You are on page 1of 91

Journal of Surgical Oncology 2008;98:215–216

GUEST EDITORIAL

Building on a Consensus

PAUL H. SUGARBAKER, MD, FACS, FRCS*


Washington Cancer Institute, Washington Hospital Center, Washington, District of Columbia

Thanks to Drs. Kusamura, Deraco, and Delphi Technology, this Esquivel and colleagues focused on carcinomatosis from colon cancer.
supplemental issue of the Journal of Surgical Oncology records a Support for a new standard of care for carcinomatosis from colorectal
current consensus for the use of cytoreductive surgery and periopera- cancer comes from the randomized controlled data of Verwaal et al.,
tive intraperitoneal chemotherapy in the management of peritoneal the multi-institutional study of Glehen et al., and at least a dozen phase
dissemination of cancer within the abdomen and pelvis. This consensus II single institution studies reported from around the world [1–3].
came from the world’s experts in peritoneal surface oncology. Consensus voters agreed that early referral of these patients when the
These experienced clinicians and investigators should have the best peritoneal cancer index was less than 20 would be a crucial step
judgments regarding the balanced application of this emerging forward in the management of carcinomatosis. Bozzetti and colleagues
technology. focused on gastric cancer. Systematic reviews and meta-analyses of
Verwaal and colleagues discussed the eligibility for these perioperative intraperitoneal chemotherapy in stage III and resectable
treatments. Not surprisingly, they suggest that the oncologist has a stage IV gastric cancer show benefit [4,5]. In the much more
great responsibility to select patients for treatment who are candidates difficult problem of gastric cancer with established carcinomatosis
for long-term benefit. Yan and colleagues focused on the proper the consensus was less optimistic. However, new approaches with
preoperative investigations. The CT of chest, abdomen, and pelvis with neoadjuvant combined intraperitoneal and systemic chemotherapy
maximal oral and intravenous contrast was the preferred radiologic test may present the optimal palliation with a ray of hope for cure with
of consensus voters. One must remember that no radiologic test is patients who have a durable response. Baratti and colleagues discussed
reliable for the detection of small volume of cancer that is layered out the cytoreductive surgery and perioperative intraperitoneal chemo-
on a peritoneal surface. Identification of patients for the combined therapy for peritoneal mesothelioma. Combined treatment at an
treatment often occurs in the operating theater. Portilla and colleagues experienced center must be considered as a standard of care. Historical
focused on the four intraoperative assessments of cancer volume. The controls show that best palliative care or systemic chemotherapy offers
consensus voters were most comfortable with the peritoneal cancer little to these patients in comparison to cytoreductive surgery and
index. However, the Gilly staging system (Lyon), the simplified perioperative intraperitoneal chemotherapy as administered by an
peritoneal cancer index (Amsterdam), and the Japanese staging system experienced group. Moran and colleagues thought that similar
of P1, P2, and P3 continue to have merit. Kusamura and colleagues conclusions could be made for mucinous appendiceal carcinomatosis.
focused on the technical aspects of cytoreductive surgery. Periton- For ovarian cancer no consensus was reached. Clinical investigation
ectomy and visceral resections should include surfaces involved by the and phase I/II trials are currently active at many institutions.
malignancy. In order to prevent suture line recurrence anastomoses Several reports strongly suggest benefit in selected patients. Rossi
should occur following completion of the HIPEC. Gonzalez-Moreno and colleagues focused on abdominal sarcomatosis. Although
and colleagues focused on assessments to quantitate postoperative little evidence exists to recommend cytoreduction and HIPEC for
residual disease. The completeness of cytoreduction score as obtained sarcomatosis there are selected patients who are likely to benefit.
by an experienced surgeon’s visual assessment of the abdomen and Finally, Bijelic and colleagues focused on treatment failures
pelvis was voted to be the preferred scoring system. Completeness of after cytoreductive surgery and intraperitoneal chemotherapy. Their
cytoreduction indicates cancer control with surgery and perioperative experience with reoperative surgery with HIPEC was extremely
chemotherapy working together; it needs to be quantitated differently positive in appendiceal malignancy but less favorable with colon
for a particular disease state (compare papillary mesothelioma or cancer. Only approximately 10% of patients experienced long-term
pseudomyxoma peritonei with gastric cancer and colon cancer). survival with second look cytoreduction in patients with colon
Glehen and colleagues focused on the technology of HIPEC delivery. carcinomatosis.
Open, closed, semi-open, or expanded abdomen techniques all have Where are we currently with this new technology applied to a large
been reported. The superiority of one technique over the other in terms number of cancers that disseminate themselves within the peritoneal
of outcome, morbidity, and preservation of a safe operating room cavity? First, a new standard of care has been achieved and recognized
environment were not apparent to the consensus voters. Kusamura by the consensus group for peritoneal mesothelioma and appendiceal
and colleagues focused on the technical aspects of hyperthermic
intraperitoneal chemotherapy. The consensus was that no consensus *Correspondence to: Dr. Paul H. Sugarbaker, MD, FACS, FRCS, 106 Irving
exists. Younan and colleagues focused on systems for classification of Street NW, Suite 3900, Washington, DC 20010, USA; Fax: (202) 877-8602
morbidity, mortality, and cancer chemotherapy toxicity. Although E-mail: paul.sugarbaker@medstar.net
protocol-specific and institution-specific assessments have been used Received 1 April 2008; Accepted 3 April 2008
with great value, the consensus voters suggested that the Common DOI 10.1002/jso.21078
Terminology Criteria for Adverse Events (CTCAE) version 3 as Published online in Wiley InterScience
proposed by the National Institutes of Health is the current standard. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


216 Sugarbaker
mucinous neoplasms. It is possible that death from pseudomyxoma recurrence of carcinomatosis remains the number one focus for
peritonei can be eliminated with this new combined treatment improved management. Current needs are optimization of cytoreduc-
modality. Serial debulking procedures should not be used. tion through surgical workshops, an increased intensity of HIPEC and
In colon cancer it is hard to deny that patients with low volume of EPIC, and protocols to investigate long-term bidirectional chemo-
peritoneal seeding are likely to benefit with long-term survival. A therapy. As clinical and laboratory projects move forward, the broad
peritoneal cancer index of less than 10 show a 50% or greater survival application of this consensus to the community of oncologists remains
at 5 years. Systemic chemotherapy alone is not going to match this the greatest challenge.
great benefit.
The total lack of consensus regarding technology requires
prospective comparisons of different surgical technologies and REFERENCES
different perioperative intraperitoneal chemotherapy technologies. 1. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of
Although hyperthermic intraperitoneal chemotherapy in the operating cytoreduction and hyperthermic intraperitoneal chemotherapy
room is the standard of care there is no standard treatment regimen. versus systemic chemotherapy and palliative surgery in patients
For example, several groups are now using 5-fluorouracil systemically with peritoneal carcinomatosis of colorectal cancer [see comment].
as a sensitizer for the intraperitoneal drugs. Also, early postoperative J Clin Oncol 2003;21:3737–3743.
intraperitoneal administration of this cell cycle-specific drug may help 2. Glehen O, Kwiatkowski F, Sugarbaker PH, et al.: Cytoreductive
eradicate small volume residual disease. The use of early postoperative surgery combined with perioperative intraperitoneal chemotherapy
for the management of peritoneal carcinomatosis from colorectal
intraperitoneal paclitaxel may improve the local control of patients
cancer: A multi-institutional study. J Clin Oncol 2004;22:3284–
with peritoneal mesothelioma and ovarian cancer. 3292.
Recently, thoughts regarding long-term bidirectional chemotherapy 3. Yan TD, Black D, Savady R, et al.: Systematic review on the
in ovarian cancer have been revised; it should be currently a standard efficacy of cytoreductive surgery combined with perioperative
of care in the United States [6]. One should not think that cancer intraperitoneal chemotherapy for peritoneal carcinomatosis from
control on peritoneal surfaces can be maintained by a single surgery colorectal carcinoma. J Clin Oncol 2006;24:4011–4019.
and a single hyperthermic intraperitoneal chemotherapy treatment. 4. Xu DZ, Zhan YQ, Sun XW, et al.: Meta-analysis of intraperitoneal
Long-term sequential treatments using both intravenous and intra- chemotherapy for gastric cancer. World J Gastroenterol 2004;10:
peritoneal chemotherapy (bidirectional) need to be incorporated into 2727–2730.
5. Yan TD, Black D, Sugarbaker PH, et al.: A systematic review and
the management of many of these peritoneal surface diseases.
meta-analysis of the randomized controlled trials on adjuvant
The challenges to the oncologist for control of peritoneal surface intraperitoneal chemotherapy for resectable gastric cancer. Ann
malignancy remain large. Even with current improvements in Surg Oncol 2007;14:2702–2713.
technology, the most common site for progression of all the diseases 6. Armstrong DK, Bundy B, Wenzel L, et al.: Intraperitoneal cisplatin
that were discussed in the consensus is at the peritoneal surface. and paclitaxel in ovarian cancer. N Engl J Med 2006;354:
Although systemic disease is occasionally a problem, peritoneal 34–43.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:217–219

REVIEWS

The Delphi Approach to Attain Consensus in Methodology of Local Regional


Therapy for Peritoneal Surface Malignancy

1
SHIGEKI KUSAMURA, MD, PhD, DARIO BARATTI, MD,1 RAMI YOUNAN, MD,
2
AND MARCELLO DERACO, MD
1
*
1
Department of Surgery, National Cancer Institute of Milan, Milan, Italy
2
Department of Surgery, Surgical Oncology Unit, CHUM, University of Montreal Health Centre, Montreal, Canada

At the Fifth International Workshop on Peritoneal Surface Malignancy (PSM), held in Milan, December 2006, the consensus on technical aspects
of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) was obtained through the Delphi process. The following
topics were discussed: pre-operative workup; eligibility to CRS þ HIPEC; intra-operative staging system; technical aspects of surgery; residual
disease classification systems; HIPEC: nomenclature and modalities; drugs, carrier solution and optimal temperature; morbidity grading systems.
Conflicting points regarding above-mentioned topics were elaborated and voted in two rounds by a panel of international experts in local-regional
therapy. The purpose of this manuscript is to describe the organization and the methodology of the consensus statements and to interpret and
discuss the implications of the most striking results.
J. Surg. Oncol. 2008;98:217–219. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal carcinomatosis; cytoreductive surgery; hyperthemic intraperitoneal chemotherapy; consensus

INTRODUCTION accumulate theoretical and practical resources and explore how to


build an international network for the design of future prospective
About three decades have passed since the first experience on controlled studies. Surgeons, medical oncologists, anesthesiologists,
hyperthermic intraperitoneal chemotherapy conducted by Spratt et al. biologists, nutrition specialists, pathologists, nurses and psychologists
[1]. In this period the treatment of peritoneal surface malignancies were involved in this process.
(PSM) with cytoreductive surgery (CRS) and intraperitoneal che- The present meeting opened the debate in three grounds: the
motherapy (IC) has gained an enormous popularity, changing methodology and the technique of the local-regional therapy, disease
positively the expectations toward a clinical condition that in specific consensus and the state of the art of the disciplines related to
former times was considered incurable. Promising results in terms of local regional therapy.
survival have been reported according to phase II studies in the The purpose of this paper is (1) to describe the organizational steps
treatment of peritoneal mesothelioma, pseudomyxoma peritonei, and and the methodology that guided the consensus achievement process of
peritoneal carcinomatosis from ovarian cancers [2–4]. Moreover, a 5th International Workshop on PSM; (2) to discuss critically the main
phase III study confirmed the efficacy of the CRS þ HIPEC in the results of this consensus statement regarding the methodological and
treatment of peritoneal carcinomatosis from colorectal cancer [5]. technical aspects of CRS þ HIPEC.
However the body of literature addressing the local regional therapy
of PSM is growing chaotically. There is a lack of uniformity in the
indications, in the methodology of the procedure and in the instruments
MATERIALS AND METHODS
to assess results in terms of side effects and outcome after the Approach
treatment.
Accordingly, there are at least five available intra-operative staging The Delphi technique consists of a consensus method designed to
systems for peritoneal surface malignancy; at least four post-operative synthesize information. It allows the inclusion of a large number of
residual disease classification systems; more than six acronyms for individuals across diverse locations and expertise and avoids the
hyperthermic intraperitoneal; more than four side effects grading
criteria.
A panel of experts named Peritoneal Surface Oncology Group Special Issue: Dedicated to the 5th International Consensus Meeting on
International (PSOGI) has been organizing a biannual meeting since Peritoneal Surface Malignancies Treatment.
1998 to share strategies, experiences, tactics and best practices to reach The authors have no financial interest related to the contents of this article to
a consensus pertaining to several conceptual and technical aspects disclose.
regarding the treatment of PSM. *Correspondence to: Marcello Deraco, MD, Fondazione IRCCS Istituto
The last version of this meeting was held in Milan, in December Nazionale dei Tumori Milano, Via Venezian 1, 20133 Milan, Italy.
2006. It was entitled ‘‘5th International workshop on PSM. Fax: þ39-02-23902404. E-mail: marcello.deraco@istitutotumori.mi.it
Integrating Cytoreductive Surgery and Hyperthermic Intraperito- Received 19 March 2008; Accepted 21 March 2008
neal Chemotherapy into the management of Peritoneal Malignancies.’’ DOI 10.1002/jso.21059
Following the same policy of the previous meetings, the PSOGI Published online in Wiley InterScience
aimed to bring together a broad expertise to generate new hypothesis, (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


218 Kusamura et al.
situation where a specific expert might be anticipated to dominate the other experts. The purpose of the voting was not to conduct a
consensus process [6,7]. It has been frequently used in health care and simple survey to take a static photograph of the general knowledge
is liable to use a wide range of study types to be considered than is among the experts. The final intention was to trigger a discussion
usual in statistical reviews. Moreover, it allows a major role for the and to suggest to each expert to do a self-reflection of own
qualitative assessment of evidence [8]. convictions and attitudes in light of the current published evidence
and the opinion of other experts.
Panel Selection and Flowchart of Scientific Boards (2) The time elapsed between the two rounds was 18 days. The experts
had 16 days to participate in each round of voting.
An Italian scientific organizing committee (ISOC) set up in the (3) Anonymity. Each expert did not have an access to the opinion of
National Cancer Institute (NCI) of Milan was vested with the function his colleagues. Only during the second round he was expected to
to launch the discussion and take the first steps to structure the vote knowing the general opinion of the entire group.
consensus achievement process. The organization of the consensus (4) During the last week of each voting period the respondents
statement started with the establishment of a International Scientific received self-generated reminder emails every other day to
Committee (ISC). Components of ISC were chosen according to their enhance the response rate of the entire panel.
involvement in the area of local-regional therapy of PSM assessed by (5) Consensus was defined as the agreement of 51% of the voters
their clinical and scientific background and participation in previous [9,10].
biannual meetings. The role of this board was to guide the scientific
activities for the maturation and establishment of common concepts
related to specific issues regarding the management of PSM. RESULTS
In total 42 experts, coming from 17 different countries participated
Literature Search and Identification of in the consensus statement on methodological aspects of the
Main Conflicting Points locoregional therapy. The response rates were 74% and 76% in the
first and second rounds, respectively. The consensus was achieved in
Parallel to the ISC formation, a Scientific Secretariat (SS) was 66% and 72% in the first and second rounds, respectively.
structured with the participation of three components, experts in local The results of voting process related to each of the above-mentioned
regional therapy, as well. The role of this board was to assist the ISC by methodological topics is outlined elsewhere in this same issue of the
means of three main tasks: (1) to identify the principal conflicting present journal.
points regarding the methodological aspects of local regional therapy; Significant change of opinion between first and second rounds
(2) to elaborate a list of possible solutions for each conflicting point; succeeded into four sub topics:
(3) to elaborate related document for each conflicting point, fully
explaining the dimension of the problematic issue. . Residual disease classification systems, in particular the R
The following issues were chosen by ISC and SS as the main not classification in pseudomyxoma peritonei.
consensual topics: . Technical aspects of the surgery, timing for bowel anastomosis.
. Ideal carrier solution for HIPEC.
. Eligibility to the procedure of CRS þ HIPEC. . Optimal temperature at HIPEC.
. Pre-operative workup.
. Intra-operative staging system. The topic in which there was a higher rate of disagreement was
. Technical aspects of surgery. drugs to use in intraperitoneal chemotherapy.
. Residual disease classification systems. Other classic conflicting topics such as intra-operative staging
. HIPEC: nomenclature and modalities. system and residual disease classification and modality of HIPEC
. Drugs, carrier solution and optimal temperature. (open vs. closed vs. etc.) were solved.
. Morbidity grading systems.
The conflicting points related with each of the above-mentioned DISCUSSION
topics were formulated as multiple choice questions. The possible
solutions for each conflicting point should have been formulated, Most of coined conflicting points regarding the methodology of
clearly, neutrally, comprehensively, accompanied by references from LRT were solved. However, this fact does not mean that the right
the literature. The above-mentioned eight conflicting points were split answer was found nor that future studies are precluded. The
into 61 multiple choice questions. Delphi technique does not substitute the scientific method. Points that
For the elaboration of related documents the SS reviewed medical remained open as well as the those not included in the present debate
literature over the past 20 years, summarized the most relevant papers are expected to be approached in the upcoming meeting of 2008.
in different grids and tables and attempted to provide the evidence for Another open question to be cleared is how willing each participant of
each conflicting point, whenever it was appropriate. the Consensus statement would be to change their everyday practice in
base of these results.
One of the criticisms raised against the locoregional therapy of PSM
The Voting is the low level of evidence that characterizes the major part of the
The whole material including the conflicting points, their respective studies on the topic. Several reasons account for this fact. First,
possible solutions and related documents were made available online several difficulties surrounds the conduction of prospective rando-
on a WEB based voting system. The panel of experts comprising mized studies in the LRT field [11]. Second, the disomogeneity of
the ISC, ISOC, SS expressed their opinion according to the Delphi the nomenclature, techniques and methodological instruments to
approach. The following elements were present in this discussion: assess the side effects and outcome. The achievement of the present
consensus allowed the establishment of a common language concern-
(1) Voting in two rounds. In the first round the expert were asked to ing basic concepts among the various centers devoted to LRT in
select the option he/she believes was the most appropriate for each the world. Moreover, it strengthened the PSOGI as an international
conflicting point. In the second round, the experts were asked to network representing a forum to maturate ideas, design and implement
vote again, having the opportunity to see the previous rating of the future prospective trials.

Journal of Surgical Oncology


Consensus in Local Regional Treatment 219

REFERENCES with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol


2003;21:3737–3743.
1. Spratt JS, Adcock RA, Muskovin M, et al.: Clinical delivery 6. Jairath N, Weinstein J: The Delphi methodology (Part one): A
system for intraperitoneal hyperthermic chemotherapy. Cancer useful administrative approach. Can J Nurs Adm 1994;7:7–
Res 1980;40:256–260. 20.
2. Deraco M, Baratti D, Inglese MG, et al.: Peritonectomy and 7. Jairath N, Weinstein J: The Delphi methodology (Part two): A
intraperitoneal hyperthermic perfusion (IPHP): A strategy that useful administrative approach. Can J Nurs Adm 1994;7:29–42.
has confirmed its efficacy in patients with pseudomyxoma 8. Jones J, Hunter D: Consensus methods for medical and health
peritonei. Ann Surg Oncol 2004;11:393–398. services research. BMJ 1995;311:376–380. Review.
3. Deraco M, Casali P, Inglese MG, et al.: Peritoneal mesothelioma 9. Loughlin KG, Moore LF: Using Delphi to achieve congruent
treated by induction chemotherapy, cytoreductive surgery, and objectives and activities in a pediatrics department. J Med Educ
intraperitoneal hyperthermic perfusion. J Surg Oncol 2003;83: 1979;54:101–106.
147–153. 10. Keeney S, Hasson F, McKenna H: Consulting the oracle: Ten
4. Ryu KS, Kim JH, Ko HS, et al.: Effects of intraperitoneal lessons rom using the Delphi technique in nursing research. J Adv
hyperthermic chemotherapy in ovarian cancer. Gynecol Oncol Nurs 2006;53:205–212.
2004;94:325–332. 11. Elias D, Delperro JR, Sideris L, et al.: Treatment of peritoneal
5. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of carcinomatosis from colorectal cancer: Impact of complete
cytoreduction and hyperthermic intraperitoneal chemotherapy cytoreductive surgery and difficulties in conducting randomized
versus systemic chemotherapy and palliative surgery in patients trials. Ann Surg Oncol 2004;11:518–521.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:220–223

The Eligibility for Local-Regional Treatment of


Peritoneal Surface Malignancy

1
VIC J. VERWAAL, MD, PhD, SHIGEKI KUSAMURA, MD, PhD,2 DARIO BARATTI, MD,
2
2
AND MARCELLO DERACO, MD *
1
Department of Surgery, The Netherlands Cancer Institute, Amsterdam, The Netherlands
2
Department of Surgery, National Cancer Institute of Milan, Milan, Italy

At the Fifth International Workshop on Peritoneal Surface Malignancy, held in Milan, the consensus on technical aspects of cytoreductive surgery
(CRS) for peritoneal surface malignancy was obtained through the Delphi process. General conflicting points concerning the eligibility to the
local-regional therapy were discussed and voted.
J. Surg. Oncol. 2008;98:220–223. ß 2008 Wiley-Liss, Inc.

KEY WORDS: eligibility; local-regional therapy

INTRODUCTION the heat [2]. This treatment has a curative intent and has a reasonable
5-year survival.
The local-regional treatment of peritoneal surface malignancies LM have been treated with curative intent and a 5-year survival of
has gained an increasing popularity thanks to the favourable results 40% was obtained in selected patients. In these patients selection
in terms of outcome in various tumour histologies. However the criteria are mostly absence of local-regional disease [3].
procedure carries a significant morbidity and is time and resource It is questionable if the combined treatment of resectable LM and
consuming. This confirms the need for the establishment of a clear operable peritoneal carcinomatosis can be successfully curative as well.
eligibility criteria defining the potential indications of the method to Elias et al. [4] conducted a prospective study on 24 patients with
assure a maximisation of the prognostic gain, improvement of quality LM and moderate PC from colorectal origin. Liver resection plus
of life and minimisation of the side-effects. CRS were combined with a curative intent. One postoperative death
The purpose of this manuscript is to approach some general con- occurred and postoperative morbidity was 58%. Three-year overall and
flicting situations concerning the eligibility criteria for the combined disease-free survival rates were respectively 41.5% (95% confidence
treatment of cytoreductive surgery (CRS) and hyperthermic intraper- interval [CI]: 23–63) and 23.6% (95% CI: 11–45). Seven patients are
itoneal chemotherapy (HIPEC). disease-free with a mean follow-up of 27.8 months after their last
surgery, 3 having a repeated hepatectomy. Three patients developed a
In General peritoneal recurrence and 13 had recurrence in the liver. The combined
treatment of LM plus PC was shown to be feasible, and beneficial in
Eligibility for a treatment of CRS þ HIPEC should be seem in the
selected patients presenting three or fewer metastases.
light of therapeutic alternatives for those eventually selected patients.
In any case the real impact of the CRS and heated intraperitoneal
Unfortunately, the data that show results of alternative treatments for
chemotherapy on survival of patients with PC and LM would be
peritoneal carcinomatosis are scarce.
defined only by a prospective controlled trial.
Taking one example from colorectal cancer. There are several
studies concerning the treatment of stage IV colorectal cancer with
systemic chemotherapy. These studies include patients with metastatic Intestinal Obstruction
colorectal cancer (metastasis of any location). However, to enable The detection of intestinal obstruction in the preoperative CT scan
proper response evaluation most studies include only those patients evaluation has been considered a predictive factor of incomplete
who have measurable disease. For practical reason measurements are cytoreduction and thus it could represent a parameter precluding an
most often made on liver metastasis (LM) or lymph nodes. Thus, the aggressive and curative surgical approach [5,6].
data of stage IV colorectal trials are dominated by the results found in Patient with an internal obstruction has poorer results then patients
the treatment of liver and lung metastases. The contribution of the without internal obstructions.
results of the patients with other sided metastasis is limited in these Shen et al. conducted an analysis of prognosis in patients with PC
studies. submitted to CRS and intraperitoneal hyperthermic chemotherapy.
This fact hampers the extrapolation of results from studies on
metastatic colorectal cancer treated with systemic chemotherapy to
cases affected with peritoneal carcinomatosis from the same origin. The authors have no financial interest related to the contents of this article to
disclose.
Liver Metastasis (LM) *Correspondence to: Marcello Deraco, MD, Fondazione IRCCS Istituto
Nazionale dei Tumori Milano, Via Venezian 1, 20133 Milano, Italy.
Cytoreduction combined with hyperthermic intraperitoneal chemo- Fax: þ39-02-23902404. E-mail: marcello.deraco@istitutotumori.mi.it
therapy is a treatment for peritoneal surface involvement. The main Received 19 March 2008; Accepted 21 March 2008
issue in this treatment is the high concentration of chemotherapy in the DOI 10.1002/jso.21060
abdominal cavity and a limited systemic concentration [1]. Besides the Published online in Wiley InterScience
selective effect of the chemotherapy there is also a selective effect of (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Consensus Statement in Peritoneal Surface Malignancy Treatment 221
Although intestinal obstruction was correlated with a poor prognosis without either of these two preoperative CT scan findings has a 94%
by univariate analysis, it was not found to be a independent predicting probability of an adequate cytoreduction.
factor of survival after multivariate analysis (HR 1.71 (95% CI: 0.93–
3.16; P-value 0.09)) [7]. De Bree et al. reported on 25 cases of colorectal cancer. Films of
On the other hand it is more difficult to treat patients affected abdominopelvic CT scans from such patients treated by CRS and
by bowel obstructions with systemic chemotherapy. Up to date there HIPEC were retrospectively analysed by two radiologists separately.
are no selective data available on the results of these patients in A simplified peritoneal cancer index (SPCI) was used to determine the
chemotherapy studies. extent of peritoneal involvement. Correlation between the preoperative
In general practice patient with bowel obstructions should be CT based SPCI-scores as well as number of involved abdominopelvic
operated on before any other treatment can be started. areas (N) and survival was examined. The preoperative SPCI- and
Averbach and Sugarbaker [8] have evaluated short- and long N-scores of one of the radiologists had no statistically significant
term results of management of recurrent intraabdominal malignancy prognostic value, while for the second radiologist SPCI  7 and N  4
causing intestinal obstruction using surgery and intraperitoneal were associated with particularly poor outcome. Additionally, the
chemotherapy. Forty-two consecutive patients with PC were treated presence of ileocaecal region involvement and, depending on the
by CRS with or without intraperitoneal chemotherapy. There were radiologist, the occurrence of tumour deposits in the left subdiaphrag-
20 patients with primary tumours of appendix, 13 with cancer of colon matic area on CT appeared to be unfavourable prognostic signs. They
or rectum and 9 patients with cancer of other origins. Surgery included concluded that the prognostic value of preoperative conventional CT
bowel resections and peritonectomy procedures. In 30 patients early appeared to be radiologist dependent and therefore may be of limited
postoperative intraperitoneal chemotherapy was administered. The value in selecting colorectal cancer patients with peritoneal carcino-
overall morbidity was 55% while mortality was 7.14%. The projected matosis who will not benefit from extensive CRS followed by HIPEC.
3-year survival for this group of patients was 32.7%. As a result of Jacquet et al. [11] evaluated retrospectively the CT scans of the
treatment patients’ performance status improved in 47.6% of cases. abdomen and pelvis in 45 patients affected by mucinous peritoneal
They concluded that an aggressive reoperative approach may be carcinomatosis who were treated with surgery and intraperitoneal
considered for palliation of selected patients with recurrent cancer chemotherapy. The following findings were correlated with complete-
causing intestinal obstruction. ness of cytoreduction: tumour volume in small bowel mesentery
(P < 0.001), tumour volume in proximal jejunum (P ¼ 0.003), tumour
Preoperative CT Findings volume in distal jejunum (P ¼ 0.002), tumour volume in proximal
ileum (P ¼ 0.003), mesentery configuration (P < 0.001) and obstruc-
Due to the fact peritoneal carcinomatosis is a surface disease it will tion of bowel segments by tumour (P < 0.001).
always stay relatively undetected by imaging studies that are depended A statistical approach using a tree-structured diagram showed that
on tumour volume such as CT MRI and even PET. patients with both obstruction of bowel segments by tumour and
Computed tomography represents a powerful instrument in the tumour diameter greater than 0.5 cm on small bowel surfaces exclusive
preoperative selection of patients for the local-regional treatment. of distal ileum on preoperative CT scan, had an 88% probability of
Studies correlating preoperative CT findings with treatment outcome incomplete resection. Patients without these two CT findings had a
has been conducted in peritoneal carcinomatosis from peritoneal 92% probability of complete resection. The conclusion was that
mesothelioma, colorectal or from appendiceal origin [6,9,10]. patients whose scans show obstruction of bowel segments by tumour
Yan et al. [6] carried out a study to identify computed tomography and tumour diameter greater than 0.5 cm on small bowel surfaces
(CT) scan images that would be useful in patient selection for CRS and exclusive of distal ileum are unlikely to be candidates for CRS for the
perioperative intraperitoneal chemotherapy. An analysis of the treatment of peritoneal carcinomatosis.
preoperative CT scans of 30 patients with peritoneal mesothelioma A brief analysis of the studies on CT allow us to make some
was performed. Characteristic interpretative CT appearances of the observations:
small bowel and its mesentery were categorised into four classes
outlined in the Table I. . Most of studies were conducted in not adequately dimensioned
small series of patients.
. The correlation of these categories with the operability revealed 9 of . For the correlation with treatment outcome only completeness of
11 patients (82%) with the suboptimal cytoreduction and 2 of cytoreduction and prognosis were taken into account in the various
19 patients (11%) with adequate cytoreduction had Class III studies. The appropriate selection of patients for local-regional
interpretative findings (P < 0.001). treatment should be based not only on survival results but also on
. Seven patients (64%) in the suboptimal cytoreduction group and two variables representing risk factor for major morbidity and mortality
patients (11%) in the adequate cytoreduction group had a >5 cm related to treatment. There is no data in the literature assessing the
tumour mass in the epigastric region (P ¼ 0.004). value of preoperative CT findings as predictor of eventual treatment
. A patient with a tumour size >5 cm in the epigastric region and with complication.
a Class III appearance of the small bowel and its mesentery has a . Conclusions obtained in one type of malignancy should not be
probability of 100% for a suboptimal cytoreduction. A patient applied arbitrarily in an another histology. For example, which was

TABLE I. Classes of Characteristic Interpretative CT Appearances of the Small Bowel and Its Mesentery

Presence of Small bowel and Loss of mesenteric


Class ascites mesentery involvement vessel clarity CT scab interpretation

0 No No No Normal appearance
I Yes No No Ascites only
II Yes Thickening, enhancing No Solid tumour present
III Yes Nodular thickening, segmental obstruction Yes Loss of normal architecture

Journal of Surgical Oncology


222 Verwaal et al.
proven to be valid in peritoneal mesothelioma, not necessary will be TABLE III. Survival of 174 Patients With Peritoneal Dissemination of
valid for gastric cancer or colorectal cancer and so on. Appendiceal Malignancy Having Incomplete Cytoreductive Surgery

HIPEC HIPEC and EPIC No


HIPEC or Early Perioperative Intraperitoneal alone EPIC alone POIC
Chemotherapy (EPIC) in Patients With Median survival time 58.6 37.6 19 12
Suboptimal Cytoreduction (months)
% 2-year survival 79.5 55.8 34.7 33.4
The proportion of patients with peritoneal carcinomatosis that % 3-year survival 66.3 53.3 24.6 21.9
remain with suboptimal residual disease after a maximum surgical % 5-yearr survival 35.4 27.2 7.3 11.7
effort is considerable. In the Table II rates of incomplete cytoreduction
HIPEC, hyperthermic intraperitoneal chemotherapy; EPIC, early postoperative
are outlined according to the histological subtype of the primary.
intraperitoneal chemotherapy; POIC, perioperative intraperitoneal chemotherapy.
Taking into account the substantial difference in terms of survival
according to the completeness of cytoreduction, one can propose not to
perform the local-regional antiblastic therapy (HIPEC or EPIC) after
the surgery. The limited benefit in terms of outcome provided by the morbidity in these patients (up to 52% of the patients had major
intraperitoneal chemotherapy does not outweigh the increase in complications) [14].
morbidity and toxicity that the local-regional antiblastic treatment
could imply, in case of suboptimal residual disease remaining in the
cavity after the tentative cytoreduction. Lymph Node Involvement
However, most of the authors of the literature have reported the Lymph nodes are the typical first side of metastasis of many
performance of perioperative intraperitoneal chemotherapy even in malignancies and remains the most frequently used parameter guiding
those suboptimally cytoreduced cases, despite the limited available the performance of adjuvant treatment. The local-regional approach
evidence supporting this policy. There is only few uncontrolled data in targets the treatment of peritoneal surface. And therefore lymph nodes
the literature giving us an idea of in which extent the performance of are out of reach of this treatment.
HIPEC and/or EPIC could add in terms of survival in patients with In the study of Glehen et al. [15] in which they reviewed the ‘‘world
incomplete cytoreduction. series’’ of CRS combined with perioperative intraperitoneal che-
Glehen et al. [16] analysed the survival of 174 patients with motherapy for peritoneal carcinomatosis of colorectal origin it was
peritoneal dissemination of appendiceal malignancy having incom- found that node positive tumours have a significant poorer survival
plete CRS (Table III). then node negative (18 and 31 months).
Extensive Disease
Sugarbaker et al. evaluated the prognostic value of PCI in patients
MATERIALS, METHODS AND RESULTS
with colorectal cancer. In clinical studies a peritoneal cancer index of In order to achieve a consensus among experts, the Delphi
<10 was associated with a 5-year survival rate of 50%, and index of methodology was employed [18]. Conflicting points were identified
11–20 with a 5-year survival rate of 20%, and an index of >20 with a and related multiple choice questions along with a scientific review
5-year survival rate of 0% (P-value < 0.0001). Based on these data they document on the topic were circulated among a panel of experts
coined a clinical pathway to treat patients with peritoneal carcinoma- on peritoneal surface oncology. Two rounds of web-based voting,
tosis from colon cancer in which the procedure of cytoreduc- the second one after disclosing the results of the first round, were
tion þ HIPEC is contra-indicated in patients with PCI > 20 [12]. carried out. A detailed description of the methodology used for this
Verwaal et al. [13] studied prognostic factors extensively. In their consensus development is available elsewhere in this issue. Results
seven-region system they found not only that the survival benefit is low were presented and discussed at the Fifth International Workshop on
in patients with more than five regions effected but also in increase in Peritoneal Surface Malignancy held in Milan, Italy, December 4–6,
2006.
The contents of the related scientific review document are outlined
TABLE II. Rates of Incomplete Cytoreduction Are Outlined According to in the Introduction Section of this article. The accompanying related
the Histological Subtype of the Primary
question circulated among the experts and the respective results of the
Result of Median survival first and second rounds are outlined below:
Primary tumour cytoreduction Rate (%) (months) The local-regional treatment of peritoneal surface malignancies has
gained an increasing popularity thanks to favourable results in terms of
Gastric cancer Cc 0/1 43.9 19.2 outcome. However the procedure carries a significant morbidity,
Cc-2/3 52.1 7.8 is time and resource consuming. This confirms the need for the
Colorectal cancer CC-0 53.5 32.4
establishment of a clear eligible criteria defining the potential
CC-1 21 24
CC-2 25.5 8.4 indications of the method to assure a maximisation of the prognostic
Appendiceal mucinous Cc 0/1 72.9 gain, improvement of the quality of life and minimisation of the side-
tumour effects. Which one of the following you do judge to be an absolute
Cc-2/3 27.1 20.5 exclusion criteria for the procedure? (more than 1 alternative allowed)
Peritoneal mesothelioma Cc 0/1 69 59%a
Cc-2/3 31 19%a (1) Presence of tumour mass >5 cm in the epigastric region evaluated
Advanced epithelial Cc 0/1 78.4 89  9 by preoperative CT scan in peritoneal surface malignancies
ovarian cancer (except Pseudomyxoma Peritonei); First round: 37.50%—Second
Cc-2/3 21.6 19  9
round: 21.88%.
a 15-19
5-Year overall survival . (2) Class III of small bowel or small bowel mesentery evaluated
The bold characters signify to highlight figures related to suboptimal by preoperative CT scan; First round: 68.75%—Second round:
cytoreduction (cc-2 or cc-3). 93.75%.

Journal of Surgical Oncology


Consensus Statement in Peritoneal Surface Malignancy Treatment 223
(3) Obstruction of bowel segments, biliary tract or ureters; First round: 3. Aloia TA, Vauthey JN, Loyer EM, et al.: Solitary colorectal liver
37.50%—Second round: 40.63%. metastasis: Resection determines outcome. Arch Surg 2006;141:
(4) Tumour diameter greater than 0.5 cm on small bowel surfaces 460–466.
exclusive of distal ileum in mucinous peritoneal carcinomatosis; 4. Elias D, Benizri E, Pocard M, et al.: Treatment of synchronous
First round: 12.50%—Second round: 0%. peritoneal carcinomatosis and liver metastases from colorectal
(5) Presence of resectable liver metastases; First round: 9.38%— cancer. Eur J Surg Oncol 2006;32:632–636.
5. Sugarbaker PH: Management of peritoneal-surface malignancy:
Second round: 21.88%. The surgeon’s role. Langenbecks Arch Surg 1999;384:576–587.
(6) Peritoneal cancer index >20 in colorectal cancer; First round: 6. Yan TD, Haveric N, Carmignani CP, et al.: Abdominal computed
53.13%—Second round: 62.50%. tomography scans in the selection of patients with malignant
(7) Residual disease >2.5 mm after the CRS. First round: 46.88%— peritoneal mesothelioma for comprehensive treatment with
Second round: 28.13%. cytoreductive surgery and perioperative intraperitoneal chemo-
(8) Gross retroperitoneal lymph node involvement First round: therapy. Cancer 2005;103:839–849.
75.00%—Second round: 90.63%. 7. Shen P, Levine EA, Hall J, et al.: Factors predicting survival after
intraperitoneal hyperthermic chemotherapy with mitomycin C
after cytoreductive surgery for patients with peritoneal carcino-
matosis. Arch Surg 2003;138:26–33.
DISCUSSION AND COMMENTS 8. Averbach AM, Sugarbaker PH: Recurrent intraabdominal cancer
Experts agreed that Class III of small bowel or small bowel with intestinal obstruction. Int Surg 1995;80:141–146.
mesentery evaluated by preoperative CT scan, peritoneal cancer index 9. de Bree E, Koops W, Kroger R, et al.: Preoperative computed
tomography and selection of patients with colorectal peritoneal
>20 in colorectal cancer and gross retroperitoneal lymph node carcinomatosis for cytoreductive surgery and hyperthermic intra-
involvement were absolute exclusion criteria for local-regional peritoneal chemotherapy. Eur J Surg Oncol 2006;32:65–71.
therapy. On the other hand the panel did not consider obstruction of 10. Yan TD, Haveric N, Carmignani CP, et al.: Computed tomo-
bowel segments, biliary tract or ureters an absolute contra-indication graphic characterization of malignant peritoneal mesothelioma.
for the procedure. Experts did not reach a consensus regarding the Tumori 2005;91:394–400.
indication of HIPEC in case of suboptimal CRS. The panel of experts 11. Jacquet P, Jelinek JS, Chang D, et al.: Abdominal computed
did not consider that the presence of preoperative CT findings of tomographic scan in the selection of patients with mucinous
unresectability (presence of tumour mass >5 cm in the epigastrium, peritoneal carcinomatosis for cytoreductive surgery. J Am Coll
tumour nodules of 0.5 cm in the distal ileum in mucinous peritoneal Surg 1995;181:530–538.
12. Sugarbaker PH: Successful management of microscopic residual
carcinomatosis) could be contra-indication for the procedure. The disease in large bowel cancer. Cancer Chemother Pharmacol
present article aimed at to identify general eligibility criteria that could 1999;43:S15–S25.
be applicable in most of cases of PSM. 13. Verwaal VJ, van Tinteren H, van Ruth S, et al.: Predicting the
The topic of eligibility is complex and is not only related to some survival of patients with peritoneal carcinomatosis of colorectal
clinical situations represented by technical unfeasibility to the origin treated by aggressive cytoreduction and hyperthermic
procedure. As already noted the analysis of prognostic factors, risk intraperitoneal chemotherapy. Br J Surg 2004;91:739–746.
factors for major morbidity and impact on quality of life should be 14. Verwaal VJ, van Tinteren H, Ruth SV, et al.: Toxicity of
globally taken into account in the decision making process of patient cytoreductive surgery and hyperthermic intra-peritoneal chemo-
selection. therapy. J Surg Oncol 2004;85:61–67.
15. Glehen O, Kwiatkowski F, Sugarbaker PH, et al.: Cytoreductive
The purpose of the present article was not to report a comprehensive surgery combined with perioperative intraperitoneal chemo-
debate in order to identify all the possible clinical situations in each therapy for the management of peritoneal carcinomatosis from
single pathology, that could represent a contra-indications for the colorectal cancer: A multi-institutional study. J Clin Oncol 2004;
combined treatment of CRS and hyperthermic intraperitoneal chemo- 22:3284–3292.
therapy. This more complex task was accomplished by the disease 16. Glehen O, Mohamed F, Sugarbaker PH: Incomplete cytoreduction
specific consensus and the results of such a discussion are outlined in in 174 patients with peritoneal carcinomatosis from appendiceal
elsewhere in this issue. malignancy. Ann Surg 2004;240:278–285.
17. Sugarbaker PH, Yan TD, Stuart OA, et al.: Comprehensive
management of diffuse malignant peritoneal mesothelioma. Eur J
REFERENCES Surg Oncol 2006;32:686–691.
18. Tentes AA, Mirelis CG, Markakidis SK, et al.: Long-term survival
1. Witkamp AJ, de Bree E, Van Goethem R, et al.: Rationale and in advanced ovarian carcinoma following cytoreductive surgery
techniques of intra-operative hyperthermic intraperitoneal chemo- with standard peritonectomy procedures. Int J Gynecol Cancer
therapy. Cancer Treat Rev 2001;27:365–374. 2006;16:490–495.
2. van Ruth S, Verwaal VJ, Hart AA, et al.: Heat penetration in 19. Yonemura Y, Kawamura T, Bandou E, et al.: Treatment of
locally applied hyperthermia in the abdomen during intra- peritoneal dissemination from gastric cancer by peritonectomy
operative hyperthermic intraperitoneal chemotherapy. Anticancer and chemohyperthermic peritoneal perfusion. Br J Surg 2005;92:
Res 2003;23:1501–1508. 370–375.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:224–227

Preoperative Investigations in the Management of Peritoneal Surface Malignancy


With Cytoreductive Surgery and Perioperative Intraperitoneal Chemotherapy:
Expert Consensus Statement

1
TRISTAN D. YAN, * DAVID L. MORRIS, MD, PhD,1 KUSAMURA SHIGEKI, MD,
BSc (Med), MBBS, PhD, PhD,
2

BARATTI DARIO, MD,2 AND DERACO MARCELLO, MD2


1
Department of Surgery, University of New South Wales, St George Hospital, Sydney, Australia
2
Istituto Nazionale dei Tumori Milano, Milano, Italy

At the Fifth International Workshop on Peritoneal Surface Malignancy, in Milan, the consensus on preoperative investigations for peritoneal
surface malignancy was obtained through the Delphi process. The results showed that 100% of the voters considered that contrast-enhanced multi-
sliced CT was the fundamental imaging modality, whereas MRI, PET, laparoscopy and serum tumor markers were regarded as useful, but not
fundamental investigational modalities.
J. Surg. Oncol. 2008;98:224–227. ß 2008 Wiley-Liss, Inc.

KEY WORDS: preoperative investigations; peritoneal surface malignancy; cytoreductive surgery; intraperitoneal
chemotherapy

INTRODUCTION individual lesions with a diameter greater than 5 cm is 60–90%, in


contrast to 10–30% for a tumor smaller than 1 cm [12,14]. There is
Cytoreductive surgery (CRS) combined with perioperative intra- also a great degree of variation in sensitivity for tumor detection in
peritoneal chemotherapy (PIC) has been suggested as a treatment different anatomical regions. The sensitivity for tumor detection in
option for patients with peritoneal surface malignancy. On December 4– epigastrium, greater omentum and undersurfaces of the diaphragms
6, 2006, the Italian National Cancer Institute convened a consensus have been reported 60–90% [8,12,13], whilst sensitivity was 50–70%
conference on the management of peritoneal surface malignancy in in retroperitoneum and pelvis [8]. With thin collimation contrast-
Milan. This conference was timely in bringing together experts in this enhanced spiral CT, Low et al. [3] demonstrated a sensitivity of 58%, a
relatively new surgical oncology field in an effort to discuss current specificity of 89%, an accuracy of 79% for parietal peritoneal lesions
approaches to this disease. The consensus focused on several important and sensitivity of 89% and specificity of 98% in detection of pelvic
patient selection criteria, preoperative investigations, prognostic disease. In contrast, sensitivity was less than 20% for mesenteric
scoring systems, treatment regimens for specific diseases and methods deposits.
of prospective data collection. Further emphasis was made on stringent At most institutions, multi-sliced CT remains the dominant imaging
patient selection utilizing current investigational modalities to identify modality for patients with intra-abdominal malignancies. It is
appropriate surgical candidates. Currently, these modalities include generally easier to perform, with shorter imaging times, less movement
contrast-enhanced multi-sliced computed tomography (CT), magnetic artifacts caused by bowel peristalsis, provides a higher spatial
resonance imaging (MRI), positron emission tomography (PET), resolution and is more accessible and familiar to radiologists and
laparoscopy and serum tumor markers. clinicians. It is limited, however, by the difficulty in early nodule
PET provides a functional image, but has the drawback of less detection in thin patients without ascites. This may be the case in many
accurate spatial resolution for small lesions when compared with patients with early tumor dissemination.
CT and MRI. The latter imaging modalities have similar diagnostic However, in the presence of a reasonable amount of fat, fine
accuracy [1–3]. However, availability, simplicity and less movement stranding and small nodules could be appreciated with the use of spiral
artifacts, make multi-sliced CT a more widely used imaging tool for thin-collimation scanning. The presence of ascites is the most common
detection of peritoneal tumors [4]. Some institutions also utilized CT-manifestation of peritoneal carcinomatosis [15]. Peritoneal thick-
laparoscopy to evaluate the extent of disease involvement [5–7]. ening and enhancement either smooth or nodular, is another useful CT-
The aims of this paper were to outline the different preoperative sign. Masses on the parietal peritoneum are best seen in presence of
investigation modalities and report the consensus statements of expert ascites (Fig. 1). Thickening of the peritoneum has a predilection for the
opinions from the Fifth International Peritonectomy Workshop.

ABDOMINOPELVIC COMPUTED The authors have no financial interest related to the contents of this article to
disclose.
TOMOGRAPHY
*Correspondence to: Tristan D. Yan, BSc (Med), MBBS, PhD, Department
Using more advanced technology including multi-sliced CT, of Surgery, University of New South Wales, St George Hospital, Sydney,
superior accuracy in detection of peritoneal implants is expected. Australia; Fax: 61-2-9350-3997. E-mail: tristan.yan@unsw.edu.au
Sensitivity of CT in diagnosing peritoneal carcinomatosis varies from Received 24 March 2008; Accepted 27 March 2008
60% to 90% [8–11]. This rate is dependent upon the quality of CT, the DOI 10.1002/jso.21069
size of tumor nodules, the abdominopelvic regions examined and the Published online in Wiley InterScience
interpretation from the radiologist [8,12,13]. Sensitivity of detecting (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Preoperative Investigations 225
previous surgery, chemo- or radiation therapy or intrinsic factors such
as inflammatory disease, may also increase peritoneal permeability,
mimicking the effect of peritoneal carcinomatosis. Hence specific
diagnosis may be impossible in patients presenting under the
conditions described [17]. In addition, small amounts of ascites
also show contrast enhancement after intravenous administration of
Gd-DTPA. In some cases, contrast enhancing ascites may not be
distinguishable from small layers of enhancing tumors.

POSITRON EMISSION TOMOGRAPHY


Anatomic imaging can detect pathologic processes only after they
reach a detectable size, become contour deforming or have abnormal
enhancement after intravenous contrast administration. PET with 2-
[18F]-fluoro-2-deoxy-D-glucose (FDG) has evolved as a useful
technique in clinical oncology, especially for tumor staging and post-
treatment monitoring. It provides metabolic information and may help
detect small metastatic lesions. Viable tumors can be differentiated
more easily from adjacent structures, as glucose metabolism in tumor
Fig. 1. Abdominal CT of the mid-abdomen showing the presence of cells is often increased beyond normal limits, causing areas of focally
peritoneal tumor implants as the peritoneum appeared to be thickened
elevated FGD tracer uptake. PET has an overall sensitivity of 97% in
and enhanced in the presence of a large volume of ascites.
the detection of colorectal metastases in the entire body, according to a
recently published meta-analysis [19]. But, the high (>85%)
sensitivity of FDG-PET for tumor localization and detection is
cul-de-sac, the pericolic gutters and the undersurfaces of the associated with a lower specificity, due to the normal physiologic
diaphragms. In clinical practice, most sites of peritoneal tumor accumulation of FDG in the stomach, bowel and urinary tract.
infiltration, including omentum, diaphragms and pelvis can be Therefore, relying solely on functional imaging may result in decrease
satisfactorily managed with peritonectomy procedures. Also ileocecal overall accuracy due to false-positive lesions.
involvement can be treated with intestinal resection. Therefore tumor Accurate anatomic localization of functional abnormalities seen on
deposits in these areas, even when missed by pre-operative CT, usually PET scans is well known to be challenging because of the lack of
have limited impact on prediction of successfulness of CRS. Whereas, detailed, high-resolution anatomy. Due to its relative scarcity of anato-
disease involving portal hepatis, small bowel and/or mesentery is a mic landmarks, however, standard PET imaging remains frequently
limiting factor for complete cytoreduction. In these regions with low unsatisfactory. This major limitation of PET imaging is overcome by
contrast flow, CT has low sensitivity in distinguishing a tumor from the the availability of integrated whole-body PET/CT imaging systems,
surrounding tissue. In most series, small bowel and mesentery yielding intrinsically fused morphological and functional data sets. An
involvement was detected correctly only in 20–50% of the cases. anatomic image can be helpful in differentiating pathological activity
Secondary CT features may be helpful in identifying the disease and due to malignant processes from physiological FDG uptake in areas
these include small bowel distortion, obstruction, wall thickening and such as the bladder, stomach or the bowel. It can also help in
enhancement. Unfortunately, when patients present with these CT identifying mucinous primary and metastatic tumors that have poor
features, the disease is usually too far advanced to be salvaged by CRS FDG uptake. This may result in a decrease in the number of false-
and PIC [8,16]. negative PET studies and improved overall accuracy of combined
PET/CT study for mucinous tumors [20].
MAGNETIC RESONANCE IMAGING
Recent advances in MRI including faster pulse sequences, breath-
hold imaging, and use of intravenous contrast agents and surface coils,
have improved image quality and shortened examination times. MRI
with its superior contrast resolution is an attractive alternative to multi-
sliced CT scanning. Decision to use CT or MRI in patients with
peritoneal malignancy will also depend on factors like MRI
availability, cost and radiologist’s expertise. An inherent advantage
of MRI compared with CT is its ability to acquire multiple image types,
each of which may be useful for depicting different forms of abdominal
disease.
The depiction of thin peritoneal tumor layers largely depends on the
presence of contrast enhancement. In a series comprising of T2 and T1-
weighted contrast enhanced sequences, Low et al., observed a
sensitivity and specificity for bowel involvement of 73% and 77%
respectively in patients with ovarian carcinoma [17]. Some reports
suggest that tumors of the pelvic floor or gynecological organs are
detected with greater accuracy on sagittal or coronal slices [18]. To
enhance the predictive value, these sequences may be added.
The contrast enhancement in peritoneal carcinomatosis is probably Fig. 2. Laparoscopic exploration of the abdomen showing small
caused by increased permeability of the peritoneum, similar to the tumor deposits layering out on the peritoneum. [Color figure can be
effect seen in diffuse peritonitis. However, exogenous factors, such as viewed in the online issue, available at www.interscience.wiley.com.]

Journal of Surgical Oncology


226 Yan et al.
TABLE I. Advantages and Disadvantages of Current Investigational Modalities for Peritoneal Surface
Malignancy

Modality Advantages Disadvantages

Contrast-enhanced spiral CT Superior spatial resolution Low sensitivity for small tumors
Shorter imaging times Low sensitivity with mesenteric
deposits
Less movement artifacts Lower contrast resolution
Readily available
Clinical familiarity
MRI Superior contrast resolution Low sensitivity for small tumors
Multiple imaging types Longer imaging times
Manipulation of signal intensities Movement artifacts
Lower spatial resolution
PET or PET/CT* Functional activity Low sensitivity for small tumors
Higher sensitivity Lower specificity
Detection of occult metastases Increased cost
Anatomical localization* Limited availability
Mismatched fusion*
Laparoscopy Direct visualization Relatively invasive
High sensitivity for small tumors Technical difficulty with adhesions
Supplement to imaging Failure to assess retroperitoneal
space
Biopsy Risk of port track seeding
Increased cost

*Correlates with PET/CT, not PET.

Despite these advantages, PET/CT may be limited by its high cost, disseminated peritoneal disease and may fail to detect early localized
availability and effects of mismatch between the CT and PET images disease at a stage where treatment is most likely to be beneficial.
due to patient respiration. This mismatch is at maximum when the
clinical CT is acquired with breath hold at full inspiration while EXPLORATORY LAPAROSCOPY
the PET is acquired with the patient breathing normally. Care must
be exercised when diagnosing patients with disease in the anatomic A reliable way of assessing tumor severity is to visualize it directly.
regions most affected by breathing artifacts. These areas include the Therefore, some patients undergo an exploratory laparotomy, in which
diaphragms, base of the lung and the upper portion of the liver. Given the combined therapy would be abandoned, where the disease is found
the spatial resolution of 4–6 mm currently available in PET and PET/ to be unresectable. Laparotomy is a simple mean of evaluating tumors
CT systems and the fact that FDG is an unspecific radioactive tracer, and performing biopsies, but may appear overly aggressive for a
micro-metastases must still be considered a challenge. FDG-PET is staging-only procedure. Laparoscopic exploration of the abdomen
known to have poor sensitivity for small (<1 cm) metastases. Negative supplements the information provided by the imaging techniques and
PET/CT results will not exclude micro-metastases. However, positive enables direct visual assessment of peritoneal involvement (Fig. 2). It
findings in PET/CT may help to optimize therapeutic strategies and is associated with less pain, shorter hospitalization, and quicker time to
reduce unnecessary surgery. recovery in comparison to laparotomy. Recent studies show that
There is little doubt that patient selection is essential to avoid futile laparoscopy is safe to treat and stage early ovarian cancers [5,6,21].
aggressive treatment, but therein lies a major problem. The well Valle and Garofalo [7] used laparoscopy to stage 97 cases of
reported benefits of resection of liver metastases have partly emanated peritoneal carcinomatosis and achieved full laparoscopic PCI assess-
from advances in preoperative imaging, in particular CT, MRI and ment in 96/97 cases, while only 2/96 cases were understaged. There
PET. These modalities allowed early detection of treatable disease, was a good correlation between the open successive surgery data and
with increasingly accurate delineation of the location and the extent of the laparoscopic PCI. There was no mortality and no trochar port
the metastases. Currently, non-invasive imaging assessing the extent and site metastases. The authors suggested that patients with massive
potential resectability of peritoneal disease are not reliable. Because involvement of their small bowel or mesentery by staging laparoscopy
peritoneal deposits usually have a low volume density, all imaging should not be considered suitable for the combined procedure. Pomel
modalities have major limitations in the assessment of low-volume et al. achieved complete cytoreduction in seven of the eight patients

TABLE II. Consensus Results on Preoperative Investigations for Peritoneal Surface Malignancy, Obtained
Through Delphi Process (Percentages in Black Were the Results From the First Round of Voting; Percentages in
Red Were the Results From the Second Round of Voting)

Investigation modality (a) Fundamental (%) (b) Useful (%) (c) Useless (%)

Multi-slice CT 96.77 3.23 0


100.00 0 0
Total body PET 19.35 64.52 16.13
6.25 84.38 9.38
MRI 12.90 70.97 16.13
3.13 81.25 15.63
Laparoscopy 16.13 70.97 12.90
9.38 78.13 12.50

Journal of Surgical Oncology


Preoperative Investigations 227
who were considered resectable by laparoscopy [6]. In the remaining 6. Pomel C, Appleyard T, Gouy S, et al.: The role of laparoscopy to
patient, there was an underestimation of the extent of the disease. evaluate candidates for complete cytoreduction of peritoneal
Despite these advantages, there are some limitations associated carcinomatosis and hyperthermic intraperitoneal chemotherapy.
with laparoscopy. It is technically difficult to evaluate patients with Eur J Surg Oncol 2005;31:540–543.
extensive prior surgery and lymph node involvement in the retro- 7. Valle M, Garofalo A: Laparoscopic staging of peritoneal surface
malignancies. Eur J Surg Oncol 2006;32:625–627.
peritoneal space. The spread of malignant cells through the trochar 8. Jacquet P, Jelinek JS, Steves MA, et al.: Evaluation of computed
tract is another major concern. To avoid this risk, all laparoscopy tomography in patients with peritoneal carcinomatosis. Cancer
trochars should be placed in the mid-line and resected at the time of 1993;72:1631–1636.
cytoreduction [6]. 9. Tempany C, Zou K, Siverman S, et al.: Staging of advanced
Table I provides a summary of the advantages and disadvantages of ovarian: Comparison of imaging modalities—Report from the
CT, MRI, PET and laparoscopy. Radiological Diagnostic Oncology Group. Radiology 2000;215:
761–767.
10. Davies J, Chalmers A, Sue-Ling H, et al.: Spiral computed
tomography and operative staging of gastric carcinoma: A
CONSENSUS RESULTS comparison with histopathological staging. Gut 1997;41:314–
319.
At the Fifth International Workshop on Peritoneal Surface 11. de Bree E, Koops W, Kroger R, et al.: Peritoneal carcinomatosis
Malignancy, in Milan in late 2006, the consensus on preoperative from colorectal or appendiceal origin: Correlation of preoperative
investigations for peritoneal surface malignancy was obtained through CT with intraoperative findings and evaluation of interobserver
the Delphi process, which is described elsewhere in this issue [22]. The agreement. J Surg Oncol 2004;86:64–73.
results showed that 100% of the voters considered that contrast- 12. de Bree E, Koops W, Kroger R, et al.: Preoperative computed
enhanced multi-sliced CT is the fundamental imaging modality, tomography and selection of patients with colorectal peritoneal
whereas MRI (84.38%), PET (81.25%), laparoscopy (78.13%) and carcinomatosis for cytoreductive surgery and hyperthermic intra-
serum tumor markers (71.88%) were regarded as useful, but not peritoneal chemotherapy. Eur J Surg Oncol 2006;32:65–71.
fundamental investigational modalities (Table II). 13. Jacquet P, Jelinek JS, Chang D, et al.: Abdominal computed
tomographic scan in the selection of patients with mucinous
In conclusion, considering these current investigational modalities, peritoneal carcinomatosis for cytoreductive surgery. J Am Coll
a high quality cross-sectional imaging study, either contrast-enhanced Surg 1995;181:530–538.
spiral CT or MRI should be performed to evaluate patients undergoing 14. Coakley F, Choi P, Gougoutas C, et al.: Detection of peritoneal
CRS and PIC. FDG-PET or preferably PET/CT should be at least metastases by spiral CT in patients with ovarian cancers.
selectively considered as part of preoperative workup in high-risk Radiology 2002;223:495–499.
patients. Laparoscopic exploration may supplement imaging modal- 15. Walkey M, Friedman A, Sohotra P, et al.: CT manifestations of
ities to allow direct visualization. However, the use of these peritoneal carcinomatosis. Am J Roentgenol 1988;150:1035–
investigations should be individualized and planned as part of a 1041.
multidisciplinary approach. The role of CT, MRI and more latterly 16. Yan TD, Haveric N, Carmignani CP, et al.: Abdominal computed
tomography scans in the selection of patients with malignant
PET or PET/CT and laparoscopy in refining patient selection and peritoneal mesothelioma for comprehensive treatment with
improving prognosis remain to be closely evaluated. cytoreductive surgery and perioperative intraperitoneal chemo-
therapy. Cancer 2005;103:839–849.
REFERENCES 17. Low R, Saleh F, Song S, et al.: Treated ovarian cancer:
Comparison of MR imaging with serum CA-125 level and
1. Low R, Semelka R, Worawattanakul S, et al.: Extrahepatic physical examination: A longitudinal study. Radiology 1999;211:
abdominal imaging in patients with malignancy: Comparison of 519–528.
MR imaging and helical CT, with subsequent surgical correlation. 18. Hamm B, Kubik-Huch R, Fleige B: MR imaging and CT of the
Radiology 1999;210:625–632. female pelvis: Radiologic–patholohic correlation. Eur Radiol
2. Prayer L, Kainz C, Stiglbauer R, et al.: CT and MR accuracy in the 1999;9:3–15.
detection of tumor recurrence in patients treated for ovarian 19. Huebner R, Park K, Shepherd JE, et al.: A meta-analysis of the
cancer. J Comput Assist Tomogr 1993;17:626–632. literature for whole-body FDG PET detection of recurrent
3. Low R, Semelka R, Worawattanakul S, et al.: Extrahepatic colorectal cancer. J Nucl Med 2000;41:1177–1189.
abdominal imaging in patients with malignancy: Comparison of 20. Whiteford M, Whiteford H, Yee L, et al.: Usefulness of FDG-PET
MR imaging and helical CT, with subsequent surgical correlation. scan in the assessment of suspected metastatic or recurrent
Radiology 1999;210:625–632. adenocarcinoma of the colon and rectum. Dis Colon Rectum
4. Raptopoulos V, Gourtsoyiannis N: Peritoneal carcinomatosis. Eur 2000;43:759–767.
Radiol 2001;11:2195–2206. 21. Tozzi R, Kohler C, Ferrara A, et al.: Laparoscopic treatment of
5. Deffieux X, Castaigne D, Pomel C: Role of laparoscopy to early ovarian cancer: Surgical and survival outcomes. Gynecol
evaluate candidates for complete cytoreduction in advanced Oncol 2004;93:199–203.
stages of epithelial ovarian cancer. Int J Gynecol Cancer 2006; 22. Jones J, Hunter D: Consensus methods for medical and health
16:35–40. services research. Br Med J 1995;311:376–380.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:228–231

The Intraoperative Staging Systems in the Management of


Peritoneal Surface Malignancy

A. GOMEZ PORTILLA, MD,1* KUSAMURA SHIGEKI, MD, PhD,2


BARATTI DARIO, MD,2 AND DERACO MARCELLO, MD2
1
Carcinomatosis Peritoneal Program, Hospital San José, Vitoria, Spain
2
IRCCS Istituto Nazionale dei Tumori Milano, Peritoneal Surface Malignancy Program, Italy

The establishment of a rationale guideline for intraoperative staging system the extent of carcinomatosis is warranted. The quantitation of tumor
found at the time of surgical exploration of the abdomen has proven to be of value in assessment of prognosis and treatment planning in patients
with peritoneal carcinomatosis. Four different assessments systems have been employed more frequently, thus far. The advantages and
disadvantages of each classification systems are described and discussed. The results of the Consensus of the last 5th International Workshop on
Peritoneal Surface malignancy of Milan, December 2006 are presented.
J. Surg. Oncol. 2008;98:228–231. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal cancer index; staging systems; peritoneal surface malignancy; peritoneal carcinomatosis

INTRODUCTION The main advantages of the Lyon staging system are simplicity and
reproducibility. Its utility has been validated and demonstrated in
The careful selection of the patients for the treatment based on the the EVOCAPE study [5], were the natural history of peritoneal
staging of the disease is mandatory in most of surgical diseases. At carcinomatosis from non-gynaecologic malignancies was determined,
the present time, we lack a universally accepted and easily with a significant difference in median survival rates from 6 to
reproducible staging/scoring system that addresses the quantity and 3 months, when stages 1 and 2 were compared to stages 3 and 4,
location of the peritoneal dissemination and its impact on achieving a respectively.
complete cytoreduction. It is used also to designate the downstaging after the cytoreduction.
The quantitation of tumor found at the time of surgical exploration Medical oncologists and radiologists consider it as a valuable tool in
of the abdomen has proven to be of value in assessment of prognosis the decision-making process regarding the therapy for the patients
and provides critical information in treatment planning in patients with affected by peritoneal surface malignancy.
peritoneal carcinomatosis. The main disadvantages are that it is not able to predict resectability
Four different assessments systems have been employed more of carcinomatosis, and the failure to quantitate the distribution of
frequently and have been published: peritoneal surface implants.
. Gilly’s Peritoneal Carcinomatosis Staging [1];
. Japanese Gastric Cancer P System [2]; THE JAPANESE CARCINOMATOSIS
. Sugarbaker’s Peritoneal Cancer Index (PCI) [3]; STAGING SYSTEM
. Dutch Simplified Peritoneal Cancer Index (SPCI) [4].
Carcinomatosis from gastric cancer is classified by the Japanese
The advantages and disadvantages of each classification systems are research society of gastric cancer as described in Table II.
described and discussed. This classification has been used as an useful quantitative pro-
The major value of these quantitative prognostic systems is to gnostic indicator validated for gastric cancer.
exclude patients estimating prognosis and the risk of morbidity/ The main advantages of the Japanese staging system are simplicity,
mortality of the treatment. reproducibility. It has been used also to designate the downstaging after
The establishment of a rationale guideline for intraoperative staging the cytoreduction and it is useful for recurrent gastric cancer.
system of the extent of carcinomatosis is warranted, major require- The main disadvantages are its inability to locate the carcino-
ments are reproducibility and simplicity. matosis, and the failure to size assessment and quantitate the
The results of the Consensus of the last 5th International Workshop distribution of peritoneal surface implants.
on Peritoneal Surface malignancy of Milan, December 2006 are
presented. None to disclose by any of the authors.
*Correspondence to: A. Gomez Portilla, MD, Director, Carcinomatosis
THE GILLY’S PERITONEAL CARCINOMATOSIS Peritoneal Program, Hospital San José, Vitoria, C/ Beato Tomás de
Zumárraga 10, 01008-Vitoria, Spain. Fax: 34-945145709.
STAGING SYSTEM E-mail: agomezpor@teleline.es
This staging system first described by Gilly et al. [1], in Lyon in Received 25 March 2008; Accepted 27 March 2008
1994, takes into account only the size of the major malignant implants DOI 10.1002/jso.21068
and their distribution on the abdominal cavity. The details of this Published online in Wiley InterScience
system are summarized in Table I. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Peroperative Staging Systems in Carcinomatosis 229
TABLE I. Gilly’s Peritoneal Carcinomatosis Staging TABLE II. Japanese Staging System

Stage Definitions Code Significance

0 No macroscopic peritoneal carcinomatosis S0 No serosal invasion


1 Regional granulations <5 mm greatest diameter localized in one part S1 Suspected serosal invasion
of the abdomen S2 Definite serosal invasion
2 Diffuse abdominal granulations <5 mm in dimension but diffuse to S3 Adjacent organ invasion
the whole abdomen N1 Perigastric LN
3 Granulations >5 mm to 2 cm in greatest diameter N2 LN around the left gastrci artery, common hepatic artery, splenic
4 Large malignant cakes, >2 cm in greatest dimension artery, and celiac axis
N3 LN in the hepatoduodenal ligament, posterior aspect of pancreas, and
root of mesentery
N4 Paraortic and middle colic LN
SUGARBAKER’S PERITONEAL P0 No peritoneal metastases
CANCER INDEX (PCI) P1 Adjacent peritoneal involvement
P2 A few scattered metastases to distant peritoneum
The PCI was proposed by Jacquet and Sugarbaker, and is P3 Many distant peritoneal metastases
represented by a score that comprises both the peritoneal implant size H0 No liver metastases
and distribution of the nodules on the peritoneal surface. H1 Metastases limited to one lobe
H2 A few bilateral metastases
To arrive at a score, the size of the peritoneal nodules must be
H3 Numerous bilateral metastases
assessed, using the lesion size score (LS): an LS0 score means that no
malignant deposits are visualized; an LS1 score means that tumor
nodules less than 0.5 are present; an LS2 score signifies that tumor shown in Figure 1. The summation of the LS score in each of the
nodules between 0.5 and 5.0 cm are present; an LS3 score signifies the abdomino-pelvic regions is the PCI of the patient, with a maximal
presence of tumor nodules greater than 5.0 cm in any dimension, or in score of 39.
case there is confluence of tumor. In order to assess the distribution of The main advantages of Sugarbaker’s staging system are that it
the peritoneal disease, the abdomino-pelvic regions are utilized as allows to estimate the probability of a complete cytoreduction in colon

Fig. 1. Sugarbaker’s PCI. The PCI (Peritoneal Cancer Index) is a clinical integration of both peritoneal implant size and distribution of
the nodules on the peritoneal surface. To arrived at a score, the size of the peritoneal nodules must be assessed. The Lesion size Score (LS) should
be use. An LS-0 score means that no malignant deposits are visualized. An LS-1 score means that tumor nodules less than 0.5 cm are present. An
LS-2 score signifies that tumor nodules between 0.5 cm and 5.0 cm are present. An LS-3 score signifies the presence of tumor nodules greater than
5.0 cm in any dimension. If there is a confluence of tumor, the lesion is scored as 3 In order to assess the distribution of the peritoneal disease, the
abdominopelvic regions are utilized. The summation of the LS score in each of the abdominopelvic regions is the PCI of the Patient. A maximal
score is 39.

Journal of Surgical Oncology


230 Gómez Portilla et al.
TABLE III. Dutch Simplified Peritoneal Cancer Index simplified peritoneal cancer index, where the size of tumor is recorded
Tumor is recorded as as large, moderate, small or no involvement as describe in Table III and
Large >5 cm the distribution of tumor is recorded in seven abdominal areas: left an
Moderate 1–5 cm right subdiaphragmatic, subhepatic, omentun/transverse colon, small
Small <1 cm intestine/mesentere, ileocecal and pelvic regions.
None The main advantages of the SPCI staging system are that it has been
Seven abdominal regions validated in PMP and in carcinomatosis from colon cancer. It allows a
I Pelvis correlation with the incidence of complications, and it is a valuable
II Right lower abdomen
guide for patient selection.
III Greater omentum, transverse colon and spleen
IV Right subdiaphragmatic area The main disadvantages are that some abdomino-pelvic regions are
V Left subdiaphragmatic area not designated, like the epigastric region, or regions of the descending
VI Subhepatic and lesser omental area and sigmoid colon AR4–AR5 (Table IV).
VII Small bowel and small bowel mesentery A summary of the staging systems is outlined in Table V.

MATERIAL AND METHODS


carcinomatosis, according to Elias et al. [6] the limit is a PCI < 16, and In order to obtain the consensus regarding the best preoperative
in Sugarbaker’s experience the limit should be a PCI <20 [7]. It has staging system among those mentioned above, an international panel of
also been useful as a guide for sequential re-operative surgery [8] and experts in local–regional therapy participated in a web-based voting
represents the most popular preoperative staging system among system, following the two-step Delphi methodology. Consensus was
surgical teams dealing with peritoneal carcinomatosis. defined as the agreement of 51% of the voters [23,24]. Further details
The main disadvantage is that it has no prognostic implication in of consensus statement methodology could be found elsewhere in the
Pseudomyxoma peritonei (PMP) and primary malignant peritoneal same issue. The following question was presented to the experts.
mesothelioma. In your opinion which one is the best intraoperative staging system
for patients affected by peritoneal carcinomatosis?

DUTCH SIMPLIFIED PERITONEAL . French system [1]


CANCER INDEX (SPCI) . Japanese system [2]
. Peritoneal cancer Index [3]
Zoetmulder and Verwaal [9], at the Netherlands Institute have used . Dutch Simplified Peritoneal Cancer Index [4]
a quantitative scoring system quite similar to the PCI, known as the . TNM system [25]

TABLE IV. Characteristics of the Main Intraoperative Staging Systems

System Survivorship prediction Advantage Disadvantage

Gilly Peritoneal In 370 patients with peritoneal Simple and reproducible [19] Should not be designated a ‘‘staging
Carcinomatosis Staging [1] carcinomatosis from non-gynecologic Downstaging system’’ because patients can only be
malignancies, a significant difference Used by Medical Oncologists staged once in the course of their
observed between stages 1/2 (median and Radiologists disease at the time of diagnosis of the
survival ¼ 6 months) and stages 3/4 primary malignancy [19]
(median survival 3 months) [5] Not able to predict resectability of
Prognostic value proven in other carcinomatosis (stage 2 disease could
clinical trials [10–12] consist of diffuse peritoneal
carcinomatosis with nodules <5 mm
that are non-resectable [19])
Failure to quantitate distribution of
peritoneal surface implants in the
stages 3 and 4 categories [19]
Japanese system [2] Prognostic value proven in other Simple [19] Inability to accurately locate the
clinical trials (gastric cancer) [13–15] Can also be applied to patients carcinomatosis
with recurrent gastric cancer [19] It has no size assessment of the
cancerous implants [19]
Peritoneal Cancer Index [3] Prognostic value proven in colon cancer Reproducible [19] PCI has no prognostic implication in the
[15–17] and ovarian cancer [18] following possible cases:
Allows to estimate the probability Non-invasive disease such as
of a complete cytoreduction [20] Pseudomyxoma peritonei [22] in
Is a guide for sequential determinations which a high PCI not necessary is
of volume of carcinomatosis correlated with a poor prognosis
over time estimating the likelihood A patient with invasive cancer at a
of a complete cytoreduction at crucial anatomic site (around the
re-operative surgery [8] common bile duct), even if the tumor
Allows a correlation with the incidence elsewhere is of low burden, the
of complications in patients who prognosis will be poor [19]
Dutch Simplified Peritoneal Prognostic value proven in receive local-regional therapy [21] The epigastric region, very important in
Cancer Index (SPCI) [4] colorectal cancer [4] Volume of tumor in each region is to determining the completeness of
be scored quantitatively [20] cytoreduction in some diseases is not
designated separately [19]

Journal of Surgical Oncology


Peroperative Staging Systems in Carcinomatosis 231
TABLE V. Results of the Panel of Experts Decision 9. Zoetmulder FAN, Verwaal V: Hyperthermic intrapoeritoneal
chemotherapy (HIPEC) with mitomycin C significantly improves
First round of Second round of survival in patients with peritoineal carcinomatosis of colorectal
Staging system the voting the voting origion (abstract). ASCO Prog Proc 2002;21:147a.
10. Beaujard AC, Glehen O, Caillot JL, et al.: Intraperitoneal
Gilly’s system 9.68% 6.25% chemohyperthermia with mitomycin C for digestive tract cancer
Japanese system 0% 0% patients with peritoneal carcinomatosis. Cancer 2000;88:2512–
Sugarbaker’s PCI 83.70% 87.50% 2519.
Dutch SPCI 6.45% 6.25% 11. Sayag-Beaujard AC, Francois Y, Glehen O, et al.: Intraperitoneal
TNM 0% 0% chemo-hyperthermia with mitomycin C for gastric cancer patients
with peritoneal carcinomatosis. Anticancer Res 1999;19:1375–
RESULTS 1382.
12. Rey Y, Porcheron J, Talabard JN, et al.: Peritoneal carcinomatosis
The decision of the panel of experts is outlined in Table V, been the treated by cytoreductive surgery and intraperitoneal chemo-
Sugarbaker’s PCI elected as the best intraoperative staging system. hyperthermia. Ann Chir 2000;125:631–642 French.
13. Hagiwara A, Togawa T, Yamasaki J, et al.: Extensive gastrectomy
and carbon-adsorbed mitomycin C for gastric cancer with
CONCLUSION peritoneal metastases. Case reports of survivors and their
implications. Hepatogastroenterology 1999;46:1673–1677.
According to the opinion of the panel of experts, the Sugarbaker’s 14. Ouchi K, Sugawara T, Ono H, et al.: Therapeutic significance of
PCI has been elected as the best intraoperative staging system and it is palliative operations for gastric cancer for survival and quality of
proposed to be used by all the investigators involved in the treatment of life. J. Surg Oncol 1998;69:41–44.
patients with peritoneal surface malignancies. 15. Fujimoto S, Takahashi M, Mutou T, et al.: Improved mortality rate
of gastric carcinoma patients with peritoneal carcinomatosis
treated with intraperitoneal hyperthermic chemoperfusion com-
REFERENCES bined with surgery. Cancer 1997;79:884–891.
16. Sugarbaker PH, Jablonski KA: Prognostic features of 51 color-
1. Gilly FN, Carry PY, Sayag AC, et al.: Regional chemotherapy ectal and 130 appendiceal cancer patients with peritoneal
(with mitomycin C) and intra-operative hyperthermia for carcinomatosis treated by cytoreductive surgery and intraperito-
digestive cancers with peritoneal carcinomatosis. Hepatogas- neal chemotherapy. Ann Surg 1995;221:124–132.
troenterology 1994;41:124–129. 17. Elias D, Blot F, El Otmany A, et al.: Curative treatment of
2. Japanese Research Society for Gastric Cancer: Jpn J Surg 1981; peritoneal carcinomatosis arising from colorectal cancer by
11:127–139. complete resection and intraperitoneal chemotherapy. Cancer
3. Jacquet P, Sugarbaker PH: Clinical research methodologies in 2001;92:71–76.
diagnosis and staging of patients with peritoneal carcinomatosis. 18. Tentes AAK, Tripsiannis G, Markakidis SK, et al.: Peritoneal
In: Sugarbaker PH, editor. Peritoneal carcinomatosis: Principles cancer index: A prognostic indicator of survival in advanced
of management. editors. Boston: Kluwer Academic Publishers ovarian cancer. Eur J Surg Oncol 2003;29:69–73.
1996:359–374. 19. Harmon RL, Sugarbaker PH: Prognostic indicators in peritoneal
4. Witkamp AJ, de Bree E, Kaag MM, et al.: Extensive cyto- carcinomatosis from gastrointestinal cancer. Int Semin Surg
reductive surgery followed by intra-operative hyperthermic Oncol 2005;2:3.
intraperitoneal chemotherapy with mitomycin-C in patients with 20. Sugarbaker PH: Intraperitoneal chemotherapy and cytoreductive
peritoneal carcinomatosis of colorectal origin. Eur J Cancer 2001; surgery for the prevention and treatment of peritoneal carcino-
37:979–984. matosis and sarcomatosis. Semin Surg Oncol 1998;14:254–261.
5. Sadeghi B, Arvieux C, Glehen O, et al.: Peritoneal carcinomatosis Review.
from nongynecological malignacies: Results of the EVOCAPE 21. Verwaal VJ, van Tinteren H, Ruth SV, et al.: Toxicity of
1 multicentric prospective study. Cancer 2000;88:358–363. cytoreduction and hyperthermic intra-peritoneal chemotherapy.
6. Elias DM, Pocard M: Treatment and prevention of peritoneal J. Surg Oncol 2004;85:61–67.
carcinomatosis from colorectal cancer. Surg Oncol Clin N Am 22. Sugarbaker PH: Management of peritoneal surface malignancy:
2003;12:543–559. The surgeon’s role. Langenbeck’s Arch Surg 1999;384:576–587.
7. Sugarbaker PH: Successful management of microscopic residual 23. Loughlin KG, Moore LF: Using Delphi to achieve congruent
disease in large bowel cancer. Cancer Chemother Pharmacol objectives and activities in a pediatrics department. J. Med Educ
1999;43(Suppl):S15–S25. 1979;54:101–106.
8. Gomez Portilla A, Sugarbaker PH, Chang D: Second-look 24. Keeney S, Hasson F, McKenna H: Consulting the oracle: Ten
surgery after cytoreduction and intraperitoneal chemotherapy lessons rom using the Delphi technique in nursing research.
for peritoneal carcinomatosis from colorectal cancer: Analysis of J. Adv Nurs 2006;53:205–212.
prognosis features. World J Surg 1999;23:23–29. 25. http://www.uicc.org. Last access 7/2006.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:232–236

Technical Aspects of Cytoreductive Surgery

SHIGEKI KUSAMURA, MD, PhD,1 SARAH T. O’DWYER, MD, FRCS,2 DARIO BARATTI, MD,1
RAMI YOUNAN, MD,3 AND MARCELLO DERACO, MD1*
1
Department of Surgery, National Cancer Institute of Milan, Milan, Italy
2
Peritoneal Tumour Service, Christie Hospital, Manchester, UK
3
Department of Surgery, Surgical Oncology Unit, CHUM, University of Montreal health centre, Montreal, Canada

At the Fifth International Workshop on Peritoneal Surface Malignancy, in Milan, the consensus on technical aspects of cytoreductive surgery
(CRS) for peritoneal surface malignancy was obtained through the Delphi process. Five conflicting points were discussed: radicality of the
peritonectomy procedure, cytoreduction of neoplastic nodules <2.5 mm, the timing of bowel anastomoses in relation to hyperthermic
intraperitoneal chemotherapy (HIPEC) and indications of protective ostomies. According to the panel of experts a partial parietal peritonectomy
restricted to the macroscopically involved regions could be indicated in all listed clinical conditions with the exception of peritoneal
mesothelioma. No expert was of the opinion that a radical parietal peritonectomy is advisable irrespective of the disease being treated. All the
experts agreed that electrovaporization of small (<2.5 mm) non-infiltrating metastatic nodules in the mesentery would be appropriate, even if
theoretically the HIPEC affords microscopic cytoreduction. The panel also agreed that in the closed technique for HIPEC administration the
intestinal anastomoses should be fashioned after completion of the perfusion. Finally when considering the place for protective ostomies the
experts voted for a flexible approach allowing the surgeon to exercise discretion for individual patients.
J. Surg. Oncol. 2008;98:232–236. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal carcinomatosis; cytoreductive surgery; consensus

INTRODUCTION invasive ability and the presence of mucinous acites. For example
moderate and high grade cancers show early local implantation of
On December 4–6, 2006, the National Cancer Institute of Milan the primary tumor to the peritoneal surface due to the presence of
organized a consensus statement on the management of peritoneal adherence molecules on the surface of the cancer cells. In these cases
surface malignancy (PSM). This conference brought together experts implantation can remain confined to a region even when a considerable
in the field of local-regional therapy in an effort to discuss current quantity of ascites is present and has been described as randomly
approaches to this PSM. proximally distributed (RPD). In contrast low grade tumor cells
The purpose of this manuscript was not to trigger a comprehensive express few adherence molecules and demonstrate limited involvement
discussion about all the technical variations involved in cytoreductive of the peritoneal surface near the primary tumors. Furthermore, where
surgery but to focus on key controversial aspects of surgical technique. there is a high production of mucin a complete redistribution
The fundamental procedures undertaken in CRS have been described phenomenon (CRD) can occur in the peritoneal cavity resulting in
and standardized by Sugarbaker [1], however with increasing number of few invasive implants and scant epithelial cells, classically character-
surgeons setting up treatment centers for peritoneal surface malignancies ized in diffuse peritoneal adenomucinosis (DPAM) and Pseudo-
across the world several modifications have been introduced. Further- myxoma Peritonei. Finally, widespread cancer dissemination (WCD)
more recognition of the potential of CRS and HIPEC applied to characterizes the invasive mucinous tumor that produces high
additional disease groups requires the adaptation of the original quantities of mucus, which in itself interferes with cell adherence
techniques with some modification aligned to specific tumor types. [6]. Recent evaluation of adhesion molecules in PMP and colorectal
Using an expert panel we explored five areas where there was cancer has demonstrated a distinct profile that helps to explain the
known variation in practice of surgical technique in an attempt to gain pattern and distribution of disease and offers potential for molecular
consensus and open opportunities for further studies and fertile targeting to prevent invasion [7]. The patterns of spread of the common
discussion in future. histotypes of peritoneal carcinomatosis are described in the Table I.
Based on these observations one could propose a sparing surgical
Radicality of the Peritonectomy Procedure approach with partial parietal peritonectomy where there is limited
peritoneal carcinomatosis as seen in the random and proximal
The extent of surgery has a direct and significant impact on the distribution (RPD) disease.
mortality and morbidity associated with the CRS [2–5]. While a
complete peritonectomy procedure seems appropriate when there is
widespread macroscopic disease (except in colorectal cancer with a The authors have no financial interest related to the contents of this article to
peritoneal cancer index >20), the same does not hold true in case of disclose.
limited or localized peritoneal carcinomatosis. The rationale for *Correspondence to: Dr. Marcello Deraco, MD, Istituto Nazionale Tumori
complete or partial peritonectomy lies with the histology of the Milano, Via Venezian 1, 20133 Milan, Italy; Fax: þ39-02-23902404.
primary tumor and the pattern of spread within the peritoneal cavity. E-mail: marcello.deraco@istitutotumori.mi.it
The extent of peritoneal carcinomatosis is determined by the ability Received 19 March 2008; Accepted 21 March 2008
of tumor cells to disseminate transcaelomically and to implant into DOI 10.1002/jso.21058
the peritoneal surface. Three patterns of dissemination have been Published online in Wiley InterScience
described by Sugarbaker and are determined by the histologic grade/ (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Consensus on Cytoreductive Surgery 233
TABLE I. Hypothesis of Cancer Spread the traditional open abdominal Coliseum technique delay the anasto-
moses until after HIPEC as they believe that manipulation of the bowel
Histologic type RPD CRP WCD
achieves better distribution of heat and drugs throughout the peritoneal
Pseudomyxoma peritonei þ cavity. When the abdomen is closed or sealed during perfusion there
Appendix cancer remains a choice as to whether to anastomose before after the treatment.
Cystoadenocarcinoma G1 þ The key advantages and disadvantages are outlined in Table II.
Cystoadenocarcinoma G2, G3 þ
Adenocarcinoma þ
Carcinoid þ Indications for Ostomies
Colorectal cancer
The incidence of major morbidity associated with CRS/HIPEC is
Mucinous G1, G2, G3 þ
Intestinal þ well documented. Many have raised concern regarding anastomotic
Gastric cancer leaks following bowel resection and have raised questions as to the
Diffuse þ indicators protective proximal/defunctioning ostomies. Key studies to
Intestinal þ be considered in this regard are (Table III):
Ovarian cancer
Serous þ Verwaal et al. evaluated toxicity and complications in 102 patients with
Mucinous þ carcinomatosis of colorectal origin treated with cytoreductive
Diffuse malignant mesothelioma þ surgery and hyperthermic intra-peritoneal chemotherapy. The
median number of small bowel resections was 2 (range: 0–7).
RPD, randomly and proximally distributed; CRP, complete redistribution Cecum, colon and rectal resections were performed in 102, 41, and
phenomenon; WCD, widespread cancer dissemination.
53 patients, respectively with protective ostomy in 42% of the cases.
Despite the stoma bowel complication was reported in 17.6% of the
An aggressive complete peritonectomy could then be reserved for cases [8]. More recently, the same group of investigators conducted
generalized peritoneal carcinomatosis resultant from the complete a similar study in 103 patients with pseudomyxoma peritonei.
redistribution phenomenon (CRP) or widespread cancer dissemination The median number of resections was 4 (range: 0–8) and gastro-
(WCD) processes. For example, patients with malignant mesothelioma intestinal fistula was reported in 19 (18.4%) cases [9].
even in the presence of a localized disease would require a complete Shen et al. despite having found an unacceptable high rate of sepsis
resection of the parietal peritoneum along with any involved structures, correlated with bowel anastomoses, adopted a more flexible policy
based on the understanding that the origin of the disease is the suggesting the performance of protective stoma as an alternative
peritoneum itself and the pattern of distribution is characterized by CRP. [2]. They performed study in 77 (32 female and 45 male)
In pseudomyxoma peritonei the distribution characteristics may support patients with nonappendiceal colorectal cancer treated with
calls for a complete peritonectomy yet the relatively low ability to CRS and HIPEC. Protective ostomy was performed in 13% of the
implant argues for a more conservative approach confining the cases and bowel complication was reported in 17.6% of the
peritonectomy to where there is evidence of more solid disease. cases [10].
In the experience of Moran et al. the preservation of rectum is
Timing of Bowel Anastomoses: Pre- or important, where an anterior resection is required during the pelvic
Post-Hyperthermic Intraperitoneal peritonectomy, a proximal defunctioning stoma is necessary due to the
Chemotherapy (HIPEC) unacceptably high risks of anastomotic leakage. Alternatively if the
rectum can be preserved, a stoma can be avoided [11,12].
There is no consensus concerning the optimal timing for bowel To primarily perform unprotected colorectal anastomoses can be
anastomoses following cytoreductive surgery (CRS). Surgeons adopting considered a feasible option. Younan et al. reported results following

TABLE II. Timing of Bowel Anastomoses in Relation to HIPEC

Timing of
anastomoses Argument for Comment

Before HIPEC
Advantages Reduce costs and operation time [15] This justification is not supported by controlled evidence
Disadvantages Mechanical traction of viscera during the perfusion could impair This justification is not supported by controlled evidence
the integrity of the anastomoses
After HIPEC
Advantages Avoidance of potential adverse effects of heat and chemotherapy The evidence supporting this justification is weak. The influence of
on the suture healing CHT on the suture healing depends on the type of drug. In
animal studies, anastomotic healing can be impaired by
intraperitoneal MMC but not by 5-FU, at normal temperature
[16,17] or paclitaxel [18]. Local hyperthermia in itself has no
adverse effect on rat anastomotic healing [19]
Diminished risk of postoperative bowel complications Rate of bowel complication and or anastomotic leak between the
two approaches (before and after) are not substantially different
according to uncontrolled series (see the next table)
Possibility to treat the bowel margins against eventual implantation There is no data in literature reporting the risk of tumor relapse in
of tumor cells [20] the anastomoses
Disadvantages After the HIPEC all the visceral and parietal tissue are embedded This justification is not supported by controlled evidence
by the perfusate. Performing a suture in a bowel tissue with
edema could be technically more difficult and could also
hamper the quality of the anastomoses

Journal of Surgical Oncology


234 Kusamura et al.
TABLE III. Overview of Studies on Bowel Complications Associated With CRS þ HIPEC

Timing of Bowel
Mean No. of Protective anastomosis in F/A ratio Median Duration complication
References Predominant histology anastomosis/patient ostomy (%) relation to HIPEC (%) of operation (hrs) rate (%)

Jacquet et al. [21] Appendix, colon 1.8 None After 9.3 10.9b 17
Stephens et al. [4] PMP, colon NA NA After NA NA 7.5
Zanon et al. [22] Ovarian, gastric 0.7 NA Before 15 4–6 10
Elias et al. [23] Colon 2.6 8 7.3 18.8
Witkamp et al. [24] PMP 2 39 After 17.4 10b 34
Moran et al. [11] PMP NA NA NA NA 10.5 5
Elias et al. [25] PMP 2.8 NA After 7.2 10 22.2
Glehen et al. [5,26] Colon, PMP, ovarian 0.6 NA Before NA 6.1b 10.7
Elias et al. [27] Colon, PMP 2a NA After NA 8 10.3
Glehen et al. [28] Colon 0.4 NA Before 17.3 5.3b 8
Verwaal et al. [10] Colon 2 ¼ 69 42 After NA 7.5 17.6
>2 ¼ 33
Shen et al. [2] Colon 13 After NA 9 >3.9
Younan et al. [29] MP, PMP, ovarian 1 0.5 Before 11.3 8.2 10.8

No., number; HIPEC, heated intraperitoneal chemotherapy; F/A, fistula/anastomoses; PMP, pseudomyxoma peritonei; PM, peritoneal mesothelioma; NA,
information not available.
a
Median.
b
Mean.

203 procedures of CRS þ HIPEC undertaken for peritoneal surface 51% of the voters [13,14]. A detailed description of the methodology
malignancies of various origin. A total of 194 anastomoses were used for this consensus development is available elsewhere in this issue.
performed with a mean of 0.96 anastomoses per patient (range: 0–4). Presentation of the results and conference discussion occurred at the
In contrast to the other groups only one patient (0.5%) had a diverting Fifth International Workshop on Peritoneal Surface Malignancy held in
colostomy and overall 22 patients (10.8%) had bowel complications [29]. Milan, Italy, December 4–6, 2006.
There remains therefore considerable variation between surgeons
regarding the use/need for stomas. The objective of the expert panel
was to explore this area more thoroughly. RESULTS
The contents of the related scientific review document are outlined
in the Introduction of this paper. The conflicting points circulated
MATERIALS AND METHODS among the experts with their respective results of the first
In order to achieve a consensus among experts, the Delphi and second round voting were as follows:
methodology was employed [18]. Conflicting points were identified 1. Peritonectomy
and related multiple choice questions along with a scientific review
document on the topic were circulated among a panel of experts who had In case of limited disease dissemination to parietal peritoneum
a special interest in peritoneal surface oncology. Two rounds of web- in which one of the following situations could a partial parietal
based voting were carried out the second conducted after disclosing the peritonectomy restricted to the macroscopically involved regions be
results of the first round. Consensus was defined as the agreement of indicated? (more than one alternative allowed).

Disease Result voting first round (%) Result voting second round (%)

Pseudomyxoma peritonei 45.16 65.63


Cystoadenocarcinoma G1 of the appendix 74.19 81.25
Cystoadenocarcinoma G2, G3 of the appendix 67.74 78.13
Adenocarcinoma of the appendix 83.87 90.63
Carcinoid tumor of the appendix 83.87 87.50
Mucinous G1, G2, G3 colorectal cancer 70.97 78.13
Intestinal colorectal cancer 87.10 90.63
Diffuse gastric cancer 64.52 75.00
Intestinal gastric cancer 70.97 87.50
Serous ovarian cancer 80.65 96.88
Mucinous ovarian cancer 61.29 75.00
Diffuse malignant mesothelioma 29.03 46.88
There is no circumstance in the above-mentioned diseases in which a surgically 0.00 0.00
sparing approach could be considered. A complete parietal peritonectomy is
indicated in all these situations even in the presence of a localized disease,
irrespective of the primary tumor histology

Journal of Surgical Oncology


Consensus on Cytoreductive Surgery 235
2. Mesenteric disease but the second vote achieved consensus (65%). Interestingly
despite the knowledge that for mesothelioma the disease origin is
In the presence of numerous small metastatic nodules (<2.5 mm) in the peritoneum no consensus was achieved. This may reflect the
the mesentery, without infiltration, after the completion of cytoreduc- experience of the experts in observing that the limitation for achieving
tion in other areas which approach is best? complete cytoreduction is more related to the presence of disease on
the small bowel than the peritoneum.
Result All experts considered electroevaporization of metastatic disease
Result voting voting second (<2.5 mm) to be recommended even where HIPEC could be effective.
Alternative first round (%) round (%) Overall deferring anastomoses to follow HIPEC administration
gained consensus (53%). Not surprisingly the indication for protective
Cytoreduction with electroevaporization 90.32 100.00
ostomies proved difficult and reflected the variation in practice already
Sparing policy leaving these residues to be 9.68 0.00
treated by the intraperitoneal reported. The experts did not agree on the need for a stoma following
chemotherapy rectal resection preferring to opt for surgical discretion.
This has been the first attempt to gain consensus and attempt
to align techniques according to disease type and to standardize
3. Timing of anastomoses approaches where possible. Many areas remain for debate when
considering application of surgical techniques as part of CRS. There
In the closed abdomen technique for administration of hyperthermic remains a need to define the place and indication for second
intraperitoneal chemotherapy which is the optimal timing for intestinal cytoreduction for disease relapse and explore a place for rectosigmoid
anastomosis after CRS? preservation and nerve sparing peritonectomy in different peritoneal
surface malignancies.

Result REFERENCES
Result voting voting second
Alternative first round (%) round (%) 1. Sugarbaker PH: Peritonectomy procedures. Ann Surg 1995;221:
29–42.
Before the perfusion 35.48 34.38 2. Shen P, Hawksworth J, Lovato J, et al.: Cytoreductive surgery and
After the perfusion 48.39 53.13 intraperitoneal hyperthermic chemotherapy with mitomycin C for
The constructive procedure could be 16.13 12.50 peritoneal carcinomatosis from nonappendiceal colorectal carci-
performed at any time after the noma. Ann Surg Oncol 2004;11:178–186.
cytoreduction, without a significant 3. Kusamura S, Younan R, Baratti D, et al.: Cytoreductive surgery
impact on the morbidity and/or mortality followed by intraperitoneal hyperthermic perfusion: Analysis of
morbidity and mortality in 209 peritoneal surface malignancies
treated with closed abdomen technique. Cancer 2006;106:1144–
1153.
4. Ostomies 4. Stephens AD, Alderman R, Chang D, et al.: Morbidity and
mortality analysis of 200 treatments with cytoreductive surgery
Indications of protective proximal ostomies after CRS are not
and hyperthermic intraoperative intraperitoneal chemotherapy
uniform, with no consensus in the literature. When is a protective using the coliseum technique. Ann Surg Oncol 1999;6:790–796.
ostomy indicated? 5. Glehen O, Osinsky D, Cotte E, et al.: Intraperitoneal chemo-
hyperthermia using a closed abdominal procedure and cytor-
eductive surgery for the treatment of peritoneal carcinomatosis:
Morbidity and mortality analysis of 216 consecutive procedures.
Result Ann Surg Oncol 2003;10:863–869.
Result voting voting second 6. Deraco M, Santoro N, Carraro O, et al.: Peritoneal carcinoma-
Alternative first round (%) round (%) tosis: Feature of dissemination. A review. Tumori 1999;85:1–5.
7. Bibi R, Pranesh N, O’Dwyer ST, et al.: A specific cadherin
Whenever a rectal resection is performed 22.58 12.50 phenotype may characterise the disseminating yet non-metastatic
In cases of low anterior resections in which the 32.26 21.88 behaviour of pseudomyxoma peritonei. Br J Cancer 2006;95:
preservation of the rectum is not possible 1258–1264.
In case of ileo-rectal anastomoses 29.03 15.63 8. Verwaal VJ, van Tinteren H, Ruth SV, et al.: Toxicity of
The policy for protective stoma could be 61.29 87.50 cytoreductive surgery and hyperthermic intra-peritoneal chemo-
flexible and the procedure done at the therapy. J Surg Oncol 2004;85:61–67.
surgeon’s discretion; the performance of 9. Smeenk RM, Verwaal VJ, Zoetmulder FA: Toxicity and mortality
unprotected colorectal anastomoses does of cytoreduction and intraoperative hyperthermic intraperitoneal
not influence the morbidity and mortality chemotherapy in pseudomyxoma peritonei—A report of 103
significantly procedures. Eur J Surg Oncol 2006;32:186–190; Epub 2005 Nov
21.
10. Verwaal VJ, van Tinteren H, Ruth SV, et al.: Toxicity of
cytoreductive surgery and hyperthermic intra-peritoneal chemo-
DISCUSSION AND COMMENTS therapy. J Surg Oncol 2004;85:61–67.
Using the Delphi methodology consensus was achieved in all areas 11. Moran BJ, Cecil TD: The etiology, clinical presentation, and
explored and for each alternative option presented except for management of pseudomyxoma peritonei. Surg Oncol Clin N Am
2003;12:585–603.
peritonectomy in mesothelioma. In all areas second round voting 12. Sugarbaker PH, Ronnett BM, Archer A, et al.: Pseudomyxoma
achieved greater consensus. peritonei syndrome. Adv Surg 1997;30:233–280.
With regard to peritonectomy, the greatest consensus was achieved 13. Loughlin KG, Moore LF: Using Delphi to achieve congruent
for serous ovarian, appendiceal and intestinal colorectal adeno- objectives and activities in a pediatrics department. J Med Educ
carcinoma. For PMP the first round demonstrated variation in opinion 1979;54:101–106.

Journal of Surgical Oncology


236 Kusamura et al.
14. Keeney S, Hasson F, McKenna H: Consulting the oracle: Ten abdomen and previously performed anastomosis. Int J Hyper-
lessons rom using the Delphi technique in nursing research. J Adv thermia 2001;17:456–464.
Nurs 2006;53:205–212. 23. Elias D, Blot F, El Otmany A, et al.: Curative treatment of
15. Zanon C, Clara R, Bortolini M, et al.: Chemohyperthermia for peritoneal carcinomatosis arising from colorectal cancer by
advanced abdominal malignancies: A new procedure with closed complete resection and intraperitoneal chemotherapy. Cancer
abdomen and previously performed anastomosis. Int J Hyper- 2001;92:71–76.
thermia 2001;17:456–464. 24. Witkamp AJ, de Bree E, Kaag MM, et al.: Extensive surgical
16. Kuzu M, Koksoy C, Kale T, et al.: Experimental study of the cytoreduction and intraoperative hyperthermic intraperitoneal
effect of preoperative 5-fluorouracil on the integrity of colon chemotherapy in patients with pseudomyxoma peritonei. Br
anastomosis. Br J Surg 1998;85:236–239. J Surg 2001;88:458–463.
17. Fumagalli U, Trabucchi E, Soligo M, et al.: Effects of intra- 25. Elias D, Laurent S, Antoun S, et al.: Pseudomyxoma peritonei
peritoneal chemotherapy on anastomotic healing in rats. J Surg treated with complete resection and immediate intraperitoneal
Res 1991;50:82–87. chemotherapy. Gastroenterol Clin Biol 2003;27:407–412.
18. Arikan AY, Gunal O, Pehlivan M, et al.: The effect of intra- 26. Glehen O, Mithieux F, Osinsky D, et al.: Surgery combined with
peritoneal paclitaxel administration on colonic anastomosis. peritonectomy procedures and intraperitoneal chemohyperther-
Hepatogastroenterology 2000;47:1273–1276. mia in abdominal cancers with peritoneal carcinomatosis: A
19. Shimizu T, Maeta M, Koga S: Influence of local hyperthermia on phase II study. J Clin Oncol 2003;21:799–806.
the healing of small intestinal anastomoses in the rat. Br J Surg 27. Elias D, Matsuhisa T, Sideris L, et al.: Heated intra-operative
1991;78:57–59. intraperitoneal oxaliplatin plus irinotecan after complete resection
20. Elias D, Sideris L, Pocard M, et al.: Efficacy of intraperitoneal of peritoneal carcinomatosis: Pharmacokinetics, tissue distribu-
chemohyperthermia with oxaliplatin in colorectal peritoneal tion and tolerance. Ann Oncol 2004;15:1558–1565.
carcinomatosis. Preliminary results in 24 patients. Ann Oncol 28. Glehen O, Cotte E, Schreiber V, et al.: Intraperitoneal chemo-
2004;15:781–785. hyperthermia and attempted cytoreductive surgery in patients
21. Jacquet P, Stephens AD, Averbach AM, et al.: Analysis of with peritoneal carcinomatosis of colorectal origin. Br J Surg
morbidity and mortality in 60 patients with peritoneal carcino- 2004;91:747–754.
matosis treated by cytoreductive surgery and heated intraoper- 29. Younan R, Kusamura S, Baratti D, et al.: Bowel complications in
ative intraperitoneal chemotherapy. Cancer 1996;77:2622–2629. 203 cases of peritoneal surface malignancies treated with
22. Zanon C, Clara R, Bortolini M, et al.: Chemohyperthermia for peritonectomy and closed-technique intraperitoneal hyperthermic
advanced abdominal malignancies: A new procedure with closed perfusion. Ann Surg Oncol 2005;12:910–918; Epub 2005 Sep 21.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:237–241

Postoperative Residual Disease Evaluation in the Locoregional Treatment of


Peritoneal Surface Malignancy

SANTIAGO GONZÁLEZ-MORENO, MD, PhD,1* SHIGEKI KUSAMURA, MD, PhD,


2

DARIO BARATTI, MD,2 AND MARCELLO DERACO, MD2


1
Department of Surgical Oncology, Peritoneal Surface Oncology Program, Centro Oncológico MD Anderson International España, Madrid, Spain
2
Surgical Department, Peritoneal Surface Malignancy Program, IRCCS Istituto Nazionale dei Tumori Milano, Italy

Background: The maximum size of the residual lesions left behind after cytoreductive surgery for peritoneal surface malignancy has consistently
been shown to be the main prognostic factor in this setting. However, a uniform assessment method and categorization for this paramount
prognostic indicator is lacking.
Methods: In order to achieve a consensus among experts, the Delphi methodology was employed. Conflicting points were identified and related
multiple choice questions were circulated among a panel of experts on peritoneal surface oncology. Two rounds of web-based voting were carried
out.
Results: The completeness of cytoreduction (CC) Score described by Sugarbaker was considered the current best classification for residual
disease size. The experienced surgeon’s naked-eye estimation was considered the ideal method to assess residual disease size. There was
agreement that the definition of CC-0 or R0 cytoreduction needs further specification. A redefinition of ‘‘completeness of cytoreduction’’
according to disease process was favored by the experts but not favored for the type of intraperitoneal chemotherapy employed.
Conclusions: Following the experts’ consensus, it is recommended that the CC score be used to categorize residual disease after cytoreductive
surgery for peritoneal surface malignancy. Pending issues for further consensus development in this area have been identified.
J. Surg. Oncol. 2008;98:237–241. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal carcinomatosis; cytoreductive surgery; prognosis; residual disease

INTRODUCTION of high precision, such as microscope, which is usually not available in


the operating room. The justification behind the cut-off value of 5 mm
The maximum size of residual disease remaining after surgical adopted by other groups [2] does not seem to be consistent with
cytoreduction has been shown to predict prognosis in patients with this rationale. Even when the limits defining the optimal residual
cancerous peritoneal dissemination treated with cytoreductive surgery disease size should be based on the intratumoral penetration ability of
and perioperative intraperitoneal chemotherapy. intraperitoneal cisplatin, the cut-off of 2.1–2.5 mm does not hold
Based on this residual lesion size, cytoreductive procedures have true for other chemotherapies, frequently used for intraperitoneal
been categorized as complete or incomplete. Results of several hyperthermic perfusion. Drugs such as doxorubicin, carboplatin and
studies [1–3] have shown a direct relation between the completeness of oxaliplatin have much lower intratumoral penetration capacity.
cytoreductive surgery and survival for peritoneal dissemination from The reproducibility of the methods used for postoperative residual
all primary cancer locations (ovary, stomach, colon, appendix), disease measurement is also of concern, as it has been demonstrated
including the peritoneum itself. However, although this finding is that intraoperative estimation of tumor size shows a significant
very consistent across groups and studies, there is a wide variation in interobserver variability. Prefontaine et al. used a simulated patient
the methodology employed by different authors to assess residual model to evaluate surgeon accuracy in estimating tumor size [15].
disease size and subsequently completeness of cytoreduction. The They found that the range of estimates was very wide. More surgeons
different residual disease classifications currently employed are shown underestimated rather than overestimated tumor diameters. He
in Figure 1. estimated, given these results, that 25% and 20% of suboptimal
Sugarbaker and colleagues found that prognosis can be estimated by and optimal patients, respectively, were erroneously categorized.
completeness of cytoreduction. For colon cancer there is a 40% chance Although such study was conducted with gynecologic oncologists, it is
of survival at 5 years in those who undergo complete cytoreduction intuitively expectable that at least the same level of inaccuracy and/or
versus 0% survival for the incomplete cytoreduction category [1,2]. lack of reproducibility could occur among peritoneal surface oncology
Numerous other groups have confirmed that complete cytoreduction is experts in the intraoperative assessment of residual tumor, unless it is
a requirement for long-term survival after treatment of peritoneal done by sterile rule or calliper at surgery.
carcinomatosis from appendiceal, colorectal and gastric cancer and
peritoneal mesothelioma [4–12]. The author has no financial interest related to the contents of this article to
There are several questions and inconsistencies that could be raised disclose.
to the currently available residual disease classification systems. One *Correspondence to: Dr. Santiago González-Moreno, MD, PhD, Calle
of them is the precision of the limits between optimal and suboptimal Gómez Hemans 2, 28033 Madrid Spain. Fax:þ34-91-768-0681.
residual tumor size. According to CC score proposed by Sugarbaker E-mail: sgonzalez@mdanderson.es
the cut-off value is 2.5 mm and represents the maximum intratumoral Received 19 March 2008; Accepted 27 March 2008
penetration ability of cisplatin when administered intraperitoneally DOI 10.1002/jso.21072
[13,14]. Obviously the value was obtained in well-controlled ex- Published online in Wiley InterScience
perimental conditions, with the employment of measuring instruments (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


238 González-Moreno et al.

Fig. 1. Residual disease size classifications currently found in the peritoneal surface oncology literature.

Furthermore, the definition of a CC-0 or R0 cytoreduction might ‘‘complete cytoreduction’’ might need to be reconsidered according to
need additional specifications. Both conceptually and in the clinical, the clinical situation and disease process.
operating room setting, a 12-hr cytoreduction down to ‘‘no visible For all the above reasons, the need for a consensus and further
tumor nodules’’ might not equal a CC-0 or R0 resection if we start with reflection on residual disease evaluation are warranted.
a large volume, widespread peritoneal disease (i.e. Peritoneal Cancer
Index (PCI) above 31 out of 39). In these cases, a very carefully MATERIALS AND METHODS
directed inspection might likely reveal tiny remaining nodules, and at
the very least microscopic tumor could be expected to remain. Some In order to achieve a consensus among experts, the Delphi
surgeons consider this situation a CC-1 cytoreduction, despite the methodology was employed [18]. Conflicting points were identified
inability by most witnesses in the operating room to see any remaining and related multiple choice questions along with a scientific review
tumor nodule, whereas other surgeons will not hesitate to label it as a document on the topic were circulated among a panel of experts on
CC-0 resection. peritoneal surface oncology. Two rounds of web-based voting,
Finally, although no formal statement in the literature is available, it the second one after disclosing the results of the first round, were
is thought that the definition of complete versus incomplete carried out. A detailed description of the methodology used for this
cytoreduction may vary with the histological type of the malignancy. consensus development is available elsewhere in this issue. Presenta-
For example, mucinous tumors are well penetrated by intraperitoneal tion and discussion of the results were done at the Fifth International
chemotherapy solutions. With minimally invasive mucinous tumors Workshop on Peritoneal Surface Malignancy held in Milan, Italy,
such as pseudomyxoma peritonei, complete cytoreduction may occur December 4–6, 2006.
in the combined treatment plan with tumor nodules up to a full The contents of the related scientific review document are outlined
centimetre in size. Likewise, Sebbag and cols [16]. found that patients in the Introduction section. The accompanying related questions
affected by peritoneal mesothelioma undergoing a CC-2 cytoreduction circulated among the experts were the following:
with perioperative intraperitoneal chemotherapy could be grouped
along with the CC-0 and CC-1 categories, also benefiting from a long- (1) Which one is the current best method to assess the residual disease
term survival. This finding holds true in the recently updated series after cytoreductive surgery in patients affected by peritoneal
from the same institution [17]. In contrast, hard fibrotic non-mucinous surface malignancies?
colon cancer is poorly penetrated by chemotherapy solutions. Only (a) AJCC/UICC R classification [19] with Dutch modification (R0
cytoreduction down to no visible evidence of disease would be and R1: no macroscopic; R2a  2.5 mm and R2b > 2.5 mm)
expected to result in long-term survival with a sclerotic malignant [20];
process. (b) AJCC/UICC R classification [19] with French modification
Also, some cancers may be remarkably more responsive to (R0 and R1: no macroscopic; R2: macroscopic disease) [21];
chemotherapy than others. This is likely the case of a majority of (c) AJCC/UICC R classification [19] with American modification
ovarian cancers. Their complete response to systemic chemotherapy (R0: no gross disease with negative microscopic margins; R1:
is also frequently seen with intraperitoneal chemotherapy solutions or no gross disease with positive microscopic margins; R2a:
a bidirectional (intraperitoneal combined with intravenous chemo- <5 mm, R2b: tumors of 6–20 mm and R2c: tumors of
therapy) approach. All these situations show that ‘‘completeness >20 mm) [2];
of cytoreduction’’ is a dynamic concept and that the definition of (d) Completeness of Cytoreduction Score [22].

Journal of Surgical Oncology


Residual Disease After Cytoreductive Surgery 239
(2) What should be the ideal method to assess residual disease size RESULTS
after cytoreductive surgery?
(a) The experienced surgeon’s naked-eye estimation; Results of the web-based voting can be found in Table I.
(b) Direct measurement by a sterile rule or calliper;
(c) The average of the naked-eye estimation of all members of the Best Classification for Residual Disease
operating team.
(3) Consider the case of a young patient affected by pseudomyxoma Sugarbakeŕs Completeness of Cytoreduction Score (so-called ‘‘CC
peritonei undergoing cytoreductive surgery. Initial PCI is 35, score’’) was overwhelmingly considered in the second voting round
operating time including hyperthermic intraoperative chemother- the best classification by 93.75% of the experts. In the first voting this
apy (HIPEC) is 11 hr, and there are no apparent visible disease option was also the most voted obtaining 74.19% of the votes. All
nodules at the end of a very laborious cytoreductive procedure. advocates of the AJCC/UICC classification with French or American
What is your completeness of cytoreduction scoring? modification, and some of the Dutch modification in the first round
(a) CC-0; gave their support to the CC score in the second round.
(b) CC-1;
(c) other. Ideal Method to Assess Residual Disease
(4) Consider the same case. What is your R scoring?
(a) R0; Experts considered that subjective assessment of the residual
(b) R1; disease size was acceptable and preferable over a more objective
(c) other. method like direct measurement by a ruler. In the first voting, the
(5) Would you favor a further specification in the definition of a CC-0 naked-eye estimation of an experienced surgeon and the average of
or R0 cytoreduction? the estimations done by all members of the operating team obtained
(a) yes; similar results, but in the second round the naked-eye estimation by
(b) no. the experienced surgeon gained a significantly better support with
(6) Would you favor a redefinition of the completeness of cytoreduc- 65.63% of the votes compared to the other two options.
tion according to the disease process?
(a) yes; Clinical Case
(b) no.
(7) Would you favor a redefinition of the completeness of cytoreduc- In the above-described case, final voting results labeled the
tion according to the type of intraperitoneal cytotoxic agent(s) cytoreduction as CC-0 and R1. Regarding the CC-score, there were
employed? no significant changes in voting between the first and second rounds.
(a) yes; However, for the R classification, an initial tie between the R0 and R1
(b) no. subsequently gave way to a very significant majority (75%) supporting

TABLE I. Results of the web-based voting of the panel of experts

Votes (%) Votes (% second


Question Possible answers first round round

Question 1
Best residual disease classification system AJCC/UICC (Dutch) 9.68 6.25
AJCC/UICC (French) 12.9 0
AJCC/UICC (American) 3.23 0
CC Score 74.19 93.75
Question 2
Ideal method to assess residual disease size Expert surgeon’s naked eye 38.71 65.63
Ruler or calliper 25.84 18.75
Average naked-eye estimation 35.48 15.63
Question 3
Clinical case, CC score CC-0 70.97 75
CC-1 25.81 25
Other 3.23 0
Question 4
Clinical case, R score R0 48.39 25
R1 48.39 75
Other 3.23 0
QUESTION 5
Need for redefinition of CC-0 or R0 Yes 58.06 75
No 41.94 25
QUESTION 6
Need for redefinition of CC according to disease Yes 54.84 78.13
No 45.16 21.88
Question 7
Need for redefinition of CC according to Yes 29.03 21.88
chemotherapy
No 70.97 78.13

CC, completeness of cytoreduction.

Journal of Surgical Oncology


240 González-Moreno et al.
the R1 option. This was the result of initial R0 proponents changing apart groups of patients with different prognosis. However, for the
their vote to R1 in the second round. great majority of experts that participated in this consensus, the CC
score proposed by Sugarbaker was considered the best residual
Need for Further Specification of disease classification. Several reasons could justify this election. The
cut-off value of 2.5 mm is well established in the literature and
CC-0 or R0 Definition corresponds to the maximum intratumoral penetration of intraperito-
The second voting round consolidated and increased the support for neal cisplatin. Although other drugs vary in their penetration capacity,
such a redefinition, already seen in the first round. none of them reach the 5 mm employed in other classifications as
the limit between the first two prognostic groups. Following this
rationale, heterogeneity in drug intratumoral penetration capacity
Need for Redefinition of Completeness of could determine different residual disease size cut-off values according
Cytoreduction According to the Disease Process to the drugs employed, but this was not considered a need by the
participating experts.
Such a redefinition was clearly favored (78.13% vs. 21.88%), and
Secondly, the CC score was specifically designed for this purpose:
differences between voting rounds were very similar to the ones
delineation of prognostic groups following cytoreductive surgery and
observed in the previous question.
perioperative intraperitoneal chemotherapy for peritoneal surface
malignant diseases. However, the R classification (AJCC/UICC) was
Need for Redefinition of Completeness of Cytoreduction created to assess residual disease in a different context, i.e. surgical
According to Chemotherapy Agent(s) Employed resection of a tumor mass, not of diffuse peritoneal involvement.
Actually, the original AJCC/UICC definition does not consider
A clear majority of experts (78.13%) rejected such a possibility, different residual disease size values within the R2 (macroscopic
with similar results in the first and second voting rounds. disease) category, a factor which we know has a key impact on
survival. For this reason different groups (except for the Lyon group)
DISCUSSION created their own subdivisions within the R2 category (Table I), adding
further confusion to this issue.
The radical treatment of peritoneal surface malignancies consists of In the third place, the AJCC/UICC R classification includes an
an inseparable combination of cytoreductive surgery and perioperative ‘‘R1’’ category, defined as microscopic residual disease. However,
intraperitoneal chemotherapy. Neither component of this therapy could because of the nature of the procedure and of the underlying neoplastic
achieve the same result alone, and need each other for optimal results. disease, microscopic residual disease is expected to be present after
Therefore, residual disease size left behind after a maximum virtually all cases of cytoreductive surgery, providing the rationale
cytoreductive effort should ideally be small enough so that the limited for the employment of regional chemotherapy. Following this line of
penetration of intraperitoneal chemotherapy could result in complete reasoning, the group from Lyon advocates using a joint R0/R1 category
cancer eradication. This criterion is the basis of the definition of a within the R classification. Furthermore, microscopic assessment
complete cytoreduction, the main goal to achieve in this therapy, and of resection margins in the numerous peritonectomy and visceral
should be honored in any attempt or proposal to establish disease size resection specimens rendered by these procedures would be an impos-
cut-off values that make up different prognostic groups. sible endeavour for a pathologist. For these reasons, a microscopic
Given the crucial importance of the measurement of residual tumor residual disease category would not need to be included in a residual
nodules, and being well aware of the high interobserver variability in disease classification after cytoreductive surgery for peritoneal surface
this topic, as demonstrated by Prefontaine and colleagues [15], experts malignancy, just as it occurs in the CC score. Putting it in different
still agreed on not using an objective measurement instrument for this words, all CC-0 cytoreductions would implicitly be CC-0/R1
purpose, but rather rely on their own naked-eye estimation. procedures, and CC-0/R0 scoring would be achieved after en-bloc
All residual disease classifications considered, although hetero- resection of a primary tumor, but rarely after a cytoreductive procedure
geneous in definition criteria, have demonstrated their ability to place for peritoneal carcinomatosis. This was reflected in the experts’

Fig. 2. A possible proposal for a redefinition of completeness of cytoreduction according to disease process. Such redefinition was favored by
the panel of experts who participated in the consensus. For each disease, the vertical line to the right of its name sets the division between complete
and incomplete cytoreduction.

Journal of Surgical Oncology


Residual Disease After Cytoreductive Surgery 241
answers to the proposed clinical case (questions 3 and 4), where an mia in abdominal cancers with peritoneal carcinomatosis: A
extensive cytoreduction down to no visible tumor nodules (the most phase II study. J Clin Oncol 2003;21:799–806.
complete we could consider) was categorized as CC-0 but R1, and 6. Sugarbaker PH, Jablonski KA: Prognostic features of 51 color-
subsequently a majority agreed on the need to further specify the ectal and 130 appendiceal cancer patients with peritoneal
definition of a CC-0 or R0 cytoreduction (question 5), although carcinomatosis treated by cytoreductive surgery and intraperito-
neal chemotherapy. Ann Surg 1995;221:124–132.
this was not formulated. It is possible that the definition of a true CC-0/ 7. Elias D, Blot F, El Otmany A, et al.: Curative treatment of
R0 cytoreduction should incorporate a statement alluding to the peritoneal carcinomatosis arising from colorectal cancer by
predictable absence of microscopic disease (or margins) and be complete resection and intraperitoneal chemotherapy. Cancer
reserved for situations with no visible disease in cases with a very low 2001;92:71–76.
initial Peritoneal Cancer Index, similar to the situation encountered 8. Witkamp AJ, de Bree E, Kaag MM, et al.: Extensive
after an en-bloc resection of a primary tumor. cytoreductive surgery followed by intra-operative hyperthermic
As outlined in the related document circulated among our experts intraperitoneal chemotherapy with mitomycin-C in patients with
and included in the above introduction, the different biological peritoneal carcinomatosis of colorectal origin. Eur J Cancer 2001;
behaviour and chemosensitivity patterns of the different neoplasms 37:979–984.
9. Sugarbaker PH: Results of treatment of 385 patients with
grouped as ‘‘peritoneal surface malignancies’’ could eventually result peritoneal surface spread of appendiceal malignancy. Ann Surg
in considering different residual disease classifications for each Oncol 1999;6:727–731.
disease (Fig. 2). Intuitively this is a reasonable, plausible statement 10. Verwaal VJ, van Tinteren H, Ruth SV, et al.: Toxicity of
and, although no scientific confirmation is available, a majority of our cytoreduction and hyperthermic intra-peritoneal chemotherapy.
experts actually favored such a redefinition. The only published J Surg Oncol 2004;85:61–67.
evidence for this comes from the peritoneal mesothelioma series 11. Yonemura Y, Fujimura T, Nishimura G, et al.: Effects of intra-
results published by the Washington Cancer Institute group, whereby operative chemohyperthermia in patients with gastric cancer with
cases with residual disease nodules up to 2.5 cm in size (equivalent to a peritoneal dissemination. Surgery 1996;119:437–444.
CC-2 cytoreduction) were included in the more favorable survival 12. Deraco M, Nonaka D, Baratti D, et al.: Prognostic analysis of
clinicopathologic factors in 49 patients with diffuse malignant
group along with CC-0 and CC-1 cases (thereby somehow considered peritoneal mesothelioma treated with cytoreductive surgery and
complete cytoreductions), in contrast with the pseudomyxoma or intraperitoneal hyperthermic perfusion. Ann Surg Oncol 2006;13:
colorectal carcinomatosis experience by the same group where CC-2 229–237.
cases are grouped with CC-3 cases in the incomplete cytoreduction 13. Los G, Verdegaal EM, Mutsaers PH, et al.: Penetration of
category. This has not been confirmed by other groups. However, it is carboplatin and cisplatin into rat peritoneal tumor nodules after
possible that in the future new, more active drugs will become available intraperitoneal chemotherapy. Cancer Chemother Pharmacol
and that their use might help modulate the cytoreductive effort 1991;28:159–165.
necessary for an optimal combined treatment result, raising the need to 14. McVie JG, Dikhoff T, Van der Heide J, et al.: Tissue concen-
consider a change in terminology from ‘‘complete’’ cytoreduction to tration of platinum after intraperitoneal cisplatinum administra-
tion in patients. Proc Am Assoc Cancer Res 1985;26:162.
‘‘optimal’’ or ‘‘adequate’’ cytoreduction. 15. Prefontaine M, Gelfand AT, Donovan JT, et al.: Reproducibility of
tumor measurements in ovarian cancer: A study of interobserver
REFERENCES variability. Gynecol Oncol 1994;55:87–890.
16. Sebbag G, Yan H, Shmookler BM, et al.: Results of treatment of
1. Elias D, Blot F, El Otmany A, et al.: Curative treatment of 33 patients with peritoneal mesothelioma. Br J Surg 2000;87:
peritoneal carcinomatosis arising from colorectal cancer by 1587–1593.
complete resection and intraperitoneal chemotherapy. Cancer 17. Yan TD, Brun EA, Cerruto CA, et al.: Prognostic indicators for
2001;92:71–76. patients undergoing cytoreductive surgery and perioperative
2. Shen P, Levine EA, Hall J, et al.: Factors predicting survival after intraperitoneal chemotherapy for diffuse malignant peritoneal
intraperitoneal hyperthermic chemotherapy with mitomycin C mesothelioma. Ann Surg Oncol 2007;14:41–49.
after cytoreductive surgery for patients with peritoneal carcino- 18. Jones J, Hunter D: Consensus methods for medical and health
matosis. Arch Surg 2003;138:26–33. services research. Br Med J 1995;311:376–380.
3. Witkamp AJ, de Bree E, Kaag MM, et al.: Extensive 19. American Joint Committee on Cancer: ‘‘Manual for Staging on
cytoreductive surgery followed by intra-operative hyperthermic Cancer’’. 4th ed. Philadelphia: Lippincott; 1992.
intraperitoneal chemotherapy with mitomycin-C in patients with 20. Verwaal VJ, van Ruth S, Witkamp A, et al.: Long-term survival of
peritoneal carcinomatosis of colorectal origin. Eur J Cancer 2001; peritoneal carcinomatosis of colorectal origin. Ann Surg Oncol
37:979–984. 2005;12:65–71.
4. Glehen O, Kwiatkowski F, Sugarbaker PH, et al.: Cytoreductive 21. Glehen O, Mithieux F, Osinsky D, et al.: Surgery combined
surgery combined with perioperative intraperitoneal chemo- with peritonectomy procedures and intraperitoneal chemohyper-
therapy for the management of peritoneal carcinomatosis from thermia in abdominal cancers with peritoneal carcinomatosis: A
colorectal cancer: A multi-institutional study. J Clin Oncol 2004; phase II study. J Clin Oncol 2003;21:799–806.
22:3284–3292. 22. Jacquet P, Sugarbaker PH: Clinical research methodologies in
5. Glehen O, Mithieux F, Osinsky D, et al.: Surgery combined with diagnosis and staging of patients with peritoneal carcinomatosis.
peritonectomy procedures and intraperitoneal chemohyperther- Cancer Treat Res 1996;82:359–374.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:242–246

Hyperthermic Intraperitoneal Chemotherapy:


Nomenclature and Modalities of Perfusion

OLIVIER GLEHEN, MD, PhD,1,2* EDDY COTTE, MD,1,2 SHIGEKI KUSAMURA, MD, PhD,3 MARCELLO DERACO, MD,3
DARIO BARATTI, MD,3 GUILLAUME PASSOT, MD,1,2 ANNIE-CLAUDE BEAUJARD, MD,2,4
1,2
AND GILLY FRANCOIS NOEL, MD, PhD,
1
Department of Oncologic surgery, Centre Hospitalo-Universitaire Lyon Sud, Pierre Bénite Cedex, France
2
EA 3738, UCBL, Faculté de médicine Lyon Sud, Oullins Cedex, France
3
Fondazione IRCCS Istituto Nazionale dei Tumori Milano, Milan, Italy
4
Department of Anesthesiology, CHLS—HCL, Pierre Bénite Cedex, France

Following international consensus, HIPEC should be the acronym used in the scientific literature to refer to the hyperthermic intraperitoneal
chemotherapy. Several modalities of perfusion are used to deliver HIPEC: open abdominal technique (Coliseum), closed abdominal technique,
peritoneal cavity expander, semi-opened abdominal technique. There is no sufficient evidence in literature confirming the superiority of one
technique over the others in terms of outcome, morbidity and safety to the personnel of the operating theatre. Each option has its own operational
advantages and disadvantages and future prospective studies must be conducted to establish which one is the best alternative. Today, the best
technique is the one which is routinely used and improved into each specialized institution involved in the management of peritoneal surface
malignancy.
J. Surg. Oncol. 2008;98:242–246. ß 2008 Wiley-Liss, Inc.

KEY WORDS: hyperthermia; peritoneal carcinomatosis; techniques; intraperitoneal chemotherapy

INTRODUCTION Modalities of Perfusion


Patients with peritoneal carcinomatosis have long been considered Early postoperative intraperitoneal chemotherapy. Early post-
as a terminal condition with no curative options. Over the past decade, operative intraperitoneal chemotherapy (EPIC) is delivered by a
novel therapeutic approaches to peritoneal surface malignancies Tenckhoff catheter or by a subcutaneous port placed through the
have emerged. Loco-regional treatments including cytoreductive abdominal wall in the approximate area at the greatest risk of
surgery and peritonectomy procedures for the macroscopic disease in recurrence after cytoreductive surgery. Closed suction drains are
combination with perioperative intraperitoneal chemotherapy for placed in dependant areas in the pelvis and below each hemi-
the microscopic residual disease have been developed for this loco- diaphragm. Intraperitoneal chemotherapy is administered postoper-
regional disease. There are different modalities for perioperative atively on postoperative days 1–5, but can be initiated immediately
intraperitoneal chemotherapy administration. Most of peritoneal postoperatively and continued in the outpatient setting [3].
surface malignancy treatment centers exclusively use hyperthermic EPIC has the advantages to administer multiple cycles of
intraperitoneal chemotherapy (HIPEC), some others only early post- chemotherapy. During each treatment, the chemotherapeutic drug is
operative administration and others use both sequentially. Several not drained for at least 24 hr, to increase the duration of exposure of
devices or technologies of HIPEC have been described and are tumor cells to therapy.
currently used into specialized centers. Although the use of HIPEC has But it also has inconvenient: because the chemotherapeutic
gained wider acceptance, the specifics of its administration lack agents persist in the peritoneum cavity, there is a greater opportunity
uniformity. Debate on the best technique for HIPEC is still open. In this for significant systemic absorption and adverse effects. This can be
review, after the report of consensus on nomenclature, the methodo- partially overcome by the use of drugs that have a high first-pass effect
logy, benefits, and risks associated with each technique are discussed. after portal absorption such as 5-fluorouracil (5-FU) which is the most
common drug used with this technique [4]. Some other toxicity
related to catheter complications (infections, bowel obstruction) has
Standardized Nomenclature been reported. EPIC was found to significantly increase the rate of
postoperative complications in the large multicentric retrospective
Multiple names and its acronyms have been used into scientific
literature to refer to the intraoperative administration of hyperthermic
intraperitoneal chemotherapy, often causing confusion. The Dutch The authors have no financial interest related to the contents of this article to
group [1] proposed in 2004 during the 4th Workshop on Peritoneal disclose.
Surface Malignancies held in Madrid, the use of the term ‘‘HIPEC.’’ *Correspondence to: Dr. Olivier Glehen, MD, PhD, Department of surgery,
The Workshop participants were already in favor of its acronym [2]. CHLS, 69495 Pierre Bénite Cedex, France. Fax: 00(33)478 863 343.
The results of the web based voting confirmed this consensus and E-mail: olivier.glehen@chu-lyon.fr
‘‘HIPEC’’ was choosed by 61.29% and 78.1% of experts during the Received 17 March 2008; Accepted 21 March 2008
first and the second round of the vote, respectively. So this acronym DOI 10.1002/jso.21061
should be used to refer to the hyperthermic intraperitoneal chemo- Published online in Wiley InterScience
therapy into scientific literature and communication. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


HIPEC: Modalities of Perfusion 243
study including 504 patients with colorectal carcinomatosis
treated with cytoreductive surgery combined with perioperative
intraperitoneal chemotherapy [5]. Its efficacy is limited by adhesions
that can result in pooling of the chemotherapeutic drugs in
intraperitoneal loculations. Not only does this sequester the treatment
from tumor cells, loculations also result in exposure of normal tissue
to high concentrations of drug, thereby adding to the morbidity of the
therapy [6].
EPIC do not involve hyperthermia. However heat has been shown to
be cytotoxic in vitro at 42.58C [7], and hyperthermia has been shown to
enhance the antitumor effect of agents such as oxapliplatin, mitomycin,
doxorubicin, and cisplatin, by augmenting cytotoxicity and increasing
the penetration of drugs into tissue [8–10]. Moreover, Elias et al.
recently compared two groups of patients with colorectal carcinoma-
tosis with characteristics as similar as possible. One was treated with
EPIC using 5-FU and mitomycin C and one with HIPEC using
oxaliplatin at 438C. All results were in favor of HIPEC group: Fig. 1. The coliseum apparatus. [Color figure can be viewed in the
mortality, morbidity, rate of peritoneal recurrence which was twice in online issue, available at www.interscience.wiley.com.]
EPIC group and overall survival [11].
Thus, EPIC may be used in the treatment of microscopic residual
peritoneal disease, but following HIPEC which seems to be more allowed an immediate thermal homogeneity, but the expander
efficient, with an increased risk of postoperative complications. isolated the abdominal wall from the instillate, resulting in early
HIPEC. To take advantage of the synergistic effect of chemo- parietal peritoneal recurrence. The use of coliseum technique was
therapy and hyperthermia, several different HIPEC devices to enable identified into this single institution as the best technique in terms of
intraoperative perfusion of the peritoneal cavity with hyperthermic thermal homogeneity and spatial diffusion. Because the surgeons can
chemotherapy have been developed. Constant hyperthermia is ob- manipulate the intra-abdominal viscera during perfusion, all peritoneal
tained by a closed continuous circuit, with pump, heater, heat surfaces are equally exposed to therapy. Furthermore, excessive
exchanger, and real-time temperature monitoring. Open circuit without heating of normal tissue that can exacerbate post-operative ileus and
recirculation and reheating of the instillate should be avoided [8,9]. To increase the incidence of postoperative perforation or fistula formation
avoid systemic hyperthermia during the perfusion procedure, core is avoided [15].
temperature have to be not more than 34–358C at the beginning of the Disadvantage of this technique is that the open abdomen naturally
perfusion. Precooling can be obtained by using cooling hat, legs or leads to heat dissipation at the surface of the instillate, making it
blankets. But for most of teams, it was not found to be necessary and more difficult to achieve hyperthermia with high temperatures. It also
precooling is simply accomplished by limited use of body warming increases exposure of operative personnel to chemotherapy. Because
during the cytoreduction, which is often a long procedure [12]. During the surgeon is required to manually manipulate the viscera, increased
the procedure, temperature probes must be placed at different sites of potential exists for contact exposure. Moreover, despite the use of
circuit and intraperitoneal cavity: heat generator, inflow and outflow the smoke evacuator, heated chemotherapy can aerosolize, creating
drains, bladder, liver, mesentery [13]. It is agreed that the desirable inhalational exposure. Stuart et al. [16] evaluated the safety of
intra-abdominal temperatures range to maintain during HIPEC is operating room personnel during the coliseum technique. All
41.5–438C, needing inflow temperatures at 46–488C [2]. The duration assessments (urine members, air sampled, and sterile gloves examina-
of the procedure varies according to investigators and drugs used from tion) were found to be in compliance with established safety standards.
30–120 min. An increased drug concentration in the instillate with a Since this study, no trial reported on increased risk of chemotherapy
shorter bathing duration would probably give similar pharmacokinetic exposure. However, guidelines concerning safety considerations for
results to a longer bathing duration with decreased drug concentration. operative room personnel are needed and have well established by
The best duration is not known and depends on the protocol used [8,9]. Gonzalez-Bayon et al. [17]: selection and education of operative room
Among the different devices reported into scientific literature we will personnel, restriction of personnel inside the operative room, used of
discuss advantages and inconvenient of open abdomen (coliseum) rigid containers, protective barrier garments, disposable, impervious
technique, closed abdomen technique, peritoneal cavity expander and gown, high power filtration mask, smoke evacuator, avoided spills.
semi-open technique. Mortality and morbidity rates may be compared following
Open abdomen technique. The open abdominal technique has also HIPEC using different techniques, but side effects from HIPEC appear
been referred to as the ‘‘Coliseum technique’’. A silastic sheet is to be principally related to the magnitude of the surgery in the largest
sutured over a Thompson retractor and to the patient’s skin over the trials of literature (Table I) [18–23]. Moreover, because of differences
abdominal incision. This suspends that abdominal wall creating a in term of patient selection, chemotherapy agents used, level of
‘‘Coliseum’’ or ‘‘soup bowl-like’’ container for instillation of the
peritoneal perfusate. An incision is made in the middle of the sheet TABLE I. Mortality and Morbidity Following Cytoreductive Surgery
to allow manual manipulation of the intra-abdominal contents Combined With HIPEC Using Closed or Open Abdomen Techniques
to prevent stasis of the heated perfusate. A smoke evacuator is Authors Technique Nb Mortality (%) Morbidity (%)
used to clear aerosolized chemotherapy liberated during the procedure
(Fig. 1). Sugarbaker et al. [22] Opened 356 2 19
Elias et al. [14] did a prospective phase II trial testing seven Verwaal et al. [23] Opened 102 8 65
different techniques in 32 patients. They found that complete closure of Elias et al. [18] Opened 64 9 55
the abdominal wall before the perfusion restricted the volume of the Shen et al. [21] Closed 109 8 36
Kusamura et al. [20] Closed 209 1 12
perfusion, decreased spatial diffusion of the instillate, and resulted in
Glehen et al. [19] Closed 216 3 24
lack of thermal homogeneity. Use of the a peritoneal cavity expander

Journal of Surgical Oncology


244 Glehen et al.

Fig. 2. HIPEC device using a closed abdomen procedure. [Color figure can be viewed in the online issue, available at www.interscience.
wiley.com.]

hyperthermia, and goal of treatment (curative or palliative intent) chemotherapy with positive pressure has been reported to enhance the
comparisons of postoperative results as well as survival results between penetration of drugs into tissue. It was reported in vivo by Jacquet et al.
the different procedures are difficult. [26] for the intraperitoneal administration of doxorubicin at pressure of
Closed abdomen technique. The intraoperative closed technique is 20–30 mm Hg. Recently, Esquis et al. [27] reported that intraperitoneal
perhaps the most widely used method of HIPEC. The closure of administration of cisplatin with increased intra-abdominal pressure
the abdominal wall may be temporary or definitive. Anastomoses are (40 mm Hg) improved the tumor accumulation and the antitumor effect
performed before or after perfusion. No increased risk of anastomotic of the drug in rats bearing advanced peritoneal carcinomatosis.
fistula or recurrence have been reported for teams which are performing The main disadvantage of the closed technique is the lack of
their anastomoses before [19,20]. This abdominal wall may be manually uniform distribution of the heated intraperitoneal chemotherapy. With
agitated during the perfusion in an attempt to promote uniform heat the instillation of methylene blue during the procedure, uneven
distribution. Figure 2 illustrated the technique used in Lyon. distribution was reported [14,15]. Inadequate circulation of heated
The major advantage of the closed technique is the ability to rapidly intraperitoneal perfusate leads to pooling and accumulation of heat and
achieve and maintain hyperthermia, because there is minimal heat chemotherapy in dependant parts of the abdomen. This may result in
loss from the closed abdomen. The technique has been refined increased systemic absorption and result in foci of hyperthermic injury
with modeling studies to optimize thermal homogeneity [24,25]. In that could contribute to postoperative ileus, bowel perforation, and
addition, there is minimal contact or aerosolized exposure of the fistula. However, the two largest clinical trials evaluating the closed
operating room staff to the chemotherapy. Moreover, intraperitoneal abdominal technique on more than 200 patients, the mortality and

Fig. 3. Peritoneal cavity expander. [Color figure can be viewed in the Fig. 4. The abdominal cavity expander. [Color figure can be viewed
online issue, available at www.interscience.wiley.com.] in the online issue, available at www.interscience.wiley.com.]

Journal of Surgical Oncology


HIPEC: Modalities of Perfusion 245
TABLE II. Techniques of Intraperitoneal Chemotherapy Administration

Technique Advantages Disadvantages

Early postoperative chemotherapy Multiple cycles of chemotherapy Increased toxicity


Uneven distribution of chemotherapy
Port malfunctions
No hyperthermia
Closed intraoperative Minimizes exposure of OR staff to chemotherapy Uneven heat distribution
Easier to achieve high perfusion temperatures Lack of uniform drug distribution
Increased drug penetration and tissue uptakes with high
intraabdominal pressure
Open intraoperative More uniform heat distribution Increased risk of OR staff exposure to chemotherapy
‘‘Coliseum technique’’ More uniform drug distribution Heat dissipation
Peritoneal cavity expander Uniform distribution of chemotherapy Increased risk of OR staff exposure to chemotherapy
More complex apparatus
Semi-closed (semi-opened) intraoperative More uniform heat distribution More complex apparatus
More uniform drug distribution Not popular
Minimizes exposure of OR staff to chemotherapy

TABLE III. Expert Votes Regarding the Best Technique of HIPEC (Milan, December 2006): Results of 1st and
2nd Round

HIPEC technique 1st round 2nd round

Open abdominal technique 25.8% 25%


Peritoneal cavity expander 3.2% 0%
Closed abdominal technique 9.7% 6.25%
Semi-opened technique 0% 3.25%
There is no sufficient evidence in literature confirming the superiority of one technique over 61.3% 62.5%
the others in terms of outcome, morbidity and safety to the personnel of the operating
theatre. Each option has its own operational advantages and disadvantages and future
prospective studies must be conducted to establish which one is the best alternative

morbidity rates were 1 to 3% and 12 to 24%, respectively (Table I) A summary of the advantages and disadvantages of the above
[19,20]. Authors did not report any complications that may be caused techniques are depicted in Table II.
by the problem of inadequate circulation.
Peritoneal cavity expander. An alternative method to increase the
distribution of heated chemotherapy involves the use of a peritoneal CONSENSUS VOTE ON MODALITIES
cavity expander (PCE), first reported by Fujimura et al. [28] The PCE OF PERFUSION
is an acrylic cylinder containing inflow and outflow catheters that
are secured over the wound. When filled with heated perfusate, the Experts were asked to vote on the best technique of HIPEC. The
PCE can accommodate the small intestine, allowing the small intestine results of two rounds of vote are reported in Table III. The majority
to float freely and be manually manipulated in the perfusate (Fig. 3). It thought that there is no sufficient evidence in literature to establish
was particularly developed by Japanese authors for the treatment or the the superiority of one technique over the others. The only way to
prevention of gastric carcinomatosis [29–31]. demonstrate it would be to conduct a prospective randomized study.
By temporarily increasing the volume of the peritoneal cavity, a Actually, each specialized team have to improve and adapt their
more uniform distribution is theoretically achieved compared with the technique (open or closed) to allow acceptable mortality and morbidity
closed technique. rates, low risk of staff exposure to chemotherapy and improved
The main disadvantage of the PCE technique is the risk of exposure survival outcomes.
to operating room personnel as discussed for the coliseum tech-
nique. Moreover, it is a complex apparatus that need experience to be
manipulated and it does not allow treatment of parietal wound. REFERENCES
Semi-opened (or semi-closed) abdominal techniques. It may be 1. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of
interesting to combine advantages of the closed and the open cytoreduction and hyperthermic intraperitoneal chemotherapy
technique. Rat et al. [32] reported the use of an abdominal cavity versus systemic chemotherapy and palliative surgery in patients
expander (Fig. 4). The skin edges are water tightly stapled with a soft with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol
‘‘abdominal cavity expander,’’ supported by a Thompson self-retaining 2003;21:3737–3743.
retractor positioned over the abdomen. So, the level of the liquid can be 2. Gonzalez-Moreno S: Peritoneal surface oncology: A progress
widely raised above the level of the skin edges. The anterior wall report. Eur J Surg Oncol 2006;32:593–596.
peritoneum, the wall edges are constantly exposed to the liquid. Large 3. Esquivel J, Vidal-Jove J, Steves MA, et al.: Morbidity and
mortality of cytoreductive surgery and intraperitoneal chemo-
amplitude movements become possible: introduction into the abdomen therapy. Surgery 1993;113:631–636.
of two forearms, even two arms, does not induce loss of any liquid. 4. Jacquet P, Averbach A, Stephens AD, et al.: Heated intraoperative
Paul Sugarbaker also reported the use of an instrument to provide intraperitoneal mitomycin C and early postoperative intraperito-
containment of intraperitoneal chemotherapy, that may allow to limit neal 5-fluorouracil: Pharmacokinetic studies. Oncology 1998;55:
heat loss and staff exposure to chemotherapy [33]. 130–138.

Journal of Surgical Oncology


246 Glehen et al.
5. Glehen OKF, Sugarbaker PH, Elias D, et al.: Cytoreductive Morbidity and mortality analysis of 216 consecutive procedures.
surgery combined whith perioperative intraperitoneal chemo- Ann Surg Oncol 2003;10:863–869.
therapy for the management of peritoneal carcinomatosis from 20. Kusamura S, Younan R, Baratti D, et al.: Cytoreductive surgery
colorectal cancer: A multi-institutional study. J Clin Oncol 2004; followed by intraperitoneal hyperthermic perfusion: Analysis of
22:3284–3292. morbidity and mortality in 209 peritoneal surface malignancies
6. Esquivel J, Angulo F, Bland RK, et al.: Hemodynamic and cardiac treated with closed abdomen technique. Cancer 2006;106:1144–
function parameters during heated intraoperative intraperitoneal 1153.
chemotherapy using the open ‘‘coliseum technique’’. Ann Surg 21. Shen P, Levine EA, Hall J, et al.: Factors predicting survival after
Oncol 2000;7:296–300. intraperitoneal hyperthermic chemotherapy with mitomycin
7. Crile G Jr: The effects of heat and radiation on cancers implanted C after cytoreductive surgery for patients with peritoneal
on the feet of mice. Cancer Res 1963;23:372–380. carcinomatosis. Arch Surg 2003;138:26–33.
8. Elias DM, Ouellet JF: Intraperitoneal chemohyperthermia: 22. Sugarbaker PH, Alderman R, Edwards G, et al.: Prospective
Rationale, technique, indications, and results. Surg Oncol Clin morbidity and mortality assessment of cytoreductive surgery plus
N Am 2000;10:915–933, xi. perioperative intraperitoneal chemotherapy to treat peritoneal
9. Glehen OMF, Gilly FN: Peritoneal carcinomatosis from digestive dissemination of appendiceal mucinous malignancy. Ann Surg
tract cancer: New management by cytoreductive surgery and Oncol 2006;13:635–644.
intraperitoneal chemohyperthermia. Lancet Oncol 2004;5:219– 23. Verwaal VJ, van Tinteren H, Ruth SV, et al.: Toxicity of
228. cytoreductive surgery and hyperthermic intra-peritoneal chemo-
10. Sugarbaker PH: Intraperitoneal chemotherapy and cytoreductive therapy. J Surg Oncol 2004;85:61–67.
surgery for the prevention and treatment of peritoneal carcino- 24. Porcheron J, Talabard JN, Breton C, et al.: Intraperitoneal
matosis and sarcomatosis. Semin Surg Oncol 1998;14:254– chemohyperthermia for peritoneal carcinomatosis: Original
261. modeling, clinical tolerance and results study about 30 patients.
11. Elias D, Benizri E, Di Pietrantonio D, et al.: Comparison of two Hepatogastroenterology 2000;47:1411–1418.
kinds of intraperitoneal chemotherapy following complete 25. Szafnicki K, Cournil M, O’Meara D, et al.: Modelling of
cytoreductive surgery of colorectal peritoneal carcinomatosis. intraperitoneal chemohyperthermia: Experimental study and
Ann Surg Oncol 2007;14:509–514. identification of certain thermal aspects. Bull Cancer 1998;85:
12. Sarnaik AA, Sussman JJ, Ahmad SA, et al.: Technology of 160–166.
intraperitoneal chemotherapy administration: A survey of techni- 26. Jacquet P, Stuart OA, Chang D, et al.: Effects of intra-abdominal
ques with a review of morbidity and mortality. Surg Oncol Clin N pressure on pharmacokinetics and tissue distribution of doxor-
Am 2003;12:849–863. ubicin after intraperitoneal administration. Anticancer Drugs
13. Szafnicki K, Narce C: Towards a ‘‘crippled-mode’’ operation of 1996;7:596–603.
an industrial wastewater treatment plant. Water Res 2006;40: 27. Esquis P, Consolo D, Magnin G, et al.: High intra-abdominal
579–587. pressure enhances the penetration and antitumor effect of
14. Elias D, Antoun S, Goharin A, et al.: Research on the best intraperitoneal cisplatin on experimental peritoneal carcinoma-
chemohyperthermia technique of treatment of peritoneal carci- tosis. Ann Surg 2006;244:106–112.
nomatosis after complete resection. Int J Surg Investig 2000;1: 28. Fujimura T, Yonemura Y, Fushida S, et al.: Continuous hyper-
431–439. thermic peritoneal perfusion for the treatment of peritoneal
15. Stephens AD, Alderman R, Chang D, et al.: Morbidity and dissemination in gastric cancers and subsequent second-look
mortality analysis of 200 treatments with cytoreductive surgery operation. Cancer 1990;65:65–71.
and hyperthermic intraoperative intraperitoneal chemotherapy 29. Fujimura T, Yonemura Y, Fujita H, et al.: Chemohyperthermic
using the coliseum technique. Ann Surg Oncol 1999;6:790–796. peritoneal perfusion for peritoneal dissemination in various intra-
16. Stuart OA, Stephens AD, Welch L, et al.: Safety monitoring of the abdominal malignancies. Int Surg 1999;84:60–66.
coliseum technique for heated intraoperative intraperitoneal 30. Hirose K, Katayama K, Iida A, et al.: Efficacy of continuous
chemotherapy with mitomycin C. Ann Surg Oncol 2002;9:186– hyperthermic peritoneal perfusion for the prophylaxis and treat-
191. ment of peritoneal metastasis of advanced gastric cancer:
17. Gonzalez-Bayon L, Gonzalez-Moreno S, Ortega-Perez G: Safety Evaluation by multivariate regression analysis. Oncology 1999;
considerations for operating room personnel during hyperthermic 57:106–114.
intraoperative intraperitoneal chemotherapy perfusion. Eur J Surg 31. Yonemura Y, Bandou E, Kinoshita K, et al.: Effective therapy for
Oncol 2006;32:619–624. peritoneal dissemination in gastric cancer. Surg Oncol Clin N Am
18. Elias D, Blot F, El Otmany A, et al.: Curative treatment of 2003;12:635–648.
peritoneal carcinomatosis arising from colorectal cancer by 32. Rat P, Benoit L, Cheynel N, et al.: Intraperitoneal chemo-
complete resection and intraperitoneal chemotherapy. Cancer hyperthermia with ‘‘overflow’’ open abdomen. Ann Chir 2001;
2001;92:71–76. 126:669–671.
19. Glehen O, Osinsky D, Cotte E, et al.: Intraperitoneal chemo- 33. Sugarbaker PH: An instrument to provide containment of
hyperthermia using a closed abdominal procedure and cytor- intraoperative intraperitoneal chemotherapy with optimized
eductive surgery for the treatment of peritoneal carcinomatosis: distribution. J Surg Oncol 2005;92:142–146.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:247–252

Drugs, Carrier Solutions and Temperature in


Hyperthermic Intraperitoneal Chemotherapy

SHIGEKI KUSAMURA, MD, PhD,1 ELIAS DOMINIQUE, MD, PhD,2*,{ DARIO BARATTI, MD,1
RAMI YOUNAN, MD,3 AND MARCELLO DERACO, MD1
1
Department of Surgery, National Cancer Institute of Milan, Italy
2
Department of Surgical Oncology, Institut Gustave Roussy, Villejuif, France
3
Department of Surgery-Surgical Oncology Unit, CHUM, University of Montreal Health Centre, Montreal, Canada

At the Fifth International Workshop on Peritoneal Surface Malignancy, in Milan, the consensus on technical aspects of cytoreductive surgery
(CRS) for peritoneal surface malignancy was obtained through the Delphi process. Conflicting points concerning drugs, carrier solution and
optimal temperature for hyperthermic intraperitoneal chemotherapy (HIPEC) were discussed.
J. Surg. Oncol. 2008;98:247–252. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal carcinomatosis; hyperthermic intraperitoneal chemotherapy; consensus

INTRODUCTION the concentration of agents in the perfusate, the volume of the carrier
solution should also be taken in consideration [1,2].
On December 4–6, 2006, the National Cancer Institute of Milan In Table III different types of carrier solution and their respective
organized a consensus statement on the management of peritoneal main characteristics are outlined.
surface malignancy (PSM). This conference brought together experts
in the field of local–regional therapy in an effort to discuss current Intraperitoneal Temperature During HIPEC
approaches to this PSM.
There is two different but synergic points to consider: the suitable
Eligible Drugs for Hyperthermic temperature to obtain and the duration of hyperthermia.
Intraperitoneal Chemotherapy What is the theoretical optimal temperature? Different levels
of target temperatures have been reported in the literature: from 40 to
Several drugs are available for intraperitoneal use, as outlined in 418C [3], from 41 to 438C [4], from 41.5 to 42.58C [20], 428C [5], from
Table I. Theoretically, only cell cycle phase non-specific agents are 42 to 438C [6] and from 42 to 458C [7].
indicated for this single-shot cancer treatment. In other words cell The establishment of the optimal temperature level during the per-
cycle phase specific agents should be not suitable for HIPEC. fusion requires the consideration of several aspects regarding the inter-
The drug (or combination of drugs) should be allocated by the action between heat and chemotherapies, method of control, besides
experts in one of the following phase of scientific investigation the risk of side-effects. Potential development of thermo-tolerance due
pathway: to the activation of heat shock proteins during short exposure time
(30 min or less) should also be taken in consideration [8].
. phase I or dose finding; Regarding the type of drugs this does not constitute a problem as all
. phase II for evaluation of tumor response; of those usually used for HIPEC are chemically stable at temperature
. phase III for evaluation of efficacy in terms of survival; as high as 508C.
. routine clinical use. Synergism between various cytotoxic drugs and hyperthermia starts
at a temperature of 398C but is stronger at higher temperatures;
However, attention should be paid to some important aspects. The according to in vitro studies on culture cells at temperature of 458C
maximum tolerated dose determined for one drug by a study on early cytotoxicity of the chemotherapies is far more intense than at 41 or
postoperative intraperitoneal chemotherapy (EPIC) should not neces- 428C; thus intuitively it is reasonable to use the highest level of
sary be applicable to hyperthermic intraperitoneal chemotherapy hyperthermia restricted only by the clinical tolerance.
(HIPEC). The same succeeds between the modalities of HIPEC: the
maximum tolerated dose of one drug defined a phase I study using the
Coliseum technique should not be adopted by those who works with
The author has no financial interest related to the contents of this article to
the closed abdomen technique. disclose.
{
Carrier Solution Chief.
*Correspondence to: Elias Dominique, MD, PhD, Département de chirurgie
The choice of a carrier solution in which the chemotherapy is générale oncologique, Institut Gustave-Roussy, 39 rue Camille-Desmoulins,
administered plays an important role in the clearance of drugs from Villejuif Cedex, France. Fax: (33) 01 42 11 5256. E-mail: elias@igr.fr
peritoneal cavity to plasma. Received 20 March 2008; Accepted 21 March 2008
However, it is necessary to underline that the chemical aspect of the DOI 10.1002/jso.21051
carrier is not the only factor that impact on pharmacokinetics and Published online in Wiley InterScience
penetration of the agents inside the tumor deposit. Other factors such as (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


TABLE I. Chemotherapy Agents Used for Intraperitoneal Chemotherapy

Normothermic intraperitoneal
HIPEC chemotherapy
248
Dose-escalation Dose-escalation
Molecular Heat Depth of study as single study as single
Drug weight AUC ratio synergy penetration agent MTD agent MTD Comments
2
Doxorubicin 580.0 230 Yes 4–6 cell No — Yes [12] 15 mg/m Sugarbaker et al. conducted a dose escalation study with early
layers postoperative intraperitoneal doxorubicin using
pharmacokinetic monitoring [12]
Melphalan 305.2 93 Marked NA No — Yes [13] 70 mg/m2 Hyperthermia affects the pharmacokinetics of intraperitoneal

Journal of Surgical Oncology


Melphalan by decreasing the AUC of peritoneal fluid Melphalan
Kusamura et al.

without increasing the plasma AUC in rat models [14]


Under investigation with hyperthermia in a clinical trial in patients
with small-volume residual carcinomatosis postcytoreduction at
a dose of 70 mg/m2 [15]
Mitomycin C 334.3 23.5 Yes 2,000 m Yes [16] 35 mg/m2 No — Used by most of the centers that participated in the Fourth
International Workshop on PSM for the treatment of
carcinomatosis of appendiceal or colonic origin [17]
According to Zoetmulder et al. the maximal tolerated dose, when
used as a single agent is 35 mg/m2 for HIPEC [18]
Cisplatin 300.1 7.8 Yes 1–3 mm Yes [19] 300 mg/m2 Yes It was demonstrated in animal models that hyperthermic perfusion
with 15 mg/ml (in 200 ml) cisplatin gave equivalent plasma drug
levels to a maximum tolerated dose (MTD] ip bolus injection
of 4 mg/kg (36 micrograms/ml in 20 ml) [20]
Gemcitabine 299.5 500 At 48 h NA No — No — The cytotoxicity of the drug is synergized by heat if the
hyperthermia is used at 48 h after intravenous drug
administration in a rat model [21]
A dose escalation study was conducted for the combination
cisplatin þ gemcitabine in normothermic condition. The MTD
was cisplatin (75 mg/m2) on day 1 and gemcitabine at 500 mg/m2
on days 1 and 8 on a 21-day schedule for four courses [22]
Mitoxantrone 517.41 115–255 Yes 5–6 cellular No — Yes [23] 28 mg/m2 Hyperthermic perfusion seems to lower mitoxantrone levels in the
layers peritoneal fluid without greatly influencing plasma levels
In phase I/II studies, Link et al. determined that a single
intraperitoneal instillation of 28 mg/m2 is well tolerated. The
study was conducted for the evaluation of the treatment with
a palliative intent in normothermic condition [24]
The only phase I study on mitoxantrone in HIPEC evaluated the
pharmacokinetics properties of the drug [25]
Oxaliplatin 397.3 16 Yes 1–2 mm Yes 460 mg/m2 No — According to a dose escalation Phase II study conducted by Elias
et al. oxaliplatin should be administered by a very short pulse of
high-dose (460 mg/m2) using a short dwell time (30 min) [26]
Etoposide 588.58 65 Yes NA No — Yes [27] 700 mg/m2 Experiences with Etoposide for HIPEC succeeded in combination
with other drugs in the treatment of gastric cancer [28,29]
Irinotecan 677.19 N/A No NA No — No — Elias et al. conducted a phase I study on pharmacokinetics of the
combination oxaliplatin and irinotecan, after intravenous
leucovorin (20 mg/m2) and 5-fluorouracil (400 mg/m2) [26]
Paclitaxel 853.9 1,000 No >80 cell No — Yes [30] 120–180 mg Up to date of the present consensus meeting there is no report in
layers total dose literature documenting the employment of paclitaxel for HIPEC
Docetaxel 861.9 552 No NA No — Yes [31] 156 mg/m2 Tested for HIPEC phase I study in gynecologic cancer patients [32]
5-Fluorouracil 130.08 250 Minimal 0.2 mm No — Yes [33] 650 mg/m2  Reported experiences of the 5-fluorouracil in combination with
5 days hyperthermia are limited to experimental studies [34,35]. The
drug has been employed mostly in early perioperative
intraperitoneal chemotherapy
Carboplatin 371.25 10 Yes 0.5 mm Yes [36] 800 mg/m2 Yes [37,38] 500 mg/m2 Steller et al. concluded that the maximum tolerated dose of
carboplatin for HIPEC with closed abdomen technique
is 800 mg/m2 [36]
Consensus in Local-Regional Treatment 249

Although not formally defined by a phase I dose escalation trial this recommended dose is popular

HIPEC: heated intraperitoneal chemotherapy; normothermic intraperitoneal chemotherapy; MTD: maximum tolerated dose; NA: not available. CDDP: cisplatin; TNF: tumor necrosis factor; Dx: doxorubicin; MMC:
The limiting factor for the employment of temperature as high as
Used by almost unanimously for advanced ovarian cancer according to the progress report of
The combination has been tested in phase II studies using HIPEC to treat a variety of tumors

The hospital mortality rate was 2.5%; grade 3-4 hematological toxicity rate was 58% [26]
458C is the tolerance of the small bowel. The only one study addressing

and irinotecan, after intravenous leucovorin (20 mg/m2) and 5-fluorouracil (400 mg/m2).
Elias et al. conducted a phase I study on pharmacokinetics of the combination oxaliplatin
the thermo-tolerance was performed in animal model (rat). It was
peritoneal mesothelioma [40,41], peritoneal sarcomatosis [42], ovarian cancer [43]

concluded that 448C during 30 min was the maximal well-tolerated


temperature [9].
However, two points remain unresolved: (1) it is not known whether
Strongly advocated by Elias D. at 4th International Workshop on PSM [17]

the conclusions drawn by experimental studies with animal models


This dose schedule was reported as a single experience by Park et al. [19]

This dose schedule was reported as a single experience by Ryu et al. [46]
could be extended to the clinical ground; (2) what is the impact of the
duration of the procedure when we used high temperature?
The duration of hyperthermia. There is clinical data demon-
strating the safety of hyperthermia with different schemes established
on empirical bases, but not from systematic experimental studies.
These schemes are the following: to use a temperature of 418C but
Observation

during 90 min, or to use 438C but during 30–40 min. Also, some teams
use a temperature of 428C during 60 min. Long duration hyperthermia
among the local–regional therapists [44,45]

needs to use cooling systems to decrease the body’s temperature.


4th International Workshop on PSM [17]

Unfortunately, there is no systematic study about the escalation of the


level and also of the duration of hyperthermia in animals or in human.
However, it appears that low hyperthermia allows long duration of the
procedure and that high hyperthermia do not allow it. At that time
nobody knows if it more efficient to privilege high temperature or long
duration. If they were equivalent, the first should be cheaper.
Also, it is necessary to obtain a thermal homogeneity in the whole
abdominal cavity to be sure that every site of the diffuse peritoneal
disease will receive the optimal treatment. It is well accepted in the
literature that only the coliseum technique with a continuous stirring of
the viscera, allows to obtain it. Even with this continuous stirring, and a
high flow rate in the pumps (2 L/min), and with a moderate volume of
perfusate (2 L/m2), in the experience of Elias et al., to obtain
a minimum of 428C in the out-drains, it is necessary to have between
44 and 458C in the in-drains.
(20 mg/m2 iv) and 5-fluorouracil (400 mg/m2 iv)
CDDP (250 mg/m ) and TNF alpha (0.1 mg) [19]

NB: using 360 mg/m2 for the two agents gives less
hematological toxicity (¼ recomanded dosage)
CDDP (25 mg/m2/L of perfusate) and MMC

The Different Parameters Impacting on


Oxaliplatin (460 mg/m2 ip) þ leucovorin

Carboplatin (350 mg/m2) þ interferon-a


CDDP (43 mg/L of perfusate) and Dx

Oxaliplatin (460 mg/m2) þ irinotecan

Pharmacokinetics and Efficacy of HIPEC


Recommended dose

(15.25 mg/L of perfusate) [39]

At last, eight parameters can be modified during HIPEC: type of


(3.3 mg/m2/L of perfusate)

drugs, concentration of drugs, combination of them, carrier solution,


(5,000,000 IU/m2) [46]

volume of the perfusate, level of the temperature, the duration, and the
technique (open or close cavity). The possible different combinations
(400 mg/m2) [26]
2

of them are unlimited. Moreover, when one parameter changes, a


pharmacokinetics study must be performed.
mitomycin C; INF: interferon; PSM: peritoneal surface malignancy.

MATERIALS AND METHODS


In order to achieve a consensus among experts, the Delphi
methodology was employed. Conflicting points were identified and
Tested in dose-escalation

related multiple choice questions along with a scientific review


study for HIPEC

document on the topic were circulated among a panel of experts on


peritoneal surface oncology. Two rounds of web-based voting,
Yes
Yes

Yes

Yes
No

No

the second one after disclosing the results of the first round, were
carried out. Consensus was defined as the agreement of 51% of the
voters [10,11]. A detailed description of the methodology used for this
consensus development is available elsewhere in this issue. Presenta-
tion and discussion of the results were done at the Fifth International
Workshop on Peritoneal Surface Malignancy held in Milan, Italy,
Oxaliplatin ip þ leucovorin

December 4–6, 2006.


Carboplatin þ INF alpha
TABLE I. (Continued )

Oxaliplatin þ Irinotecan
iv þ 5-fluorouracil iv
CDDP þ TNF alpha

RESULTS
CDDP þ MMC
Combinations

CDDP þ Dx

The contents of the related scientific review document are outlined in


Introduction Section. The accompanying conflicting points circulated
among the experts with their respective results of the second voting
round were the following:

Journal of Surgical Oncology


250 Kusamura et al.

Fig. 1. Scientific pathway for drug development.

Drugs DISCUSSION AND COMMENTS


Several drug schedules have been used in the last two decades for The panel of experts reached a consensus in only 2 out of 19 points
HIPEC in the treatment of peritoneal surface malignancies. Usually the submitted to voting with respect to the allocation of the drugs in the
scientific pathway expected to be followed before the introduction of different phases of scientific investigational pathway. About seventy
a antiblastic drug (alone or in combination) in the routine clinical percent of the voters agreed that Cisplatin and Mitomycin-C could be
practice is outlined in Figure 1. employed in the routine clinical practice of local–regional therapy as
However, this pathway has not been strictly respected in the local– single agents.
regional therapy scenario. As an example, some drugs have been tested Although no consensus was established in the following points (as
in phase II studies without having been adequately tested in phase I the minimum cut-off level of 51% was not reached) the voters
trials. Fortuitously such situations could have resulted in procedures preferences could be summarized as follows: (1) drugs not eligible for
with acceptable morbidity, which accords with the conceptual goal of HIPEC: paclitaxel and 5-fluoruracil; (2) drugs elected for experimental
local–regional therapy of minimal systemic antiblastic side-effects. studies: gemcitabine, mitoxantrone, etoposide, docetaxel, cispla-
However, the disadvantage of such a skipping policy is that the drug tin þ TNF alpha and carboplatin þ INF alpha; (3) drugs elected for
(alone of combination) could be adopted by local–regional therapist phase I studies: melphalan, irinotecan, carboplatin, oxalipla-
with a ill-defined suboptimal dose, which does not exploit the whole tin þ irinotecan; (4) drugs elected for phase II studies: none; (5) drugs
antiblastic potential of the chemotherapy(ies). Thus, taking into elected for phase III studies: oxaliplatin, oxaliplatin ip þ leucovorin
account the state of the art relative to each of the following drug (or iv þ 5-fluorouracil iv; (6) drugs elected for routine clinical use:
combination) for HIPEC, we would like to ask you in which phase it doxorubicin, cisplatin and mitomycin-C.
should be allocated to be adequately studied (the results of the voting
are outlined in Table II). TABLE II. Types of Carrier Solution for Hyperthermic Intraperitoneal
Chemotherapy

Type of carrier Characteristic


Carrier Solutions
Isotonic salt solutions Rapidly absorbed due to low molecular weight [47]
The choice of carrier solution in which the antineoplastic drug is and dextrose solutions
administered can play an important role in the clearance of drugs from Inability to maintain a prolonged high intraperitoneal
peritoneal cavity to plasma. In the circumstance of a high molecular fluid volume
weight drugs with delayed systemic metabolism or excretion, which Hypotonic solutions According to experimental evidences, hypotonic
one of the following alternatives may present more advantages, solution was promising in vitro as it increases the
according to experimental data? Cisplatin accumulation in tumor cells and
enhances its cytotoxicity [48–50]
On the other hand, the administration of heated
(a) isotonic salt solutions and dextrose solutions: 53.13%; intraoperative oxaliplatin in increasing hypotonic
(b) hypotonic solutions: 0%; solutions resulted in a high incidence of
(c) hypertonic solutions: 0%; unexplained postoperative peritoneal bleeding
(d) isotonic high molecular weight solutions: 6.25%; and no pharmacokinetics advantage [51]
(e) the use of carrier solutions of varying tonicity requires further Hypertonic solutions Allows a prolonged high intraperitoneal volume [52]
investigation: 40.63%. Slows down the clearance of intraperitoneal fluid
Main disadvantage is dilution of intraperitoneal drug
due to fluid shift inward to the peritoneal
cavity [53]
Temperature Isotonic high molecular Solutions such as icodextrin allows a prolonged high
weight solutions intraperitoneal volume [54], a reduced drug
The optimal range of temperature (8C) levels during the HIPEC is: clearance from the peritoneal cavity
However, the reduced drug clearance from the
(a) 40–41; 0%; peritoneal cavity does not necessary imply an
(b) 41–43; 62.50%; increased cancerours surfaces exposure to the
(c) 41.5–42.5; 21.88%; drug. Up to date there is no proof that icodextrin
(d) 42; 0%; releases the agents and that the agents penetrates
(e) 42–42.5; 0%; inside the tumor and bathed tissues
Indication for HIPEC based on this elements is
(f) 42–43; 15.63%;
questionable
(g) 42–45; 0%.

Journal of Surgical Oncology


Consensus in Local-Regional Treatment 251
TABLE III. Results of Voting Regarding the Allocation of Chemotherapies in the Scientific Pathway for Drug Development (only the Results of the Second
Round Are Outlined)

(a) Not eligible (b) Experimental (f) Routine


Drug/Combination for HIPEC phase (c) Phase I (d) Phase II (e) Phase III clinical use

Doxorubicin 12.50% 6.25% 9.38% 12.50% 12.50% 46.88%


Melphalan 25.00% 21.88% 37.50% 6.25% 0% 9.38%
Mitomycin-C 12.50% 3.13% 0% 3.13% 9.38% 71.88%
Cisplatin 6.25% 6.25% 0% 6.25% 12.50% 68.75%
Gemcitabine 18.75% 37.50% 31.25% 6.25% 3.13% 3.13%
Mitoxantrone 28.13% 34.38% 15.63% 12.50% 3.13% 6.25%
Oxaliplatin 9.38% 15.63% 9.38% 18.75% 31.25% 15.63%
Etoposide 28.13% 34.38% 12.50% 9.38% 6.25% 9.38%
Irinotecan 18.75% 21.88% 31.25% 15.63% 9.38% 3.13%
Paclitaxel 31.25% 21.88% 21.88% 9.38% 3.13% 12.50%
Docetaxel 18.75% 34.38% 31.25% 3.13% 6.25% 6.25%
5-Fuorouracil 37.50% 15.63% 12.50% 0% 0% 34.38%
Carboplatin 15.63% 15.63% 21.88% 15.63% 15.63% 15.63%
CDDP þ TNF alpha 15.63% 37.50% 25.00% 12.50% 6.25% 3.13%
CDDP þ Dx 9.38% 12.50% 18.75% 6.25% 21.88% 31.25%
CDDP þ MMC 9.38% 15.63% 12.50% 6.25% 21.88% 34.38%
Oxaliplatin ip þ leucovorin 3.13% 15.63% 15.63% 25.00% 28.13% 12.50%
iv þ 5-fluorouracil iv
Oxaliplatin þ irinotecan 9.38% 12.50% 37.50% 12.50% 18.75% 9.38%
Carboplatin þ INF alpha 18.75% 43.75% 28.13% 6.25% 3.13% 0%

The figures highlighted in bold represent the preferred option by the experts after the voting.

The drugs to be used in local–regional therapy was the most 2. Sugarbaker PH, Stuart OA, Carmignani CP: Pharmacokinetic
complex topic approached in the present methodological consensus changes induced by the volume of chemotherapy solution in
statement. In fact, the panel of experts achieved the lowest rate of patients treated with hyperthermic intraperitoneal mitomycin C.
agreement in allocating the chemotherapies (as single agent or in Cancer Chemother Pharmacol 2006;57:703–708.
3. Smeenk RM, Verwaal VJ, Zoetmulder FA: Toxicity and mortality
combination) in different phases of scientific pathway. Most of them of cytoreduction and intraoperative hyperthermic intraperitoneal
voted an option without taking into account the state of the art related chemotherapy in pseudomyxoma peritonei—A report of 103
to this issue, that was outlined in Table II. This material was available procedures. Eur J Surg Oncol 2006;32:186–190. Epub November
to the panel, during the first and second rounds. 21, 2005.
Other aspects that should have been carefully considered is that 4. Shido A, Ohmura S, Yamamoto K, et al.: Does hyperthermia
conclusions drawn with respect to pharmacokinetics or MTD of a induce peritoneal damage in continuous hyperthermic peritoneal
determinate chemotherapy should not be applied in all technical and perfusion? World J Surg 2000;24:507–511.
modalities variations of local regional therapy. What is true for the 5. Moran BJ, Mukherjee A, Sexton R: Operability and early
open abdomen technique of HIPEC is not necessarily applicable for the outcome in 100 consecutive laparotomies for peritoneal malig-
nancy. Br J Surg 2006;93:100–104.
closed technique. This topic of drugs should be a priority for discussion 6. Glehen O, Cotte E, Brigand C, et al.: Therapeutic innovations in
in the upcoming 6th International Workshop on Peritoneal Surface the management of peritoneal carcinomatosis from digestive
Malignancy as little has been cleared with the present debate. origin: Cytoreductive surgery and intraperitoneal chemotherapy
Isotonic salt solutions and dextrose solutions were elected the [Article in French]. Rev Med Int 2006;27:382–391. Epub
advisable type of perfusate for HIPEC. Only 41% of the experts were October 5, 2005.
of the opinion that further studies should be carried out on this matter. 7. Elias D, Raynard B, Boige V, et al.: Impact of the extent and
The optimal range of temperature was 41–438C. This decision is duration of cytoreductive surgery on postoperative hematological
close to that of previous Consensus meeting held in Madrid in 2004. toxicity after intraperitoneal chemohyperthermia for peritoneal
At that occasion the experts elected 41.5–42.58C as the optimal carcinomatosis. J Surg Oncol 2005;90:220–225.
8. Christophi C, Winkworth A, Muralihdaran V, et al.: The treatment
range [17]. of malignancy by hyperthermia. Surg Oncol 1998;7:83–90.
Review.
9. Shimizu T, Maeta M, Koga S: Influence of local hyperthermia on
CONCLUSION the healing of small intestinal anastomoses in the rat. Br J Surg
1991;78:57–59.
A partial consensus was obtained on these different points of
10. Loughlin KG, Moore LF: Using Delphi to achieve congruent
techniques of HIPEC. Eight different parameters impact on the objectives and activities in a pediatrics department. J Med Educ
pharmacokinetics and the efficacy, defining infinite combinations, 1979;54:101–106.
which cannot be studied one by one. A strategic choice of the most 11. Keeney S, Hasson F, McKenna H: Consulting the oracle: Ten
promising combinations to be tested will be determinant for the future. lessons rom using the Delphi technique in nursing research. J Adv
Nurs 2006;53:205–212.
12. Sugarbaker PH: Early postoperative intraperitoneal adriamycin as
REFERENCES an adjuvant treatment for advanced gastric cancer with lymph
node or serosal invasion. In: Sugarbaker PH, editor. Management
1. Elias D, Bonnay M, Puizillou JM, et al.: Heated intra-operative of Gastric Cancer. Boston: Kluwer; 1991: pp. 277–284.
intraperitoneal oxaliplatin after complete resection of peritoneal 13. Sugarbaker PH, Mora JT, Carmignani P, et al.: Update on
carcinomatosis: Pharmacokinetics and tissue distribution. Ann chemotherapeutic agents utilized for perioperative intraperitoneal
Oncol 2002;13:267–272. chemotherapy. Oncologist 2005;10:112–122.

Journal of Surgical Oncology


252 Kusamura et al.
14. Glehen O, Stuart OA, Mohamed F, et al.: Hyperthermia modifies 36. Steller MA, Egorin MJ, Trimble EL, et al.: A pilot phase I trial of
pharmacokinetics and tissue distribution of intraperitoneal melpha- continuous hyperthermic peritoneal perfusion with high-dose
lan in a rat model. Cancer Chemother Pharmacol 2004;54:79–84. carboplatin as primary treatment of patients with small-volume
15. Washington Cancer Institute Protocol #X02380. residual ovarian cancer. Cancer Chemother Pharmacol 1999;43:
16. van Ruth S, Verwaal VJ, Zoetmulder FAN: Pharmacokinetics of 106–114.
intraperitoneal mitomycin C. Surg Oncol Clin N Am 2003;12: 37. Malmstrom H, Larsson D, Simonsen E: Phase I study of
771–780. intraperitoneal carboplatin as adjuvant therapy in early ovarian
17. Gonzalez-Moreno S: Peritoneal surface oncology: A progress cancer. Gynecol Oncol 1990;39:289–294.
report. Eur J Surg Oncol 2006; [Epub ahead of print]. 38. Speyer JL, Beller U, Colombo N, et al.: Intraperitoneal
18. Zoetmulder FA, van der Vange N, Witkamp AJ, et al.: Hyper- carboplatin: Favorable results in women with minimal residual
thermic intra-peritoneal chemotherapy (HIPEC) in patients with ovarian cancer after cisplatin therapy. J Clin Oncol 1990;8:1335–
peritoneal pseudomyxoma or peritoneal metastases of colorectal 1341.
carcinoma: Good preliminary results from the Netherlands 39. Rossi CR, Foletto M, Mocellin S, et al.: Hyperthermic intra-
Cancer Institute [Article in Dutch]. Ned Tijdschr Geneeskd operative intraperitoneal chemotherapy with cisplatin and doxor-
1999;143:1863–1868. ubicin in patients who undergo cytoreductive surgery for
19. Park BJ, Alexander HR, Libutti SK, et al.: Treatment of primary peritoneal carcinomatosis and sarcomatosis: Phase I study. Cancer
peritoneal mesothelioma by continuous hyperthermic peritoneal 2002;94:492–499.
perfusion (CHPP). Ann Surg Oncol 1999;6:582–590. 40. Deraco M, De Simone M, Rossi CR, et al.: An Italian Multicentric
20. Zeamari S, Floot B, van der Vange N, et al.: Pharmacokinetics and Phase II study on peritonectomy and intra peritoneal hyper-
pharmacodynamics of cisplatin after intraoperative hyperthermic thermic perfusion (IPHP) to treat patients with peritoneal
intraperitoneal chemoperfusion (HIPEC). Anticancer Res 2003; mesothelioma. J Exp Clin Cancer Res 2003;22:41–45.
23:1643–1648. 41. Deraco M, Casali P, Inglese MG, et al.: Peritoneal mesothelioma
21. Van Bree C, Beumer C, Rodermond HM, et al.: Effectiveness of treated by induction chemotherapy, cytoreductive surgery, and
20 ,20 difluorodeoxycytidine (Gemcitabine) combined with hyper- intraperitoneal hyperthermic perfusion. J Surg Oncol 2003;83:
thermia in rat R-1 rhabdomyosarcoma in vitro and in vivo. Int J 147–153.
Hyperthermia 1999;15:549–556. 42. Rossi CR, Deraco M, De Simone M, et al.: Hyperthermic
22. Sabbatini P, Aghajanian C, Leitao M, et al.: Intraperitoneal intraperitoneal intraoperative chemotherapy after cytoreductive
cisplatin with intraperitoneal gemcitabine in patients with surgery for the treatment of abdominal sarcomatosis: Clinical
epithelial ovarian cancer: Results of a phase I/II trial. Clin Cancer outcome and prognostic factors in 60 consecutive patients. Cancer
Res 2004;10: 2962–2967. 2004;100:1943–1950.
23. Oza AM, ten Bokkel Huinink W, Dubbelman R, et al.: Phase I/II 43. Raspagliesi F, Kusamura S, Campos Torres JC, et al.: Cytor-
study of intraperitoneal mitoxantrone in refractory ovarian cancer. eduction combined with intraperitoneal hyperthermic perfusion
Ann Oncol 1994;5:343–347. chemotherapy in advanced/recurrent ovarian cancer patients: The
24. Link KH, Roitman M, Holtappels M, et al.: Intraperitoneal experience of National Cancer Institute of Milan. Eur J Surg
chemotherapy with mitoxantrone in malignant ascites. Surg Oncol 2006.
Oncol Clin N Am 2003;12:865–872. 44. Pilati P, Mocellin S, Rossi CR, et al.: Cytoreductive surgery
25. Nicoletto MO, Padrini R, Galeotti F, et al.: Pharmacokinetics of combined with hyperthermic intraperitoneal intraoperative che-
intraperitoneal hyperthermic perfusion with mitoxantrone in motherapy for peritoneal carcinomatosis arising from colon
ovarian cancer. Cancer Chemother Pharmacol 2000;45:457–462. adenocarcinoma. Ann Surg Oncol 2003;10:508–513.
26. Elias D, Sideris L, Pocard M, et al.: Efficacy of intraperitoneal 45. Deraco M, Baratti D, Inglese MG, et al.: Peritonectomy and
chemohyperthermia with oxaliplatin in colorectal peritoneal intraperitoneal hyperthermic perfusion (IPHP): A strategy that
carcinomatosis. Preliminary results in 24 patients. Ann Oncol has confirmed its efficacy in patients with pseudomyxoma
2004;15:781–785. peritonei. Ann Surg Oncol 2004;11:393–398.
27. O’Dwyer PJ, LaCreta FP, Daugherty JP, et al.: Phase I 46. Ryu KS, Kim JH, Ko HS, et al.: Effects of intraperitoneal
pharmacokinetic study of intraperitoneal etoposide. Cancer Res hyperthermic chemotherapy in ovarian cancer. Gynecol Oncol
1991;51:2041–2046. 2004;94:325–332.
28. Yonemura Y, Fujimura T, Nishimura G, et al.: Effects of 47. Torres IJ, Litterst CL, Guarino AM: Transport of model
intraoperative chemohyperthermia in patients with gastric cancer compounds across the peritoneal membrane in the rat. Pharma-
with peritoneal dissemination. Surgery 1996;119:437–444. cology 1978;17:330–340.
29. Yonemura Y, Bandou E, Kinoshita K, et al.: Effective therapy for 48. Groose E, Walker L, Masters JR: The influence of osmolarity on
peritoneal dissemination in gastric cancer. Surg Oncol Clin N Am drug cytotoxicity in vitro. Br J Cancer 1986;54:181.
2003;12:635–648. Review. 49. Kondo A, Maeta M, Oka A, et al.: Hypotonic intraperitoneal
30. Mohamed F, Sugarbaker PH: Intraperitoneal taxanes. Surg Oncol cisplatin chemotherapy for peritoneal carcinomatosis in mice. Br
Clin N Am 2003;12:825–833. J Cancer 1996;73:1166–1170.
31. Morgan RJ Jr, Doroshow JH, Synold T, et al.: Phase I trial of 50. Tsujitani S, Oka A, Kondo A, et al.: Administration in a hypotonic
intraperitoneal docetaxel in the treatment of advancedmalignan- solution is preferable to dose escalation in intraperitoneal
cies primarily confined to the peritoneal cavity: Dose-limiting cisplatin chemotherapy for peritoneal carcinomatosis in rats.
toxicity and pharmacokinetics. Clin Cancer Res 2003;9:5896–5901. Oncology 1999;57:77–82.
32. de Bree E, Romanos J, Michalakis J, et al.: Intraoperative 51. Elias D, El Otmany A, Bonnay M, et al.: Human pharmacokinetic
hyperthermic intraperitoneal chemotherapy with docetaxel study of heated intraperitoneal oxaliplatin in increasingly hypo-
as second-line treatment for peritoneal carcinomatosis of tonic solutions after complete resection of peritoneal carcinoma-
gynaecological origin. Anticancer Res 2003;23:3019–3027. tosis. Oncology 2002;63:346–352.
33. Speyer JL, Collins JM, Dedrick RL, et al.: Phase I and 52. Pestieau SR, Schnake KJ, Stuart OA, et al.: Impact of carrier
pharmacological studies of 5-fluorouracil administered intra- solutions on pharmacokinetics of intraperitoneal chemotherapy.
peritoneally. Cancer Res 1980;40:567–572. Cancer Chemother Pharmacol 2001;47:269–276.
34. Maeta M, Sawata T, Kaibara N: Effects of hyperthermia on the 53. Litterst CL, Torres IJ, Arnold S, et al.: Adsorption of antineo-
metabolism of 5-fluorouracil in vitro. Int J Hyperthermia 1993;9: plastic drugs following large-volume ip administration torats.
105–113. Cancer Treat Rep 1982;66:147–155.
35. Tsumura M, Yoshiga K, Takada K: Enhancement of antitumor 54. McArdle CS, Kerr DJ, O’Gorman P, et al.: Pharmacokinetic study
effects of 1-hexylcarbamoyl-5-fluorouracil combined with hyper- of 5-fluorouracil in a novel dialysate solution: A long-term
thermia on Ehrlich ascites tumor in vivo and Nakahara-Fukuoka intraperitoneal treatment approach for advanced colorectal
sarcoma cell in vitro. Cancer Res 1988;48:3977–3980. carcinoma. Br J Cancer 1994;70:762–766.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:253–257

Morbidity, Toxicity, and Mortality Classification Systems in the


Local Regional Treatment of Peritoneal Surface Malignancy

RAMI YOUNAN, MD,1 SHIGEKI KUSAMURA, MD, PhD,2 DARIO BARATTI, MD,2
ALEXIS-SIMON CLOUTIER, MD,1 AND MARCELLO DERACO, MD2*
1
Department of Surgery, Surgical Oncology Unit, University of Montreal Health Center (CHUM), Montreal, Canada
2
Department of Surgery, Peritoneal Surface Malignancy Unit, Istituto dei tumori, Milano, Italy

To reach a consensus for reporting complications related to cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy
(HIPEC). Reporting the adverse events related to CRS þ HIPEC is not standardized yet. Post-operative complications can be divided in two
categories: the effects of surgical manipulation per se and the toxic effects of the heated intraoperative chemotherapy. Additive and/or synergistic
effects also exist. Different centers have published their experience with regard to the complications associated with the procedure. Various
classification systems have been used which makes a temptative comparison of the different techniques and results almost impossible. An effort
was made here to review the existing major classification systems: The Bozzetti classification, the Clavien classification (and two proposed
modifications from Feldman et al. and Elias et al.) and the Common terminology criteria for adverse events (CTCAE) version 3.0 of the National
Institute of Health (NIH) criteria. A related document was sent to an international panel of experts. The CTCAE was adopted by the panel of
experts as the unique classification system to be used for reporting complications related to CRS þ HIPEC.
J. Surg. Oncol. 2008;98:253–257. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal surface malignancy; HIPEC; cytoreductive surgery; morbidity; toxicity; intraperitoneal
chemotherapy

INTRODUCTION DISCUSSION
Surgical literature lacks a uniform classification system for surgical Death and complication rates remain the most comprehensive
complications. No standard classification system has been agreed on measures used to assess short-term outcomes of a specific procedure.
by the surgical community. This makes the comparison of negative Surgical complications can be in many cases the prime reason for
outcomes of a specific procedure difficult to analyze. Mostly modifying patient management and ultimately getting wide acceptance
misreporting and to a lesser extent underreporting of complications for a specific procedure throughout the medical community. However,
is a frequent happening in the day to day care of patients. these two outcome measures remain hard to assess and define
Cytoreductive surgery (CRS) and hyperthermic intraperitoneal che- throughout the surgical literature because of the lack of standardization
motherapy (HIPEC) is not an exception to the rule. In spite of this, and underreporting. Most probably underreporting of complications
appreciable effort has been developed by investigators performing is the consequence of the absence of a clear system to classify
CRS þ HIPEC to report morbidity, toxicity, and mortality related to the complications. Martin et al. [1] analyzed reports from various centers
procedure and push further the concept of morbidity reporting. describing complications related to three major surgical procedures:
However, non-standardized classification systems exist for morbidity pancreatectomy, esophagectomy, and hepatectomy. They also develop-
and mortality which further complicates this task. A definite effort ed 10 criteria used to assess the quality of reporting complications
should be undertaken to better report complications across the surgical in published series. Interestingly their criteria included the use of a
literature and specifically regarding peritoneal surface malignancy severity grading system for major and minor complications. When
treatment. The aim of the last consensus meeting in Milan was to find a reviewing the 119 studies they analyzed, only 24 graded the severity of
consensus for morbidity reporting to: (a) better compare techniques the complications; furthermore, none of these used a numeric grading
and results between centers; (b) set standards in the surgical literature; system as recommended by the authors. Accurate assessment of the
and (c) demonstrate leadership in the field. Postoperative complica- morbidity related to CRS þ HIPEC is of major concern. The issue is
tions after CRS þ HIPEC are related to surgical manipulation, the relatively complex for CRS þ HIPEC because the emergence of
intraperitoneal chemotherapy component or a combination of both postoperative complications related to surgical manipulation can be
(overlap). Most authors classify surgical morbidities separately from
the toxicity related to the chemotherapy perfusion. Before the Milan
Conference, North American and European series were the largest None to disclose by any of the authors.
reporting on complications. Multiple classification systems were used. *Correspondence to: Marcello Deraco, MD, Fondazione IRCCS Istituto
No conclusion could be drawn from these reports, no comparison could Nazionale dei Tumori Milano, Via Venezian 1, 20133 Milano, Italy.
be made and no consensus was obvious. In this report, we discuss the Fax: þ39-02-23902404. E-mail: marcello.deraco@istitutotumori.mi.it
major classification systems of morbidity and toxicity related to Received 19 March 2008; Accepted 21 March 2008
CRS þ HIPEC, review the main series from various institutions DOI 10.1002/jso.21057
reporting these complications and elaborate a consensus with regard Published online in Wiley InterScience
to reporting complications associated with CRS þ HIPEC. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


254 Younan et al.
TABLE I. Classification of Complications According to Clavien et al. [3]
Grade I Events carrying minor risks, that is, complication if left untreated resolves spontaneously or at most requires a simple bedside procedure. Analgesics,
antipyretics, antiemetics, oral antibiotics and antidiarrheals are permitted
Grade II Potentially life-threatening; require some form of intervention. Grade IIa require other drugs than above, parenteral nutrition or transfusions. Grade IIb
require invasive procedures (radiological or endoscopic) or reoperation
Grade III Events with residual or lasting disability including organ resection
Grade IV Death

confounded with toxic side effects of the intraperitoneal chemotherapy malignancies and found a 30-day mortality rate of 0.9%. Their
(IES). For example, the emergence of postoperative anemia depending major (grade 3 or 4) morbidity rate was 12% overall. The Bozzetti and
on the timing of its onset could be attributed to bleeding or to bone modified Clavien classifications are not intended to account for
marrow suppression or both. Another evidence favoring the existence toxicities related to the use of chemotherapy during CRS þ HIPEC.
of an overlap between side effects related to surgery and to The investigators from Milan used the World Health Organization
intraperitoneal chemotherapies was given by a recent study conducted (WHO) toxicity scale for that purpose. It is a relatively simple four-
by Kusamura et al. [2] They found that a dose of Cisplatin > 240 mg grade scale that includes twelve categories for toxicity: Hematological,
used for HIPEC, using the closed abdomen technique, is an gastrointestinal, pulmonary, allergic, cutaneous, infection, cardiovas-
independent risk factor for the emergence of major surgical morbidity cular, neurotoxicity, fever with drug, hair and renal. Pre-determined
related to CRS þ HIPEC. Despite these considerations, most authors cut-offs are determined within each category to grade the adverse
propose to report and classify the surgical morbidities separately from event. Ten (4.8%) patients presented Grade 3/4 toxicity in their series.
the toxicity related to the chemotherapy component, adopting two The investigators from Institut Gustave-Roussy [4] used the joint
separate classifications. We define here morbidity as any adverse National Cancer Institute(NCI)/National Institute of Health(NIH)
event related to surgical manipulation during the procedure. Toxicity Common Terminology Criteria for Adverse Events (CTCAE) version
is defined as any adverse event that can be clearly related to the 3.0 [7] to report the toxicity related to the procedure. This classification
chemotherapy component. Mortality is finally an adverse event system was initially designed as a clinical trials research tool. It is
resulting in death. Some investigators have employed for morbidity widely used by investigators to accurately record medication toxicity
the modified Clavien system [3,4] while others have adopted more in randomized control trials. It is a very complete and extensive guide
simple criteria derived from their local institutional experience like the regrouping 310 types of complications within 28 categories based on
Bozzetti classification [2,5,6]. And others have used the National the anatomy and/or pathophysiology of the complication (Table V).
Institute of Health (NIH) common terminology criteria to report on Examples of that include separate definitions for fistula, anastomotic
both morbidity and toxicity [7–9]. This heterogeneity in the adopted leak and perforation. All the categories are very well defined and
systems hampers the comparison of treatment-related complication detailed within the 70-pages booklet produced by the NCI/NIH.
rates among the various studies. Clavien et al. [3], in 1992, proposed a Complications are graded within a 5-scale system well detailed in the
classification system for complications in surgery. It included a four- booklet with predetermined definitions (Table VI). One of the main
grade severity scale (Table I). Their proposed system’s relevance was advantages of the CTCAE version 3.0 is that it can be used to
tested and validated on 650 cases of elective cholecystectomy. The determine both toxicity and surgical morbidity. Two more representa-
system was later slightly modified by Feldman et al. [10] to exclude the tive series on CRS þ HIPEC have used exclusively the CTCAE
prolongation of hospital stay as a criterion to increase the grade of the classification system for both toxicity and surgical morbidity. Smeenk
complication (Table II). The latter was then used and modified by et al. [8] from the National Cancer Institute of Holland have reported
investigators reporting on complications related to CRS þ HIPEC. on 103 procedures performed for pseudomyxoma peritonei. They
Elias et al. [4] reported on surgical complications using their found a treatment-related mortality of 11% and an overall complica-
modification scheme (Table III). They found a 2.5% mortality rate tions rate of 54% for grade 3–5 events. This included both toxicity
(one patient). The morbidity rate (grade 3–5 complications only) was and surgical morbidity. Surgical morbidity occurred after 38% of the
69% for major events. Only 12 patients or 30.5% did not present any procedures. Their conclusion was that ‘‘. . .differences in the method
postoperative complication. Other investigators have used the Bozzetti of grading toxicity make it difficult to compare these reports. . .’’
system for surgical complications [5]. It was developed at the Italian Finally Glehen et al. [9] from Lyon published their results after
National Cancer Institute in Milan to report on their local institutional 216 procedures performed for various peritoneal surface malignancies.
experience. It is a very simple four-grade scale to report complications They found a 3.2% mortality rate and a combined grade III/IV toxicity
(Table IV). Kusamura et al. [2] reported their morbidity results using and morbidity rate of 24.5%. The authors used the previous version of
this system. They analyzed 209 patients with various peritoneal surface the CTCAE to report their results.

TABLE II. Feldman et al. [10] Classification of Complications


Grade I Minor complications that resolve spontaneously if left untreated or require a simple bedside procedure. Analgesics, antipyretics, antiemetics, oral
antibiotics and antidiarrheals are permitted. Examples include superficial surgical site infection, urinary retention, lower urinary tract infection, ileus
requiring nasogastric tube
Grade II Potentially life-threatening; require intervention that carry some risks. Grade IIa require other drugs, parenteral nutrition or transfusions. Examples
include pneumonia, arrhythmia or acute pancreatitis. Grade IIb require invasive procedures (radiological or endoscopic) or reoperation. Examples
include CT-guided abcess drainage
Grade III Events with residual or lasting disability or organ loss. Examples include cerebrovascular events with residual disability and iatrogenic splenectomy.
All complications in this classification are categorized as cardiac, respiratory, gastrointestinal, urinary, local, or ‘‘other’’
Grade IV Death

Journal of Surgical Oncology


Classification of Locoregional Treatment 255
TABLE III. Elias et al. [4] Classification of Surgical Complications Related to CRS þ HIPEC
Grade 0 No complications
Grade 1 Complications requiring either no intervention or minor interventions such as oral antibiotics, bowel rest, or basic monitoring
Grade 2 Complications requiring moderate interventions such intravenous medication (e.g., antibiotics or antiarrythmics), total parenteral nutrition, prolonged
tube feeding, or chest tube insertion
Grade 3 Complications are those requiring hospital readmission, surgical intervention, or radiologic intervention
Grade 4 Complications are those producing chronic disability, organ resection, or enteric diversion
Grade 5 Complications result in death

A numeric grading system seems the most appropriate to report (1) They are different in terms of the total number of classes: they can
complications related to CRS þ HIPEC based on the recommendation be of 4 grades or 5 grades.
from Martin et al. [1] and after reviewing the major published series. (2) There is no correspondence between the grades: For example grade
As discussed above, many classification systems have been used when 2 in Feldman classification means a potentially life-threatening
reporting complications. That makes a temptative comparison of the complication usually requiring some form of intervention while in
above reports impossible to make and renders the rates of complica- the Bozzetti classification grade 2 means a mild complication.
tions almost meaningless. In that sense, the high rate of complications (3) One interesting thing in common between the various classifica-
reported by Smeenk et al. can be explained by the fact that the CTCAE tions is that the grades can be grouped into two main subdivisions:
version 3.0 is a system that accounts for a myriad of parameters and is Minor complications and Major complications.
then expected to produce such a high complications rate. On the other
hand, the other classifications are more simplistic and less extensive In order to achieve the consensus among experts, the Delphi
and would expectedly produce less complications as a final outcome. methodology was employed. [11] A conflicting point was identified
Agreeing on a single classification system for complications is in that and related multiple choice questions along with a scientific review
sense of up most importance. A quick evaluation of the various document on the topic were circulated among the panel of experts on
classification systems (Table VII) allows us to raise the following peritoneal surface oncology. Two rounds of web-based voting, the
considerations: second one after disclosing the results of the first round, were carried
out. Consensus was defined as the agreement of 51% of the voters.
[11,12] A detailed description of the methodology used for this
TABLE IV. Bozzetti et al. [5] Classification of Surgical Complications consensus development is available elsewhere in this issue. Presenta-
tion and discussion of the results were done at 5th International
Grade I No complications Workshop on Peritoneal Surface Malignancy held in Milan, Italy, 4–6,
Grade II Minor complications: Wound infection, Urinary tract infection,
December 2006. The question presented to the experts was the follow:
pancreatitis, ileus, deep vein thrombosis
Grade III Major complications (requiring re-operation or Intensive care For the assessment of morbidity, toxicity and mortality, which
admission or interventional radiology treatment) classification system is the most appropriate for a patient undergoing a
Grade IV In-hospital or intensive care unit mortality local-regional therapy?
Three answers were possible and included:

1. Bozzetti et al. criteria for surgical complications and WHO criteria


TABLE V. Categories Included in the CTCAE Version 3.0 [7] for toxicity?
2. Feldman (Clavien or Elias) et al. criteria for surgical complications
Allergy/immunology Infection
and WHO criteria for toxicity?
Auditory/ear Lymphatics
Blood/bone marrow Metabolic/laboratory 3. NCI CTC version 3.0 criteria for both surgical complications and
Cardiac arrhythmia Musculoskeletal/soft tissue toxicity?
Cardiac general Neurology
Coagulation Ocular/visual
Constitutional symptoms Pain The first round of results were as follows: the Bozzetti/WHO
Death Pulmonary/upper respiratory combination 25.81%, the Feldman/WHO combination 12.90% which
Dermatology/skin Renal/genitourinary left the NCI CTC version 3.0 with 61.29% of the vote. The results of
Endocrine Secondary malignancy the second round of voting were even more definitive giving the NCI
Gastrointestinal Sexual/reproductive function CTCAE version 3.0 an approval of 87.5% of the expert members. This
Growth and development Surgery/intraoperative injury classification system was adopted as the consensus system to report all
Hemorrhage/bleeding Syndromes future complications related to CRS þ HIPEC.
Hepatobiliary/pancreas Vascular

CONCLUSION
TABLE VI. Morbidity and Toxicity Grading According to CTCAE Version
A numeric grading system seems to be the most appropriate way to
3.0 [7]
report the complications related to CRS þ HIPEC. It is well known that
Grade 1 Mild adverse event the CTCAE version 3.0 five-level grading system is used for
Grade 2 Moderate adverse event chemotherapy and radiation therapy associated toxicities. The same
Grade 3 Severe adverse event
scale could possibly be used for cytoreductive surgery in an effort to
Grade 4 Life-threatening or disabling adverse event
Grade 5 Death
standardize across those three interdisciplinary oncologic specialties.
Grade 5 would certainly represent death and grade 0 no complications

Journal of Surgical Oncology


256
Younan et al.

Journal of Surgical Oncology


TABLE VII. Summary of the Various Morbidity and Toxicity Classifications

Clavien et al. system [3]

Common terminology criteria for


Bozzetti et al. criteria [5] Feldman et al. modification [10] Elias et al. modification [4] adverse events version 3.0 [7]

Grade I No complications Grade 0 No complications Grade 0 No adverse event or Minor


within normal limits complications
Grade II Minor Grade 1 Complications are minor and are likely Grade 1 Complications requiring either no Grade 1 Mild Adverse Event
complications to resolve spontaneously or with intervention or minor interventions such
antipyretics, analgesics, antidiarrheals as oral antibiotics, bowel rest, or basic
or oral antibiotics monitoring
Grade 2 Complications are potentially Grade 2 Complications requiring moderate Grade 2 Moderate adverse event
life-threatening and interventions such intravenous
usually require some form of medication (e.g., antibiotics or
intervention. Subsets of this include 2a, antiarrythmics), total parenteral
requiring medications other than above nutrition, prolonged tube feeding,
or total parenteral nutrition for treatment or chest tube insertion
2b implies the need for reoperation or an
invasive procedure
Grade III Major Grade 3 Complications have lasting disability Grade 3 Complications are those requiring Grade 3 Severe and undesirable Major
complications or require an organ resection hospital read mission, surgical adverse event complications
(requiring intervention, or radiologic intervention
re-operation or
ICU admission
or interventional
radiology)
Grade IV In-hospital or Grade 4 Death Grade 4 Complications are those producing Grade 4 Life-threatening or
intensive care chronic dis ability, organ resection, disabling adverse event
unit mortality or enteric diversion
Grade 5 Complications result in death Grade 5 Death related to adverse event
Classification of Locoregional Treatment 257
resulting from the treatment. The CTCAE version 3.0 was indeed 6. Younan R, Kusamura S, Baratti D, et al.: Bowel complications
adopted by an international panel of experts as the definitive in 203 cases of peritoneal surface malignancies treated with
classification system for complications resulting from CRS þ HIPEC. peritonectomy and closed-technique intraperitoneal hyperthermic
perfusion. Ann Surg Oncol 2005;12:910–918.
7. https://webapps.ctep.nci.nih.gov/webobjs/ctc/webhelp/welcome_
REFERENCES to_ctcae.htm.
8. Smeenk RM, Verwaal VJ, Zoetmulder FA: Toxicity and mortality
1. Martin RC, Brennan MF, Jaques DP: Quality of complication of cytoreduction and intraoperative hyperthermic intraperitoneal
reporting in the surgical literature. Ann Surg 2002;235:803– chemotherapy in pseudomyxoma peritonei–A report of 103
813. procedures. Eur J Surg Oncol 2006;32:186–190.
2. Kusamura S, Younan R, Baratti D, et al.: Cytoreductive surgery 9. Glehen O, Osinsky D, Cotte E, et al.: Intraperitoneal chemo-
followed by intraperitoneal hyperthermic perfusion: Analysis of hyperthermia using a closed abdominal procedure and cytor-
morbidity and mortality in 209 peritoneal surface malignancies eductive surgery for the treatment of peritoneal carcinomatosis:
treated with closed abdomen technique. Cancer 2006;106:1144– Morbidity and mortality analysis of 216 consecutive procedures.
1153. Ann Surg Oncol 2003;10:863–869.
3. Clavien PA, Sanabria JR, Strasberg SM: Proposed classification 10. Feldman L, Barkun J, Barkun A, et al.: Measuring postoperative
of complications of surgery with examples of utility in complications in general surgery patients using an outcomes-
cholecystectomy. Surgery 1992;111:518–526. based strategy: Comparison with complications presented at
4. Elias D, Matsuhisa T, Sideris L, et al.: Heated intra-operative morbidity and mortality rounds. Surgery 1997;122:711–719;
intraperitoneal oxaliplatin plus irinotecan after complete resection discussion 719–720.
of peritoneal carcinomatosis: Pharmacokinetics, tissue distribu- 11. Loughlin KG, Moore LF. Using Delphi to achieve congruent
tion and tolerance. Ann Oncol 2004;15:1558–1565. objectives and activities in a pediatrics department. J Med Educ
5. Bozzetti F, Braga M, Gianotti L, et al.: Postoperative enteral 1979;54:101–106.
versus parenteral nutrition in malnourished patients with gastro- 12. Keeney S, Hasson F, McKenna H. Consulting the oracle: Ten
intestinal cancer: A randomised multicentre trial. Lancet 2001; lessons from using the Delphi technique in nursing research.
358:1487–1492. J Adv Nurs 2006;53:205–212 (Review).

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:258–262

The Fifth International Workshop on Peritoneal Surface Malignancy (Milan, Italy,


December 4–6, 2006): Methodology of Disease-Specific Consensus

DARIO BARATTI, MD, SHIGEKI KUSAMURA, MD, PhD, AND MARCELLO DERACO, MD*
Department of Surgery, National Cancer Institute, Milan, Italy

Peritoneal surface malignancies (PSM) have been traditionally regarded as uniformly terminal conditions. The combination of cyto-reductive
surgery and perioperative intraperitoneal chemotherapy has changed PSM management from palliation to possible cure. Due to the inherent
differences in biological and clinical behavior, the optimal adaptation of comprehensive treatment to each PSM is still a matter of debate. A
session of ‘‘The Fifth International Workshop on Peritoneal Surface Malignancy’’ (Milan, Italy, December 4–6, 2006) was committed to reach a
consensus pertaining to conceptual and technical aspects of the loco-regional treatment of each PSM. The consensus developing process was
based on principles of the Delphi method. A total of 103 international experts from 17 countries were included in six Working Groups (WG) for
each of the following PSM: peritoneal mesothelioma, abdominal sarcomatosis, carcinomatosis of gastric, colo-rectal, appendiceal, and ovarian
origin. Evidence reports were written by the respective WG. The main conflicting points (CP) regarding preoperative evaluation, patient
eligibility, combined treatment methodology, postoperative follow-up and future investigational perspectives were summarized as a list of
multiple-choice questions. Overall, 160 CP were identified. A consensus 51% of voters favoring one option was reached in 143/160 CP (89.4%).
The general treatment guidelines and future investigational perspectives were defined.
J. Surg. Oncol. 2008;98:258–262. ß 2008 Wiley-Liss, Inc.

KEY WORDS: consensus; peritoneal surface malignancies; cytoreductive surgery; hyperthermic intraperitoneal
chemotherapy; HIPEC

INTRODUCTION In the third session, a discussion has been conducted regarding the
disciplines indirectly involved in the local-regional therapy, such as
The Fifth International Workshop on Peritoneal Surface cellular and molecular biology, anaesthesiology, clinical nutrition
Malignancy took place in Milan (Italy) on December 4, 5 and 6, and quality of life, nursing, to define the state of the art and future
2006. The meeting was entitled ‘‘Integrating Cytoreductive Surgery prospects.
and Hyperthermic Intra-peritoneal Chemotherapy into the Manage- The consensus developing process was based on the principles of
ment of Peritoneal Malignancies: a Consensus Meeting.’’ the Delphi method, supported by summarized evidence reports. The
Since the first International Workshops was held in London in Delphi method has been used widely in health research and in clinical
1998, a meeting of international centres and health care professionals practice developing as a mean to assess the extent of agreement
currently involved or interested in the management of peritoneal (consensus measurement) and to resolve disagreement (consensus
surface malignancy (PSM) took place every 2 years [1]. International development). The method involves the participation of experts who
collaboration has played a prominent role in the progress of peritoneal are invited to provide opinions on a specific topic, based on their
surface oncology. The Peritoneal Surface Oncology Group Interna- knowledge and experience, and the use of anonymous questionnaires
tional (PSOGI) includes primarily surgical oncologists, but also where the main headings and statements are draft for circulation and
pathologists, gynaecologists, medical oncologists, radiologists, anaes- ranking to all participants. The process occurs in consecutive rounds,
thesiologists, biologists, nutrition specialists, as well as nurses and allowing the participants to know the distribution of the group’s
perfusionists from all over the world. PSOGI was founded in order to response and to change their opinions [2,3].
facilitate the collaboration among groups from different countries, This article summarizes and discusses the most relevant methodo-
bring together a broad expertise, generate new scientific hypothesis, logical issues of the Specific Disease Consensus Session, along with
accumulate theoretical and practical resources and build an interna- the results pertaining to the organization of the consensus attain-
tional network for the design of future prospective controlled studies. ment process. Selected topics regarding each PSM discussed during
The workshop was organized into three main sessions. The the Workshop are covered in detail by the articles contained in
Methodological Consensus Session was committed to reach a consensus this monography and authored by the speakers who presented each of
pertaining to conceptual and technical aspects of the local-regional them.
treatment of PSM. The objectives of the Specific Disease Consensus
Session were to define the state of the art of the operative methodology,
the general treatment guidelines and the future investigational per-
spectives for the following PSM: The authors have no financial interest related to the contents of this article to
disclose.
(1) peritoneal mesothelioma; *Correspondence to: Dr. Marcello Deraco, MD, Istituto Nazionale Tumori
(2) peritoneal dissemination from mucinous appendiceal neoplasms Milano, Via Venezian,1 20133 Milano, Italy. Fax: þ39-02-23902404.
(pseudomyxoma peritonei); E-mail: marcello.deraco@istitutotumori.mi.it
(3) carcinomatosis from colorectal cancer; Received 19 March 2008; Accepted 21 March 2008
(4) carcinomatosis from gastric cancer; DOI 10.1002/jso.21056
(5) carcinomatosis from ovarian cancer; Published online in Wiley InterScience
(6) abdominal sarcomatosis. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Consensus on Peritoneal Malignancies 259

DEVELOPING THE identification of the main conflicting points, along with the possible
DISEASE-SPECIFIC CONSENSUS alternatives supported by the current scientific literature.
The consensus document elaboration guidelines included the
The disease-specific consensus attainment process has been following topics:
conducted during the whole year of 2006. It was organized into a
few consecutive steps, which took place partly during the months . Preoperative evaluation: to define a brief clinical pathway guiding
before the meeting and partly during the Workshop itself. The whole the physician to choose the best instruments (imaging studies,
process was coordinated by the Workshop President, the Workshop laboratory tests, invasive procedures) for the diagnosis and the
Director (MD) and the International Scientific Committee (ISC), which staging of the disease entity being considered.
included previous Workshop Directors and internationally renown . Eligibility: to identify the circumstances of the natural history of
experts on local-regional therapy from the main centres. The Scientific each single disease that could be amenable to be treated by cyto-
Secretariat (SS) and the Italian Organizing and Scientific Committee reductive surgery (CRS) and peri-operative intra-peritoneal che-
(IOSC) provided scientific and organizational support. motherapy (PIC), as well as the selection criteria of patients most
likely to benefit from a combined modality treatment approach. The
identification of quantitative prognostic indicators and risk factors
Working Groups
for major morbidity was also addressed.
The first step of the consensus development process was the . State of the art of the treatment methodology: to delineate the
composition of six Working Groups (WG). The WG are multi- technical variations of the procedure of CRS and PIC that could best
disciplinary international teams which received the responsibility to fit in the treatment of each single disease, based on the results of the
conduct the discussion about the role of loco-regional therapy in the Methodological Consensus. A special attention was paid on the
clinical management of each single PSM. Two coordinators for each possible role of adjuvant and neo-adjuvant systemic treatments.
WG, as well as the other members, were elected by the SS and the ISC . Definition of the scientific evidence: the degree of the scientific
based on their expertise in the management of PSM and additional evidence of the indication and methodology of the combined
parameters, such as the institutional role in the country of origin and modality treatment approach of PSM, according to a five level
the scientific publications regarding the disease being considered. ranking, as shown in Table II [3].
To guarantee a multidisciplinary feature and to enlarge the base of . Follow-up: to identify the most rational postoperative surveillance
the consensus, not only surgical oncologists directly involved in the schedule and the diagnostic tools most suitable to assess therapeutic
local-regional treatment of PSM, but also medical oncologists, response or disease progression after comprehensive local-regional
pathologists and other specialists were included in the WG. Analo- treatment.
gously, also physicians not necessarily favorable to the local-regional . Future perspectives: to define both basic and clinical potential
comprehensive treatment were involved to strengthen the external investigational lines, with a special focus on drafting prospective
validity of the consensus. The Working Group Coordinators had the right randomized controlled trials.
to modify the list of the members of their WG according to their personal
All the consensus documents were available on the Workshop web
criteria.
site (http://www.peritonealworkshop2006.com) for consultation.
The composition of the six disease specific Working Groups is
The main conflicting points were organized according to the same
detailed in Table I, along with the represented peritoneal malignancy
topics addressed in the consensus documents (preoperative evaluation,
treatment centres and countries of origin. Overall, 12 WG coordinators
patient eligibility, combined treatment methodology, postoperative
and 91 members from 69 centres of 17 different nations were involved to
follow-up, and future investigational perspectives) and were summarized
express their opinion through the web-based vote. They were 58 surgical
as a list of multiple choice questions. The respective possible solutions
oncologists, 16 gynaecologic oncologists, 20 medical oncologists,
were given to be examined and selected by WG members. In Table III
5 pathologists, and 3 other health care professionals.
the conflicting points are displayed by specific topics and disease
process. A total of 160 conflicting points were identified.
Consensus Elaboration
Web-Based Voting
On August 11, 2006 the beginning of the activity of the six Working
Groups was announced. The goal of this phase was the elaboration of a Once the consensus documents and the conflicting points were
consensus document regarding the specific disease of interest and the available on the web, the members of each WG were involved to

TABLE I. Working Groups Composition

WG WG Represented Represented
Working groups coordinators members centres countries

Abdominal sarcomatosis 2 15 12 5
Colo-rectal cancer carcinomatosis 2 21 17 10
Gastric cancer carcinomatosis 2 17 16 7
Ovarian cancer carcinomatosis 2 26 22 11
Peritoneal mesothelioma 2 19 15 5
Pseudomyxoma peritonei 2 15 14 8

12 91a 69b 16a

WG, working group.


a
Some members were involved in more than 1 WG.
b
More members were affiliated to the same center.

Journal of Surgical Oncology


260 Baratti et al.
TABLE II. Evidence Based Methodology: Type of Basis
‘‘TYPE C basis’’ (General consensus)
There is a widespread consolidated consensus. Randomized trials have not been carried out or have been inadequate, but the issue is settled without major
controversy: currently, no (further) experimental evidence is felt to be needed.
‘‘TYPE 1 evidence’’ (Randomized trial(s) available, strong evidence)
Consistent results have been provided by more than one randomized trials, and/or a reliable meta-analysis was performed. In some instances, one randomized trial
can be considered sufficient to support this type of evidence.
Further confirmatory trials do not seem necessary.
‘‘TYPE 2 evidence’’ (Randomized trial(s) available, weak evidence)
One or more randomized trials have been completed, but the evidence they provide is not considered definitive (their results are not consistent, and/or they are
methodologically unsatisfactory, etc.). Some controlled evidence has therefore been provided, but confirmatory trials would be desirable.
‘‘TYPE 3 evidence’’ (External controlled comparisons available)
Evidence is available from non-randomized studies, with external controls allowing comparisons. Some uncontrolled evidence has therefore been provided, but
trials would be desirable.
‘‘TYPE R basis’’(Rational inference)
Little or no direct evidence from clinical studies is available. Yet clinical conclusions can be rationally inferred from available data and knowledge (e.g., by
rationally combining pieces of information from published studies and observations; for a rare neoplasm, or presentation, through analogy with a related, more
common tumor, or presentation; etc.). The inference can be more or less strong, and trials may, or may not, be desirable (although sometimes unfeasible).

express their opinion through a web-based voting system. They were In Table IV the results of the consensus attainment process are
asked to browse the sequence of options and choose the one they found summarized. A consensus of at least 51% of voters favoring one of the
more appropriate to every specific conflicting point. The literature proposed options was reached in 143/160 conflicting points (89.4%). In
references were accessible online during the voting procedure, as well detail, consensus was obtained in 96.1% of the conflicting points
as the consensus document pertaining to every single PSM. The pertaining to pseudomyxoma peritonei and abdominal sarcomatosis.
definition of consensus was at least 51% of voters favoring one option. Concerning the specific topics, the best agreement was observed for
The WG coordinators were informed on the results of the web-based follow-up. By contrast, the higher rate of disagreement was recorded
voting. for ovarian cancer carcinomatosis and preoperative evaluation.
The Workshop President were responsible for the final approval of
Workshop Discussion and Voting the Specific Disease Consensus.

The final phases of the consensus attainment process took place


DISCUSSION
during the Workshop. A distinct plenary session was dedicated to
every single peritoneal malignancy. The state of the art and the basic During the last decades, better knowledge of the natural history,
and clinical research perspectives pertaining to each PSM were innovative surgical techniques and the advances in loco-regional
summarized by the coordinators of the respective WG. During chemotherapy have resulted in a new treatment strategy for peritoneal
each Disease Consensus plenary session, the preliminary results of surface malignancies. Improved understanding of the predictable
the expert web-based vote were presented and discussed by all the patterns of intra-abdominal disease redistribution, the observation that
Workshop participants. mobile surfaces of small bowel and its mesentery may lastingly remain
According to the Delphi method, a second round of voting was free of tumor and the notion that PSM may be confined within the
accomplished during the Workshop. All the participants expressed peritoneal cavity during most of their clinical course have made an
their opinion through a digital voting system on the same conflicting intensive loco-regional treatment approach attractive [4]. Periton-
points voted during the preliminary phase. ectomy procedures to remove all the visible peritoneal tumor
dissemination have been greatly popularized by Sugarbaker [5] and
Consensus Approval standardized methods to deliver intra-operative hyperthermic intra-
peritoneal chemotherapy have been developed to prevent the progres-
The results of the digital vote was immediately available for sion of microscopic residual disease [6].
evaluation and further discussion during each session. The Interna- The strong treatment rationale and striking clinical results have led
tional Scientific Committee had the responsibility to synthesize the to the establishment of many highly qualified PSM management
discussion about state of the art and the future investigational centers in the USA, Australia, Asia, and nearly all countries in Europe.
perspectives of the local-regional treatment for each peritoneal The most common peritoneal surface malignancies which have
malignancy and to analyze and validate the results of voting. been treated are carcinomatosis of colorectal, gastric, ovarian and

TABLE III. Conflicting Points by Specific Topics and Peritoneal Surface Malignancy

Preoperative Patient State of the art of Future


Working groups evaluation eligibility the methodology Follow-up perspectives Total

Abdominal sarcomatosis 4 6 10 4 2 26
Colo-rectal cancer carcinomatosis 3 7 9 4 3 26
Gastric cancer carcinomatosis 3 11 10 4 4 32
Ovarian cancer carcinomatosis 2 9 11 2 2 26
Peritoneal mesothelioma 2 7 9 4 2 24
Pseudomyxoma peritonei 4 5 12 4 1 26

18 45 61 22 14 160

Journal of Surgical Oncology


Consensus on Peritoneal Malignancies 261
TABLE IV. Results of the Disease Specific Consensus Attainment Process

Preoperative State of the art of


Working groups evaluation Patient eligibility the methodology Follow-up Future perspectives Total

Abdominal sarcomatosis 4/4 5/6 10/10 4/4 2/2 25/26 (96.1%)


Colo-rectal cancer carcinomatosis 2/3 6/7 9/9 4/4 3/3 24/26 (92.3%)
Gastric cancer carcinomatosis 2/3 10/11 10/10 4/4 4/4 30/32 (93.7%)
Ovarian cancer carcinomatosis 2/2 6/9 8/10 1/2 1/2 18/26 (69.2%)
Peritoneal mesothelioma 2/2 6/7 7/9 4/4 2/2 21/24 (87.5%)
Pseudomyxoma peritonei 3/4 5/5 12/12 3/3 1/1 25/26 (96.1%)

15/18 (83.3%) 38/45 (84.4%) 58/61 (95.1%) 21/22 (95.4%) 13/14 (92.8%) 143/160 (89.4%)

appendiceal origin, peritoneal mesothelioma and peritoneal sarcoma- through the use of intra-peritoneal chemotherapy was declared the
tosis. These disease processes represent different epidemiological, standard of practice by the National Cancer Institute, Bethesda, USA
biological, and clinical conditions. The optimal adaptation of CRS after a recent phase III study [20].
and PIC to each PSM is still a matter of clinical investigations. This Peritoneal involvement as an isolated site of tumor progression most
prompted us to address the main conceptual and methodological issues commonly results from GISTs or retroperitoneal sarcomas. At the
separately for each disease entity. moment, medical therapy is the standard of care for these patients and
Colorectal cancer ranks third in incidence in both men and women the combination of surgery and PIC should be considered investi-
in the USA and western Europe as well (http://seer.cancer.gov/) [7]. gational [21]. A clear distinction can be made between GIST and other
Isolated peritoneal carcinomatosis is encountered in 7–10% of patients adult soft-tissue sarcomas, because the natural history of GIST has
at primary surgery and in 4–19% of those who recur after curative been deeply affected by the medical therapy [22]. The major benefit
surgery [8]. Ovarian cancer ranks seventh in women in USA and up to gained from Imatinib, the availability of further second-line targeted
75% of patients are diagnosed with an advanced stage disease (http:// agents and the lack of formal proofs of efficacy of surgery for residual
seer.cancer.gov/). Gastric cancer accounts for 9.9% of all new cancer disease, makes it difficult, at the moment, to define the role of
diagnoses in the USA and it is the second leading cause of cancer- combined loco-regional therapies in advanced GIST patients, even
related mortality worldwide [9]. It has been estimated that 15–50% of when the disease is confined to the peritoneum.
patients have a peritoneal dissemination at surgical exploration, The results of CRS and PIC should be compared cautiously to
especially when there is serosal involvement by the tumor [10]. traditional therapies since, with only few exceptions, these treatment
Conversely, pseudomyxoma peritonei and peritoneal mesothelioma are strategies have not been directly compared [19,20]. More randomized
extremely uncommon tumor with an annual incidence of approxi- controlled studies may be difficult to undertake with exceedingly
mately 1/1,000,000 and 2.2/1,000,000 [11,12]. Sarcomas represent 1% uncommon tumors such as pseudomyxoma peritonei, sarcoma and
of all newly diagnosed cancer and abdominal sarcomas account for peritoneal mesothelioma. Furthermore, phase III trials would compare
10–15% of all-type sarcoma (http://seer.cancer.gov/). a potentially curative treatment with merely palliative procedures and
Every PSM is a clinicopathologic entity with a unique natural patients would presumably be not prone to accept randomization.
history. Before the era of cytoreduction and PIC, such conditions were Particularly with a slow progressing condition such as pseudomyxoma
generally treated by palliative/debulking surgery and systemic peritonei, a long-term follow-up of at least 10–20 years is required to
chemotherapy. In historical series, median survival was 5–6 months demonstrate a statistical difference. However, the fact that randomized
for colorectal cancer carcinomatosis [13], 9–12 months for peritoneal trials are difficult to perform does not imply that more information
mesothelioma [12,14], 1.5–3.1 months for gastric cancer carcinoma- from multi-institutional international phase III trial would not be
tosis [15], 35–38 months for advanced ovarian cancer [16]. The desirable.
majority of pseudomyxoma peritonei are minimally aggressive and In an attempt to overcome such difficulties, we planned a Specific
they rarely cause lymphatic or hematogenous metastases. However, Disease Consensus Session to define the state of the art of the operative
previous reports demonstrated that serial debulking surgery with methodology, the general treatment guidelines and the future investi-
various adjuvant therapies can only achieve a long-term survival of gational perspectives for each single PSM. Consensus making is an
20–30% [11,17]. alternative method of dealing with conflicting scientific evidence.
Results of comprehensive treatment approach vary by indication. Considerable experience in using Delphi consensus technique exists,
Pseudomyxoma peritonei is the ‘‘classic’’ indication. In prospective since it has been widely used in health research, especially within the
case-series treated by cytoreductive surgery and PIC, 5-year survival fields of technology assessment, education training or nursing and
rates varied from 52% to 96% and median survival from 51 to clinical practice developing [23].
156 months [17]. To date, there is an unquestionable consensus that Consensus methods consider a wider range of scientific studies
this combined treatment can be considered standard of care for all than conventional statistical reviews and allow a greater role for the
cases of mucinous appendiceal neoplasms with peritoneal dissemina- qualitative assessment of evidence [2,3]. They try to overcome
tion amenable to potentially complete surgical cytoreduction [11,18]. the disadvantages normally found with decision making in groups
Especially in recent years, as the comprehensive approach or committees, which are commonly dominated by individuals or
pioneered by Sugarbaker has expanded, the treatment results of coalitions representing specific interests. Delphi method enables a
peritoneal mesothelioma have dramatically improved. The combina- large group of experts to be contacted inexpensively by mail or by
tion of CRS and PIC has reportedly resulted in a median survival of internet with a self administered questionnaire, with few or no
34–92 months [14]. geographical limitations. Rounds in which the participants meet to
Multiple trials suggest that outcomes after CRS and PIC are better discuss the process and resolve uncertainty or any ambiguities in the
for colorectal than for gastric cancer [8,15]. In patients with colon wording of the questionnaire may be included.
cancer carcinomatosis, a phase III trial have confirmed the superiority For studies concerned with defining criteria for clinical interven-
of such treatment approach over standard palliative therapies for tion, the most appropriate experts will be clinicians practising in the
cancer [19]. Treatment of ovarian cancer peritoneal dissemination field under discussion. However, the inclusion of other clinicians, not

Journal of Surgical Oncology


262 Baratti et al.
necessarily favorable to the debated topic may be appropriate to 11. Sugarbaker PH: New standard of care for appendiceal epithelial
provide an alternative clinical view, particularly when the study is neoplasms and pseudomyxoma peritonei syndrome? Lancet
expected to have an impact beyond a particular specialist field. Oncol 2006;7:69–76.
In conclusion, the disease-specific consensus process brought 12. Deraco M, Baratti D, Zaffaroni N, et al.: Advances in clinical
research and management of diffuse peritoneal mesothelioma.
together internationally renowned experts in the field of peritoneal Recent Results Cancer Res 2007;169:137–155.
neoplasms, team members of existing PSM treatment centers and 13. Sadeghi B, Arvieux C, Glehen O, et al.: Peritoneal carcinomatosis
physicians planning to start new programs. Controversial issues from non-gynecologic malignancies: Results of the EVOCAPE 1
regarding the state of the art of the operative methodology, the general multicentric prospective study. Cancer 2000;88: 358–363.
treatment guidelines and the future investigational perspectives 14. Yan TD, Welch L, Black D, et al.: A systematic review on the
were summarized and discussed through a multiple choice question efficacy of cytoreductive surgery combined with perioperative
digital system. A high rate of agreement was reached by the intraperitoneal chemotherapy for diffuse malignant peritoneal
participants. mesothelioma. Ann Oncol 2007;18:827–834.
15. Yan TD, Black D, Sugarbaker PH, et al.: A systematic review and
meta-analysis of the randomized controlled on adjuvant intra-
REFERENCES peritoneal chemotherapy for resectable gastric cancer. Ann Surg
Oncol 2007;14:2702–2713.
1. González-Moreno S: Peritoneal surface oncology: A progress 16. McGuire WP, Hoskins WJ, Brady MF, et al.: Cyclophosphamide
report. Eur J Surg Oncol 2006;32:593–596. and cisplatin compared with paclitaxel and cisplatin in patients
2. Jones J, Hunter D: Consensus methods for medical and health with stage III and stage IV ovarian cancer. N Engl J Med 1996;
services research. Br Med J 1995;311:376–380. 334:1–6.
3. Casali P, Licitra L, Tondini C, et al.: START: A European state-of- 17. Yan TD, Black D, Savady R, et al.: A systematic review on the
the-art on-line instrument for clinical oncologists. Ann Oncol efficacy of cytoreductive surgery and perioperative intraperitoneal
1999;10:769–773. chemotherapy for pseudomyxoma peritonei. Ann Surg Oncol
4. Esquivel J, Sticca R, Sugarbaker P, et al.: Society of Surgical 2007;14:484–492.
Oncology Annual Meeting. Cytoreductive surgery and hyper- 18. Sugarbaker PH, Yan TD, Stuart OA, et al.: Comprehensive
thermic intraperitoneal chemotherapy in the management of management of diffuse malignant peritoneal mesothelioma. Eur J
peritoneal surface malignancies of colonic origin: A consensus Surg Oncol 2006;32:686–691.
statement. Society of Surgical Oncology. Ann Surg Oncol 2007; 19. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of
14:128–133. cytoreduction and hyperthermic intraperitoneal chemotherapy
5. Sugarbaker PH: Peritonectomy procedures. Ann Surg 1995;221: versus systemic chemotherapy and palliative surgery in patients
29–42. with peritoneal carcinomatosis of colorectal origin. J Clin Oncol
6. Stewart JH IV, Shen P, Levine EA: Intraperitoneal hyperthermic 2003;21:37–43.
chemotherapy for peritoneal surface malignancy: Current status 20. Armstrong DK, Bundy B, Wenzel L, et al.: Intraperitoneal
and future directions. Ann Surg Oncol 2005;12:765–777. cisplatin and paclitaxel in ovarian cancer. N Engl J Med 2006;
7. Boyle P, Ferlay J: Cancer incidence and mortality in Europe, 354:77–79.
2004. Ann Oncol 2005;16:481–488. 21. Bonvalot S, Cavalcanti A, Le Péchoux C, et al.: Randomized trial
8. Koppe MJ, Boerman OC, Oyen WJG, et al.: Peritoneal of cytoreduction followed by intraperitoneal chemotherapy versus
carcinomatosis of colorectal origin: Incidence and current treat- cytoreduction alone in patients with peritoneal sarcomatosis. Eur
ment strategies. Ann Surg 2006;243:212–222. J Surg Oncol 2005;31:917–923.
9. Parkin DM, Pisani P, Ferlay J: Global cancer statistics. CA Cancer 22. Rankin C, Von Mehren M, Blanke C, et al.: Dose effect of
J Clin 1999;49:33–64. imatinib in patients with metastatic GIST: Phase III sarcoma
10. Moriguchi S, Maehara Y, Korenaga D, et al.: Risk factors group studyS0033. J Clin Oncol 2004;22:819.
which predict pattern of recurrence after curative surgery for 23. Keeney S, Hasson F, McKenna H: Consulting the oracle: Ten
patients with advanced gastric cancer. Surg Oncol 1992;1:341– lessons from using the Delphi technique in nursing research.
346. J Adv Nurs 2006;53:205–212.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:263–267

Consensus Statement on the Loco Regional Treatment of Colorectal


Cancer With Peritoneal Dissemination

JESUS ESQUIVEL, MD, FACS,1* DOMINIQUE ELIAS, MD,2 DARIO BARATTI, MD,3
SHIGEKI KUSAMURA, MD, PhD,3 AND MARCELLO DERACO, MD3
1
St. Agnes Hospital, Baltimore, Maryland,
2
Département de chirurgie générale oncologique, Institut Gustave-Roussy, Villejuif, France
3
Department of Surgery, National Cancer Institute of Milan, Italy

Medical management with combinations of cytotoxic chemotherapy, and/or biological agents, has resulted in an unprecedented median survival
>20 months in patients with Stage IV colorectal cancer. The management of disease limited to the peritoneal cavity has been controversial and at
the present time, there is no published data that outlines the impact of these new therapeutic regimens when given to patients with colorectal
cancer with metastatic disease confined to the peritoneum. Over the last 5 years, an increasing number of international treatment centers
have published their prospective results using cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) in the management
of peritoneal surface malignancies of colorectal origin and have shown that good long-term results can be achieved with a complete cytoreduction
and HIPEC. However, most of the surgical data comes from Phase II studies from single institutions and there is a wide range on inclusion/
exclusion criteria, drugs, temperatures and methods of delivering the heated chemotherapy. This manuscript will analyze and discuss the results of
a group of health care providers trying to achieve a consensus statement in the management of this group of patients.
J. Surg. Oncol. 2008;98:263–267. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal carcinomatosis; intraperitoneal chemotherapy; cytoreductive surgery

INTRODUCTION techniques that include Peritonectomy Procedures, standardized


methods to deliver intraoperative HIPEC and better patient selection
Surgical resection remains the hallmark therapy for primary colon criteria, along with the strong treatment rationale and superior
cancer. It allows the patients to become clinically disease free, provides results when compared to historical controls, have lead to the
proper staging, and determines who should receive adjuvant systemic establishment of numerous treatment centers in the United States
chemotherapy. Treatment options for patients with unresectable and Europe. Over the last 5 years, an increasing number of
metastatic disease have improved significantly in the past few years. international treatment centers have published their prospective results
A review of the published data in the treatment of patients with Stage using cytoreductive surgery and HIPEC in the management of
IV colorectal cancer, outlining the surgical and medical therapeutic peritoneal surface malignancies of colorectal origin [6–10]. In 2003,
options demonstrates that medical management, with combinations of the Dutch group conducted a randomized controlled trial comparing
cytotoxic chemotherapy, and/or biological agents, has resulted in an systemic chemotherapy with cytoreductive surgery combined with
unprecedented Median Survival >20 months [1]. Peritoneal involve- HIPEC and this trial clearly demonstrated the superiority in survival of
ment in colorectal cancer occurs in approximately 30% of patients. the combined treatment group [11]. In 2004, a multi-institutional
Approximately 8% of patients are diagnosed with synchronous registry study from 28 international treatment centers showed that the
peritoneal dissemination at the time of primary colorectal surgery median survival was 19 months and 3-year survival was 39% after
and 25% of patients have recurrence confined to the peritoneal cavity cytoreductive surgery and HIPEC for 506 patients with colorectal
[2]. The management of disease limited to the peritoneal cavity has peritoneal carcinomatosis [12].
been controversial. However, at the present time, there is no published An analysis of evidence indicates prolongation of survival and
data that outlines the impact of these new therapeutic regimens when potential for cure in patients with low volume metastatic adeno-
given to patients with colorectal cancer with metastatic disease carcinoma of colonic origin limited to the peritoneal cavity using
confined to the peritoneum. Therefore, systemic treatment alone is an cytoreductive surgery and HIPEC. However, as pointed out by Yan et al.
unproven therapeutic strategy for this particular group of Stage IV in a recent systematic review of the quality of evidence in 14 rigorously
patients with limited peritoneal dissemination from a primary or
recurrent colon cancer.
The present paper will focus on the available scientific evidence of
the role of cytoreductive surgery and hyperthermic intraperitoneal The authors have no financial interest related to the contents of this article to
chemotherapy (HIPEC) in the management of peritoneal surface disclose.
malignancies of colorectal origin and will include the results of the *Correspondence to: Jesus Esquivel, MD, FACS, Surgical Oncology, St.
voting on conflicting points by 22 experts in the disease specific Agnes Hospital, 900 Caton Avenue, Baltimore, MD 21229. Fax: 410-951-
workshop group in colorectal cancer. 4007. E-mail: jesusesquivel@yahoo.com
In the 1990s, Sugarbaker and colleagues proposed cytoreductive Received 19 March 2008; Accepted 21 March 2008
surgery and perioperative intraperitoneal chemotherapy as a definitive DOI 10.1002/jso.21053
treatment for peritoneal dissemination from appendiceal neoplasms Published online in Wiley InterScience
and diffuse malignant peritoneal mesothelioma [3–5]. Better surgical (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


264 Esquivel et al.
selected series, there were 2 randomized controlled trials, 1 randomized Reviewing the published data helps us learn about the outcome of
comparative study, and 11 observational studies without control groups, patients with colorectal cancer with peritoneal dissemination treated
including one multi-institutional study [13]. In addition, the indications with cytoreductive surgery and HIPEC. There is no question that
for, the technique of HIPEC, the drugs used and the degree of heat, vary 2-year survival rates are around 60% and 5-year survival rates approach
among physicians using these methods of treatment. 40% in many series when a complete cytoreduction can be achieved.
A recent international conference on this subject was convened However, review of the data also teaches us about the biology of the
and a consensus statement on the appropriate use of cytoreductive tumor as we all continue to have incomplete cytoreductions with an
surgery and HIPEC was developed and adopted by the Peritoneal expected median survival of about 6 months. At the present time, we
Surface Malignancy Group (PSMG) in an attempt to standardize the lack a universally accepted and easily reproducible prospective staging/
indications and techniques for this treatment, Figure 1. We agreed that scoring system that addresses the quantity and location of the peritoneal
the main strategy would be to use the peritonectomy procedures dissemination and its impact on achieving a complete cytoreduction. We
necessary to achieve a complete cytoreduction. After all resections also do not have a universal agreement on inclusion/exclusion criteria.
were done then the hyperthermic chemotherapeutic perfusion would be Some centers would consider patients with a few liver metastases
carried out. We agreed that most of us use Mitomycin C with some not candidates for this procedure while other centers would consider this
significant variations regarding dosage, temperatures and methods of situation an absolute contraindication.
delivery. We also recognize that there is some exciting and provocative In the absence of a large prospective randomized trial that addresses
data with the use of intraperitoneal Oxaliplatin and with the systemic all the above-mentioned issues, it is going to be difficult to demonstrate
use of biological agents [14]. the true impact of cytoreductive surgery and HIPEC on the natural

Fig. 1. Clinical pathway for the management of peritoneal surface malignancies of colonic origin.

Journal of Surgical Oncology


Colorectal Cancer With Carcinomatosis 265
biologic behavior of colorectal cancer with peritoneal dissemination, peritoneal carcinomatosis is accurate to assess the complete re-
especially in the era of newer systemic therapy, keeping cytoreductive sectability of peritoneal carcinomatosis in patients for which there
surgery and HIPEC still as an unproven and frequently questioned is inadequate or contradictory information concerning disease
therapeutic treatment modality. extent.

PRE-OPERATIVE EVALUATION VARIABLES ASSOCIATED WITH INCREASED


CHANCES OF HAVING A COMPLETE
Proper patient selection remains a crucially important aspect in
the treatment of patients with peritoneal dissemination from colo- CYTOREDUCTION
rectal cancer. As previously mentioned, long-term survival can only The following represent clinical and/or radiographic variables that
be achieved with cytoreductive surgery and HIPEC when a complete are usually associated with an increase chance of achieving a complete
cytoreduction is accomplished. Review of the data in multiple removal of all tumors during cytoreductive surgery.
series shows that those patients that have an incomplete removal of
their peritoneal dissemination have a median survival of about
ECOG Performance status 2 or less
6 months and therefore these patients do not benefit from a surgical
No evidence of extra-abdominal disease
procedure [11,12]. In addition, they represent a financial burden to the
Up to 3 small, resectable parenchymal hepatic metastases
institution.
No evidence of biliary obstruction
Once a patient has been diagnosed with colorectal cancer with
No evidence of ureteral obstruction
peritoneal involvement, the work-up usually includes a complete
No evidence of intestinal obstruction at more than one site
colonoscopy as well as a CT scan of the chest, abdomen and pelvis
Small bowel involvement: No evidence of gross disease in the
with maximum oral and intravenous contrast to evaluate the extent of
mesentery with several segmental sites of partial obstruction.
peritoneal dissemination. A PET scan can be considered if there is any
Small volume disease in the gastro-hepatic ligament.
question of extra-abdominal disease. Review of two published series
trying to address the diagnostic accuracy of the CT scan as an imaging
modality can be summarized by stating that the detection of peritoneal These data represent characteristics that most peritoneal surface
carcinomatosis by CT scan is only moderately useful and that it malignancy centers agreed upon but it has not been submitted to a
has severe limitations in detecting small peritoneal implants, especially prospective analysis. With the establishment of this consensus and the
in the small intestine. In addition, CT scan was considered of participation of most of the international groups, we hope we can
limited value in selecting colorectal patients with peritoneal carcino- validate these clinical and/or radiographic variables.
matosis who will not benefit from cytoreductive surgery with HIPEC
[15,16].
ELIGIBILITY CRITERIA
Establishing clear eligibility criteria for any given treatment
ROLE OF ABDOMINAL LAPAROSCOPY IN plan seems obvious in order to maximize benefits and minimize
THE PRE-TREATMENT EVALUATION OF unnecessary treatments. The indications for cytoreductive surgery
PERITONEAL CARCINOMATOSIS and HIPEC in the treatment of peritoneal surface malignancies
of colorectal origin have not been clearly defined and at the
Garofalo et al. reported on 97 cases of peritoneal carcinomatosis present time range from being indicated because the patient is in
submitted to video laparoscopy to stage the disease. They achieved full good physical condition to withstand this particular treatment to the
laparoscopic Peritoneal Cancer Index assessment in 96/97 cases, while patients that have failed every other possible treatment and are looking
only 2/96 cases were under staged. There was a good correlation for a last chance.
between the open successive surgery data and the laparoscopic Up to date, there are no published indications for the role of
Peritoneal Cancer Index. There was no mortality and no neoplastic cyotreductive surgery and HIPEC in the treatment of peritoneal surface
colonization at the trocar port site. Patients with massive involvement malignancies of colorectal origin. The medical oncology community,
of their small bowel or mesentery by staging laparoscopy should be labels these patients as Stage IV disease and extrapolate data from the
considered not amenable for peritonectomy. They considered laparo- efficacy of modern combinations of cytotoxic and biological agents as
scopy a useful tool in peritoneal surface malignancies. It allows direct their rationale to treat patients with colon cancer metastatic to the
visualization even of small cancer nodules and provides a reliable peritoneum. Unfortunately, they fail to realize that the vast majority of
assessment of the feasibility of peritonectomy [17]. the patients on the trials that generated these data, were patients with
A group of French investigators also evaluated the role of metastatic extra-abdominal disease and the second largest group were
explorative laparoscopy to evaluate candidates for complete resection patients with liver metastases.
of peritoneal carcinomatosis combined with HIPEC. Eleven patients On the other hand, an analysis of the published data by the surgical
planned to undergo a cytoreductive surgery þ HIPEC underwent an oncology community shows that the indications are very vague and
explorative laparoscopy. Laparoscopic evaluation was successful in all none specific like, having a good performance status and few
11 patients. The median operating time was 38 min (range 23–75 min). comorbidities in order to survive the operation. We have tried to
The laparoscopic examinations were well tolerated in all cases. For identify those patients in whom this particular treatment option is not
3 patients, the peritoneal carcinomatosis was as judge as unresectable. indicated, like in patients with extra-abdominal disease, or patients
A complete resection of the peritoneal carcinomatosis combined with multiple sites of intestinal obstruction or patients with too much
with HIPEC was performed in seven out of the eight patients with disease. Sugarbaker et al. evaluated the prognostic value of the
peritoneal carcinomatosis considered resectable at laparoscopy. One Peritoneal Cancer Index (PCI) in patients with colorectal cancer
patient was diagnosed with more extensive disease than that as undergoing cytoreductive surgery plus HIPEC. A PCI < 10 was
assessed by the evaluative laparoscopy [18]. Of note, in 20% of the associated with a 5-year survival rate of 50% while it was 0% if the
patients taken directly for a laparotomy, a complete cytoreduction PCI > 20 (P-value < 0.0001). Based on these data, they concluded that
was not possible. The conclusion was that laparoscopic scoring of cytoreductive surgery plus HIPEC were contraindicated in patients

Journal of Surgical Oncology


266 Esquivel et al.
with a PCI > 20 [19]. Verwaal et al. using their seven regions system Do you think there should be a PCI determination on CT scan?
found not only that the survival benefit was low in patients with more Yes 50%
than five regions involved but also that the morbidity was increased in Not sure 33%
No 16%
these patients [20].
Should a Peritoneal Cancer Index > 20 be an absolute exclusion
There are a few points on this subject that deserve attention.
criteria for CRS þ HIPEC?
The story of surgical treatment of colorectal liver metastases is Yes 33%
being rewritten with a different title. It would be impossible to think No 66%
that a medical oncologist would recommend systemic therapy for a Should Liver Metastases be an absolute exclusion criteria for
patient with a single liver metastasis 18 months after resection of the CRS þ HIPEC?
primary tumor as well as it would be impossible to think that a Yes 0%
surgical oncologist would favor surgical treatment in a patient with a No 66%
primary tumor and more than 20 liver metastases. In this particular Cannot answer 33%
Should ALL asymptomatic patients with CRC with PC receive
setting of metastatic disease, the indications appear to be well
systemic chemotherapy prior to CRS þ HIPEC?
described.
Yes 41.7%
When it comes to metastatic disease to the peritoneum, we face the No 58.3%
problem that not only we do not have a universally agreed upon scoring
system to follow the patients from time zero, but that it is very difficult
to measure with the current image modalities the amount and location
of the disease. Most of the patients that we treat, at least in the United T 4 N 0 M 1 (limited peritoneal dissemination)
States, have failed every possible combination of cytotoxic and
biological agents and come to us with a debilitated body and a depleted Best systemic therapy followed by surgery only to relieve symptoms 0%
bone marrow. However, we most address that these patients represent Best systemic therapy  3months followed by CRS þ HIPEC and BST 8.3%
the numerator and we do not know the denominator, making it very CRS þ HIPEC upfront followed by best systemic therapy 91.6%
difficult to have meaningful conclusions on the effect of systemic
therapy in patients with colorectal cancer metastatic to the peritoneum.
In addition, it is also difficult to measure the benefit of the procedure. T 4 N 2 (8/12) M1 (limited peritoneal dissemination)
We all have seen patients with intractable ascites that require weekly
paracentesis and that after cytoreductive surgery and HIPEC, live only Best systemic therapy followed by surgery only to relieve symptoms 0%
6 months but never needed to be tapped again. Palliative cytoreduction Best systemic therapy  3 months followed by CRS þ HIPEC and BST 50%
with HIPEC, plays a tremendous role in improving quality of life in CRS þ HIPEC upfront followed by best systemic therapy 50%
these patients.
In summary, we know that most of the patients with an incomplete
tumor removal will not benefit from cytoreductive surgery and HIPEC Asymptomatic patient with large omental cake, ascites, minimal
and face a median survival of 6 months. We also know that long-term small bowel involvement.
survival can only be achieved with a complete removal of the
peritoneal disease. Best systemic therapy followed by surgery only to relieve symptoms 0%
Best systemic therapy  3 months followed by CRS þ HIPEC and BST 33%
CRS þ HIPEC upfront followed by best systemic therapy 66%

RESULTS OF WEB-BASED VOTING ON


CONFLICTING POINTS Future Investigations
Pre-Operative Work-Up
Do you think it is necessary to perform a large Phase III trial in order to make
In addition to the basic work-up that includes a good history and
CRS þ HIPEC standard of care?
physical, blood work and a colonoscopy, 100% of the experts Yes 75%
agreed that the preferred imaging modality is a CT scan of the No 25%
abdomen and pelvis with maximum oral and intravenous contrast.
Regarding a CT scan of the chest, the vast majority, 83%, thought
that it was necessary to obtain such a study. Fifty-eight percent Of the following trials, which one would you recommend?
thought that a CT/PET scan should be performed in all patients. MRI
scans of the abdomen and pelvis, Ultrasound, Laparoscopy with
CRS and then HIPEC vs. no HIPEC both followed by best systemic 81.82%
biopsy, PET scan and upper endoscopy, were considered necessary by
therapy
less than 50% of the experts. More than 66% of the experts agreed that Best systemic therapy vs. CRS þ HIPEC 36.36%
it is necessary to obtain a CEA and a CA 19-9 level pre-operatively. All Best systemic therapy vs. CRS þ HIPEC vs. CRS with no HIPEC 18.18%
other tumor markers were recommended by less than 50% of the with the last two arms followed by best systemic therapy
experts.

Would you favor the following Phase II trial over a Phase III trial?
Eligibility
Best systemic therapy for 3 months followed by CRS þ HIPEC if a complete
Seven questions were asked regarding some of the conflicting
cytoreduction is achieved and then BST for 3 more months
indications and/or situations that would make a patient with colorectal Yes 33%
cancer and peritoneal dissemination a candidate for cytoreductive No 66%
surgery and HIPEC.

Journal of Surgical Oncology


Colorectal Cancer With Carcinomatosis 267

DISCUSSION OF RESULTS 2. Sadeghi B, Arvieux C, Glehen O, et al.: Peritoneal carcinomatosis


from non-gynecologic malignancies: Results of the EVOCAPE 1
Understanding that the very vast majority of the experts in this multicentric prospective study. Cancer 2000;88:58–63.
voting were surgical oncologists, we can make an analysis of where 3. Sugarbaker PH: New standard of care for appendiceal epithelial
we are in characterizing the role of a combination of surgery and neoplasms and pseudomyxoma peritonei syndrome. Lancet Oncol
2006;7:69–76.
intraperitoneal chemotherapy in the management of patients with
4. Sugarbaker PH, Graves T, DeBruijn EA, et al.: Rationale for early
colorectal cancer and peritoneal dissemination. There was universal post-operative intraperitoneal chemotherapy (EPIC) in patients
agreement regarding the imaging modality of choice in the pre- with advanced gastrointestinal cancer. Cancer Res 1990;50:
operative evaluation of a patient considered a surgical candidate. Fifty 5790–5794.
percent even though that there should be an attempt at trying to 5. Look M, Chang D, Sugarbaker PH: Long-term results of
quantify the amount of carcinomatosis by determining the PCI in the cytoreductive surgery for advanced and recurrent epithelial
CT scan. Even though the published data shows that the extent of ovarian cancers and papillary serous carcinoma of the perito-
carcinomatosis is a prognostic indicator of outcome, and that patients neum. Int J Gynecol Cancer 2004;14:35–41.
with incomplete cytoreductions in general do not benefit from surgery, 6. Carmignani CP, Ortega-Perez G, Sugarbaker PH: The manage-
ment of synchronous peritoneal carcinomatosis and hematoge-
two thirds of the experts would not consider a PCI > 20 or the presence
nous metastasis from colorectal cancer. Eur J Surg Oncol 2004;
of liver metastatses an absolute contraindication to cytoreductive 30:391–398.
surgery and HIPEC. 7. Glehen O, Cotte E, Schreiber V, et al.: Intraperitoneal chemo-
Regarding the role of systemic therapy, it appears that the majority hyperthermia and attempted cytoreductive surgery in patients
of the experts believe that the role of systemic therapy should be in the with peritoneal carcinomatosis of colorectal origin. Br J Surg
‘‘adjuvant’’ setting in patients with peritoneal dissemination and 2004;91:747–754.
negative nodes and even in patients with a large number of positive 8. Elias D, Delperro JR, Sideris L, et al.: Treatment of peritoneal
nodes, only half recommended a 3 month course of neoadjuvant carcinomatosis from colorectal cancer: Impact of complete
therapy. cytoreductive surgery and difficulties in conducting randomized
trials. Ann Surg Oncol 2004;11:518–521.
Seventy-five percent of the experts are in agreement that we need a
9. Elias D, Raynard B, Farkhondeh F, et al.: Peritoneal carcinoma-
large scale Phase III trial in order to make cytoreductive surgery and tosis of colorectal origin: Long-term results of intraperitoneal
HIPEC standard of care. However, after a critical analysis and review chemohyperthermia with oxaliplatin following complete cytor-
of the published data, we can conclude that we have surgical outcome eductive surgery. Gastroenterol Clin Biol 2006;30:1200–1204.
data that it is mostly from Phase II experience of single institutions and 10. Verwaal VJ, van Tinteren H, van Ruth S, et al.: Predicting the
that there is no data when it comes to evaluating the role of the newer survival of patients with peritoneal carcinomatosis of colorectal
combinations of cytotoxic chemotherapy and biological agents in the origin treated by aggressive cytoreduction and hyperthermic
management of patients with colorectal cancer with limited peritoneal intraperitoneal chemotherapy. Br J Surg 2004;91:739–746.
dissemination. Therefore, it is not surprising that the majority of the 11. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of
cytoreduction and hyperthermic intraperitoneal chemotherapy
experts believe that the real question to be asked in such a trial is what
versus systemic chemotherapy and palliative surgery in patients
is the role of HIPEC after a complete cytoreduction as most of us with peritoneal carcinomatosis of colorectal origin. J. Clin Oncol
believe that the primary treatment modality and the only one that can 2003;21:3737–3743.
lead to a good long-term result is achieving a complete cytoreduction. 12. Glehen O, Kwiatkowski F, Sugarbaker PH, et al.: Cytoreductive
Only 36% thought that the right question would be to compare the role surgery combined with perioperative intraperitoneal chemo-
of the newer systemic therapy versus surgery and HIPEC. therapy for the management of peritoneal carcinomatosis from
We have presented the available data and the strategy on how to colorectal cancer: A multi-institutional study. J. Clin Oncol
obtain the data that is necessary to make cytoreductive surgery and 2004;22:3284–3292.
HIPEC the standard of care in patients with colorectal cancer with 13. Yan TD, Black D, Savady R, et al.: A systematic review on the
efficacy of cytoreductive surgery combined with perioperative
peritoneal dissemination. However, the reality is that patients with
intraperitoneal chemotherapy for peritoneal carcinomatosis from
colorectal cancer and peritoneal dissemination, even in the absence colorectal carcinoma. J. Clin Oncol 2006;24:4011–4019.
of hematogenous dissemination, are still labeled as Stage IV, and 14. Esquivel J, Sticca R, Sugarbaker PH, et al.: Cytoreductive surgery
therefore the vast majority of our medical oncology colleagues will and hyperthermic intraperitoneal chemotherapy in the manage-
agree that the best treatment modality is the combination of cytotoxic ment of peritoneal surface malignancies of colonic origin: A
chemotherapy and biological agents because of the dramatic improve- consensus statement. Ann Surg Oncol 2007;14:128–133.
ment in outcome using this regimens in patients with Stage IV 15. de Bree E, Koops W, Kroger R, et al.: Preoperative computed
colorectal cancer as a result of hematogenous and/or lymphatic tomography and selection of patients with colorectal peritoneal
dissemination. Therefore, all of the above can be summarized in saying carcinomatosis for cytoreductive surgery and hyperthermic intra-
peritoneal chemotherapy. Eur J Surg Oncol 2005.
that we as surgeons know that there is a definitive role for cytoreductive
16. Yan TD, Haveric N, Carmignani CP, et al.: Computed tomo-
surgery and HIPEC in the management of patients with colorectal graphic characterization of malignant peritoneal mesothelioma.
cancer and peritoneal dissemination, we have the data to prove it. Our Tumori 2005;91:394–400.
medical oncology colleagues have very impressive data from large 17. Garofalo A, Valle M: Staging videolaparoscopy of peritoneal
Phase III trials in patients with Stage IV colorectal cancer. Only with a carcinomatosis Tumori 2003;89:70–77. Review, Italian.
continued interaction between both groups, will we be able to find the 18. Pomel C, Appleyard TL, Gouy S, et al.: The role of laparoscopy to
best treatment modality for that subset of patients who are Stage IV as a evaluate candidates for complete cytoreduction of peritoneal
consequence of peritoneal dissemination. carcinomatosis and hyperthermic intraperitoneal chemotherapy.
Eur J Surg Oncol 2005;31(5):540–543.
19. Sugarbaker PH: Successful management of microscopic residual
REFERENCES disease in large bowel cancer. Cancer Chemother Pharmacol
1999;43:S15–S25.
1. Hurwitz HI, Fehrenbacher L, Hainsworth JD, et al.: Bevacizumab 20. Verwaal VJ, van Tinteren H, van Ruth S, et al.: Predicting the
in combination with fluorouracil and leucovorin: An active survival of patients with peritoneal carcinomatosis of colorectal
regimen for first-line metastatic colorectal cancer. J. Clin Oncol origin treated by aggressive cytoreduction and hyperthermic
2005;23:3502–3508. intraperitoneal chemotherapy. Br J Surg 2004;91:739–746.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:268–272

Consensus Statement on Peritoneal Mesothelioma

MARCELLO DERACO, MD1* DAVID BARTLETT, MD,2 SHIGEKI KUSAMURA, MD, PhD,1 AND DARIO BARATTI, MD,1
1
Department of Surgery, National Cancer Institute, Milan, Italy
2
Department of Surgery, Division of Surgical Oncology, UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania

Diffuse malignant peritoneal mesothelioma (DMPM) has been traditionally regarded as a rapidly lethal disease. Recently, several independent
prospective trials have reported improved survival with an intensive loco-regional treatment strategy including cytoreductive surgery (CRS) along
with peri-operative intra-peritoneal chemotherapy (PIC). However, most of the surgical data comes from mono-institutional phase I or II studies
and there is a broad range of variability regarding inclusion criteria, cytoreductive surgical procedures, drugs, temperatures and methods of
delivering the heated chemotherapy (open vs. closed abdomen). This manuscript critically analyze and discuss the results of a group of health care
providers trying to achieve a consensus statement in the management of this group of patients. The main conflicting points regarding preoperative
evaluation, patient eligibility, combined treatment methodology, postoperative follow-up and future investigational perspectives were
summarized as a list of multiple-choice questions. A questionnaire was placed on the website of the ‘‘5th International Workshop on Peritoneal
Surface Malignancies’’ and the group members voted via internet. The results were presented for further debate during a dedicated session of the
Workshop. The general treatment guidelines and future investigational perspectives were defined.
J. Surg. Oncol. 2008;98:268–272. ß 2008 Wiley-Liss, Inc.

KEY WORDS: consensus; peritoneal mesothelioma; cytoreductive surgery; hyperthermic intra-peritoneal chemotherapy;
HIPEC

INTRODUCTION The present paper focuses on the results of a group of health care
providers trying to achieve a consensus statement in the management
Malignant mesothelioma is an uncommon tumor arising from the of peritoneal mesothelioma.
serosal layer of pleura, peritoneum, pericardium, and tunica vaginalis
testis [1]. The incidence of the disease has been rising worldwide since
1970 and is not expected to peak for another 10–20 years, due to the
PREOPERATIVE EVALUATION
widespread exposure to asbestos during the last decades [2]. Early diagnosis of DMPM is traditionally challenging [21,22]. Due
In the United States, where the incidence peak has likely already to its rarity and unspecific presentation, the disease is commonly
been reached, about 2,500 new cases of DPM are registered each year diagnosed at advanced stage or confused with ovarian cancer or other
[3]. Diffuse malignant peritoneal mesothelioma (DMPM) accounts for peritoneal disseminations, often resulting in disease mistreatment. The
10–20% of all forms of malignant mesothelioma [3]. goal of a rationale diagnostic pathway is to start an adequate and timely
In the past, DMPM has been regarded as rapidly lethal disease. treatment, thus optimizing the clinical results. Since DMPM has a
Clinical results of conventional treatments, such as palliative great propensity to implant at needle tracts, laparoscopic port sites
surgery, systemic or intra-peritoneal chemotherapy, have been far or surgical incisions, preventing disease dissemination as a con-
from satisfactory in terms of both cure or palliation. Recently, sequence of inappropriate invasive procedures is an additional primary
several independent phase I/II prospective trials have reported objective [15].
improved survival with an intensive loco-regional treatment CT-scan is the imaging test of choice for DMPM. The findings
strategy including cytoreductive surgery (CRS) along with peri- associated to the disease have been recently reviewed. Diffuse disease
operative intra-peritoneal chemotherapy (PIC) in the form of distribution throughout the peritoneal cavity with large tumor volume
hyperthermic intra-peritoneal chemotherapy (HIPEC) þ/ early in the mid-abdomen and in the pelvis may increase the level of clinical
post-operative intra-peritoneal chemotherapy (EPIC). suspicion. Additional findings that could be of help in the differential
There are objective difficulties in planning a phase III clinical trial diagnosis from other gastrointestinal or gynaecologic malignancies are
in this setting, since DMPM is a rare disease and a randomized study the lack of a primary site and the absence of lymph node or distant
would compare a potentially curative treatment with a palliative one. metastases [23]. The role of other imaging studies, such as magnetic
Nevertheless, an extensive literature search of the available retro- resonance or positron emission tomography is presently unclear.
spective historical series has shown that the median survival after
palliative surgery and systemic and/or intra-peritoneal chemotherapy is The authors have no financial interest related to the contents of this article to
about 1 year, ranging from 9 to 15 months [4–11]. Conversely, the disclose.
median survival after aggressive surgery combined with HIPEC has *Correspondence to: Dr. Marcello Deraco, MD, Fondazione IRCCS,
approached 5 years and seems to improve with subsequent reports Istituto Nazionale Tumori Milano, Via Venezian 1, 20133 Milano, Italy.
[4,12–19]. Fax: þ39-02-23902404. E-mail: marcello.deraco@istitutotumori.mi.it
Taken together the aforementioned data suggest that treatment Received 19 March 2008; Accepted 21 March 2008
of PMP by means of CRS and PIC is supported by ‘‘Type 3 evidence,’’ DOI 10.1002/jso.21055
as scientific evidence is available from non-randomized studies, with Published online in Wiley InterScience
external controls allowing comparisons [20]. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Consensus on Peritoneal Mesothelioma 269
Laboratory tests are not useful in the diagnosis of DMPM since only PATIENT ELIGIBILITY TO CYTOREDUCTIVE
unspecific alterations may be occasionally observed [1,2]. In a recent SURGERY AND PIC
study CA125 was preoperatively elevated in the majority of patients
with DMPM, while CEA and CA19.9 were normal in all of them. The combination cytoreductive surgery and PIC is expensive in
These data indicate that CA125 elevation is not a valid criterion to terms of financial resources, operative time and major morbidity [28].
exclude DMPM from diagnosis, although the tumor is less common Therefore, patient selection is important to maximize the clinical
than ovarian cancer [24]. outcome and to exclude patients who will not benefit from a potentially
In patients with suspected DMPM or peritoneal dissemination of life threatening procedure. Candidates have to be medically fit in order
unknown origin, laparoscopy rather than surgical exploration is a to undergo safely to CRS with HIPEC. Furthermore, performance
recommended minimally invasive tool to obtain tumor tissue samples. status has a prognostic relevance, since Eastern Cooperative Oncology
Allowing direct inspection of peritoneal surfaces, laparoscopy may Group (ECOG) score of 0 has been demonstrated to be independently
also facilitate disease staging and the selection of surgical candidates. related to better progression-free survival in patients with DMPM [18].
If DMPM is incidentally discovered during abdominal surgery carried Hepatic or extra-abdominal metastases, as well as peritoneal disease
out for other causes, the surgeon should perform only multiple tumor not amenable of macroscopically complete cytoreduction are exclusion
biopsies, in an effort to preserve as much as possible the integrity of the criteria for CRS and HIPEC. Unnecessary laparotomy can be avoided
peritoneal barrier and to not hinder a future multimodality approach. by accurate radiological evaluation. Preoperative CT-scan can reliably
Pathological confirmation is always required for definitive diag- assess size and distribution of tumor deposits within the abdominal
nosis [22]. In spite of the quality of modern immunocytochemical and cavity. This is useful for the physicians not only to plan the surgical
ultra-structural methods, cytological analyses of ascitic fluid are often cytoreductive procedures but also to identify patients to be excluded
inconclusive [15,21,22]. The pathological differentiation from benign from comprehensive treatment. Tumor mass >5 cm in the epigastric
mesothelial proliferations or from other peritoneal malignancies may region and loss of normal architecture of the small bowel and its
be difficult and requires appropriate immunohistochemical studies. mesentery (segmental obstruction and nodular thickening) are related
DMPM is characterized by positive staining for calretinin, epithelial to failure in adequately removing all the macroscopic tumor. In a
membrane antigen (EMA), Wilms tumor 1 antigen (WT1), cytokeratin composite analysis, none of the patients with both these radiological
5/6, human mesothelial cell 1 (HBME-1), mesothelin and negative features had an adequate cytoreduction. Patients lacking these two
staining for CEA, B72.3, MOC-31, TTF-1 and Ber-EP4. In most cases preoperative CT-scan findings had a 94% probability of adequate
positive calretinin and EMA with negative CEA is highly suggestive of cytoreduction [29].
DPM [25]. Electron microscopy may be of help. Confirmation of the Biological aggressiveness is one of the most relevant prognostic
diagnosis in highly qualified referral centers is recommended [1,2]. determinant for either pleural or peritoneal mesothelioma [1,2].
The definition of the histological sub-variant is prognostically and Patients affected by border-line malignant potential variants (well-
therapeutically relevant. Diffuse malignant mesothelioma is the most differentiated papillary and multicystic) are generally good candidates
common primary peritoneal tumor. Localized forms usually show a for CRS and HIPEC. By contrast, biphasic and sarcomatoid histology
benign clinical behavior. According to the WHO classification, DMPM is associate to poor prognosis [2]. Epithelial variants with intermediate
can be broadly divided as displayed in Table I [26,27]. prognosis represents the great majority of DMPM. In this setting,
During the workshop, 100% of the voters stated that CT-scan of the additional selection factors would be needed to identify patients
abdomen and pelvis has the greatest relevance in the preoperative who will benefit from the procedure and to design an individualized
work-up. CT-scan of the chest was voted by 83.3% of the experts, multimodality treatment plan.
clinical examination by 58.3%, ultrasound by 54.5% and laparoscopy To date, only few centers have clinically investigated the potential
by 50%. No significant consensus was reached about the role of prognostic factors for patients with DMPM treated by cytoreductive
colonscopy, EGDS and PET-scan. Concerning laboratory tests, surgery and PIC, as summarized in Table II along with the statistical
consensus was achieved for complete blood count (75%), coagulation methods used. There is a substantial agreement among the different
studies (67.6%) and comprehensive metabolic panel with liver function groups about the variables related to reduced overall survival: male sex,
tests (75%). Apparently, there was no agreement on the role of incomplete surgical cytoreduction and aggressive tumor histology.
urinalysis pulmonary function tests and tumor serum markers. Tumor invasion of deep tissues, no history of prior surgical debulking,
non-incidental diagnosis and high mitotic count can also be considered
markers of aggressive biological behavior. Finally, Cerruto have re-
cently demonstrated by multivariate analysis that the size of the nucleus
of the DPM cells is independently associated with survival [30].
Correlation to progression-free survival was analyzed in only two
TABLE I. Pathological Classification of Diffuse Malignant Peritoneal
Mesothelioma (DMPM)
studies. No prior debulking, deep tissue invasion, ECOG performance
status >0 and mitotic count >5/50 HPF were recognized by
% of multivariate analysis as independent predictor of adverse prognosis
Tumor type DMPM [14,18].
During the workshop, 81.7% of the experts agreed that histological
Epithelial 75%
subtype has the maximal relevance in patient selection. In a scale from
Malignant DMPM Tubulo-papillary
Nonglandular (solid) 13% 1 to 5, 41.7% and 58.3% of the voters stated that mitotic count and
Sarcomatous 6% nuclear grade or size has a relevance of 5 and 4, respectively.
Biphasic (mixed) 6% Concerning the radiological assessment, 63.6% and 58.3% of the
Undifferentiated experts agreed on the unfavorable prognostic value of tumor mass in
Desmoplastic the epigastric region and small bowel segmental obstruction. Patient
Lympho-histiocitoid performance status was recognized as a useful selection criteria by the
Small cells 50% of voters.
Deciduoid Available literature data suggest that systemic chemotherapy (CT)
Border-line/low malignant DMPM Well-differentiated papillary Rare
may be integrated with the combined loco-regional treatment strategy
Multicystic
for DMPM. Cisplatin has shown the better activity rates as a

Journal of Surgical Oncology


270 Baratti et al.
TABLE II. Prognostic Factors for Peritoneal Mesothelioma After Cytoreductive Surgery and Hyperthermic
Intraperitoneal Chemotherapy (HIPEC)
Principal investigator (Ref.) Feldman [14] Deraco [18] Yan-Sugarbaker [19]

Year 2003 2006 2006


Statistical analysis Multivariate Multivariate Multivariate
N. of patients 49 49 62

Age > 60 yrs X


Previous debulking X
Deep tissue invasion X
Nuclear Size X(3)
Completeness of cytoreduction X(1) X(2)
Mitotic count > 5/50 HPF X
(1)
residual disease >1 cm vs 1 cm; (2)residual disease >2.5 mm vs 2.5 mm; (3)Nuclear size I (10–20 mm) vs II (21–
30 mm) vs III (31–40 mm) vs IV (>40 mm).

single agent and particularly in combination with gemcitabine [31]. A the aforementioned clinical data, the classification involves the
phase III clinical trial testing a new antifolates drug (pemetrexed) in following parameters: (1) presence of extra-abdominal and/or hepatic
combination with cisplatin versus cisplatin alone showed increased metastases; (2) disease potentially suitable for complete surgical
response rate and overall survival. Such combination is currently cytoreduction at preoperative imaging studies; (3) prognostic factors
considered by many oncologists the regimen of choice for pleural (histology, nuclear grade, mitotic count). Prospective studies would
mesothelioma [32]. There are little information on the effectiveness be needed to validate such classification (see Table III). The developing
of this combination for DPMP. The preliminary results of a non- of a new staging system was favored by 75% of the experts and 66.7%
randomized trial started in June 2002 account for an overall objective of them would agree on the opportunity of planning prospective trials
response rate of 26% among 73 patients with DMPM [33]. On these to validate new staging systems.
bases, patients at high risk for postoperative failure may be potential
candidate for adjuvant systemic CT. Furthermore, patients not suitable
for immediate cytoreduction and HIPEC may theoretically benefit of STATE OF THE ART OF THE METHODOLOGY
induction systemic CT to reduce disease extent and undergo a second
comprehensive surgical evaluation. Accurate exploration of the abdominal cavity and lyses of
Different hypotheses of integrated treatment were debated during adherences are needed to assess peritoneal disease extent. The surgical
the workshop for each clinical setting. Adequate surgical cytoreduction cytoreduction is aimed at removing all the macroscopic tumor by
and HIPEC was the favored treatment for low malignant potential means of formal diaphragmatic, parietal and pelvic peritonectomies,
mesothelioma (multicystic and papillary well-differentiated), being complete greater and lesser omentectomy. Small volume tumor
voted by 66.7% of experts. Concerning epithelial DMPM, 33.3% of implants on bowel serosa and mesentery can be electro-fulgurated.
the participants voted for cytoreduction with HIPEC and adjuvant/ Conversely, the opportunity to perform multivisceral resections in case
neo-adjuvant systemic CT, 41.7% for cytoreduction with HIPEC and of massive involvement should be carefully evaluated in light of the
EPIC and only 25% for cytoreduction with HIPEC. CRS with HIPEC deriving functional consequences and the individual prognosis.
and adjuvant/neo-adjuvant systemic CT was the treatment of choice for The definition of adequate cytoreduction as residual tumor nodules
biphasic and sarcomatoid DMPM according to 83.3% of voters. 2.5 mm (cc-1 or R2a, according to the two main scoring systems) was
The best treatment options for patients not amenable to adequate favored by 75% of voters, while 58.3% of the experts agreed on the
cytoreduction were discussed. The following hypothesis were favored opportunity to perform complete parietal peritonectomy even in case of
by the experts: debulking surgery for multicystic and papillary well- disease involvement confined to a limited area, due to the possibility of
differentiated mesothelioma (83.3%); primary systemic CT followed microscopic disease spread.
by re-evaluation for surgical cytoreduction with HIPEC in selected Technique of HIPEC (open vs. closed), type and dose of antiblastic
cases with significant therapeutic response for epithelial peritoneal drugs, duration of treatment and degree of hyperthermia vary from
mesothelioma (66.7%) and biphasic/sarcomatoid mesothelioma one series to other, as summarized in Table IV. Cisplatin has a
(50%). Since no reliable staging system for DMPM is currently favorable pharmacological profile for intra-peritoneal administration
available, we propose a rationale operative prognostic classification for and is likely the most active drug against either pleural or peritoneal
patients candidate to comprehensive loco-regional treatment. Based on mesothelioma [31]. Theoretically, Mitomycin-C has some advantage
for intra-peritoneal administration, since pharmacokinetic studies have
demonstrated a better area under the curve ratio of peritoneal fluid to
plasma [34]. Doxorubicin has shown a great activity in primary tissue
TABLE III. Proposal for Preoperative Prognostic Classification for Diffuse cultures obtained from surgical specimens of DPM (unpublished data).
Peritoneal Mesothelioma A major technical variation is represented by EPIC but the contribution
Peritoneal disease suitable for of such therapeutic tool in addition to CRS and HIPEC is presently
Stage complete cytoreduction Prognostic factors unknown. A comprehensive treatment plan including induction intra-
peritoneal CT, second-look surgery, HEPIC and total abdomen
I Yes No unfavorable prognostic factor radiation have been tested in a phase I/II trial [16].
IIA Yes 1 unfavorable prognostic factors According to the voting results, a combination rather than a
IIB Yes 2/3 unfavorable prognostic factors single drug schedule (75% vs. 25%) was preferred by the experts; the
III No Any combination of cisplatin plus doxorubicin, with the 88.9% of votes,
IV Presence of extra-abdominal
was defined as the regimen of choice. Finally, a temperature of 428C
and/or hepatic metastases
was considered the standard for HIPEC.

Journal of Surgical Oncology


Consensus on Peritoneal Mesothelioma 271
TABLE IV. Technical Features of Cytoreductive Surgery and HIPEC

Residual HIPEC HIPEC


Center Pts (no.) disease status technique HIPEC agents duration Temperature (8C)

Wake Forest Un. 12 60% <2.5cm Closed Mitomycin-C 120 min 42.5
NCI Bethesda, MD 49 88% <1cm Open Cisplatin 90 min 41
EPIC (5-FU, paclitaxel)
Milan, Italy 49 82% <2.5mm Closed Cisplatin þ doxorubicin 90 min 42.5
Lyon, France 15 66% <2.5mm Closed Cisplatin 90 min 42
Mitomycin-C
Washington, DC 62 69%<2.5cm Open Cisplatin þ doxorubicin 90 min 42
EPIC (paclitaxel)
Columbia Univ.-Presbiterian 27 NS Open 1st stage: debulking þ IP cisplatin and 60 min 40
Hospital doxorubicin þ IP gamma-interferon (4 courses)
2nd stage: 2nd look surgery þ HIPEC (cisplatin,
mitomycin-C) þ whole abdominal radiation

EPIC, early postoperative intra-peritoneal chemotherapy; 5-FU, 5-fluoruracil; IP, intra-peritoneal.

FOLLOW-UP epidermal growth factor (EGF) signaling pathway have been


demonstrated to be active in malignant mesothelioma. Imatinib and
Clinical follow-up after CRS and HIPEC can be carried out on out- gefitinib, respectively, block these pathway. However, early studies
patient base. CT-scan was voted as the best imaging study by 75% of with these compounds have not yielded convincing results in the
experts and should be regularly performed along with clinical treatment of mesothelioma [39]. Data from extensive studies in
examination and serum CA125 determination. A recent study suggests mouse models have showed that mesothelioma is apparently sensitive
the utility of circulating CA125 to assess postoperative disease to immunotherapy. These findings have been supported by studies in
progression in patients with elevated baseline levels [24]. In human beings with interferon-alpha, interleukin-2 and macrophage
asymptomatic patients, follow-up visits may be performed every stimulating factors, although standard use in clinical practice is not yet
3–4 months during the first 2 years and every 6 months afterward. If warranted [40]. Recent studies in animals suggest the potential efficacy
asymptomatic recurrences occur, treatment by means of early surgery of the combination of immunotherapy with apoptotic-inducing agents,
or chemotherapy was considered adequate by 83.3% and 75% of the such as gemcitabine [41].
experts, respectively. Two separate gene therapy approaches to mesothelioma have been
tested, involving respectively ‘‘suicide gene’’ and immunomodulatory
gene therapy. Encouraging response in small groups of men has been
FUTURE PERSPECTIVES observed [42]. Preliminary results indicate that the presence of
multiple telomere maintenance mechanisms and the over-expression
Advances in diagnosis of DPM may come form new imaging of survivin, an inhibitors of apoptosis protein, could be involved,
or laboratory tests. Positron emission tomography combined with CT- respectively, in the dysregulation of the apoptotic pathways and in the
scan is a new imaging technique which may be able to preserve immortalization of mesothelioma cells (unpublished data). This could
the high resolution of CT-scan and to provide at the same time open new opportunities for the design of novel therapeutic strategies.
more functional information then CT-scan. This technique has been Other novel agents are involved in clinical trials: bevacizumab,
suggested to reliably detect extra-abdominal disease and to accurately thalidomide and PTK/ZK 787 are new antiangiogenetic drugs targeting
assess response to chemotherapy in patients with mesothelioma [2]. in part the vascular endothelial growth factor (VGEF) pathway;
Serum mesothelin-related proteins are a soluble form of mesothelin histone deacetylase and proteosome inhibitors are other drugs blocking
which is elevated in 84% of patients with pleural mesothelioma and in specific mesothelioma pathway [1,2].
only 2% with other pulmonary diseases [35]. Serum osteopontin levels The experts were asked to express their opinion on potential fields
were shown to be significantly higher in patients with pleural of future investigational studies. The following topics were favored by
mesothelioma than in those with asbestos exposure [36]. No data are the voters: biological and pathological prognostic factors (100%),
presently available about the clinical utility of these antigens in DPM biological target for gene therapy (100%), gene expression profile by
management. RNA microarray (100%), evaluation of asbestos concentration in the
Microarray techniques can assess simultaneously the expression of specimens (70%).
thousands of gene in a single tumor sample. Mesothelioma and normal
pleural samples have been compared in a recent analysis providing
information about gene expression differences between the paired REFERENCES
normal and neoplastic tissues [37]. A microarray study yielded a 99% 1. Robinson BWS, Lake RA: Advanced in malignant mesothelioma.
accuracy in the difficult pathological distinction between pleural N Engl J Med 2005;353:1591–1603.
malignant mesothelioma and lung carcinoma. Such result was obtained 2. Robinson BW, Musk AW, Lake RA: Malignant mesothelioma.
by measuring the expression levels of three pair of genes encoding for Lancet 2005;366:397–408.
calretinin and TTF-1, which are widely used for the immunohisto- 3. Price B: Analysis of current trends in the United States
chemical characterization of mesothelioma [38]. Microarrays are now of mesothelioma incidence. Am J Epidemiol 1997;45:211–
218.
being used to systematically investigate the molecular changes that
4. Sugarbaker PH, Yan TD, Stuart OA, et al.: Comprehensive
may predict the individual response to specific drugs and correlate to management of diffuse malignant peritoneal mesothelioma. Eur
clinical outcome [2]. J Surg Oncol 2006;32:686–691.
Due to the limited activity of traditional drugs, new therapeutic 5. Chailleux E, Dabouis G, Pioche D, et al.: Prognostic factors in
agents are presently matter of both clinical and experimental diffuse malignant pleural mesothelioma, A. study of 167 patients.
investigations. The platelet-derived growth factor (P-DGF) and the Chest 1988;93:159–162.

Journal of Surgical Oncology


272 Baratti et al.
6. Antman K, Shemin R, Ryan L, et al.: Malignant mesothelioma: 24. Baratti D, Kusamura S, Martinetti A, et al.: Circulating CA125 in
Prognostic variables in a registry of 180 patients, the Dana- patients with peritoneal mesothelioma treated by cytoreductive
Farber Cancer Institute and Brigham and Women’s Hospital surgery and intraperitoneal hyperthermic perfusion. Ann Surg
experience over two decades, 1965–1985. J Clin Oncol 1988;6: Oncol 2007;14:500–508.
147–153. 25. Nonaka D, Kusamura S, Baratti D, et al.: Diffuse malignant
7. Sridhar KS, Doria R, Raub WA, Jr., et al.: New strategies are mesothelioma of the peritoneum: A clinicopathological study of
needed in diffuse malignant mesothelioma. Cancer 1992;70: 35 patients treated locoregionally at a single institution. Cancer
2969–2979. 2005;104:2181–2188.
8. Markman M, Kelsen D: Efficacy of cisplatin-based intraperitoneal 26. Battifora H, McCaughey WTE: Atlas of tumor pathology. Third
chemotherapy as treatment of malignant peritoneal mesothe- series. Fascicle 15. Tumors of the serosal membranes. Wash-
lioma. J Cancer Res Clin Oncol 1992;118:547–550. ington, DC: Armed Forces Institute of Pathology; 1995.
9. Yates DH, Corrin B, Stidolph PN, et al.: Malignant mesothelioma 27. Weiss SW, World Health Organization, International Histological
in south east England: Clinicopathological experience of Classification of Tumours: Histological typing of soft tissue
272 cases. Thorax 1997;52:507–512. tumours. 2nd edition. Berlin: Springer-Verlag; 1994.
10. Neumann V, Muller KM, Fischer M: Peritoneal mesothelioma— 28. Kusamura S, Younan R, Baratti D, et al.: Cytoreductive surgery
Incidence and etiology. Pathologe 1999;20:169–176. followed by intraperitoneal hyperthermic perfusion in the treat-
11. Eltabbakh GH, Piver MS, Hempling RE, et al.: Clinical picture, ment of peritoneal surface malignancies: Analysis of morbidity
response to therapy, and survival of women with diffuse and mortality in 209 cases treated with closed abdomen
malignant peritoneal mesothelioma. J Surg Oncol 1999;70:6– technique. Cancer 2006;106:1144–1153.
12. 29. Yan TD, Haveric N, Carmignani CP, et al.: Abdominal computed
12. Park BJ, Alexander HR, Libutti SK, et al.: Treatment of tomography scans in the selection of patients with malignant
primary peritoneal mesothelioma by continuous hyperthermic peritoneal mesothelioma for comprehensive treatment with
peritoneal perfusion (CHPP). Ann Surg Oncol 1999;6:582– cytoreductive surgery and perioperative intraperitoneal chemo-
590. therapy. Cancer 2005;103:839–849.
13. Loggie BW, Fleming RA, McQuellon RP, et al.: Prospective trial 30. Cerruto C, Brun E, Chang D, et al.: Prognostic significance of
for the treatment of malignant peritoneal mesothelioma. Am Surg histo-morphological parameters in diffuse malignant peritoneal
2001;67:999–1003. mesothelioma. Arch Pathol Lab Med 2006;130:1654–1661.
14. Feldman AL, Libutti SK, Pingpank JF, et al.: Analysis of 31. Garcia Carbonero R, Paz-Ares L: Systemic chemotherapy in the
factors associated with outcome in patients with malignant management of malignant peritoneal mesothelioma. Eur J Surg
peritoneal mesothelioma undergoing surgical debulking and Oncol 2006;32:676–681.
intraperitoneal chemotherapy. J Clin Oncol 2003;21:4560– 32. Vogelzang NJ, Rusthoven JJ, Symanowsky J, et al.: Phase III
4567. study of pemetrexed in combination with cisplatin versus cisplatin
15. Sugarbaker PH, Welch LS, Mohamed F, et al.: A review of alone in patients with malignant pleural mesothelioma. J Clin
peritoneal mesothelioma at the Washington Cancer Institute. Surg Oncol 2003;21:2636–2644.
Oncol Clin North Am 2003;12:605–621. 33. Janne PA, Wozniak AJ, Belani CP, et al.: Open-label study of
16. Wagmiller JA, Keohan ML, Chabot JA, et al.: Peritoneal pemetrexed alone or in combination with cisplatin for the
mesothelioma: The Columbia experience. In: Pass HI, Vogelzang treatment of patients with peritoneal mesothelioma: Outcomes
NJ, Carbone M, editors. Malignant mesothelioma. Advances in of an expanded access program. Clin Lung Cancer 2005;7:40–46.
pathogenesis, diagnosis, and translational therapies. NY: Springer 34. Stewart JH IV, Shen P, Levine EA: Intraperitoneal hyperthermic
Verlag; 2005. pp. 723–731. chemotherapy for peritoneal surface malignancy: Current
17. Brigand C, Monneuse O, Mohamed F, et al.: Malignant peritoneal status and future directions. Ann Surg Oncol 2005;12:765–777.
mesothelioma treated by cytoreductive surgery and intraperito- 35. Pass HI, Lott D, Lonardo F, et al.: Asbestos exposure, pleural
neal chemohyperthermia: Results of a prospective study. Ann mesothelioma, and serum osteopontin levels. N Engl J Med 2005;
Surg Oncol 2006;13:405–412. 353:1564–1573.
18. Deraco M, Nonaka D, Baratti D, et al.: Prognostic analysis of 36. Robinson BWS, Creaney J, Lake R, et al.: Mesothelin-family
clinicopathologic factors in 49 patients with diffuse malignant proteins and diagnosis of mesothelioma. Lancet 2003;362:1612–
peritoneal mesothelioma treated with cytoreductive surgery and 1616.
intraperitoneal hyperthermic perfusion. Ann Surg Oncol 2006;13: 37. Singhal S, Wiewrodt R, Malden LD, et al.: Gene expression
229–237. profiling of malignant mesothelioma. Clin Cancer Res 2003;9:
19. Yan TD, Brun EA, Cerruto CA, et al.: Prognostic indicators for 3080–3097.
patients undergoing cytoreductive surgery and perioperative 38. Gordon GJ, Jensen RV, Hsiao LL, et al.: Translation of microarray
intraperitoneal chemotherapy for diffuse malignant peritoneal data into clinically relevant cancer diagnostic tests using gene
mesothelioma. Ann Surg Oncol 2007;14:41–49. expression ratios in lung cancer and mesothelioma. Cancer Res
20. Casali P, Licitra L, Tondini C, et al.: START: A European state-of- 2002;62:4963–4967.
the-art on-line instrument for clinical oncologists. Ann Oncol 39. Nowak AK, Lake RA, Kindler HL, et al.: New approaches for
1999;10:769–773. mesothelioma: Biologics, vaccines, gene therapy, and other novel
21. Hassan R, Alexander R, Antman K, et al.: Current treatment agents. Semin Oncol 2002;29:82–96.
options and biology of peritoneal mesothelioma: Meeting 40. Mukherjee S, Robinson BWS: Immunotherapy of malignant
summary of the first NIH peritoneal mesothelioma conference. mesothelioma. In: Robinson BWS, Chahinian AP, editors.
Ann Oncol 2006;17:1615–1619. Mesothelioma. London: Martin Dunitz; 2002. pp 325–338.
22. Acherman YI, Welch LS, Bromley CM, et al.: Clinical 41. Nowak AK, Robinson BSW, Lake RA: Synergy between
presentation of peritoneal mesothelioma. Tumori 2003;89:269– chemotherapy and immunotherapy in the treatment of established
273. murine solid tumor. Cancer Res 2003;63:4490–4496.
23. Yan TD, Haveric N, Carmignani CP, et al.: Computed tomo- 42. Sterman DH, Kaiser LR, Albelda SM: Gene therapy for malignant
graphic characterization of malignant peritoneal mesothelioma. pleural mesothelioma. Hematol Oncol Clin North Am 1998;12:
Tumori 2005;91:394–400. 553–568.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:273–276

Locoregional Treatment of Peritoneal Carcinomatosis From Gastric Cancer

F. BOZZETTI, MD,1 W. YU, MD, FACS,2* DARIO BARATTI, MD,3


SHIGEKI KUSAMURA, MD, PhD,3 AND MARCELLO DERACO, MD3
1
Department of Surgery, Hospital of Prato, 59100 Prato, Italy
2
Department of Surgery, Kyungpook National University, Daegu, South Korea
3
Department of Surgery, National Cancer Institute of Milan, Italy

The authors reviewed the natural history and the main features of the peritoneal carcinomatosis from gastric cancer briefly and analyzed the
pertinent literature concerning the locoregional modalities for prevention and for treatment. Results of the web based voting by experts were also
summarized. As regards the peritoneal perfusion with cytotoxic drugs with or without hyperthermia for preventing peritoneal carcinomatosis in
high risk patients, there are some randomized clinical trials and one meta-analysis supporting a benefit of the procedure. However, disparity in
methodology (drugs, dosage, duration of the treatment, addition of hyperthermia, etc.) precludes the adoption of a shared protocol to be used in the
clinical practice in high risk patients. Once the peritoneal carcinomatosis is established, the approach reported in literature is the peritonectomy
associated with hyperthermic perfusion. However, data supporting benefits are scanty, and limited to few centers with a specific experience in this
field. With regard to the main questions addressed to the experts’ panel and concerning the indications for treatment and methodology, there was a
general consistency among the experts and agreement with the findings of the literature. The need for a large multicenter trial to confirm the
benefit and risk of intraperitoneal chemotherapy was recognized by both the experts and the authors.
J. Surg. Oncol. 2008;98:273–276. ß 2008 Wiley-Liss, Inc.

KEY WORDS: peritoneal carcinomatosis; gastric neoplasm; intraperitoneal chemotherapy; peritonectomy

INTRODUCTION Staging laparoscopy has become an accepted part of the pretreat-


ment evaluation of patients who are thought to have advanced gastric
Peritoneal carcinomatosis (PC) from gastric cancer is a frequent cancer. Laparoscopy allows for direct inspection of the peritoneal and
event even in the early phase of the disease. It has been estimated that a visceral surfaces for detection of CT-occult small-volume metastases.
percentage of patients ranging from 15% to 50% or more have a Staging laparoscopy also allows for assessment of peritoneal cytology
peritoneal disease at the surgical exploration, especially when there is and intraperitoneal evaluation with adjunctive diagnostic techniques
serosal involvement by the tumor [1,2]. The main risk factors for PC such as laparoscopic ultrasound (LUS). The accuracy of laparoscopy
are the serosal involvement by the tumor [3–5], the lymph nodes for peritoneal metastasis is reported to be 92% [17,18]. Two
involvement [6], the infiltrative growth [7], the big size [8], the gross major unresolved issues remain regarding the timing and extent of
appearance [9] and the scirrhous type reaction [5]. These rates can laparoscopy. Laparoscopy can be performed as a separate staging
dramatically increase if exfoliated neoplastic cells or their markers are procedure before definitive treatment planning or immediately before
actively searched in the peritoneal lavage cytology as well as if one planned laparotomy. LUS and extended laparoscopy are techniques
considers autopsy findings. that may increase the diagnostic yield of laparoscopy. LUS involves
PC, once established, is associated with a poor survival with median examination of the stomach, perigastric region, and peritoneal cavity
values ranging from 1–1.6 months [10,11] to 3.1 and 9 months [1,12] using a laparoscopic ultrasound probe, whereas extended laparoscopy
and no survival at 5 years [13,14]. Peritoneal dissemination is the most involves a more detailed laparoscopic examination of the perigastric
common reason for failure after intensive chemotherapy [15] and region that includes laparoscopic examination of the lesser sac and
systemic chemotherapy is largely ineffective against peritoneal retrogastric space.
carcinomatosis. The majority of patients with peritoneal carcinomatosis are
diagnosed and assessed at laparotomy. Among several systems for
DIAGNOSIS AND STAGING assessing the distribution of peritoneal surface disease, peritoneal
cancer index (PCI) is commonly used. This is a clinical summary of
The preoperative diagnosis and staging of peritoneal disease is both lesion size and distribution of peritoneal surface malignancy [19].
limited by the insensitivity of traditional imaging modalities such as The lesion size (LS) is used to assess the size of nodules. In order to
computed tomography (CT), ultrasonography (US), and magnetic assess the distribution of peritoneal surface disease, the abdomino-
resonance imaging (MRI). Although there is increased accuracy in pelvic regions are utilized. For each of these 13 regions, an LS score is
the assessment of extragastric disease with modern multiphase,
multidetector spiral CT imaging, sensitivity of CT for detection of The authors have no financial interest related to the contents of this article to
extragastric disease declines with the size of metastases. Current CT disclose.
techniques cannot consistently identify low-volume macroscopic *Correspondence to: W. Yu, MD, FACS, Department of Surgery,
metastases that are 5 mm or less in size. However, CT scans are Kyungpook National University, 50 Samduk-dong, Daegu 700-721, South
sensitive for the detection of omental metastases or indirect evidence of Korea. Fax: 82-53-421-0510. E-mail: wyu@mail.knu.ac.kr
tumor such as the presence of ascites, mesenteric thickening, or Received 19 March 2008; Accepted 21 March 2008
matting of loops of bowel. DOI 10.1002/jso.21052
Lee et al. [16] reported that EUS was more sensitive than combined Published online in Wiley InterScience
US and CT scan examinations in diagnosing ascites. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


274 Bozzetti et al.
determined. The summation of the LS in each of the 13 regions is the of hospital stay, but not mortality, were significantly higher in the
PCI for that patient. It is basically a recording of what the surgeon treated group.
visualizes as he explores the abdomen. PCI can be easily applied to the
nodular type of peritoneal seeding from gastric cancer. However, it is
difficult to assess the diameter of peritoneal surface disease in diffuse PERITONECTOMY (P) AND
infiltrating type of dissemination. HYPERTHERMIC INTRAPERITONEAL
CHEMOTHERAPY (HIPEC)
CLINICAL PATHWAY
The experience with P-HIPEC is limited in number of studies
Patients who are found to have occult metastatic disease at and number of patients undergoing the procedure. To our knowledge
laparoscopy or laparotomy are considered incurable and only palliative there are three institutional studies on P-HIPEC in patients with
procedures are recommended for these patients [20]. gastric cancer, one retrospective (42 patients) [23], one prospective
In patients with peritoneal carcinomatosis from colon cancer, the (49 patients) [28] and one comparative non-randomized (34 patients)
prognosis of patients with low PCI scores is significantly better than [29]. The median survival ranged from 8 months [28] to 10–11 months
that of patients with high PCI scores [21]. The value of the PCI as a [23,28] and the 5-year survival ranged from 6% [23,29] to 16%
selection tool in this patient population is obvious, however, PCI as a according to Glehen et al. [28] Surgical mortality was acceptable,
selection tool in gastric cancer has not been evaluated. namely 2–7.1% [23,28,29]. Interestingly, a multivariate analysis
The peritonectomy is indicated for the patients with peritoneal showed that completeness of cytoreduction (and hence initial stages
dissemination which is considered to be able to macroscopically of peritoneal dissemination versus advanced stages) and absence of
completely resected. Patients with a limited number of metastatic ascites were favorable prognostic factors for survival. More precisely,
nodules distributed in the peritoneal cavity, and those without evidence in patients with CC-0 or CC-1 median survival was 21.3 months and
of the involvement of the liver or distant lymph nodes are eligible for 5-year survival was 29.4% [28], and Yonemura et al. [23] reported
peritonectomy [22]. Peritonectomy is contraindicated for patients with quite similar figures: a complete cytoreduction was associated with a
many big nodules distributed on the mesentery of small bowel. In other median survival of 19.2 months and a 5-year survival of 27%. It is
words, PCI should be converted to 0 by cytoreduction, that means noteworthy, however, that a complete cytoreduction was possible in
completeness of cytoreduction score 0 (no visible peritoneal only 44–51% of patients operated on by Yonemura et al. [23] and
carcinomatosis remains after cytoreduction). Glehen et al. [28] The most important prognostic factors for survival
Invisible dissemination already spreads everywhere in the perito- are absence of ascites, completeness of cytoreduction, a low complete-
neal cavity, even if the complete resection of peritoneal dissemination ness of cytoreduction score and a low peritoneal dissemination class.
is achieved by aggressive surgical cytoreduction. A major role for
intraperitoneal chemotherapy is the prevention of subsequent perito- EXPERTS’ CONSENSUS
neal carcinomatosis. Intraperitoneal chemotherapy is an important
treatment option for patients with T3 or T4 lesions as well as for A number of questions were addressed to 19 experts from 7 different
patients who are undergoing peritonectomy. Locoregional treatment countries regarding IPEC (with or without hyperthermia), P-HIPEC
consists in peritoneal perfusion (with or without hyperthermia) as a and some methodological issues.
single procedure or following cytoreductive surgery. The two
procedures are not comparable since the first has usually been used Questions Regarded the Eligibility for IPEC
in the earliest stages and sometimes as an adjuvant treatment,
the second one has been exclusively used in advanced stages. All the experts agreed that adjuvant IPEC should be recommended
The treatment strategy composed of peritonectomy, intraoperative in patients after curative gastrectomy if there was a positive cytology
and early postoperative intraperitoneal chemotherapy is expected to be and the 83% of them advised the procedure to be done for T3–T4
a powerful therapy for patients with peritoneal dissemination from tumors. Moreover 4/5 of them accepted that T classification could
gastric cancer. be defined on the basis of the simple intraoperative gross appearance
without the help of frozen sections or sophisticated instrumentation.
INTRAPERITONEAL CHEMOTHERAPY (IPEC) Should PC be discovered during laparotomy and gastric tumor
be adequately resectable patients might be eligible for IPEC if
A meta-analysis of 11 randomized clinical trials (1980–2003) PCI is less than 10 (92% consensus) and size of the single lesions
involving 1,161 patients receiving IPEC (usually without hyperthermia is 0.2 cm or less (92% consensus). The vast majority of the experts
and mainly with mitomycin C, but also cisplatinum or fluorouracil) (92%) supported the concept that IPEC should be associated with
after curative resection for locally advanced gastric cancer showed a hyperthermia, should be associated with postoperative adjuvant
benefit in the overall survival (pooled odds ratio 0.51, 95% CI 0.40– systemic chemotherapy (77%), and should be delivered as a single
0.60) [24]. We reanalyzed the Xu’s study [24] to find out which drug cycle (77%). Only 54% advised to prolong IPEC also in the
combination proved to be the most active, but we found none of the postoperative period.
various drug combinations (mitomycin C-carbon particles, mitomycin Two questions explored the opinion of the experts about multicenter
C and cisplatinum, fluorouracil, mitomycin C and fluorouracil) proved randomized clinical trials on IPEC: 77% of them would be available to
to be consistently active. Similarly there was no consistent effect with take part in the trials of IPEC versus no-IPEC in CC-0 patients or in
the use of IPEC or hyperthermic IPEC (HIPEC). patients at high risk for PC. A lesser percentage (62%) was available to
More recently two phase II studies were published [25,26], but the take part in a trial of IPEC versus other therapies in patients with PC
only new report of a randomized clinical trial is from Yu [27] They discovered during laparotomy.
reported on 248 patients randomized to early postoperative intraper-
itoneal chemotherapy with mitomycin C and 5-fluorouracil for 5 days Questions Regarding P þ HIPEC
versus control. There was a benefit in the 5- and 10-year survival in the
treated arm which was mainly confined to stage III. A multivariate The experts recommended the procedure in primary tumors and in
analysis confirmed that IPEC was a statistically significant prognostic recurrent tumors in the following conditions: only in CC-0/CC-1
factor for survival and rate of postoperative complications and length resections (100% in primary tumors, 75% in recurrent tumors); only in

Journal of Surgical Oncology


Peritoneal Carcinomatosis From Gastric Cancer 275
absence of distant metastasis (83% in primary tumors, 73% in recurrent 4. Moriguchi S, Maehara Y, Korenaga D, et al.: Risk factors
tumors); only in absence of ascites (33% in primary tumors, 19% in which predict pattern of recurrence after curative surgery for
recurrent tumors); in stages PCI < 10 or 15, 38% or 71%. patients with advanced gastric cancer. Surg Oncol 1992;1:341–
The majority of the experts (92%) favored the association of 346.
P þ HIPEC with systemic therapy. Again, 92% considered necessary a 5. Wu CW, Lo Wu SS, Shen KH, et al.: Incidence and factors
associated with recurrence patterns after intended curative surgery
large phase III trial to make P þ HIPEC the standard therapy in PC for gastric cancer. World J Surg 2003;27:153–158.
from gastric cancer, and 77% would accept to take part in the trial 6. Roviello F, Marrelli D, de Manzoni G, et al.: Prospective study of
which compares P þ HIPEC with other therapies. peritoneal recurrence after curative surgery for gastric cancer. Br
J Surg 2003;90:1113–1119.
7. Maehara Y, Hasuda S, Koga T, et al.: Postoperative outcome and
Miscellaneous Questions sites of recurrence in patients following curative resection of
gastric cancer. Br J Surg 2000;87:353–357.
Three quarters—two thirds of the experts advised not to attempt any 8. Marrelli D, Roviello F, de Manzoni G, et al.: Different patterns of
peritonectomy if a radical resection is not attainable and 89% suggest recurrence in gastric cancer depending on Lauren’s histological
limiting peritonectomy to the affected area only. Furthermore, 77% of type: Longitudinal study. World J Surg 2002;26:1160–1165.
them were in favor of using multiple drugs for HIPEC, and in the order, 9. Lee CC, Lo SS, Wu CW, et al.: Peritoneal recurrence of gastric
mitomycin C (85%), cisplatinum (77%), fluorouracil (46%) and adenocarcinoma after curative resection. Hepatogastroenterology
doxorubicin (31%). 2003;50:1720–1722.
10. Chu DZ, Lang NP, Thompson C, et al.: Peritoneal carcinomatosis
in nongynecologic malignancy. A. prospective study of prognos-
DISCUSSION tic factors. Cancer 1989;63:364–367.
11. Yamada S, Takeda T, Matsumoto K: Prognostic analysis of
It appears that there was, in general, a good agreement between data malignant pleural and peritoneal effusions. Cancer 1983;51:136–
of the literature and opinion of the experts and a good internal 140.
consistency within the experts’ consensus. 12. Sadeghi B, Arvieux C, Glehen O, et al.: Peritoneal carcinomatosis
Certainly IPEC or HIPEC as adjuvant treatment is a procedure more from non-gynecologic malignancies: Results of the EVOCAPE 1
multicentric prospective study. Cancer 2000;88:358–363.
validated than P-HIPEC, and there are randomized clinical trials and 13. Yonemura Y, Fujimura T, Nishimura G, et al.: Effects of
also a meta-analysis supporting its efficacy. However, these trials are intraoperative chemohyperthermia in patients with gastric cancer
often underpowered, their quality is suboptimal and studies from with peritoneal dissemination. Surgery 1996;119:437–444.
eastern countries were more successful than those from Europe. All 14. Elias D, Blot F, El Otmany A, et al.: Curative treatment of
these factors contribute to a limited spread of the procedure and to a peritoneal carcinomatosis arising from colorectal cancer by
general agreement of the experts who recognize the need to plan a large complete resection and intraperitoneal chemotherapy. Cancer
multicenter randomized clinical trial which definitely might prove the 2001;92:71–76.
efficiency of an adjuvant intraperitoneal chemotherapy versus the 15. Ajani JA, Ota DM, Jackson DE: Current strategies in the
standard systemic chemotherapy. management of locoregional and metastatic gastric carcinoma.
Cancer 1991;67:260–265.
Although the majority of experts claim that the addition of 16. Lee YT, Ng EK, Hung LC, et al.: Accuracy of endoscopic
hyperthermia improves the efficacy of the IPEC, centers where ultrasonography in diagnosing ascites and predicting peritoneal
hyperthermia is readily available and feasible when a high-risk metastases in gastric cancer patients. Gut 2005;54:1541–1545.
patient is intraoperatively identified are relatively few. Probably the 17. Ribeiro U, Jr., Gama-Rodrigues JJ, Safatle-Ribeiro AV, et al.:
larger number of patients who could undergo the simplest IPEC Prognostic significance of intraperitoneal free cancer cells
would compensate for a lesser impact of this procedure and would obtained by laparoscopic peritoneal lavage in patients with
account for a higher number of patients who finally benefit from the gastric cancer. J Gastrointest Surg 1998;2:244–249.
treatment. 18. Fujimura T, Kinami S, Ninomiya I, et al.: Diagnostic laparoscopy,
Since there is a continuous progress in the field of chemotherapy a serum CA125, and peritoneal metastasis in gastric cancer.
Endoscopy 2002;34:569–574.
reasonable approach for a multicenter randomized clinical trial would 19. Jacquet P, Sugarbaker PH: Clinical research methodologies in
be a two-arm study with following scheme: for patients at high risk for diagnosis and staging of patients with peritoneal carcinomatosis.
PC (T3–T4), undergone a R0 gastrectomy, randomization to adjuvant Cancer Treat Res 1996;82:359–374.
IPEC versus no-IPEC, regardless to any systemic therapy which could 20. Yoshikawa T, Kanari M, Tsuburaya A, et al.: Should gastric
be administered to both the groups. Previous experience has shown that cancer with peritoneal metastasis be treated surgically? Hepato-
a 3-year follow up could be enough to demonstrate a potential benefit gastroenterology 2003;50:1712–1715.
in survival. 21. Sugarbaker PH: Successful management of microscopic residual
In contrast, P-HIPEC remains an approach less mature and to be disease in large bowel cancer. Cancer Chemother Pharmacol
reserved for centers which are familiar with the procedure to maintain 1999;43:S15–S25.
22. Kim T, Yu W: Indications for surgical resection of stage IV gastric
an acceptable operative risk and select the patient population who cancer. J Korean Surg Soc 2004;67:183–187.
presumably can have the best benefit-to-risk option. 23. Yonemura Y, Kawamura T, Bandou E, et al.: Treatment of
peritoneal dissemination from gastric cancer by peritonectomy
REFERENCES and chemohyperthermic peritoneal perfusion. Br J Surg 2005;92:
370–375.
1. Okajima K, Yamada S: Surgical treatment of far-advanced gastric 24. Xu DZ, Zhan YQ, Sun XW, et al.: Meta-analysis of intra-
cancer. Jpn J Cancer Clin 1986;32:1203–1209. peritoneal chemotherapy for gastric cancer. World J Gastroenterol
2. Sugarbaker PH, Yonemura Y: Clinical pathway for the manage- 2004;10:2727–2730.
ment of resectable gastric cancer with peritoneal seeding: Best 25. Newman E, Potmesil M, Ryan T, et al.: Neoadjuvant chemo-
palliation with a ray of hope for cure. Oncology 2000;58:96–107. therapy, surgery, and adjuvant intraperitoneal chemotherapy in
3. Boku T, Nakane Y, Minoura T, et al.: Prognostic significance of patients with locally advanced gastric or gastroesophageal
serosal invasion and free intraperitoneal cancer cells in gastric junction carcinoma: A phase II study. Semin Oncol 2005;32:
cancer. Br J Surg 1990;77:436–439. S97–S100.

Journal of Surgical Oncology


276 Bozzetti et al.
26. Kunisaki C, Shimada H, Akiyama H, et al.: Therapeutic outcomes 28. Glehen O, Schreiber V, Cotte E, et al.: Cytoreductive surgery
of continuous hyperthermic peritoneal perfusion against advanced and intraperitoneal chemohyperthermia for peritoneal car-
gastric cancer with peritoneal carcinomatosis. Hepatogastroenter- cinomatosis arising from gastric cancer. Arch Surg 2004;139:
ology 2006;53:473–478. 20–26.
27. Yu W: A review of adjuvant therapy for resected primary gastric 29. Hall JJ, Loggie BW, Shen P, et al.: Cytoreductive surgery with
cancer with an update on Taegu’s phase III trial with intra- intraperitoneal hyperthermic chemotherapy for advanced gastric
peritoneal chemotherapy. EJSO 2006;32:655–660. cancer. J Gastrointest Surg 2004;8:454–463.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:277–282

Consensus Statement on the Loco-Regional Treatment of Appendiceal Mucinous


Neoplasms with Peritoneal Dissemination (Pseudomyxoma Peritonei)

BRENDAN MORAN, MD,1 DARIO BARATTI, MD,2 TRISTAN D. YAN, BSc(Med), MBBS, PhD,
3

SHIGEKI KUSAMURA, MD, PhD,2 AND MARCELLO DERACO, MD2*


1
Colorectal Research Unit, North Hampshire Hospital, Basingstoke, Hampshire, UK
2
Department of Surgery, National Cancer Institute of Milan, Italy
3
University of New South Wales, Department of Surgery, St. George Hospital, Sydney, Australia

Pseudomyxoma peritonei (PMP) is a rare condition mostly originating from low malignant potential mucinous tumours of the appendix. Although
this disease process is minimally invasive and rarely causes haematogenous or lymphatic metastases, expectation of long-term survival are limited
with no prospect of cure. Recently, the combined approach of cytoreductive surgery (CRS) and perioperative loco-regional chemotherapy (PLC)
has been proposed as the standard of treatment for the disease. The present paper summarizes the available literature data and the main features of
the comprehensive loco-regional treatment of PMP. The controversial issues concerning the indications and technical methodology in PMP
management were discussed through a web-based voting system by internationally known experts. Results were presented for further evaluation
during a dedicated session of ‘‘The Fifth International Workshop on Peritoneal Surface Malignancy (Milan, Italy, December 4–6, 2006)’’. The
experts agreed that multiple prospective trials support a benefit of the procedure in terms of improved survival, as compared with historical
controls. Concerning the main controversial methodological questions, there was an high grade of consistency among the experts and agreement
with the findings of the literature.
J. Surg. Oncol. 2008;98:277–282. ß 2008 Wiley-Liss, Inc.

KEY WORDS: pseudomyxoma peritonei; appendiceal tumours; cytoreductive surgery; hyperthermic intraperitoneal
chemotherapy; HIPEC

Pseudomyxoma peritonei (PMP) is a rare condition characterized controls [8–18]. Sugarbaker published a large series of 385 patients in
by copious mucinous ascites and peritoneal mucinous implants [1–2]. 1999 [8]. Of these, 205 received hyperthermic intra-peritoneal
The earliest description of the condition was by Rokitansky in 1842 in chemotherapy (HIPEC). He showed survival advantages in those
a patient with a benign mucocele of the appendix [3]. who had complete versus incomplete cytoreduction (80% vs. 20%) and
Traditionally patients with PMP have been treated with repeated in those with low-grade versus high-grade tumours (80% vs. 28%) but
interval debulking procedures for relief of symptoms, but with limited did not comment on whether the introduction of HIPEC made any
expectation of long-term survival and no prospect of cure [1–2]. difference to survival. Most recent updates by Sugarbaker demon-
However, accurate historical controls of uniformly treated patients are strated a median survival of 156 months and 5- and 10-year survival of
scarce, partly due to the rarity of the disease. In 1994, Gough reported a 72% and 55% in 501 patients [11]. Taken together these data suggest
10-year survival of 32% in 56 PMP patients who underwent serial that treatment of PMP by means of CRS and PLC is supported by a
debulking procedures and selectively treated with intra-peritoneal ‘‘type 3 evidence’’, coming from non-randomised studies with external
radiotherapy or chemotherapy between 1957 and 1983 [4]. In 2005, controls allowing comparisons [19].
Miner reported a 10-year survival of 21% in 97 PMP patients treated The present paper addresses the available scientific evidence and
with serial debulking, systemic chemotherapy and/or delayed inter- the results of a group of health-care providers trying to achieve a
mittent intra-peritoneal 5-fluorouracil over a 22-year period [5]. consensus statement in the management of PMP by means of CRS and
Misdraji reported on 107 patients with a median survival of about PLC regimens, including intra-operative HIPEC and/or early post-
7.5 years, and a 20-year survival of 25% after serial debulking and operative intra-peritoneal chemotherapy (EPIC) within 7 days from
intra-peritoneal chemotherapy. The number of patients in this group surgery.
who received aggressive locoregional treatment is unknown [6].
Although a subset of patients may remain asymptomatic for
many years, the disease almost always recurs and patients often re-
present with gastrointestinal obstructive symptoms. Over time each
repeated debulking procedure becomes more ineffective and sometimes Disclosures: The authors have no financial interest related to the contents of
more dangerous due to the risk of bowel injury [1]. In addition, in some this article to disclose.
patients the disease may not remain indolent throughout its clinical *Correspondence to: Dr. Marcello Deraco, MD, Istituto Nazionale Tumori
course. Yan and colleagues showed that some patients underwent Milano, Via Venezian, 1 20133 Milano, Italy. Fax: þ39-02-23902404.
transitions from a less aggressive to a more aggressive histopathologic E-mail: marcello.deraco@istitutotumori.mi.it
type over time and with repeated surgical interventions [7]. Received 19 March 2008; Accepted 21 March 2008
The structured approach of cytoreductive surgery (CRS) and DOI 10.1002/jso.21054
perioperative loco-regional chemotherapy (PLC) regimens has shown Published online in Wiley InterScience
in multiple studies improved survival, as compared with historical (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


278 Moran et al.

TERMINOLOGY AND abdominal cavity is dictated by gravity, pressure changes associated


PATHOLOGICAL CLASSIFICATION with respiration and physical boundaries of the peritoneal reflections.
The mucin-producing cells in PMP are poorly adherent and freely
There has been considerable confusion in the literature about the circulate with peritoneal fluid. This process has been termed as
site of origin and pathological classification of PMP [1,6]. High-grade redistribution phenomenon [33]. Accordingly, the predictable pattern
colonic mucinous neoplasms, adenocarcinomas of the appendix of accumulation of mucinous ascites instead of the individual deposits
and mucinous adenocarcinomas originating from any other intra- can suggest the diagnosis of early-stage PMP. When the peritoneal
abdominal organ can simulate the clinical, radiological and patho- cavity is completely, or almost completely filled with PMP, CT-scan
logical features of PMP. Additionally, there appears to be a histological findings become less specific. In most cases, the striking feature is the
spectrum of disease from low to high-grade [1]. Recent pathological, relative sparing of the small bowel and its mesentery and their
molecular genetic and immunohistochemical evidence supports the compartimentalization in the central abdomen by a large omental cake
theory that the majority of classical PMP originate from ruptured low- and massive mucinous ascites.
grade appendiceal tumours [20–22]. Undoubtedly a small proportion The role of magnetic resonance imaging (MRI) in this clinical
of cases arise from other organs and it is possible that an ovarian setting is presently under investigation. According to a preliminary
primary mucinous tumour may be the commonest in this diverse group report, the use of MRI seems promising in disease staging and patient
arising from the stomach, colon, pancreas, gallbladder, urachus and selection for cytoreductive surgery [34]. PET is of very limited value
other intra-abdominal organs [23–26]. for low-grade mucinous lesions, such as pseudomyxoma [35]. CT-scan
These difficulties in pathological classification of the clinical entity or US-guided biopsy may be useful, although the relatively acellular
of PMP have led to ongoing confusion as to the outcomes following material is often difficult to diagnose with certainty.
intervention. Thus, many series include all cases of PMP, of whatever Most patients are referred from other surgical or gynaecological
origin, and include patients with mucinous adenocarcinoma of the units and have usually had variable degrees of surgery prior to formal
appendix whereas others have reported only on classical pseudo- attempts at complete cytoreduction. Extensive previous attempts to
myxoma from appendiceal cystadenomas. Ronnett, in a retrospective reduce tumour load were shown by one large study to have a negative
review of a series of patients undergoing complete cytoreduction impact on survival [8]. It advisable, therefore, that the referring
reported a pathological system commonly quoted in the literature teams perform the minimal amount of surgery required to establish a
[20]. They classified low-grade tumours as disseminated peritoneal histological diagnosis prior to referral to a specialist centre.
adenomucinosis (DPAM) and high-grade tumours as peritoneal In total 78% of the panel agreed that PMP should always be
mucinous carcinomatosis (PMCA), with an intermediate group (IG). considered a cancer and therefore malignant. However, even when
Survival was significantly higher in DPAM group as compared with the the pathology was stated as ‘‘Malignant PMP syndrome-mucinous
IG and PMCA, with actuarial 5-year survivals of 84%, 37.6% and Appendix Carcinoma and Mucinous Carcinoma from any origin’’
6.7%, respectively. They were unable to show a statistically significant surprisingly 11% of the panel still considered this not to be a cancer.
difference between the IG and PMCA groups and in subsequent Regarding pathological classification, no consensus was reached
articles have grouped these together [27]. with 44% favouring Ronnetts three-category classification. A further
In a recent study by Misdraji, a different histopathologic classifica- 44% voted for a two-category system of Low-Grade Adenocarcinoma
tion of appendiceal neoplasm was proposed [6]. They classified the and High-Grade Adenocarcinoma. The novel recent terminology of
appendiceal mucinous tumours into low-grade appendiceal mucinous Mucinous Carcinomatosis Pertitonei (MCP) low-grade and MCP high-
neoplasms (LAMN) and high-grade mucinous adenocarcinomas grade, which is in reality new terminology similar to the two grade
(MACA). The former diagnosis corresponds to mucinous cysto- system, was favoured by 12%.
adenoma and the latter to mucinous cysto-adenocarcinoma in the These discrepancies in terminology and pathological classification
conventional classification system [28]. However, in this study, follow- are important and really require universal agreement due to the
up data were available only in 66 of 107 patients and patients underwent difficulties in comparing series, credibility in justifying major
a variety of treatments by different surgeons over a 50-year period. More surgical intervention, utilization of chemotherapy and difficulty in
recently, based on the clinically malignant behaviour of all PMP establishing treatment programmes for what even experts consider a
varieties, Bradley has proposed that mucinous carcinoma peritonei, ‘‘benign disease’’. Based on the terminology of uncontrolled growth
either low-grade or high-grade, is best applied to all cases of PMP [29]. of abnormal cells it is the authors opinion that PMP is at best a
‘‘borderline malignancy’’ with optimal treatment being that of a
PREOPERATIVE EVALUATION cancer entailing complete macroscopic tumour removal combined with
intraperitoneal chemotherapy. An interesting comment to one of us at
The clinical presentation of PMP has been poorly defined due to the consensus meeting was ‘‘Based on this statement on PMP do you
few reports with large patient populations. The recent publication equally consider endometriosis a malignancy?’’.
by Esquivel and Sugarbaker is the most enlightening [30]. In a series of There was general agreement concerning pre-operative work-up
410 patients with appendiceal tumours, 27% presented with suspected with 100% considering abdominal and pelvic CT-scan very important,
appendicitis, 23% with increasing abdominal distension and 14% 66% considering clinical examination very important with very
with a new onset hernia. In women, a diagnosis of PMP was most little role for PET-scan, CT-PET, MRI or US. The panel felt that
commonly made while being evaluated for ovarian mass (39%). colonoscopy and laparoscopy and biopsy might help on occasion but
Nevertheless, the majority of patients are diagnosed at a laparotomy were not essential. Important laboratory work-up was considered to be
performed for either suspected appendicitis, peritonitis or gynaeco- basic metabolic panel urea, creatinine and electrolytes, CEA, complete
logical cancer. blood count and coagulation studies by 75% of the panel with
More recently, the diagnosis of PMP is suspected in an increasing little importance attached to pulmonary function, urinalysis or CA125
number of patients at imaging studies, particularly based on radio- measurement.
logical features at CT-scan [31,32]. Appropriate radiologic techniques
and a combination of oral, rectal and venous contrast are needed. On ELIGIBILITY FOR SURGICAL TREATMENT
CT-scan PMP appears of low attenuation but areas of high attenuation,
due to solid elements within the mucinous material or compressed Careful selection for this expensive, time consuming and high-risk
mesentery, can be seen [32]. The circulation of peritoneal fluid in the surgery is essential. However, the potential surgical morbidity, along

Journal of Surgical Oncology


Consensus on Pseudomyxoma Peritonei 279
with the spectrum of disease aggressiveness, makes preoperative paper, recurrence-free interval was statistically reduced for patients
selection of patients for CRS and PLC critical. Current selection with increased CEA, but also for patients with at least one positive
criteria predominantly revolves around clinical assessment and CT- marker (among CEA, CA125 and CA19.9) [40].
scan imaging [32]. CT imaging and CT-scan with tumour marker measurements were
Patients affected by peritoneal carcinomatosis with performance chosen by 71% and 83% as key eligibility criteria with only 50%
score ¼ 2/3 according to the Eastern Cooperative Oncology Group support for laparoscopy or other investigations. With regards to disease
(ECOG) have shown significantly poorer overall survival, compared to extension 100% felt that all asymptomatic patients with PMP
those with ECOG score ¼ 1 [35]. with resectable peritoneal dissemination should have CRS and PLC,
In an effort to correlate the role of previous surgery in the as should patients with minimal small bowel involvement and node
establishment of peritoneal implants, Sugarbaker introduced the negative primary (100%). There was 100% agreement that peritoneal
Prior Surgical Score (PSS). In patients with a PSS ¼ 0, diagnosis of carcinomatosis index (PCI) >20 was not an absolute exclusion
peritoneal carcinomatosis was obtained through biopsy or laparoscopy criterion in PMP, with a low grade tumour (87%), young age (63%),
only. PSS ¼ 1 indicates only a previous exploratory laparotomy. absence of mesenteric invasion (88%) or liver metastases (75%) were
PSS ¼ 2 indicates exploratory laparotomy with some resections. important factors to consider in conjunction with PCI.
Usually this was a greater omentectomy and/or right colectomy.
PSS ¼ 3 indicates patients had an attempt at a complete cytoreduction. STATE OF THE ART OF THE METHODOLOGY
Patients with PSS ¼ 0–2 had a significantly improved survival
compared with those with a PSS ¼ 3 [8]. The aim of surgery in PMP is complete cytoreduction as described
The current best method to assess operability is contrast-enhanced by Sugarbaker [2]. This involves up to six different peritonectomy
CT-scan. Radiological features predicting the likelihood to adequate procedures in combination with visceral resections as required, to
remove all the peritoneal tumour deposits have been described by remove all visible tumour, or if this was not possible, to leave tumour
Jacquet and validated by Sulkin [31,32]. In the study by Jacquet two deposits less than 2.5 mm (2.5 mm being the maximum direct
radiologic findings predicted complete versus suboptimal cytoreduc- penetration of locally applied chemotherapy). In brief, peritonectomy
tion: the segmental obstruction of the small bowel and tumour masses includes a greater omentectomy and splenectomy, left upper quadrant
>5 cm on small bowel and its mesentery exclusive of distal ileum. A peritonectomy, right upper quadrant peritonectomy, lesser omen-
statistical approach using a tree-structured diagram showed that tectomy and cholecystectomy, appendicectomy or right/total colec-
patients with both these features on preoperative CT-scan, had an tomy, partial or total gastrectomy, and pelvic peritonectomy with
88% probability of incomplete resection. Patients without these two anterior resection of the recto-sigmoid colon. Females require
findings had a 92% probability of complete resection [31]. hysterectomy and bilateral salpingo-oophorectomy.
Allowing direct inspection of peritoneal surfaces, laparoscopy may No study has reported on the effect of hyperthermic compared to
also facilitate disease staging and the selection of surgical candidates. normothermic intra-peritoneal chemotherapy in patients in whom a
The role of laparoscopy for PMP has never been specifically addressed. complete cytoreduction was performed. The chemotherapy agents
The experiences with patients affected by carcinomatosis from utilized, the dosage, the temperature or duration of intra-peritoneal
colorectal cancer has led to the conclusion that laparoscopy is accurate chemotherapy have not been subject to randomised trials but have been
to score peritoneal involvement and to assess the complete resectability chosen on knowledge of the agents’ intra-peritoneal pharmacokinetics.
of carcinomatosis in patients for which there is inadequate or The commonly used intra-operative agents are mitomycin, cisplatin,
contradictory information [36]. 5-fluorouracil or a combination of these and are usually administered
Finally, in recent years the prognostic value of markers has been for 30-120 min [41]. For EPIC, 5-fluorouracil, cyclophosphamide and
extensively addressed. In the paper by Baratti and colleagues: normal mitomycin C are most frequently used for up to 6 days (see Table I).
preoperative CA125 correlated to adequate CRS, but only at univariate To date, all the English language articles regarding PMP treated by
analysis; in the same study, increased baseline CA19.9 was an CRS combined with locoregional chemotherapy are observational
independent predictor of worse progression-free survival at multi- studies without control groups performed by tertiary referral centers.
variate analysis [37]. In 63 patients, van Ruth found only weak Most are serial publications reporting accumulating numbers of
statistical association between baseline CA19.9 and disease-free patients or increased follow-up. There are no randomized trials or
interval, but elevated CA19.9 after the procedure or rising during comparative studies. Six studies included 100 patients [8,11,14–
follow-up was related to disease recurrence [38]. Survival correlated to 16,18] and the remaining series <100 patients. Five studies exclusively
preoperative CEA and CA19.9 among 532 patients studied by reported PMP originating from appendiceal mucinous neoplasms
Carmignani. Both markers, measured at the time of disease recurrence, [8,11,14,15,17] and the remaining PMP originating mainly from
correlated to survival after a second cytoreduction [39]. In a recent appendiceal mucinous neoplasms, representing 86–97% of their

TABLE I. Technical Modalities of Perioperative Intra-Peritoneal Chemotherapy for Pseudomyxoma Peritonei Combined with Cytoreductive Surgery

Principal investigator Center Year Perioperative intra-peritoneal chemotherapy

McGregor [9] Vancouver 2002 EPIC (MMCþ5-FU)


Elias [10] Paris 2003 HIPEC (MMC or OX or MMCþCisplatin) þ/ EPIC (5FU or 5FUþ FA)
Sugarbaker [11] Washington DC 2004 HIPEC (MMC)þ EPIC (5-FU)
Piso [12] Regensburg 2005 HIPEC (MMC or Cisplatin)
Glehen [13] Lyon 2005 HIPEC (MMCþCisplatin)
Loggie [14] Winston-Salem 2006 HIPEC (MMC)
Moran [15] Basingstoke 2006 HIPEC (MMC)þ EPIC (5-FU)
Zoetmulder [16] Amsterdam 2007 HIPEC (MMC)
Morris [17] Sydney 2006 HIPEC (MMC)þ EPIC(5-FU)
Deraco [18] Milan 2007 HIPEC (MMCþcisplatin)

HIPEC: hyperthermic intra-peritoneal chemotherapy; EPIC: early postoperative intra-peritoneal chemotherapy; MMC: Mitomycin-C; 5-FU: 5-fluoruracil; OX:
oxaplatin; FA: folinic acid.

Journal of Surgical Oncology


280 Moran et al.
TABLE II. Effectiveness of Cytoreductive Surgery and Perioperative Intra-Peritoneal Chemotherapy for Pseudomyxoma Peritonei

Overall survival (%) Disease status (%)

Principal investigator Pts (no.) Median survival (months) 1 year 2 years 3 years 5 years 10 years NED AWD DFD

McGregor [9] 11 NA — — 60 — — — — —
Elias [10] 36 NA — — — 66 — 23 3 4
Sugarbaker [11] 501 156 — — — 72 55 47 15 32
Piso [12] 28 51 — — — — — — —— —
Glehen [13] 27 NA 100 88 78 52 — — — —
Loggie [14] 110 64 80 — 59 53 — — — —
Moran [15] 123 NA — 57* 758 — — — —
Zoetmulder [16] 103 NA 90 83 71 60 — 61 5 19
Morris [17] 50 NA 89 76 69 69 — — — —
Deraco [18] 104 NA — — — 72 — 68 15 18

NED: no evidence of disease; AWD: alive with disease; DFD: died from disease; NA: not reached; 8: 83 patients undergoing complete cytoreduction; *: in 34 patients
undergoing major palliative resections.

sample population. The rate of complete cytoreduction is 40–91%. not amenable to complete cytoreduction was endorsed by 78% of
This supports the benefits of centralising this aggressive surgical the panel, but 89% felt not for all cases, and 67% agreement
treatment at institutions with an interest in PMP. Effectiveness of CRS for appendiceal tumours. In limited disease, 65% felt a limited
and PLC on survival and recurrence is demonstrated in Table II. peritonectomy was not appropriate, though 63% felt a complete
A number of recent reports in the literature have addressed the parietal peritonectomy was not necessary for patients with a limited
morbidity and mortality in patients with PMP treated by CRS and PLC affected area. In patients with palliative or inoperable disease, 56% felt
(see Table III). The overall morbidity rate varied from 33 to 56%, PLC should not be used compared to 44% in favour suggesting that this
hematological toxicity from 4 to 9%, blood loss from 2100 to aspect might be amenable to a randomized trial. There was ambiguity
8000 cm3, mean operation duration from 6.0 to 12.6 hr. Re-operation concerning the optimum drug and relative temperature though 428C
rates for postoperative adverse events were 11–21%, as reported in two was considered optimal for irinotecan and gemcitabine by 100% and
studies. Overall mortality rates ranged from 0 to 18%. The median and for mitomycin and doxorubicin by 63% and 67%, respectively. The
mean hospital stay ranged from 16 to 21 days and 26 to 29 days, optimal single agent was felt to be mitomycin-C by 100% but 66% felt
respectively (see Table III). that combination therapy was optimal. If combination therapy was
One aspect that has not been fully addressed is a strategy for the used 83% favoured cisplatin and mitomycin-C.
many patients whose tumours were preoperatively considered unlikely A common dilemma is the aspect of systemic chemotherapy in
to be completely removable, either due to tumour extent and PMP. There was unanimous consensus that systemic chemotherapy
distribution, or as a result of serious co-morbidity or age. There is should not be used whilst awaiting surgery and HIPEC, but should be
increasing evidence that many of these may benefit from a major given in non-resectable cases and those with high tumour grade. High
palliative resection with reasonable intermediate-term survival of 43% tumour markers were considered an indication to consider systemic
at 2 years and 15% at 5 years and improved quality of life [42]. In these chemotherapy by 71%.
situations an approach involving extended right hemicolectomy, There was 100% agreement that the preferred imaging modality for
greater omentectomy and splenectomy with an ileocolic anastomosis follow-up was CT-scan with no role for MRI, PET or CT PET. Tumour
may be advisable [43]. Glehen recommended combination of markers, CT imaging and clinical examination at intervals of
comprehensive surgical debulking with HIPEC except for patients 0–12 months were favoured by 89% with 11% suggesting an
with signet ring histology or lymph node involvement in his experience interval of 12–24 months. For asymptomatic recurrence, 66%
of 174 patients with incomplete cytoreduction [42]. felt should have early surgery and 33% voted for no surgery and
There remains confusion as to the definition of complete systemic chemotherapy. There was 100% support for a clinical trial of
cytoreduction with 100% stating this should be CC0 but 100% also cytoreductive surgery and HIPEC followed by best systemic
agreeing it should include CC1. Maximal palliative surgery for patients chemotherapy in patients with high-grade disease if a complete

TABLE III. Morbidity and Mortality of Cytoreductive Surgery and Perioperative Intra-Peritoneal Chemotherapy for Pseudomyxoma Peritonei

Principal Hematol. Hospital


investigator Pts (no.) Morbidity (%) toxicity (%) Blood loss (ml) Operation duration (hr) Reoperation (%) Mortality (%) stay (days)

McGregor [9] 11 56 — — — — 18 —
Elias [10] 36 44 19 1200 10.08 14 248
Piso [12] 28 36 — 2100 6.0* 21 7 29*
Glehen [13] 27 44 — — — — 0 168
Loggie [14] 110 38 4 — 10.5* — — —
Moran [15] 123 — — — 108 — 5 228
Sugarbaker [46] 356 40 5 — — 11 2 218
Zoetmulder [47] 103 54 — 8000 9.0 — 3 218
Morris [17] 50 48 — — 10.0* — 4 26*
Deraco [18] 104 19 4 — — — 1 —

* mean; 8 median.

Journal of Surgical Oncology


Consensus on Pseudomyxoma Peritonei 281
cytoreduction was achieved. There was a 50/50 split concerning 10. Elias D, Lasser P, Reynard B, et al.: Pseudomyxoma peritonei
systemic chemotherapy after complete cytoreduction in patients with treated with complete resection and immediate intraperitoneal
high tumour markers suggesting an area that could be considered for a chemotherapy. Gastroenterol Clin Biol 2003;27:407–412.
randomized trial. 11. Gonzalez-Moreno S, Sugarbaker PH: Right hemicolectomy does
not confer a survival advantage in patients with mucinous
carcinoma of the appendix and peritoneal seeding. Br J Surg
CONCLUSION 2004;91:304–311.
12. Guner Z, Schmidt U, Dahlke MH, et al.: Cytoreductive surgery
Recent evidence suggests that optimal surgical resection (complete and intraperitoneal chemotherapy for pseudomyxoma peritonei.
cytoreduction if possible) combined with HIPEC is the most Int J Colorectal Dis 2005;20:155–160.
13. Loungnarath R, Causeret S, Bossard N, et al.: Cytoreductive
fundamentally based strategy for PMP. A combined approach makes
surgery with intraperitoneal chemohyperthermia for the treatment
both common and scientific sense, in that surgery attempts to remove of pseudomyxoma peritonei: a prospective study. Dis Colon
all macroscopic disease whilst HIPEC addresses residual microscopic Rectum 2005;48:1372–1379.
disease. This treatment strategy is a complex procedure, associated 14. Stewart JH, Shen P, Russell GB, et al.: Appendiceal neoplasms
with significant morbidity and mortality with a substantial institutional with peritoneal dissemination: outcomes after cytoreductive
and individual ‘‘learning curve’’ phenomenon [44,45]. surgery and intraperitoneal hyperthermic chemotherapy. Ann
The clinical results for cytoreduction and HIPEC in PMP show Surg Oncol 2006;13:624–634.
good survival for those patients with low-grade histology amenable to 15. Murphy EM, Sexton R, Moran BJ: Early results of surgery in 123
complete cytoreduction. Poorly defined and often confusing terms in patients with pseudomyxoma peritonei from a perforated
appendiceal neoplasm. Dis Colon Rectum 2006;50:37–42.
the literature have resulted in most series being a heterogeneous
16. Smeenk RM, Verwaal VJ, Antonini N, et al.: Survival analysis of
population. This has limited the quality of the evidence with a lack of pseudomyxoma peritonei patients treated by cytoreductive
randomised controlled trials [41]. This deficiency is not unique to PMP surgery and hyperthermic chemotherapy. Ann Surg 2007;245:
surgery as few, if any, major surgical techniques are amenable to 104–109.
randomisation. Recent publications in the last 15 years with increasing 17. Yan TD, Links M, Xu ZY, et al.: Cytoreductive surgery and
numbers of medium to large case-series in the literature reflect an perioperative intraperitoneal chemotherapy for pseudomyxoma
improved awareness and understanding of the disease. This trend peritonei from appendiceal mucinous neoplasms. Br J Surg 2006;
should continue with the development of centralised treatment centres 93:1270–1276.
throughout the world such that the quality of care and information 18. Baratti D, Kusamura S, Nonaka D, et al.: Pseudomyxoma
peritonei: clinical pathological and biological prognostic factors
available for patients with PMP should ultimately improve.
in patients treated with cytoreductive surgery and hyperthermic
An emerging network of specialized centres may facilitate multi- intraperitoneal chemotherapy (HIPEC). Ann Surg Oncol 2008;15:
centre studies on aspects of chemotherapy type, duration and 526–534.
temperature to help allay the criticisms of many surgical, and in 19. Casali P, Licitra L, Tondini C, et al.: START: a European state-of-
particular, medical oncologists on the lack of hard scientific evidence the-art on-line instrument for clinical oncologists. Ann Oncol
in PMP management. 1999;10:769–773.
Meanwhile good evidence is rapidly accumulating and surgical 20. Ronnett BM, Zahn CM, Kurman RJ, et al.: Disseminated
nihilism is no longer acceptable in this inexorably progressive, peritoneal adenomucinosis and peritoneal mucinous carcinoma-
universally fatal, but eminently treatable disease. tosis: a clinicopathologic analysis of 109 cases with emphasis on
distinguishing pathologic features, site of origin, prognosis and
relationship to ‘‘pseudomyxoma peritonei. Am J Surg Pathol
1995;19:1390–1408.
REFERENCES 21. Szych C, Staebler A, Connolly DC, et al.: Molecular genetic
evidence supporting the clonality and appendiceal origin of
1. Moran BJ, Cecil TD: The etiology, clinical presentation, and pseudomyxoma peritonei in women. Am J Path 1849–1855.
management of pseudomyxoma peritonei. Surg Oncol Cin N Am 22. Carr NJ, Emory TS, Sobin LH: Epithelial neoplasms of the
2003;12:585–603. appendix and colorectum: an analysis of cell proliferation,
2. Sugarbaker PH: New standard of care for appendiceal epithelial apaptosis and expression of p53, CD44, bcl-2. Arch Pathol Lab
neoplasms and pseudomyxoma peritonei syndrome? Lancet Med 2002;126:837–841.
Oncol 2006;7:69–76. 23. Kahn MA, Demopoulos RI: Mucinous ovarian tumors
3. Weaver CH: Mucocele of the appendix with pseudomucinous with pseudomyxoma peritonei: a clinicopathological study. Int
degeneration. Am J Surg 1937;36:523–526. J Gynecol Pathol 1992;11:15–23.
4. Gough DB, Donohue JH, Schutt AJ, et al.: Pseudomyxoma 24. Chejfec G, Rieker WJ, Jablokow VR, et al.: Pseudomyxoma
peritonei. Long-term patient survival with an aggressive regional peritonei associated with colloid carcinoma of the pancreas.
approach. Am Surg 1994;219:112–119. Gastroenterology 1986;90:202–205.
5. Miner TJ, Shia J, Jaques DP, et al.: Long-term survival following 25. Hawes D, Robinson R, Wira R: Pseudomyxoma peritonei from
treatment of pseudomyxoma peritonei: an analysis of surgical metastatic colloid carcinoma of the breast. Gastrointest Radiol
therapy. Ann Surg 2005;241:300–308. 1991;16:80–82.
6. Misdraji J, Yantiss RK, Graeme-Cook FM, et al.: Appendiceal 26. Ronnett BM, Seidman JD: Mucinous tumors arising in ovarian
mucinous neoplasms: a clinicopathologic analysis of 107 cases. mature cystic teratomas: relationship to the clinical syndrome of
Am J Surg Pathol 2003;27:1089–1103. pseudomyxoma peritonei. Am J Surg Pathol 2003;27:650–657.
7. Yan H, Pestieau SR, Shmookler BM, et al.: Histopathologic 27. Ronnett BM, Schmooker BM, Diener-West M, et al.: Immuno-
analysis in 46 patients with pseudomyxoma peritonei syndrome: histochemical evidence supporting the appendiceal origin of
failure versus success with a second-look operation. Mod Pathol pseudomyxoma peritonei in women. Int J Gynecol Pathol 1997;
2001;14:164–171. 16:1–19.
8. Sugarbaker PH, Chang D: Results of treatment of 385 patients 28. Higa E, Rosai J, Pizzimbono CA, et al.: Mucosal hyperplasia,
with peritoneal surface spread of appendiceal malignancy. Ann mucinous cystadenoma, and mucinous cystadenocarcinoma of the
Surg Oncol 1999;6:727–731. appendix. A. re-evaluation of appendiceal ’mucocele’. Cancer
9. Butterworth SA, Panton NM, Klaassen DJ, et al.: Morbidity and 1973;32:1525–1541.
mortality associated with intraperitoneal chemotherapy for 29. Bradley RF, Stewart JH, Russell GB, et al.: Pseudomyxoma
pseudomyxoma peritonei. Am J Surg 2002;183:529–532. peritonei of appendiceal origin: a clinicopathological analysis of

Journal of Surgical Oncology


282 Moran et al.
101 patients uniformly treated at a single institution, with 39. Carmignani CP, Hampton R, Sugarbaker CE, et al.: Utility of
literature review. Am J Surg Pathol 2006;30:551–559. CEA and CA19-9tumor markers in diagnosis and prognostic
30. Esquivel J, Sugarbaker PH: Clinical presentation of pseudomy- assessment of mucinous epithelial cancers of the appendix. J Surg
xoma peritonei syndrome. Br J Surg 2000;87:1414–1418. Oncol 2004;87:162–166.
31. Jacquet P, Jelinek JS, Chang D, et al.: Abdominal computed 40. Alexander-Sefre F, Chandrakumaran K, Banerjee S, et al.:
tomographic scan in the selection of patients with mucinous Elevated tumour markers prior to complete tumour removal in
peritoneal carcinomatosis for cytoreductive surgery. J Am Coll patients with pseudomyxoma peritonei predict early recurrence.
Surg 1995;181:530–538. Colorectal Dis 2005;7:382–386.
32. Sulkin TV, O’Neill H, Amin AI, et al.: CT in pseudomyxoma 41. Yan TD, Black D, Savady R, et al.: A systematic review on the
peritonei: A review of 17 cases. Clin Radiol 2002;57:608–613. efficacy of cytoreductive surgery and perioperative intraperitoneal
33. Sugarbaker PH: Pseudomyxoma peritonei. A. Cancer whose chemotherapy for pseudomyxoma peritonei. Ann Surg Oncol
biology is characterized by a redistribution phenomenon. Ann 2006;14:484–492.
Surg 1994;219(2):109–111. 42. Glehen O, Mohamed F, Sugarbaker PH: Incomplete cytoreduction
34. Low RN, Barone RM, Gurnet JM, et al.: Mucinous appendiceal in 174 patients with peritoneal carcinomatosis from appendiceal
neoplasms: preoperative MR staging and tumor classification malignancy. Ann Surg 2004;240:278–285.
compared to surgical and histopathological findings. Ann Surg 43. Farquharson SM, Murphy E, Sexton R, et al.: What happens to
Oncol 2006;13:P182. patients following surgery for peritoneal malignancy with
35. Stewart JH, Perry S, Levine EA: Intraperitoneal hyperthermic incomplete tumour removal? Analysis of 100 consecutive
chemotherapy for peritoneal surface malignancy: current status laparotomies. J Roy Coll Surg Ed & Ireland 2005;92: Supp, p. 7.
and future directions. Ann Surg Oncol 2005;12:765–777. 44. Moran BJ: Establishment of a peritoneal malignancy treatment
36. Pomel C, Appleyard TL, Gouy S, et al.: The role of laparoscopy to centre in the United Kingdom. Eur J Surg Oncol 2006;32:614–
evaluate candidates for complete cytoreduction of peritoneal 618.
carcinomatosis and hyperthermic intraperitoneal chemotherapy. 45. Smeenk RM, Verwaal VJ, Zoetmulder FA: Learning curve of
Eur J Surg Oncol 2005;31(5):540–543. combined modality treatment in peritoneal surface disease. Br
37. Baratti D, Kusamura S, Martinetti A, et al.: Prognostic value of J Surg 2007;94:1408–1414.
circulating tumor markers in patients with pseudomyxoma 46. Sugarbaker PH, Alderman R, Edwards G, et al.: Prospective
peritonei treated with cytoreductive surgery and hyperthermic morbidity and mortality assessment of cytoreductive surgery plus
intra-peritoneal chemotherapy (HIPEC). Ann Surg Oncol 2007; perioperative intraperitoneal chemotherapy to treat peritoneal
14:500–508. dissemination of appendiceal mucinous malignancy. Ann Surg
38. Van Ruth S, Hart AA, Bonfrer JM, et al.: Prognostic value of Oncol 2006;13:1–11.
baseline and serial carcinoembryonic antigen and carbohydrate 47. Smeenk RM, Verwaal VJ, Zoetmulder FAN: Toxicity and
antigen 19.9 measurements in patients with pseudomyxoma mortality of cytoreduction and intraoperative hyperthermic intra-
peritonei treated with cytoreduction and hyperthermic intra- peritoneal chemotherapy in pseudomyxoma peritonei—a report
peritoneal chemotherapy. Ann Surg Oncol 2002;9:961–967. of 103 procedures. Eur J Surg Oncol 2006;32:186–190.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:283–290

Hyperthermic Intraperitoneal Chemotherapy With and


Without Cytoreductive Surgery for Epithelial Ovarian Cancer

C. WILLIAM HELM, MB.BChir,1* ROBERT E. BRISTOW, MD,2 SHIGEKI KUSAMURA, MD, PhD,
3

DARIO BARATTI, MD,3 AND MARCELLO DERACO, MD3


1
Division of Gynecologic Oncology, James Graham Brown Cancer Center, University of Louisville, Kentucky
2
The Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions,
Baltimore, Maryland
3
Department of Surgery, National Cancer Institute of Milan, Milan, Italy

Women with epithelial ovarian cancer (EOC) usually present with advanced disease and overall only just over half survive 5 years. Even following
a complete response to front-line treatment two-thirds will recur, with a resultant dismal prognosis. We review and discuss the role of surgery and
hyperthermic intraperitoneal chemotherapy (HIPEC) in EOC and present the results of the ovary consensus panel (OCP) convened for the 5th
International Workshop on Peritoneal Surface Malignancy.
J. Surg. Oncol. 2008;98:283–290. ß 2008 Wiley-Liss, Inc.

KEY WORDS: hyperthermic intraperitoneal chemotherapy (HIPEC); cytoreductive surgery; epithelial ovarian cancer;
hyperthermia; loco-regional therapy; intraperitoneal chemotherapy

INTRODUCTION AND BACKGROUND RATIONALE FOR HYPERTHERMIA WITH


Worldwide there are approximately 204,000 new cases and
INTRAPERITONEAL CHEMOTHERAPY
125,000 deaths per annum from epithelial ovarian cancer (EOC) [1]. Hyperthermia alone is tumoricidal [6] and it increases the
The disease most often presents in advanced stage by which time cure cytotoxicity of many chemotherapeutic agents in both human cell
is difficult. This review addresses the role that hyperthermic culture and animal models [7–11]. Much of the work in this area has
intraperitoneal chemotherapy (HIPEC) with or without cytoreductive been performed on cisplatin with investigations of the cellular effects
surgery (CRS) might play in improving the treatment of EOC and of the combination demonstrating increased DNA cross-linking and
reports the results of the ovary consensus panel (OCP) convened for increased DNA adduct formation [9,12] and deeper penetration into
the 5th International Workshop on Peritoneal Surface Malignancy. peritoneal tumor implants when delivered intraperitoneally with
The OCP responded to a series of clinical questions set by the co- hyperthermia [12]. However, the precise underlying molecular
chairmen (REB and CWH) with answers rated from 1 to 5 (strongly mechanisms of these effects are still unknown.
disagree to strongly agree). Opinions reported here are the percentage The possible synergy between hyperthermia and chemotherapy
of the OCP answering 4 or 5 to the question. Levels of evidence are treatment combined with the possibility of delivering these two
classified by that described by the Center for Evidence-Based modalities directly into the peritoneal cavity has sparked clinical trials
Medicine, Oxford [2] and ‘State-of-the-Art Oncology in Europe’ in many cancers including, gastric [13], mesothelioma [14], appendi-
(START) [3]. ceal [15], endometrial [16] and colorectal [17]. A phase III randomized
study of HIPEC following CRS surgery compared with traditional
RATIONALE FOR REGIONAL intravenous chemotherapy and palliative care in patients with
(INTRAPERITONEAL) CHEMOTHERAPY peritoneal spread of colo-rectal carcinoma showed a statistically
significant prolongation of life in the experimental arm [17].
IN OVARIAN CANCER
HIPEC and CRS include the following modalities: hyperthermia,
intraperitoneal chemotherapy and CRS. Dedrick proposed that the The authors have no financial interest related to the contents of this article to
intraperitoneal delivery of certain chemotherapeutic agents could lead disclose.
to a significant increase in peritoneal cavity drug exposure compared to *Correspondence to: Dr. C. William Helm, MB.BChir, Associate Professor,
that in the systemic vascular compartment [4]. For drugs most active in Division of Gynecologic Oncology, James Graham Brown Cancer Center,
EOC the ratio of their intraperitoneal to plasma concentrations varies University of Louisville, 529 South Jackson Street, Louisville, KY 40207.
from 18 to 20 for carboplatin and cisplatin to 120 to >1,000 for the Fax 502-561-7205. E-mail: cwhelm@uoflobgyn.com
taxanes, docetaxel and paclitaxel (see Table I) [5]. EOC should be a Received 15 April 2008; Accepted 15 April 2008
good target for intraperitoneal treatment because it is relatively chemo- DOI 10.1002/jso.21083
sensitive and it remains confined within the peritoneal cavity for much Published online in Wiley InterScience
of its natural history. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


284 Helm et al.
TABLE I. Peritoneal to Plasma Ratios for Peak Concentration and AUC of 2.5–5 mm even with hyperthermia [27–29], the largest residual
lesion size prior to HIPEC should be no more than 5 mm and optimally
Peritoneal to plasma ratio
less. Some drugs such as doxorubicin, carboplatin and oxaliplatin have
Drug Peak AUC
much lower intratumoral penetration capacity. Some 90% of the OCP
felt that the objective should be a residual lesion size of 2.5 mm or less
Cisplatin 20 12 prior to HIPEC.
Paclitaxel >1,000 >1,000 There appears to be a clear theoretical rationale for the utilization of
Gemcitabinea 500 CRS with HIPEC in ovarian cancer but who should be considered for
Doxorubicin 474 230 this and at what time-points in the natural history of the disease?
Melphalan 93 65
5-Fluoruracilb 298 367
Oxaliplatin 25 16
PRE-OPERATIVE EVALUATION
Carboplatin 18 18 AND ELIGIBILITY
Docetaxel 120 552
Mitomycin 71 23 In order to undergo CRS and/or HIPEC, eligible patients must be
Mitoxantrone 57 115 sufficiently healthy to withstand the surgery and chemotherapy and
Interferon-a 100 their disease should be resectable to <5mm residual and confined to
Etoposide 65 the peritoneal cavity. Since in the front-line setting, malignant pleural
effusions in the absence of bulk disease in the chest and resectable
Adapted from [5].
a groin node disease are not considered contraindications to CRS, they
Cytotoxic activity augmented if given before hyperthermia.
b
Activity not augmented by hyperthermia. could be considered within the criteria of eligibility for HIPEC pending
further investigation. In fact, metastatic disease to groin nodes does
not upstage the disease from IIIC in the front-line setting [30]. The
The standard initial treatment of EOC following an attempt OCP were equally split on this issue. Those that said they would give
at maximal cytoreductive surgery involves combination normothermic HIPEC would do so for sub-renal retroperitoneal nodes 89%, liver
platinum/taxane regimens previously given intravenously only and parenchymal disease 78%, groin nodes 67%, and positive pleural
now often including combined intravenous/intraperitoneal [18]. For effusion 56%.
the treatment of recurrent EOC a wide range of agents have shown Pre-operative assessment to identify patients whose disease is not
activity in addition to platinums and taxanes including topotecan, likely to be optimally resectable would enable such patients to avoid
etoposide, liposomal doxorubicin, doxorubicin, epirubicin, gemcita- the morbidity of unnecessary surgery. Although several studies have
bine, hexamethylmelamine, navelbine, ifosfamide, irinotecan, 5- investigated the accuracy of computed tomography (CT) scans in
fluorouracil (5FU), melphalan and mitoxantrone with response rates determining the resectability of EOC [31–35] the factors associated
between 20% and 77% in platinum-sensitive disease and up to 28% in with prediction of suboptimal surgery vary between studies and centers
platinum-resistant disease [19]. Until newer agents come on-line any of probably reflecting the surgical philosophy at individual institutions
these agents with proven activity could conceivably be used for HIPEC [36]. Magnetic resonance imaging (MRI) is equally as accurate in the
provided that they fit with the Dedrick theory, have activity enhanced staging of EOC as CT [35,37]. 18F-FDG PET may add accuracy
by heat and have acceptable side-effects. in evaluating peritoneal carcinomatosis [38]. The level of surgical
expertise is of major importance with regard to the operability of EOC
and this will ultimately determine the chance of optimal CRS [39].
RATIONALE FOR CYTOREDUCTIVE SURGERY
Although the serum CA125 level has been correlated with the
The role of cytoreductive surgery (CRS) in the front-line treatment achievement of optimal CRS, it is not accurate enough to be used
of EOC was initially demonstrated in a study reporting an inverse alone [40–42]. The OCP consensus on investigations to determine the
relationship between the largest diameter of disease remaining after extent of disease is given in Figure 1.
CRS and survival [20]. Multiple subsequent retrospective reports
have confirmed this finding [21–23]. The role of surgery in this dual
modality approach was supported in a meta-analysis of 6885 patients
undergoing CRS during the platinum era where on an institutional
basis for each 10% increase in the percentage of patients undergoing
maximal CRS there was a 5.5% increase in median survival
duration [24].
The reason CRS is thought to be effective when combined
with chemotherapy is that it removes bulky disease containing poorly
oxygenated, non-proliferating cells which are either resistant or
potentially resistant to chemotherapy and leaves small volume tumors
with a higher proportion of cells in the proliferative phase making them
more susceptible to chemotherapy.
Even though ‘optimal’ residual disease at completion of initial CRS
for EOC was accepted as being any nodule <2 cm in dimension [25], it
is now established that the most favorable prognosis is in patients
with no macroscopic residual disease at all [26]. Currently, there is no
universally accepted working definition of optimal cytoreduction
among gynecologic oncologists. A survey of members of the Society
of Gynecologic Oncologists reported that the majority believed Fig. 1. Optimum investigations to define extent of disease—% of
optimal CRS was a residual maximal lesion size of 1 cm [26]. Based respondents answering 4 or 5. CT, computed tomography; C/A/P,
on in vitro and in vivo evidence that diffusion of intraperitoneally chest, abdomen, and pelvis; PET, positron emission tomography; CXR,
delivered chemotherapy agents into peritoneal tumors is to a maximum chest X-ray; MRI, magnetic resonance imaging.

Journal of Surgical Oncology


HIPEC With and Without CRS in Ovarian Cancer 285
therapy in ovarian cancer involving 1819 patients also revealed an
overall benefit of intraperitoneal chemotherapy [49].
HIPEC performed at the time of initial surgery would have the
following advantages over standard intraperitoneal chemotherapy:
(1) intraperitoneal adhesions have been divided allowing the HIPEC
solution to reach all surfaces of the peritoneal cavity, (2) residual tumor
is at the smallest possible volume, optimally leaving only microscopic
and free floating cells to be treated and (3) treatment would be
delivered many days prior to the usual time when postoperative
chemotherapy is given.
The first report of the use of HIPEC at the time of front-line CRS
was from the United States National Cancer Institute when a Phase 1
study was performed to examine the feasibility, toxicity and
pharmacokinetics of carboplatin given to six patients, five of whom
had small-volume residual disease following optimal CRS [50]. There
are reports of only around 50 patients treated in this fashion [51–
56,91]. These numbers are currently too small to assess any meaningful
effect. See Table II for a list of studies reporting HIPEC in ovarian
cancer in English.
Fig. 2. Pre-operative criteria to defer attempt at cytoreductive The precedent for treatment at the time of front-line surgery has
surgery until after neoadjuvant therapy—% responding 4 or 5. been set and further study is needed. Such studies would ideally be
randomized phase III design but this would require several hundred
patients and require collaboration between many centers. The natural
The OCP agreed broadly that involvement of the porta hepatis, history time-points that the OCP considered HIPEC to be relevant
liver parenchyma, mesenteric root, high small bowel obstruction were interval debulking and recurrent platinum-sensitive disease 79%,
(SBO), and extra-abdominal metastasis identified pre-operatively were consolidation 63%, front-line failure 58%, front-line 53%, and for
of concern to preclude a successful resection. These are not absolute platinum-resistant recurrent disease 42%.
contraindications to successful CRS and every case should be
individualized (Fig. 2). The OCP were concerned that the following Interval Debulking
medical morbidity indicators would preclude CRS and HIPEC: heart
failure 94%, pulmonary compromise 94%, previous pulmonary Although the standard of care for patients presenting with ovarian
embolus 63%, previous deep venous thrombosis without pulmonary cancer is initial maximal CRS, this is not always possible due to
embolus 26%, body mass index (BMI) >35 48%, BMI 30–34 26%. factors such as medical unfitness for major surgery, cardiac disease,
pulmonary compromise, recent pulmonary embolus, poor nutritional
status, and other co-morbidities. In this circumstance, neoadjuvant
TIME-POINTS FOR POSSIBLE USE chemotherapy for two to three cycles may allow time for performance
OF CRS AND HIPEC status to be improved whilst reducing the extent of disease and
increasing tumor resectability. This may lead to less intraoperative
There are five time-points in the natural history of EOC at which blood loss, shorter operative times, less intensive care unit admissions,
discussion of treatment with HIPEC and research can usefully be and shorter length of hospital stay [69,70]. Another indication for
directed: front-line, interval debulking following initial neo-adjuvant neoadjuvant chemotherapy may be the absence of a surgical specialist
chemotherapy, consolidation following complete response to front-line to perform the necessary CRS in the local setting.
therapy and at the time of recurrence, including front-line failure with Individual case series on outcome for neoadjuvant chemotherapy
disease still present at the end of initial treatment. followed by interval debulking involve relatively small numbers and
heterogeneous groups of patients and surgeons [70] but outcomes are
Front-line overall not as good as those achieved with front-line maximal CRS
followed by chemotherapy [71,72].
Standard front-line therapy for ovarian carcinoma evolved to A useful time to investigate the incorporation of HIPEC would be in
be CRS followed by intravenously administered chemotherapy with a those situations where interval debulking surgery is performed. In
platinum and taxane combination, usually carboplatin/paclitaxel. addition to the theoretical reasons stated for using front-line, there
Although response rates of EOC to front-line chemotherapy are high, would be the advantage of lead-time to arrange for the HIPEC
20–30% of tumors are inherently platinum-resistant [43] and 60–70% perfusion team and equipment to be available at this surgery. Its use
of all patients with ovarian carcinoma will recur [44–47] with the may add progression-free or survival benefit to these patients whose
overall 5-year survival still being only around 50%. overall outcome is inferior to those treated with initial maximal CRS
Clinical research interest in regional treatment for ovarian followed by chemotherapy. Several series have included patients
carcinoma has existed for many years but in January 2006 interest treated with HIPEC at the time of interval debulking but the numbers
rose when a Gynecologic Oncology Group study (GOG-172) are too small for meaningful analysis and conclusions[50,54,62,64,66];
randomizing 415 patients to either standard intravenous chemotherapy the level of evidence is 4 [2] and R [3].
only, or experimental intravenous/intraperitoneal chemotherapy fol- Ryu [64] reported on 57 women who underwent HIPEC at the time
lowing optimal CRS for ovarian cancer, showed a significant increase of either interval debulking or second-look surgery following front-line
in median progression-free and overall survival in the experimental therapy but it is not known how many were treated in each group.
arm from 18 to 23 months and 49 to 66 months, respectively.[18] Others have reported small numbers. [50,54,62,66] HIPEC at
GOG-172 was the third of three, large randomized studies [18,28,48] the time of interval debulking would be a fruitful area for research.
reporting a benefit for intraperitoneal chemotherapy. A Cochrane Currently, there is a randomized phase III trial under coordination by
Systematic Review of all randomized trials of intraperitoneal chemo- the Netherlands Cancer Center, Amsterdam randomizing patients to

Journal of Surgical Oncology


286 Helm et al.
TABLE II. Studies of HIPEC for EOC
Time Temperature
Author Year Reference Type Situation n Agent Dose (min) (8C)

Loggie 1994 [57] Pilot Advanced 1 Mitomycin 30 mg 120 39–40.5


Steller 1999 [50] Pilot Front-line 6 Carboplatin 800–1,200 mg/m2 90 41–43
van der Vange 2000 [58] Pilot Recurrent/persistent 5 Cisplatin 50/75 mg/m2 90 40
Cavalieri 2000 [59] Retrospective Recurrent/persistent 20 Cisplatin 25 mg/m2/L 90 41.5–42.5
Deraco 2001 [60] Phase II Recurrent/persistent 27 Cisplatin 25 mg/m2/L 60 42.5
Panteix 2002 [61] pK Recurrent 16 Cisplatin 60/80/100 mg 90 41–43
de Bree 2003 [62] Prospective Recurrent/persistent 19 Docetaxel 75 mg/m2 120 41–43
Zanon 2004 [63] Phase II Recurrent/persistent 30 Cisplatin 100/150 mg/m2 60 41.5–42.5
Look 2004 [52] Retrospective Front-line, recurrent/persistent 12a Cisplatin mitomycin b
60 41.5–42.5
Piso 2004 [53] Retrospective Primary/recurrent 19 Cisplatin or mitoxantrone 75 mg/m2 90 41.5
15 mg/m2
Ryu 2004 [64] Retrospective Interval 57 Carboplatin interferon-a 350 mg/m2 90 43–44
debulking/second-look 5  106 IU/m2
Gori 2005 [65] Retrospective Second-look 32 Cisplatin 100 mg/m2 60 41–43
Reichman 2005 [66] Retrospective Recurrent/interval debulking 13 Cisplatin 50 mg/m2 90 40
Yoshida 2005 [54] Retrospective Front-line/interval 10 Cisplatin mitomycin 100 mg 20 mg 50 41–44.5
debulking/second-look etoposide 100 mg
Raspagliesi 2006 [67] Retrospective Recurrent/persistent 40c Cisplatin/MMC or 25/3.3 mg/m2/L b
42.5
cisplatin/ doxorubicin 43/15.25 mg L
Rufian 2006 [55] Retrospective Front-line/recurrent 33 Paclitaxel 60 mg/m2 60 41–43
Helm 2007 [68] Retrospective Recurrent/persistent 18 Cisplatin or mitomycin 100 mg/m2 40 mg 90 42–43
Lentz 2007 [91] Phase I Front-line/second-look 17 Carboplatin 400–1200 mg/m2 90 39–43
Cotte 2007 [56] Prospective Front-line, recurrent/persistent 81 Cisplatin 20 mg/m2/L 90 44–46 inflow
Max. 80 mg

Excludes case report [51].


a
Some patients received EPIC.
b
Not stated.
c
Some previously reported in [60].

receive HIPEC or no HIPEC at the time of interval debulking (W. van review of patients treated with intraperitoneal chemotherapy agents
Driel, personal communication). including cisplatin and paclitaxel [76]. In a group of 89 patients with
The Consensus Group’s opinion with regard to the pre-operative complete pathologic response following front-line therapy treated for
criteria to defer an attempt at cytoreductive surgery until after consolidation, the median survival was 8.7 years. In a phase II study
neoadjuvant chemotherapy is detailed in Figure 2. There was broad from the same institution using IP cisplatin and etoposide, 36 patients
agreement that involvement of the porta hepatis, liver parenchyma, were compared with 46 historical controls and recurrence occurred in
mesenteric root, high small bowel obstruction, and extra-abdominal 39% versus 54%, respectively [77,78].
metastasis identified pre-operatively were of concern to preclude a There is some non-randomized evidence that disease found
successful resection. These are not absolute contraindications to at second-look surgery following front-line therapy may be helped
successful CRS and every case should be individualized. by CRS if it can be reduced to microscopic or small volume [79–82].
CRS alone has no role in this situation and chemotherapy would
Consolidation always be required for best outcome. Secondary CRS for persistent
disease following a previous attempt at maximal CRS has been
Despite the high initial response rates to front-line CRS and investigated and shown not to significantly extend survival [83].
chemotherapy in EOC in most patients the disease will recur. In GOG- Patients with platinum-resistant disease do extremely poorly with both
172, 65% of those in the experimental IP/IV arm (which had the best CRS and chemotherapy. There have been no randomized studies
outcome) developed recurrent disease [18]. Almost two decades ago it investigating the role of HIPEC as consolidation therapy at second-
was reported that stage III/IV patients with a complete pathologic look laparotomy in EOC but some promising work has been reported in
response confirmed at second-look surgery experienced recurrence this setting, although the level of evidence is 4 [2] and 3 [3]. Ryu
rates of 28% at 2 years, 56% at 5 years, and 60% at 10 years [73]. One et al.[64] reported on 57 women who underwent front-line cytor-
way of improving outcomes in EOC is to consider methods of eductive surgery followed by intravenous chemotherapy and were then
consolidating front-line therapy in those patients with no clinical treated with a second surgery with HIPEC who were compared with
evidence of disease. a historical control group of 60 patients who underwent initial cyto-
There is no established method of consolidation following front- reductive surgery followed by intravenous chemotherapy only. HIPEC
line therapy for EOC. A randomized study, investigating additional was given in the form of carboplatin 350 mg/m2 and interferon-alpha at
intravenous paclitaxel was stopped early when interim analysis showed a temperature of 43–448C. The disease-free survival and overall
a significantly improved disease-free survival in patients randomized to survival were significantly better in the experimental arm but the study
the 12-month arm (in comparison to the 3-month arm) and the trial included patients with germ cell tumors and it is not stated how many
was discontinued early making full analysis difficult [74]. Investigators received treatment at the time of interval debulking or as consolidation.
have looked at the use of chemotherapy agents intraperitoneally Gori [65] reported on 51 patients with ovarian carcinoma who
for consolidation. In 2003, the European Research Group (EORTC) underwent primary surgery with optimal cytoreduction to <2 cm
reported a randomized study of consolidation with four cycles of followed by chemotherapy with intravenous cisplatin and cyclopho-
intraperitoneal cisplatin versus no further treatment following com- sphamide. Of this group, 32 patients then underwent second-look
plete response to front-line therapy. The study was closed due to poor laparotomy with HIPEC cisplatin and 19 who refused second-look
recruitment but over 150 patients were entered and randomized. The laparotomy formed the control group. There was a trend to improved
recurrence rates were 49% in the experimental arm and 55% in the outcome in the HIPEC group with median survival of 64.4 months
control arm. [75] Memorial Hospital, New York reported a long-term versus 46.4 months in the control arm (P ¼ 0.29). In a Phase I study,

Journal of Surgical Oncology


HIPEC With and Without CRS in Ovarian Cancer 287
Lentz [91] performed HIPEC at the time of second-look laparotomy in Where reported, progression-free survival was 10, 23.9, and 31 months
14 patients to determine the maximum tolerated dose of carboplatin. [55,67,68]. Cotte [56] reported on 81 patients (65 with recurrent
The perhaps inappropriately named, ‘second-look laparotomy’, is a disease) with overall disease-free survival of 19.2 months and survival
relatively safe and accepted procedure for patients on clinical trials for of 28.4 months; 47 patients had three or more surgeries and it is not
ovarian carcinoma. As a marker of its safety, there were no deaths clear how many received more than one HIPEC treatment.
reported in seven major series including 682 operations [84]. In this The peri-operative mortality rate for CRS and HIPEC was 3.7%
era, it is not a procedure that is performed to merely assess the presence (8/236 cases) [50,53–55,58,60,62–65,67,68] compared with 1.4%
or absence of disease. Surgery is the only setting in which disease (0–3.4%) out of 631 patients undergoing CRS for recurrent EOC [88].
status can be accurately assessed due to the lack of accuracy of clinical Neither the risks nor risk/benefit ratio associated with HIPEC þ CRS
investigations to detect small volume disease. It is also the optimum versus CRS alone have been clearly defined. However, patients
way of delivering intraperitoneal chemotherapy in the total absence of historically selected for secondary CRS are likely patients with better
adhesions. The use of the laparoscope to assess the extent of disease disease characteristics and performance status than many who
and deliver HIPEC [85] could facilitate the incorporation of HIPEC underwent CRS þ HIPEC as shown in Table II.
research protocols in this setting. Where postoperative morbidity information for CRS þ HIPEC in
A randomized phase III study of HIPEC for consolidation would recurrent/persistent EOC is detailed in Table III [50,53–55,58,60,62–
require several hundred patients but would be the best way to 65,67,68] grade 3 or 4 hematologic toxicity was 4.7% (11/236) and
investigate it. Meanwhile, Phase I and II studies could investigate the raised creatinine, mainly associated with the use of cisplatin, was
tolerability and results of various drugs following front-line IV and 3.4% (8/236). The rate of anastomotic leaks was 1.7% (4/236) and
front-line IV/IP chemotherapy. Of the OCP, 63% felt that HIPEC gastrointestinal perforations 2.5% (6/236). The incidence of anasto-
should be considered at second-look laparotomy after complete motic leaks appears low but it is difficult to be sure of the exact
response to cisplatin/taxane therapy even if the patients had up to six percentage since the performance of diverting stomas is not always
courses of intraperitoneal cisplatin and taxane. A Phase I trial is mentioned. The occurrence of ‘spontaneous’ intestinal perforations
underway at the University of Louisville to investigate whether following HIPEC is of concern and may be related to the effect of
patients can tolerate HIPEC following standard front-line therapy with heat and chemotherapy agents on bowel serosa traumatized even
surgery followed by combined intravenous/intraperitoneal chemo- mildly during enterolysis. Sepsis is of concern when peri-operative
therapy. The OCP was equally split as to whether the consolidation
should be single agent or double agent. TABLE III. Complications Associated With CRS and Delivery of HIPEC
(n ¼ 236) Patientsa

Recurrence n

Most patients with EOC will initially respond to front-line therapy Hematologic
and then recur. The mainstay of treatment for recurrent EOC has been DIC 1
chemotherapy and a wide range of active agents which are listed in the Leucopenia grade three 6
‘‘Introduction’’ section [19]. Thrombocytopenia 4
Metabolic
Recurrence is most often not curable but duration of survival
Hypoalbuminemia 8
depends greatly on the time interval from initial treatment to the Raised liver enzymes 4
recurrence [86,87]. Patients with platinum-resistant tumors have Renal toxicity 8
response rates of up to 28% with an overall median survival of Renal failure 6
6–12 months and platinum-sensitive patients have response rates of up Unknown grade 2
to 77% with median survival of 12–40 months [19,88]. Pulmonary and neck
For patients with disease confined to the peritoneal cavity, Embolus 3
secondary CRS may have a role in extending survival with or without Pleural effusions 9
post-operative normothermic chemotherapy [88,89]. Patients with no Pneumonia 4
Central vein thrombosis 2
gross residual after CRS for recurrent disease have survival of
Surgical
44–60 months [88] with the best survival associated with a disease- Anastomotic leaks 4
free interval of more than 12 months, platinum-sensitive tumors, Intestinal perforation 5
absence of ascites, the number of recurrence sites, and the extent of Gastric perforation 1
residual disease [89,90]. Bowel obstruction 7
Since CRS with post-operative normothermic chemotherapy has Paralytic ileus 1
shown promise, the incorporation of HIPEC with CRS in the context of Intra-abdominal bleeding 5
recurrent disease should be investigated. All studies to date (Table II) Hematuria 2
have been non-randomized and are difficult to meaningfully compare Sepsis 10
Peritonitis 2
and assess due to their heterogeneity. Although the defined levels of
Wound infection 11
evidence are 4 [2] and 3 [3] it is possible to obtain some useful Wound seroma 1
information from this work. Abscess 3
The first paper to report the use of HIPEC in recurrent ovarian Dehiscence 2
cancer was that of van der Vange and colleagues in 2000. Studies with Total 1
the most clearly definable results for recurrent/persistent disease Deep 1
report median overall survivals of 28.1, 31, 41.4, and 57 months, Hernia 2
[55,63,67,68], overall 2-year survival 55% [60] and 60% [63], 3-year a
Excluding Grades 1 and 2 complications.
survival 37.5%, [61] 51%, [55] and 55% [66] and overall 5-year Other complications have included hematoma from the inferior epigastric
survival 15% [67]. Rufian reported 2- and 5-year survival of 75% for vessels, abdominal pain 4, temporary cutaneous fistula after removal of
patients with recurrent disease 55 years of age [55]. Panteix reported gastrostomy tube, and a femoral nerve neuropathy due to retraction at surgery,
an overall 7-year survival rate of 12.5% [61] and De Bree [62] reported complications of ileostomy, pancreatitis and neurological complication. Table
25% (3 of 12) of patients surviving a mean of 74 (72–79) months. incorporates data from [50,53–55,58,60,62–65,67,68].

Journal of Surgical Oncology


288 Helm et al.
chemotherapy is given and septic complications were reported in formation in patients and ovarian cancer cell lines. Eur J Cancer
26 (11%) patients with wound infection in 11 (4.7%), peritonitis in 1998;34:148–154.
2 (0.8%) and abscess in 3 (1%). Overall, the rates of severe morbidities 13. Fujimoto S, Takahashi M, Mutou T, et al.: Improved mortality rate
reported are not much higher than that of 11% (67/631) reported for of gastric carcinoma patients with peritoneal carcinomatosis
CRS alone [88]. treated with intraperitoneal hyperthermic chemoperfusion com-
bined with surgery. Cancer 1997;79:884–891.
14. Loggie BW, Fleming RA, McQuellon RP, et al.: Prospective trial
Prognostic Factors for the treatment of malignant peritoneal mesothelioma. Am Surg
2001;67:999–1003.
Reported prognostic factors for survival in recurrent/persistent 15. Sugarbaker PH, Chang D: Results of treatment of 385 patients
disease include the interval from diagnosis to HIPEC [60,68], the with peritoneal surface spread of appendiceal malignancy. Ann
peritoneal cancer index (PCI) [60,63,66], the extent of residual disease Surg Oncol 1999;6:727–731.
prior to HIPEC [55,60,63,66–68], patient age [55,60], WHO 16. Helm CW, Toler CR, Martin RS III, et al.: Surgical cytoreduction
performance status [67], and the presence of lymph node metastases and intraperitoneal heated chemotherapy (IPHC) for endometrial
[55]. carcinoma recurrent within the peritoneal cavity. Int J Gynecol
Cancer 2007;17:204–209.
Much more research is still required to improve outcomes in 17. Witkamp AJ, de Bree E, Kaag MM, et al.: Extensive
recurrent/persistent EOC. Randomized studies in this setting are cytoreductive surgery followed by intra-operative hyperthermic
extremely difficult to accomplish. A recent study of the Italian Society intraperitoneal chemotherapy with mitomycin-C in patients with
of Integrated Loco Regional Therapy (SITILO), which randomized peritoneal carcinomatosis of colorectal origin. Eur J Cancer 2001;
patients with persistent EOC following front-line therapy to CRS and 37:979–984.
HIPEC versus chemotherapy, only closed due to lack of accrual and 18. Armstrong DK, Bundy BN, Wenzel L, et al.: Intraperitoneal
suboptimal outcomes. Patients in this setting have a very poor survival cisplatin and paclitaxel in ovarian cancer. N Engl J Med 2006;
with standard therapy and further work is needed. The challenge will 354:34–43.
be to compare CRS with and without HIPEC in patients with 19. Berek JS, Hacker NF: Epithelial Ovarian Cancer. In: Practical
Gynecologic Oncology. Philadelphia: Lippincott Williams and
recurrence with favorable prognostic factors. Wilkins 2000; 500.
20. Griffiths CT: Surgical resection of tumor bulk in the primary
CONCLUSION treatment of ovarian carcinoma. Natl Cancer Inst Monogr 1975;
42:101–104.
HIPEC delivered at the time of surgery for ovarian cancer has 21. Eisenkop SM, Friedman RL, Wang HJ: Complete cytoreductive
definite potential. The growing literature documents its relative surgery is feasible and maximizes survival in patients with
tolerability and supports the continuation of further research in EOC advanced epithelial ovarian cancer: a prospective study. Gynecol
where outcomes remain so poor with current therapy. Oncol 1998;69:103–108.
22. Hoskins WJ, Bundy BN, Thigpen JT, et al.: The influence of
cytoreductive surgery on recurrence-free interval and survival in
REFERENCES small-volume stage III epithelial ovarian cancer: A Gynecologic
Oncology Group study. Gynecol Oncol 1992;47:159–166.
1. Parkin DM, Bray F, Ferlay JPP: Global cancer statistics. CA 23. Guidozzi F, Ball JH: Extensive primary cytoreductive surgery for
Cancer J Clin 2005;55:74–108. advanced epithelial ovarian cancer. Gynecol Oncol 1994;53:326–
2. Phillips R, Ball C, Sackett D: Oxford-Center for Evidence Based 330.
Medicine http://www. cebm.net/levels_of_evidence.asp [accessed 24. Bristow RE, Tomacruz RS, Armstrong DK, et al.: Survival effect
April 6, 2008]. 2001. of maximal cytoreductive surgery for advanced ovarian carci-
3. Casali P, Licitra L, Tondini C, et al.: START: A European state-of- noma during the platinum era: a meta-analysis. J Clin Oncol 2002;
the-art on-line instrument for clinical oncologists. Ann Oncol 20:1248–1259.
1999;10:769–773. 25. Hoskins WJ, McGuire WP, Brady MF, et al.: The effect of
4. Dedrick RI, Myers CE, Bungay PM, et al.: Pharmacokinetic diameter of largest residual disease on survival after primary
rationale for peritoneal drug administration in the treatment of cytoreductive surgery in patients with suboptimal residual
ovarian cancer. Cancer Treat Rep 1978;62:1–11. epithelial ovarian carcinoma. Am J Obstet Gynecol 1994;170:
5. Markman M: Intraperitoneal chemotherapy in the management of 974–979.
malignant disease. Expert Rev Anticancer Ther 2001;1:142– 26. Eisenkop SM, Spirtos NM: What are the current surgical
148. objectives, strategies, and technical capabilities of gynecologic
6. Giovanella BC, Stehlin JS Jr, Morgan AC: Selective lethal effect oncologists treating advanced epithelial ovarian cancer? Gynecol
of supranormal temperatures on human neoplastic cells. Cancer Oncol 2001;82:489–497.
Res 1976;36:3944–3950. 27. Los G, Verdegaal EM, Mutsaers PH, et al.: Penetration of
7. Hahn GM: Potential for therapy of drugs and hyperthermia. carboplatin and cisplatin into rat peritoneal tumor nodules after
Cancer Res 1979;39:2264–2268. intraperitoneal chemotherapy. Cancer Chemother Pharmacol
8. Alberts DS, Peng YM, Chen HS, et al.: Therapeutic synergism 1991;28:159–165.
of hyperthermia-cis-platinum in a mouse tumor model. J Natl 28. Alberts DS, Liu PY, Hannigan EV, et al.: Intraperitoneal cisplatin
Cancer Inst 1980;65:455–461. plus intravenous cyclophosphamide versus intravenous cisplatin
9. Meyn RE, Corry PM, Fletcher SE, et al.: Thermal enhancement of plus intravenous cyclophosphamide for stage III ovarian cancer.
DNA damage in mammalian cells treated with cis-diamminedi- N Engl J Med 1996;335:1950–1955. [see comment].
chloroplatinum(II). Cancer Res 1980;40:1136–1139. 29. Deraco M, Raspagliesi F, Kusamura S: Management of peritoneal
10. Los G, van Vugt MJ, Pinedo HM: Response of peritoneal solid surface component of ovarian cancer. Surg Oncol Clin N Am
tumours after intraperitoneal chemohyperthermia treatment with 2003;12:561–583.
cisplatin or carboplatin. Br J Cancer 1994;69:235–241. 30. FIGO. Annual report on the results of treatment in gynaecological
11. Mohamed F, Marchettini P, Stuart OA, et al.: Thermal enhance- cancer. J Epidemiol Biostat 1998;3:75–101.
ment of new chemotherapeutic agents at moderate hyperthermia. 31. Forstner R, Hricak H, Occhipinti KA, et al.: Ovarian cancer:
Ann Surg Oncol 2003;10:463–468. staging with CT and MR imaging. Radiology 1995;197:619–626.
12. van de Vaart PJ, van der Vange N, Zoetmulder FA, et al.: 32. Meyer JI, Kennedy AW, Friedman R, et al.: Ovarian carcinoma:
Intraperitoneal cisplatin with regional hyperthermia in advanced value of CT in predicting success of debulking surgery. AJR Am J
ovarian cancer: pharmacokinetics and cisplatin-DNA adduct Roentgenol 1995;165:875–878.

Journal of Surgical Oncology


HIPEC With and Without CRS in Ovarian Cancer 289
33. Bristow RE, Duska LR, Lambrou NC, et al.: A model for Database Syst Rev 2006;1:DOI:10.1002/14651858.CD005340.
predicting surgical outcome in patients with advanced ovarian pub2.
carcinoma using computed tomography. Cancer 2000;89:1532– 50. Steller MA, Egorin MJ, Trimble EL, et al.: A pilot phase I trial of
1540. continuous hyperthermic peritoneal perfusion with high-dose
34. Dowdy SC, Mullany SA, Brandt KR, et al.: The utility of carboplatin as primary treatment of patients with small-volume
computed tomography scans in predicting suboptimal cytoreduc- residual ovarian cancer. Cancer Chemother Pharmacol 1999;43:
tive surgery in women with advanced ovarian carcinoma. Cancer 106–114. [erratum appears in Cancer Chemother Pharmacol
2004;101:346–352. 1999;44(1):90].
35. Qayyum A, Coakley FV, Westphalen AC, et al.: Role of CT and 51. Ajisaka H, Yonemura Y, Bando E, et al.: Long-term survival of a
MR imaging in predicting optimal cytoreduction of newly patient with primary papillary serous carcinoma of the peri-
diagnosed primary epithelial ovarian cancer. Gynecol Oncol toneum treated by subtotal peritonectomy plus intraoperative
2005;96:301–306. chemohyperthermia. Hepato-Gastroenterology 2002;49:1027–
36. Axtell AE, Lee MH, Bristow RE, et al.: Multi-institutional 1029.
reciprocal validation study of computed tomography predictors of 52. Look M, Chang D, Sugarbaker PH: Long-term results of
suboptimal primary cytoreduction in patients with advanced cytoreductive surgery for advanced and recurrent epithelial
ovarian cancer. J Clin Oncol 2007;25:384–389. ovarian cancers and papillary serous carcinoma of the perito-
37. Kurtz AB, Tsimikas JV, Tempany CM, et al.: Diagnosis and neum. Int J Gynecol Cancer 2004;14:35–41.
staging of ovarian cancer: comparative values of Doppler and 53. Piso P, Dahlke M-H, Loss M, et al.: Cytoreductive surgery and
conventional US, CT, and MR imaging correlated with surgery hyperthermic intraperitoneal chemotherapy in peritoneal carci-
and histopathologic analysis—report of the Radiology Diagnostic nomatosis from ovarian cancer. World J Surg Oncol 2004;2:21–
Oncology Group. Radiology 1999;212:19–27. 27.
38. Turlakow A, Yeung HW, Salmon AS, et al.: Peritoneal 54. Yoshida Y, Sasaki H, Kurokawa T, et al.: Efficacy of intra-
carcinomatosis: role of (18)F-FDG PET. J Nucl Med 2003;44: peritoneal continuous hyperthermic chemotherapy as consolida-
1407–1412. tion therapy in patients with advanced epithelial ovarian cancer: a
39. Eisenkop SM, Spirtos NM, Lin WC: ‘‘Optimal’’ cytoreduction for long-term follow-up. Oncol Rep 2005;13:121–125.
advanced epithelial ovarian cancer: a commentary. Gynecol 55. Rufian S, Munoz-Casares FC, Briceno J, et al.: Radical surgery-
Oncol 2006;103:329–335. peritonectomy and intraoperative intraperitoneal chemotherapy
40. Chi DS, Venkatraman E, Masson V, et al.: The ability of for the treatment of peritoneal carcinomatosis in recurrent or
preoperative serum CA-125 to predict optimal primary tumor primary ovarian cancer. J Surg Oncol 2006;94:316–324.
cytoreduction in stage III epithelial ovarian carcinoma. Gynecol 56. Cotte E, Glehen O, Mohamed F, et al.: Cytoreductive surgery and
Oncol 2000;77:227–231. intraperitoneal chemo-hyperthermia for chemo-resistant and
41. Gemer O, Lurian M, Gdalevich M, et al.: A multicenter study of recurrent advanced epithelial ovarian cancer: prospective study
CA 125 level as a predictor of non-optimal primary cytoreduction of 81 patients. World J Surg 2007;31:1813–1820. [see comment].
of advanced epithelial ovarian cancer. Eur J Surg Oncol 2005;31: 57. Loggie BW, Sterchi JM, Rogers AT, et al.: Intraperitoneal
1006–1010. hyperthermic chemotherapy for advanced gastrointestinal and
42. Memarzadeh S, Lee SB, Berek JS, et al.: CA125 levels are a weak ovarian cancers. Reg Cancer Treat 1994;2:78–81.
predictor of optimal cytoreductive surgery in patients with 58. van der Vange N, van Goethem AR, Zoetmulder FA, et al.:
advanced epithelial ovarian cancer. Int J Gynecol Cancer 2003; Extensive cytoreductive surgery combined with intra-operative
13:120–124. intraperitoneal perfusion with cisplatin under hyperthermic
43. Cannistra SA: Cancer of the ovary. N Engl J Med 1993;329: conditions (OVHIPEC) in patients with recurrent ovarian cancer:
1550–1559. a feasibility pilot. Eur J Surg Oncol 2000;26:663–668.
44. McGuire WP, Hoskins WJ, Brady MF, et al.: Cyclophosphamide 59. Cavaliere F, Perri P, Di Filippo F, et al.: Treatment of peritoneal
and cisplatin versus paclitaxel and cisplatin: a phase III carcinomatosis with intent to cure. J Surg Oncol 2000;74:41–
randomized trial in patients with suboptimal stage III/IV ovarian 44.
cancer (from the Gynecologic Oncology Group). Semin Oncol 60. Deraco M, Rossi CR, Pennacchioli E, et al.: Cytoreductive
1996;23:40–47. surgery followed by intraperitoneal hyperthermic perfusion in the
45. Muggia FM, Braly PS, Brady MF, et al.: Phase III randomized treatment of recurrent epithelial ovarian cancer: a phase II clinical
study of cisplatin versus paclitaxel versus cisplatin and paclitaxel study. Tumori 2001;87:120–126.
in patients with suboptimal stage III or IV ovarian cancer: A 61. Panteix G, Beaujard A, Garbit F, et al.: Population pharmacoki-
gynecologic oncology group study. J Clin Oncol 2000;18:106– netics of cisplatin in patients with advanced ovarian cancer during
115. [see comment]. intraperitoneal hyperthermia chemotherapy. Anticancer Res 2002;
46. Ozols RF, Bundy BN, Greer BE, et al.: Phase III trial of 22:1329–1336.
carboplatin and paclitaxel compared with cisplatin and paclitaxel 62. de Bree E, Romanos J, Michalakis J, et al.: Intraoperative
in patients with optimally resected stage III ovarian cancer: a hyperthermic intraperitoneal chemotherapy with docetaxel as
Gynecologic Oncology Group study. J Clin Oncol 2003;21:3194– second-line treatment for peritoneal carcinomatosis of gynaeco-
3200. [see comment]. logical origin. Anticancer Res 2003;23:3019–3027.
47. Alberts DS, Green S, Hannigan EV, et al.: Improved therapeutic 63. Zanon C, Clara R, Chiappino I, et al.: Cytoreductive surgery and
index of carboplatin plus cyclophosphamide versus cisplatin plus intraperitoneal chemohyperthermia for recurrent peritoneal car-
cyclophosphamide: final report by the Southwest Oncology cinomatosis from ovarian cancer. World J Surg 2004;28:1040–
Group of a phase III randomized trial in stages III and IV ovarian 1045.
cancer. J Clin Oncol 1992;10:706–717. [see comment; erratum 64. Ryu KS, Kim JH, Ko HS, et al.: Effects of intraperitoneal
appears in J Clin Oncol 1992 Sep;10(9):1505]. hyperthermic chemotherapy in ovarian cancer. Gynecol Oncol
48. Markman M, Bundy BN, Alberts DS, et al.: Phase III trial of 2004;94:325–332.
standard-dose intravenous cisplatin plus paclitaxel versus mod- 65. Gori J, Castano R, Toziano M, et al.: Intraperitoneal hyperthermic
erately high-dose carboplatin followed by intravenous paclitaxel chemotherapy in ovarian cancer. Int J Gynecol Cancer 2005;15:
and intraperitoneal cisplatin in small-volume stage III ovarian 233–239.
carcinoma: an intergroup study of the Gynecologic Oncology 66. Reichman TW, Cracchiolo B, Sama J, et al.: Cytoreductive
Group, Southwestern Oncology Group, and Eastern Cooperative surgery and intraoperative hyperthermic chemoperfusion for
Oncology Group. J Clin Oncol 2001;19:1001–1007. [see com- advanced ovarian carcinoma. J Surg Oncol 2005;90:51–56.
ment]. 67. Raspagliesi F, Kusamura S, Campos Torres JC, et al.: Cytor-
49. Jaaback K, Johnson N: Intraperitoneal chemotherapy for the eduction combined with intraperitoneal hyperthermic perfusion
initial management of primary epithelial ovarian cancer. Cochr chemotherapy in advanced/recurrent ovarian cancer patients: The

Journal of Surgical Oncology


290 Helm et al.
experience of National Cancer Institute of Milan. Eur J Surg advanced ovarian cancer with pathological complete remission.
Oncol 2006;32:671–675. Gynecol Oncol 2003;91:341–345.
68. Helm CW, Randall-Whitis L, Martin RS III, et al.: Hyperthermic 79. Podratz KC, Schray MF, Wieand HS: Evaluation of treatment and
intraperitoneal chemotherapy in conjunction with surgery for the survival after positive second-look laparotomy. Gynecol Oncol
treatment of recurrent ovarian carcinoma. Gynecol Oncol 2007; 1988;31:9–24.
105:90–96. 80. Hoskins WJ, Rubin SC, Dulaney E, et al.: Influence of secondary
69. Surwit E, Childers J, Atlas I, et al.: Neoadjuvant chemotherapy cytoreduction at the time of second-look laparotomy on the
for advanced ovarian cancer. Int J Gynecol Cancer 1996;6:356– survival of patients with epithelial ovarian carcinoma. Gynecol
361. Oncol 1989;34:365–371.
70. Huober J, Meyer A, Wagner U, et al.: The role of neoadjuvant 81. Williams L, Brunetto VL, Yordan E, et al.: Secondary cyto-
chemotherapy and interval laparotomy in advanced ovarian reductive surgery at second-look laparotomy in advanced ovarian
cancer. J Cancer Res Clin Oncol 2002;128:153–160. cancer: A Gynecologic Oncology Group Study. Gynecol Oncol
71. Spirtos NM, Pisani AL, Chen ND, et al.: Commenting on ‘‘Future 1997;66:171–178. [see comment].
directions in the surgical management of ovarian cancer’’. 82. Obermair A, Sevelda P: Impact of second look laparotomy
Gynecol Oncol 2004;94:236–238. and secondary cytoreductive surgery at second-look laparotomy
72. Bristow RE, Eisenhauer EL, Santillan A, et al.: Delaying the in ovarian cancer patients. Acta Obstet Gynecol Scand 2001;80:
primary surgical effort for advanced ovarian cancer: A systematic 432–436.
review of neoadjuvant chemotherapy and interval cytoreduction. 83. Rose PG, Nerenstone S, Brady MF, et al.: Secondary surgical
Gynecol Oncol 2007;104:480–490. cytoreduction for advanced ovarian carcinoma. N Engl J Med
73. Rubin SC, Randall TC, Armstrong KA, et al.: Ten-year follow-up 2004;351:2489–2497. [see comment].
of ovarian cancer patients after second-look laparotomy with 84. Rubin SC, Lewis JJ: Second-look surgery in ovarian carcinoma.
negative findings. Obstet Gynecol 1999;93:21–24. Crit Rev Oncol-Hematol 1988;8:75–91.
74. Markman M, Liu PY, Wilczynski S, et al.: Phase III randomized 85. Chang E, Alexander HR, Libutti SK, et al.: Laparoscopic
trial of 12 versus 3 months of maintenance paclitaxel in patients continuous hyperthermic peritoneal perfusion. J Am Coll Surg
with advanced ovarian cancer after complete response to platinum 2001;193:225–229.
and paclitaxel-based chemotherapy: A Southwest Oncology 86. Gore ME, Fryatt I, Wiltshaw E, et al.: Cisplatin/carboplatin
Group and Gynecologic Oncology Group trial. J Clin Oncol cross-resistance in ovarian cancer. Br J Cancer 1989;60:767–769.
2003;21:2460–2465. [see comment]. 87. Markman M, Rothman R, Hakes T, et al.: Second-line platinum
75. Piccart MJ, Floquet A, Scarfone G, et al.: Intraperitoneal cisplatin therapy in patients with ovarian cancer previously treated with
versus no further treatment: 8-year results of EORTC 55875, a cisplatin. J Clin Oncol 1991;9:389–393.
randomized phase III study in ovarian cancer patients with a 88. Munkarah AR, Coleman RL: Critical evaluation of secondary
pathologically complete remission after platinum-based intra- cytoreduction in recurrent ovarian cancer. Gynecol Oncol 2004;
venous chemotherapy. Int J Gynecol Cancer 2003;2:196–203. 95:273–280.
76. Barakat RR, Sabbatini P, Bhaskaran D, et al.: Intraperitoneal 89. Chi DS, McCaughty K, Diaz JP, et al.: Guidelines and selection
chemotherapy for ovarian carcinoma: results of long-term follow- criteria for secondary cytoreductive surgery in patients with
up. J Clin Oncol 2002;20:694–698. recurrent, platinum-sensitive epithelial ovarian carcinoma. Can-
77. Barakat RR, Almadrones L, Venkatraman ES, et al.: A phase II cer 2006;106:1933–1939.
trial of intraperitoneal cisplatin and etoposide as consolidation 90. Salom E, Almeida Z, Mirhashemi R: Management of recurrent
therapy in patients with Stage II–IV epithelial ovarian cancer ovarian cancer: evidence-based decisions. Curr Opin Oncol 2002;
following negative surgical assessment. Gynecol Oncol 1998;69: 14:519–527.
17–22. 91. Lentz SS, Miller BE, Kucera GL: Intraperitoneal hyperthermic
78. Tournigand C, Louvet C, Molitor JL, et al.: Long-term survival chemotherapy using carboplatin: A phase I analysis in ovarian
with consolidation intraperitoneal chemotherapy for patients with carcinoma. Gynecol Oncol 2007;106:207–210.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:291–294

The Consensus Statement on the Locoregional Treatment


of Abdominal Sarcomatosis

CARLO RICCARDO ROSSI, MD,1 PAOLO CASALI, MD,2 SHIGEKI KUSAMURA, MD, PhD,
3

DARIO BARATTI, MD,3 AND MARCELLO DERACO, MD3*


1
Sarcoma and Melanoma Unit, Clinica Chirurgica II, University of Padova Italy
2
Department of Medical Oncology, National Cancer Institute of Milan, Italy
3
Department of Surgery, National Cancer Institute of Milan, Italy

Abdominal sarcomatosis (AS) is a rare condition characterized by soft tissue sarcoma spreading throughout the abdomen, in the absence of extra-
abdominal dissemination. Retroperitoneal sarcomas, pelvic sarcomas, particularly uterine leiomyosarcoma, and gastrointestinal stromal tumors
(GISTs) most frequently give rise to AS. Systemic chemotherapy is the standard of care for AS from non-GIST sarcomas, but with an essentially
palliative aim and major limitations. Innovative targeted therapies has deeply affected the natural history of GIST, at least in prolonging
significantly survival in responsive patients. In this context, the notion that abdominal spread in the lack of extra-peritoneal lesions may typically
occur in a number of patients, along with the dismal prognosis generally carried by AS, has prompted a few centers to perform cytoreductive
surgery and perioperative intraperitoneal chemotherapy. To date, the rarity of these presentations makes it difficult to evaluate the clinical results
and the role of combined local-regional treatment is still a matter of debate. This article presents the results of a group of experts from around the
World trying to achieve a consensus statement in AS comprehensive management. A questionnaire was placed on the website of the 5th
International Workshop on Peritoneal Surface Malignancy and the experts voted via internet.
J. Surg. Oncol. 2008;98:291–294. ß 2008 Wiley-Liss, Inc.

KEY WORDS: consensus; local regional therapy; abdominal sarcomatosis

INTRODUCTION therapies by definition. At the moment, chemotherapy of adult soft


tissue sarcomas is based on anthracyclines and Ifosfamide, given either
Abdominal sarcomatosis (AS) may be defined as the spread of a soft alone or in combination (Type 1 evidence). The response rate is in
tissue sarcoma throughout the abdomen, in the absence of any extra- the 20–40% range, and median survival is in the 1-year range.
abdominal dissemination, or at least other sites of disease of major Gemcitabine plus/minus Docetaxel may exert a palliative effect
clinical concern. This gives rise to presentations in which the especially in leiomyosarcoma (Type 3 evidence). The new marine
abdominal spread of disease is the dominating clinical factor, thus agent Trabectedin (ET-743) is active in liposarcoma and leiomyosar-
the main therapeutic challenge [1]. The perspective may be palliative coma, that is, the two main retroperitoneal mesenchymal entities, with
or curative, the former situation being more frequent than latter. an exceedingly high activity in myxoid liposarcoma, which however is
Retroperitoneal soft tissue sarcomas are the most frequent sarcomas rare in the retroperitoneum (but an abdominal widespread extent of
to give rise to disseminated abdominal disease [2]. Indeed, the local disease, even in the lack of lesions elsewhere, may occur following a
regional risk of a retroperitoneal sarcoma may well be in the 70% range primary tumor in a limb; Type 3 evidence). Imatinib is exceedingly
in the long term. Liposarcomas are especially at risk, given their effective in GIST, with a progression-free interval and an overall
inherent anatomic characteristics (average tumor size, multinodularity survival, respectively, in the 2-year and 5-year range if the disease is
or even multifocality, etc.) and their tendency to remain confined to metastatic. Surgery of residual disease is often carried out following
the abdomen in many cases throughout their natural history. Even optimal response of GIST to Imatinib, though, possibly, peritoneal
pelvic tumors may result in peritoneal sarcomatosis, such as a uterine lesions may benefit less than liver metastases from such an approach,
leiomyosarcoma relapsing with scattered tumor masses throughout the which in any case remains investigational (Type R basis evidence).
abdomen. Gastrointestinal stromal tumors (GISTs) essentially meta- Medical therapy is active, but generally of limited prognostic meaning,
stasize to two sites, peritoneum and the liver, the former in the lack of in desmoplastic small round cell abdominal tumor. Therefore,
the latter in a number of patients. Other rare mesenchymal entities may medical therapies have some activity against peritoneal sarcomatosis,
metastasize throughout the abdomen, such as the desmoplastic small but always with an essentially palliative aim, major limitations,
round cell abdominal tumor of young adults or adolescents, typically and variability across tumor entities. A huge distinction can be made
spreading with multiple tumor nodules over the peritoneal surface. All between GIST and other adult soft tissue sarcomas, because the natural
these tumors are rare. Their abdominal spread in the lack of lesions
outside is even less frequent, though it may typically occur, indeed, in a
number of patients. Finally, the prognosis of AS is generally dismal, The authors have no financial interest related to the contents of this article to
although liposarcomas may run a slow course, and GIST are now disclose.
amenable to effective medical therapies which at least prolong *Correspondence to: Marcello Deraco, MD, Istituto Nazionale Tumori
significantly survival. Milano, Via Venezian 1, 20133 Milano, Italy. Fax: þ39-02-23902404.
E-mail: marcello.deraco@istitutotumori.mi.it
Received 25 March 2008; Accepted 27 March 2008
Medical Therapies
DOI 10.1002/jso.21067
Disseminated disease spreading throughout the abdomen Published online in Wiley InterScience
makes the patient metastatic, and thus a candidate for medical (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


292 Rossi et al.
history of GIST has been deeply affected by the medical therapy, while Treatments in Oncology (SITILO) [14]. In a study, 60 patients affected
this is still not the case for other sarcomas. Integrated approaches are by advanced (multifocal primary or locally recurrent) intraabdominal
often resorted to, often using surgery in the case of tumor response visceral or retroperitoneal soft tissue sarcomas underwent optimal
to medical therapies, and/or medical therapy in the case surgery is cytoreductive surgery (tumor remnants diameter <3 mm) followed by
unfeasible. Surgery is obviously the treatment mainstay in most cases HIPEC with doxorubicin and cisplatin. The postoperative complication
with a localized disease stage, but the surgeon is well aware of the rate was 23%, locoregional toxicity 15%, and overall morbidity 33%.
risk of sarcomatosis, which may be inherent to the natural history The median time to local disease progression and the median
of disease, and also a consequence of complications occurring overall survival were 22 and 34 months, respectively. A multivariate
during surgery (tumor rupture, etc.). Therefore, the value of adjuvant analysis showed a significant advantage for patients who had
procedures has been long explored. Unfortunately, as of today, no undergone complete versus near-complete cytoreduction. Moreover,
adjuvant medical therapy has been proved of value in these diseases the importance of inherent tumor aggressiveness was underscored by
[3]. Prospective trials on adjuvant Imatinib are ongoing in inter- the different prognosis of high versus low grade malignancies.
mediate-high risk GIST patients [4,5]. On the other hand, patients with Although in this group of patients an improved median overall survival
adult soft tissue sarcomas are often offered adjuvant chemotherapy in was observed in comparison to other case series (34 vs. 20–29 months),
several institutions across the world when the disease is high-risk, as is these results should be interpreted by considering: (1) heterogeneity of
the case by definition in most cases of high-grade retroperitoneal, or histological types across the case series; (2) different inclusion criteria;
pelvic, sarcomas. However, we lack any formal proof of efficacy (3) different treatments (HIPEC end EPIC). One possible explanation
thereof. Local regional drug delivery systems can yield high for these poor results in PS might lie in the natural history of sarcomas,
intraperitoneal concentrations of chemotherapeutic agents, while which tend to spread across anatomical structures as nerves and
keeping low their systemic levels. As the intraperitoneal route cannot vessels, which in the abdomen are retroperitoneal, and are therefore not
guarantee an adequate drug penetration into tumor deposits larger than accessible to peritoneal bathing. It may be concluded that, at present,
1–3 mm, cytoreductive surgery is the main prerequisite for any there is no sufficient evidence supporting the treatment of patients with
intraperitoneal chemotherapy. In addition, by entrapping cancer cells peritoneal sarcomatosis with HIPEC or EPIC, even if the former may
and preventing them from being reached by chemotherapeutic agents be slightly more effective (Type 3 evidence). This treatment modality
[6], early postoperative adhesions may limit the effect of intraper- is highly demanding both for patients and health resources, and thus
itoneal therapies when given postoperatively. This has led to place should be all the more considered investigational for patients with
these therapies immediately after surgery (early postoperative multiple peritoneum implants (true peritoneal sarcomatosis).
intraperitoneal chemotherapy: EPIC). Furthermore, the notion that
some antineoplastic drugs act synergistically with others, as well as Perspectives
with heat, led some authors to propose hyperthermic intraperitoneal
chemotherapy (HIPEC) for the treatment of locally advanced The rarity of these presentations makes it difficult to undertake
intraabdominal malignancies [7,8]. So far, aggressive cytoreductive formal clinical trials. However, only the joined efforts of several
surgery combined with EPIC or HIPEC has been mainly considered for institutions might achieve the goal. At the moment, medical therapy is
peritoneal carcinomatosis from gastrointestinal cancers, with encoura- the standard of care for these patients. The histological entity dictates
ging results [9,10]. Based on the same rationale, studies have been which medical treatment to select, and/or clinical studies may be open
performed also in patients with PS. in the various presentations. Following optimal tumor response,
surgery is often resorted to in sarcomas, and may well be used also
in these patients. Its combination with HIPEC should be considered
Intraperitoneal Chemotherapy investigational in retroperitoneal and/or pelvic sarcomas. The major
benefit gained from Imatinib, and the availability of further second-line
The first published study investigating this therapeutic approach in molecularly targeted agents, in the lack of any formal proof of efficacy
patients with PS is attributed to Berthet et al. [11]. These authors of surgery for residual disease, makes it difficult, at the moment, to
combined complete gross resection through a peritonectomy procedure foresee that HIPEC may make any difference in advanced GIST
with early peritoneal chemotherapy with or without hyperthermia. patients, even when the disease is confined to the peritoneum.
Only 16 out of 43 underwent HIPEC. Overall, the 43 patients achieved A prospective study on the combination of aggressive cytoreductive
a median survival of 20 months. Only the completeness of cyto- surgery with HIPEC might be foreseen in principle, if a collaborative
reductive surgery had a significant impact on survival. Eilber et al. [12] effort can be put in place. Eligible for such a trial could be
used a similar aggressive surgical approach for recurrent abdominal patients with: (1) histologically proven multifocal peritoneal implants
sarcoma, including first recurrence with/without sarcomatosis, but of non-GIST adult soft tissue sarcoma; (2) tumor remnants <3 mm
intraperitoneal chemotherapy was delayed (1–2 weeks after complete following cytoreductive surgery; (3) absence of distant metastasis on
surgery) with the risk of postoperative adhesions preventing complete CT/MR imaging. Patients’ selection should be based on an accurate
bathing of the peritoneal cavity. The 35 patients having intraperitoneal preoperative work-up encompassing thoraco-abdominal CT scan.
disease alone showed a median survival of 24 months. The median Intraoperative staging should include a peritoneal cancer index.
survival of patients treated in these two studies does not differ from that Cytoreductive surgery should be performed according to the criteria
reported by Bilimoria, who treated 51 patients with sarcomatosis or by Sugarbaker [15]. HIPEC should be based on Doxorubucin and
GISTs at the MD Anderson Cancer Centre by means of surgery and Cisplatin, according to the results of a previously reported Phase I
conventional chemo/radiotherapy, thus obtaining a median survival of study [16,17]. In the event such a clinical trial provides interesting
around 22 months [1]. Bonvalot et al. [13] recently published a results, further investigations with randomized confirmatory studies
randomized trial on 19 patients with peritoneal sarcomatosis treated and/or on new pharmacological regimens could be pursued.
with surgery alone, versus 19 who underwent surgery followed by
intraperitoneal chemotherapy with doxorubicin and cisplatin for Results of Web Based Voting on Conflicting Points
5 days. After a median follow-up of 60 months, the local relapse-
free, metastatic relapse-free and overall survival were similar in the This section reports and comments on the results of the peritoneal
two groups. So far, the most relevant experience reported with surgery sarcomatosis consensus group, which was composed of 13 physicians
combined with HIPEC is from the Italian Society for Locoregional from around the World. A questionnaire was placed on the website of

Journal of Surgical Oncology


Consensus in Local Regional Treatment 293
the 5th International Workshop on Peritoneal Surface Malignancy Eligibility
and the group members voted via internet. Questions generally asked
for a ranking of answers on a 1–5 scale where 1 was considered Six questions were asked regarding some of the conflicting
NOT important and 5 MOST important. For the purposes of clarity indications and/or situations that would make a patient with peritoneal
in presentation of the results of the consensus, the answers 4 and 5 sarcomatosis a candidate for cytoreductive surgery and HIPEC.
have been added together as were answers 1 and 2 and the results
are expressed as percentages of the total respondents. The percentages (1) With regard to the non-GIST sarcomas, may we foresee a role for HIPEC in
were rounded up and down to the nearest whole number. the era of molecularly targeted therapies?
In some cases meaningful data could not be obtained from the YES 67%
questionnaire and this data is not reported here. NO 33%
(2) With regard to the GIST model, may we foresee a role for HIPEC in the era
of molecularly targeted therapies?
Preoperative Work-Up YES 50%
NO 50%
What investigations are optimal to define extent of disease?
(3) With regard to the GIST model, may we foresee a role for HIPEC in patient
(Tables I–III). non-responsive to targeted therapies?
YES 67%
NO 33%
(4) Referring to retroperitoneal sarcomas, pelvic sarcomas, GIST, is there any
clinical presentation in which abdominal sarcomatosis could be treated
today with HIPEC outside a clinical study? In other words, as of today,
TABLE I. Optimal Investigations to Define Extent of Disease: 1 (Not should we consider HIPEC:
Important) to 5 (Most Important) Investigational only 58%
Suitable for individual clinical use in selected patients 42%
% of respondents (5) As of today’s knowledge, which is the selective contribution of cytoreductive
surgery, antiblastic perfusion and hyperthermia to the potential efficacy of
Exam 1–2 3 4–5 HIPEC, if any, in abdominal sarcomatosis?
Only for palliation 33%
Clinical examination 0 10 90 For locoregional control 67%
CT chest 0 8 92 For improvement on survival 0%
CT abdomen/pelvis 0 0 100 (6) With regard to non-GIST sarcomas, which role for HIPEC may we foresee
MRI abdomen/pelvis 42 16 42 within combined approaches incorporating pre/postoperative chemotherapy?
PET/CT 25 50 25 At the time of primary tumor treatment 9%
Laparoscopy with biopsy 25 42 33 At the time of recurrence 73%
Ultrasound 42 50 8 Both 18%
Colonoscopy 33 42 25
OGDS 45 55 0

State of the Art of the Methodology


The following questions were presented to the voters concerning the
TABLE II. Recommended Laboratory Work-Up Prior to Surgery: 1 (Not technical aspects of the local-regional therapy.
Important) to 5 (Most Important) Sixty-two percent of the voters defined complete cytoreduction as
% of respondents
the presence of a residual disease <2.5 mm (CC-1).
Three quarters of the voters were of the opinion that there would be
Exam 1–2 3 4–5 a role of maximal palliative cytoreduction in cases not amenable to
radical surgery.
Basic metabolic panel urea, creatinine, electrolytes 9 9 82 Regarding the advisable extent of radicality the majority of the
Comprehensive metabolic panel above plus liver function 0 0 100 panel (92%) voted that a limited peritonectomy to affected area would
Complete blood count 0 0 100 be sufficient, rather than performing a complete peritoneal resection
Coagulation studies 8 0 92 even in the absence of macroscopic disease.
Urinalysis 25 17 58
Seventy-three percent of the panel was of the opinion that HIPEC
Pulmonary function testing 17 41 42
CA125 50 33 17
would not be performed in inoperable cases.
CEA 75 17 8 Hundred percent of the panel voted that the combination of
CA 19-9 58 17 25 chemotherapies would be better than single agent to use in HIPEC. The
CA 15.3 56 17 17 preferred combination was cisplatin þ doxorubicin.

Follow-Up
Ninety-one percent of the panel voted CT scan as the preferred
imaging modality for the follow-up.
In case of asymptomatic recurrence three quarters of the voters favored
TABLE III. Do You Think That There Should be an Effort to Document a
the surgical approach and 92% favored the systemic chemotherapy.
Peritoneal Cancer Index on the Pre-Operative CT Scan? (Just One
Alternative Allowed)
Definitely 25% FUTURE INVESTIGATIONS
Not sure or does not matter 67%
Ninety-two percent of the panel was of the opinion that a
No, it is too cumbersome 8%
prospective multicentric randomized trial testing the efficacy of

Journal of Surgical Oncology


294 Rossi et al.
CRS þ HIPEC in the treatment of peritoneal sarcomatosis would be of prospective, randomized, clinical trial. Arch Surg 1993;128:
concern. The same rate of voters agreed that the patients should be 402–410.
randomized to CRS þ HIPEC versus CRS alone. 4. Plaat BE, Hollema H, Molenaar WM, et al.: Soft tissue
leiomyosarcomas and malignant gastrointestinal stromal tumors:
Differences in clinical outcome and expression of multidrug
DISCUSSION OF RESULTS resistance proteins. J Clin Oncol 2000;18:3211–3220.
5. Weichselbaum RR, Beckett MA, Hallahan DE, et al.: Molecular
The great majority of the experts, accounting for more than 90% of targets to overcome radioresistance. Semin Oncol 1992;19:14–20.
voters, agreed that physical examination and CT-scan of the chest 6. Markman M: Intraperitoneal drug delivery of antineoplastics.
abdomen and pelvis are of the utmost importance to define the extent of Drugs 2001;61:1057–1065.
peritoneal involvement by sarcomatous tumor. Furthermore, there was a 7. Sugarbaker P: Peritoneal carcinomatosis: Principles of manage-
ment. Boston: Kluwer; 1996.
general agreement that additional diagnostic tools, such as MRI
8. Spratt JS, Adcock RA, Muskovin M, et al.: Clinical delivery
imaging, PET-scan, laparoscopy and endoscopy play a limited role in system for intraperitoneal hyperthermic chemotherapy. Cancer
the preoperative work-up of potential candidates to local-regional Res 1980;40:256–260.
treatment. However, it is surprising that only 25% stated that there 9. Verwaal VJ, Ruth SV, De Bree E, et al.: Randomized trial of
should be an effort to document a Peritoneal Cancer Index on the pre- cytoreduction and hypertermic intraperitoneal chemotherapy
operative CT scan, while for 67% of voters such an issue is not relevant. versus systemic chemotherapy and palliative surgery in patients
Concerning the local-regional treatment of abdominal sarcoma, the with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol
most controversial issue is presently represented by the availability of 2003;21:3737–3743.
innovative targeted therapies which has shown favorable effectiveness 10. Elias D, Blot F, El Otmany A, et al.: Curative treatment of
peritoneal carcinomatosis arising from colorectal cancer by
at least in prolonging significantly survival for patients affected by
complete resection and intraperitoneal chemotherapy. Cancer
GIST [18]. This, along with the lack of formal proof of the efficacy of 2001;92:1.
the combined modality approach in this clinical setting, led the 11. Berthet B, Sugarbaker TA, Chang D, et al.: Quantitative
majority of the experts to conclude that today HIPEC should be methodologies for selection of patients with recurrent abdomi-
considered a merely investigational option to be performed in case of nopelvic sarcoma for treatment. Eur J Cancer 1999;35:413–419.
recurrent disease presentation with the aim to achieve better local 12. Eilber FC, Rosen G, Forscher C, et al.: Surgical resection and
control, but with no impact of long term survival. Accordingly, when intraperitoneal chemotherapy for recurrent abdominal sacrcoma.
the role of surgical cytoreduction and HIPEC in the management of the Ann Surg Oncol 1999;6:645–650.
different sub-types of sarcoma was discussed, no agreement was 13. Bonvalot S, Cavalcanti A, Le Péchoux C, et al.: Randomized trial
of cytoreduction followed by intraperitoneal chemotherapy versus
reached on the use of the intraperitoneal perioperative chemotherapy in
cytoreduction alone in patients with peritoneal sarcomatosis. Eur
patients affected by GIST responsive to the molecular therapy. By J Surg Oncol 2005;31:917–923.
contrast, two-third of voters favored the hypothesis of a combined 14. Rossi CR, Deraco M, De Simone M, et al.: Hyperthermic
treatment approach to treat non-GIST sarcomas or GIST not responsive intraperitoneal intraoperative chemotherapy after cytoreductive
to the molecular therapy. surgery for the treatment of abdominal sarcomatosis: Clinical
The rarity of abdominal sarcomas and the controversies surrounding outcome and prognostic factors in 60 consecutive patients. Cancer
their local-regional aggressive management have resulted in a limited 2004;100:1943–1950.
number of patients treated by a multimodality approach being 15. Sugarbaker PH: Peritonectomy procedure. Ann Surg 1995;221:
reported in the literature [12–14,19–24]. Consequently, many technical 29–42.
16. Rossi CR, Foletto M, Mocellin S, et al.: Hyperthermic intra-
aspects of the comprehensive procedure still have to be optimized.
operative intraperitoneal chemotherapy with cisplatin and dox-
Nevertheless, according to the base principles of peritoneal surface orubicin in patients who undergo cytoreductive surgery for
tumor management, the expert panel emphasized that maximal peritoneal carcinomatosis and sarcomatosis: Phase I study. Cancer
cytoreductive surgical efforts should be paid to reduce the volume of 2002;94:492–499.
residual disease as much as possible, or at least to diameter less than 17. Sugarbaker PH, Stuart OA: Pharmacokinetic and phase II
2.5 mm. Analogously, due to the well-known limited tumor penetration study of heated intraoperative intraperitoneal melphalan.
of locally applied chemotherapy, they agreed that HIPEC would Cancer Chemother Pharmacol 2007;59:151–155. Epub, 2006.
not be performed in patients not amenable to significant surgical 18. Eilber FC, Rosen G, Forscher C, et al.: Recurrent gastrointestinal
cytoreduction. stromal sarcomas. Surg Oncol 2000;9:71–75.
19. Eilber FC, Rosen G, Forscher C, et al.: Surgical resection and
In conclusion, treatment of peritoneal recurrence following surgical
intraperitoneal chemotherapy for recurrent abdominal sarcomas.
resection of intraabdominal sarcomas presents a significant challenge Ann Surg Oncol 1999;6:645–650.
to clinicians. Although the rarity of this disease presentation makes it 20. Yoshida Y, Inoue K, Ohsako T, et al.: Weekly paclitaxel therapy is
difficult to undertake formal clinical trials a prospective multicentric curative for patients with retroperitoneal liposarcoma. Gan To
randomized trial testing the efficacy of CRS þ HIPEC in the treatment Kagaku Ryoho 2007;34:465–467.
of peritoneal sarcomatosis was considered advisable by the majority 21. Edmonson JH, Ebbert LP, Nascimento AG, et al.: Phase II study
of the expert panel and it would ideally compare the combination of of docetaxel in advanced soft tissue sarcomas. Am J Clin Oncol
cytoreductive surgery and HIPEC with cytoreductive surgery alone. 1996;19:574–576.
22. Van Hoesel QG, Verweij J, Catimel G, et al.: Phase II study with
docetaxel (taxotere) in advanced soft tissue sarcomas of the adult.
REFERENCES EORTC Soft Tissue and Bone Sarcoma Group. Ann Oncol 1994;
5:539–542.
1. Bilimoria MM, Holtz DJ, Mirza NQ, et al.: Tumor volume as a 23. Elias D, Bonnay M, Puizillou JM, et al.: Heated intra-operative
prognostic factor for sarcomatosis. Cancer 2002;94:2441–2446. intraperitoneal oxaliplatin after complete resection of peritoneal
2. Pirayesh A, Chee Y, Helliwell T, et al.: The management of carcinomatosis: Pharmacokinetics and tissue distribution. Ann
retroperitoneal soft tissue sarcoma: A single institution experi- Oncol 2002;13:267–272.
ence with a review of the literature. Eur J Surg Oncol 2001;27: 24. Bartlett DL, Buell JF, Libutti SK, et al.: A phase I trial of
491–497. continuous hyperthermic peritoneal perfusion with tumor necrosis
3. Sindelar WF, Kinsella TJ, Chen PW, et al.: Intraoperative factor and cisplatin in the treatment of peritoneal carcinomatosis.
radiotherapy in retroperitoneal sarcomas. Final results of a Cancer 1998;83:1251–1261.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:295–299

Treatment Failure Following Complete Cytoreductive Surgery and


Perioperative Intraperitoneal Chemotherapy for Peritoneal Dissemination
From Colorectal or Appendiceal Mucinous Neoplasms

1 2
LANA BIJELIC, MD, TRISTAN D. YAN, BSc(Med), MBBS, PhD, AND PAUL H. SUGARBAKER, MD1*
1
Peritoneal Surface Malignancy Program, Washington Cancer Institute, Washington Hospital Center, Washington, District of Columbia
2
University of New South Wales, Department of Surgery, St. George Hospital, Sydney, Australia

Background: Peritonectomy combined with perioperative intraperitoneal chemotherapy is a successful treatment option for patients with
peritoneal dissemination of appendiceal and colorectal malignancy. Despite its efficacy, recurrences remain a common problem.
Methods: Patients with peritoneal dissemination from appendiceal or colorectal malignancy who underwent complete cytoreduction and
perioperative intraperitoneal chemotherapy were included in this study. Data regarding recurrent disease found on abdominal exploration and/or
diagnostic studies was extracted from a prospective database and analyzed.
Results: Seventy patients with colorectal cancer carcinomatosis and 402 with appendiceal neoplasm were analyzed. Forty-nine of 70 and 111 of
402 patients developed documented recurrences. The median survival of 49 patients with colorectal cancer was 33 months while the median
survival for patients with appendiceal neoplasms was not reached. The most common type of recurrent disease was a localized intra-abdominal
recurrence for both appendiceal and colon cancer patients. Patients who underwent second surgery for recurrent disease had an improved survival.
Conclusion: Additional treatments should be strongly considered in patients who fail an initial cytoreduction combined with perioperative
intraperitoneal chemotherapy. This resulted in 5-year survival in 17% of colorectal patients and 70% of the appendiceal mucinous neoplasm
patients.
J. Surg. Oncol. 2008;98:295–299. ß 2008 Wiley-Liss, Inc.

KEY WORDS: cytoreductive surgery; intraperitoneal chemotherapy; appendiceal neoplasms; pseudomyxoma peritonei;
colorectal cancer; recurrence; survival

INTRODUCTION Eligibility Criteria


The management of peritoneal carcinomatosis has progressed over Patient characteristics, pathologic features and treatment-related
the last two decades. The long-term survival of patients with data were obtained from a prospective database. The inclusion criteria
disseminated mucinous appendiceal neoplasms has led the way to of this study consisted of patients who had a diagnosis of appendiceal
defining this success [1]. Another rare disease, peritoneal mesothe- mucinous neoplasm or colorectal cancer with peritoneal dissemination
lioma, now shows long-term survival in a group of patients who in the and underwent a complete cytoreduction. All sites and volumes
past all rapidly died of disease progression within the abdomen and of residual disease following the CRS were prospectively recorded
pelvis [2]. Currently, cytoreductive surgery (CRS) and perioperative using the Completeness of Cytoreduction Score (CCR Score). A
intraperitoneal chemotherapy (PIC) is being explored as a treatment complete cytoreduction was defined as CCR-0 or CCR-1 (residual
option for colorectal cancer [3,4]. tumor < 2.5 mm). These eligibility requirements were designed to
However, even with these improvements in the management of assess patterns of treatment failure and progression-free survival after
gastrointestinal malignancies, most patients who are currently treated go optimal combined treatment. In this failure analysis, all analyzed
on to progress on peritoneal surfaces. In all of these peritoneal surface patients had documented recurrent disease.
malignancies treatment failure is most commonly seen on the peritoneal
surface and at the site of the surgeons’ best efforts. This present study is Preoperative Management
designed to clarify the sites of treatment failure for appendiceal and
colorectal carcinomatosis. It is an attempt to improve and if possible All patients underwent extensive preoperative investigations which
eliminate peritoneal surface progression of disease in these two groups included physical examination, abdominal, pelvic and chest computed
of patients. It is hoped that an improved understanding of treatment tomography (CT) scans to assess the extent of the disease involved.
failure following CRS and PIC will allow new and more beneficial CT scans were performed following the administration of oral and
management strategies to be initiated. Our satisfaction with these intravenous contrast media. Extent of prior surgery was determined
treatments should not occur until the peritoneal surface component of
appendiceal and colorectal cancers have been eliminated.
*Correspondence to: Dr. Paul H. Sugarbaker, MD, 106 Irving Street, NW,
Suite 3900N, Washington, DC 20010; Fax: 202-877-8602.
METHODS E-mail: paul.sugarbaker@medstar.net
Between June 1981 and July 2006, CRS combined with Received 3 April 2008; Accepted 15 April 2008
perioperative intraperitoneal chemotherapy was used to treat patients DOI 10.1002/jso.21084
with peritoneal carcinomatosis, by the same surgeon (P.H.S.) at the Published online in Wiley InterScience
Washington Cancer Institute, Washington, DC. (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


296 Bijelic et al.
according the number of abdominopelvic regions dissected during region, left pericolic gutter region, left inguinal region, pelvic region,
previous surgery. right inguinal region, right pericolic gutter region, proximal portion
of jejunum, distal portion of jejunum, proximal portion of ileum
Cytoreductive Surgery and distal portion of ileum. Treatment failure in the abdominal wall
incisions and/or other systemic sites found on imaging was also
The volume and extent of the tumor deposits were prospectively recorded prospectively and included in the analysis if they occurred
recorded using the Peritoneal Cancer Index (PCI), as described within 3 months of the initial site.
previously. All patients underwent CRS by the same surgical team with The statistical analyses of clinicopathologic and treatment-related
intent to remove all visible intraperitoneal tumor deposits. Periton- parameters used progression-free and overall survival as the endpoints.
ectomy procedures included total anterior parietal peritonectomy, All survival analyses were performed using Kaplan–Meier method and
omentectomy  splenectomy, right and left subphrenic peritonectomy, compared using the log-rank test. Statistical analyses were performed
pelvic peritonectomy, and lesser omentectomy with stripping of the by the intent-to-treat principle, using SPSS for Windows (Version 14.5;
omental bursa  cholecystectomy [5]. These resections were used at SPSS GmbH, Munich, Germany). A significant difference was
the anatomic sites where there was visible evidence of disease. assumed for P < 0.05.

Perioperative Intraperitoneal Chemotherapy RESULTS


For colorectal cancer patients mitomycin C was administered as a Failure Analysis of Progressive Colorectal Cancer
normothermic solution on postoperative day 1 in the first 36 patients
and as a hyperthermic solution intraoperatively in the subsequent Between June 1981 and November 2004, 70 patients with
34 patients treated after 1997. The 5-fluorouracil was administered on carcinomatosis from colorectal cancer underwent a complete cytor-
postoperative days 2–6 in the first 36 patients and on days 1–5 in the eduction. One patient suffered postoperative death and was eliminated
subsequent 34 patients. Patients with appendiceal neoplasms treated from further data analysis. The mean and median follow-up of this
between January 1994 and July 2006 with CRS and hyperthermic group of 70 patients was 40.8 and 29.5 months, respectively. Forty-nine
intraperitoneal chemotherapy (HIPEC) followed by early postopera- patients developed documented recurrences during follow-up while
tive intraperitoneal chemotherapy (EPIC) were included. The coliseum an additional six patients had inadequate documentation of their
technique was used for administration of HIPEC with mitomycin recurrences. Four of these six patients have died of disease, one is alive
C (10 mg/m2 for females and 12.5 mg/m2 for males) at approximately with disease as of last follow-up in January 2005 and one was lost to
428C in 2 L of 1.5% dextrose peritoneal dialysis solution for 90 min. follow-up. These six patients were excluded from further data analysis
During postoperative days 1–5, 5-fluorouracil (650 mg/m2) was making 49 patients with documented recurrence eligible for further
administered into the peritoneal cavity in 1 L of 1.5% dextrose study. The median time to progression defined as the time from
peritoneal dialysis solution. By the principle of intention-to-treat, all definitive cytoreduction to time of first documentation of recurrent
patients were to receive EPIC with 5-fluorouracil. However, patients disease was 9 months. The median survival for this group of patients
with postoperative bleeding, hemodynamic instability, prior extensive was 30 months and the 5-year survival was 17%. The time to
chemotherapy treatment or other complications experienced early in progression and the survival curves for patients with documented
the postoperative period were exempt from EPIC. For appendiceal recurrences is shown in Figure 1.
and colorectal cancer patients, further systemic chemotherapy was There were 12 patients who recurred within 6 months and 37 who
administered as clinically indicated. recurred after 6 months. Those who developed recurrence within
Patients having repeat CRS for recurrence did not receive HIPEC or 6 months of cytoreduction had a significantly shorter overall survival
EPIC, if gross residual disease remained after a best surgical effort at (17 months) than patients who developed recurrence more then
complete cytoreduction. 6 months after cytoreduction (36 months, P ¼ 0.001) (Fig. 2).
Twenty-six of the 49 patients (53%) had at least one reoperation after
the initial cytoreduction. Eighteen of the 26 patients had a complete
Follow-Up and Study Methods
All patients were followed prospectively until the last time of
contact or death. The follow-up review included clinical examination,
measurement of relevant tumor markers (every 3 months performed by
the treating medical oncologist) and assessment of abdominopelvic
CT scans every 6 months or when recurrence was suspected
clinically. Other diagnostic modalities or procedures were used based
on the patients’ symptoms. Colorectal cancer patients were offered
elective second-look surgery approximately 6 months following
cytoreduction unless they declined surgery or had obvious progressive
disease. Patients undergoing ostomy closure underwent full abdominal
exploration (second-look surgery) at the time of the ostomy closure.
Patients with a rise in tumor markers and no obvious metastatic disease
underwent abdominal exploration.
For colon cancer patients, recurrences found within 3 months of
the initial recurrence, either by imaging studies or by abdominal
exploration were included in the analysis. For patients with
appendiceal neoplasms, the sites of treatment failure were determined
from those patients who underwent repeat CRS and PIC and recorded
prospectively in the 13 abdominopelvic regions during repeat Fig. 1. Time to progression and overall survival for 49 patients
laparotomy: central abdomen/parietal peritoneum of anterior abdo- who failed complete cytoreduction and perioperative intraperitoneal
minal wall, perihepatic region, epigastric/lesser sac region, perisplenic chemotherapy as treatment for colorectal peritoneal carcinomatosis.

Journal of Surgical Oncology


Failures After Carcinomatosis Treatment 297
TABLE I. Anatomic Location and Character of Disease Recurrence,
Number of Patients and Median Survival for 49 Patients Who Failed
Complete Cytoreduction and Perioperative Intraperitoneal Chemotherapy
as Treatment for Colorectal Peritoneal Carcinomatosis

Median
Character of Number of survival
Location of disease recurrence disease recurrence patients (months)

Localized intra-abdominal with Localized 18 37


or without abdominal wall
Intra-abdominal and pelvic Diffuse 11 29
Isolated distant metastases Systemic 10 13
Distant metastases and other site Systemic þ other 10 30

Institute. One hundred and eleven patients (26%) after the initial
Fig. 2. Overall survival of 49 patients with documented recurrence complete cytoreduction were found to have subsequent disease
according to the time to first evidence of recurrence (P ¼ 0.001). All
progression. Of these 111 patients, 98 patients underwent second
patients failed complete cytoreduction and perioperative intraperitoneal
chemotherapy as treatment for colorectal peritoneal carcinomatosis. CRS and PIC. Seventy-six patients had complete second cytoreduction
(CCR-0/CCR-1) and 22 patients had incomplete second cytoreduction
second cytoreduction (no visible tumor or tumor nodules <2.5 mm in (CCR-2/CCR-3).
diameter). The median survival of patients who had a second operation Figure 4 demonstrates overall survival and actual progression-free
was significantly longer then that of patients who did not have a second survival of these 111 patients. It shows that median survival was not
operation (39 months vs. 20 months, P ¼ 0.0003) (Fig. 3). reached and 5-year overall survival was 74%. This was associated with
The anatomic sites of recurrence, character of recurrence and their a median actual progression-free survival of 15 months. Analysis of
associated survival are shown in Table I. Eighteen patients had prognostic factors for survival in these 11 patients showed that
localized intra-abdominal recurrence (including retroperitoneum and progression-free interval of 2 years was not associated with an
pelvis) with or without abdominal wall recurrence. The recurrence was improved survival (vs. <2 years, P ¼ 0.836) (Fig. 5). However, in
categorized as localized if three or less non-contiguous abdomino- patients who underwent a repeat CRS and PIC (n ¼ 96), the survival
pelvic regions were involved by discrete tumor masses. Ten patients results was significantly improved when compared to those who were
had diffuse peritoneal disease and ten had isolated distant metastases. not selected for repeat surgery P < 0.001) (Fig. 6).
Eleven patients had both distant metastases and another form of Table II demonstrates the distribution of treatment-failure sites and
recurrence (diffuse or localized peritoneal disease). their associated median survival. Fifty-seven patients had diffuse intra-
abdominal disease progression. Forty-one patients had focal intra-
Failure Analysis of Progressive Appendiceal Cancer abdominal disease progression. Twenty-four patients had systemic
progression, including 11 patients had progression only at a systemic
Four hundred and two patients had complete cytoreduction (CCR-0 site and 13 patients had progression both at systemic and intra-
or CCR-1) after initial CRS and PIC at the Washington Cancer abdominal sites. Two patients had abdominal wall recurrence only.

Fig. 3. Overall survival in 49 patients with documented recurrences Fig. 4. Time to progression and overall survival for 111 patients who
according to second surgery (P ¼ 0.0003). All patients failed complete failed complete cytoreduction and perioperative intraperitoneal
cytoreduction and perioperative intraperitoneal chemotherapy as chemotherapy as treatment for appendiceal neoplasms with peritoneal
treatment for colorectal peritoneal carcinomatosis. dissemination.

Journal of Surgical Oncology


298 Bijelic et al.
patients treated with cytoreductive surgery and heated intraoperative
intraperitoneal chemotherapy followed by systemic chemotherapy
compared to 12.6 months for the group treated with systemic
chemotherapy without combined treatment [4]. However, it has to be
realized that the clinical evidence on treatment failure after the
combined treatment is still limited, especially regarding peritoneal
dissemination from appendiceal neoplasms where a long-term follow-
up is needed to achieve mature results due to the biology of the disease.
Some, even though limited data on the timing and distribution of
recurrences following cytoreductive surgery and perioperative intra-
peritoneal chemotherapy for colorectal cancer is available. Glehen
et al. reported an overall incidence of recurrence of 73.3% in a multi-
institutional retrospective study of 506 patients [3]. In this study, there
were 377 patients who underwent CCR-0 or CCR-1 and had a
recurrence rate of 64.2% with 41.9% having peritoneal recurrence.
Verwaal et al. published their results of 106 patients treated
Fig. 5. Overall survival of 111 patients with documented recurrence with cytoreductive surgery, heated intraoperative intraperitoneal
according to the time to first evidence of recurrence (P ¼ 0.863). All chemotherapy followed by systemic chemotherapy showing that
patients failed complete cytoreduction and perioperative intraperito- recurrences developed in 69/106 patients (65%) [8]. The
neal chemotherapy as treatment for appendiceal neoplasms with median time to development of recurrences differed according to the
peritoneal dissemination. completeness of cytoreduction: patients with a macroscopically
complete cytoreduction had a median time to recurrence of 13.7 months
DISCUSSION while patients with a cytoreduction to <2.5 mm had a median time of
10.8 months. The majority of patients recurred in the abdomen but
The evidence that supports a combined treatment of peritoneal more precise anatomic characterization of intra-abdominal recurrences
carcinomatosis aimed at long-term survival has grown significantly in was not given.
the last two decades. A number of referral centers have been developed The present study demonstrated that after initial complete CRS and
in the USA, Europe and Australia that have reported on phase II data PIC for appendiceal neoplasm, the overall 5- and 10-year progression-
regarding the use of cytoreductive surgery and perioperative free survival rates were 70% and 67%, respectively. One hundred and
intraperitoneal chemotherapy for peritoneal carcinomatosis from eleven patients (28%) developed progressive disease and disease
appendiceal neoplasms as well as colorectal cancer [2,6]. The Fourth progression was the only independent risk factor associated with a
International Workshop on Peritoneal Surface Malignancy that was reduced overall survival. In the colorectal cancer group, 70% of
held in Spain in December 2004 determined as one of the key patients developed disease recurrence with a median progression-free
consensus points that ‘‘cytoreductive surgery combined with perio- survival of 9 months. These strikingly different results emphasize the
perative intraperitoneal chemotherapy is considered the current differences in cancer biology and aggressiveness between appendiceal
standard of care for all cases of mucinous appendiceal neoplasms and colorectal tumors but also indicate that further efforts to optimize
with peritoneal dissemination, in an otherwise fit patient in the absence the results of combined treatment for both of these diseases need to be
of distant metastases’’ [7]. The only available level I evidence on undertaken.
the treatment of colorectal carcinomatosis examined the role of The pattern of recurrence after an initial optimal treatment of
cytoreductive surgery combined with intraperitoneal chemotherapy. peritoneal carcinomatosis (that includes complete cytoreduction and
This study showed a median survival of 22.3 months for the group of perioperative intraperitoneal chemotherapy) may be suggestive of the
underlying cause of failure and thus indicate the direction for possible
changes in management. It is possible that a localized form of
recurrence within the abdomen is a result of ‘‘surgical failure’’ to
completely eradicate disease even though patients had complete
adhesiolysis before the administration of intraperitoneal chemo-
therapy. Tumor cells entrapped in scar tissue are less likely to be
eradicated with intraperitoneal chemotherapy than free cancer cells.
Another potential cause for surgical failure is the disease burden on the
small bowel during initial cytoreduction as electroevaporative surgery
cannot be used in the same manner as in other anatomic locations in the
abdomen, which may result in higher failure rates at these sites. Indeed,
an interesting observation on patterns of treatment failure is the
progression of disease on the small bowel surface at reoperation in the
appendiceal group. On the other hand, a diffuse intra-abdominal
recurrence probably suggests a failure of intraperitoneal chemotherapy
to eradicate tumor cells remaining after initial cytoreduction. This is an
important type of failure since it is associated with worse outcomes
both in the colorectal and the appendiceal group of patients.
This study clearly shows that additional efforts to reduce the
incidence of intra-abdominal recurrences after cytoreductive surgery
Fig. 6. Overall survival in 111 patients with documented recurrences and HIPEC need to occur. Our suggestion is that improved local-
according to second surgery (P < 0.001). All patients failed complete regional methods to maintain a disease-free peritoneal surface after
cytoreduction and perioperative intraperitoneal chemotherapy as complete cytoreduction are needed. This should include efforts to
treatment for appendiceal neoplasms with peritoneal dissemination. improve HIPEC by determining optimal agents, maximal doses and

Journal of Surgical Oncology


Failures After Carcinomatosis Treatment 299
TABLE II. Anatomic Location and Character of Disease Recurrence, Number of Patients and Median Survival
for 111 Patients Who Failed Complete Cytoreduction and Perioperative Intraperitoneal Chemotherapy as
Treatment for Appendiceal Neoplasms With Peritoneal Dissemination

Character of Number of Median survival


Location of disease recurrence disease recurrence patients (months)

Abdominal wall only Localized 2 NR


Focal intra-abdominal Localized 57 NR
Diffuse intra-abdominal Diffuse 41 NR
Systemic only Systemic 11 62
Systemic and others Systemic þ other 13 74

optimal levels of hyperthermia to be used. Also, considering the very intraperitoneal chemotherapy for diffuse malignant peritoneal
high propensity for recurrence, especially in high-grade cancers, mesothelioma. Ann Oncol 2007;18:827–834.
additional intraperitoneal treatments administered as early post- 3. Glehen O, Kwiatkowski F, Sugarbaker PH, et al.: Cytoreductive
operative intraperitoneal chemotherapy are likely to be beneficial. surgery with perioperative intraperitoneal chemotherapy for
Cell cycle-specific agents such as paclitaxel and 5-fluorouracil should the management of peritoneal carcinomatosis from colorectal
cancer: a multi-institutional study. J Clin Oncol 2004;22:3284–
be chosen for this purpose. The concept of biphasic treatment, utilizing 3292.
the synergy between different chemotherapeutic agents as well as 4. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of
between heat and some systemic agents should be employed in the cytoreduction and hyperthermic intraperitoneal chemotherapy
operating room when the cancerous nodules in the peritoneum have versus systemic chemotherapy and palliative surgery in patients
been heated [9]. The intraoperative bidirectional chemotherapy is a with peritoneal carcinomatosis of colorectal origin. J Clin Oncol
definite direction for optimization of HIPEC. Multiple controlled trials 2003;21:3737–3743.
in ovarian cancer showed clear benefits for long-term bidirectional 5. Sugarbaker PH: Peritonectomy procedures. Ann Surg 1995;221:
chemotherapy [10,11,12]. The same principle may need to be applied 29–42.
in carcinomatosis of appendiceal and colorectal origin in order to 6. Yan TD, Black D, Savady R, et al.: A systematic review on the
efficacy of cytoreductive surgery and perioperative intraperitoneal
maintain control of disease on peritoneal surfaces. chemotherapy for pseudomyxoma peritonei. Ann Surg Oncol
This study also showed that a second attempt at cytoreduction in 2007;14:484–492.
patients with recurrence is of benefit. This is especially true for 7. Gonzalez-Moreno S: Peritoneal surface oncology: A progress
appendiceal neoplasms where prolonged survivals were achieved report. Eur J Sur Oncol 2006;32:593–596.
with second cytoreduction. Considering the risk of recurrence, perhaps 8. Verwaal VJ, Boot H, Aleman BMP, et al.: Recurrences after
all patients with high-grade cancers such as colorectal cancer should be peritoneal carcinomatosis of colorectal origin treated by cytor-
scheduled for second-look surgery to help accurately select patients eduction and hyperthermic intraperitoneal chemotherapy: Loca-
who may benefit from additional surgery. Certainly, strict protocols for tion, treatment and outcome. Ann Surg Oncol 2003;11:375–379.
follow-up that include surgery for evidence of recurrent disease need to 9. Elias D, Sideris L, Pocard M, et al.: Efficacy of intraperitoneal
chemohyperthermia with oxaliplatin in colorectal peritoneal
be part of the management plan of this group of patients. carcinomatosis. Preliminary results in 24 patients. Ann Oncol
2004;15:781–785.
ACKNOWLEDGMENTS 10. Armstrong DK, Bundy B, Wenzel L, et al.: Intraperitoneal
cisplatin and paclitaxel in ovarian cancer. N Engl J Med 2006;
Lana Bijelic and Tristan D. Yan are surgical oncology research 354:34–43.
fellows, supported by the Foundation for Applied Research in 11. Markman M, Bundy B, Alberts D, et al.: Phase III trial of
Gastrointestinal Oncology. The authors thank David Chang for his standard-dose intravenous cisplatin plus paclitaxel versus
moderately high-dose carboplatin followed by intravenous
assistance in statistical analysis. paclitaxel and intraperitoneal cisplatin in small-volume stage III
ovarian carcinoma: an intergroup study of the Gynecologic
REFERENCES Oncology Group, Southwestern Oncology Group and Eastern
Cooperative Oncology Group. J Clin Oncol 2001;19:1001–
1. Sugarbaker PH: New standard of care for appendiceal epithelial 1007.
neoplasms and pseudomyxoma peritonei syndrome? Lancet 12. Alberts DS, Liu PY, Hannigan EV, et al.: Intraperitoneal
Oncol 2006;7:69–76. cisplatin plus intravenous cyclophosphamide versus intra-
2. Yan TD, Welch L, Black D, et al.: A systematic review on the venous cisplatin plus intravenous cyclophosphamide for
efficacy of cytoreductive surgery combined with perioperative stage III ovarian cancer. N Engl J Med 1996;335:1950–1955.

Journal of Surgical Oncology


Journal of Surgical Oncology 2008;98:300–305

Quality of Life and Nutritional Assessment in Peritoneal Surface


Malignancy (PSM): Recommendations for Care

RICHARD MCQUELLON, PhD,1* CECILIA GAVAZZI, MD,2 POMPILIU PISO, MD, PhD,
3

DAVID SWAIN, BSc (Hons), PGDip RD,4 AND EDWARD LEVINE, MD5
1
Department of Internal Medicine, Section of Hematology and Oncology, Wake Forest University School of Medicine,
Winston-Salem, North Carolina
2
Clinical Nutrition Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
3
Department of Surgery University of Regensburg, Regensburg, Germany
4
Department of Nutrition and Dietetics, Basingstoke and North Hampshire NHS Foundation Trust, England, UK
5
Surgical Oncology Service, Wake Forest University School of Medicine, Winston-Salem, North Carolina

Quality of life (QOL) and nutritional status of patients treated for peritoneal surface malignancy are important areas for ongoing assessment.
A working group of clinicians including a dietitian, physicians, and quality of life researchers was formed as part of the Fifth International
Workshop on Peritoneal Surface Malignancy. The purpose of the group was to form a consensus statement on both quality of life and nutritional
assessment in PSM. The relevant literature from the quality of life and nutritional assessment in peritoneal surface malignancy was reviewed and
integrated to form a consensus statement. Baseline and ongoing assessment of both quality of life and nutritional status of patients undergoing
cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy (HIPEC) is recommended.
J. Surg. Oncol. 2008;98:300–305. ß 2008 Wiley-Liss, Inc.

KEY WORDS: quality of life; nutrition; peritoneal surface malignancy

INTRODUCTION as long as possible. It is not good enough to extend life if this is at the
expense of quality living. Prior to the systematic evaluation of PSM
Cytoreductive surgery (CS) plus hyperthermic intraperitoneal patients with QOL instruments, there was only anecdotal and clinical
chemotherapy (HIPEC) has been associated with high rates of data to evaluate patient outcome following CS plus HIPEC. Clinical
morbidity and mortality. However, a recent report of a randomized trials [4–8] now provide data on which to base judgments about QOL
trial by Verwaal and colleagues and more extensive systematic following this procedure. There is much work to do, particularly
evaluation of this procedure have suggested that mortality and since there has been no randomized clinical trial on the procedure with
morbidity can be reduced with improved selection criteria [1–3]. a companion quality of life component.
Nevertheless, most patients undergoing this treatment will experience,
at the very least, short periods of physical discomfort and impairment
to their overall quality of life and nutritional status [4–6].
Health Related Quality of Life—Definition
The purpose of this article is to establish guidelines for assessing Health related quality of life (HRQL) refers to how a person’s
and maintaining patient overall quality of life and nutritional status health affects their ability to carry out normal social and physical
before and after this extensive surgical and chemotherapeutic activities. There is a general consensus among researchers that the
procedure. It should be noted that nutritional status has a direct quality of life concept is measurable, multi-dimensional, and
effect on the patients overall quality of life. In fact most quality of life subjective or meaningful to the individual patient [9]. Prior to the
instruments include questions about appetite since both disease and development of psychometrically sound QOL instruments, perfor-
treatment affect eating and nutritional status. This article is written in mance status or toxicity ratings were thought to represent QOL. While
two parts: Part I, quality of life issues and Part II, nutritional status. these are dimensions of quality of life, and while performance status
We recognize that this is an artificial distinction and that the two may serve as a crude proxy, there is no substitute for a standardized
areas are closely related. Integration of these important areas of patient QOL questionnaire properly administered.
functioning remains to be accomplished. The QOL construct is measurable in that its dimensions can be
assessed reliably over time and have been shown to be valid with
PART I: HEALTH RELATED QUALITY reference to other validated instruments. There are at least four areas,
OF LIFE FOR PATIENTS WITH PERITONEAL sometime referred to as domains, that can be measured in quality of
SURFACE MALIGNANCY (PSM)
*Correspondence to: Richard McQuellon, PhD, Comprehensive Cancer
Surgical, radiological, and chemotherapeutic treatments of cancer Center of Wake Forest University/Baptist Medical Center, Medical Center
have a potential to do great harm and impair the patient’s quality of Blvd., Winston-Salem, NC 27157-1082. Fax: 336-716-5687.
life both in the short- and long-term. However, they also provide the E-mail: rmcquell@wfubmc.edu
possibility for cure in some instances and prolonged life with quality in Received 21 March 2008; Accepted 21 March 2008
others. For patients with PSM, life is precious since almost everyone DOI 10.1002/jso.21050
enters treatment with a poor prognosis and limited lifespan. In this Published online in Wiley InterScience
setting, it is very important to help patients maintain a life of quality for (www.interscience.wiley.com).

ß 2008 Wiley-Liss, Inc.


Quality of Life and Nutrition in PSM 301
life assessment (Fig. 1) [10]. These include physical well-being at the initial consultation are often symptomatic and anxious. For
(symptoms and side effects such as pain, nausea, etc.), social/family successful QOL assessment, it is necessary to have a nurse or a
well-being (sociability and intimacy, including feeling close to friends, research assistant introduce the questionnaires to the patient and
satisfaction with family communication, etc.), functional well-being explain the rationale for conducting the assessment. Often this is done
(role performance and activities of daily living such as work, sleep etc.) at the time of the outpatient consultation with the surgeon or at
emotional well-being (sadness, nervousness, etc.). There are other some time prior to surgery. At the initial assessment, it is very
important factors in a patient’s life that may modify how they important for the research assistant to check all the data to make sure
experience their overall QOL. These include spirituality, financial and the patient has answered each question. These data are not recoverable
support resources, psychological resilience, sexuality, and family at a later time point. Subsequent assessments can either be conducted at
functioning. follow-up outpatient appointments or by mail. Often patients travel a
At one time, subjectivity was thought to be a detriment in the significant distance for their treatment and will need to complete
assessment of the patient’s status. However, asking the patient to assess follow-up questionnaires at home and mail them back to the treatment
the quality of their life has proven to be a valuable source of data for center. To ensure the completion of the follow-up questionnaires, it
clinicians in making treatment related recommendations as well as may be necessary to repeatedly send questionnaires out to patients and/
providing ongoing care. In fact, patient ratings have been shown to be or phone them to answer any questions they may have.
very useful relative to clinician ratings which may overestimate or Oftentimes, patients are confronted with recurrent disease, signi-
underestimate the patient’s functioning and overall quality of life. ficant debilitating symptoms, and/or may be hospitalized at the time
Additionally, this subjective assessment of the patient’s QOL provides that they received the questionnaires. Also, they may assume that if a
for a measure of meaningfulness. great deal of time has passed since the questionnaires arrived, that there
Why is it important to measure quality of life in the PSM is no longer any use to complete them. Thus, it is helpful to talk with
context? Most patients with peritoneal surface malignancies will die of them to encourage continued participation.
their disease. Nevertheless, CS plus HIPEC allows for extended life far Systematic assessment of patient rated outcomes including health
beyond what might be predicted with no treatment [11–13]. However, related quality of life in the PSM context is new. At this time, to our
extended survival is often at the expense of short-term health since knowledge, there are few published reports of the health related
morbidity rates of up to 40% have been reported in this population. quality of life of patients following cytoreductive surgery plus HIPEC
There are common surgical complications which may result in lengthy [4–8,10]. The general methodology is simple with a pre-surgery
hospitalizations, far beyond the predicted course with no complica- assessment followed by post-surgery assessments at various time
tions. Even without complications, patients will likely face a points ranging from 2 to 4 weeks (at the first follow-up appointment
significant period of convalescence after the procedure. Nevertheless, after surgery) to up to 2 years. Several studies have reported on long-
depending on how symptomatic the patient is at presentation, they may term survivors from 2 to 8 years post-HIPEC [4,11].
show improvements in QOL within several weeks of treatment. We QOL instruments can be thought of as generic, disease specific, or
have reported that patients without major symptoms report a drop in symptom specific. Depending on the purpose of the assessment, one or
QOL scores not long after surgery plus HIPEC while those with more of these instruments could be used to inform both clinicians and
significant symptoms such as ascites show improvement in QOL at patients.
post-surgery [5]. Instruments that have been reported in the literature in this context
What methods and instruments have been used to measure quality of are the Functional Assessment of Cancer Therapy-C (FACT-C), the
life in this context? While the methodology for assessing QOL in the Short Form-36 (SF-36) of the medical outcome study, the European
PSM context is simple, it is not easy. Patients presenting with PSM Organization for Research on Treatment-Cancer (EORT-C) instru-

Fig. 1. Quality of life domains and modifiers [10].

Journal of Surgical Oncology


302 McQuellon et al.
ment. Additionally, the Eastern Cooperative Oncology Group patient reference. However, there has been no systematic evaluation of
Performance Status measure has been reported in the literature. Both how these scores might affect patient decision-making regarding the
the FACT and EORTC are specific to cancer while the SF-36 has been acceptance or rejection of cytoreductive surgery. Also the baseline
used in the general population and can be used for comparison evaluation has not been used to predict morbidity and mortality.
purposes. The utility of assessment after surgery could be in helping the patient
What do Quality of Life Scores mean? Statistical and clinical who has reduced QOL scores to get additional support resources as
significance are not the same thing. While P values are important, necessary such as physical therapy, nutritional counseling and/or
especially in randomized trials which compare two different groups, psychosocial care. While clinical assessment by the surgeon or other
the P-value in and of itself does not indicate whether a particular result medical team members is crucial, systematic patient-related outcomes
has clinical significance. A P-value simply indicates whether or not the using a QOL instrument will provide additional data upon which to
results of the study are more or less likely to be found by chance. make rehabilitation treatment decisions.
In clinical trials using very large samples, there will be considerable What is the tradeoff between short-term (1–3 months) and longer-
statistical power to detect very small and perhaps meaningless term (4–12 months) quality of life in the PSM setting? Patients who
differences between two different treatments. In the PSM setting, opt for this aggressive treatment are likely to have, at minimum,
large sample sizes with QOL data at 1 year post-CS plus HIPEC are significant pain and disability immediately post-surgery and for up to
non-existent. Thus, statistically significant differences over time on a 7–10 days. Their functional status will be severely limited and
QOL questionnaire with smaller samples are likely to indicate clinical they may be bedridden for a significant amount of time should
significance as well. complications develop. Nevertheless, when faced with certain
There has been much progress in implementing QOL measurements progressive disease and death, many patients will opt for short-term
in oncology clinical practice and trials. The literature suggests that loss of function and impairment in quality of life with the hopes of
both patients and clinicians appreciate knowing more about the having longer-term survival and restoration of life’s quality. To date,
patients’ QOL in this context [9]. Nevertheless, once a specific QOL we have not systematically assessed patient quality of life in the period
score has been observed, what does it mean? At present there is no immediately following surgery (1–10 days). It is simply assumed that
indication of the appropriate clinical pathway for patients with a this is a time of misery for most patients. In general, patients can be
particular QOL score. In other words, the use of QOL scores for counseled to expect a return to baseline levels of QOL (if they
determining individual rehabilitation programs outside of clinical trials are nearly symptom free when CS plus HIPEC is performed) in the
is rare if not non-existent in clinical practice. 3–6 months post-surgery time period. However, that the return to pre-
Relatively small gains in HRQL can have significant value for surgery functioning may occur as late as between 6 and 12 months [2].
individual patients. Comparable declines may be less meaningful,
since patients may minimize personal negative evaluations about their Analytic Strategies for Handling QOL Data
condition. This has important implications for the interpretation of the
meaningfulness of change scores on QOL questionnaires. Factors such A serious challenge to understanding QOL results in PSM trials is
as adaptation to disease, response shift, dispositional optimism and the the fact that many patients are either too sick to complete
need for signs of clinical improvement may be contributing to the questionnaires at follow-up time points or deceased. For example,
results and should be investigated in future studies [9]. one study reported 64 patients completing QOL questionnaires at
The minimally important difference (MID) has been defined as the baseline with only 31 completing questionnaires at the 12-month
smallest change in a patient related outcome measure that is perceived follow-up due to the fact that 24 patients had died [5]. Because of this
by patients as beneficial or that will result in the change in treatment. rate of mortality, it is necessary to use analytic techniques that account
There are a number of anchor based and distribution based methods for data that are missing, not at random, to avoid a biased estimate of
that have been used to determine MID for QOL measures. The anchor the QOL in the study population. (See Fairclough et al. [14] for an
based methods require an external patient base or clinical criteria for excellent discussion of this analytic problem.) Similar results with
comparison to determine what changes in QOL scores are meaningful. regard to drop-outs have been reported by Alexander et al., [6] who
The distribution based methods reflect one of several statistical indices began with a total of 73 patients at baseline and reported 45 patients at
of change. The method for determining the MID is still evolving [9]. the 9-month follow-up and Schmidt et al. [4] with 67 patients assessed
This is particularly true in the PSM context where patients are choosing at baseline and 20 patients being assessed ranging 1–8 years following
major surgery with the high likelihood of significant morbidity. They treatment.
are choosing short-term (1–3 months) reduction in quality of life for
long-term gain in the form of additional life span. Clinical experience Using HIPEC Quality of Life Data in Clinical Practice
suggests that for the patients with minimal morbidity, this is a
reasonable option. However, for those with significant morbidity and The current literature suggests that a sub sample of patients may
presumed deficits in quality of life, it remains an open question as to return to near baseline levels of functioning on quality of outcome
whether or not the decision to seek this intensive treatment was in fact measures at between 6 and 12 months, with some reporting
the ‘‘right’’ decision for the patient. improvement to baseline as early as 3 months. It is important to
What time periods need to be studied? More patient-rated QOL data underscore this is likely the best of outcomes since attrition rates from
in the 1- to 3-week time period following surgery are needed as well as baseline to 6 months are substantial and range from 50% to 60%. Thus,
the time period over the first year where data on recurrent disease and the sickest patients may not be represented in these data. In other
accompanying QOL are sparse. There is very little information on words, the data may be overly optimistic. Nevertheless, it is useful for
patients who do not have a reasonably successful course following CS patients to see this information. Figure 2 illustrates how such data can
plus HIPEC. In one study, survivors reported having no regrets be utilized in patient counseling prior to and after cytoreductive
following the CS plus HIPEC procedure [6]. Nevertheless, clinical surgery plus HIPEC. The graph illustrates physical functioning, role
experience suggests that for a substantial minority of patients, post- functioning and general health as reported by patients in the study
surgery recovery is very difficult and fraught with numerous conducted by McQuellon, et al. [13]. Also, normative sample scores of
complications, severely compromising short-term QOL. patients age 45–54 are reported for comparison. This is necessarily an
How can quality of life scores be used? To date, the pre-surgery QOL imprecise comparison. The ideal comparison would be a patient of
assessments have only been used to establish a baseline score for similar age without cancer who was identified by each of the study

Journal of Surgical Oncology


Quality of Life and Nutrition in PSM 303
subjects in the HIPEC sample. Nevertheless, this gives us some general PART II: NUTRITIONAL CARE OF PATIENTS
framework for comparison and suggests that even though patients do WITH PSM AND CANDIDATES
relatively well and return to greater than baseline levels of physical
function and role functioning following HIPEC, these scores are still FOR CS PLUS HIPEC
considerably lower than the comparison sample. Further, their reported It is well documented that malnutrition is associated with
general health is significantly lower than a sample of the general depression of humoral and cellular immune function, alterations in
population. On the one hand, the ‘‘glass is half full,’’ and patients return the inflammatory response and delay or failure of the wound healing
to baseline functioning as early as 3–6 months. On the other hand, process. As a result, increased complication rates, length of stay,
the ‘‘glass is half empty,’’ in that, at least on physical measures, this cost, and mortality occur in surgical patients [15]. Moreover there is a
group of patients scores lower than the normative sample comparison strong relationship between nutritional status and various clinical
group. outcomes, including quality of life, survival and the ability to tolerate
treatment [16,17].
Recommendations for QOL Assessment in PSM We know of no published nutritional data on patients selected for
CS plus HIPEC for PSM. Nutritional assessment and adequate
(1) All patients being evaluated for CS plus HIPEC for PSM should nutritional treatment should be considered in these patients in the
have their QOL and performance status evaluated. This can be perioperative period and nutritional follow-up should be planned for at
utilized to help evaluate the risk of the proposed procedure. least 1 year after surgical procedures [18,19].
(2) Evaluation of quality of life after surgery should be systematically
evaluated by treating centers. Prevalence of Malnutrition in Patients
(3) Standardized instruments such as the FACT or EORTC should Affected by PSM
be used as core instruments. Additional general quality of life
measurers, that is, the SF-36 or symptom specific questionnaires Few data are available on the prevalence of malnutrition in patients
(the Brief Pain Inventory), can be used at the clinicians discretion affected by PSM. Nutritional status varies considerably based on the
based on patient’s symptoms. different underlying diseases associated with PSM. For example,
(4) Clinical trials assessing the efficacy of CS plus HIPEC should pseudomyxoma peritonei (PMP) is rarely associated with malnutrition.
incorporate a quality of life patient-rated outcome instrument. This Patients affected by PMP and admitted to Instituto Nazionale Dei
is particularly important in any randomized trials since the impact Tumori (INT) for surgery, reported a history of involuntary weight loss
of treatment in two different trials can be assessed. This is crucial if and reduced food intake in less than 3% of cases. No data are available
there are significant survival benefits for one treatment over the for peritoneal mesothelioma. In contrast, peritoneal carcinomatosis in
other and can answer the question as to whether or not survival ovarian, colon and gastric cancer is usually a situation of a long lasting
benefit is at the expense of the patient’s quality of life. and advanced disease; when it is diffuse, it creates difficulties in
(5) Future studies should focus on answering the following questions: regular oral alimentation and it is generally associated with a high
(a) What is the minimally important difference in quality of life prevalence of malnutrition.
measures in this context? This is a developing field of inquiry Among patients affected by different types of gynaecologic cancer,
where much work is needed. What exactly do these differences malnutrition has been reported significantly higher in women with
on specific instruments mean to patients? ovarian cancer, with a prevalence of 67% [20,21]. The percentage of
(b) Can preoperative quality of life data contribute to clinical patients affected by colon cancer malnutrition has been reported
parameters in predicting survival? to range from 34% to 54% [16,22]. The highest prevalence of

Fig. 2. Selected SF-36 Scales at four time points.

Journal of Surgical Oncology


304 McQuellon et al.
malnutrition has been always reported in patients affected by gastric (3) Prospective randomized studies are needed to define the best
cancer, ranging from 62% to 83% [16]. timing and route perioperative nutrition.
(4) Nutritional counselling should be given to patients before
Standard Methods for Nutritional Status Assessment discharge and ambulatory follow-up should be planned with
a time schedule correlated to number and type of resections.
Standard nutritional screening is needed for all candidates for major
elective surgery and especially patients affected by PSM. More in-
depth nutritional assessment should be considered in malnourished REFERENCES
patients only. A variety of nutritional indices have been found to be
valuable in predicting outcome when used alone, but there is no 1. Yan TD, Black D, Savady R, et al.: A systematic review on the
consensus on the best method for preoperative nutritional risk efficacy of cytoreductive surgery and perioperative intraperitoneal
assessment in patients undergoing major surgery [23]. The European chemotherapy for pseudomyxoma peritonei. Ann Surg Oncol
Society for Clinical Nutrition and Metabolism has stated that recent 2007;14:484–492.
2. Verwaal VJ, van Ruth S, de Bree E, et al.: Randomized trial of
weight loss, body mass index and reduced dietary intake are considered
cytoreduction and hyperthermic intraperitoneal chemotherapy
the more important indicators for nutritional screening in the hospital versus systemic chemotherapy and palliative surgery in patients
setting, by Laviano and Meguid [22]. with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol
Malnutrition should be considered in the presence of weight loss 2003;21:3737–3743.
greater than 5% within 3 months, body-mass index below 18.5 Kg/m2, 3. Esquivel J, Sticca R, Sugarbaker P, et al.: Cytoreductive surgery
and/or food intake below 50% of normal requirement. A more in- and hyperthermic intraperitoneal chemotherapy in the manage-
depth nutritional assessment can be performed using biochemical ment of peritoneal surface malignancies of colonic origin: A
and functional measures. Biochemical nutritional indicators include: consensus statement. Ann Surg Oncol 2007;14:128–133.
serum levels of total protein, albumin, prealbumine, transferrin, 4. Schmidt U, Dahlke MH, Klempnauer J, et al.: Perioperative
morbidity and quality of life in long-term survivors following
cholinesterase, and lymphocytes. Serum albumin <30 g/L, with no
cytoreductive surgery and hyperthermic intraperitoneal chemo-
evidence of hepatic or renal dysfunction should be considered an therapy. Eur J Surg Oncol 2005;31:53–58.
indicator of potential morbidity [24]. A functional measure that could 5. McQuellon RP, Loggie BW, Fleming RA, et al.: Quality of
assess nutritional status indirectly is evaluated muscle strength by hand life after intraperitoneal hyperthermic chemotherapy (IPHC)
grip dynamometer. for peritoneal carcinomatosis. Eur J Surg Oncol 2001;27:65–
73.
6. Alexander R, Mavroukakis S, Libutti S, et al.: Impact of tumor
Perioperative Nutrition Interventions resection (Rxn) and intraperitoneal (IP) chemotherapy (CHRx) on
Although no data are available on nutrition support in patients with health related quality of life (HRQL) in patients (Pts) with
peritoneal surface malignancies (PSM). Proc Soc Surg Oncol
PSM and undergoing CS and HIPEC, perioperative artificial nutrition
2004;11:S109.
should be planned considering the extent of abdominal surgery and 7. Tuttle TM, Zhang Y, Greeno E, et al.: Toxicity and quality of life
the important postoperative catabolic state, following international after cytoreductive surgery plus hyperthermic intraperitoneal
guidelines for surgical patients [25–26]. All malnourished patients chemotherapy. Ann Surg Oncol 2006;13:1627–1632.
should be supported in pre- and post-operative phases. In the 8. McQuellon RP, Loggie BW, Lehman AB, et al.: Long-term
preoperative phase, oral nutritional supplements should be given if survivorship and quality of life after cytoreductive surgery plus
the gut is functioning; total parenteral nutrition (TPN) should be intraperitoneal hyperthermic chemotherapy for peritoneal carci-
administrated only in presence of mechanical bowel obstruction. nomatosis. Ann Surg Oncol 2003;10:155–162.
Postoperative artificial nutrition should start as soon as possible, when 9. Sloan JA, Frost MH, Berzon R, et al.: The clinical significance of
quality of life assessments in oncology: A summary for clinicians.
general conditions are stable. In line with international guidelines early
Support Care Cancer 2006;14:988–998.
enteral nutrition (EN) should be planned; however, the lack of 10. Cella DF: Functional status and quality of life: Current views on
knowledge on the small bowel physiology after HIPEC indicates that measurement and intervention. Functional status and quality of
EN nutrition should be started with caution and initially to overlap life in persons with cancer. American Cancer Society, 1–12. First
with TPN. National Conference on Cancer Nursing Research, American
Cancer Society, Atlanta, GA, November 30–December 2, 1989;
1991.
Nutrition Interventions After Discharge 11. Levine EA: Problems of success and problems of failure:
Metabolic and mechanical effects of surgery can have a negative Recurrent disease after cytoreductive surgery and intraperitoneal
impact on nutritional status and quality of life for a long time after chemoperfusion. Ann Surg Oncol 2004;11:351–353.
12. Stewart JH IV, Shen P, Levine EA: Intraperitoneal hyperthermic
discharge [27]. All patients with gastrectomy, colectomy and ileal chemotherapy for peritoneal surface malignancy: Current
resection have a progressive weight loss for several months after status and future directions. Ann Surg Oncol 2005;12:765–
surgery. Weight loss can be so severe in patients with multiple 777.
resections that home parenteral nutrition should be considered. More 13. McQuellon RP, Danhauer SC, Russell GB, et al.: Monitoring
systematic study of the post-discharge time period is necessary. health outcomes following cytoreductive surgery plus intra-
peritoneal hyperthermic chemotherapy for peritoneal carcinoma-
tosis. Ann Surg Oncol 2007;14:1105–1113.
Recommendations for Nutritional Management 14. Fairclough DL, Peterson HF, Cella D, et al.: Comparison of
several model-based methods for analysing incomplete quality of
(1) Systematic study of the nutritional status of PSM surgery
life data in cancer clinical trials. Stat Med 1998;17:781–796.
candidates is needed before during and after CS plus HIPEC. 15. Mullen JL, Buzby GP, Waldman MT, et al.: Prediction of
(2) Nutritional screening should be performed in all patients affected operative morbidity and mortality by preoperative nutritional
by PSM and candidates to major elective surgery, while a more in- assessment. Surg Forum 1979;30:80–82.
depth nutritional assessment should be considered in malnourished 16. Andreyev HJ, Norman AR, Oates J, et al.: Why do patients with
patients only. weight loss have a worse outcome when undergoing chemo-

Journal of Surgical Oncology


Quality of Life and Nutrition in PSM 305
therapy for gastrointestinal malignancies? Eur J Cancer 1998;34: 22. Laviano A, Meguid MM: Nutritional issues in cancer manage-
503–509. ment. Nutrition 1996;12:358–371.
17. DeWys WD, Begg C, Lavin PT, et al.: Prognostic effect of weight 23. Kuzu MA, Terzioglu H, Genc V, et al.: Preoperative nutritional
loss prior to chemotherapy in cancer patients. Eastern Coopera- risk assessment in predicting postoperative outcome in patients
tive Oncology Group. Am J Med 1980;69:491–497. undergoing major surgey. World J Surg 2006;30:378–390.
18. Huhmann MB, Cunningham RS: Importance of nutritional 24. Martindale RG, Maerz LL: Management of perioperative
screening in the treatment of cancer-related weight loss. Lancet nutrition support. Curr Opin Crit Care 2006;12:290–294.
Oncol 2005;6:334–343. 25. Weimann A, Braga M, Harsanyi L, et al.: ESPEN Guidelines on
19. Kondrup J, Allison SP, Elia M, et al.: Educational and clinical enteral nutrition: Surgery including organ transplantation. Clin
Practice Committee, European Society of Parenteral and Enteral Nutr 2006;25:224–244 [Epub 2006 May 15].
Nutrition (ESPEN). ESPEN guidelines for nutrition screening 26. Guidelines for the use of parenteral and enteral nutrition in
2002. Clin Nutr 2003;22:415–421. adult and pediatric patients. J Parenter Enteral Nutr 2002;26:
20. Tunca JC: Nutritional evaluation of gynaecologic cancer patients 15A–65A.
during initial diagnosis of their disease. Am J Obstet Gynecol 27. Beattie AH, Prach AT, Baxter JP, et al.: A randomised controlled
1983;147:893–896. trial evaluating the use of enteral nutritional supplements
21. Laky B, Janda M, Bauer J, et al.: Malnutrition among postoperatively in malnourished surgical patients. Gut 2000;46:
gynaecological cancer patients. Eur J Clin Nutr 2007;61:642–646. 813–818.

Journal of Surgical Oncology

You might also like