You are on page 1of 20

PERITONEAL SPREAD OF COLORECTAL CANCER: PREVENTION AND

TREATMENT

WP Ceelen, MD, PhD

Department of Gastrointestinal Surgery

Ghent University Hospital


De Pintelaan 185, B-9000 Ghent, Belgium

Wim.ceelen@ugent.be

Introduction

With an annual worldwide mortality rate of over half a million, colorectal cancer
(CRC) remains a major cause of cancer related mortality.1 Since malignant disease
ultimately causes death by distant organ invasion, the unravelling of molecular
mechanisms underlying hematogenous and lymphatic metastasis is a topic of
intensive research activity.2 In parallel, the introduction of targeted biological agents
has met with considerable survival prolongation in patients with metastatic disease.3
On the other hand, intraperitoneally located tumors may be at the origin of
locoregional peritoneal spread. Although often coexisting with systemic disease, it is
increasingly realised that colorectal tumor dissemination within the peritoneal cavity
may represent a separate phenotypic and molecular entity. Established carcinomatosis
from CRC is much less responsive to systemic therapy and causes considerable
morbidity in affected patients. Recognition of the causes and mechanisms of
peritoneal metastasis may contribute to strategies to effectively prevent the
development of PC in colorectal cancer. Moreover, in a small group of patients with
low volume peritoneal disease, a locoregional treatment strategy combining surgery
with intracavitary cytotoxic therapy has been shown to improve outcome.4

Incidence and Prognostic Significance of Peritoneal Tumor Spread from


Colorectal Cancer

Synchronous peritoneal metastases are found at the time of surgery with curative
intent in about five to six percent of patients, and are more frequently observed in
right sided cancers.5 Peritoneal carcinomatosis is present in 25-30% of patients with
recurrent or metastatic colorectal cancer; in approximately 3% isolated peritoneal
disease without systemic spread is observed.6, 7 The prognostic outlook of patients
with palliatively managed PC from colorectal malignancy is grim: the French
multicenter EVOCAPE 1 study found a median survival of 5.2 months.8 Systemic
chemotherapy improves survival in metastatic colorectal cancer, but the presence of
PC has been shown to be an adverse determinant of response and prognosis in patients
treated with fluorouracil or irinotecan based systemic chemotherapy.9-11 Contrary to
those with visceral metastasis, patients with PC are at risk of develop debilitating
symptoms such as obstruction and ascites formation, while the risk of perforation
induced by the VEGF inhibitor bevacizumab may be more pronounced.12
Although established peritoneal metastases are found in only a limited percentage of
patients undergoing surgery, it is likely that microscopic spread occurs much more
frequently.13 Several authors have studied the presence of free peritoneal cancer cells
immediately before or after colorectal cancer resection, and related their presence to
local recurrence and survival (Table 1). Overall, the results of these studies suggest
that microscopic peritoneal disease at the time of surgery represents a prognostic
factor and thus a possible therapeutic target in resectable CRC. A similar conclusion
was formulated by the authors of a recent meta-analysis showing that the presence of
free peritoneal cancer cells pre CRC resection was associated with a higher risk of
overall recurrence (Odds Ratio 0.19–0.88) and local recurrence (Odds Ratio 0.21–
0.82) while post resection, the presence of free cancer cells resulted in a significantly
higher risk of overall recurrence (Odds Ratio 0.03–0.18).14 In parallel, in vitro studies
and animal tumorigenicity assays have shown that exfoliated CRC cells have the
potential to proliferate and invade.15, 16
In many CRC patients, the occurrence of carcinomatosis is a late manifestation of
disseminated cancer. There is, however, a subset of patients in whom peritoneal
disease progression appears without any systemic spread. The prototypic syndrome in
this regard is pseudomyxoma peritonei (PMP), which was shown to originate from
mucinous neoplasms of the appendix and whose clinical course is characterized by
progressive panabdominal mucinous carcinomatosis without any systemic metastasis.
Bibi and coworkers recently demonstrated that a specific expression profile of
cadherins underlies the intriguing behaviour of PMP.17 Compared to adenocarcinoma
of the colon, PMP samples showed increased N-cadherin, reduced E-cadherin, and
increased vimentin expression, suggesting that the propensity for peritoneal spread is
caused by an epithelial to mesenchymal transition (EMT) state. Similarly, differences
in gene expression profile have been described between systemic (hepatic) and
isolated peritoneal metastases.18, 19 In a preliminary analysis, we compared gene
expression between CRC liver metastases and isolated peritoneal metastases, and
found 179 genes related to immune response, cellular differentiation, (EMT), and cell
growth to be differentially expressed.20 Pathway analysis showed that IL-6 and TGF-β
signalling were upregulated in peritoneal metastases. Taken together, these results
suggest that in some patients, peritoneal progression of CRC is driven by a specific
set of genetic alterations.

Mechanisms of Peritoneal Tumor Spread of Colorectal Cancer

The development of peritoneal carcinomatosis can be viewed as several well defined


steps.

1. Detachment of cells from the primary tumor

The first step in the cascade resulting in peritoneal carcinomatosis (PC) is liberation
of tumour cells from the primary cancer mass (Fig). This process can occur
spontaneously, or can be iatrogenically caused. First, downregulation of cell-cell
adhesion molecules, such as E-cadherin via the transcription factor TWIST, has been
reported to promote cancer cell detachment21, 22 Second, spontaneous shedding of
loose cells is facilitated by the elevated interstitial fluid pressure (IFP) in most solid
tumours.23 This hydrodynamic property of malignant tissue is caused by rapid cellular
proliferation, defective lymphatic drainage, fibrosis and contraction of the interstitial
matrix, and increased osmotic pressure generated by anaerobic glycolysis and leakage
of plasma proteins.24, 25 Several clinical studies showed a significantly poorer outcome
in CRC patients in whom visceral peritoneal invasion was identified (pT4a) on
pathological examination of the resection specimen (Tabel 2). Similarly, spontaneous
bowel perforation has been shown to represent an adverse prognostic event and to
more than double the risk of postoperative carcinomatosis compared to non perforated
cancers.26-29
Third, cancer cell spillage by inadvertent cutting into tumor tissue or by sectioning
draining blood, lymphatic, or biliary channels has been shown to promote
locoregional tumor dissemination.30, 31

2. Peritoneal transport

Free cancer cells in the peritoneal cavity are subject to passive movement dictated by
gravity and by the excursion of the diaphragm. As a result, a predictable path is
usually followed towards the pelvis and from the pelvis, along the right paracolic
gutter, towards the subdiaphragmatic space.32 Cancer cells also possess active motility
provided by lamellipodia and filipodia, whose mechanical force is generated by
polymerization of actin microfilaments.33

3. Mesothelial adhesion

Free peritoneal cancer cells adhere to either the mesothelial lining or to the underlying
extracellular matrix (ECM) through specific adhesion molecules. Mesothelial cells
express VCAM-1, ICAM-1, and PECAM-1, but not ICAM-2 or E-selectin.34, 35 Ziprin
et al. showed that in vitro, colorectal tumour - mesothelial adhesion was mediated by
the interaction of mesothelial ICAM-1 and CD43 (sialophorin) rather than β2 integrin,
the most ubiquitous ligand of ICAM-1.36 Adhesion between tumor cells and ECM
components seems to be mediated primarily by the beta 1 integrin subunit.37
Interestingly, the major omentum is a preferential site of peritoneal tumor growth. The
mechanisms underlying this specificity are not fully elucidated. It has been suggested
that cancer growth is stimulated by the pro-angiogenic environment of the omental
‘milky spots’, which consist of immune aggregates and a dense capillary network.38
Sorensen et al. showed that tumor cell binding may be mediated by a network of
collagen I fibers overlaying the milky spots, and that omental microvessels express
the pro-angiogenic vascular endothelial growth factor receptor 3 (VEGFR3).39
The expression of mesothelial adhesion molecules (and the resulting cancer cell
adhesion) may be considerably enhanced by inflammatory stimuli induced by
infection or surgical trauma.40 Thus, mesothelial expression of ICAM-1 is stimulated
by proinflammatory cytokines such as TNF-α, IL-1β, IL-6, and the epidermal growth
factor.41 Also, malignant cells may become entrapped in exudated fibrin matrices, and
exudated plasma proteins such as fibronectin and vitronectin may act as bridging
molecules between endothelial cells, smooth muscle cells and cancer cells via the α5β1
and αvβ3 receptors.42 Alterations in mesothelial binding may also be caused by
mechanical factors. In vitro, elevation of the ambient pressure (for instance by
establishing a pneumoperitoneum) increases adhesion of colon cancer cells to matrix
proteins.43 Also, elevated intraperitoneal pressures cause contraction of mesothelial
cells resulting of increased exposure of ECM binding sites.44 Apart from the
mechanical effects on mesothelial structure, the acidification and dehydration
associated with CO2 gas inflation during laparoscopic surgery were shown to promote
tumour growth and invasiveness in a number of preclinical studies.45, 46 Despite the
concerns raised by these preclinical models, the incidence of port site metastases has
been noted to decrease with appropriate protective measures such as trocar fixation,
adequate wound protection, and avoidance of desufflation through a skin incision. In
colorectal cancer, the results of recently completed large randomized trials comparing
open with laparoscopic colectomy demonstrated that the rate of wound recurrence is
low (<1%) and does not differ between the open and laparoscopic technique.47, 48

4. Invasion of the submesothelial tissue

Loose tumour cells gain access to submesothelial tissue at areas of peritoneal


discontinuity or mesothelial cell contraction. Alternatively, tumour cells can induce
apoptosis of mesothelial cells. Heath et al. demonstrated FAS dependent apoptosis of
cultured human mesothelial cells induced by SW480 colorectal tumour cells.49 Once
the mesothelial barrier is breached, tumor cells degrade the underlying matrix by
secretion of several proteases such as the matrix metalloproteinases (MMP).50, 51

5. Systemic metastasis

Peritoneal cancer cells can gain access to the lymphatic circulation through
specialized lymphatic stomata, which are localized mainly on the diaphragmatic
surface, falciform ligament of the liver, and pelvic side wall.52 These stomata are 8-10
µm2 round to oval gaps between cuboidal mesothelial cells and communicate directly
with the lumen of a lymphatic vessel or lacuna.53 In a rabbit model, passage from
cancer cells from the peritoneal cavity via the stomata into the lymphatic cisterna was
demonstrated.54 Importantly, both the density and diameter of the stomata and thus the
peritoneal absorptive capacity may increase by raised intraperitoneal pressure or by
molecular mediators such as VEGF and nitric oxide (NO).55

Prevention of Peritoneal Colorectal Metastasis

1. Reduction of Surgical Trauma

Surgical trauma may be prevented by physical measures such as the use of atraumatic
gauze or non powdered gloves.56, 57 Compared to open surgery, the use of laparoscopic
techniques minimizes peritoneal trauma and reduced tumour growth compared to
open surgery in several animal models, a finding usually attributed to a difference in
postoperative immune competence.58-60 Sylla et al. studied splenic T cell gene
expression following laparotomy versus CO2 pneumoperitoneum in a mouse model
and found that 177 genes were increased and 15 decreased at least 2-fold after
laparotomy relative to pneumoperitoneum.61

2. Peritoneal irrigation
Exfoliated cancer cells may be eradicated by irrigation of the peritoneal cavity, either
mechanically or by using a tumoricidal solution. There is some evidence from
preclinical studies that instillation of povidone-iodine (PVD) is beneficial.62 Basha et
al. showed that instillation of Povidone-iodine was effective in preventing tumour
take in a rat model when a limited tumour inoculum was used.63 In an animal model of
laparoscopy assisted tumour splenectomy, abdominal irrigation with dilute PVD
significantly reduced the number of animals with peritoneal implant metastases.64
Many of the commonly used antiseptics, however, are known to be inactivated by the
presence of blood.65 Huguet et al. used distilled water to achieve osmotic lysis of
cancer cells.66 Complete lysis in vivo took significantly longer (more than 30 minutes)
compared to in vitro instillation. Clinical studies using these approaches have not
been reported, and potential toxicity of intraperitoneal PVD should be kept in mind.67

3. Intraperitoneal Chemotherapy

In animal models, intraperitoneal instillation of chemotherapy was shown to be ef-


fective in preventing cancer cell implantation.68, 69 Nordlinger et al. reported a
randomized trial in which 753 patients with stage II or III colorectal cancer were
assigned to adjuvant systemic chemotherapy alone or immediate postoperative
regional chemotherapy (5-fluorouracil intraperitoneal or intraportal according to
treatment center) followed by systemic chemotherapy.70 No differences were observed
in overall survival or disease free survival, although the incidence of peritoneal
recurrence is not reported. Since intraperitoneal chemotherapy was administered
postoperatively, one may assume that postoperative adhesions prevented uniform
access of the instilled drug to the peritoneal cavity. This obstacle may be overcome by
intraoperative administration of chemotherapy, usually as a component of
hyperthermic intraoperative chemoperfusion (HIPEC), a strategy shown to benefit
colorectal cancer patients with resectable peritoneal carcinomatosis.4, 71, 72 A similar
approach consisting of ‘prophylactic’ HIPEC could reduce the risk of peritoneal
recurrence in high risk CRC patients. This hypothesis was provided initial
confirmation Elias et al., who performed second look surgery in 29 asymptomatic
CRC patients at high risk of peritoneal recurrence (resected minimal peritoneal
implants, synchronous ovarian metastases, or perforated tumours).73 Peritoneal
carcinomatosis undetected on imaging studies was present in 16 (55%) patients and
treated with surgery followed by intraperitoneal hyperthermic chemoperfusion,
resulting in a disease free status in half of the patients after a median follow up of 27
months. No signs of carcinomatosis were found in the remaining 13 patients, six of
whom underwent ‘prophylactic’ HIPEC. Interestingly, none of these six patients
recurred intraperitoneally while three out of seven patients who did not undergo the
HIPEC procedure did recur in the peritoneal cavity. These preliminary findings
formed the rationale of an ongoing randomised trial by the US National Cancer
Institute (ClinicalTrials.gov ID: NCT01095523).

4. Inhibition of Postoperative Growth of Peritoneal Residual Cancer Cells

It has been realised that the mechanisms involved in postoperative wound healing
may stimulate the growth of residual cancer cells.74 One of the essential features of
both wound healing and tumor growth is angiogenesis. Therefore, postoperative
inhibition of angiogenesis may prevent the outgrowth of peritoneal residual cancer.
Understandably, however, inhibition of VEGF in the postoperative setting carries the
risk of anastomotic and wound healing complications.75 Thus, in the National Surgical
Adjuvant Breast and Bowel Project (NSABP) C-08 trial which compared adjuvant
chemotherapy (FOLFOX6) with or without bevacizumab in stage II or III CRC, the
proportion of wound complications (incisional hernia, wound dehiscence, and port-
site dehiscence) was significantly higher in patients who received bevacizumab (1.7%
versus 0.3%, P < 0.01).76
Alternatively, one may avoid or suppress the inflammatory response after surgery. In
a mouse model, Roh et al. found that the selective cyclooxygenase-2 (Cox-2) inhibitor
celecoxib had a significant inhibitory effect on tumour growth in the surgical wound
when administered daily from 1 day before surgical wounding and tumour
implantation.77, 78 Also, restauration of surgery induced immunosuppression may
prevent postoperative locoregional cancer growth.40, 79 Small clinical trials in CRC
patients have shown that perioperative systemic administration using IL-2, GM-CSF,
or interferon restores postoperative immune function.80-82 However, their effect on
postoperative recurrence or survival is not known. In preclinical models,
intraperitoneal immunotherapy using cytokines, monoclonal antibodies, or
radionucleotide antibody conjugates has been successfully used to treat or prevent
peritoneal carcinomatosis.83

5. Inhibition of Adhesion of Free Intraperitoneal Cancer Cells

Specific therapy targeting the various mechanisms of tumour-mesothelial interaction


has been addressed in preclinical studies. One approach has been directed towards the
binding sites of the extracellular matrix (ECM). Alkhamesi et al. showed that
intraperitoneal application of heparin caused a significant decrease in tumour cell
adhesion accompanied by a decrease in ICAM-1 expression.84 Expression of ICAM-1
is also blocked in vitro by the grape polyphenol, resveratrol.85 Similarly, low
molecular weight heparin significantly reduced tumour growth following laparoscopy
in a rat colorectal cancer model.86 Heparins also inhibit tumour induced production of
thrombin, fibrin, and tissue factor, all of which have been implicated in primary and
metastatic tumour growth, and block P-and L-selectin mediated cell adhesion.87, 88
Alternatively, covering the ECM binding sites with a phospholipid emulsion reduced
tumour-mesothelial cell adhesion in animal models.89 Targeting of specific adhesion
molecules such as integrins90-92, L1 cell adhesion molecule93 and JAM-C94 with
monoclonal antibodies has shown to be effective in preventing tumour adhesion
and/or growth in preclinical models and may represent a future clinical therapeutic
tool.

Conclusion

Patients with locally advanced intraperitoneally located CRC are at risk of developing
peritoneal metastatic disease. It is increasingly acknowledged that peritoneal spread
represents a distinct disease manifestation, characterised by well defined stepwise
progression. The risk of peritoneal recurrence may be reduced by avoiding or
minimising surgical trauma, reducing the postoperative inflammatory response, and
by avoiding postoperative immunosuppression. Outgrowth of peritoneal cancer cells
may be abolished by intraperitoneal chemotherapy or tumoricidal solutions; clinical
trials are underway to establish the merit of these approaches.
Table 1. Incidence and prognostic significance of minimal residual disease in colorectal cancer

Author year N method markers or antibodies Detection rate Comments or Conclusion

Juhl95 1995 67 ICC C1P83 (CEA), Ra96, CA19-9 27% Positive ICC correlates with OS in univariate analysis
17-1A, C54-0, Kl-1
Hase96 1998 140 CYT 16% CYT positivity at the end of surgery predicts local recurrence

Schott97 1998 109 ICC C1P83 (CEA), CA19-9, 17-1A, 31% Positive ICC correlates with OS in univariate analysis
Ra96, C54-0, Kl-1
Wind98 1999 88 CYT 28% Presence of CYT+ cells provides no prognostic information

Vogel99 2000 90 CYT and ICC** HEA-125 47% Positive ICC does not predict local recurrence or outcome
100
Vogel 2001 135 ICC C1P83 (CEA), Ra96, CA19-9 23% Detection of ip tumor cells correlates with prognosis in univariate analysis
Worst prognosis in the presence of CEA positive intraperitoneal cells

Broll101 2001 75* ICC and RT-PCR CEA 63% CEA mRNA detection by RT-PCR not recommended due to high FP rate; presence
of ICC + cells represents independent prognostic factor in multivariate analysis

Lucha102 2002 56 CYT 2% Only 1 patient with + cytology

Aoki103 2002 20 RT-PCR CEA, CK20 24% Detection rate increased in parallel with invasion depth
104
Guller 2002 39 qRT-PCR CEA, CK20 28% Presence of PCR+ cells associated with worse DFS and OS in multivariate
analysis

Yamamoto105 2003 189 CYT 6% Presence of CYT+ cells predicted peritoneal recurrence and represents an independent
prognostic factor in multivariate analysis

Kanellos106 2003 110 CYT 20% Presence of CYT+ cells predicts locoregional recurrence but not survival

Bosch107 2003 53 CYT and ICC Ks20.8 (CK20), Ber-Ep4 17% Presence of CYT and/or ICC+ cells in blood and peritoneal lavage fluid
associated with DFS and OS in multivariate analysis

Lloyd108 2006 125 immunobead CK20, CEA, EphB4,LAMγ2, 29% Presence of PCR+ cells in post resection lavage fluid predicts DFS in multivariate ana-
RT-PCR matrilysin lysis (stage I and II cancers)

Kanellos109 2006 95 CYT and [CEA] 26% Presence of CYT+ cells and high peritoneal CEA level predicts recurrence but not OS

Gozalan110 2007 88 CYT 15% Presence of CYT + cells did not predict locoregional or systemic recurrence and does
not provide prognostic information

Hara111 2007 128 qRT-PCR CEA, CK20 23% Presence of PCR + cells is not an independent prognostic factor in multivariate
analysis and does not predict peritoneal recurrence

Kristensen112 2008 237 PCR for k-RAS mutation 8% Mutated k-RAS in peritoneal lavage samples following rectal cancer resection
Is associated with worse OS

Noura113 2009 697 CYT 2.2% Positive CYT is an independent prognosticator of cancer specific survival and
Peritoneal recurrence
Fujii114 2009 298 CYT 6% CYT status before resection does not affect survival or peritoneal recurrence

CYT, cytology; ICC, immunocytochemistry; RT-PCR, reverse transcriptase polymerase chain reaction; OS, overall survival; DFS, disease free survival; *includes 17 stomach cancer and 9 pancreas cancer cases;
** in a subset of 36 patients
Table 2. Incidence and prognostic significance of peritoneal involvement (pT4) of the resected colorectal cancer specimen

Author year N Stage II Stage III Detection rate Comments or Conclusion

Shepherd115 1997 412 42% 52% 27%* Peritoneal involvement significantly associated with OS in multivariate analysis; present in 45 of 46
patients who developed peritoneal recurrence

Solomon116 1997 103 47% 53% 14.6% Only the type of surgery predicted peritoneal involvement in univariate analysis; higher incidence in
rectal procedures

Lennon117 2003 118 100% - 13.6% Peritoneal involvement significantly associated with 5 year survival in multivariate analysis

Baskaranathan118 2004 281 63% 37% 9.3% Peritoneal involvement significantly associated with cancer specific survival in multivariate analysis

Keshava119 2007 665 49.9% 50.1% 5.3% Serosal involvement significantly associated with local recurrence and OS in multivariate analysis

Stewart120 2007 82 100% - 22% Serosal invasion associated with 5 year survival in univariate analysis
References

1. Herszenyi L, Tulassay Z. Epidemiology of gastrointestinal and liver tumors.


Eur Rev Med Pharmacol Sci;14(4): 249-258.
2. Royston D, Jackson DG. Mechanisms of lymphatic metastasis in human
colorectal adenocarcinoma. J Pathol 2009;217(5): 608-619.
3. Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B,
Starling N. Colorectal cancer. Lancet 2010;375(9719): 1030-1047.
4. Ceelen WP, Flessner MF. Intraperitoneal therapy for peritoneal tumors:
biophysics and clinical evidence. Nature Reviews Clinical Oncology 2010;7(2): 108-
115.
5. Benedix F, Kube R, Meyer F, Schmidt U, Gastinger I, Lippert H, Colon
Rectum Carcinomas Primary T. Comparison of 17,641 Patients With Right- and Left-
Sided Colon Cancer: Differences in Epidemiology, Perioperative Course, Histology,
and Survival. Diseases of the Colon & Rectum;53(1): 57-64.
6. Knorr C, Reingruber B, Meyer T, Hohenberger W, Stremmel C. Peritoneal
carcinomatosis of colorectal cancer: incidence, prognosis, and treatment modalities.
International Journal of Colorectal Disease 2004;19(3): 181-187.
7. Koppe MJ, Boerman OC, Oyen WJG, Bleichrodt RP. Peritoneal
carcinomatosis of colorectal origin - Incidence and current treatment strategies.
Annals of Surgery 2006;243(2): 212-222.
8. Sadeghi B, Arvieux C, Glehen O, Beaujard AC, Rivoire M, Baulieux J,
Fontaumard E, Brachet A, Caillot JL, Faure JL, Porcheron J, Peix JL, Francois Y,
Vignal J, Gilly FN. Peritoneal carcinomatosis from non-gynecologic malignancies -
Results of the EVOCAPE 1 multicentric prospective study. Cancer 2000;88(2): 358-
363.
9. Folprecht G, Lutz M, Rougier P, Van Cutsem E, Saltz L, Douillard J,
Cunningham D, Hecker H, C K. Effect of systemic chemotherapy (CTx) in patients
with known peritoneal carcinomatosis (PC) from colorectal cancer. In: 2007 ASCO
Gastrointestinal Cancers Symposium Abstract nr 294.
10. Kohne CH, Cunningham D, Di Costanzo F, Glimelius B, Blijham G, Aranda
E, Scheithauer W, Rougier P, Palmer M, Wils J, Baron B, Pignatti F, Schoffski P,
Micheel S, Hecker H. Clinical determinants of survival in patients with 5-
fluorouracil-based treatment for metastatic colorectal cancer: results of a multivariate
analysis of 3825 patients. Annals of Oncology 2002;13(2): 308-317.
11. Mitry E, Douillard JY, Van Cutsem E, Cunningham D, Magherini E, Mery-
Mignard D, Awad L, Rougier P. Predictive factors of survival in patients with
advanced colorectal cancer: an individual data analysis of 602 patients included in
irinotecan phase III trials. Annals of Oncology 2004;15(7): 1013-1017.
12. Saif MW, Elfiky A, Salem RR. Gastrointestinal perforation due to
bevacizumab in colorectal cancer. Annals of Surgical Oncology 2007;14(6): 1860-
1869.
13. Ceelen WP, Bracke ME. Peritoneal minimal residual disease in colorectal
cancer: mechanisms, prevention, and treatment. Lancet Oncology 2009;10(1): 72-79.
14. Rekhraj S, Aziz O, Prabhudesai S, Zacharakis E, Mohr F, Athanasiou T, Darzi
A, Ziprin P. Can intra-operative intraperitoneal free cancer cell detection techniques
identify patients at higher recurrence risk following curative colorectal cancer
resection: a meta-analysis. Ann Surg Oncol 2008;15(1): 60-68.
15. Fermor B, Umpleby HC, Lever JV, Symes MO, Williamson RCN.
Proliferative and Metastatic Potential of Exfoliated Colorectal-Cancer Cells. J Natl
Cancer Inst 1986;76(2): 347-349.
16. Skipper D, Cooper AJ, Marston JE, Taylor I. EXFOLIATED CELLS AND
INVITRO GROWTH IN COLORECTAL-CANCER. Br J Surg 1987;74(11): 1049-
1052.
17. Bibi R, Pranesh N, Saunders MP, Wilson MS, O'Dwyer ST, Stern PL,
Renehan AG. A specific cadherin phenotype may characterise the disseminating yet
non-metastatic behaviour of pseudomyxoma peritonei. Br J Cancer 2006;95(9): 1258-
1264.
18. Kleivi K, Lind GE, Diep CB, Meling GI, Brandal LT, Nesland JM, Myklebost
O, Rognum TO, Giercksky KE, Skotheim RI, Lothe RA. Gene expression profiles of
primary colorectal carcinomas, liver metastases, and carcinomatoses. Molecular
Cancer 2007;6: -.
19. Varghese S, Burness M, Xu H, Beresnev T, Pingpank J, Alexander HR. Site-
specific gene expression profiles and novel molecular prognostic factors in patients
with lower gastrointestinal adenocarcinoma diffusely metastatic to liver or
peritoneum. Ann Surg Oncol 2007;14: 3460-3471.
20. Debergh I, Van Damme N, Peeters M, Van Hummelen P, Pattyn P, Ceelen W.
Differential gene expression between metastatic colorectal tumours in liver and
peritoneum. Br J Surg 2008;95(S6): 13.
21. Terauchi M, Kajiyama H, Yamashita M, Kato M, Tsukamoto H, Umezu T,
Hosono S, Yamamoto E, Shibata K, Ino K, Nawa A, Nagasaka T, Kikkawa F.
Possible involvement of TWIST in enhanced peritoneal metastasis of epithelial
ovarian carcinoma. Clin Exp Metastasis 2007;24(5): 329-339.
22. Kokenyesi R, Murray KP, Benshushan A, Huntley ED, Kao MS. Invasion of
interstitial matrix by a novel cell line from primary peritoneal carcinosarcoma, and by
established ovarian carcinoma cell lines: role of cell-matrix adhesion molecules,
proteinases, and E-cadherin expression. Gynecol Oncol 2003;89(1): 60-72.
23. Hayashi K, Jiang P, Yamauchi K, Yamamoto N, Tsuchiya H, Tomita K,
Moossa AR, Bouvet M, Hoffman RM. Real-time Imaging of tumor-cell shedding and
trafficking in lymphatic channels. Cancer Res 2007;67(17): 8223-8228.
24. Rutz HP. Hydrodynamic consequences of glycolysis - Thermodynamic basis
and clinical relevance. Cancer Biology & Therapy 2004;3(9): 812-815.
25. Heldin CH, Rubin K, Pietras K, Ostman A. High interstitial fluid pressure -
An obstacle in cancer therapy. Nature Reviews Cancer 2004;4(10): 806-813.
26. McArdle CS, McMillan DC, Hole DJ. The impact of blood loss, obstruction
and perforation on survival in patients undergoing curative resection for colon cancer.
Br J Surg 2006;93(4): 483-488.
27. Komatsu S, Shimomatsuya T, Nakajima M, Amaya H, Kobuchi T, Shiraishi S,
Konishi S, Ono S, Maruhashi K. Prognostic factors and scoring system for survival in
colonic perforation. Hepatogastroenterology 2005;52(63): 761-764.
28. Chen HS, Sheen-Chen SM. Obstruction and perforation in colorectal
adenocarcinoma: An analysis of prognosis and current trends. Surgery 2000;127(4):
370-376.
29. Cheynel N, Cortet M, Lepage C, Ortega-Debalon P, Faivre J, Bouvier AM.
Incidence, Patterns of Failure, and Prognosis of Perforated Colorectal Cancers in a
Well-Defined Population. Diseases of the Colon & Rectum 2009;52(3): 406-411.
30. Hansen E, Wolff N, Knuechel R, Ruschoff J, Hofstaedter F, Taeger K. Tumor-
Cells in Blood Shed from the Surgical Field. Arch Surg 1995;130(4): 387-393.
31. Tanaka N, Nobori M, Suzuki Y. Does bile spillage during an operation present
a risk for peritoneal metastasis in bile duct carcinoma? Surgery Today-the Japanese
Journal of Surgery 1997;27(11): 1010-1014.
32. Meyers MA. Distribution of Intraabdominal Malignant Seeding - Dependency
on Dynamics of Flow of Ascitic Fluid. Am J Roentgenol 1973;119(1): 198-206.
33. Lindberg U, Karlsson R, Lassing I, Schutt CE, Hoglund AS. The
microfilament system and malignancy. Semin Cancer Biol 2008;18(1): 2-11.
34. Jonjic N, Peri G, Bernasconi S, Sciacca FL, Colotta F, Pelicci P, Lanfrancone
L, Mantovani A. EXPRESSION OF ADHESION MOLECULES AND
CHEMOTACTIC CYTOKINES IN CULTURED HUMAN MESOTHELIAL
CELLS. J Exp Med 1992;176(4): 1165-1174.
35. Bittinger F, Klein CL, Skarke C, Brochhausen C, Walgenbach S, Rohrig O,
Kohler H, Kirkpatrick CJ. PECAM-1 expression in human mesothelial cells: An in
vitro study. Pathobiology 1996;64(6): 320-327.
36. Ziprin P, Alkhamesi NA, Ridgway PF, Peck DH, Darzi AW. Tumour-
expressed CD43 (sialophorin) mediates tumour-mesothelial cell adhesion. Biol Chem
2004;385(8): 755-761.
37. Oosterling SJ, van der Bij GJ, Boegels M, ten Raa S, Post JA, Meijer GA,
Beelen RHJ, van Egmond M. Anti-beta 1 integrin antibody reduces surgery-induced
adhesion of colon carcinoma cells to traumatized peritoneal surfaces. Ann Surg
2008;247: 85-94.
38. Gerber SA, Rybalko VY, Bigelow CE, Lugade AA, Foster TH, Frelinger JG,
Lord EM. Preferential attachment of peritoneal tumor metastases to omental immune
aggregates and possible role of a unique vascular microenvironment in metastatic
survival and growth. Am J Pathol 2006;169(5): 1739-1752.
39. Sorensen EW, Gerber SA, Sedlacek AL, Rybalko VY, Chan WM, Lord EM.
Omental immune aggregates and tumor metastasis within the peritoneal cavity.
Immunol Res 2009;45(2-3): 185-194.
40. van der Bij GJ, Oosterling SJ, Beelen RHJ, Meijer S, Coffey JC, van Egmond
M. The Perioperative Period is an Underutilized Window of Therapeutic Opportunity
in Patients With Colorectal Cancer. Ann Surg 2009;249(5): 727-734.
41. Klein CL, Bittinger F, Skarke CC, Wagner M, Kohler H, Walgenbach S,
Kirkpatrick CJ. Effects of cytokines on the expression of cell adhesion molecules by
cultured human omental mesothelial cells. Pathobiology 1995;63(4): 204-212.
42. Ikari Y, Yee KO, Schwartz SM. Role of alpha 5 beta 1 and alpha v beta 3
integrins on smooth muscle cell spreading and migration in fibrin gels. Thromb
Haemost 2000;84(4): 701-705.
43. Basson MD, Yu CF, Herden-Kirchoff O, Ellermeier M, Sanders MA, Merrell
RC, Sumpio BE. Effects of increased ambient pressure on colon cancer cell adhesion.
J Cell Biochem 2000;78(1): 47-61.
44. Volz J, Koster S, Spacek Z, Paweletz N. Characteristic alterations of the
peritoneum after carbon dioxide pneumoperitoneum. Surgical Endoscopy-Ultrasound
and Interventional Techniques 1999;13(6): 611-614.
45. Ridgway PF, Smith A, Ziprin P, Jones TL, Paraskeva PA, Peck DH, Darzi
AW. Pneumoperitoneum augmented tumor invasiveness is abolished by matrix
metalloproteinase blockade. Surgical Endoscopy and Other Interventional
Techniques 2002;16(3): 533-536.
46. Jacobi CA, Sabat R, Bohm B, Zieren HU, Volk HD, Muller JM.
Pneumoperitoneum with carbon dioxide stimulates growth of malignant colonic cells.
Surgery 1997;121(1): 72-78.
47. Lacy AM, Garcia-Valdecasas JC, Delgado S, Castells A, Taura P, Pique JM,
Visa J. Laparoscopy-assisted colectomy versus open colectomy for treatment of non-
metastatic colon cancer: a randomised trial. Lancet 2002;359(9325): 2224-2229.
48. Nelson H, Sargent D, Wieand HS, Fleshman J, Anvari M, Stryker SJ, Beart
RW, Hellinger M, Flanagan R, Peters W, Ota D, Hellinger M. A comparison of
laparoscopically assisted and open colectomy for colon cancer. N Engl J Med
2004;350(20): 2050-2059.
49. Heath RM, Jayne DG, O'Leary R, Morrison EE, Guillou PJ. Tumour-induced
apoptosis in human mesothelial cells: a mechanism of peritoneal invasion by Fas
Ligand/Fas interaction. Br J Cancer 2004;90(7): 1437-1442.
50. Carreiras F, Rigot V, Cruet S, Andre F, Gauduchon P, Marvaldi J. Migration
properties of the human ovarian adenocarcinoma cell line IGROV1: Importance of
alpha v beta 3 integrins and vitronectin. International Journal of Cancer 1999;80(2):
285-294.
51. Lubbe WJ, Zuzga DS, Zhou ZY, Fu WL, Pelta-Heller J, Muschel RJ,
Waldman SA, Pitari GM. Guanylyl Cyclase C Prevents Colon Cancer Metastasis by
Regulating Tumor Epithelial Cell Matrix Metalloproteinase-9. Cancer Res
2009;69(8): 3529-3536.
52. Wang ZB, Li M, Li JC. Recent Advances in the Research of Lymphatic
Stomata. Anatomical Record-Advances in Integrative Anatomy and Evolutionary
Biology;293(5): 754-761.
53. Li JC, Yu SM. Study on the Ultrastructure of the Peritoneal Stomata in
Humans. Acta Anat (Basel) 1991;141(1): 26-30.
54. Namba Y. [An electron microscopic demonstration of the invasion of tumor
cells into the diaphragm]. Nippon Geka Gakkai Zasshi 1989;90(11): 1915-1921.
55. Li YY, Li JC. Cell signal transduction mechanism for nitric oxide regulating
lymphatic stomata and its draining capability. Anatomical Record-Advances in
Integrative Anatomy and Evolutionary Biology 2008;291(2): 216-223.
56. van den Tol RM, van Rossen EME, van Eijck CHJ, Bonthuis F, Marquet RL,
Jeekel H. Reduction of peritoneal trauma by using nonsurgical gauze leads to less
implantation metastasis of spilled tumor cells. Ann Surg 1998;227(2): 242-248.
57. van den Tol MP, Haverlag R, van Rossen MEE, Bonthuis F, Marquet RL,
Jeekel J. Glove powder promotes adhesion formation and facilitates tumour cell
adhesion and growth. Br J Surg 2001;88(9): 1258-1263.
58. Allendorf JDF, Bessler M, Horvath KD, Marvin MR, Laird DA, Whelan RL.
Increased tumor establishment and growth after open vs laparoscopic bowel resection
in mice. Surgical Endoscopy-Ultrasound and Interventional Techniques 1998;12(8):
1035-1038.
59. Allendorf JDF, Bessler M, Horvath KD, Marvin MR, Laird DA, Whelan RL.
Increased tumor establishment and growth after open vs laparoscopic surgery in mice
may be related to differences in postoperative T-cell function. Surgical Endoscopy-
Ultrasound and Interventional Techniques 1999;13(3): 233-235.
60. Carter JJ, Feingold DL, Kirman I, Oh A, Wildbrett P, Asi Z, Fowler R, Huang
E, Whelan RL. Laparoscopic-assisted cecectomy is associated with decreased
formation of postoperative pulmonary metastases compared with open cecectomy in a
murine model. Surgery 2003;134(3): 432-436.
61. Sylla P, Nihalani A, Whelan RL. Microarray analysis of the differential effects
of open and laparoscopic surgery on murine splenic T-cells. Surgery 2006;139(1): 92-
103.
62. Pattana-Arun J, Wolff BG. Benefits of povidone-iodine solution in colorectal
operations: Science or legend. Dis Colon Rectum 2008;51(6): 966-971.
63. Basha G, Ghirardi M, Geboes K, Yap SH, Penninckx F. Limitations of
peritoneal lavage with antiseptics in prevention of recurrent colorectal cancer caused
by tumor-cell seeding - Experimental study in rats. Dis Colon Rectum 2000;43(12):
1713-1718.
64. Lee SW, Gleason NR, Bessler M, Whelan RL. Peritoneal irrigation with
povidone-iodine solution after laparoscopic-assisted splenectomy significantly
decreases port-tumor recurrence in a murine model. Dis Colon Rectum 1999;42(3):
319-326.
65. Docherty JG, McGregor JR, Purdie CA, Galloway DJ, Odwyer PJ. Efficacy of
Tumoricidal Agents in-Vitro and in-Vivo. Br J Surg 1995;82(8): 1050-1052.
66. Huguet EL, Keeling NJ. Distilled water peritoneal lavage after colorectal
cancer surgery. Dis Colon Rectum 2004;47(12): 2114-2119.
67. Keating JP, Neill M, Hill GL. Sclerosing encapsulating peritonitis after
intraperitoneal use of povidone iodine. Aust N Z J Surg 1997;67(10): 742-744.
68. Hribaschek A, Kuhn R, Pross M, Meyer F, Fahlke J, Ridwelski K, Boltze C,
Lippert H. Intraperitoneal versus intravenous CPT-11 given intra- and postoperatively
for peritoneal carcinomatosis in a rat model. Surg Today 2006;36(1): 57-62.
69. Abaza R, Keck RW, Selman SH. Intraperitoneal chemotherapy for the
prevention of transitional cell carcinoma implantation. J Urol 2006;175(6): 2317-
2322.
70. Nordlinger B, Rougier P, Arnaud JP, Debois M, Wils J, Ollier JC, Grobost O,
Lasser P, Wals J, Lacourt J, Seitz JF, dos Santos JG, Bleiberg H, Mackiewickz R,
Conroy T, Bouche O, Morin T, Baila L, van Cutsem E, Bedenne L. Adjuvant regional
chemotherapy and systemic chemotherapy versus systemic chemotherapy alone in
patients with stage II-III colorectal cancer: a multicentre randomised controlled phase
III trial. Lancet Oncology 2005;6(7): 459-468.
71. Elias D, Lefevre JH, Chevalier J, Brouquet A, Marchal F, Classe JM, Ferron
G, Guilloit JM, Meeus P, Goere D, Bonastre J. Complete Cytoreductive Surgery Plus
Intraperitoneal Chemohyperthermia With Oxaliplatin for Peritoneal Carcinomatosis
of Colorectal Origin. Journal of Clinical Oncology 2009;27(5): 681-685.
72. Verwaal VJ, Bruin S, Boot H, van Slooten G, van Tinteren H. 8-year follow-
up of randomized trial: Cytoreduction and hyperthermic intraperitoneal chemotherapy
versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal
cancer. Annals of Surgical Oncology 2008;15(9): 2426-2432.
73. Elias D, Goere D, Di Pietrantonio D, Boige V, Malka D, Kohneh-Shahri N,
Dromain C, Ducreux M. Results of systematic second-look surgery in patients at high
risk of developing colorectal peritoneal carcinomatosis. Ann Surg 2008;247(3): 445-
450.
74. Harless WW. Revisiting perioperative chemotherapy: the critical importance
of targeting residual cancer prior to wound healing. Bmc Cancer 2009;9: 9.
75. McCormack PL, Keam SJ. Bevacizumab - A review of its use in metastatic
colorectal cancer. Drugs 2008;68(4): 487-506.
76. Allegra CJ, Yothers G, O'Connell MJ, Sharif S, Colangelo LH, Lopa SH,
Petrelli NJ, Goldberg RM, Atkins JN, Seay TE, Fehrenbacher L, O'Reilly S, Chu L,
Azar CA, Wolmark N. 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(20):
3385-3390.
77. Roh JL, Sung MW, Kim KH. Suppression of accelerated tumor growth in
surgical wounds by celecoxib and indomethacin. Head and Neck-Journal for the
Sciences and Specialties of the Head and Neck 2005;27(4): 326-332.
78. Roh JL, Sung MW, Park SW, Heo DS, Lee DW, Kim KH. Celecoxib can
prevent tumor growth and distant metastasis in postoperative setting. Cancer Res
2004;64(9): 3230-3235.
79. Coffey JC, Wang JH, Smith MJF, Bouchier-Hayes D, Cotter TG, Redmond
HP. Excisional surgery for cancer cure: therapy at a cost. Lancet Oncology
2003;4(12): 760-768.
80. Mels AK, Muller MGS, van Leeuwen PAM, von Blomberg BME, Scheper RJ,
Cuesta MA, Beelen RHJ, Meijer S. Immune-stimulating effects of low-dose
perioperative recombinant granulocyte-macrophage colony-stimulating factor in
patients operated on for primary colorectal carcinoma. Br J Surg 2001;88(4): 539-
544.
81. Brivio F, Lissoni P, Rovelli F, Nespoli A, Uggeri F, Fumagalli L, Gardani G.
Effects of IL-2 preoperative immunotherapy on surgery-induced changes in
angiogenic regulation and its prevention of VEGF increase and IL-12 decline.
Hepatogastroenterology 2002;49(44): 385-387.
82. Oosterling SJ, Mels AK, Teunis BHG, van der Bij GJ, Tuk CW, Vuylsteke R,
van Leeuwen PAM, Meijer GA, Meijer S, Beelen RHJ, van Egmond M. Preoperative
granulocyte/macrophage colony-stimulating factor (GM-CSF) increases hepatic
dendritic cell numbers and clustering with lymphocytes in colorectal cancer patients.
Immunobiology 2006;211(6-8): 641-649.
83. Ströhlein M, Heiss M. Immunotherapy of Peritoneal Carcinomatosis. In:
Peritoneal Carcinomatosis: a Multidisciplinary Approach, Ceelen W (ed). Springer:
New York, 2007; 483-491.
84. Alkhamesi NA, Ziprin P, Pfistermuller K, Peck DH, Darzi AW. ICAM-1
mediated peritoneal carcinomatosis, a target for therapeutic intervention. Clin Exp
Metastasis 2005;22(6): 449-459.
85. Park JS, Kim KM, Kim MH, Chang HJ, Baek MK, Kim SM, Do Jung Y.
Resveratrol Inhibits Tumor Cell Adhesion to Endothelial Cells by Blocking ICAM-1
Expression. Anticancer Res 2009;29(1): 355-362.
86. Pross M, Lippert H, Misselwitz F, Nestler G, Kruger S, Langer H, Halangk W,
Schulz HU. Low-molecular-weight heparin (reviparin) diminishes tumor cell
adhesion and invasion in vitro, and decreases intraperitoneal growth of colonadeno-
carcinoma cells in rats after laparoscopy. Thromb Res 2003;110(4): 215-220.
87. Niers TMH, Klerk CPW, DiNisio M, Van Noorden CJF, Buller HR, Reitsma
PH, Richel DJ. Mechanisms of heparin induced anti-cancer activity in experimental
cancer models. Critical Reviews in Oncology Hematology 2007;61(3): 195-207.
88. Borsig L. Antimetastatic activities of heparins and modified heparins.
Experimental evidence. Thromb Res;125: S66-S71.
89. Jansen M, Jansen PL, Otto J, Kirtil T, Neuss S, Treutner KH, Schumpelick V.
The inhibition of tumor cell adhesion on human mesothelial cells (HOMC) by
phospholipids in vitro. Langenbecks Arch Surg 2006;391(2): 96-101.
90. Oosterling SJ, van der Bij GJ, Boegels M, ten Raa S, Post JA, Meijer GA,
Beelen RHJ, van Egmond M. Anti-beta 1 integrin antibody reduces surgery-induced
adhesion of colon carcinoma cells to traumatized peritoneal surfaces. Ann Surg
2008;247(1): 85-94.
91. Takatsuki H, Komatsu S, Sano R, Takada Y, Tsuji T. Adhesion of gastric
carcinoma cells to peritoneum mediated by alpha 3 beta 1 integrin (VLA-3). Cancer
Res 2004;64(17): 6065-6070.
92. Heyder C, Gloria-Maercker E, Hatzmann W, Niggemann B, Zanker KS,
Dittmar T. Role of the beta(1)-integrin subunit in the adhesion, extravasation and
migration of T24 human bladder carcinoma cells. Clin Exp Metastasis 2005;22(2):
99-106.
93. Arlt MJE, Novak-Hofer I, Gast D, Gschwend V, Moldenhauer G, Grunberg J,
Honer M, Schubiger PA, Altevogt P, Kruger A. Efficient inhibition of intra-peritoneal
tumor growth and dissemination of human ovarian carcinoma cells in nude mice by
anti-L1-cell adhesion molecule monoclonal antibody treatment. Cancer Res
2006;66(2): 936-943.
94. Lamagna C, Hodivala-Dilke KM, Imhof BA, Aurrand-Lions M. Antibody
against junctional adhesion molecule-C inhibits angiogenesis and tumor growth.
Cancer Res 2005;65(13): 5703-5710.
95. Juhl H, Kalthoff H, Kruger U, Hennebruns D, Kremer B. Immunocytological
Detection of Micrometastatic Cells in Gastrointestinal Cancer-Patients. Zentralbl
Chir 1995;120(2): 116-122.
96. Hase K, Ueno H, Kuranaga N, Utsunomiya K, Kanabe S, Mochizuki H.
Intraperitoneal exfoliated cancer cells in patients with colorectal cancer. Dis Colon
Rectum 1998;41(9): 1134-1140.
97. Schott A, Vogel I, Krueger U, Kalthoff H, Schreiber HW, Schmiegel W,
Henne-Bruns D, Kremer B, Juhl H. Isolated tumor cells are frequently detectable in
the peritoneal cavity of gastric and colorectal cancer patients and serve as a new
prognostic marker. Ann Surg 1998;227(3): 372-379.
98. Wind P, Nordlinger B, Roger V, Kahlil A, Guin E, Parc R. Long-term
prognostic value of positive peritoneal washing in colon cancer. Scand J
Gastroenterol 1999;34(6): 606-610.
99. Vogel P, Ruschoff J, Kummel S, Zirngibl H, Hofstadter F, Hohenberger W,
Jauch KW. Prognostic value of microscopic peritoneal dissemination - Comparison
between colon and gastric cancer. Dis Colon Rectum 2000;43(1): 92-100.
100. Vogel I, Francksen H, Soeth E, Henne-Bruns D, Kremer B, Juhl H. The
carcinoembryonic antigen and its prognostic impact on immunocytologically detected
intraperitoneal colorectal cancer cells. Am J Surg 2001;181(2): 188-193.
101. Broll R, Weschta M, Windhoevel U, Berndt S, Schwandner O, Roblick U,
Schiedeck THK, Schimmelpenning H, Bruch HP, Duchrow M. Prognostic
significance of free gastrointestinal tumor cells in peritoneal lavage detected by
immunocytochemistry and polymerase chain reaction. Langenbecks Arch Surg
2001;386(4): 285-292.
102. Lucha PA, Ignacio R, Rowley D, Francis M. The incidence of positive
peritoneal cytology in colon cancer: A prospective randomized blinded trial. Am Surg
2002;68(11): 1018-1021.
103. Aoki S, Takagi Y, Hayakawa M, Yamaguchi K, Futamura M, Kunieda K, Saji
S. Detection of peritoneal micrometastases by reverse transcriptase-polymerase chain
reaction targeting carcinoembryonic antigen and cytokeratin 20 in colon cancer
patients. J Exp Clin Cancer Res 2002;21(4): 555-562.
104. Guller U, Zajac P, Schnider A, Bosch B, Vorburger S, Zuber M, Spagnoli GC,
Oertli D, Maurer R, Metzger U, Harder F, Heberer M, Marti WR. 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(6): 768-775.
105. Yamamoto S, Akasu T, Fujita S, Moriya Y. Long-term prognostic value of
conventional peritoneal cytology after curative resection for colorectal carcinoma.
Jpn J Clin Oncol 2003;33(1): 33-37.
106. Kanellos I, Demetriades H, Zintzaras E, Mandrali A, Mantzoros I, Betsis D.
Incidence and prognostic value of positive peritoneal cytology in colorectal cancer.
Dis Colon Rectum 2003;46(4): 535-539.
107. Bosch B, Guller U, Schnider A, Maurer R, Harder F, Metzger U, Marti WR.
Perioperative detection of disseminated tumour cells is an independent prognostic
factor in patients with colorectal cancer. Br J Surg 2003;90(7): 882-888.
108. Lloyd JM, McIver CM, Stephenson SA, Hewett PJ, Rieger N, Hardingham JE.
Identification of early-stage colorectal cancer patients at risk of relapse post-resection
by immunobead reverse transcription-PCR analysis of peritoneal lavage fluid for
malignant cells. Clin Cancer Res 2006;12(2): 417-423.
109. Kanellos I, Zacharakis E, Kanellos D, Pramateftakis MG, Betsis D. Prognostic
significance of CEA levels and positive cytology in peritoneal washings in patients
with colorectal cancer. Colorectal Disease 2006;8(5): 436-440.
110. Gozalan U, Yasti AC, Yuksek YN, Reis E, Kama NA. Peritoneal cytology in
colorectal cancer: incidence and prognostic value. Am J Surg 2007;193(6): 672-675.
111. Hara M, Nakanishi H, Jun Q, Kanemitsu Y, Ito S, Mochizuki Y, Yamamura
Y, Kodera Y, Tatematsu M, Hirai T, Kato T. Comparative analysis of intraperitoneal
minimal free cancer cells between colorectal and gastric cancer patients using
quantitative RT-PCR: possible reason for rare peritoneal recurrence in colorectal
cancer. Clin Exp Metastasis 2007;24(3): 179-189.
112. Kristensen AT, Wiig JN, Larsen SG, Giercksky KE, Ekstrom PO. Molecular
detection (k-ras) of exfoliated tumour cells in the pelvis is a prognostic factor after
resection of rectal cancer? Bmc Cancer 2008;8: 8.
113. Noura S, Ohue M, Seki Y, Yano M, Ishikawa O, Kameyama M. Long-Term
Prognostic Value of Conventional Peritoneal Lavage Cytology in Patients Undergoing
Curative Colorectal Cancer Resection. Dis Colon Rectum 2009;52(7): 1312-1320.
114. Fujii S, Shimada H, Yamagishi S, Ota M, Kunisaki C, Ike H, Ichikawa Y.
Evaluation of intraperitoneal lavage cytology before colorectal cancer resection. Int J
Colorectal Dis 2009;24(8): 907-914.
115. Shepherd NA, Baxter KJ, Love SB. The prognostic importance of peritoneal
involvement in colonic cancer: A prospective evaluation. Gastroenterology
1997;112(4): 1096-1102.
116. Solomon MJ, Egan M, Roberts RA, Philips J, Russell P. Incidence of free
colorectal cancer cells on the peritoneal surface. Dis Colon Rectum 1997;40(11):
1294-1298.
117. Lennon AM, Mulcahy HE, Hyland JMP, Lowry C, White A, Fennelly D,
Murphy JJ, O'Donoghue DP, Sheahan K. Peritoneal involvement in stage II colon
cancer. Am J Clin Pathol 2003;119(1): 108-113.
118. Baskaranathan S, Philips J, McCredden P, Solomon MJ. Free colorectal cancer
cells on the peritoneal surface: Correlation with pathologic variables and survival. Dis
Colon Rectum 2004;47(12): 2076-2079.
119. Keshava A, Chapuis PH, Chan C, Lin BPC, Bokey EL, Dent OF. The
significance of involvement of a free serosal surface for recurrence and survival
following resection of clinicopathological stage B and C rectal cancer. Colorectal
Disease 2007;9(7): 609-618.
120. Stewart CJR, Morris M, de Boer B, Iacopetta B. Identification of serosal
invasion and extramural venous invasion on review of Dukes' stage B colonic
carcinomas and correlation with survival. Histopathology 2007;51(3): 372-378.

You might also like