.01). Clinical outcome data of ORFA vs. LRFA are presented in Table 2. As compared with the LRFA-group, the ORFA-group was associated with significantly higher intra-operative blood loss, longer duration of surgery, more postoperative complications, and longer postoperative hospital stay (p < 0.01). According to the TOSGS classification, postoperative complications in the ORFA-group were more severe than those in the LRFA-group (p < 0.01).

After exclusion of patients who underwent simultaneous colorectal and/or hepatic resection, 103 patients remained for subgroup analysis. Clinical outcome parameters were in favour of LRFA (n 77). However, tumour diameter in the ORFA-group was significantly larger as compared to that in the LRFA-group [Tables 3 and 4]. Therefore, in order to better assess the role of laparotomy vs. laparoscopy, only patients with liver tumours measuring ≤ 30mm in diameter were evaluated. As compared with the LRFA-group (n 61), the ORFA-group (n 12) was associated with higher intra-operative blood loss (22.5 (0-600) vs. 10 (0-200)ml; p < 0.01), longer duration of surgery (147 (90-370) vs. 70 (30-230) min; p <0.01), more postoperative complications (3 vs. 3; p = 0.02), more NSSC (1 vs. 0; p = 0.02), and longer postoperative hospital stay (7(3-15) vs. 3 (1-10) d; p < 0.01). According to the TOSGS classification, postoperative complications in the ORFA-group were more severe than those in the LRFA-group (p < 0.01).

Predictors of postoperative complications In univariate analysis the following variables were significantly (p < 0.01) related to the presence of postoperative complications: simultaneous colorectal resection, laparotomy, duration of surgery, tumour location in right liver, liver segment 7 (p = 0.01), absence of cirrhosis (p = 0.02), liver segment 8 (p = 0.03), and metastatic liver cancer (p = 0.04).

Page 201: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

192

Discussion Morbidity of RFA

Radiofrequency ablation of hepatic malignancies by laparotomy or laparoscopy provides superior oncological outcome as compared to percutaneous RFA8. Surgical RFA of liver cancer is mostly performed by laparotomy while only few reports are available on RFA through minimally invasive surgery or laparoscopy1-6. Reported complication rates after ORFA for liver cancer range from 8.6% to 9.9%, whereas the complication rate of ORFA combined with hepatic resection is around 31%.7,11. Indeed, additional surgery may increase complication rate of RFA for hepatic cancer, as observed in the present study. The complication rate after RFA with simultaneous hepatic resection was 13.6% and 45.4% after simultaneous colorectal resection. Several factors were found as potential (univariate) predictors of postoperative complications: simultaneous colorectal resection, laparotomy, duration of surgery, tumour location in the right liver, involvement of liver segment 7, absence of cirrhosis, involvement of liver segment 8, and metastatic liver cancer. Due to the low rate of events and high number of variables, no multivariate analysis was made.

Type of surgical approach for RFA With advancing technology laparoscopic liver surgery has become feasible and safe, but not yet routinely implemented. The potential minimally invasive benefits of laparoscopic surgery seem to be applicable for RFA of hepatic malignancies as well. In the present study, as compared with ORFA, the LRFA-group was associated with significantly lower intra-operative blood loss, shorter duration of surgery, fewer postoperative complications, and shorter postoperative hospital stay. The complications after LRFA were also less severe than those in the ORFA-group. These benefits were most pronounced in patients with HCC and cirrhosis, patients that

Page 202: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

193

are considered to be at high risk for postoperative complications. The favourable clinical outcome of LRFA remained consistent in patients without simultaneous colorectal and/or hepatic resection and even in patients with small liver tumours (≤ 3cm). Patients with liver tumours measuring 3 cm or less seem to be excellent candidates for LRFA, whereas larger tumours are treated preferably via laparotomy because of technical considerations. However, the non-randomized nature and the small number of patients, especially in the subgroup analyses, are limitations of the present study to draw final conclusions.Therefore, larger patient series in randomized controlled trials are needed to better evaluate LRFA compared with ORFA.

Survival after RFA In terms of postoperative morbidity, LRFA for hepatic

malignancies seems to be preferable above ORFA, provided that the oncological outcome is not jeopardised. The longterm outcome of patients treated in the present study is under evaluation. The effectiveness of RFA with respect to local tumour control has been evaluated in several studies2-4,8. Sufficient data are available with regard to excellent long-term outcome in using RFA to treat small HCC whereas only few report on CRLM12-15,17. In patients with colorectal liver metastases RFA appeared to have a positive impact on overall survival and to achieve excellent local control for metastases smaller than 3 cm in diameter2,16. On the other hand, RFA alone or in combination with resection for unresectable patients did not provide survival comparable to resection only17,18. These data indicate the heterogeneity of the published series and the emerging need of randomized controlled trials on the role of RFA in patients with resectable CRLM. Indeed, today hepatic resection still remains the treatment of choice for CRLM.

Page 203: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

194

Conclusion Laparoscopic radiofrequency ablation for hepatic

malignancies is associated with better short-term clinical outcome as compared to open RFA, especially for hepatocellular carcinoma in cirrhosis. Simultaneous colorectal and/or hepatic resection results in an increased postoperative complication rate.

Acknowledgements Many thanks to staff members of the Departments of

Hepatobiliary Diseases (D. Cassiman, F. Nevens, W. Vansteenbergen, C. Verslype, and P. Yap) and Digestive Oncology (S. Tejpar, E. Van Cutsem) for including patients in this study.

Page 204: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

195

Table 1: Demographics of patients treated by open vs. laparoscopic RFA for liver cancer

ORFA (n 49) LRFA (n 93) p-Value

M/F 31/18 62/31 0.68 Age (y) 63 (37-80) 61 (35-84) 0.15 Primary/metastatic cancer 11/38 45/48 0.003 HCC/CRLM/other 10/28/11 44/37/12 0.007 Cirrhosis 9 45 0.0005 Child-Pugh class A/B/C 8/1/0 28/14/3 0.003 Number of tumours 1 (1-8) 1 (1-9) 0.89 Tumour diameter (mm) 25 (8-90) 22 (8-58) 0.25 Additional surgical procedure 30 43 0.09 Colorectal resection 14 8 0.002 Hepatic resection 12 10 0.03

Continuous variables are presented as median (range). CRLM, colorectal liver metastasis; HCC, hepatocellular carcinoma; M, male; F, female; RFA, radiofrequency ablation; LRFA, laparoscopic RFA; ORFA, open RFA.

Page 205: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

196

Table 2: Clinical outcome after open vs. laparoscopic RFA of liver cancer

ORFA (n 49) LRFA (n 93) p-Value

Duration surgery (min) 180 (25-440) 75 (30-390) <0.0001 Blood loss (ml) 20 (0-1700) 10 (0-900) 0.0001 Mortality 1 1 0.64 Postoperative complication 17 8 0.0001 NSSC 9 1 0.0001 SSC 10 7 0.02 HSC 4 3 0.20 NHSC 12 5 0.0009 TOSGS 1/2/3/4/5 0/8/6/2/1 0/2/5/0/1 0.001 LOS (d) 8 (1-127) 4 (1-51) <0.0001

Continuous variables are presented as median (range). HSC, hepatic site complication; NHSC, non-hepatic site complication; LOS, length of hospital stay; RFA, radiofrequency ablation; LRFA, laparoscopic RFA; ORFA, open RFA; SSC, surgical site complication; NSSC, non-surgical site complication;TOSGS, therapy-oriented severity grading system.

Page 206: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

197

Table 3: Demographics of patients treated by open vs. laparoscopic RFA for liver cancer, without simultaneous colorectal and/or hepatic resection

ORFA (n 26) LRFA (n 77) p-Value

M/F 14/12 50/27 0.31 Age (y) 67 (47-80) 62 (35-80) 0.05 Primary/metastatic cancer 8/18 43/34 0.03 HCC/CRLM/other 7/12/7 42/24/11 0.046 Cirrhosis 8 44 0.02 Number of tumours 1 (1-7) 1 (1-9) 0.62 Tumour diameter (mm) 33.5 (10-90) 25 (8-58) 0.001 N of segments involved 2 (1-6) 1 (1-6) 0.04

Continuous variables are presented as median (range). CRLM, colorectal liver metastasis; HCC, hepatocellular carcinoma; M, male; F, female; RFA, radiofrequency ablation; LRFA, laparoscopic RFA; ORFA, open RFA.

Page 207: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

198

Table 4: Clinical outcome after open vs. laparoscopic RFA for liver cancer, without simultaneous colorectal and/or hepatic resection

ORFA (n 26) LRFA (n 77) p-Value

Duration of RFA (min) 50 (10-210) 20 (8-125) <0.0001 Duration surgery (min) 182 (60-385) 75 (30-230) <0.0001 Blood loss (ml) 37 (0-1700) 10 (0-200) <0.0001 Mortality 1 1 0.42 Postoperative complication 9 6 0.0008 NSSC 5 0 <0.0001 TOSGS 1/2/3/4/5 0/5/2/1/1 0/2/3/0/1 0.008 LOS (d) 7 (1-32) 4 (1-25) <0.0001

Continuous variables are presented as median (range). LOS, length of hospital stay; RFA, radiofrequency ablation; LRFA, laparoscopic RFA; ORFA, open RFA; NSSC, non-surgical site complication; TOSGS, therapy-oriented severity grading system.

Page 208: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

199

References 1. Fahy B, Jarnagin W. Evolving techniques in the

treatment of liver colorectal metastases: role of laparoscopy, radiofrequency ablation, microwave coagulation, hepatic arterial chemotherapy, indications and contraindications for resection, role of transplantation, and timing of chemotherapy. Surg Clin North Am 2006; 86: 1005–1022.

2. Berber E, Pelley R, Siperstein A. Predictors of survival after radiofrequency thermal ablation of colorectal cancer metastases to the liver: a prospective study. J Clin Oncol 2005; 23: 1–7.

3. Santambrogio R, Opocher E, Costa M. Survival and intra-hepatic recurrences after laparoscopic radiofrequency of hepatocellular carcinoma in patients with liver cirrhosis. J Surg Oncol 2005; 89: 218–226.

4. Decadt B, Siriwardena A. Radiofrequency ablation of liver tumours: systematic review. Lancet Oncol 2004; 5: 550–560.

5. Chung M, Wood T, Tsioulias G, Rose D, Bilchik A. Laparoscopic radiofrequency ablation of unresectable hepatic malignancies. A phase 2 trial. Surg Endosc 2001; 15: 1020–1026.

6. Siperstein A, Garland A, Engle K. Laparoscopic radiofrequency ablation of primary and metastatic liver tumors. Technical considerations. Surg Endosc 2000; 14: 400–405.

7. Curley S, Marra P, Beaty K. Early and late complications after radiofrequency ablation of malignant liver tumors in 608 patients. Ann Surg 2004; 239: 450–458.

8. Mulier S, NiY, Jamart J. Local recurrence after hepatic radiofrequency coagulation. Multivariate meta-analysis and review of contributing factors. Ann Surg 2005; 242: 158–171.

9. Topal B, Aerts R, Penninckx F. Laparoscopic radiofrequency ablation of unresectable liver

Page 209: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

200

malignancies: feasibility and clinical outcome. Surg Laparosc Endosc Percutan Tech 2003; 13: 11–15.

10. Dindo D, Demartines N, Clavien P. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg 2004; 240: 214–215.

11. Mulier S, Mulier P, Ni Y. Complications of radiofrequency coagulation of liver tumours. Br J Surg 2002; 89: 1206–1222.

12. Wood T, Rose D, Chung M. Radiofrequency ablation of 231 unresectable hepatic tumors: indications, limitations, and complications. Ann Surg Oncol 2000; 7: 593–600.

13. Curley SA, Izzo F, Delrio P. Radiofrequency ablation of unresectable primary and metastatic hepatic malignancies: results in 123 patients. Ann Surg 1999; 230: 1–8.

14. Chow D, Sinn L, Kelvin K. Radiofrequency ablation for hepatocellular carcinoma and metastatic liver tumors: a comparative study. J Surg Oncol 2006; 94: 565–571.

15. De Baere T, Elias D, Dromain L. Radiofrequency ablation of 100 hepatic metastases with a mean follow-up of more than 1 year. AJR Am J Roentgenol 2000; 175: 1619–1625.

16. Abitabile P, Hartl U, Lange J. Radiofrequency ablation permits an effective treatment for colorectal liver metastasis. Eur J Surg Oncol 2007; 33: 67–71.

17. Abdalla E, Vauthey J, Ellis L. Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases. Ann Surg 2004; 239: 818–825; discussion 825-827.

18. Poston GJ, Adam R, Alberts S. OncoSurge: a strategy for improving resectability with curative intent in metastatic colorectal cancer. J Clin Oncol 2005; 23: 7125–34.

Page 210: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

201

IV

3. Laparoscopic liver resection using radiofrequency coagulation

Hompes D, Aerts R, Penninckx F, Topal B

Surg Endosc 2007; 21: 175-180.

Page 211: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

202

Page 212: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

203

Laparoscopic liver resection using radiofrequency coagulation

D.Hompes, R.Aerts, F.Penninckx, B.Topal

Department of Abdominal Surgery, University Hospital Gasthuisberg, Leuven, Belgium

Introduction Intraoperative blood loss is a major concern in liver

surgery. In classical, open, hepatic resection, several surgical and anaesthesiological measures are known to limit intraoperative blood loss. The use of radiofrequency energy has been described to perform liver resection safely and with minimal blood loss. However, these results are based on few studies with small patient series, in which only open surgical procedures were performed17,27,32,37. With improvements in technology and hepatobiliary surgery, laparoscopic liver resection has become feasible and safe. Although only few reports are available on laparoscopic liver resection (LLR), this type of resection may offer the advantages of minimally

invasive surgery, provided there is adequate patient selection, surgical expertise, technological equipment,

and infrastructure1–11,13,14,18,21,29,35. No data are available on the potential contribution of RF

energy to the limitation of blood loss during laparoscopic liver resection. The aim of the present study was to investigate the role of radiofrequency energy to reduce intraoperative blood loss in patients undergoing laparoscopic liver resection.

Patients and methods From October 2002 until September 2005, forty-five

consecutive patients were enrolled in a prospective, nonrandomized study to undergo laparoscopic partial hepatectomy. The male/female ratio was 22/23, with a median

Page 213: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

204

age of 57 years (range 26–80 years). A laparoscopic procedure was considered in patients without severe systemic disease according to the American Society of Anaesthesiology (ASA score < 3). Patient selection was based on the judgment of the hepatobiliary surgeon. Radiofrequency energy was applied in 20 patients, whereas 25 patients underwent LLR without RF assistance. The surgeon decided at random whether or not to use RF for parenchymal transection. Fifty-eight hepatic tumors were resected (47 malignant, 11 benign), of which the majority were metastases from colorectal cancer [Table 1].

Two hepatobiliary surgeons performed the laparoscopic procedures. In order to prevent the occurrence of clinically significant gas embolism, the intra-abdominal pressure (CO2 pneumoperitoneum) was kept as low as possible, aiming for 6 to 8 mmHg during hepatic parenchymal transection. All procedures were assisted by laparoscopic ultrasound. In the RF-assisted LLR, the transection plane was pre-coagulated using radiofrequency energy. Parenchymal transection was performed through this precoagulated plane, moving ahead step by step, and allowing the direction of transection to be altered when necessary. The radiofrequency energy was applied during 1 to 2 minutes every 2 cm using a monopolar RF generator and a single cool-tip electrode (Tyco Healthcare, Radionics Europe NV). Parenchymal transection of the precoagulated liver tissue was accomplished with the harmonic scalpel and the ultrasonic aspirator. Blood vessels measuring less than 4 mm in diameter were controlled with bipolar diathermy. Major portal and hepatic veins were controlled with endoclips or endoscopic stapling devices. The resected specimen was extracted through a suprapubic incision by means of an endobag.

Patient, tumor, and surgery characteristics were comparable in patients undergoing hepatic resection with or without RF-assistance (p ≥ 0.1370), that is, gender, age, number and type of tumors (malignant vs benign), maximum tumor diameter, tumor location in right/left liver, presence/absence

Page 214: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

205

of cirrhosis, preoperative chemotherapy (yes/no), duration of hepatic vascular occlusion, other additional surgical procedures (yes/no), surgeon, and duration of surgery [Table 2]. At the time of surgery, macroscopic invasion of the diaphragm (2) or a hepatic vein (2) was encountered equally in both groups. Cirrhosis was present in 5 patients who underwent LLR (segmental hepatectomy 4, left lateral lobectomy 1) for a hepatocellular carcinoma (HCC) with a median maximum diameter of 37 (20–45) mm. Preoperative chemotherapy was given in 12 out of 19 patients with colorectal liver metastases.

Any hepatic segment involved with a tumor was considered for the laparoscopic approach. The location of the liver tumors is shown in Table 3. Thirty-seven minor hepatectomies (resection of 2 segments or less) were performed, of which 22 were segmental resections (segmentectomy/metastasectomy) and 15 were left lateral lobectomies (LLL). Major hepatectomy (resection of 3 segments or more) consisted of 6 right hemihepatectomies (RHH), 2 left hemihepatectomies (LHH), and 1 central hepatectomy (Sg4-5-8). Eighteen patients underwent additional surgery simultaneously with laparoscopic liver resection [Table 3].

Analysis of the statistical significance of differences between groups of data was performed using two-tailed Fisher exact test or Wilcoxon / Kruskal-Wallis test, as appropriate. A p-value of ≤ 0.05 was considered statistically significant. All analyses were performed with the statistical software package JMP (version 5.1.2).

Results No intra- or postoperative mortality occurred. The

median duration of surgery was 115 (45–360) minutes. The median intra-operative blood loss was 200 ml (range 5–4000 ml) and was similar in patients undergoing hepatic resection with or without RF assistance [Table 4]. The type of surgical

Page 215: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

206

procedure was a determinant for the amount of intra-operative blood loss (p = 0.0002). Laparoscopic right/left hemihepatectomy was associated with a median blood loss of 2000 ml, left lateral lobectomy with 200 ml and segmental hepatectomy with 100 ml [Table 5]. Bleeding was the only intra-operative complication and was observed in 3/25 (12%) of patients in the RF– group and in 7/20 (35%) patients in the RF+ group. In these 10 (22.2%) patients, bleeding occurred from large hepatic vessels and was encountered in 6 out of 9 (66.7%) patients who underwent a major hepatic resection, and in 4 out of 36 (11.1%) patients undergoing minor LLR [Table 6]. No significant (Spearman) correlations were found among patient, tumor, and surgery-associated variables and the amount of intraoperative blood loss. Median intra-operative blood loss in patients with cirrhosis and/or preoperative chemotherapy (RF+ in 8 vs RF– in 9 patients) was 150 (5–3000) ml as compared to 200 (10–4000) ml in patients without cirrhosis and/or preoperative chemotherapy (RF+ in 12 vs RF– in 16 patients). This difference was not statistically significant (p = 0.2175).

Pringle’s maneuver was performed in 6 patients for a median duration of 18 (7–40) minutes, that is, in one LLL, in one LLL combined with a segmental resection in Sg4-5, and in 4 patients undergoing segmental hepatectomy (respectively in Sg4-5, Sg5, Sg5-6 + Sg6-7 and Sg5 + Sg6-7). Segmental hepatectomy for a tumor involving Sg7 or Sg8 was performed in 3 and 2 patients, respectively. In one patient (Sg5+Sg6-7), Pringle’s maneuver was performed to control the hemorrhage. Median blood loss in these patients was 100 (10–1400) ml. Microscopic resection margins [8 (1–10) mm] were free of tumor in all patients.

In patients who underwent a right (n = 6) or left (2) hemihepatectomy, the inflow vessels were controlled selectively prior to parenchymal transection, without a Pringle’s maneuver. The draining hepatic veins were divided with an endoscopic vascular stapling device after completing the

Page 216: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

207

parenchymal transection. The site of significant hemorrhage in patients undergoing a major LLR was from a hepatic vein in 4 patients, diaphragmatic vein in 1, and portal vein in 1. During minor LLR, hemorrhage occurred from a hepatic vein in 3 and from a portal vein in 1 patient.

Conversion from a laparoscopic procedure to an open hepatic resection was necessary in 3 patients because of uncontrollable intraoperative bleeding from the right hepatic vein (at RHH), the middle hepatic vein (at segmental hepatectomy Sg4a) and the right portal vein (at RHH) [Table 6].

Postoperative complications were observed in 2 patients, that is, 1 biliary leakage with intra-abdominal abscess formation and 1 pulmonary infection. The median length of hospital stay (LOS) was 7 days following LLR with and without RF assistance. In all resection specimens, the margins were free of tumor on macroscopic and pathologic examination. The magnitude of the tumor-free margin [median 10 (1–30) mm] was comparable after LLR with or without RF-assistance, less than a centimeter in 16 and greater than a centimeter in 29 patients.

Discussion

In open liver surgery, major blood loss implicates the risk of higher morbidity and mortality as well as shorter long-term survival17,27,32,37. In order to reduce parenchymal bleeding during the actual transection phase in open liver surgery, several measures have been described, including radiofrequency energy to create a ‘‘zone of dessication’’ prior to performing hepatic transection. The potential advantages of radiofrequency-assisted hepatectomy might be a ‘‘virtually bloodless’’ procedure, shorter operation time, and reduced morbidity17,27,32,37. The RF technique cannot be used close to the liver hilum, since heat might traumatize biliary structures with subsequent bile leakage and/or abscess formation27,28,32,37. The RF technique is also unable to control blood vessels

Page 217: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

208

measuring larger than 4 mm in diameter, such as hepatic or portal vein branches24. The major shortcomings of previously published data are that these studies primarily cover small patient series, and often information on the number or size of the liver tumors, as well as the exact type of hepatic resection, is omitted or is not clearly specified17,32,37.

The implementation of radiofrequency-assisted hepatectomy in laparoscopy was assumed possibly to represent a considerable step in making laparoscopic liver resection safer and more feasible30,37,38. However, this hypothesis is supported by few case reports9,38. The present study is the first to report laparoscopic liver resection prospectively in a substantial number of patients over a relatively short period of time, and to evaluate the contribution of radiofrequency energy to the amount of blood loss during minor and major laparoscopic hepatectomy.

In the present study, laparoscopic liver resection indeed appears safe and feasible, without mortality, and with a morbidity rate of 24% (11/45). With a median blood loss of 200 ml, an operative duration of 113 minutes, and a length of hospital stay of 7 days, the short-term results concur with those reported in literature1,2,4,13,15,18,25,29,31,35,38.

Losing control on blood loss is obviously the most important concern for all authors reporting on LLR1,2,4,9,13,15,19,22,25,30,35,38. The only intra-operative complication retained in the present study was bleeding that was observed in 66.7% (6/9) of the major LLR and in 11.1% (4/36) of the minor resections. Significant blood loss occurred from a hepatic vein in 7 patients, from a portal vein branch in 2, and from a diaphragmatic vein in 1 patient. It is clear that this type of bleeding can neither be prevented nor managed by using RF energy. On the other hand, the use of Pringle’s maneuver would only have been beneficial in the 2 cases with hemorrhage from the portal vein, since this maneuver does not control blood loss from the draining hepatic veins. Indeed, this high percentage of significant intraoperative bleeding indicates

Page 218: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

209

that laparoscopic major hepatectomy needs thorough evaluation before it can be implemented into daily practice.

The application of RF energy during parenchymal transection in LLR was not associated with reduced intra-operative blood loss, operation time, or complication rate. Only the type of surgical procedure appeared to determine significantly the amount of blood loss. No other significant correlations were found between the analyzed patient, tumor- and surgery-associated variables, and the amount of intraoperative blood loss. Intra-operative blood loss in patients with a ‘‘normal liver’’ was comparable to blood loss in patients with cirrhosis or with preoperative chemotherapy.

Patient selection, experience in hepatobiliary surgery, adequate technology, and infrastructure are vital in defining the safety, feasibility, and outcome of laparoscopic liver resection. In the present study, patient selection and the use of RF energy were based on the surgeon’s judgment in a nonrandomized fashion, suggesting a potential bias. However, it is a difficult task to randomize a large number of patients with similar characteristics, who are suffering from liver tumors, to undergo comparable surgical procedures. All LLRs were performed by surgeons with experience in both hepatobiliary and advanced laparoscopic surgery as well as in hepatic radiofrequency application. Although patients with severe systemic disease were excluded, any patient can be eligible for LLR as long as they can endure an advanced laparoscopic procedure. Anterior and left lateral hepatic segments and tumors smaller than 5 cm seem to be most suitable for LLR1,2,4,13,15,18,25,29,35,38. However, tumor size and localization don’t have to be a contraindication for LLR as such, provided the tumor isn’t in close relationship to the portal bifurcation or the suprahepatic junction and provided a sufficient tumor-free margin can be warranted. In the present study, any hepatic segment involved by tumor was considered for laparoscopic approach. Benign lesions were judged to be an indication for LLR when they were symptomatic, a suspected

Page 219: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

210

adenoma, or of uncertain histopathologic diagnosis. Malignant lesions eligible for LLR were HCC in cirrhosis and liver metastasis, mostly colorectal in origin. One gallbladder cancer in situ and 4 intrahepatic cholangiocarcinomas were resected. Five patients (11%) suffered from cirrhosis. Several reports on LLR suggest that this population could benefit from minimally invasive surgery, as it seems to mean significant reduction in perioperative blood loss and surgical trauma as compared to open surgery1,4,7,11,15,21,26,35. In order to prevent cancer recurrence caused by technical failure, the same oncologic rules should be respected for LLR as for open hepatectomy1,2,4,11,15,18,21,25,29,34–36. In the present study, the resection margins were tumor-free on macroscopic and pathologic examination in all resected specimens. The LLR was accomplished according to the ‘‘no touch’’ technique, without hand-assistance. The resected liver tissue was extracted using a specimen bag, avoiding direct contact of the tumor with the abdominal wall.

As far as postoperative outcome is concerned, the complications observed in the present study are comparable with those most frequently reported in literature4,10,15,20,25,29,35. The same remark can be made for the conversion rate, as the reported conversion rates vary between 0 and 15.4%1,2

5,7,9,15,21,25,29. The authors believe that conversion of the LLR to an open procedure must be acknowledged as a safety measure and not as a technical failure. Major intra-operative bleeding during LLR will always remain difficult to control laparoscopically, even with extensive surgical experience. Therefore, a low threshold for conversion to open hepatic resection is indicated.

Conclusions Laparoscopic liver resection is feasible and safe,

provided that adequate equipment and extensive experience of the surgical team in both open liver surgery and advanced

Page 220: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

211

laparoscopy are available. LLR can be performed with minimal amounts of intraoperative blood loss, which is determined by the type of hepatectomy. Significant intraoperative hemorrhage occurs from large hepatic vessels during major LLR. The high percentage of significant bleeding that is observed at major hepatectomy indicates that the role of laparoscopy in major liver resections needs further evaluation. Radiofrequency-assisted parenchymal transection in LLR does not seem to reduce blood loss, operation time, or peri-operative morbidity. Therefore, RF-assisted LLR seems not to be advocated as the preferred surgical technique in laparoscopic hepatectomy, also taking into account the additional costs of the RF equipment. The present, prospective study, however, only analyzes the short-term outcome and has a nonrandomized nature. The impact of laparoscopic surgery on the long-term outcome of patients with hepatic tumors has yet to be evaluated.

Page 221: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

212

Table 1. Pathology of hepatic tumors

Number of patients ----------------------------------------------- Pathology Number of lesions RF- RF+

Benign Adenoma 2 1 1 Hemangioma 7 3 4 Focal nodular hyperplasia 1 1 0 Biliary cysta 1 0 1

Malignant CRLM 32 10 9 Hepatocellular carcinoma 8 6 2 Intrahepatic cholangiocarcinoma 4 2 2 Gallbladder CIS 1 0 1 Metastasis of ET 1 1 0

Metastasis of RCC 1 1 0

CRLM, colorectal liver metastases; CIS, carcinoma in situ; ET, endocrine tumor; RCC, renal cell carcinoma; RF, radiofrequency aBiliary cyst was preoperatively interpreted as a tumor mass with an uncertain histopathologic diagnosis

Page 222: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

213

Table 2. Patient and tumor characteristics

RF- RF+ p

N patients 25 20 Male/female 13/12 9/11 Age (years) 57 (28–80) 55 (26–74)

N lesions 32 26 Benign/malignant 5/27 6/20 NS Location, right/left 16/16 9/19 Median diameter (mm) 36 (8–120) 51 (12–170) Invasion: diaphragm 1 1 Hepatic vein 1 1 Cirrhosis 4 1

N, number; RF, radiofrequency; NS, not significant

Page 223: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

214

Table 3. Type of surgical procedure

RF- RF+

Segmental hepatectomy 15 8 Segment involved by tumor: Sg2 5 – Sg3 – 1 Sg4 3 4 Sg5 5 5 Sg6 4 2 Sg7 2 1 Sg8 – 1 Left lateral lobectomy (LLL) 6 9 Left hemihepatectomy (LHH) 1 1 Right hemihepatectomy (RHH) 3 3 Additional surgery 12 6 Type LCCE 4x LND and CCE

Lap. CPAA Lap. CPAA and ileostomy Right HC (M.Crohn) LCCE 2x Adhesiolysis Left iliacal LND Adhesiolysis & LCCE Lap. anteriorresection + lap.RFA CRLM Sg5 Drainage hemoperitoneum + vesiculae seminales Lap.anteriorresection resection + bilateral ureteric stenting RFA 3 HCCs Closure loop-ileostomy

CRLM, colorectal liver metastasis; (L)CCE, (laparoscopic) cholecystectomy; lap. CPAA laparoscopic colonic pouch anal anastomosis; right HC, right hemicolectomy; LND, lymph node dissection; RF, radiofrequency; RFA, radiofrequency ablation; Sg, segment

Page 224: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

215

Table 4. Clinical outcome of laparoscopic liver resection in 45 patients

RF- RF+ Total group

Procedure Blood loss (ml) 200 (20–4000) 200 (5–3000) 200 (5–4000) OP time (min) 105 (45–360) 120 (50–310) 115 (45–360) Pringle (N) // (min) 2 // 25 (20–30) 4 // 15 (7–45) 6 // 17.5 (7–45)

Complications * Intraoperative - bleeding 3 6 9 - other 0 0 0 * Postoperative 0 2 2 * Mortality 0 0 0 LOS (days) 7 (3–18) 7 (5–41) 7 (3–41) Tumor free margins (mm) 10 (1–30) 10 (1–20) 10 (1–30)

OP, operation; N, number of patients; LOS, length of hospital stay

Page 225: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

216

Table 5. Type of surgical procedure and intraoperative blood loss Median blood loss (ml) (range) -------------------------------------------------------------------------------- Type surgical procedure N of patients Total RF+ RF- p value

a

Segmental hepatectomy 23 100 (5–2600) 25 (5–2600) 100 (20–700) 0.456 Right/left hemihepatectomy 8 2000 (600–4000) 2000 (1500–3000) 1650 (600–4000) 0.886 Left lateral lobectomy 15 200 (10–1200) 200 (10–1200) 200 (20–850) 0.633

RF, radiofrequency a Statistically significant difference in blood loss following different types of surgical procedure (p value = 0.0002)

Page 226: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

217

Table 6. Analysis of intraoperative hemorrhage Surgical Procedure Site of bleeding Blood loss (ml) Conversion to laparotomy

RF- RHH Middle HV 4000 0 RHH Right PV 2500 1 LLL Left HV 850 0

RF+ RHH Branch right HV 3000 1 RHH Right HV 1500 0 RHH Right HV 2300 0 LHH Left diaphragm vein 1700 0 Resection Sg4a Middle HV 2600 1 Resection Sg5 + Sg6-7 Branch HV 1400 0 LLL Portal pedicle Sg2-3 1200 0

RHH, right hemihepatectomy; LHH, left hemihepatectomy; LLL, left lateral lobectomy; RF, radiofrequency; HV, hepatic vein; PV, portal vein

Page 227: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

218

References 1. Belli G, Fantini C, D’Agostino A. Laparoscopic liver

resections for hepatocellular carcinoma (HCC) in cirrhotic patients. HPB 2004; 6: 236–246.

2. Biertho L, Waage A, Gagner M. Hépatectomies sous laparopscopie. Ann Surg 2002; 127: 164–170.

3. Buell J, Koffron A, Thomas M. Laparoscopic liver resection. J Am Col Surg 2005; 200: 472–480.

4. Cherqui D, Husson E, Hammoud R. Laparoscopic liver resections: a feasibility study in 30 patients. Ann Surg 2000; 232: 753–762.

5. Cherqui D. Résections hépatiques par voie coelioscopic: utiles ou futiles? Ann Chir 2002; 127: 171–174.

6. Cherqui D, Yachouchi-Chouillard E. Lobectomy gauche par abord coelioscopic. J Chir (Paris) 2002; 139: 25–28.

7. Cherqui D. Laparoscopic liver resection. Br J Surg 2003; 90: 644–646.

8. Consten E, Gagner M. Perioperative outcome of laparoscopic left lateral liver resection is improved by using staple line reinforcement technique: a case report. J Gastrointest Surg 2005; 9: 360–364.

9. Croce E, Olmi S, Bertolini A. Laparoscopic liver resection with radiofrequency. Hepatogastroenterology 2003; 50: 2088–2092.

10. Descottes B, Glineur D, Gigot J. Laparoscopic liver resection of benign liver tumors. Surg Endosc 2003; 17: 23–30.

11. Dulucq J, Wintringer P, Stabilini C. Laparoscopic liver resections: a single center experience. Surg Endosc 2005; 19: 886–891.

12. Elias D, Cavalcanti A, Sabourin J. Results of 136 curative hepatectomies with a safety margin of less than 10 mm for colorectal liver metastases. J Surg Oncol 1998; 69: 88–93.

13. Fujita F, Fujita R, Kanematsu T. New approaches to the minimally invasive treatment of liver cancer. Cancer J 2005; 11: 52–56.

Page 228: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

219

14. Gagner M. Pioneers in laparoscopic solid organ surgery. Surg Endosc 2003; 17: 1853–1854.

15. Gagner M, Rogula T, Selzer D. Laparoscopic liver resection: benefits and controversies. Surg Clin North Am 2004; 84: 451–462.

16. Gigot J, Glineur D, Azagra J. Laparoscopic liver resection for malignant liver tumors: preliminary results of a multicenter European study. Ann Surg 2002; 236: 90–97.

17. Haghighi K, Wang F, King J. In-line radiofrequency ablation to minimize blood loss in hepatic parenchymal transection. Am J Surg 2005; 190: 43–47.

18. Hansen P. Laparoscopic resections of liver and pancreas. J Gastrointest Surg 2004; 8: 925–926.

19. Kalil A, Mastalir E. Laparoscopic hepatectomy for benign liver tumors. Hepatogastroenterology 2002; 49: 803–805.

20. Kaneko H, Takagi S, Otsuka Y. Laparoscopic liver resection of hepatocellular carcinoma. Am J Surg 2005; 189: 190–194.

21. Laurent A, Cherqui D, Lesurtel M. Laparoscopic liver resection for subcapsular hepatocellular carcinoma complicating chronic liver disease. Arch Surg 2003; 138: 763–769.

22. Lesurtel M, Cherqui D, Laurent A. Laparoscopic versus open left lateral lobectomy: a case-control study. J Am Coll Surg 2003; 196: 236–242.

23. Linden B, Humar A, Sielaff T. Laparoscopic stapled left lateral segment liver resection – technique and results. J Gastrointest Surg 2003; 7: 777–782.

24. Machi J. New technique for liver resection using heat coagulative necrosis – letter to the editor. Ann Surg 2004; 241: 194–196.

25. Mala T, Edwin B, Gladhaug I. A comparative study of the short-term outcome following open and laparoscopic liver resection of colorectal metastases. Surg Endosc 2002; 16: 1059–1063.

Page 229: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

220

26. Morino M, Morra I, Rosso E. Laparoscopic vs open hepatic resection: a comparative study. Surg Endosc 2003; 17: 1914–1918.

27. Navarra G, Spalding D, Zacharoulis D. Bloodless hepatectomy technique. HPB Surg 2002; 4: 95–97.

28. Navarra G, Lorenzini C, Currò G. Early results after radiofrequency-assisted liver resection. Tumori 2004; 90: 32–35.

29. O’Rourke N, Fielding G. Laparoscopic right hepatectomy: surgical technique. J Gastrointest Surg 2004; 8: 213–216.

30. O’Rourke N, Shaw I, Nathanson L. Laparoscopic resection of hepatic colorectal metastases. HPB 2004; 6: 230–235.

31. Shimada M, Harimoto N, Maehara S. Minimally invasive hepatectomy: modulation of systemic reactions to operation or laparoscopic approach? Surgery 2002; 131(Suppl 1): S312–S317.

32. Stella M, Percivale A, Pasqualini M. Radiofrequency-assisted liver resection. J Gastrointest Surg 2003; 7: 797–801.

33. Tepel J, Klomp H, Habib N. Modification of the liver resection technique with radiofrequency coagulation. Chirurg 2004; 75: 53–55.

34. Topal B, Kaufman L, Aerts R. Patterns of failure following curative resection of colorectal liver metastases. Eur J Surg Oncol 2003; 29: 248–253.

35. Vibert E, Kouider A, Gayet B. Laparoscopic anatomic liver resection. HPB 2004; 6: 222–229.

36. Vittimberga F, Foley D, Meyers W. Laparoscopic surgery and the systemic immune response. Ann Surg 1998; 227: 326–334.

37. Weber JC, Navarra G, Jiao LR. New technique for liver resection using heat coagulative necrosis. Ann Surg 2002; 236: 560–563.

38. Weber JC, Navarra G, Habib NA. Laparoscopic radiofrequency-assisted liver resection. Surg Endosc 2003; 17: 834.

Page 230: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

221

V

Discussion

Page 231: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

222

Page 232: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

223

Worldwide, colorectal cancer (CRC) is the fourth most common cause of cancer death, with more than 1.200.000 new diagnoses every year and resulting in 600.000 deaths/year. In Europe alone, CRC is responsible for 200.000 deaths/year. Long-term survival is dependent on the stage of the disease at diagnosis, ranging from an average 5-year overall survival (OS) of approximately 90% for stage I disease to 5% or less for stage IV (metastatic) CRC. About 20-30% of CRC patients present with stage IV disease at diagnosis and about 50-60% of CRC patients will eventually develop metastases at some point in the course of their disease. So, obviously this involves a large number of CRC patients. In general, about 50% of CRC patients will develop colorectal liver metastases (CRLM). Peritoneal carcinomatosis (PC) is reported to occur in about 12-13% of patients with CRC and PC often occurs in combination with other metastatic sites.

Unresectable PC from CRC and neo-adjuvant

chemotherapy Resectable PC from CRC, without systemic metastases, is considered a locoregional phenomenon, which can be treated with complete cytoreductive surgery (CCRS) + hyperthermic intra-peritoneal chemotherapy (HIPEC), resulting in 5-year OS rates of up to 50%1-9. But the majority of patients with PC present with unresectable disease. Literature on the outcome of unresectable PC from CRC is scarce and reports a very poor prognosis10-15 [Table 116]. Two prospective studies focused on the prognostic factors in PC of non-gynecologic malignancy10,11: the EVOCAPE 1 trial by Sadeghi et al. included only patients undergoing palliative treatment for PC of CRC. A median OS of 5.2 months [range 0.6-44.8 months] was reported and patients with PC of digestive origin were found to have a very poor prognosis10. Only the extent of PC was significantly correlated to survival10, whereas in the study of Chu et al. tumor burden did not affect survival11. In a retrospective analysis by Jayne et al. 349 (13%) out of 3019 patients had PC, of whom 61% had synchronous PC and 39% developed

Page 233: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

224

metachronous PC17. A resection of the primary tumor and the PC could be performed in the synchronous PC group in 72%, whereas debulking of the PC was only possible in 2% of patients with metachronous PC. Furthermore, the extent of PC and T-stage were identified as predictors of survival for synchronous PC17. Our retrospective study found a median OS of 6.3 months [0.4-33.1 months] for 43 patients with unresectable PC from CRC15. Univariate analysis could only find palliative systemic chemotherapy to have a statistically significant influence on (p<0.001): a median OS of 9.3 months [range 0.9-33.1 months] [Table 315] was registered under palliative chemotherapy versus 3.1 months [range 0.4-6.5 months] without palliative chemotherapy. This concurs with current literature2,10-13. However, a negative Pearson correlation was found between the administration of palliative chemotherapy on the one hand and ASA score ≥ 2 as well as the occurrence of postoperative complications after surgical exploration on the other hand. This suggests that a better general condition or performance status (PS) facilitates patients to undergo and tolerate chemotherapy and it’s side effects [Table 415]. Literature data support this theory. A retrospective analysis of 3823 patients with advanced CRC by Folprecht et al. already stressed the importance of PS18 and a systematic review by Stillwell et al. showed that a patient’s PS, age and ASA-score, as well as the postoperative morbidity and mortality after palliative surgery are important prognostic factors related to OS in patients with stage IV CRC and unresectable metastases19. Pelz et al. stated that the biologically aggressive nature of PC impairs the functional status of patients to an extent that makes them only eligible for palliative care20. A review by Köhne et al. and the analysis by Folprecht et al. identified PC in itself as an independent negative predictive factor for outcome18,21. In our analysis lymph node involvement was not identified as a predictive factor adversely affecting survival15. This concurs with the results of EVOCAPE 110. Possibly, this indicates that the local phenomenon of PC might have a

Page 234: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

225

stronger adverse influence on outcome than the systemic spread of advanced CRC by nodal involvement.

Currently, systemic chemotherapy is considered the only available treatment option for patients with unresectable PC from CRC. For some time now, neo-adjuvant chemotherapy is accepted as a down-sizing technique for initially unresectable systemic metastases from CRC, such as colorectal liver metastases (CRLM)22-29, with response rates as high as 50%24. For CRLM conversion rates of 10-20% have been described23,24,26-28, allowing resection and resulting 5-year OS rates of 30-40%23. It is very difficult to determine whether this finding for metastatic CRC in general also applies to the local phenomenon of PC. In order to compare the results of palliative chemotherapy (+/- palliative surgery) with the outcome after CCRS+HIPEC Elias et al. performed a multicenter retrospective study on 2 highly selected patient groups with a comparable, limited (resectable) extent of PC from CRC. Median survival was 23.9 months in the systemic chemotherapy-group versus 62.7 months in the HIPEC-group30. But literature results on systemic chemotherapy for PC are far less optimistic when patients with resectable and unresectable PC are combined in one trial. For this patient population the randomized controlled trial by Verwaal et al. reports a disease-specific survival of 12.6 months31. This concurs with the median survival of 5.2-12.6 months after 5-FU/LV-based chemotherapy by as reported by Koppe et al.32. Results are even worse for unresectable PC15,20. Even with modern systemic therapy regimens a survival of almost 2 years is only rarely reached15. But “unresectable” PC implies more extensive disease15, whereas the best results with chemotherapy are reached in patients with low tumor load and in patients with PC only (i.e. without systemic metastases)20. Another important obstacle to interpret the available data is the inability to image sub-centimetric peritoneal lesions and assess tumor response on the RECIST-criteria. This makes accurate documentation or monitoring of treatment response impossible20. Therefore, retrospective evaluation of the extent of

Page 235: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

226

PC is very difficult. At present, laparoscopy is the only tool available that to reliably assess the extent of PC. In an attempt to formulate an answer to the difficulties summarized above, we performed the first prospective observational pilot study to assess and document the actual response rate and response characteristics of unresectable PC from CRC under neo-adjuvant systemic chemotherapy with capecitabine and oxaliplatin with or without bevacizumab16. A laparoscopy was performed to assess and document the extent of PC before and after this combined chemotherapy. All 10 patients that were initially included into the trial, had a good PS and ASA-score at PC diagnosis. One patient had to be excluded because of systemic metastases. Of the 9 patients that proceeded within the trial protocol, 1 developed early progressive disease, 2 had macroscopically stable disease, 5 had progressive disease at laparoscopy and 1 patient had proven progressive disease at laparotomy after palliative chemotherapy outside of protocol. In other words, 78% of patients had progressive disease under chemotherapy and 22% had stable disease16. Thus, no clear macroscopic response to chemotherapy could be demonstrated. For this reason trial inclusion was discontinued. Of course, an important shortcoming of this trial is the small number of patients included. Furthermore, despite their good PS at inclusion, the majority of patients completed their systemic chemotherapy with dose reduction. This indicates that PC indeed has a substantial influence on the patient’s functional status. Finally, for safety reasons, 2 important concessions were done on the trial protocol: First, due to tumor load and adhesions, not all 7 abdominal regions were always fully accessible for macroscopic assessment and documentation. Nevertheless, PC unresectability criteria were met in all cases. Second, peritoneal biopsies could not always be safely taken, especially at second laparoscopy. But all patients had histopathological proof of PC from CRC at trial inclusion16.

Page 236: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

227

To evaluate the prize in complication rate that comes with the administration of neo-adjuvant chemotherapy, we performed a review of literature to assess the incidence and clinical significance of Bevacizumab(BV)-related non-surgical and surgical serious adverse events (SAE)33. The BV-related SAE reported in literature are mild to moderate in severity and manageable using standard therapies. Furthermore, the incidence of SAE under BV-based chemotherapy is low: even very large, multicenter prospective and randomized controlled trials reported only very small numbers of SAE [Table 233]. In most trials the incidence of hemorrhage (2.0-3.0%), gastro-intestinal (GI) perforation (<1.0-2.0%) and arterial thrombo-embolic events (1.0-2.0%) was higher under BV-based treatment, but overall the absolute number of patients affected remained very low. Also, BV-related grade 5 toxicity was rare. Thus, the addition of BV to chemotherapy causes no clinically relevant aggravation of chemotherapy-related SAE34-43. Leaving the primary tumor in situ during BV-treatment appears safe: BV-related GI perforations occur in about 3% of patients, but virtually all of these are observed in the first 3 months of treatment and occur throughout the entire GI tract, hardly ever involving the primary tumor site37,40,42-44. Finally, the risk emergency surgery might be needed due to BV-related SAE, such as bleeding or perforation, is very low (estimated 2.0%)37,38,44,45. The relation between postoperative SAE and BV-based chemotherapy can be divided in 3 groups:

First, for surgery after BV discontinuation, trials showed very low rates of SAE if a TTS of 5-6 weeks is respected [Table 433 and Table 533]. The majority of SAE reported are wound healing complications (WHC). Bleeding and GI perforation occur infrequently41-43,46-48. In most of these trials 30-40% of patients undergo major surgery, which suggests the low rate of SAE reported should be reliable41-43,46,48-52. For minor surgery, WHC are the main issue, but rates are low if BV is discontinued within a 10-day peri-operative period53.

Second, major or emergency surgery during BV-treatment seemed to represent a higher risk (SAE 1.3-2.7% vs. 0.0%), but

Page 237: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

228

this was rated statistically insignificant49. The SAE incidence remained relatively small in this setting, but they could still be clinically relevant. Thus, close monitoring of patients undergoing surgery under BV is advisable.

Third, for the start of BV-treatment after major surgery few data are available. Small rates of SAE were found, especially if a period of minimally 28 days after surgery is respected before starting BV49. However, in case of comorbidity or wound-healing issues after surgery, the time point for starting BV should be individualized to allow more time for wound healing.

Cytoreductive surgery and HIPEC for PC from CRC Many of the larger series in literature report on overall

safety and efficacy of CCRS + HIPEC, including both patients who underwent HIPEC with Oxaliplatin as well as patients who received HIPEC with Mitomycin C (MMC) in one and the same publication4,5,8,54. This might complicate the interpretation and comparison of results. The number of papers reporting only on CCRS followed by HIPEC with Oxaliplatin is limited and quite often include relatively small numbers of patients7,30,55. Therefore, we performed a prospective observational Belgian multicenter trial to assess the safety and efficacy of HIPEC with Oxaliplatin56. Complete cytoreduction (R0/1 or CC-0) was achieved in all included patients. There was no 30-day mortality. The overall complication rate was 52.1%, with anastomotic leakage in 10.4% of patients, bleeding in 6.3% and prolonged ileus in 22.9%. Surgical reintervention was needed in 20.8% of cases. The median hospital stay was 20 days [range 5-65 days]56. These results concur with literature. Elias et al. reported grade 3-5 SAE in up to 50% of patients who underwent CCRS, resulting in CC-0 resection, followed by HIPEC with Oxaliplatin7,55. Larger series on CCRS + HIPEC with Oxaliplatin and/or MMC reported mortality rates of <1-6%, grade 3-5 SAE rates of 31-66% and reintervention in 11-30% of cases4,5,8,57. The anastomotic leakage rate is often not clearly specified. Elias et al. reported a gastrointestinal tract complications/fistula rate of 9-10%4,5,55. Obviously, these high morbidity rates after CCRS+HIPEC imply

Page 238: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

229

that patients undergoing such extensive surgery should be well-selected regarding general PS, as well as extent of disease. Before the combination treatment of CCRS + HIPEC became available, no long-term survivors were reported for PC of CRC. The OS found in this multicenter Belgian registry was 97.9% at 1 year and 88.7% at 2 years, at a median follow-up of 22.7 months [range 3.2-55.7months] [Fig.1A56]. Table 156 gives an overview of the available literature on survival data reached with CCRS + HIPEC56. If an R0/1 resection can be reached by CCRS, the overall survival rates for CCRS + HIPEC are comparable to the 5-year overall survival rates of 35-60% reported by several large single- and multi-institutional experiences after curative liver resection for colorectal liver metastases58-63. This could be considered as level 1c evidence for this treatment strategy. In the largest HIPEC series reported by Elias et al. positive independent prognostic factors identified by multivariable analysis were complete debulking, a low peritoneal carcinomatosis index (PCI), no invaded lymph nodes and the use of adjuvant chemotherapy5. The Belgian registry found a significant difference in DFS and OS when PCI is dichotomized between patients with PCI≤15 and patients with PCI>15 [Fig.2A56, 2B56]. This concurs with earlier papers published by Elias and Glehen et al.54,64. Furthermore, resection small bowel (p=0.004), post-operative extra-abdominal complications (p=0.0005) and a lesion size>5cm (p=0.043) were found to be significantly correlated with OS56. Due to the low number of deceased patients and the low number of patients for some risk factors, results for OS need to be interpreted with care. For DFS, there is a significant correlation with transfusion (p=0.018) and a trend was observed for the resection of small bowel at debulking56.

At present, both Oxaliplatin and MMC are used as intraperitoneal chemotherapy agents in HIPEC for PC of CRC. Oxaliplatin has become standard systemic treatment in CRC65-68, whereas MMC is only used as salvage treatment in advanced progressive CRC69-72. The conference on Peritoneal Surface Malignancies in Milan in 2006 failed in it’s attempt to reach a

Page 239: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

230

methodological consensus on the intraperitoneal drugs to be used for HIPEC8, which illustrates the complexity of this topic. Oxaliplatin and MMC are both alkylating chemotherapeutic agents, interfering with DNA and DNA-synthesis without being cell cycle dependant73-75. Because Oxaliplatin and MMC have a large molecular weight (397.3 Dalton and 334.3 Dalton, respectively), they can reach high intraperitoneal drug concentrations during HIPEC, with limited systemic absorption74,76. Furthermore, they have enhanced cytotoxicity under hyperthermia and a maximal tissue penetration depth of 2mm76. These characteristics make both Oxaliplatin and MMC suitable as intraperitoneal agents for HIPEC. The recommended intraperitoneal dose for Oxaliplatin is 460mg/m² and perfusion time is limited to 30 minutes, whereas the advised dose for MMC is 35mg/m² with a perfusion duration of 90 minutes55,76-78. The intraperitoneal half-life for Oxaliplatin is 29.5 minutes and 49 minutes for MMC [Table 479]. In order to potentiate the Oxaliplatin activity, patients in the Oxaliplatin-group receive intravenous 5-Fluorouracil and Folinic Acid approximately 1 hour before starting the HIPEC procedure, to bathe the tumor and healthy tissue before HIPEC80. The 5- fluorouracil is administered intravenously, because it cannot be mixed with Oxaliplatin inside the peritoneal cavity (because of pH incompatibility)81. In contrast to MMC, Oxaliplatin it is not stable in chloride-containing solutions: it can only be administered in a 5% dextrose perfusion solution82. This results in biochemical disturbances, which are manageable with a compensation of the sodium loss into the perfusate and a good control of hyperglycemia82,83. Oxaliplatin is easily detectable and therefore thouroughly studied for its distribution55,73. Besides an excellent regional exposure, high drug concentrations were found in colonic tissues73. Elias et al. reported a high uptake of Oxaliplatin in local tissues (tumoral tissue and peritoneum) after HIPEC55. So far, there are no trials are available in literature, which perform a straight-forward comparison of toxicity and survival data after CCRS+HIPEC for PC of CRC with Oxaliplatin versus MMC as intraperitoneal chemotherapy-agents. Furthermore,

Page 240: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

231

there is a wide variation in toxicity as well as survival data [Table 579 and Table 179] after CCRS+HIPEC for patients with PC from CRC due to multiple variations in factors such as HIPEC indication, patient selection, extent of disease, completeness of cytoreduction2-9,30,31, administration of neo-adjuvant systemic chemotherapy3-5,30 and presence of systemic metastases3-5. Therefore, we performed a retrospective analysis of prospectively gathered data on 2 patient cohorts from 2 different HIPEC centers, who underwent CCRS+HIPEC with Oxaliplatin (39 patients) and MMC (56 patients), respectively79. The extent of PC (assessed using the Dutch 7-region count85) was found to be the only tumor-related parameter that is significantly different between the Oxaliplatin- and MMC-group (p<0.004)79. Therefore, it was taken into account in the statistical analysis, to enable adequate comparison between the HIPEC results in both patient cohorts. A 3-year OS of 54.0% was observed for Oxaliplatin-patients and 3- and 5-year OS for MMC-patients was 41.1% and 25.6%. Median OS was 37.1 months [IQR: 22.4-52.8] for Oxaliplatin-patients and 26.5 months [IQR: 16.9-64.8] for MMC-patients (p=0.45)79. The survival analysis (corrected for extent of disease) showed curves that appear slightly in favour of the Oxaliplatin-group, but statistical analysis showed no difference in OS (HR=1.37 (95%CI: 0.74-2.54), p=0.32) [Fig.179] and RFS (HR=1.24 (95%CI: 0.75-2.05), p=0.39) [Fig.279] for the Oxaliplatin- and MMC-group79. Univariate analysis shows no statistical differences across the Oxaliplatin- and MMC-groups for intra-abdominal complications (IAC) (p=0.64), extra-abdominal complications (EAC) (p=0.15) or the need for surgical reintervention (p=0.51)79 [Table 379]. But, after correction of the toxicity data for extent of disease with a logistic regression model, there is a higher overall complication rate in the MMC-group (OR=2.68 (95%CI: 1.04-6.91), p=0.04), with a tendency towards more EAC (OR=2.23 (95%CI: 0.91-5.43), p=0.079). The IAC rate remains comparable across groups (OR=0.78 (95%CI: 0.30-2.03), p=0.61)79. Literature data on morbidity and mortality were already mentioned above. Overall,

Page 241: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

232

the occurrence of fistulae after CCRS+HIPEC is reported to be 17.6% and 10% for MMC and Oxaliplatin, respectively55,75,78,86. MMC is known to cause severe neutropenia in 28% of patients55,75,78,86. In this paper 26.8% of MMC-patients developed to have neutropenia or leucopenia. So despite the very limited systemic absorption of intraperitoneal MMC, there must be a systemic effect, which is probably due to systemic accumulation. This accumulation results not only from systemic absorption, but also from the hepatic metabolization of MMC75. For Oxaliplatin the predominant route for the elimination is by urinary excretion87. When administered systemically, Oxaliplatin is known to cause severe neurotoxicity88,89, but no neurotoxicity was found in this study79. Finally, when the recommended protective garment is used and safety considerations are followed MMC90,91 and Oxaliplatin92 do not represent a detectable safety hazard to the surgeon or other operating room personnel.

Shepherd et al. have shown that transserosal invasion of colorectal tumors (T4) was shown to increase the risk for PC and/or local recurrence93-96. It would be interesting to assess the burden of PC in T4 tumors and the risk for local intraperitoneal recurrence as the only site of metastatic disease. This is crucial to adequately estimate the potential benefits of HIPEC as an adjuvant treatment strategy in T4 patients. Literature on this issue is scarce. Therefore, we retrospectively analyzed prospectively collected data on 379 consecutive patients who underwent elective or emergency surgery for colon cancer between January 2004 and January 2007 at the department of abdominal Surgery of the University Hospitals Leuven97. The relapse analysis was restricted to patients with stage II-III colon cancer for whom the data on relapse were completely available. Out of 215 patients with stage II-III CRC 26 patients (12.1%) developed peritoneal metastases [Table 297], which is in agreement with published data17,18. Furthermore, a clear and significant (p=0.008) correlation was shown between transserosal invasion of colonic

Page 242: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

233

cancer (T4) and peritoneal recurrence [Fig.597]. The majority of these T4 tumors had a T4a staging [Table 397]. In 5 of 8 T4 tumors that developed PC (proportion 0.625, CI [0.245-0.915]), PC was the only metastatic site, without synchronous systemic metastases. This could implicate that 5 out of the 19 T4 patients might eventually benefit from adjuvant HIPEC in order to prevent PC. This is 1.3% (5/379) of the total patient population presenting with colon cancer97. Based on our own retrospective analysis a sample size calculation showed that a randomized controlled trial to demonstrate the benefit of adjuvant HIPEC in stage II and III T4 colon cancers, defined as a 50% risk reduction for the development of PC without systemic metastases, with a power of 80%, would require a sample size of 352 patients. This is an impossible task for a single institute. Even with a multicentric approach inclusion of 176 stage II-III T4 colonic tumors in each trial arm would be an ambitious undertaking97. Moreover, there is no adequate diagnostic tool available at present to intra-operatively identify tumors with transserosal invasion. Noura et al. performed a large prospective trial on peritoneal lavage to identify intraperitoneal tumor cells (IPTC), which showed that a very large patient cohort is needed to identify a very small number of patients (2.2.%) with positive cytology98. There are more sensitive IPTC detection methods, such as immunocytochemistry99 or RT-PCR100,101 (detection rates of 12-47%), but despite this higher sensitivity, the significance of IPTC for survival remains unclear. Additionally, the ability to estimate the amount of free IPTC might have a higher clinical value than the mere detection of their presence98. To our knowledge, only one study, performed by Noura et al., explored the effects of intraperitoneal chemotherapy (IPC) with MMC on the prevention of peritoneal recurrence in CRC in patients with positive peritoneal lavage cytology102. IPC appeared effective in preventing peritoneal recurrence and prolonging cancer-specific survival, with recurrence rates 12.5% in the IPC(+) group versus 50% in the IPC(-) group. This difference was rated significant, but only a small number of patients was included into

Page 243: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

234

the trail over a long time period and the total number of recurrences was low102. Two important obstacles for “prophylactic“ HIPEC should be mentioned: First, HIPEC comes with the price of very high morbidity4,55,57,63,74,86,103-105. Second, in the first 24-72h after surgery fibrin tissue will inevitably be formed in the resection planes and adhesions will start to develop, thus entrapping any free IPTC that might be present in the abdomen and making them unattainable for HIPEC. Therefore, pathologic diagnosis of transserosal invasion should be made within 24h after resection. An alternative to neo-adjuvant HIPEC for T4 tumors could be the performance of a second look laparotomy plus HIPEC 1 year after resection of the primary tumor for asymptomatic patients at high risk of developing colorectal PC as proposed by Elias e al. The criteria for high-risk were defined based as: synchronous macroscopic PC, synchronous ovarian metastases and perforation63,106,107.

Radiofrequency ablation (RFA) for CRLM

We performed a review of literature to summarize the available data on the application of RFA as a treatment tool for CRLM108. The efficacy and reliability of RFA depends mainly on the diameter of the targeted lesions, the applied approach (surgical or percutaneous) and the distance between the targeted lesion and large vessels (because of the heat sink phenomenon). Obviously, larger CRLM require overlapping treatment zones, i.e. multiple RFA sessions, which makes the result less reliable109. RFA for lesions greater than 5 cm in diameter is even questionable109,110. Furthermore, the experience of the physician performing RFA appears to be an important factor109,111, the use of CT or MRI guidance improves the accuracy of the treatment110 and newer generation probes seem to result in lower local recurrence111. Therefore, in some highly specialized centers, a percutaneous approach in well-selected patients seems to produce equivalent results to liver resection109.

Page 244: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

235

For unresectable CRLM, current literature data strongly suggest a benefit of any combined modality treatment with RFA over a chemotherapy-only approach112,113. At present, there is a lack of evidence for the use of RFA as an alternative to surgery in patients with resectable CRLM. The available data on RFA for small solitary lesions are contradictory [Table 2108]108,114. Nevertheless, a shift in patient selection from unresectable to resectable CRLM, implying more favorable tumor biology, better localization etc, might result in a different (better) outcome after RFA for small solitary lesions,. Furthermore, this could avoid extended liver resections for small, centrally located lesions111,115. Thus, a far larger liver remnant is left in place, which leaves more room for retreatment of future local recurrences or newly developed CRLM at other sites. Because most of the currently available papers compare RFA treatment for unresectable CRLM with liver resection for resectable CRLM, interpretation of the results in any direction remains very difficult and dangerous. The only real solution to this persistent deadlock is the design of an RCT on RFA versus resection for carefully chosen, resectable, small (≤3 cm), solitary CRLM. Several authors have expressed their sincere concerns about the dangers of conducting such a trial, as it might encourage inappropriate use of RFA by the occasional practitioner of thermal tumor ablation116. But the survey of Birth et al.117 shows that physicians are already autonomously interpreting the available data and, despite the lack of real evidence, deciding for themselves to treat selected resectable patients with RFA. Thus, if an RCT is not performed soon, it will become impossible to determine what is really good clinical practice.

The RFA approach might progressively become a less interesting discussion point. Basically the indication for each approach is a matter of patient selection110. For grossly resectable disease with a few unresectable lesions, there might be a preference for open RFA (ORFA), whereas for patients with unresectable CRLM, who are unfit for major surgery due to

Page 245: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

236

extensive comorbidity, a laparoscopic approach (LRFA) should be used. Furthermore, the experience of the physician performing RFA appears to be an important factor influencing outcome109,111. We performed a retrospective study including 154 patients to compare the morbidty and mortality after LRFA versus ORFA for 291 hepatic malignancies (81 HCC, 157 CRLM, 53 other)118. The LRFA-group was associated with significantly lower intra-operative blood loss, shorter duration of surgery, fewer postoperative complications, and shorter postoperative hospital stay (p<0.01). The complications after LRFA were also less severe than those in the ORFA-group (p<0.01). However, tumor diameter in the ORFA-group was significantly larger as compared to that in the LRFA-group [Table 3118 and Table 4118]. Nevertheless, when only patients with liver tumors measuring ≤3cm were evaluated, the original conclusions remained the same118.Thus, patients with liver tumors ≤3 cm seem to be excellent candidates for LRFA, whereas larger tumors are treated preferably via laparotomy because of technical considerations. However, this is a non-randomized study, including a small number of patients, especially in the subgroup analyses. Therefore, larger patient series in RCTs are needed to better evaluate LRFA versus ORFA for hepatic malignancies.

In open liver surgery, major blood loss implicates a risk of

higher morbidity and mortality as well as shorter long-term survival119-122. Several measures have been described to reduce parenchymal bleeding during the transection phase in open liver surgery, including radiofrequency (RF) energy to create a ‘‘zone of dessication’’ prior to performing hepatic transection. The potential advantages of RF-assisted hepatectomy might be a ‘‘virtually bloodless’’ procedure, shorter operation time, and reduced morbidity119-122. This technique cannot be used close to the liver hilum, since heat might traumatize biliary structures with subsequent bile leakage and/or abscess formation120-123. The RF technique is also unable to control blood vessels

Page 246: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

237

measuring larger than 4 mm in diameter, such as hepatic or portal vein branches124. We performed a prospective study on laparoscopic liver resection (LLR) in 45 patients to evaluate the contribution of RF energy to the amount of blood loss during minor and major laparoscopic hepatectomy125. LLR appears safe and feasible, without mortality, and with a morbidity rate of 24% (11/45) 125. With a median blood loss of 200 ml [range 5-4000ml], an operative duration of 115 minutes [range 45-360 minutes], and a length of hospital stay of 7 days [range 3-41 days], the short-term results concur with literature data126-136. Losing control on blood loss is obviously the most important concern for all authors reporting on LLR126-130,132,135-140. The only intra-operative complication found in our study was bleeding, which was registered in 66.7% (6/9) of the major LLR and in 11.1% (4/36) of the minor LLR. Significant blood loss occurred from large hepatic vessels125. Obviously, this type of bleeding can neither be prevented nor managed by using RF energy. RF-assistance during parenchymal transection in LLR was not associated with reduced intraoperative blood loss, operation time, or complication rate125. Only the type of surgical procedure appeared to determine significantly the amount of blood loss. No other significant correlations were found between the analyzed patient, tumor- and surgery-associated variables, and the amount of intraoperative blood loss125. The resection margins were tumor-free on macroscopic and pathologic examination in all resected specimens. The LLR was accomplished according to the ‘‘no touch’’ technique, without hand-assistance125. As far as postoperative outcome is concerned, the complications observed in the present study are comparable with those most frequently reported in literature128,130,132,133,135,141,142. The same remark can be made for the conversion rate (6.7%)125, as the reported conversion rates vary between 0 and 15.4%126,127,130,132,133,137,143-145. Major intraoperative bleeding during LLR will always remain difficult to control laparoscopically, even with extensive surgical experience. Therefore, a low threshold for conversion to open hepatic resection is indicated.

Page 247: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

238

Tables & Figures [Table x#] = Table x in paper with reference number # [Fig.x#] = Figure x in paper with reference number # References

1. Yan T, Black D, Savady R. Systematic review on the

efficacy of cytoreductive surgery combined with

perioperative intraperitoneal chemotherapy for

peritoneal carcinomatosis fran colorectal cancer. J Clin

Oncol 2006; 24: 4011-4019.

2. Verwaal V, Bruin S, Boot H. 8-year follow-up of

randomized trial: cytoreduction and hyperthermic

intraperitoneal chemotherapy versus systemic

chemotherapy in patients with peritoneal

carcinomatosis of colorectal cancer. Ann Surg Oncol

2008; 15: 2426-2432.

3. Franko J, Ibrahim Z, Gusani N. Cytoreductive surgery

and hyperthermic intraperitoneal chemoperfusion

versus systemic chemotherapy alone for colorectal

peritoneal carcinomatosis. Cancer 2010; 116: 3756-

3762.

4. Elias D, Glehen O, Pocard M. A comparative study of

complete cytoreductive surgery plus intraperitoneal

chemotherapy to treat peritoneal dissemination from

colon, rectum, small bowel and nonpseudomyxoma

appendix. Ann Surg 2010; 251: 896-901.

5. Elias D, Gilly F, Boutitie F. 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.

Page 248: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

239

6. Verwaal V, van Ruth S, Witkamp A. Long-term survival

of peritoneal carcinomatosis of colorectal origin. Ann

Surg Oncol 2005; 12: 65-71.

7. Elias D, Pocard M. Treatment and prevention of

peritoneal carcinomatosis from colorectal cancer. Surg

Oncol Clin N Am 2003; 12: 543-559.

8. van Leeuwen B, Graf W, Pahlman L. Swedish experience

with peritonectomy and HIPEC in peritoneal

carcinomatosis. Ann Surg Oncol 2008; 15: 745-753.

9. Gomes da Silva R, Sugarbaker P. 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.

10. Sadeghi B, Arvieux C, Gilly F. Peritoneal carcinomatosis

from non-gynecologic malignancies. Results of the

EVOCAPE I multicentric prospective study. Cancer 2000;

88: 358–363.

11. Chu D, Lang N, Thompson C. Peritoneal carcinomatosis

in nongynecologic malignancy. A prospective study of

prognostic factors. Cancer 1989; 63: 364–367.

12. Elias D, Gilly F, Glehen O. Peritoneal 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.

13. Davies J, O’Neil B. Peritoneal carcinomatosis of

gastrointestinal origin: Natural history and treatment

options. Exp Opin Invest Drugs 2009; 18: 913–919.

14. Bloemendaal A, Verwaal V, van Ruth S. Conventional

surgery and systemic chemotherapy for peritoneal

Page 249: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

240

carcinomatosis of colorectal origin: A prospective study.

Eur J Surg Oncol 2005; 31: 1145–1151.

15. Hompes D, Boot H, van Tinteren H. Unresectable

Peritoneal Carcinomatosis From Colorectal Cancer: A

Single Center Experience J Surg Oncol 2011; 104: 269–

273.

16. Hompes D, Aalbers A, Boot H. A prospective pilot study

to assess neo-adjuvant chemotherapy for unresectable

peritoneal carcinomatosis from colorectal cancer.

Submitted to Br J Cancer.

17. Jayne D, Fook S, Seow-Choen F. Peritoneal

carcinomatosis from colorectal cancer. Br J Surg 2002;

89: 1545-1550.

18. Folprecht G, Köhne C, Lutz M. Systemic chemotherapy

in patients with peritoneal carcinomatosis from

colorectal cancer. Cancer Treat Res 2007; 134: 425-440.

19. Stillwell A, Ho Y, Veitch C. Systemic review of prognostic

factors related to overall survival in patients with stage

IV colorectal cancer and unresectable metastases.

World J Surg 2011; 35: 684-692.

20. Pelz J, Chua T, Esquivel J. Evaluation of best supportive

care and systemic chemotherapy as treatment stratified

according to the retrospective Peritoneal Surface

Disease Severity Score (PSDSS) for peritoneal

carcinomatosis of colorectal origin. BMC Cancer 2010;

10: 689.

21. Köhne C, Vanhoefer U, Hartung G. Clinical predictive

factors. Eur J Cancer 2009; 45 suppl 1: 43-49.

22. Malik H, Farid S, Al-Mukthar A. A critical appraisal of the

role of neoadjuvant chemotherapy for colorectal liver

metastases: a case-controlled study. Ann Surg Oncol

2007; 14: 3519-3526.

Page 250: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

241

23. Adam R, Vibert E, Pitombo M. Induction chemotherapy

and surgery for colorectal liver metastases. Bull Cancer

2006; 93 Suppl 1: S45-49.

24. Fusai G, Davidson B. Strategies to increase the

respectability of liver metastases from colorectal

cancer. Dig Surg 2003; 20: 481-496.

25. Ruers T, Bleichrodt R. Treatment of liver metastases, an

update on the possibilities and results. Eur J Cancer

2002; 38: 1023-1033.

26. Alberts S, Poston G. Treatment advances in liver-limited

metastatic colorectal cancer. Clin Colorectal Cancer

2011; 10: 258-265.

27. Bismuth H, Adam R, Levi F. Resection of nonresectable

liver metastases from colorectal cancer after

neoadjuvant chemotherapy. Ann Surg 1996: 224: 509-

520.

28. Adam R, Delvart V, Pascal G. Rescue surgery for

unresectable colorectal liver metastases downstaged by

chemotherapy: a model to predict long-term survival.

Ann Surg 2004; 240: 644-657.

29. Van Cutsem E, Nordlinger B, Adam R. Towards a pan-

European consensus on the treatment of patients with

colorectal liver metastases. Eur J Cancer 2006; 42: 2212-

2221.

30. Elias D, Lefevre J, Chevalier J. Complete cytoreductive

surgery plus intraperitoneal chemohyperthermia with

oxaliplatin for peritoneal carcinomatosis of colorectal

origin. J Clin Oncol 2009; 27: 681–685.

31. Verwaal V, van Ruth S, de Bree E. Randomized trial of

cytoreduction and hyperthermic intraperitoneal

chemotherapy versus systemic chemotherapy and

palliative surgery in patients with peritoneal

Page 251: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

242

carcinomatosis of colorectal cancer. J Clin Oncol 2003;

21: 3737-3743.

32. Koppe M, Boerman O, Oyen W. Peritoneal

carcinomatosis of colorectal origin: incidence and

strategies. Ann Surg 2006; 243: 212-222.

33. Hompes D, Ruers T. Review: Incidence and clinical

significance of Bevacizumab-related non-surgical and

surgical serious adverse events I metastatic colorectal

cancer. Eur J Surg Oncol 2011; 37: 737-746.

34. Saltz L, Clarke S, Diaz-Rubio E. Bevacizumab in

combination with oxaliplatin-based chemotherapy as

first-line therapy in metastatic colorectal cancer: a

randomized phase III study. J Clin Oncol 2008; 26: 2013–

9.

35. Kabbinavar F, Hurwitz H, Rosen O. Addition of

bevacizumab to fluorouracil-based first-line treatment

of metastatic colorectal cancer: pooled analysis of

cohorts of older patients from two randomized clinical

trials. J Clin Oncol 2009; 27: 199–205.

36. Hochster H, Hart L, Hedrick E. Safety and efficacy of

oxaliplatin and fluoropyrimidine regimens with or

without bevacizumab as first-line treatment of

metastatic colorectal cancer: results of the TREE Study. J

Clin Oncol 2008; 26: 3523–3529.

37. Giantonio B, Catalano P, Meropol N. Bevacizumab in

combination with oxaliplatin, fluorouracil, and

leucovorin (FOLFOX4) for previously treated metastatic

colorectal cancer: results from the Eastern Cooperative

Oncology Group Study E3200. J Clin Oncol 2007; 25:

1539–1544.

38. Kabbinavar F, Schulz J, Novotny W. Addition of

bevacizumab to bolus fluorouracil and leucovorin in

Page 252: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

243

first-line metastatic colorectal cancer: results of a

randomized phase II trial. J Clin Oncol 2005; 23: 3697–

3705.

39. Kabbinavar F, Hambleton J, Hurwitz H. Combined

analysis of efficacy: the addition of bevacizumab to

fluorouracil/leucovorin improves survival for patients

with metastatic colorectal cancer. J Clin Oncol 2005; 23:

3706–3712.

40. Hurwitz H, Fehrenbacher L, Kabbinavar F. Bevacizumab

plus irinotecan, fluorouracil and leucovorin for

metastatic colorectal cancer. N Engl J Med 2004; 350:

2335–2342.

41. Gruenberger B, Tamandl D, Gruenberger T.

Bevacizumab, capecitabine and oxaliplatin as

neoadjuvant therapy for patients with potentially

curable metastatic colorectal cancer. J Clin Oncol 2008;

26: 1830–1835.

42. Van Cutsem E, Rivera F, Cunningham D. Safety and

efficacy of first-line bevacizumab with folfox, xelox,

folfiri and fluoropyrimidines in metastatic colorectal

cancer: the BEAT study. Ann Oncol 2009; 20: 1842–

1847.

43. Kozloff M, Yood M, Grothey A. Clinical outcomes

associated with bevacizumab-containing treatment of

metastatic colorectal cancer: the BRiTE observational

cohort study. The Oncologist 2009; 14: 862–870.

44. Poultsides G, Servais E, Saltz L. Outcome of primary

tumor in patients with synchronous stage IV colorectal

cancer receiving chemotherapy without surgery as

initial treatment. J Clin Oncol 2009; 27: 3379–3384.

Page 253: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

244

45. Wagner A, Arnold D, Grothey A. Anti-angiogenic therapy

for metastatic colorectal cancer. Cochrane Database

Syst Rev 2009; 8(3): CD005392.

46. Willett C, Boucher Y, di Tomaso E. Direct evidence that

the VEGFspecific antibody bevacizumab has antivascular

effects in human rectal cancer. Nat Med 2004; 10: 145–

147.

47. Gruenberger T, Gruenberger B, Scheithauer W.

Neoadjuvant therapy with bevacizumab. J Clin Oncol

2006; 24: 2592–2593.

48. Willett C, Boucher Y, Duda D. Surrogate markers for

antiangiogenic therapy and dose-limiting toxicities for

bevacizumab with radiation and chemotherapy:

continued experience of a phase I trial in rectal cancer

patients. J Clin Oncol 2005; 23: 8136–8139.

49. Scappaticci F, Fehrenbacher L, Hurwitz H. Surgical

wound healing complications in metastatic colorectal

cancer patients treated with bevacizumab. J Surg Oncol

2005; 91: 173–180.

50. Kesmodel S, Ellis L, Lin E. Preoperative bevacizumab

does not significantly increase postoperative

complication rates in patients undergoing hepatic

surgery for colorectal cancer liver metastases. J Clin

Oncol 2008; 26: 5254–5260.

51. Reddy S, Morse M, Hurwitz H. Addition of bevacizumab

to irinotecan-and oxaliplatin-based preoperative

chemotherapy regimens does not increase morbidity

after resection of colorectal liver metastases. J Am Coll

Surg 2008; 206: 96–106.

52. D’Angelica M, Kornprat P, Saltz L. Lack of evidence for

increased morbidity after hepatectomy with

Page 254: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

245

perioperative use of bevacizumab: a matched case-

control study. Ann Surg Oncol 2007; 14: 759–765.

53. Zawacki W, Walker G, DeVasher E. Wound dehiscence

or failure to heal following venous access port

placement in patients receiving bevacizumab therapy. J

Vasc Interv Radiol 2009; 20: 624–627.

54. Glehen O, Elias D, Deraco M. Cytoreductive surgery

combined with perioperative intraperitoneal

chemotherapy for the management of peritoneal

carcinomatosis from colorectal cancer: a

multiinstitutional study. J Clin Oncol. 2004; 22: 3284–

3292.

55. Elias D, Bonnay M, Puizillou J. Heated intra-operative

intraperitoneal oxaliplatin after complete resection of

peritoneal carcinomatosis: pharmacokinetics and tissue

distribution. Ann Oncol. 2002; 13: 267–272.

56. Hompes D, D'Hoore A, Van Cutsem E. The Treatment of

Peritoneal Carcinomatosis of Colorectal Cancer with

Complete Cytoreductive Surgery and Hyperthermic

Intraperitoneal Peroperative Chemotherapy (HIPEC)

with Oxaliplatin: A Belgian Multicenter Prospective

Phase II Clinical Study. Ann Surg Oncol 2012; 19: 2186-

2194.

57. Verwaal V, Zoetmulder F. Long-term survival of

peritoneal carcinomatosis of colorectal origin. Ann Surg

Oncol 2005; 12: 65–71.

58. Andres A, Majno P, Morel P. Improved long-term

outcome of surgery for advanced colorectal liver

metastases: reasons and implications for management

on the basis of a severity score. Ann Surg Oncol. 2008;

15: 134–143.

Page 255: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

246

59. Arru M, Aldrighetti L, Castoldi R. Analysis of prognostic

factors influencing long-term survival after hepatic

resection for metastatic colorectal cancer. World J Surg.

2008; 32: 93–103.

60. Choti M, Sitzmann J, Tiburi M. Trends in long-term

survival following liver resection for hepatic colorectal

metastases. Ann Surg. 2002; 235: 759–766.

61. Zakaria S, Donohue JH, Que F. Hepatic resection for

colorectal metastases: value for risk scoring systems?

Ann Surg 2007; 246: 183–191.

62. House M, Ito H, Fong Y. Survival after hepatic resection

for metastatic colorectal cancer: trends in outcomes for

1,600 patients during two decades at a single

institution. J Am Coll Surg. 2010; 210: 744–752.

63. Maggiori L, Elias D. Curative treatment of colorectal

peritoneal carcinomatosis: current status and future

trends. Eur J Surg Oncol. 2010; 36: 599–603.

64. Elias D, Blot F. Curative treatment of peritoneal

Carcinomatosis arising from colorectal cancer by

complete resection and intraperitoneal chemotherapy.

Cancer. 2001; 92: 71–76.

65. Soulié P, Raymond E, Brienza S. Oxaliplatin: the first

DACH platinum in clinical practice. Bull Cancer 1997; 84:

665-673.

66. de Gramont A, Figer A, Seymour M. Leucovorin and

fluorouracil with or without oxaliplatin as first-line

treatment in advanced colorectal cancer. J Clin Oncol

2000; 18: 2938-2947.

67. Makatsoris T, Kalafonos H, Aravantinos G. A phase II

study of capecitabine plus oxaliplatin (XELOX): a new

first-line option in metastatic colorectal cancer. Int J

Gastrointest Cancer 2005; 35: 103-109.

Page 256: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

247

68. Scheithauer W, Kornek G, Raderer M. Randomized

multicenter phase II trial of two different schedules of

capecitabine plus oxaliplatin as first-line treatment in

advanced colorectal cancer. J Clin Oncol 2003; 21: 1307-

1312.

69. Alkis N, Demerci U, Benekli M. Mitomycin-C in

combination with fluoropyrimidines in the treatment of

metastatic colorectal cancer after oxaliplatin and

irinotecan failure. J BUON 2011; 16: 80-83.

70. Kang E, Choi Y, Kim J. Mitomycin-C, 5-fluorouracil, and

leucovorin as a salvage therapy in patients with

metastatic colorectal adenocarcinoma. Asia Pac J Clin

Oncol 2010; 6: 286-291.

71. Michalaki V, Gennatas S, Gennatas C. Mitomycin C and

UFT/leucovorin as salvage treatment in patients with

advanced colorectal cancer. J BUON 2010; 15: 270-273.

72. Vormittag L, Kornek G, Gruhsmann B. UFT/leucovorin

and mitomicyn C as salvage treatment in patients with

advanced colorectal cancer – a retrospective analysis.

Anticancer Drugs 2007; 18: 709-712.

73. Pestieau S, Belliveau J, Griffin H. Pharmacokinetics of

intraperitonealoxaliplatin: experimental studies. J Surg

Oncol 2001; 76: 106-114.

74. Lambert L, Armstrong T, Lee J. Incidence, risk factors,

and impact of severe neutropenia after hyperthermic

intraperitoneal mitomycin C. Ann Surg Oncol 2009; 16:

2181-2187.

75. van Ruth S, Verwaal V, Zoetmulder F. Pharmacokinetics

of intraperitoneal mitomycin C. Surg Oncol Clin N Am

2003; 12: 771-780.

Page 257: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

248

76. Kusamura S, Elias D, Baratti D. Drugs, carrier solutions

and temperature in hyperthermic intraperitoneal

chemotherapy. J Surg Oncol 2008; 98: 247-252.

77. van Ruth S, Mathôt R, Sparidans R. Population

pharmacokinetics and pharmacodynamics of mitomycin

during intraoperative hyperthermic intraperitoneal

chemotherapy. Clin Pharmacokinet 2004; 43: 131-143.

78. Elias D, Pocard M, Goere D. HIPEC with oxaliplatin in the

treatment of peritoneal carcinomatosis of colorectal

origin. Cancer Treat Res 2007; 134: 303-318.

79. Hompes D, D’Hoore A, Mirck B. The use of Oxaliplatin or

Mitomycin C in HIPEC treatment for peritoneal

carcinomatosis from colorectal cancer: a comparative

study. Submitted to Eur J Cancer.

80. Bécouarn Y, Ychou M, Ducreux M. Phase II trial of

oxaliplatin as first-line chemotherapy in metastatic

colorectal cancer patients: Digestive Group of French

Federations of Cancer Centers. J Clin Oncol 1998; 16:

2739-2744.

81. Elias D, Benizri E, Di Pietrantonio D. Comparison of two

kinds of intraperitoneal chemotherapy following

complete cytoreductive surgery of colorectal peritoneal

carcinomatosis. Ann Surg Oncol 2007; 14: 509-514.

82. De Somer F, Ceelen W, Delanghe J. Severe

hyponatremia, hyperglycemia, and hyperlactatemia are

associated with intraoperative hyperthermic

intraperitoneal chemoperfusion with oxaliplatin. Perit

Dial Int 2008; 28: 61-66.

83. Ceelen W, Peeters M, Houtmeyers P. Safety and efficacy

of hyperthermic intraperitoneal chemoperfusion with

high-dose oxaliplatin in patients with peritoneal

carcinomatosis. Ann Surg Oncol 2008; 15: 535-541.

Page 258: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

249

84. Verwaal V, van Tinteren H, van Ruth S. Predicting the

survival of patients with peritoneal carcinomatosis of

colorectal origing treated by aggressive cytoreduction

and hyperthermic intraperitoneal chemotherapy. Br J

Surg 2004; 91: 739-746.

85. Swellengrebel H, Zoetmulder F, Verwaal V. Quantitative

intra-operative assessment of peritoneal carcinomatosis

- a comparison of three prognostic tools. Eur J Surg

Oncol 2009; 35: 1078-1084.

86. Verwaal V, van Tinteren H, van Ruth S. Toxicity of

cytoreductive surgery and hyperthermic intra-

peritoneal chemotherapy. J Surg Oncol 2004; 85: 61-67.

87. Graham M, Lockwood G, Greenslade D. Clinical

pharmacokinetics of oxaliplatin: a critical review. Clin

Cancer Res 2000; 6: 1205-1218.

88. Kautio A, Haanpää M, Kautiainen H. Oxaliplatin Scale

and National Cancer Institute-Common Toxicity Criteria

in the Assessment of Chemotherapy-induced Peripheral

Neuropathy. Anticancer Res 23011; 31: 3493-3496.

89. Grothey A. Oxaliplatin-safety: neurotoxicity. Semin

Oncol 2003; 30: 5-13.

90. Stuart O, Stephens A, Welch L. Safety monitoring of the

coliseum technique for heated intraoperative

intraperitoneal chemotherapy with mitomycin C. Ann

Surg Oncol 2002; 9: 186-191.

91. Schmid K, Boettcher M, Pelz J. Investigations on safety

of hyperthermic intraoperative intraperitoneal

chemotherapy (HIPEC) with mitomycin C. Eur J Surg

Oncol 2006; 32: 1222-1225.

92. Näslund Andréasson S, Anundi H, Thorén S. Is platinum

present in blood and urine from treatment givers during

Page 259: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

250

hyoerthermic intraperitoneal chemotherapy? J Oncol

2010; 2010: 649719.

93. Shepherd N, Baxter K, Love S. The prognostic

importance of peritoneal involvement in colonic cancer:

a prospective evaluation. Gastroenterology 1997; 112:

1096-1102.

94. Ludeman L, Shepherd N. Serosal involvement in

gastrointestinal cancer: its assessment and significance.

Histopathology 2005; 47: 123-131.

95. Shepherd N, Baxter K, Love S. Influence of local

peritoneal involvement on pelvic recurrence and

prognosis in rectal cancer. J Clin Pathol 1995; 48: 849-

855.

96. Petersen V, Baxter K, Shepherd N. Identification of

objective pathological prognostic determinants and

models of prognosis in Dukes’ B colon cancer. Gut 2002;

51: 65-69.

97. D.Hompes, J.Tiek, A.Wolthuis. HIPEC in T4a colon

cancer: a defendable treatment to improve oncologic

outcome? Accepted to Ann Oncol 2012

98. Noura S, Ohue M, Seki Y. Long-term prognostic value of

conventional peritoneal lavage cytology in patients

undergoing curative colorectal cancer resection. Dis

Colon Rectum 2009; 52: 1312-1320.

99. Bosch B, Guller U, Schnider A. perioperative detection

of disseminated cells is an independent prognostic risk

factor in patients with colorectal cancer. Br J Surg 2003;

90: 882.

100. Guller U, Zajac P, Schnider A. Disseminated single tumor

cells as detected by real-time quantitative polymerase

chain reaction represent a prognostic factor in patients

Page 260: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

251

undergoing surgery for colorectal cancer. Ann Surg

2002; 236: 768-75; discussion 775-776.

101. Hara M, Nakanishi H, Jun Q. Comparative analysis of

intraperitoneal minimal free cancer cells between

colorectal and gastric patients: using quantitative RT-

PCR: possible reason for rare peritoneal recurrence in

colorectal cancer. Clin Exp Metastasis 2007; 24: 179-

189.

102. Noura S, Ohue M, Shingai T. Effects of intraperitoneal

chemotherapy with Mitomycin C on the prevention of

peritoneal recurrence in colorectal cancer patients with

positive peritoneal lavage cytology findings. Ann Surg

Oncol 2010; 18: 396-404.

103. Van Ruth S, Mathôt R, Verwaal V. Population

pharmacokinetics and pharmacodynamics of mitomycin

during intraoperative hyperthemic intraperitoneal

chemotherapy. Clin Pharmacokinet 2004; 43: 131-143.

104. Elias D, Goere D, Blot F. Optimization of hyperthermic

intraperitoneal chemotherapy with oxaliplatin plus

irinotecan at 43 degrees C after complete cytoreductive

surgery: mortality and morbidity in 106 consecutive

patients. Ann Surg Oncol 2007; 14: 1818-1824.

105. Elias D, Sideris L. Pharmacokinetics of heated

intraoperative intraperitoneal oxaliplatin after complete

resection of peritoneal carcinomatosis. Surg Oncol Clin

N Am 2003; 12: 755-69.

106. Elias D, Goere D, Di Pietrantonio D. Results of

systematic second-look surgery in patients at high risk

of developing colorectal peritoneal carcinomatosis. Ann

Surg 2008; 247: 445-450.

107. Elias D, Honoré C, Dumont F. Results of systematic

second-look surgery plus HIPEC in asymptomatic

Page 261: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

252

patients presenting a high risk of developing colorectal

peritoneal carcinomatosis. Ann Surg 2011; 254: 289-

293.

108. Hompes D, Prevoo W, Ruers T. Review: Radiofrequency

ablation as a treatment tool for liver metastases of

colorectal origin. Cancer Imaging 2011; 11: 23-30.

109. Mulier S, Ni Y, Jamart J. Local recurrence after hepatic

radiofrequency coagulation: multivariate meta-analysis

and review of contributing factors. Ann Surg 2005; 242:

158-171.

110. Joosten J, Ruers T. Local radiofrequency ablation

techniques for liver metastases of colorectal cancer. Crit

Rev Oncol Hematol 2007; 62: 153-163.

111. Mulier S, Ni Y, Ruers T. Radiofrequency ablation versus

resection for resectable colorectal liver metastases:

time for a randomized trial? Ann Surg Oncol 2008; 15:

144-157.

112. Ruers T, Joosten J, Wiering B. Comparison between local

ablative therapy and chemotherapy for non-resectable

colorectal liver metastases: a prospective study. Ann

Surg Oncol 2007; 14: 1161-1169.

113. Abdalla E, Vauthey J, Ellis L. Recurrence and outcomes

following hepatic resection, radiofrequency ablation

and combined resection/ablation for colorectal liver

metastases. Ann Surg 2004; 239: 818-827.

114. Abdalla E. Commentary: Radiofrequency ablation for

colorectal liver metastases: do not blame the biology

when it is the technology. Am J Surg 2009; 197: 737-

739.

115. Mulier S, Ruers T, Jamart J. Radiorequency ablation

versus resection for resectable colorectal liver

Page 262: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

253

metastases: time for a randomized trial? An update. Dig

Surg 2008; 25: 445-460.

116. Curley A. Radiofrequency ablation versus resection for

resectable colorectal liver metastases: time for a

randomized trial? Ann Surg Oncol 2007; 15: 11-13.

117. Birth M, Hildebrand P, Dahmen G. Aktueller Stand der

Radiofrequenzablation von lebertumoren: eine

deutschlandweite Umfrage. Chirurg 2004; 75: 417-423.

118. Topal B, Hompes D, Aerts R. Morbidity and mortality of

laparoscopic vs. open radiofrequency ablation for

hepatic malignancies. Eur J Surg Oncol 2007; 33: 603-

607.

119. Haghighi K, Wang F, King J. In-line radiofrequency

ablation to minimize blood loss in hepatic parenchymal

transection. Am J Surg 2005; 190: 43–47.

120. Navarra G, Spalding D, Zacharoulis D. Bloodless

hepatectomy technique. HPB Surg 2002; 4: 95–97.

121. Stella M, Percivale A, Pasqualini M. Radiofrequency-

assisted liver resection. J Gastrointest Surg 2003; 7:

797–801.

122. Weber J, Navarra G, Jiao L. New technique for liver

resection using heat coagulative necrosis. Ann Surg

2002; 236: 560–563.

123. Navarra G, Lorenzini C, Currò G. Early results after

radiofrequency-assisted liver resection. Tumori 2004;

90: 32–35.

124. Machi J. New technique for liver resection using heat

coagulative necrosis – letter to the editor. Ann Surg

2004; 241: 194–196.

125. Hompes D, Aerts R, Penninckx F. Laparoscopic liver

resection using radiofrequency coagulation. Eur J Surg

Oncol 2007; 33: 603-607.

Page 263: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

254

126. Belli G, Fantini C, D’Agostino A. Laparoscopic liver

resections for hepatocellular carcinoma (HCC) in

cirrhotic patients. HPB 2004; 6: 236–246.

127. Biertho L, Waage A, Gagner M. Hépatectomies sous

laparopscopie. Ann Surg 2002; 127: 164–170.

128. Cherqui D, Husson E, Hammoud R. Laparoscopic liver

resections: a feasibility study in 30 patients. Ann Surg

2000; 232: 753–762.

129. Fujita F, Fujita R, Kanematsu T. New approaches to the

minimally invasive treatment of liver cancer. Cancer J

2005; 11: 52–56.

130. Gagner M, Rogula T, Selzer D. Laparoscopic liver

resection: benefits and controversies. Surg Clin North

Am 2004; 84: 451–462.

131. Hansen P. Laparoscopic resections of liver and pancreas.

J Gastrointest Surg 2004; 8: 925–926.

132. Mala T, Edwin B, Gladhaug I. A comparative study of the

short-term outcome following open and laparoscopic

liver resection of colorectal metastases. Surg Endosc

2002; 16: 1059–1063.

133. O’Rourke N, Fielding G. Laparoscopic right

hepatectomy: surgical technique. J Gastrointest Surg

2004; 8: 213–216.

134. Shimada M, Harimoto N, Maehara S. Minimally invasive

hepatectomy: modulation of systemic reactions to

operation or laparoscopic approach? Surgery 2002;

131(Suppl 1): S312–S317.

135. Vibert E, Kouider A, Gayet B. Laparoscopic anatomic

liver resection. HPB 2004; 6: 222–229.

136. Weber J, Navarra G, Habib N. Laparoscopic

radiofrequency-assisted liver resection. Surg Endosc

2003; 17: 834.

Page 264: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

255

137. Croce E, Olmi S, Bertolini A. Laparoscopic liver resection

with radiofrequency. Hepatogastroenterology 2003; 50:

2088–2092.

138. Kalil A, Mastalir E. Laparoscopic hepatectomy for benign

liver tumors. Hepatogastroenterology 2002; 49: 803–

805.

139. Lesurtel M, Cherqui D, Laurent A. Laparoscopic versus

open left lateral lobectomy: a case-control study. J Am

Coll Surg 2003; 196: 236–242.

140. O’Rourke N, Shaw I, Nathanson L. Laparoscopic

resection of hepatic colorectal metastases. HPB 2004; 6:

230–235.

141. Descottes B, Glineur D, Lachachi F. Laparoscopic liver

resection of benign liver tumors. Surg Endosc 2003; 17:

23–30.

142. Kaneko H, Takagi S, Otsuka Y. Laparoscopic liver

resection of hepatocellular carcinoma. Am J Surg 2005;

189: 190–194.

143. Cherqui D. Résections hépatiques par voie coelioscopic:

utiles ou futiles? Ann Chir 2002; 127: 171–174.

144. Cherqui D. Laparoscopic liver resection. Br J Surg 2003;

90: 644–646.

145. Laurent A, Cherqui D, Lesurtel M. Laparoscopic liver

resection for subcapsular hepatocellular carcinoma

complicating chronic liver disease. Arch Surg 2003; 138:

763–769.

Page 265: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

256

Page 266: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

257

VI

Conclusions

Page 267: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

258

Page 268: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

259

The scope of this clinical thesis is “advanced” and metastatic colorectal cancer (CRC). About 20-30% of patients with CRC present with stage IV disease at diagnosis and about 50-60% of patients with CRC will develop metastases at some point in the course of their disease. Until 2 decades ago chemotherapy was the only available treatment option for this substantial patient group, but as systemic treatment progressively improved, a large number of initially unresectable patients were rendered resectable. Subsequently, the surgeon’s active participation in the treatment of metastatic disease has increased and changed substantially. This thesis focused on peritoneal carcinomatosis (PC) and liver metastases (CRLM) and addressed the 3 following subjects:

Unresectable PC from CRC and neo-adjuvant chemotherapy

A retrospective study of patients with unresectable PC from CRC at a single institute found the administration of systemic chemotherapy to be the only factor to significantly influence survival, resulting in a median overall survival (OS) of 9.3 months. But this benefit in survival probably largely attributable to the fact that this patient group has a better performance status at diagnosis, thus enabling them to tolerate the systemic treatment.

Subsequently, an observational pilot study was performed to progressively assess the actual effect of modern systemic chemotherapy on unresectable PC from CRC, by performing a laparoscopy before and after “neo-adjuvant” chemotherapy. Trial inclusion was discontinued, because none of the included patients showed macroscopic response to the chemotherapy. All patients showed either progressive or stable disease. This indicates that PC, which is in itself a local phenomenon, does not respond well enough to chemotherapy to render unresectbale PC from CRC resectable. A review on the incidence and clinical significance of Bevacizumab(BV)-related non-surgical and surgical serious adverse events (SAE) was performed to assess the morbidity and

Page 269: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

260

mortality that result from the administration of neo-adjuvant treatment with BV. Even trials consisting of large patient populations report small numbers of BV-related SAE in relationship to surgery. This makes statistical analysis and definitive conclusions difficult. On the other hand, the fact that even large populations show low absolute numbers of BV-related SAE in any setting, suggests that BV-based treatment causes few clinically significant problems, even when surgical procedures are involved.

Complete cytoreductive surgery and HIPEC for PC from CRC

A Belgian multicentre prospective phase II clinical study was performed to assess the treatment of PC from CRC with complete cytoreductive surgery (CCRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) with Oxaliplatin. This invasive combined modality treatment can be implemented with acceptable morbidity and in well-selected patients, in whom complete cytoreduction can be achieved, it can result in real long-term DFS and OS.

A retrospective study of prospectively gathered data from 2 different HIPEC centers compared CCRS+HIPEC with Oxaliplatin versus Mitomicyn C (MMC) as the intraperitoneal drugs. Based on the available data on safety and efficacy of CCRS+HIPEC in patient with PC from CRC, no strong plea can be made for the use of either Oxaliplatin or MMC. Although Oxaliplatin has become standard systemic treatment in CRC, no clear benefit in OS and RFS can be demonstrated for HIPEC in this trial or in literature. MMC often results in neutropenia, but the clinical significance of this finding is minimal. Oxaliplatin and MMC result in equally high morbidity rates. Indirect arguments for which one might prefer Oxaliplatin over MMC as the intraperitoneal drug are the proven efficacy of (systemic) Oxaliplatin in CRC, the proven high Oxaliplatin concentrations in the targeted colonic tissues after HIPEC, the fact that Oxaliplatin is not metabolized hepaticly and - in this time of cost and facility restrictions - the shorter HIPEC perfusion time (only 30 minutes).

Page 270: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

261

A retrospective analysis of a large cohort of consecutive patients undergoing surgery for colon cancer showed clearly that T4a colon tumors are at significantly higher risk of developing PC. Furthermore, 25% of stage II and III T4 tumors developed PC as the only site of metastases. This defines the window of opportunity for adjuvant HIPEC to prevent peritoneal recurrence. However, to allow the implementation of HIPEC as an adjuvant treatment in this high risk group of patients 2 important hurdles need to be overcome: First, to demonstrate the benefit of HIPEC in this population, a RCT with a large sample size is needed. Second, there is an urgent need to evaluate and validate diagnostic methods to allow peroperative detection of IPTC and/or transserosal invasion (pT4a). For the time being, a strategy as proposed by Elias et al. (second look surgery) could be the better approach. Radiofrequency ablation (RFA) for CRLM

A review of literature on RFA as a treatment tool for CRLM showed the efficacy and reliability of RFA depends mainly on the diameter of the targeted lesions (RFA in lesions >5cm is questionable), the applied approach (surgical or percutaneous RFA), the distance between the targeted lesion and large vessels (because of the heat sink phenomenon) and the experience of the physician performing the RFA procedure. For unresectable CRLM, any combined modality treatment with RFA should be prefered over chemotherapy-only. For the use of RFA in patients with resectable CRLM, as an alternative to surgery, there is a lack of evidence in current literature and the results for series on RFA for small solitary lesions are contradictory. Nevertheless, for small CRLM, located centrally in the liver, RFA would result in a more parenchymal sparing policy, which would leave more room for retreatment of future recurrent or new lesions. Most papers currently available in the literature compare RFA for unresectable CRLM with liver resection for resectable CRLM, which makes interpretation of data very difficult and dangerous. An RCT, comparing resection to RFA for carefully chosen,

Page 271: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

262

resectable, small (≤3 cm), solitary CRLM, would bring answers. But in reality, conducting such a trial is difficult and possibly dangerous. On the other hand, physicians are already autonomously interpreting the available data and, despite the lack of real evidence, deciding for themselves to treat selected resectable patients with RFA. The approach for RFA (laparoscopic [LRFA] versus open [ORFA]) might progressively become a less interesting discussion point. Basically, the indication for each approach is a matter of patient selection, with a preference for ORFA in the case of grossly resectable disease with a few unresectable lesions and for LRFA in patients with unresectable CRLM, unfit for major surgery because of extensive comorbidity. We performed a retrospective analysis to assess morbidity and mortality of LRFA versus ORFA for hepatic malignancies. LRFA was associated with better short-term clinical outcome as compared to ORFA. Simultaneous colorectal and/or hepatic resection resulted in an increased postoperative complication rate. Losing control on blood loss is obviously the most important concern of surgeon’s performing laparoscopic liver resections (LLR). We performed a prospective, observational trial on LLR using radiofrequency(RF)-coagulation in an attempt to decrease intra-operative blood loss. Laparoscopic liver resection is feasible and safe, if adequate equipment and extensive experience of the surgical team are available. LLR can be performed with minimal amounts of intraoperative blood loss, which is determined by the type of hepatectomy. Significant intraoperative hemorrhage occurs from large hepatic vessels during major LLR, which cannot be controled by RF. RF-assisted parenchymal transection in LLR does not seem to reduce blood loss, operation time, or perioperative morbidity.

Page 272: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

263

Acknowledgements

Page 273: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

264

Page 274: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

265

Verspreid over meerdere decennia schreef Marguerite Yourcenar (Amerikaanse schrijfster van Frans-Belgische oorsprong, 1903-1987), de historisch-psychologische roman “Mémoires d’Hadrien”. Voor deze denkbeeldige autobiografie van keizer Hadrianus, geschreven in de ik-persoon, verdiepte Yourcenar zich jarenlang bijna obsessief in het leven en de tijd van deze Romeinse Keizer en de eerste 2 versies van haar roman liet ze eigenhandig integraal in de prullemand belanden. Toen dit indrukwekkende werk dan toch eindelijk een in haar ogen publiceerbare versie had bereikt, schreef ze in het laatste hoofdstuk de volgende opdracht aan een vriend: “… zelfs de langste opdracht is nog een te onvolledige en te alledaagse manier om hulde te brengen aan een zo ongewone vriendschap. Als ik probeer dit geluk dat me sinds jaren is geschonken te omschrijven, zeg ik tegen mezelf dat een dergelijk voorrecht, hoe zeldzaam ook, toch niet uniek kan zijn; dat er soms, een beetje op de achtergrond, in het ontstaansproces van een tot een goed einde gebracht boek of in een gelukkig schrijversleven iemand moet zijn die de onjuist of zwak geformuleerde zin, die wij uit vermoeidheid wilden laten staan, niet laat passeren; iemand die als het moet twintig keer met ons een onzekere bladzijde overleest; iemand die voor ons van de bibliotheekplanken de lijvige boekdelen pakt waarin we hopen een nuttige aanwijzing te vinden, en die er nog harnekkiger in blijft zoeken op het moment dat wij ze al ontmoedigd hebben gesloten; iemand die de vreugden van de kunst en van het leven, hun vele, nooit vervelende en nooit gemakkelijke werkzaamheden in gelijk enthousiasme met ons deelt; iemand die noch onze schaduw noch ons spiegelbeeld en zelfs niet ons complement is, maar zichzelf; iemand die ons goddelijk vrijlaat en ons toch verplicht geheel en al te zijn wat we zijn. Hospes comesque. …” Hospes comesque. Je kan het, denk ik, niet mooier samenvatten. En nu het mijn beurt is om een dankwoord neer te schrijven, dringt het tot mij door dat dit inderdaad een vrijwel onmogelijke opdracht is, omdat ik zo ongeveer iedere minuut

Page 275: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

266

van mijn leven omringd ben geweest door liefde, warmte, vriendschap, goed advies en vertrouwen. Dus welke woorden ik hierna ook op papier zet, ze zullen hoe dan ook majeur tekortschieten, maar ik ga het – tegen beter weten in – toch proberen…

Lieve mama en papa, ik zou nooit in staat geweest zijn om deze lange en eerlijk gezegd soms erg moeizame weg te gaan zonder jullie onvoorwaardelijke liefde en steun. Ik ben wie ik ben omdat ik steeds, no matter what time of day and no matter what the circumstances are, echt kan “thuiskomen”, omdat alles altijd bespreekbaar is, omdat jullie mij elke dag voorgeleefd hebben om anderen met warmte, respect, een open vizier en oprechte belangstelling tegemoet te treden, omdat jullie de dingen met het nodige geduld, relativeringsvermogen, rechtvaardigheid en heel veel zin voor humor beluisterd en (gevraagd en soms ook ongevraagd ) van rustige commentaar voorzien hebben en omdat jullie Titia en mij elke dag het gevoel geven dat we iets bijzonders zijn. We are two very lucky girls!

Lieve Titia, je bent de puurste, eerlijkste, warmste en

meest oprechte mens die ik ken. Bedankt voor al die onvoorwaardelijke liefde en interesse waarmee je elke dag opnieuw mijn lief en leed en mijn kwetsbaarheid hebt gedeeld. Over “lange, moeilijke weg” gesproken: jij bent zelf ook meer dan ervaringsdeskundig! Ik ben ongelooflijk trots op jou en oneindig dankbaar dat je er bent, “mijn zus”. Lieber Matthias, danke dir für deine Freundschaft, deine Ruhe, deine Geduld und dein fantastischen Sinn für Humor: wir sind alle sehr froh das du jetzt zur Familie gehörst!

En natuurlijk bedankt aan mijn familie (in ’t bijzonder alle

Bomanskes!): voor alle liefde, steun en oprechte interesse die ik steeds weer van jullie kreeg.

En toen… “Inspiration is everything”, in het leven en

zeker ook in een opleiding tot chirurg:

Page 276: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

267

LEUVEN Ik koester uiteraard diepe dankbaarheid voor alle stafleden Abdominale Heelkunde van het UZ Leuven, die mij gedurende meer dan 3 jaren opgeleid, bijgestuurd en gedragen hebben. Maar ik wil toch graag 2 van mijn vroegere “bazen” in het bijzonder vermelden:

Prof. André d’Hoore: André, bedankt voor ALLES! - voor je consequente dagelijkse aanwezigheid met kritische commentaren op elke ochtendkrans (een onwaarschijnlijk waardevol en onvervangbaar dagelijks trainingsmoment!) - voor het doorgeven van je doorgedreven streven naar kwaliteit (soms met het nodige ongeduld als de dingen niet snel genoeg de richting uitgingen die jij voor ogen had ) - voor je feilloze gevoel voor timing en schitterende one-liners: net op het moment dat je als assistent/resident geen vooruitgang meer maakte in een operatie en dus een beetje rond de pot aan ’t draaien was, bleek er plots iemand achter je schouder mee te kijken: “wa zijt ge aan ’t doen?... Geen grote operatie van maken, hè…” - voor de vele kansen die ik kreeg, chirurgisch-technisch en wetenschappelijk - voor het vertrouwen dat ik kreeg op een moment dat ik er zelf geen vertrouwen meer in had dat het ooit een keer zou goedkomen - voor de vele overlegmomenten, vaak op zondagmiddag in het studiekot van Abdominale, om mijn projecten/papers vooruit te helpen en bij te sturen - … Ik ben heel trots dat je mijn co-promotor wil zijn. Dit manuscript zou er niet liggen zonder jou. De opleiding was soms keihard en een ongelooflijke uitputtingsslag, maar ik ben ontzettend dankbaar om ieder moment ervan. Chirurgisch ben ik wie ik ben door jou. And you will always be the little voice in my head, pushing me to do better.

Page 277: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

268

Dr.Raymond Aerts: beste dokter Aerts, ik schreef het al een keer in een mailtje aan u toen ik naar Amsterdam vertrok in augustus 2009: sommige mensen laten zonder veel lawaai te maken voor altijd een diepe indruk op je na, gewoon door hun kennis, hun technisch kunnen, hun dagelijks functioneren, hun manier van met mensen omgaan en zich engageren voor patiënten. Eén van uw directe collega’s zei een keer over u: “Als je iets niet weet, vraag het aan Raymond. Als je een goed advies nodig hebt, vraag het aan Raymond. En als je wil dat iemand een keer écht goed naar je patiënt kijkt, vraag het aan Raymond.” Mooier kan ik het niet zeggen. Bedankt voor alle kansen, al uw geduld en al het vertrouwen dat ik kreeg.

Lieve Guido en Isabelle, bedankt voor jullie support en vriendschap!

Ik zeg hier ook uit de grond van mijn hart “bedankt” aan die vele, vele anderen die, elk op hun manier, op enig moment van mijn opleiding een substantiële inspiratie betekend hebben. Ik noem er hier enkelen en excuseer mij al op voorhand voor de vele anderen die ik hier ongetwijfeld vergeet: Dr. Michel Van Pelt, Dr. Karel Mulier, Dr. Peter Reynaert, Dr. Heidi Weyns, Dr. Joep Knol, Dr. Tom Feryn, Dr. Michel Van Brussel, Dr. Marc Vandevoort, Dr. Johan Raymakers, Dr. Jef Van Baal, Dr. Srdjan Rakic, Dr. Peter Smits, Prof. Chris Verslype, Prof. Alexander Wilmer, Prof. Patrick Ferdinande, Prof. Paul Sergeant, Prof. Wim Daenen, de ganse staf Thoraxheelkunde UZ Leuven en vele anderen…

AMSTERDAM Beste promotor, beste Emiel, je was de eerste met wie ik sprak tijdens mijn sollicitatiegesprek in het AVL en ik was meteen op mijn gemak. Dat is daarna altijd zo gebleven. In een Vlaamse opleiding geldt het motto “geen nieuws, is goed nieuws”: als er niks fundamenteels de mist ingaat, hoeft er met supervisie ook niets besproken te worden. Dus toen ik na 1 jaar in het AVL voor het eerst in mijn leven voor een evaluatiegesprek werd

Page 278: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

269

uitgenodigd, dacht ik: “ik heb echt niks te melden… dat gaat hier wat mij betreft geen 5 minuten duren…” Niets bleek minder waar: we spraken meer dan een uur, jij bleek verdomd goed op de hoogte te zijn en ik vond het nog een zeer goed gesprek ook! Je begreep blijkbaar ook heel goed dat het voor een rasechte Vlaamse onder die extraverte Amsterdamse “overlegcultuur-Nederlanders” niet altijd zo evident communiceren was. Bedankt voor al je steun en bijsturing op de achtergrond als dat nodig was.

Beste co-promotor, beste Victor, ik was op voorhand al erg geïnteresseerd in peritoneale maligniteiten en HIPEC en dat is in Amsterdam alleen maar “erger” geworden. Bedankt voor de inspiratie en de grenzeloze mogelijkheden en back-up die ik kreeg om operatief-technisch te leren en kilometers te maken. We hebben beiden niet bepaald een eenvoudig karakter en schoten dus ook niet altijd “probleemloos” op. Toen je in ons afscheidsgesprek op mijn laatste AVL-dag vroeg naar mijn opmerkingen, zei ik : “We hebben er wel ruim een jaar over gedaan om onze weg naar goeie communicatie te vinden, hè, Vic…”. Je antwoordde met een grijns: “Zo! Dat heb je dan snel voor mekaar gekregen!” Typisch jij ! Ik heb ontzettend veel geleerd en je hebt mij meer dan eens getoond wat het betekent om voor een patiënt tot het uiterste te gaan en het onderste uit de kan te halen. Bedankt voor alles!

Beste Theo, ik heb op heel veel manieren ontzettend veel van jou geleerd. Bedankt voor het vertrouwen: je hebt mij in heel korte tijd een fenomenaal aantal levers en rectums laten opereren. Bedankt voor je begeleiding bij het schrijven van de review en de editorial in dit proefschrift. We hadden nog heel veel andere projectideeën, maar de tijd was wat te kort om het allemaal te verwezenlijken. Bedankt ook voor je rust, diplomatie en strategisch denken. Je hebt me geleerd om goed na te denken, vooraleer ergens vol enthousiasme aan te beginnen: “Waar wil ik naartoe?”. And last but not least: bedankt voor je steun in de extra 6 maanden die ik in het AVL mocht

Page 279: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

270

doorbrengen: zonder jou zou de bijkomende sarcomenopleiding financieel niet mogelijk geweest zijn en de onderhandelingen met UZ Leuven over mijn verdere toekomst zouden een stuk moeilijker geweest zijn.

Lieve Frits, wat ik van jou gekregen heb, heeft het voor mij mentaal en technisch mogelijk gemaakt het werk te doen dat ik nu elke dag doe. Je leerde mij nadenken over sarcoompathologie, opereren met systematiek, met oog voor detail en precisie, met fascinatie voor de pathologie en respect voor de tumor, met een ongelooflijk engagement voor de patiënt, met een grote liefde voor rust en bescheidenheid en met een onevenaarbare zin voor humor. Bovendien heb ik van jou geleerd om met gezond verstand te redeneren en te beslissen in the best interest of the patient: het is niet omdat iets technisch mogelijk is, dat het ook zinvol is om het te doen. En daarbij was ik ook nog eens welkom bij jou en Hetta thuis! Jullie zijn beiden heel bijzonder! Dikke kus en bedankt voor je geduldige support tot op vandaag. Uiteraard ook dank aan alle andere leden van de Sarcomen-groep in het NKI-AVL en met name aan Rick Haas voor de teaching en begeleiding.

Geachte leden van de Leescommissie, hartelijk dank aan u allen dat u bereid bent aanwezig te zijn en te participeren aan mijn promotieceremonie.

Prof. Eric Van Cutsem, dank voor de persistente, rustige support op de achtergrond en de vele deuren die u voor mij geopend hebt. Ook heel bijzondere dank aan Prof. Chris Verslype voor de trouwe steun. Lieve Loes, dank voor het geduld tijdens de vele uren die we in het kader van de leverprojecten samen doorgebracht hebben achter de microscoop en dank voor je waardevolle bijdrage aan een aantal papers in dit proefschrift en je immer constructieve opmerkingen.

Page 280: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

271

In dit kader uiteraard ook heel veel dank aan Warner Prevoo, Wouter Vogel, Henk Boot, Harm van Tinteren en Arend Aalbers. Lieve Pieter, dank aan jou en Marlene voor jullie vriendschap en (erg muzikale!) luisterend oor.

En dan zijn er die vele goeie vrienden waarmee ik gezegend ben… In Leuven Matthi, bedankt voor je eindeloos geduldige aanwezigheid, met name op momenten dat de weg moeilijk is. Je hebt mij zo vaak opnieuw en opnieuw gemotiveerd om ondanks alles toch door te zetten. Je nuchtere kijk, inschattingsvermogen en fenomenale zin voor humor hebben mij meer dan eens “on track” gehouden. I couldn’t have done this without you! Je bent een ongelooflijke levenskunstenaar, die er mij steeds weer aan herinnert dat ik tussendoor niet mag vergeten het leven ten volle te leven. Liebe Christine, ich brauche dir nicht zu erklären wie wichtig du für mich bist. Ich danke dir, für dein aufrechtes Interesse daran, wie es mir wirklich geht, für deine zuverlässige Anwesenheit, deine Wärme und Freundschaft, deinen super-Kaffee, deine unvergleichliche Pasta à la matricana und unsere vielen, vielen langen Gespräche bis tief in die Nacht. Ich habe dich sehr lieb! Kuβ! Lia, bedankt voor wat je mij het afgelopen jaar aan begeleiding, coaching en perspectief hebt gegeven. Goed kunnen luisteren en vervolgens de juiste vragen stellen, is een talent dat aan niet vele mensen gegeven is. My life will never be the same. Lieve Joyce, Willem en Alexandre en lieve Tom, Katrien, Sophie en Anna, bedankt dat jullie mij als ‘part of the family’ deel laten hebben aan jullie leven. Lieve Myriam en Laurent-Jan, Katarina, Cathelijne, Birgit en Katrien, bedankt voor jullie vriendschap en interesse.

Page 281: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

272

en in Amsterdam: Lieve Houke, de laatste van mijn stellingen is niet voor niks aan jou gewijd:

“Het komt allemaal goed…” “Ja… , maar…” “Nee, luister nou! Het komt écht allemaal goed… ”

Dat is een gesprek dat we inderdaad ongeveer zo’n duizend keer over-and-over-again gevoerd hebben, bij de zoveelste kop capuccino in jouw kamer of in de nachtelijke uurtjes bij een glas wijn bij jou thuis aan de keukentafel. Je zei eens tegen mij: “ Je weet toch waar dit over gaat, hè?... Volhouden!” Bedankt voor je steun, je kritische blik, je sterke gevoel voor wat écht belangrijk is in het leven en je eindeloze geduld. Jij hebt mij hierdoorheen gedragen in Amsterdam, dus ik ben erg trots dat je mijn paranimf wil zijn! Dikke kus. Dear Patricia, I don’t think I need to explain much. Thanks for always being there for me, for inviting me into your life and home and for caring so deeply. I Love you very much! Lieve Carolien, uren samen koken… en eten!, uren samen kletsen, over het leven, de liefde, de mannen, het werk, kunst, literatuur, architectuur, … uren samen batterijtjes opladen dus! Thanks! Dikke kus! Lieve Jan, bedankt voor de vele gezellige Vlaams-Nederlandse culinaire momenten, de vele filosofische gesprekken over het leven, de liefde en de kunst (niet van IKEA!) en je verwoede pogingen om er mij aan te herinneren dat er nog iets anders bestaat in het leven dan opereren en promoveren. Misschien leek het op het eerste gezicht niet zo, maar ik heb het allemaal heel goed gehoord én ook begrepen. Kus. Mijn dierbare AVL-flexplek-buddies, Boj en Danny, thanks for letting me pick your brains every once in a while en bedankt voor alle vriendschap en de gezelligheid tijdens onze vele koffietjes en kooksessies.

Page 282: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

273

En aan mijn vele andere Amsterdamse “familieleden”: Feikje, Rachel, Judit, Deborah, Cindy, Glynis, Frouwe, Monique, Miranda, Annemieke, Michi, Patricia, Marjolein, Rogier, Koen, Tineke, … Bedankt voor jullie gezelligheid, steun en vriendschap!

And last but not least: Aan mijn inmiddels alweer vertrouwde nieuwe ONC/AMO/RTO/KNC/ABD/URO/THO/GNC/DIO-familie in het UZ Leuven: bedankt aan iedereen die mij in mijn startjaar met heel veel geduld en enthousiasme gesteund heeft. Heel, heel bijzondere dank aan jou, Rose: jij hebt mij echt gedragen het afgelopen jaar.

Tot slot nog dit: Toon Hermans schreef:

Ik dacht Telkens als ik iets verzon Ik ben het water Niet de bron

Dat behoeft, geloof ik, geen verdere uitleg. Daphne

Page 283: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

274

Page 284: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

275

Curriculum Vitae

Page 285: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

276

Page 286: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

277

Daphne Hompes werd op 26-06-1976 geboren te Leuven, waar ze ook opgroeide en school liep. Daarna studeerde ze aan de Faculteit Geneeskunde van de Katholieke Universiteit Leuven en studeerde er in juni 2001 af met grote onderscheiding. Vervolgens werd dokter Hompes opgeleid tot algemeen chirurg, waarna ze aansluitend 2 jaren resident was op de afdeling Abdominale Heelkunde van het UZ Gasthuisberg te Leuven om een bijzondere bekwaming in de Abdominale Chirurgie te behalen. Van augustus 2009 tot en met februari 2012 werkte dokter Hompes in het Nederlands Kanker-Instituut / Antoni van Leeuwenhoek-ziekenhuis (NKI-AVL) te Amsterdam als research-fellow in de Oncologische Chirurgie. Ze was er gedurende 2 jaren halftijds klinisch actief, met als bijzondere focus cytoreductieve chirurgie & HIPEC, alsook leverchirurgie voor gemetastaseerde colorectale kanker. In ditzelfde aandachtsgebied deed ze ook halftijds clinical research met het oog op dit proefschrift. In de laatste 6 maanden die dokter Hompes in het NKI-AVL doorbracht, lag de klinische klemtoon op sarcoma-pathologie en –chirurgie. Sinds maart 2012 is dokter Hompes werkzaam als staflid Oncologische Heelkunde in het Universitaire Ziekenhuis Gasthuisberg te Leuven, met als specifieke aandachtspunten sarcomachirurgie, pediatrische oncochirurgie, cytoreductieve chirurgie en HIPEC.

Page 287: UvA-DARE (Digital Academic Repository) Advanced colorectal ... · in patients with peritoneal carcinomatosis from colorectal cancer. Cancer Treat Res 2007; 134: 425-440. 9. Jayne

278