Editorial Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Oct 7, 2011; 17(37): 4153-4156
Published online Oct 7, 2011. doi: 10.3748/wjg.v17.i37.4153
Selection criteria for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in gastric cancer
Ingmar Königsrainer, Department of General, Visceral and Transplant Surgery, Tübingen University Hospital, 72076 Tübingen, Germany
Author contributions: Königsrainer I was the sole contributor to this paper.
Correspondence to: Ingmar Königsrainer, MD, Department of General, Visceral and Transplant Surgery, Tübingen University Hospital, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany. ingmar.koenigsrainer@med.uni-tuebingen.de
Telephone: +49-7071-2985073 Fax: +49-7071-295588
Received: January 24, 2011
Revised: June 9, 2011
Accepted: June 16, 2011
Published online: October 7, 2011

Abstract

Peritoneal carcinomatosis in gastric cancer is associated with a dismal prognosis. Systemic chemotherapy is not effective because of the existence of a blood-peritoneal barrier. Cytoreductive surgery and intraperitoneal chemotherapy can improve survival and quality of life in selected patients. Patient selection for this multimodal approach is one of the most critical issues, and calls for interdisciplinary evaluation by radiologists, medical and surgical oncologists, and anaesthetists. This article sets forth criteria for selection of gastric cancer patients suffering from peritoneal carcinomatosis.

Key Words: Peritoneal carcinomatosis, Gastric cancer, Hyperthermic intraperitoneal chemotherapy, Cytoreductive surgery, Selection criteria



INTRODUCTION

Gastric cancer is one of the most frequent causes of cancer-related mortality worldwide[1,2]. Surgical resection after neoadjuvant chemotherapy in primary locally extended cases remains the mainstay for treating patients suffering from this disease. Surgery is limited by various factors: impaired general status, severe concomitant diseases, and distant metastases. One form is peritoneal dissemination of cancer cells within the abdominal cavity. Peritoneal carcinomatosis (PC) is detected in more than 30% of patients with advanced gastric cancer, and almost 60% of deaths are caused by peritoneal dissemination. In contrast to lymphatic and hematogenous metastasis, peritoneal carcinomatosis can be considered a local disease limited to the peritoneal cavity. Based on this rationale, cytoreductive surgery and intraabdominal chemotherapy have become a relevant treatment option for patients.

Various intraabdominal chemotherapy protocols have been established, varying from hyperthermic intraperitoneal chemotherapy (HIPEC), to early postoperative intraperitoneal chemotherapy, normothermic intraperitoneal chemotherapy, and delayed postoperative intraperitoneal chemotherapy[3]. They differ in the heat of the administered agent, the chemotherapy dosage, and time of administration of the chemotherapy. HIPEC seems to have the most beneficial impact on overall survival[3]. Retrospective analyses of patients treated with cytoreductive surgery plus HIPEC show a clear survival benefit when complete cytoreduction was possible. The randomized trial by Fujimoto et al[4] in 141 gastric cancer patients, who were curatively resected, showed a significantly reduced peritoneal recurrence rate and improved long-term survival when HIPEC was part of the treatment as compared to the “surgery alone” group. This observation was confirmed by Kim et al[5], who showed a significantly lower peritoneal recurrence rate and an improvement in the five-year survival rate in the multimodally treated patients as compared to the “surgery alone” group.

Systemic chemotherapy is not as effective as surgery plus HIPEC because of the blood-peritoneal barrier[6].

For metachronous peritoneal carcinomatosis from gastric cancer, there is no evidence to show which patient should be treated with the presented multimodal strategy. The dilemma arises when patients are very young and systemic chemotherapy is ineffective. In such cases, an individual approach with maximal tumor debulking may be an option and is justified in highly selected patients. However, in metachronous peritoneal carcinomatosis, the tumor often extensively involves the abdominal cavity with infiltration of the retroperitoneum, liver hilus, etc., which makes surgery impossible.

The aim of this article is to summarize the recent knowledge on patient selection for cytoreductive surgery and perioperative intraperitoneal chemotherapy for primary gastric cancer with peritoneal carcinomatosis or positive cytology.

GENERAL STATUS

Patients with limited peritoneal carcinomatosis from gastric cancer do not suffer from symptoms such as dysphagia or dysmotility, and PC is frequently found even in low T and negative N stages. Therefore, if the general status is acceptable, the option of radical surgical treatment should always be considered.

A detailed preoperative anesthesiological check-up is of importance and all patients should undergo a preoperative lung and cardiac function test. Concomitant diseases, that may influence surgical and anesthesiological risks, should be identified. As for most general elective operations, a low ASA score is mandatory. Age is still a matter of concern, because biological age does not always correlate with numerical age. However, most groups dealing with cytoreductive surgery and HIPEC restrict patient selection to an age below 65. Most importantly, informed consent with discussion of all alternative therapies must be obtained from the patients, and patients should be offered psychooncological support.

PREOPERATIVE DIAGNOSTICS

A high-end computed tomography is actually the standard and should be performed also to exclude extraabdominal spread and liver metastases. Recently, we demonstrated that positron emission tomography computed tomography(PET-CT) correlates well with intraoperative tumor load in peritoneal carcinomatosis[7]. However, gastric cancer with PC is frequently of mucinous character; therefore, PET-CT is not helpful in selecting candidates for radical resection. Nodules smaller than 5-8 mm cannot be consistently detected[8,9]. In particular, nodules on the small bowel and its mesentery are difficult to diagnose, but relevant for indication.

To date, there is no imaging method that can sufficiently predict intraoperative tumor load. Therefore, explorative laparoscopy is an invasive alternative for candidates in whom radiological work-up was not sufficient to determine operability.

Laparoscopy permits determination of the peritoneal carcinomatosis index (PCI) and cytology in locally advanced cases. Laparoscopy is highly accurate for the diagnosis of peritoneal carcinomatosis, with good correlation to the open surgical exploration found by Yonemura et al[9].

Therefore, every patient should undergo explorative laparoscopy before neoadjuvant therapy or primary gastrectomy. As the first step of treatment, some groups even administer HIPEC via laparoscopy in patients with synchronous PC or positive cytology[10,11].

PCI

PCI describes the tumor load in the abdomen and varies from 0 to 39, depending on the compartments involved[12].

In contrast to colorectal cancer, where the PCI should be lower than 20 so that patients potentially profit in terms of overall survival[13], in gastric cancer, the PCI should be much lower, because the biological behaviour of the tumor is more aggressive.

In a recent work by Yonemura et al[14], complete cytoreduction was achieved in 91% of the patients when the PCI was lower than 6, but in only 42% of the patients with a PCI ≤ 7. Overall survival was also better in the PCI ≤ 6 group. In gastric cancer with peritoneal carcinomatosis, lymph nodes should be removed only if they are infiltrated; however, prophylactic D2 lymphadenectomy is unnecessary.

Patients with liver metastasis, involving para-aortic lymph nodes and extraabdominal metastases, are not candidates for cytoreductive surgery and HIPEC. The treatment of metachronous metastases remains controversial, and cancer masses tend to infiltrate the retroperitoneum and liver hilus with vascular structures, which makes surgery impossible.

COMPLETENESS OF CYTOREDUCTION SCORE

The completeness of cytoreduction score CC score describes the completeness of cytoreduction after operation. Ideally, all tumor nodules can be removed macroscopically (CC0). Otherwise, a CC1, CC2, or CC3 score describes non-resectable tumor nodules that vary in size and influence on prognosis. Non-resectability is caused either by diffuse peritoneal carcinomatosis with a high PCI, where surgical resection is oncologically not justified, or by diffuse infiltration of the small bowel or the mesenterial axis, and infiltration of the retroperitoneum.

NEW PROTOCOLS

Neoadjuvant intraperitoneal systemic chemotherapy protocol (NIPS) is a newly developed neoadjuvant intraperitoneal treatment modality developed by Yonemura et al[15]. A good predictor of the possibility of achieving a CC0 status is preoperative cytology. Yonemura et al[16] performed NIPS and achieved CC0 status in 27 of 52 patients with negative cytology, but only in four of 27 with positive cytology. Peritoneal wash cytology may, therefore, be a good predictor of the potential for CC0 status.

SUMMARY AND FUTURE PERSPECTIVES

Gastric cancer with peritoneal carcinomatosis is a biologically aggressive tumor, and surgery is still the gold standard of treatment if abdominal spread is limited and PCI is low, ideally < 10. HIPEC may have a potential impact on remaining free cancer cells, although it has not been proven in randomized trials. In metachronous peritoneal carcinomatosis, the surgical approach is often limited by the extensive intraabdominal tumor load and by the aggressive biological behaviour of the tumor itself. NIPS is a promising therapy, and may improve resectability and survival. Intense research is currently being done in experimental peritoneal carcinomatosis, which will eventually modulate current indications.

Concerning promising biomarkers, Phosphoglycerate-kinase 1 (PGK1), an adenosine-triphosphate (ATP)-generating enzyme of the glycolytic pathway, which also affects DNA replication and repair, seems to be an interesting enzyme that is significantly involved in the pathogenesis of gastric cancer and PC[17-19]. Concerning tumorigenesis, it is assumed that genes involved in the glycolytic pathway are responsible for providing solid tumor cells with ATP. A newly discovered link between metabolic changes, including PGK1, and differentiation, has intriguing connections to an old hypothesis advocated by Otto Warburg for tumor metabolism. Further, recent in vitro and in vivo studies showed that PGK1 overexpression is associated with an elevated tumor invasion and metastatic rate in gastric cancer[17-19]. Those results demonstrate that PGK1 might be a crucial enzyme enabling cancer cells to metastasize, and, therefore, may serve as a target molecule for therapy in gastric cancer in the near future.

CONCLUSION

Nowadays, a radical combined treatment should be considered for a motivated patient with good performance status and low-grade peritoneal carcinomatosis. In addition, the patient should be sent to a peritoneal surface malignancy center.

Footnotes

Peer reviewer: Kevin Michael Reavis, MD, Assistant Clinic Professor, Department of Surgery, Division of Gastrointestinal Surgery, University of California, Irvine Medical Center, 333 City Boulevard West, Suite 850, Orange, CA, United States

S- Editor Sun H L- Editor Stewart GJ E- Editor Xiong L

References
1.  Kelley JR, Duggan JM. Gastric cancer epidemiology and risk factors. J Clin Epidemiol. 2003;56:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Parkin DM, Pisani P, Ferlay J. Global cancer statistics. CA Cancer J Clin. 1999;49:33-64, 1.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Yan TD, Black D, Sugarbaker PH, Zhu J, Yonemura Y, Petrou G, Morris DL. A systematic review and meta-analysis of the randomized controlled trials on adjuvant intraperitoneal chemotherapy for resectable gastric cancer. Ann Surg Oncol. 2007;14:2702-2713.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Fujimoto S, Takahashi M, Mutou T, Kobayashi K, Toyosawa T. Successful intraperitoneal hyperthermic chemoperfusion for the prevention of postoperative peritoneal recurrence in patients with advanced gastric carcinoma. Cancer. 1999;85:529-534.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Kim JY, Bae HS. A controlled clinical study of serosa-invasive gastric carcinoma patients who underwent surgery plus intraperitoneal hyperthermo-chemo-perfusion (IHCP). Gastric Cancer. 2001;4:27-33.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, Rodrigues A, Fodor M, Chao Y, Voznyi E. Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. J Clin Oncol. 2006;24:4991-4997.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Pfannenberg C, Königsrainer I, Aschoff P, Oksüz MO, Zieker D, Beckert S, Symons S, Nieselt K, Glatzle J, Weyhern CV. (18)F-FDG-PET/CT to select patients with peritoneal carcinomatosis for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg Oncol. 2009;16:1295-1303.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Koh JL, Yan TD, Glenn D, Morris DL. Evaluation of preoperative computed tomography in estimating peritoneal cancer index in colorectal peritoneal carcinomatosis. Ann Surg Oncol. 2009;16:327-333.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Yonemura Y, Bando E, Kawamura T, Ito H, Endo Y, Miura M, Kiyosaki K, Sasaki T. Cytoreduction and intraperitoneal chemotherapy for carcinomatosis from gastric cancer. Cancer Treat Res. 2007;134:357-373.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Valle M, Garofalo A. Laparoscopic staging of peritoneal surface malignancies. Eur J Surg Oncol. 2006;32:625-627.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Facchiano E, Scaringi S, Kianmanesh R, Sabate JM, Castel B, Flamant Y, Coffin B, Msika S. Laparoscopic hyperthermic intraperitoneal chemotherapy (HIPEC) for the treatment of malignant ascites secondary to unresectable peritoneal carcinomatosis from advanced gastric cancer. Eur J Surg Oncol. 2008;34:154-158.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res. 1996;82:359-374.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Esquivel J, Sticca R, Sugarbaker P, Levine E, Yan TD, Alexander R, Baratti D, Bartlett D, Barone R, Barrios P. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in the management of peritoneal surface malignancies of colonic origin: a consensus statement. Society of Surgical Oncology. Ann Surg Oncol. 2007;14:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Yonemura Y, Elnemr A, Endou Y, Hirano M, Mizumoto A, Takao N, Ichinose M, Miura M, Li Y. Multidisciplinary therapy for treatment of patients with peritoneal carcinomatosis from gastric cancer. World J Gastrointest Oncol. 2010;2:85-97.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Yonemura Y, Bandou E, Kinoshita K, Kawamura T, Takahashi S, Endou Y, Sasaki T. Effective therapy for peritoneal dissemination in gastric cancer. Surg Oncol Clin N Am. 2003;12:635-648.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Yonemura Y, Endou Y, Shinbo M, Sasaki T, Hirano M, Mizumoto A, Matsuda T, Takao N, Ichinose M, Mizuno M. Safety and efficacy of bidirectional chemotherapy for treatment of patients with peritoneal dissemination from gastric cancer: Selection for cytoreductive surgery. J Surg Oncol. 2009;100:311-316.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Zieker D, Königsrainer I, Weinreich J, Beckert S, Glatzle J, Nieselt K, Bühler S, Löffler M, Gaedcke J, Northoff H. Phosphoglycerate kinase 1 promoting tumor progression and metastasis in gastric cancer - detected in a tumor mouse model using positron emission tomography/magnetic resonance imaging. Cell Physiol Biochem. 2010;26:147-154.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Zieker D, Königsrainer I, Tritschler I, Löffler M, Beckert S, Traub F, Nieselt K, Bühler S, Weller M, Gaedcke J. Phosphoglycerate kinase 1 a promoting enzyme for peritoneal dissemination in gastric cancer. Int J Cancer. 2010;126:1513-1520.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Zieker D, Königsrainer I, Traub F, Nieselt K, Knapp B, Schillinger C, Stirnkorb C, Fend F, Northoff H, Kupka S. PGK1 a potential marker for peritoneal dissemination in gastric cancer. Cell Physiol Biochem. 2008;21:429-436.  [PubMed]  [DOI]  [Cited in This Article: ]