UNIVERSITY OF MEDICINE AND PHARMACY „GRIGORE … · UNIVERSITY OF MEDICINE AND PHARMACY...

46
UNIVERSITY OF MEDICINE AND PHARMACY „GRIGORE T. POPA” IAȘI PhD Thesis Summary HYPERTHERMIC INTRAPERITONEAL CHEMOTHERAPY ASSOCIATED WITH CYTOREDUCTIVE SURGERY IN PERITONEAL CARCINOMATOSIS OF GASTROINTESTINAL ORIGYN SCIENTIFIC COORDINATOR, PROFESSOR DR. SCRIPCARIU Viorel PhD STUDENT HUŢANU Ionuţ 2013

Transcript of UNIVERSITY OF MEDICINE AND PHARMACY „GRIGORE … · UNIVERSITY OF MEDICINE AND PHARMACY...

UNIVERSITY OF MEDICINE AND PHARMACY „GRIGORE T. POPA” IAȘI

PhD Thesis

Summary

HYPERTHERMIC INTRAPERITONEAL CHEMOTHERAPY ASSOCIATED

WITH CYTOREDUCTIVE SURGERY IN PERITONEAL

CARCINOMATOSIS OF GASTROINTESTINAL ORIGYN

SCIENTIFIC COORDINATOR,

PROFESSOR DR. SCRIPCARIU Viorel

PhD STUDENT

HUŢANU Ionuţ

2013

Research funded by the project “Burse doctorale pentru creşterea

competitivităţii în domeniul medical şi farmaceutic” POSDRU/88/1.5/S/63117,

cofounded from the European Social Fund through the Operational Program for

Human Resources Development 2007-2013

Main intervention field: 1.5 „Doctoral and post-doctoral program

supporting the research”

Project title: „Doctoral fellowships for the increase of competitiveness in

the medical and pharmaceutical field”

Beneficiary: “Gr. T. Popa” University of Medicine and Pharmacy of Iasi

Partner : University of Medicine and Pharmacy of Timişoara

The thesis consists of:

- theory (40 pages )in four Chapters

- personal research (95 pages) in five Chapters

- 352 references

- 63 pictures

- 25 tables

- 2 B+ aticles and 5 ISI indexed published or accepted

In this abstract, the table of contents and the number for each figure is the same as in

the thesis

Key words

Peritoneal carcinomatosis, HIPEC, peritonectomy, cytoreduction,

pseudomyxoma peritonei, diffuse malingnant mesothelioma

Table of contents

Personal motivations 9

General Part.Level of acknowledge 11

CHAPTER 1 Considerations on peritoneal carcinomatosis

Anatomy and psysiopathology 13

1.1.I. Ultrastructure of peritoneum 13

1.1.II. Molecular biology of peritoneal carcinomatosis 14

1.2. Clinical aspects in peritoneal carcinomatosis 16

1.3. Imaging of peritoneal carcinomatosis 16

CHAPTER 2 Hyperthermic intraperitoneal chemotherapy 19

2.1. Intraperitoneal assessment of the extent of peritoneal carcinomatosis 19

2.2. Hyperthermic intraperitoneal chemotherapy (HIPEC) 20

2.3. The evolution of the treatment of peritoneal carcinomatosis 21

2.4. Peritonectomy procedures – technique 22

CHAPTER 3 Role of HIPEC in the treatment of peritoneal carcinomatosis 25

3.1. Colorectal cancer 25

3.1.I. The incindence of colorectal peritoneal carcinomatosis 25

3.1.II. Survival after systemic chemotherapy 27

3.1.III. Survival after systemic chemotherapy associated with biologic agents 28

3.1.IV. Survival after HIPEC 29

3.1.V. Peritoneal carcinomatosis associated with liver metastases 33

3.1.VI. Strategy in incidental findins of peritoneal carcinomatosis of colorectal origin 33

3.1.VII. Cytoreductive surgery without HIPEC 33

3.1.VIII Tumor relapse after HIPEC 34

3.2. Gastric cancer 34

3.3. Ovarian cancer 37

3.4. Pseudomyxoma peritonei 39

3.4.I. Pathology os pseudomyxoma peritonei 39

3.4.II. Clinical aspects of pseudomyxoma peritonei 42

3.4.III. Treatment of pseudomyxoma peritonei 42

3.5. Diffuse malignant peritoneal mesothelioma 44

CHAPTER 4 Considerations on HIPEC 48

4.1. Patient selection for HIPEC 48

4.2. Morbidity and mortality after HIPEC 49

4.3. Quality of life after HIPEC 53

4.4. Costs of HIPEC procedure 56

4.5. Learning curve in cytoreductie surgery 59

Personal contributions 61

CHAPTER 5. Morbidity and mortality after HIPEC for peritoneal carcinomatosis of colorectal

cancer and pseudomyxoma peritonei. Experience of National Cancer Institute of Milano, Italy

61

5.1. Aims of the study 61

5.2. Material and methods 62

5.3. Results 83

5,4. Discussions 93

CHAPTER 6 The role of circulating tumor markers in pseudomyxoma peritonei 99

6.1. Aims of the study 99

6.2. Material and Methods 100

6.3. Results 105

6.4. Discussions 127

CHAPTER 7. Treatment of peritoneal carcinomatosis of gastro-intestinal origin-a retrospective

study of 203 cases 131

7.1. Introduction. Aim of the study 131

7.2. Material and methods 132

7.3. Results 132

7.4. Discussions 137

7.5. Conclusions 138

CHAPTER 8 The role of perioperative systemic chemotherapy in diffuse malignant peritoneal

mesothelioma patients treated with cytoreductive surgery associated with hyperthermic

intraperitoneal chemotherapy (HIPEC) 141

8.1. Introduction. Aims of the study 141

8.2. Material and methods 141

8.3. Results 147

8.4. Discussions 154

CHAPTER 9 Discussions and conclussions 157

9.1. Discussions 157

9.2. Conclusions 163

References 165

Addenda 185

Abbreviations 185

Informed Consent for HIPEC treatment 186

List of scientific papers issued during PhD studies 191

Personal Motivations

Peritoneal carcinomatosis (PC) is a disease with a dismal prognosis, difficult

to treat wich represent a permanent problem for surgeons, medical oncologists, and

other specialities.

Overall survival of the patiens with peritoneal carcinomatosis is very poor.

It‘s natural history is very unpredicteble with long hospitalisation, low quality of life, ,

palleative surgery to treat various complications, ascites, and finally death.

Primary peritoneal carcinomatosis is a very rare disease wich include diffuse

malignant peritoneal mesothelioma and serous peritoneal carcinomatosis. Secondary

pesitoneal carcinomatosis is more frecquent and coul be found in advanced intra or

extraperitoneal disease. The most frecquent cancers are gastric, colorectal, pancreatic

and ovarian. Rare cancers are billiary, prostatic, renal and breast cancer and a very

rare appendicular cancer - pseudomyxoma peritonei.

EVOCAPE study on 370 patients with PC of gastrointestinal origyn revealed

aoverall survival of 3 to 9 month for the patients with 3 months for those with nodules

greater than 2 cm and 9 months for those with nodules less tan 5 mm. [1].

Hiperthermic intraperitoneal chemotherapy (HIPEC) has become during the

last 30 years standard therapy fost PC of colorectal origun and for pseudomyxoma

peritonei and peritoneal mesothelioma. There are reports favoring the complex

procedure for other entities like ovarian, gastric carcinomatosis.

One of the concerning of this complex procedures reguard the lack of high

evidence base medicine with only one randomised prospective study comparing the

survival of patients with peritoneal carcinonamtosis of colorectal origyn with or

without HIPEC. Other concernings are high morbidity and mortality, costs, quality of

life, learning curve.

In Romania HIPEC procedure is not performed. The aim of this study is to

investigate the morbidity and mortality of this procedure, the morbidity and mortality

of „traditionally treated patient‖ with carcinomatosis and various aspects of the

treattment of pseudomyxoma peritonei and peritoneal diffuse mesothelioma.

Level of aknowledge

CHAPTER 1. Peritoneal carcinomatosis

Mechanisms of peritoneal carcinomatosis

Nowadays the mechanism of CP is incompletely understood. It is considered

that it‘s different from the hematogenous or lymphatic pathways involved in the

development of distant metastases in parenchymal organs or lymph nodes. In order to

explain the occurrence of CP, there have been proposed several theories. One

hypothesis suggests that the tumor cells once they are infiltrating the serous and once

they are free into the peritoneal cavity, then they are able to generate the appearance

of CP. In this process may be involved also the tumor manipulation during surgery or

biopsy[10]. This theory can not explain, however, a rare occurrence of CP in T1 or T2

tumors [1]. It has been described the presence of free tumor cells in colorectal cancer

in stage I before their surgical manipulation [11].

I.2. Clinical aspects in peritoneal carcinomatosis

The clinical aspects in CP usually overlaps with the primary tumor signs and

symptoms. To those pacients where the primitive tumor hasn‘t been diagnosed, it may

occure signs as ascites, enlargement of the abdomen, weight, physical or intellectual

ability loss, irritability, changes in eating habits, flatulence, presence of palpable

tumors. Often, especially in pseudomixoma peritonei and ovarian cancer, the palpation

of the abdomen reveals a central tumor mass because of the tumoral implants on the

great omentum, also known as "omental-cake". In advanced cases, the patients

become cachectic, and the clinical picture may be complicated by the appearance of

intestinal obstruction because of the invasion of the small or large bowel, local

peritonitis in case of tumor invasion or diastatic perforation, bleeding.Sometimes

clinical signs and symptoms may be lesions to other secondary (metastatic) sites: liver,

lung, bone, brain.

.

CHAPTER 2. Hyperthermic intraperitoneal chemotherapy (HIPEC) treatment of

peritoneal carcinomatosis

2.2. Hiperthermic intraperitoneal chemotherapy (HIPEC)

Hiperthermic intraperitoneal therapy belongs to locoregional therapies. With

other procedures like locoregional treatment of unresectable liver metastases [50-51],

isolated limb perfusion for advanced tumors with the goal to preserve the function

[52-53], intraoperative radiotherapy for advanced breast cancer [54] HIPEC provides

high dose of chemotherapic agents locally avoiding systemic complications. In the last

3 decennia HIPEC proved its efficacity in various loaclly advanced tumors like

colorectal cancer [55], ovarian [56], gastric [57-58] and has became golden standard

for rare tumors like pseudomyxoma peritonei (PMP) [59-60] and peritoneal

mesothelioma (MP) [61].

2.3. The evolution of the surgical treatment of peritoneal carcinomatosis

The evolution of intraperitoneally administred chemotherapy [69]:

Weisberger and col. in 1955 [70] administred nitrogen mustard for the threatment of

ascites of in advanced ovarian cancer. Authors observed a good palliation of ascites

but no improvements in survival pentru tratamentul ascitei neoplazice.

1980 Spratt and col. [71] suggested intraperitoneal hiperthermic chemotherapy after

animal studies.Then they performed for the first time the procedure in a 37 years old

male with pseudomixoma peritonei

1981 Spreyer and col. [72] described the farmacology of intraperitoneally

adminiustred 5-florouracyl in liver cancer patients

1984 Hamazoe and col. [73] first study of hiperthermic intraperitoneal chemotherapy

with Mitomicin C in patients with gastric and colorectal carcinomatosis proving the

efficacity of the method

1985 Sugarbaker and col. [74] published the results of a randomized study

(systemic chemotherapy vs intraperitoneal chemotherapy) for advanced colorectal

cancer with no macroscopic carcinomatosis. Authors observed a low incidence of

peritoneal carcinomatosis in patients treated with intraperitoneal chemotherapy.

1988 Koga and col. [75] reported first study of prophylactic intraperitoneal

chemotherapy with Mitomicin C in advanced gastric cancer (T3). The authors

observed an improvement of survival.

1988 Kiuchi and col. [76] reported an overall survival of 380 days and 62%-one

year survival in a population of pacients with gadvanced gastric cancer treated with

HIPEC and overall survival of 160 days and 16% one years survival in patients

traditionally treated.

1995 Sugarbaker [77] described peritonectomy procedure

2003 Verwaal and col. [78] published the results of the first randomized study on

HIPEC in peritonel carcinomatosis of colorectal cancer. The authors observed an

overall survival of 22 monts in patients treated with hiperthermic intraperitoneal

chemotherapy and 12 month in patients treated with surgery plus systemic

chemotherapy.

CHAPTER 3. The role of HIPEC in the treatement of peritoneal carcinomatosis

3.1.The Colorectal cancer

Colorectal cancer in Europe has estimated an incidence of 415 000 cases per

year. It is estimated that approximately 210 000 patients die because of this

neoplasia[105].CP is one of the most common ways of dissemination in colorectal

cancer having symptoms which may overlay on the primary tumor condition, only if

it‘s limited. It‘s diagnosed in 7-10% of patients who had surgery for colorectal

cancer, it develops in 4-19% of patients who underwent surgery with curative visa and

occurs in approximately 44% of patients with postoperative tumor recurrence.In a

retrospective study of 3019 patients [106] , 349 of them (13%) developed CP of which

214 (61%) had a CP synchronous and 135 (39%) CP metachronous.

Treatment of metastatic colorectal cancer is multimodal,surgery is the only curative

intent, despite the new class of chemotherapy which significantly improved survival.

3.1.IV.Results in CP‘s multimodal treatment (cytoreduction + HIPEC) of colorectal

etiology.

In 1995, Sugarbaker et al. [81] published the first results of multimodal

therapy including tumor cytoreduction associated with hyperthermic intraperitoneal

chemotherapy in a group of 181 patients, who uderwent surgery for colo-rectal CP (51

patients), and for apendicular etiology for CP (130 patients). The authors identified

colonic origin, nods status, tumor differentiation and extent of CP as negative

prognostic factors. In those colo-rectal patients with incomplete cytoreduction,

authors have reported a 3-year survival of 20%, significantly lower than those with a

full cytoreductive (60%).

There is only one study [119] randomized and completed so far on the colo-

rectalCP treated by HIPEC. In this study, between february 1998 - august 2001, 105

patients were randomized into two groups. All 105 patients had histological diagnosis

of colo-rectal CP and no liver or extra-abdominal metastases, age under 71years old,

with acceptable performance status and good renal, liver and bone marrowfunction.

The patients were randomized to standard therapy or experimental treatment. The

patients in the standard group (51 patients after randomization) went surgery only if

there were casesof complications and in that situation they performed bypass, stenting

and exceptional resection. Otherwise, in a total of 44 patients, there were performed

standard chemotherapies from that that period in Netherlands, after a schema

containing 5-Fluorouracil and Leucovorin. In the experimental group (54 patients after

randomization) 49 patients wentsurgerypracticing tumor cytoreduction and

hyperthermic intraperitoneal chemotherapy for a period of 90 minutes with mitomycin

C.Subsequently these patients (33 patients) had systemic chemotherapy following the

same pattern as those in the standard arm.All patients were subsequently re-evaluated

every 3-6 months. Study results showed a median survival of 12 months in arm with

systemic chemotherapy and 22 months in the experimental arm so a benefit of about

10 months

3.4. Pseudomixoma peritonei

Pseudomixoma peritonei (PMP) is localized or generalized progressive

accumulation of a gelatinous consistency ascite in the abdominal cavity, usually a

consequence of chistadenoma appendix rupture.PMP is a generic term used to

describe clinical and radiological syndrome, but not a histopathologic diagnosis.

Rokitansky was the first to describe an appendix mucocel, he named it hydrops

vermiform process [185]. In 1884 [186], Werth introduced the PMP term describing

the first case in a patient with an ovarian tumor.In 1901 Frankel had described a case

ofPMP associated with an appendicular cyst.

PMP prevalence in the general population is difficult to assess. It is estimated

at 1-2 cases per 1 million inhabitants.In a retrospective study [187] for a period of 10

years from 167 744 appendectomies carried out in Holland in 1482 were found

appendix tumors (0.9%) of which 574 were epithelial tumors. Because it is a rare

condition there is no prospective study on the treatment of this heterogeneous

disease.Nowadays,in the world are 8 reference centers with more than 100 patients

[59], the greatest casework of 500 cases being reported in Basingstoke.

CHAPTER 4. Considerations about HIPEC

4.1. Patient selection for HIPEC

Patient selection for HIPEC is not easy. Surgical treatment decision should

not belong to a single physician, it should be taken in consensus by a multidisciplinary

team [222].Piso et al. [224] have sketched some indicative criteria for selection of

patients with CP proposed for surgery by identifying factors related to tumor and

patient: factors related tumors: primary tumor location, histological type and grading;

presence of extra-abdominal metastatic disease, the presence of para-aortic lymph

nodes metastasis, Peritoneal Cancer Index, bowel and hepatogastric

ligamentinvolvement, bladder or ureteral stenosis, response to prior systemic

chemotherapy. Other factors: ECOG performance status,patient comorbidities, the

learning curve of the surgeon, the patient's informed consent, quality of life expected

postoperatively.

4.2. Morbidity and mortality after HIPEC

The complications, such as fistula or the perforation, are at a rate of 7-17%

[88, 237-238]. Compared to the 2-4% incidence of fistula in elective colorectal

surgery [239] the relatively large number of anastomotic anastomotic fistulas is

justified by thepatients‘status, often with a history of multiple interventions with

postoperative adhesions that make dissection difficult,but also by the fact that in some

cases, in order to obtain anproper cytoreductive result, we actually increase the risk of

complications by performing a large number of anastomosis. Often we practice serous

tumor cytoreduction in bowel or mesentery, which increase the risks of bowel necrosis

and devascularisation.

4.5. The learning curve in cytoreductive surgery.

Smeenk et al. [223] believes that learning curve peak is reached after

approximately 130 cases.The authors have noticeda decreaing morbidity from 71 to

34%, a decrease in the length of stay for 24 to 17 days and an increased proportion of

patients with complete cytoreductive from 35 to 65% in a group of 323 patients who

went for surgery for colo-rectal CP or PMP, dividing the patients into 3 groups

according to the period in which they were operating.

In the group of patients the ultimate complications of gastrointestinal

complications have been observed (32%), infection (20%), lung (13%), bone marrow

suppression (12%).Post-operative mortality decreased from 8% to 4% for the first 75

patients operated on.One of the authors explanations for better results over time is

related to a more rigorous selection of patients excluding patients with extensive

abdominal CP (6-7 regions), becausecomplete citoreduction is difficult and because of

thepostoperator riscks and complications, due to large excision necessary for optimal

cytoreduction.Yan et al. [271] in a group of 140 patients operated by cytoreduction

combined with HIPEC has been a net improvement in the parameters of the last 70

patients from the first 70 patients.

PERSONAL PART

CONTRIBUTIONS

CHAPTER 5. Morbidity and mortality in patients operated after HIPEC with

peritoneal carcinomatosis of colorectal etiology and Pseudomixoma peritonei at

National Tumor Institute in Milan, Italy

5.1. The purpose of the study

The main objective of this study is to identify the main risk factors for the occurrence

of severe postoperative complications in patients with CR and PMP peritoneal

carcinomatosis. The second objective is to evaluate the oncological outcome of

patients with CR CP, when performind peritonectomie associated with intraperitoneal

chemotherapy.

5.2.I. The group of patients

The study was conducted on a group of patients operated at the National

Cancer Institute (INT) in Milan between june 1996 - march 2012.In this period went

for surgery a total of 473 patients with various diseases of the peritoneum.All patients

were operated by a single surgical team, having as principal surgeon the same surgeon

(Marcello Deraco) who also performed patient selection for curative intervention visa.

The decision was taken after surgery consent multidisciplinary team consisting of a

surgeon, oncologist, pathologist, anesthesiologist and nutritionist.

All patients were operated after a protocol developped by INT and met the

following criteria: CR CP or PMP, good renal function, hepatic and hematopoietic,

age under 75 years old, ECOG performance status less than 3, the absence of distant

metastases and retroperitoneal lymphadenopathy blocks, imaging investigations that

suggest the possibility of an optimal citoreductive surgery, signed informed consent of

patients.

Fig. 5.7 Right upper quadrant peritonectomy

Collection of Dr. Marcello Deraco

Fig. 5. 14 Parietal and pelvic peritonectomy

Collection of Dr. Marcello Deraco

TABLE 5.IV

Risk factors for postoperative high grade mororbidity after HIPEC in PMP and

colorectal peritoneal carcinomatosis patients

Factors Morbidity Grade 3-5

Univariate analysis*

Multivariate**

p Odds Ratio

Confidence interval P

Tumor (Colon vs PMP) 0,91 - - -

Sex (Male vs Female) 0,51 - - -

Age >52 years 0,30 - - -

Body mass index <22

( yes vs no)

0,14 - - -

Performance status (0 vs 1-2) <0,01 3,73 1,50-9,30 <0,01

Charlson morbidity index > 3 (da vs nu) 0.600 - - -

Histologic subtype (Colon + PMCA vs

DPAM)

0,12 - - -

Prior surgical score 0-1 vs 2-3 0,40 - - -

Preoperative chemotherapy

(yes vs no)

0,20 - - -

Preoperative albumin

(<3,5 vs >3,5)

0,09 - -

Preoperative lymphocite count < 1500 (da vs nu)

0,02 - -

Preoperative platelets count > 400.000

( da vs nu)

0,48 - - -

Peritoneal Cancer Index (> 20 vs < 20) <0,01 - - -

Anastomoses( yes vs no) 0,04 - - -

Anastomoses > 2 (yes vs no) <0,01 - -

Ttransfusions > 2 units ( yes vs no)

1 - - -

Transfusion ( yes vs no) 0,70 - - -

Duration of procedure > 600 minutea ( yes vs no)

0.23 - - -

Splenectomy (yes vs no) 0,06 - - -

Colectomy ( yes vs no) 0,09 - - -

Enterectomy ( yes vs no) 0,01 2,49 1,16-5,31 0,02

Histerectomy 0,14 - - -

Number of procedures > 8

(yes vs no)

0,31 - - -

Complete cytoreduction

(yes vs no)

0,46 - - -

CDDP intraperitoneal > 220 mg

( yes vs no)

<0,01 2,96 1,31-6,67 <0,01

TABEL 5.V Risk factors fost postoperative fistulas after HIPEC in PMP and colorectal carcinomatosis

patients

Risk factors Fistulae

Univariate

analysis*

Multivariate analysis**

p Odds

Ratio

Confidence interval P

(Colon vs PMP) 0,69 - - -

Sex (male vs female) 0,31 - - -

Age >52 (yes vs no) 0,59 - - -

BMI <22 ( yes vs no) 0,17 - - -

Performance status (0 vs 1-2) 0,046 - -

Charlson comorbidity index

> 3 (yes vs no)

0,94 - - -

(Colon + PMCA vs DPAM) 0,83 - - -

Prior surgical score 0-1 vs 2-3 0,21 - - -

Preoperative chemotherapy (da vs nu) 0,58 - - -

Preoperative albumin (<3,5 vs >3,5) 0,61 - -

Preoperative lymphocitic count < 1500

(da vs nu)

<0,01 3,90 1,45-10,46 <0,01

Preoperative platelets > 400.000

( da vs nu)

0,45 - - -

Peritoneal Cancer Index >20

(yes vs < no)

<0,01 4,57 1,40-14,56 0,01

Anastomoses ( yes vs no) 0,31 - - -

Anastomoses > 2 (yes vs no) <0,01 - - -

Intraoperative blood units > 2 unităti

( yes vs no)

1 - - -

Blood transfusion ( yes vs no) 0,70 - - -

Durata operaţiei > 600 minute ( da vs

nu)

0.23 - - -

Splenectomy (yes vs no) 0,08 - - -

Colectomy ( yes vs no) 0,09 - - -

Enterectomy (yes vs no) <0,01 2,94 1,18-7,34 0,02

Histerectomie (yes vs no) 0,14 - - -

Nomber of procedures

> 8 ( yes vs no)

0,14 - - -

CC score >1 (yes vs no) 0,09 - - -

Cisplatin dose > 220 mg

( yes vs no)

<0,05 - - -

5.4. Discussions

Univariate analysis showed the following factors: ECOG performance status,

preoperative lymphocyte count, Peritoneal Cancer Index> 20, enterectomy,

colectomy, splenectomy, CDDP dose and complete cytoreduction. After performing

multivariate analysis (logistic regression - backward conditional) only Peritoneal

Cancer Index >20, enterectomy and preoperative lymphocytic count remained as

independent factors. One explanation for Peritoneal Cancer could be : more

involvement of peritoneal cavity implies more procedures and thus higher risk for

bleeding, fistulas, anastomotic leak. Preoperative albumin, number of procedures,

perioperative blood transfusion were not amongst the independent factors.

CHAPTER 6. Circulating tumor markers: predictors of incomplete

cytoreduction and powerful determinants of outcome in pseudomyxoma

peritonei

6.1. Introduction Pseudomyxoma peritonei (PMP) is a rare condition characterized by

a slow accumulation of peritoneal deposits of mucinous tumors and mucin. With

systemic chemotherapy and palleative surgery overall survival is limited.

The aim of this study is to evaluate the ability of preoperative serum markers (Ca125,

CEA, and Ca19-9) to predict incomplete cytoreduction in patients PMP patients

elected eligible to cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy deemed for hyperthermic intraperitoneal chemotherapy associated with

peritonectomy.The secondary aim of this study is to evaluate their prognostic role for

oncologic outcome .

6.3. Results

Mean follow up was 42 months (1-170). 30 patients died (mean follow-up 31

months range 1-132. 34 patients were alive (mean follow-up 46 months range 13-

143). Five year overall survival was 73%. Mean age was 53 years (±13,6). There were

60 males and 96 females. Mean Charlson comorbidity index was 3,6 (±1).

Preoperative serum albumin was 4.3 g/dl (±0,7). PSS was 0 to1 in 47% patients and

was > 2 in 53% of patients. 23% patients underwent preoperative systemic

chemotherapy. 34% patients had PMCA/PMVA-I histologic type. Mean Peritoneal

Cancer Index was 22,6 (±11,1). Most frecquent procedure was omentectomy.

(TABLE 6.I) 14 patients (9%) underwent incomplete cytoreduction. Mean operative

time was 600 minutes (75-1260 minute). During the intervention were recquired 3

units of blood (range 0-37) Mean lenght of stay was 18 days (7-101). One patient

presented postoperative leukopenia ( TABLE 6.II)

6.4. Discussions

Univariate analysis revealed the following negative prognostic factors:

Charlson comorbidity index >4, male gender, ECOG performance status >2

preoperative systemic chemotherapy, histologic subtype PMCA, preoperative albumin

< 3,5mg/dl, Peritoneal Cancer Index >20, incomplete cytoreduction, Ca125>125

U/ml, Ca19,9 >89 U/ml. After multivariate analysis (multivariate logistic regression)

only the following factors remained: ECOG performance status, serum albumin < 3,5

mg/dl, Ca125 >125 and Ca19.9 > 89

TABLE 6.II.

Changes in laboratory values after HIPEC

Creatini >1,3mg/dl 36

Creatin > 3mg/dl 5

TGO 276 (38-1157)

TGP 311 (35-1503)

Total bilirubin 1 (0,4-4,49)

Amilase 239 (46-1226)

Leukopenia <3000 /mmc 1

Anemia <8 gHb/dl 31

Platelets count < 120.000mmc 9

TABLE 6.V

Risk factors for death in PMP patients after HIPEC Factors Univariate analysis

(Log-Rank)

Cox regression

p HR ( p

Gender (Male vs Female) 0,03 NS

Age ( > 52 vs 52) 0,8 -

Preoperative chemotherapy (Yes vs No)

0,03 NS

Histologic subtype ( DPAM vs

PMCA)

<0,01 NS

Serum albumin (≤3,5mg/dl vs >3,5

mg/dl)

<0,01 3,9 (1,45-10,49) <0,01

Ca 125 ( Ca125 >125U/ml vs ≤125

U/ml

<0,01 4,56 (1,98-10,51) <0,01

CA 19.9 ( CA19.9 > 89U/ml vs ≤

89 U/ml)

0,04 2,82 (1,17-6,71) 0,02

ACE ( > 18ng/dl vs ≤18 ng/dl) 0,35 -

Performance status ECOG (1-2 vs

0)

0,03 NS

Charlson Index ( >3 vs ≤3) 0,30 -

Preoperative platelets (>450.000 vs

≤ 450.000)

0,89 -

Prior Surgical Score ( 2-3 vs 0-1) 0,75 -

Peritoneal Cancer Index ( >20 vs ≤ 20)

<0,01 NS

No procedures ( >8 vs ≤ 8) 0,38 -

Cytoreduction (CC0-1 vs CC2-3)

<0,01 8,15 (2,73-23,86) <0,01

Postoperative morbidity (grade III-IV vs 0-II)

0,19 -

The following variables were proven to be independently associated with

shorter PFS: charlson comorbidity index >4, preoperative systemic chemotherapy,

incomplete cytoreduction, morbidity G3-5, and Ca125>125 U/ml. On the other hand,

variables independently associated with shorter OS were as follows: ECOG

performance status >2, serum preoperative albumin<3.5 g/dl, CC, Ca125>125U/ml,

and Ca19-9>89U/ml.

Neither the histological subtype nor the PCI>20 were proven to be

independent predictors of survival.

Despite presenting a reasonable discriminant power in predicting incomplete

cytoreductive surgery others clinicopathlogic data are more important. On the other

hand preoperative Ca125 and Ca19.9 were proven to be important oncological

prognostic markers of poor prognosis in operated PMP patients.

CHAPTER 7. Treatment of peritoneal carcinomatosis of gastro-intestinal

origin a retrospective study on 203 cases

7.1. Introduction.Aim of the study

Peritoneal carcinomatosis of gastrointestinal origin (PC-GI) is an advanced

digestive tumor and is found in 10-30% of patients (P) with primary surgery for

cancer (C) and up to 50% of C recurrences.

Patients with peritoneal carcinomatosis of gastrointestinal origyn have a very

poor prognosis. With systemic therapy the overall survival is limited to 3 to 9 months.

New drugs improved the survival of colorectal patients to 22 monts.

The aim of this studz is to evaluate the main characteristics, ethio-pathogenesis,

prognosis and imaging to track of P with PC-GI admitted to the Third Surgical Clinic,

"St. Spiridon" Hospital, Iaşi.

7.2. Material and methods

A retrospective study was carried out on series of 203 patients admitted in the

period June 2006 - March 2011. The patients were aged between 27-80 years (average

62), with a women/men ratio of 95/108. The duration of hospitalization was between 1

and 61 days, with an average of 13.5 days for emergency cases and 15 days for

elective cases. The data from observation files, the operating protocols, pathology

reports and follow-up files were collected and analyzed.

7.3. Results

136 patients were hospitalized with synchronous PC (the most common

gastric N = 60) and 67 with metachronous PC (the most common colon N = 29).

Imaging investigations consisted of ultrasound and computer tomography that showed

a sensibility and specificity of 80% and 73% respectively, mainly in regard to ascites

but less in assessing the presence of peritoneal deposits. The most common

complication was septic shock and mortality was 9.5% (17 patients). Average survival

was 5.7 months.

The ethiology of sincronous peritoneal carcinomatosis is summarised in TABLE 7.II

TABLE 7.II

Ethiology of syncronous carcinomatosis 136 patients with syncronous carcinomatosis

Stomac 60

Colon 31 Right 14

Trasverse 4

Left 13

Pancreas 19 Head 5

Body and tail 14

Rectum 8

Esophagus 6

Duodenum 2

Liver 4

Billiary tree 7

Small bowell 1

Others 6

The ethiology os metacronous carcinomatosis is summarised in TABLE 7.III 53

patients with metacronous carcinomatosis were operated

TABLE 7.III

Ethiology of metacronous carcinomatosis 67 patients metacronous carcinomatosis

Stomac 13

Colon 29 Right 10

Transverse 5

Left 14

Pancreas 7 Head 5

Body and tail 2

Rectum 8

Esophagus -

Duodenum 3

Liver -

Billiary tree -

Others 7

7.4. Discussions

Intraperitoneal normothermic chemotherapy was performed only in 5 patients

2 with right colon cancer with sincronous peritoneal carcinomatosis and 3 with

metactonous carcinomatosis). One patient presented a transitory renal failure. Two

patient were lost to follow-up and the other three survived between 6 to 9 months.

Conclusion

PC-GI is a disease with a poor prognosis, posing difficulties in early

diagnosis, establishing the surgical indication and protocol. Consistent advances in

systemic and locoregional chemotherapy, surgical techniques, intraoperative

radiotherapy, as well as immunotherapy are expected to improve prognosis.

CHAPTER 8. The role of perioperative systemic chemotherapy in diffuse malignant

peritoneal mesothelioma patients treated with cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy

8.1.Introduction. Aim of the study

Peritoneal mesothelioma is a rare disease with an incidence of 1-2 cases per

million in western countries.70% of all mesotheliomas arises from pleura, 20-25%

from peritoneum and in very rare cases the disease arises from pericardum or from

tunica vaginalis testis. It is a disease with a poor prognosis- overall survival with

systemic chemotherapy is one year [327]. There is no gold standard therapy for this

disease. Best systemic chemotherapy includes cisplatin and doxorubicin.

HIPEC has became in the last 30 years standard therapy for diffuse malignat

mesothelioma with 5 years survival exceeding 50% [328].

Despite good results this aggresive method is still performed in only a few

centers. High costs, high morbidity and mortality long learning curve are arguments

for the centralisation of this procedure[329] .

The aim of this study was to evaluate the effects of perioperative systemic

chemotherapy on short-term surgical and long-term oncologic results in diffuse

malignant peritoneal patients treated with HIPEC at National Cancer Institute of

Milan.

8.3. Results

There were 116 with diffuse malignant peritoneal mesotheliomapatients

operated between February 1995 and October 2011, 60 males and 56 females. Main

characteristics are summarized in TABLE 8.I. Main procedures during the

intervention are summarized in TABLE 8.II

Fig. 8.2 Diffuse malignant mesothelioma. Peritoneal Cancer Index 36. Operative time 870 minutes.

Collection of dr. Marcello Deraco

TABLE 8.I.

Clinicopathologic characteristics of DMPM patients

Characteristics

Agexx 58 (22-76)

Sex (m\f) 60\56

BMIxx 24 (15-56)

ECOG 0-1 vs >2 105\11

Hystological type epitelial \byphasic + sarcomatos 104\12

Albumin (g/dl)xx 4,03 (2,5-5,7)

Total Proteins (g/dl)xx 7,2 (5,7-8,8)

Preoperative platelets /mmcxx 373000 (87000-1400000)

Hospitalisationxx 25 (9-110)

Morbiditz grade III-Vx 47 ( 40,5%)

Mortality ( grade V)x 3 (2,5%)

Follow-up (months)xx 39 (1-119)

TABLE 8.IV.

Univariate and multivariate analysis of risk fators for severe morbidity and incomplete

cytoreduction in patients operated for diffuse malignant peritoneal mesothelioma Factor Morbidity (Grade III-V) Optimal cytoreduction vs incomplete

cytoreduction

Univariatex Multivariatexx Univariatex

Multivariatexx

p OR p p OR p

Age

0,53 0,47

Sex 0,28 0,57

ECOG 0,03 5,29

(1,27-22,6)

0,02 <0,01 4,25

(1,03-

17,47)

0,045

Charlson index (> 3 vs <4)

0,9 0,7

Prior surgical score ( > 2 vs < 2)

0,72 0,34

( epiteliod vs

biphazic/sarcomat

os)

0,71 0,07 NS

Platelet

preoperatice

>400.000

Yes vs no

0,39 0,08 NS

Preoperative chemotherapy (da

vs nu)

0,41 0,14

Peritoneal cancer

index > 20 (Yes vs

No)

0,06 NS 0,01 5,64

(1,45-

21,89)

0,1

Anastomosis (yes vs no)

0,02 3,49 (1,51-8,2)

<0,01 0,87

No. Procedures > 7

(Yes vs no)

0,01 4,25 (1,43-12,6)

<0,01 0,26

Optimal

cytoreduction (Yes vs No)

0,44

X – chi-square test/ Fisher exact test xx- multivariate logistic regression

8.4.Discussions

With the setting up of the division for peritoneal diseases at INT Milano,

patients form all Italy have been examined and treated.

After the finalizations of the investigations the patients are put on waiting

lists. The waiting time is approximately 6 months. Some of them have received

therapeutic schemes before arriving at INT and smaller percentage underwent

chemotherapy between first medical evaluation and the surgical intervention time.

The analysis of risk factors in the studied patient group for postoperative

complications and incomplete cytoreduction reveals together with the disease

extension through the peritoneum (Peritoneal Cancer index) the role of the ECOG

performance status.

The disease extension quantified by the peritoneal cancer index usually

translates in patients with high values, a diffuse involvement of the peritoneal cavity,

therefore the necessity of complex surgical gesture with the possibly of visceral

multiple resection. Other important complications are the prolonged intraoperative

time, intra and postoperative bleeding, fistula, due to intestinal serosa peritonectomy

or due to the necessity of anastomosis creation after intestinal or colic resection.

ECOG performance status role is most obvious for low performance status patients,

which means limited organism resources and deficient nutritional status.

On the studied group of patients preoperative chemotherapy did not influence

the optimal cytoreduction possibilities and post operative morbidity. Unlike the

ovarian cancer or the hepatic metastasis originating in colorectal cancer, where

neoadjuvant chemotherapy might lead to diminishing tumor dimensions and therefore

raise the cytoreduction possibilities, chemotherapy didn‘t influence the optimal

treatment possibilities. One of the explanations might be the relative chemoresistence

of PM.[336].

CHAPTER 9. Discussions and conclusions

9.1. Discussions

The survival of cancer patients already found in the metastatic phase has been

reported taking into consideration all types of metastasis. Therefore there is a lack of

data regarding the survival of the peritoneal metastatic disease patients. Tumor

biological variety and different evolutionary paths can lead to cerebral, hepatic,

pulmonary, bone, retroperitoneal and quite frequent peritoneal metastasis. Due to ease

of management the general therapeutic approach has been that of treating all these

entities as metastatic disease. In reality all these locations can have a different impact

over survival rate. The existence of 1 cm cerebral metastasis can significantly

influence the patient survival, sometimes limited to only a few weeks in the absence of

a surgical gesture. One, a few centimeters large, hepatic metastasis can evolve without

any clinical symptoms for several years. Due to intestinal lumen proximity PC can

also have an unforeseeable evolution. Occasionally a small singular nodule located on

the small bowel serosa can cause serious complications, whereas sometimes patients

can present multiple intraperitoneal nodules that do not concern the intestinal

peritoneum. Apparently these do not generate transit changes and sometimes even the

imagistic diagnosis is challenging.

There is no standard treatment for PC. It is a disease with a poor prognosis and for a

long time patients underwent treatment only for palliative purposes. Once new

chemotherapeutic agents were introduced the ovarian and colorectal PC prognosis has

been improved. In spite of a good initial response most patients present cancer

recurrence and five year survival cases are extremely rare. The idea of bringing

tumorous cells into contact with the chemotherapeutic agent had to undergo several

stages before being accepted by most surgeons. The first data concerning the

cytoreduction benefits in ovarian cancer drew interest for further research regarding

the PC‘s biology and made possible the development of new techniques designed to

achieve the maximum therapeutic benefits for patients [341]. Another goal of the

conducted studies has been indentifying patients that could best benefit from surgical

interventions.

Few I and II phase and only one III phase study that were carried out

according to Spratt intraperitoneal chemotherapy scheme in the case of a young

patient with PMP have shown obvious benefits as far as survival and life quality in

patients suffering from PC RC and PMP is concerned, using the complex procedure

that includes both cytoreduction and HIPEC. Intraoperative chemotherapy initially

normothermic and afterwards done hyperthermicly in association or not with

postoperative intaperitoneal chemotherapy has become a complex multidisciplinary

treatment method in PC. This change represents an alternative to systemic

chemotherapy used with palliative purposes.

Numerous retrospective studies have shown the advantages of the method for

PMP, peritoneal malignant mezothelioma, primary peritoneal tumors. Unfortunately

the complex treatment method has not yet been proven through prospective

randomized or multicentric studies. The majority of the studies are in reference to

personal experience which is rarely constituted by more than 100 cases of operated

patients. Furthermore most of them are not considered high reference points.

Wervaal and col‗s [119] randomized prospective study is the only EBM first

class study that has yet been published. Numerous systematic reviews [231,342] have

shown the method‘s advantages in comparison to systemic chemotherapy associated

with palliative surgical intervention.

In the retrospective study „Treatment of peritoneal carcinomatosis of gastro-

intestinal origin--a retrospective study of 203 cases” (Rev Med Chir Soc Med Nat

Iasi. 2012 Jan-Mar;116(1):150-6 PMID:23077888) [343] conducted at III Surgical

Clinic of Sf Spriridon Hospital Iaşi, data about morbidity and mortality has been

assessed after the admission of 203 patients over a period of 5 years. There have been

numerous problems in conducting the study. The patient lot has been heterogenic –

electively admitted patients or as emergencies with PC of different etiology but with

GC and RC predominance with or without preoperative chemotherapy. The

retrospective nature of the study didn‘t allow the evaluation of some clinical and

epidemiological factors such as nutritional and performance status, postoperative

complications in CTCAE classification and tumor markers.

The relatively small patient lot with a heterogenic pathology has not

permitted uni/multivariate analysis to identify the risk factors for postoperative

complications.

During hospitalization patients were not given life quality questionnaires.

Also the postoperative follow up has not been done actively therefore data about life

quality, undertaken chemotherapy and other possible surgeries, is missing. Patient or

family compliance to the sent questionnaires and also the difficulties in the

collaboration process with the Population Evidence Bureau have not allowed survival

estimation with Kaplan Meier curve.

Due to patient group heterogeneity and lacking data we could not make a

comparison with the witness group. In the absence of a regional or national register for

cancer patients and in the absence of a proactive systematic follow up, the

retrospective recuperation of data was deficitary.

One of the observations made after the study regarded the mortality during

patient hospital admission which was rather high 9,5% and did not include 30 or 60

days mortality according to recommendations for accurately done estimations over

postoperative mortality.

One of the reasons for high mortality could be the specific of the department

that has many emergencies cases. For occlusive RC and cephalic cancer the absence

of minimally invasive palliative treatment through stent implantation has probably

influenced the immediate survival of these patients.

Many of these patients underwent surgery regardless of the performance

status which may have lead to discrepancies between the high mortality rate and the

reported rate for ample interventions such as HIPEC.

The patient group cannot be compared to a witness group treated with

intaperitoneal chemotherapy because of data inconsistency due to the small number of

intraperitoneal chemotherapy conducted (5 patients).

During the time spent at the National Institute for Tumors in Milano as part

as the Doctoral scholarship I have conducted several studies, most of them

retrospective.

The Circulating tumor markers: Predictors of incomplete cytoreduction and

powerful determinants of outcome in pseudomyxoma peritonei”. [344] (J Surg Oncol.

2013 May 29. doi: 10.1002/jso.23329) evaluates the role of the tumoral markers in the

management of the patients that underwent surgery for PMP. There is little

mentioning in medical literature of the tumoral markers in PMP.

In 2007 Barrati and col. [345] published a retrospective study on a 62 patient

group that underwent surgery at INT underlining the preoperative role of Ca125 in

optimal cytoreduction possibility prediction and prognostic role of Ca 19.9. In the

present study there were included 156 patients with PMP treated at INT. With the help

of ROC curve –un useful instrument in evaluating the diagnosis capacity of a test we

have established threshold limits of the main tumor markers CEA ca 125 and Ca19.9.

The analysis on a larger group of patients highlighted the minor preoperative role of

tumoral markers in the prediction of optimal cytoreduction possibility.

The preoperative value of tumoral markers didn‘t significantly influence the

tumoral cytoreduction possibilities. In the group of patients with optimal

cytoreduction, a value of the Ca 125 and Ca 19.9 markers over the threshold limit

represented a negative prognosis factor with a precocious apparition of tumoral

recurrence and also a shorter survival rate. Even though the patient group is small and

patient subgroups with histological different subtypes are small, the study shows the

importance of tumoral markers in the oncologic prognosis of PMP patients operated

by HIPEC.

Even if some patients had an optimal cytoreduction they had tumoral

recurrence and a continuously evolving disease. One of the possibilities for future

research might be the role of neoadjuvant chemotherapy in patients with high levels of

tumor markers at the moment of diagnosis. The study could not follow the tumoral

markers dynamic through the pre and post operative period nor could it follow the

impact of perioperative chemotherapy in these patients.

Although MP is not a digestive disorder, this rare pathology is one of the

HIPEC beneficiaries. The low incidence of the disorder in the Western countries is

probably underrated due to the lack of diagnosis of all types of the disorder and to

confusing it with other disorders whose starting point are the abdominal parts.

Between 2003 and 2011, while I was working within the III Surgical Clinic of Sfantul

Spiridon Hospital of Iasi only 2 MP patients have been diagnosed and treated. This

slightly low number of patients treated in a surgical clinic with addressability from a

region with more than 4 million people contradicts the incidence of the illness for our

country. Considering that in real life a high number of MP patients receive an

inaccurate diagnosis, I was interested in this pathology during my external traineeship

with INT Milano. The department of peritoneum disorders within INT is a reference

centre in Italy for this disorder and probably one of the first four centres in the world

in terms of number of operated patients.

In the research ―The role of perioperative systemic chemotherapy in diffuse

malignant peritoneal mesothelioma patients treated with cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy‖ (Ann Surg Oncol, 2013 Apr; 20(4):1993-

100, two: 10.1245/s10434-012-2845-x) [2013] on a batch of 150 operated patients I

assessed the role of chemotherapy in MP patients subject to HIPEC. Considering that

the role of perioperative chemotherapy has not been studied before, the research data,

under the limitation caused by the low number of cases, showed a small impact of

chemotherapy on the survival rate of the patients, but there was highlighted a survival

rate of approximately 50% in patients with optimal cytoreduction. The low incidence

of the disorder in general population, namely 1-2 cases in one million of inhabitants,

should not be a counter-argument to the utility of setting up a HIPEC programme in

Romania. At least in theory, this disorder has an incidence that equals those in the

Western countries, where it is estimated that the incidence peak will be reached in

2030. In Romania, by dropping the use of asbestos materials later, the peak of this

incidence will probably occur after this year. Between the period May 2012 and

March 2013 in the I Surgical Clinic of the Regional Institute of Oncology Iasi 2 MP

patients were operated. A 70-year-old patient was investigated for undetermined

etiology ascites. The laparascopic examination diagnosis with biopsy of peritoneal

nodules raised the suspicion of CP with digestive, maybe pancreatic starting point.

Under the paliative treatment applied with Gemcitabina, the condition of the patient

improved after 2 chemotherapy sessions. A re-assessment of the biopsy parts revealed

suggestive aspects of peritoneal mesothelioma biphasic type. Three months after the

surgery, the patient was undergoing systemic chemotherapy. Another patient, aged 56

years, operated in another service for a right retroperitoneal tumour with

anatomopathologic examination suggested an extra digestive neuroendocrine tumour,

under chemotherapeutic treatment, has been assessed in the I Clinic for a relapse

retroperitoneal tumour. The patient was suggested for surgical intervention taking into

consideration the tumour cytoreduction in order to facilitate the systemic

chemotherapy. Due to the surgery, there were revealed a large retroperitoneal tumour

that was making common block with the ascending colon and the last ileal ansa, as

well as several peritoneal nodules located on the great epiploon and on the mesentery

serosa. It was applied an expanded right hemicolectomy and an epiplonectomy, and

the anatomopathologic examination revealed an epithelioid mesothelioma aspect with

a low degree of differentiation. Four months after the surgery, the patient undergoes

systemic chemotherapy.

In the 6th

chapter of the paper there were investigated the post-surgery

morbidity and mortality on the patients operated at the National Institute of Tumours

in Milano for CP of CR and PMP etiology. Although there had been concerns in the

CP treatment of gastric etiology, they were abandoned at the end of 2000 because of

the unsatisfactory results of the operated patients. Intraperitoneal chemotherapy for

gastric cancer should be reserved according to the opinion of the doctors responsible

of experimental trials within INT. Another suggestion of the European research is that

once CP occurs in a case of gastric cancer, chemotherapy would have limited benefits.

Intraperitoneal chemotherapy should be practised when there are no peritoneal

tumours, when there is no tumour residue, but patients have an increased risk of

relapsing with localisation in the peritoneum only (T3, T4, N+ tumours).

The two types of pathologies included in the research are chiefly different.

The PMP diagnosis is different from the CP CR one. On the other hand, within PMP

there are also histological subtypes with different diagnoses – PMCA has a reserved

diagnosis similar to the CP CR one. The two entities were included in the research

because the batches with CP CR or PMP patients were similar from point of view of

the clinical-epidemiological features, the number of intra-operative procedures being

similar in both cases. It would have been impossible to carry out a study only on the

batch of patients with CP CR because of the low number of patients.

Although it is a more complex intervention, the tumoral cytoreduction

associated to intraperitoneal chemotherapy for the batch of patients from INT has not

been followed in the postoperative period by increased morbidity or mortality. The

data is comparable to the data available in literature for HIPEC [233, 346-347], with

severe morbidity of 23% and intrahospital mortality of 3.5%. The multi-varied

analysis of the risk factors for the postoperative severe morbidity identified a factor

related to the patient and to the physician that carries out the selection for the patient –

ECOG performance status, a factor that is related to the extension of the peritoneal

affectation – the number of performed enterectomies and a physician-related factor –

the dosage of the chemotherapy used. The performance status during the preoperative

period is highly important since it probably reflects the duration of the illness

evolution, functional reserves and the ability of the patient to recover during the

postoperative period. It is arguable whether a patient who needs recovery in bed for

more than 12 hours or a patient who is incapable of taking care of himself can be

submitted to a large intervention. In relation to the number of anastomoses carried out

it is obvious in every surgical intervention that every anastomosis with various

localisations can increase the occurrence risk of the anastomotic fistula. There is not

much data known about the role of chemotherapy applied intraoperatively on

anastomoses. Research conducted on animals revealed a more difficult recovery of

anastomoses under hyperthermia [348]. All anastomoses are carried out at INT before

performing regular chemotherapy on the small intestine, generally mechanic suture in

cases of ileotransverse, colocolical and colorectal or ileorectal anastomosis, with

protective ileostomy in the last case.

In the research The Importance of the Learning Curve and Surveillance of

Surgical Performance in Peritoneal Surface Malignancy Programs (Surg Oncol Clin N

Am, 2012 Oct; 21 (4):559-76, two: 10.1016/j.soc.2012.07.011) [244] the assessment

was retrospectively on the cases operated within the National Institute for Tumours in

Milano between 1995 and 2011. The learning curve is a concept that appeared almost

30 years ago in medical practice and mainly refers to the stages necessary to reach

expertise in a field when starting from scratch. It is a rough guide, but it has a

subjectivity degree. It is hard to believe that a surgeon does only a certain type of

surgeries in order to gain experience. While earning experience, the surgeon assists

other colleagues as well or performs other interventions and thus he always makes

progress. There are few studies related to the learning curve in cytoreductive surgery.

Since in most retrospective cases the learning curve is assessed by dividing the batch

into periods or sub-batches selected arbitrarily, there can be discrepancies between the

capacity of the surgeon reflected on his position on the learning curve and the

postoperative results. The classical method of assessing the learning curve, by

dividing the patients into batches does not take into account other factors, such as

patient-dependent factors. The performance of the surgeon must also be adjusted on

the patient. The patients‘ selection is highly important and depends on the qualities of

the surgeon, but patients who could benefit from HIPEC should at least be assessed

for surgery. It is obvious that if a surgeon selected for a complex intervention only

young patients, with a high probability of favourable postoperative evolution, he

would probably reach the peak of the learning curve faster than a surgeon that would

select patients with increased probabilities of complications. A 70-year-old patient

with multiple co-morbidities that has a Peritoneal Cancer Index of 30 and might

undergo cytoreduction would need a complex intervention and would have a higher

probability of developing postoperative complications as compared to a 40-year-old

patient, who has not important co-morbidities and has a limited affectation of the

peritoneum. If a surgeon operates the 2 patients successively, the impact on the

surgeon‘s performance is different if patients have a postoperative severe

complication. For the patient with important co-morbidities the occurrence of a

postoperative complication reflects least the surgeon‘s performances and more his

performance status. On the same patient, the simple postoperative evolution is mainly

due to the surgeon‘s performances. The situation is rather opposite in the case of the

young patient. Therefore, in order to assess the performances of a surgeon in carrying

out an intervention the patient should also be taken into consideration. It is likewise

for other objectives aimed at during surgery (optimal cytoreduction etc.).

Severe morbidity was 29% in the study group. The mortality rate was 2.4%. RA-

SPRT curve showed a peak reaching the learning curve in the control of severe

postoperative complications after approximately 150 cases. For incomplete

cytoreduction learning curve peak is reached after 140 cases. The most common

disorders included in the study were PMP and MP. The RA-SPRT curve indicates an

improvement of performances after approximately 80 PMP cases operated and for

approximately 100 operated MP cases.

In the study "Learning Curve for cytoreductive Surgery and Hyperthermic

Intraperitoneal Chemotherapy in Peritoneal Surface malignancies: Analysis of Two

Centres (J Surg Oncol. 2013 Mar, 107 (4) :312-9. Doi: 10.1002/jso.23231. Epub 2012

August 23) [350]" we evaluated the mentor‘s role in the learning curve of a surgeon

who has initiated, under supervision, a treatment program for peritoneal diseases.

From 2003 to 2006 the main surgeon from INT oversaw the development of a new

center of peritonectomy at Bentivoglio Hospital from Bologna. Through repeated trips

from Bologna to INT and assisting the surgeon at INT, and then by initiating at

Bologna the peritonectomy program + HIPEC by the surgeon here, assisted in the

operations by the surgeon from INT, from 2003 to 2006 the Bologna center

peritonectomy was founded.

The goal of the study was to examine whether the support of the experienced

doctor influenced the surgeon's learning curve from the new center. 628 patients were

included in the study, operated at two hospitals: 67.4% at INT and 32.6% at the

Hospital in Bologna. With pre-operative risk factors for both groups of patients a

multivariate regression logistic pattern was created. With this pattern were then

evaluated and compared learning curves (RA-SPRT) for the two doctors - the

experienced and the initiated. After comparing the two curves RA-SPRT the

conclusion was that the initiated doctor has shortened the learning period with about

20 cases for optimal cytoreduction, severe morbidity and postoperative mortality. The

determining factor in this difference probably lies in the different ways of initiating a

HIPEC program. The initiated surgeon from Milan witnessed at the beginning of his

career a few interventions in the service where cytoreduction was sporadically used

and then visited the Cancer Institute in Washington, and then initiated the program

HIPEC unassisted by any experienced surgeon. The initiated surgeon after witnessing

a numerous interventions made in Milan was then assisted by an experienced surgeon.

The main conclusion is that in order to improve the results of HIPEC in the early

initiation of a program, the best solution is that the initiated surgeon is attended by an

experienced surgeon during his first operations. This is the only study that compares

the learning process through two different ways of initiation in the cytoreduction

tumors.

In Diffuse malignant peritoneal mesothelioma: Long-term survival with complete

cytoreductive surgery followed by hyperthermic intraperitoneal chemotherapy

(HIPEC)( Eur J Cancer. 2013 Jul 4. pii: S0959-8049(13)00435-8. doi:

10.1016/j.ejca.2013.05.027. [Epub ahead of print]) 108 patients were operated for

malignant peritoneal mesothelioma at INT where HIPEC was used. In the group of

patients studied postoperative mortality was 1.9%, and the severe morbidity was 39%.

Survival and disease free interval were 63 and 25 months. Survival reached a plateau

after about 84 months for 19 patients after that these patients were disease free.

Multivariate analysis revealed a direct correlation between survival and histological

subtype, nodal status, Ki67 positive presence in more than 10% of cells, and a

correlation between disease-free interval and the positive staining at podoplanin.

The absence of randomized multicentre prospective studies and of met analyses

is responsible for the skepticism that HIPEC is viewed. The rarity of PMP and MP are

making almost impossible the conduction of prospectively randomized studies. It

gradually imposed the necessity of specialized treatment centers [351]. The reason for

these centers is that more cases benefit the expertise of a surgical team involved in a

complex procedure.

In 2008, RC incidence (GLOBOCAN International Agency for Research on

Cancer) was approximately 1.2 million, representing 10% of all newly diagnosed

cancers. In Romania in the same year almost 9000 patients were diagnosed with RC.

Transposing Jayne's study results at Romanian's population results that about 1,000

patients are diagnosed with CP and of these, approximately 250 have limited HP.

They are a subgroup of patients in which systemic chemotherapy / possibly associated

with biological agents and HIPEC would make a superior survival rate compared to

other patients.

In an article published in 2003 Bayon and col [352] revealed several necessary

factors to create a HIPEC center : implementation in progressive stages under institute

tutelage with permanent evaluation and improvement of the human and material

resources, the creation of multidisciplinary support, training the involved staff and

starting a program through a feasibility study.

Technical difficulties and high costs are the reasons why the HIPEC technique is

used in a limited number of centers. It is almost impossible for a surgeon to reach the

peak of the learning curve after 10-20 cases. Probably the best option would be that

after being instructed in an experienced center to be then helped by doctors who have

reached the curve's peak and then to operate by themselves on the easier cases.

9.2 Conclusions

The classical surgical treatment of peritoneal carcinomatosis is difficult with

a high postoperative morbidity and mortality rate that goes up to 10% when unselected

patients are included.

Careful patient selection and treatment opportunities in specialized centers

provided by experienced physicians in peritoneum disease may limit morbidity and

mortality to 30 and respectively 3%

Common tumor markers play a limited part in patient selection for tumor

cytoreduction associated with HIPEC but are associated with negative prognosis when

high levels are reached in the preoperative stage.

Preoperative chemotherapy does not bring significant advantages over the

surgical approach in patients with peritoneal mezotelioma who are treated by

cytoreduction associated with HIPEC. A 50% survival rate after 5 years in patients

operated in reference centers validates the aggressive treatment protocol of this poor

prognostic malignancy.

The learning curve in cytoreductive surgery is a long one of over 130 cases

needed for efficient management in the selection, surgical treatment and postoperative

care of patients with peritoneal disease that require peritonectomy associated with

HIPEC.

This learning curve can be shortened with the help of a mentor that is initially

assisted and then assists the one that wishes to specialize in cytoreduction surgery.

Through the setting up of peritonectomy centers in Romania for a number of

patients with peritoneal carcinomatosis of peritoneal mesothelioma, apendicular and

colorectal etiology, the oncologic prognosis may be improved.

References

1. Sadeghi, B., et al., Peritoneal carcinomatosis from non-gynecologic malignancies: results of the EVOCAPE 1 multicentric prospective study. Cancer, 2000. 88(2): p. 358-63.

2. McCawley, L.J. and L.M. Matrisian, Tumor progression: defining the soil round the tumor seed.

Curr Biol, 2001. 11(1): p. R25-7. 3. Herrick, S.E. and S.E. Mutsaers, Mesothelial progenitor cells and their potential in tissue

engineering. Int J Biochem Cell Biol, 2004. 36(4): p. 621-42.

4. Yung, S., et al., Source of peritoneal proteoglycans. Human peritoneal mesothelial cells synthesize and secrete mainly small dermatan sulfate proteoglycans. American Journal of Pathology, 1995.

146(2): p. 520-9.

5. French, J.E., H.W. Florey, and B. Morris, The absorption of particles by the lymphatics of the diaphragm. Q J Exp Physiol Cogn Med Sci, 1960. 45: p. 88-103.

6. Negrini, D., et al., Distribution of diaphragmatic lymphatic stomata. J Appl Physiol, 1991. 70(4):

p. 1544-9. 7. Bettendorf, U., Electronmicroscopic studies on the peritoneal resorption of intraperitoneally

injected latex particles via the diaphragmatic lymphatics. Lymphology, 1979. 12(2): p. 66-70.

8. Shimotsuma, M., et al., Milky spots in the human greater omentum. Macroscopic and histological identification. Acta Anat (Basel), 1989. 136(3): p. 211-6.

9. Shimotsuma, M., et al., Morpho-physiological function and role of omental milky spots as

omentum-associated lymphoid tissue (OALT) in the peritoneal cavity. Lymphology, 1993. 26(2): p. 90-101. 10. Eggermont, A.M., E.P. Steller, and P.H. Sugarbaker, Laparotomy enhances intraperitoneal tumor

growth and abrogates the antitumor effects of interleukin-2 and lymphokine-activated killer cells. Surgery,

1987. 102(1): p. 71-8. 11. Schott, A., et al., Isolated tumor cells are frequently detectable in the peritoneal cavity of gastric

and colorectal cancer patients and serve as a new prognostic marker. Ann Surg, 1998. 227(3): p. 372-9. 12. Yonemura, Y., et al., E-cadherin and urokinase-type plasminogen activator tissue status in

gastric carcinoma. Cancer, 1995. 76(6): p. 941-53.

13. Pocard, M., et al., Single alteration of p53 or E-cadherin genes can alter the surgical resection benefit in an experimental model of colon cancer. Dis Colon Rectum, 2001. 44(8): p. 1106-12.

14. Elloul, S., et al., Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease

aggressiveness in metastatic ovarian and breast carcinoma. Cancer, 2005. 103(8): p. 1631-43. 15. Yonemura, Y., et al., Inverse expression of S100A4 and E-cadherin is associated with metastatic

potential in gastric cancer. Clin Cancer Res, 2000. 6(11): p. 4234-42.

16. Kanellos, I., et al., Incidence and prognostic value of positive peritoneal cytology in colorectal cancer. Dis Colon Rectum, 2003. 46(4): p. 535-9.

17. Gozalan, U., et al., Peritoneal cytology in colorectal cancer: incidence and prognostic value. Am

J Surg, 2007. 193(6): p. 672-5. 18. Nishikawa, T., et al., Prognostic value of peritoneal cytology and the combination of peritoneal

cytology and peritoneal dissemination in colorectal cancer. Dis Colon Rectum, 2009. 52(12): p. 2016-21.

19. Noura, S., et al., Long-term prognostic value of conventional peritoneal lavage cytology in patients undergoing curative colorectal cancer resection. Dis Colon Rectum, 2009. 52(7): p. 1312-20.

20. Kristensen, A.T., et al., Molecular detection (k-ras) of exfoliated tumour cells in the pelvis is a

prognostic factor after resection of rectal cancer? BMC Cancer, 2008. 8: p. 213. 21. van den Tol, P.M., et al., Reduction of peritoneal trauma by using nonsurgical gauze leads to less

implantation metastasis of spilled tumor cells. Ann Surg, 1998. 227(2): p. 242-8.

22. Zoetmulder, F.A., Cancer cell seeding during abdominal surgery: experimental studies. Cancer Treat Res, 1996. 82: p. 155-61.

23. Sugarbaker, P.H., Peritoneal carcinomatosis: natural history and rational therapeutic

interventions using intraperitoneal chemotherapy. Cancer Treat Res, 1996. 81: p. 149-68. 24. Jacquet, P. and P.H. Sugarbaker, Peritoneal-plasma barrier. Cancer Treat Res, 1996. 82: p. 53-

63.

25. Yonemura, Y., et al., A possible role of cytokines in the formation of peritoneal dissemination. Int J Oncol, 1997. 11(2): p. 349-58.

26. Sacchi, G., et al., Possible role of milky spots in mesothelial transplantation. Int J Artif Organs,

2007. 30(6): p. 520-6.

27. Sugarbaker, P.H., Observations concerning cancer spread within the peritoneal cavity and

concepts supporting an ordered pathophysiology. Cancer Treat Res, 1996. 82: p. 79-100. 28. Krause, J., et al., Ultrasonography findings and tumour quantification in patients with

pseudomyxoma peritonei. European Journal of Radiology, 2012. 81(4): p. 648-51.

29. Que, Y., et al., Ultrasound-guided biopsy of greater omentum: an effective method to trace the origin of unclear ascites. European Journal of Radiology, 2009. 70(2): p. 331-5.

30. Sulkin, T.V., et al., CT in pseudomyxoma peritonei: a review of 17 cases. Clin Radiol, 2002.

57(7): p. 608-13. 31. Bechtold, R.E., et al., CT appearance of disseminated peritoneal adenomucinosis. Abdom

Imaging, 2001. 26(4): p. 406-10.

32. Esquivel, J., et al., Accuracy and clinical relevance of computed tomography scan interpretation of peritoneal cancer index in colorectal cancer peritoneal carcinomatosis: a multi-institutional study. J Surg

Oncol, 2010. 102(6): p. 565-70.

33. Risson, J.R., et al., Cirrhotic and malignant ascites: differential CT diagnosis. Diagn Interv Imaging, 2012. 93(5): p. 365-70.

34. Koh, J.L., et al., Evaluation of preoperative computed tomography in estimating peritoneal

cancer index in colorectal peritoneal carcinomatosis. Ann Surg Oncol, 2009. 16(2): p. 327-33.

35. Alvarez Moreno, E., M. Jimenez de la Pena, and R. Cano Alonso, Role of New Functional MRI

Techniques in the Diagnosis, Staging, and Followup of Gynecological Cancer: Comparison with PET-CT.

Radiol Res Pract, 2012. 2012: p. 219546. 36. Kim, D.W., S.A. Park, and C.G. Kim, Detecting the recurrence of gastric cancer after curative

resection: comparison of FDG PET/CT and contrast-enhanced abdominal CT. J Korean Med Sci, 2011.

26(7): p. 875-80. 37. Sun, L., et al., Clinical value of F-FDG PET/CT in assessing suspicious relapse after rectal

cancer resection. World J Gastrointest Oncol, 2009. 1(1): p. 55-61.

38. Yang, Q.M., et al., The diagnostic value of PET-CT for peritoneal dissemination of abdominal malignancies. Gan To Kagaku Ryoho, 2006. 33(12): p. 1817-21.

39. Iversen, L.H., P.C. Rasmussen, and S. Laurberg, Value of laparoscopy before cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis. Br J Surg, 2013.

100(2): p. 285-92.

40. Gilly, F.N., et al., Regional chemotherapy (with mitomycin C) and intra-operative hyperthermia for digestive cancers with peritoneal carcinomatosis. Hepatogastroenterology, 1994. 41(2): p. 124-9.

41. Beaujard, A.C., et al., Intraperitoneal chemohyperthermia with mitomycin C for digestive tract

cancer patients with peritoneal carcinomatosis. Cancer, 2000. 88(11): p. 2512-9. 42. Harmon, R.L. and P.H. Sugarbaker, Prognostic indicators in peritoneal carcinomatosis from

gastrointestinal cancer. Int Semin Surg Oncol, 2005. 2(1): p. 3.

43. Jacquet, P. and P.H. Sugarbaker, Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res, 1996. 82: p. 359-74.

44. Elias, D., et al., Curative treatment of peritoneal carcinomatosis arising from colorectal cancer by

complete resection and intraperitoneal chemotherapy. Cancer, 2001. 92(1): p. 71-6. 45. Sugarbaker, P.H., Successful management of microscopic residual disease in large bowel cancer.

Cancer Chemother Pharmacol, 1999. 43 Suppl: p. S15-25.

46. Sugarbaker, P.H., Intraperitoneal chemotherapy and cytoreductive surgery for the prevention and treatment of peritoneal carcinomatosis and sarcomatosis. Semin Surg Oncol, 1998. 14(3): p. 254-61.

47. Fujimoto, S., et al., Improved mortality rate of gastric carcinoma patients with peritoneal

carcinomatosis treated with intraperitoneal hyperthermic chemoperfusion combined with surgery. Cancer,

1997. 79(5): p. 884-91.

48. Witkamp, A.J., et al., Extensive cytoreductive surgery followed by intra-operative hyperthermic

intraperitoneal chemotherapy with mitomycin-C in patients with peritoneal carcinomatosis of colorectal origin. Eur J Cancer, 2001. 37(8): p. 979-84.

49. Portilla, A.G., et al., The intraoperative staging systems in the management of peritoneal surface

malignancy. J Surg Oncol, 2008. 98(4): p. 228-31. 50. Boone, B.A., D.L. Bartlett, and A.H. Zureikat, Isolated hepatic perfusion for the treatment of

liver metastases. Curr Probl Cancer, 2012. 36(2): p. 27-76.

51. van Iersel, L.B., et al., Management of isolated nonresectable liver metastases in colorectal

cancer patients: a case-control study of isolated hepatic perfusion with melphalan versus systemic chemotherapy. Ann Oncol, 2010. 21(8): p. 1662-7.

52. Bhangu, A., et al., Outcomes of isolated limb perfusion in the treatment of extremity soft tissue

sarcoma: A systematic review. Eur J Surg Oncol, 2013. 53. Deroose, J.P., et al., Long-term outcome of isolated limb perfusion with tumour necrosis factor-

alpha for patients with melanoma in-transit metastases. Br J Surg, 2011. 98(11): p. 1573-80.

54. Bitterman, A., et al., Intraoperative radiotherapy for breast cancer. Isr Med Assoc J, 2012. 14(4): p. 256-9.

55. Elias, D., et al., Peritoneal colorectal carcinomatosis treated with surgery and perioperative

intraperitoneal chemotherapy: retrospective analysis of 523 patients from a multicentric French study. J Clin Oncol, 2010. 28(1): p. 63-8.

56. Mulier, S., et al., Survival Benefit of Adding Hyperthermic IntraPEritoneal Chemotherapy

(HIPEC) at the Different Time-points of Treatment of Ovarian Cancer: Review of Evidence. Curr Pharm Des, 2012. 18(25): p. 3793-803.

57. Glehen, O., et al., Peritoneal carcinomatosis from gastric cancer: a multi-institutional study of

159 patients treated by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy.

Ann Surg Oncol, 2010. 17(9): p. 2370-7.

58. Yang, X.J., et al., Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy

improves survival of patients with peritoneal carcinomatosis from gastric cancer: final results of a phase III randomized clinical trial. Ann Surg Oncol, 2011. 18(6): p. 1575-81.

59. Chua, T.C., et al., Early- and long-term outcome data of patients with pseudomyxoma peritonei

from appendiceal origin treated by a strategy of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. J Clin Oncol, 2012. 30(20): p. 2449-56.

60. Deraco, M., et al., Surgical technique of parietal and visceral peritonectomy for peritoneal

surface malignancies. J Surg Oncol, 2009. 100(4): p. 321-8. 61. Yan, T.D., et al., Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for

malignant peritoneal mesothelioma: multi-institutional experience. J Clin Oncol, 2009. 27(36): p. 6237-42. 62. Sticca, R.P. and B.W. Dach, Rationale for hyperthermia with intraoperative intraperitoneal

chemotherapy agents. Surg Oncol Clin N Am, 2003. 12(3): p. 689-701.

63. Song, C.W., J.G. Rhee, and S.H. Levitt, Blood flow in normal tissues and tumors during hyperthermia. J Natl Cancer Inst, 1980. 64(1): p. 119-24.

64. Overgaard, J., Effect of hyperthermia on malignant cells in vivo. A review and a hypothesis.

Cancer, 1977. 39(6): p. 2637-46. 65. Lepock, J.R., How do cells respond to their thermal environment? Int J Hyperthermia, 2005.

21(8): p. 681-7.

66. Takemoto, M., et al., The effect of various chemotherapeutic agents given with mild hyperthermia on different types of tumours. Int J Hyperthermia, 2003. 19(2): p. 193-203.

67. Tannock, I.F., et al., Limited penetration of anticancer drugs through tumor tissue: a potential

cause of resistance of solid tumors to chemotherapy. Clin Cancer Res, 2002. 8(3): p. 878-84. 68. Los, G., et al., Platinum distribution in intraperitoneal tumors after intraperitoneal cisplatin

treatment. Cancer Chemother Pharmacol, 1990. 25(6): p. 389-94.

69. González-Moreno, S., Advances in peritoneal surface oncology. Recent results in cancer research 169. 2007, Berlin ; New York: Springer. xiii, 176 p.

70. Weisberger, A.S., B. Levine, and J.P. Storaasli, Use of nitrogen mustard in treatment of serous

effusions of neoplastic origin. J Am Med Assoc, 1955. 159(18): p. 1704-7.

71. Cascales Campos, P.A., et al., Perioperative fast track program in intraoperative hyperthermic

intraperitoneal chemotherapy (HIPEC) after cytoreductive surgery in advanced ovarian cancer. Eur J Surg

Oncol, 2011. 37(6): p. 543-8. 72. Parson, E.N., et al., Outcomes after cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy for peritoneal surface dissemination from ovarian neoplasms. Am J Surg, 2011. 202(4): p.

481-6. 73. Yang, X.J., Y. Li, and Y. Yonemura, Cytoreductive surgery plus hyperthermic intraperitoneal

chemotherapy to treat gastric cancer with ascites and/or peritoneal carcinomatosis: Results from a Chinese

center. J Surg Oncol, 2010. 101(6): p. 457-64.

74. McQuellon, R. and K.E. Duckworth, Health-related quality of life and cytoreductive surgery

plus hyperthermic intraperitoneal chemotherapy. Curr Probl Cancer, 2009. 33(3): p. 203-18. 75. Shen, P., J.H.t. Stewart, and E.A. Levine, Cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy for peritoneal surface malignancy: overview and rationale. Curr Probl Cancer,

2009. 33(3): p. 125-41. 76. Gammon, D.C., et al., Cytoreductive surgery and intraperitoneal hyperthermic chemotherapy in

the treatment of peritoneal carcinomatosis. Am J Health Syst Pharm, 2009. 66(13): p. 1186-90.

77. Eveno, C., et al., [Treatment of peritoneal carcinomatosis with surgery and hyperthermic peroperative intraperitoneal chemotherapy (HIPEC): new aspects and validated indications]. Bull Cancer,

2008. 95(1): p. 141-5.

78. Capone, A., et al., Postoperative infections in cytoreductive surgery with hyperthermic intraperitoneal intraoperative chemotherapy for peritoneal carcinomatosis. J Surg Oncol, 2007. 96(6): p.

507-13.

79. Van der Speeten, K., O.A. Stuart, and P.H. Sugarbaker, Pharmacokinetics and pharmacodynamics of perioperative cancer chemotherapy in peritoneal surface malignancy. Cancer J, 2009.

15(3): p. 216-24.

80. Sugarbaker, P.H., Surgical responsibilities in the management of peritoneal carcinomatosis. J

Surg Oncol, 2010. 101(8): p. 713-24.

81. Sugarbaker, P.H. and K.A. Jablonski, Prognostic features of 51 colorectal and 130 appendiceal

cancer patients with peritoneal carcinomatosis treated by cytoreductive surgery and intraperitoneal chemotherapy. Ann Surg, 1995. 221(2): p. 124-32.

82. Glehen, O., et al., Cytoreductive surgery combined with perioperative intraperitoneal

chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multi-institutional study. J Clin Oncol, 2004. 22(16): p. 3284-92.

83. Glehen, O., et al., Surgery combined with peritonectomy procedures and intraperitoneal

chemohyperthermia in abdominal cancers with peritoneal carcinomatosis: a phase II study. J Clin Oncol, 2003. 21(5): p. 799-806.

84. Verwaal, V.J., et al., Predicting the survival of patients with peritoneal carcinomatosis of colorectal origin treated by aggressive cytoreduction and hyperthermic intraperitoneal chemotherapy. Br J

Surg, 2004. 91(6): p. 739-46.

85. Sugarbaker, P.H., W. Yu, and Y. Yonemura, Gastrectomy, peritonectomy, and perioperative intraperitoneal chemotherapy: the evolution of treatment strategies for advanced gastric cancer. Semin Surg

Oncol, 2003. 21(4): p. 233-48.

86. Stephens, A.D., et al., Morbidity and mortality analysis of 200 treatments with cytoreductive surgery and hyperthermic intraoperative intraperitoneal chemotherapy using the coliseum technique. Ann

Surg Oncol, 1999. 6(8): p. 790-6.

87. Stuart, O.A., et al., Safety monitoring of the coliseum technique for heated intraoperative intraperitoneal chemotherapy with mitomycin C. Ann Surg Oncol, 2002. 9(2): p. 186-91.

88. Jacquet, P., et al., Analysis of morbidity and mortality in 60 patients with peritoneal

carcinomatosis treated by cytoreductive surgery and heated intraoperative intraperitoneal chemotherapy. Cancer, 1996. 77(12): p. 2622-9.

89. Elias, D., et al., Research on the best chemohyperthermia technique of treatment of peritoneal

carcinomatosis after complete resection. Int J Surg Investig, 2000. 1(5): p. 431-9. 90. Flessner, M.F., Intraperitoneal drug therapy: physical and and biological principles. Cancer Treat

Res, 2007. 134: p. 131-52.

91. Sugarbaker, P.H., O.A. Stuart, and C.P. Carmignani, Pharmacokinetic changes induced by the

volume of chemotherapy solution in patients treated with hyperthermic intraperitoneal mitomycin C. Cancer

Chemother Pharmacol, 2006. 57(5): p. 703-8.

92. Elias, D.M. and L. Sideris, Pharmacokinetics of heated intraoperative intraperitoneal oxaliplatin after complete resection of peritoneal carcinomatosis. Surg Oncol Clin N Am, 2003. 12(3): p. 755-69, xiv.

93. Park, B.J., et al., Treatment of primary peritoneal mesothelioma by continuous hyperthermic

peritoneal perfusion (CHPP). Ann Surg Oncol, 1999. 6(6): p. 582-90. 94. van Ruth, S., V.J. Verwaal, and F.A. Zoetmulder, Pharmacokinetics of intraperitoneal mitomycin

C. Surg Oncol Clin N Am, 2003. 12(3): p. 771-80.

95. Sugarbaker, P.H., Early postoperative intraperitoneal adriamycin as an adjuvant treatment for advanced gastric cancer with lymph node or serosal invasion. Cancer Treat Res, 1991. 55: p. 277-284.

96. Elias, D., et al., Efficacy of intraperitoneal chemohyperthermia with oxaliplatin in colorectal

peritoneal carcinomatosis. Preliminary results in 24 patients. Ann Oncol, 2004. 15(5): p. 781-5. 97. Steller, M.A., et al., A pilot phase I trial of continuous hyperthermic peritoneal perfusion with

high-dose carboplatin as primary treatment of patients with small-volume residual ovarian cancer. Cancer

Chemother Pharmacol, 1999. 43(2): p. 106-14. 98. Speyer, J.L., et al., Phase I and pharmacological studies of 5-fluorouracil administered

intraperitoneally. Cancer Res, 1980. 40(3): p. 567-72.

99. Maeta, M., T. Sawata, and N. Kaibara, Effects of hyperthermia on the metabolism of 5-fluorouracil in vitro. Int J Hyperthermia, 1993. 9(1): p. 105-13.

100. Morgan, R.J., Jr., et al., Phase I trial of intraperitoneal docetaxel in the treatment of advanced

malignancies primarily confined to the peritoneal cavity: dose-limiting toxicity and pharmacokinetics. Clin Cancer Res, 2003. 9(16 Pt 1): p. 5896-901.

101. de Bree, E., et al., Intraoperative hyperthermic intraperitoneal chemotherapy with docetaxel as

second-line treatment for peritoneal carcinomatosis of gynaecological origin. Anticancer Res, 2003. 23(3C): p. 3019-27.

102. Mohamed, F. and P.H. Sugarbaker, Intraperitoneal taxanes. Surg Oncol Clin N Am, 2003. 12(3):

p. 825-33.

103. Yonemura, Y., et al., Effects of intraoperative chemohyperthermia in patients with gastric cancer

with peritoneal dissemination. Surgery, 1996. 119(4): p. 437-44.

104. Yonemura, Y., et al., Effective therapy for peritoneal dissemination in gastric cancer. Surg Oncol Clin N Am, 2003. 12(3): p. 635-48.

105. Bae, J.H., et al., Treatment of ovarian cancer with paclitaxel- or carboplatin-based intraperitoneal

hyperthermic chemotherapy during secondary surgery. Gynecol Oncol, 2007. 106(1): p. 193-200. 106. Kianmanesh, R., et al., Iterative cytoreductive surgery associated with hyperthermic

intraperitoneal chemotherapy for treatment of peritoneal carcinomatosis of colorectal origin with or without

liver metastases. Ann Surg, 2007. 245(4): p. 597-603. 107. Jayne, D.G., et al., Peritoneal carcinomatosis from colorectal cancer. Br J Surg, 2002. 89(12): p.

1545-50. 108. Elias, D., et al., Optimization of hyperthermic intraperitoneal chemotherapy with oxaliplatin plus

irinotecan at 43 degrees C after compete cytoreductive surgery: mortality and morbidity in 106 consecutive

patients. Ann Surg Oncol, 2007. 14(6): p. 1818-24. 109. Chu, D.Z., et al., Peritoneal carcinomatosis in nongynecologic malignancy. A prospective study

of prognostic factors. Cancer, 1989. 63(2): p. 364-7.

110. Jayne, D.G., et al., Peritoneal carcinomatosis from colorectal cancer. British Journal of Surgery, 2002. 89(12): p. 1545-1550.

111. Lemmens, V.E., et al., Predictors and survival of synchronous peritoneal carcinomatosis of

colorectal origin: a population-based study. Int J Cancer, 2011. 128(11): p. 2717-25. 112. Segelman, J., et al., Incidence, prevalence and risk factors for peritoneal carcinomatosis from

colorectal cancer. Br J Surg, 2012. 99(5): p. 699-705.

113. Gion, M., et al., How tumor markers are used in the routine follow-up of breast and colorectal cancer. A survey of 29 Italian hospitals. Int J Biol Markers, 1998. 13(3): p. 124-38.

114. Pelz, J.O., et al., Evaluation of best supportive care and systemic chemotherapy as treatment

stratified according to the retrospective peritoneal surface disease severity score (PSDSS) for peritoneal carcinomatosis of colorectal origin. BMC Cancer, 2010. 10: p. 689.

115. Franko, J., et al., Treatment of colorectal peritoneal carcinomatosis with systemic chemotherapy:

a pooled analysis of north central cancer treatment group phase III trials N9741 and N9841. J Clin Oncol,

2012. 30(3): p. 263-7.

116. Goldberg, R.M., et al., NCCTG Study N9741: leveraging learning from an NCI Cooperative

Group phase III trial. Oncologist, 2009. 14(10): p. 970-8. 117. Zani, S., et al., Modest advances in survival for patients with colorectal-associated peritoneal

carcinomatosis in the era of modern chemotherapy. J Surg Oncol, 2012.

118. Klaver, Y.L., et al., Addition of Biological Therapies to Palliative Chemotherapy Prolongs Survival in Patients With Peritoneal Carcinomatosis of Colorectal Origin. Am J Clin Oncol, 2012.

119. Verwaal, V.J., et al., Randomized trial of cytoreduction and hyperthermic intraperitoneal

chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol, 2003. 21(20): p. 3737-43.

120. Markman, M., Hyperthermic intraperitoneal chemotherapy in the management of ovarian cancer:

A critical need for an evidence-based evaluation. Gynecol Oncol, 2009. 113(1): p. 4-5. 121. De Simone, M., et al., Treatment of pseudomyxoma peritonei with two times--cytoreduction and

hypertermic antiblastic peritoneal perfusion (HAPP). J Exp Clin Cancer Res, 2003. 22(4 Suppl): p. 25-8.

122. Glehen, O., et al., Toward curative treatment of peritoneal carcinomatosis from nonovarian origin by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy: a multi-

institutional study of 1,290 patients. Cancer, 2010. 116(24): p. 5608-18.

123. Perez, J., et al., [Anesthetic management for scheduled peritonectomy and hyperthermic intraperitoneal chemotherapy in 20 patients]. Rev Esp Anestesiol Reanim, 2006. 53(9): p. 550-5.

124. Saegesser, F., [Treatment of peritoneal pseudomyxoma due to the rupture of an appendicular

mucocele]. Lyon Chir, 1965. 61(4): p. 535-8. 125. Elias, D., et al., Results of systematic second-look surgery plus HIPEC in asymptomatic patients

presenting a high risk of developing colorectal peritoneal carcinomatosis. Ann Surg, 2011. 254(2): p. 289-

93. 126. Taniguchi, M., et al., [Experimental study of intraperitoneal hyperthermic chemotherapy]. Gan

To Kagaku Ryoho, 1998. 25(7): p. 1083-6.

127. Cavaliere, F., et al., Prognostic factors and oncologic outcome in 146 patients with colorectal

peritoneal carcinomatosis treated with cytoreductive surgery combined with hyperthermic intraperitoneal

chemotherapy: Italian multicenter study S.I.T.I.L.O. Eur J Surg Oncol, 2011. 37(2): p. 148-54.

128. Bretcha-Boix, P., et al., Cytoreductive surgery and perioperative intraperitoneal chemotherapy in patients with peritoneal carcinomatosis of colonic origin: outcomes after 7 years' experience of a new centre

for peritoneal surface malignancies. Clin Transl Oncol, 2010. 12(6): p. 437-42.

129. Vaira, M., et al., Treatment of peritoneal carcinomatosis from colonic cancer by cytoreduction, peritonectomy and hyperthermic intraperitoneal chemotherapy (HIPEC). Experience of ten years. In Vivo,

2010. 24(1): p. 79-84.

130. Scaringi, S., et al., The role of cytoreductive surgery alone for the treatment of peritoneal carcinomatosis of colorectal origin. A retrospective analysis with regard to multimodal treatments.

Hepatogastroenterology, 2009. 56(91-92): p. 650-5. 131. Shen, P., et al., Cytoreductive surgery and intraperitoneal hyperthermic chemotherapy with

mitomycin C for peritoneal carcinomatosis from nonappendiceal colorectal carcinoma. Ann Surg Oncol,

2004. 11(2): p. 178-86. 132. Elias, D., et al., Complete cytoreductive surgery plus intraperitoneal chemohyperthermia with

oxaliplatin for peritoneal carcinomatosis of colorectal origin. J Clin Oncol, 2009. 27(5): p. 681-5.

133. Hagendoorn, J., et al., Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis from colorectal and gastrointestinal origin shows acceptable morbidity and high

survival. Eur J Surg Oncol, 2009. 35(8): p. 833-7.

134. Yan, T.D. and D.L. Morris, Cytoreductive surgery and perioperative intraperitoneal chemotherapy for isolated colorectal peritoneal carcinomatosis: experimental therapy or standard of care?

Ann Surg, 2008. 248(5): p. 829-35.

135. Yonemura, Y., E. Canbay, and H. Ishibashi, Prognostic Factors of Peritoneal Metastases from Colorectal Cancer following Cytoreductive Surgery and Perioperative Chemotherapy.

ScientificWorldJournal, 2013. 2013: p. 978394.

136. Turrini, O., et al., Initial experience with hyperthermic intraperitoneal chemotherapy. Arch Surg, 2012. 147(10): p. 919-23.

137. Cashin, P.H., et al., Cytoreductive surgery and intraperitoneal chemotherapy for colorectal

peritoneal carcinomatosis: prognosis and treatment of recurrences in a cohort study. Eur J Surg Oncol, 2012.

38(6): p. 509-15.

138. Hompes, D., et al., The treatment of peritoneal carcinomatosis of colorectal cancer with

complete cytoreductive surgery and hyperthermic intraperitoneal peroperative chemotherapy (HIPEC) with oxaliplatin: a Belgian multicentre prospective phase II clinical study. Ann Surg Oncol, 2012. 19(7): p. 2186-

94.

139. Chua, T.C., et al., Peritoneal carcinomatosis and liver metastases from colorectal cancer treated with cytoreductive surgery perioperative intraperitoneal chemotherapy and liver resection. Eur J Surg

Oncol, 2009. 35(12): p. 1299-305.

140. Elias, D., et al., Treatment of synchronous peritoneal carcinomatosis and liver metastases from colorectal cancer. Eur J Surg Oncol, 2006. 32(6): p. 632-6.

141. Maggiori, L., et al., Should Patients With Peritoneal Carcinomatosis of Colorectal Origin With

Synchronous Liver Metastases Be Treated With a Curative Intent?: A Case-Control Study. Ann Surg, 2012. 142. Liberale, G., et al., Recommendations for general surgeons facing incidental peritoneal

carcinomatosis of colorectal origin. Eur J Surg Oncol, 2008. 34(7): p. 725-6.

143. Look, M., D. Chang, and P.H. Sugarbaker, Long-term results of cytoreductive surgery for advanced and recurrent epithelial ovarian cancers and papillary serous carcinoma of the peritoneum. Int J

Gynecol Cancer, 2004. 14(1): p. 35-41.

144. Matsuda, K., et al., Clinical impact of a macroscopically complete resection of colorectal cancer with peritoneal carcinomatosis. Surgery, 2012. 151(2): p. 238-44.

145. Verwaal, V.J. and F.A. Zoetmulder, Follow-up of patients treated by cytoreduction and

chemotherapy for peritoneal carcinomatosis of colorectal origin. Eur J Surg Oncol, 2004. 30(3): p. 280-5. 146. Sugarbaker, P.H., et al., A simplified plan for follow-up of patients with colon and rectal cancer

supported by prospective studies of laboratory and radiologic test results. Surgery, 1987. 102(1): p. 79-87.

147. Yonemura, Y., et al., Safety and efficacy of bidirectional chemotherapy for treatment of patients with peritoneal dissemination from gastric cancer: Selection for cytoreductive surgery. J Surg Oncol, 2009.

100(4): p. 311-6.

148. Juhl, H., et al., Immunocytological detection of micrometastatic cells: comparative evaluation of

findings in the peritoneal cavity and the bone marrow of gastric, colorectal and pancreatic cancer patients.

Int J Cancer, 1994. 57(3): p. 330-5.

149. Yonemura, Y., et al., The natural history of free cancer cells in the peritoneal cavity. Recent Results Cancer Res, 2007. 169: p. 11-23.

150. Bamias, A. and N. Pavlidis, Systemic Chemotherapy in Gastric Cancer: Where Do We Stand

Today? Oncologist, 1998. 3(3): p. 171-177. 151. Viudez-Berral, A., et al., Current management of gastric cancer. Rev Esp Enferm Dig, 2012.

104(3): p. 134-41.

152. Fujimoto, S., et al., Intraperitoneal hyperthermic perfusion combined with surgery effective for gastric cancer patients with peritoneal seeding. Ann Surg, 1988. 208(1): p. 36-41.

153. Fujimoto, S., et al., Positive results of combined therapy of surgery and intraperitoneal hyperthermic perfusion for far-advanced gastric cancer. Ann Surg, 1990. 212(5): p. 592-6.

154. Yonemura, Y., et al., Hyperthermo-chemotherapy combined with cytoreductive surgery for the

treatment of gastric cancer with peritoneal dissemination. World J Surg, 1991. 15(4): p. 530-5; discussion 535-6.

155. Yonemura, Y., et al., Prophylaxis with intraoperative chemohyperthermia against peritoneal

recurrence of serosal invasion-positive gastric cancer. World J Surg, 1995. 19(3): p. 450-4; discussion 455. 156. Hirose, K., et al., Efficacy of continuous hyperthermic peritoneal perfusion for the prophylaxis

and treatment of peritoneal metastasis of advanced gastric cancer: evaluation by multivariate regression

analysis. Oncology, 1999. 57(2): p. 106-14. 157. Glehen, O., et al., Cytoreductive surgery and intraperitoneal chemohyperthermia for peritoneal

carcinomatosis arising from gastric cancer. Arch Surg, 2004. 139(1): p. 20-6.

158. Wu, X.J., et al., Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves the survival of gastric cancer patients with ovarian metastasis and peritoneal dissemination. Tumour Biol,

2013. 34(1): p. 463-9.

159. Strohlein, M.A., D.R. Bulian, and M.M. Heiss, Clinical efficacy of cytoreductive surgery and hyperthermic chemotherapy in peritoneal carcinomatosis from gastric cancer. Expert Rev Anticancer Ther,

2011. 11(10): p. 1505-8.

160. Scaringi, S., et al., Advanced gastric cancer with or without peritoneal carcinomatosis treated

with hyperthermic intraperitoneal chemotherapy: a single western center experience. Eur J Surg Oncol,

2008. 34(11): p. 1246-52.

161. De Roover, A., et al., Adjuvant hyperthermic intraperitoneal peroperative chemotherapy (HIPEC) associated with curative surgery for locally advanced gastric carcinoma. An initial experience.

Acta Chir Belg, 2006. 106(3): p. 297-301.

162. Zhibing, W., et al., Clinical study of cisplatin hyperthermic intraperitoneal perfusion chemotherapy in combination with docetaxel, 5-flourouracil and leucovorin intravenous chemotherapy for

the treatment of advanced-stage gastric carcinoma. Hepatogastroenterology, 2013. 60(128).

163. Yonemura, Y., et al., Intraoperative chemohyperthermic peritoneal perfusion as an adjuvant to

gastric cancer: final results of a randomized controlled study. Hepatogastroenterology, 2001. 48(42): p. 1776-82.

164. Fujimoto, S., et al., Successful intraperitoneal hyperthermic chemoperfusion for the prevention

of postoperative peritoneal recurrence in patients with advanced gastric carcinoma. Cancer, 1999. 85(3): p. 529-34.

165. Landis, S.H., et al., Cancer statistics, 1999. CA Cancer J Clin, 1999. 49(1): p. 8-31, 1.

166. Heintz, A.P., et al., Carcinoma of the ovary. Int J Gynaecol Obstet, 2003. 83 Suppl 1: p. 135-66. 167. Rubin, S.C., et al., Ten-year follow-up of ovarian cancer patients after second-look laparotomy

with negative findings. Obstet Gynecol, 1999. 93(1): p. 21-4.

168. Griffiths, C.T., Surgical resection of tumor bulk in the primary treatment of ovarian carcinoma. Natl Cancer Inst Monogr, 1975. 42: p. 101-4.

169. Bristow, R.E., et al., Survival effect of maximal cytoreductive surgery for advanced ovarian

carcinoma during the platinum era: a meta-analysis. J Clin Oncol, 2002. 20(5): p. 1248-59. 170. Alberts, D.S., et al., Intraperitoneal cisplatin plus intravenous cyclophosphamide versus

intravenous cisplatin plus intravenous cyclophosphamide for stage III ovarian cancer. N Engl J Med, 1996.

335(26): p. 1950-5.

171. Markman, M., et al., Phase III trial of standard-dose intravenous cisplatin plus paclitaxel versus

moderately high-dose carboplatin followed by intravenous paclitaxel and intraperitoneal cisplatin in small-

volume stage III ovarian carcinoma: an intergroup study of the Gynecologic Oncology Group, Southwestern Oncology Group, and Eastern Cooperative Oncology Group. J Clin Oncol, 2001. 19(4): p. 1001-7.

172. Helm, C.W., The role of hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer.

Oncologist, 2009. 14(7): p. 683-94. 173. Yoshida, Y., et al., Efficacy of intraperitoneal continuous hyperthermic chemotherapy as

consolidation therapy in patients with advanced epithelial ovarian cancer: a long-term follow-up. Oncol

Rep, 2005. 13(1): p. 121-5. 174. de Bree, E., et al., Cytoreductive surgery and intraoperative hyperthermic intraperitoneal

chemotherapy with paclitaxel: a clinical and pharmacokinetic study. Ann Surg Oncol, 2008. 15(4): p. 1183-92.

175. Ryu, K.S., et al., Effects of intraperitoneal hyperthermic chemotherapy in ovarian cancer.

Gynecol Oncol, 2004. 94(2): p. 325-32. 176. Argenta, P.A., et al., Hyperthermic intraperitoneal chemotherapy with carboplatin for optimally-

cytoreduced, recurrent, platinum-sensitive ovarian carcinoma: a pilot study. Gynecol Oncol, 2013. 129(1):

p. 81-5. 177. Bakrin, N., et al., Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy

(HIPEC) for persistent and recurrent advanced ovarian carcinoma: a multicenter, prospective study of 246

patients. Ann Surg Oncol, 2012. 19(13): p. 4052-8. 178. Ansaloni, L., et al., Evaluation of extensive cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy (HIPEC) in patients with advanced epithelial ovarian cancer. Int J Gynecol

Cancer, 2012. 22(5): p. 778-85. 179. Deraco, M., et al., Secondary cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy for recurrent epithelial ovarian cancer: a multi-institutional study. BJOG, 2012. 119(7): p.

800-9. 180. Tentes, A.A., et al., Cytoreductive surgery combined with hyperthermic intraperitoneal

intraoperative chemotherapy in the treatment of advanced epithelial ovarian cancer. J Oncol, 2012. 2012: p.

358341.

181. Spiliotis, J., et al., The role of cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy in the management of recurrent advanced ovarian cancer: a prospective study. J BUON,

2011. 16(1): p. 74-9. 182. Melis, A., et al., Reappraisal of the role of hyperthermic intraperitoneal chemotherapy (HIPEC)

in the management of ovarian cancer: a single institutional experience. Bull Cancer, 2011.

183. Fagotti, A., et al., HIPEC in recurrent ovarian cancer patients: morbidity-related treatment and long-term analysis of clinical outcome. Gynecol Oncol, 2011. 122(2): p. 221-5.

184. Frenel, J.S., et al., Oxaliplatin-based hyperthermic intraperitoneal chemotherapy in primary or

recurrent epithelial ovarian cancer: A pilot study of 31 patients. J Surg Oncol, 2011. 103(1): p. 10-6.

185. Fujii, M., et al., [Two cases of intussusception of appendiceal mucocele: diagnostic value of

preoperative ultrasonography]. Nihon Geka Gakkai Zasshi, 1986. 87(7): p. 808-12. 186. Cheng, K.K., An experimental study of mucocele of the appendix and pseudomyxoma peritonei.

J Pathol Bacteriol, 1949. 61(2): p. 217-25, 4 pl.

187. Smeenk, R.M., et al., Appendiceal neoplasms and pseudomyxoma peritonei: a population based study. Eur J Surg Oncol, 2008. 34(2): p. 196-201.

188. Lyss, A.P., Appendiceal malignancies. Semin Oncol, 1988. 15(2): p. 129-37.

189. Misdraji, J., et al., Appendiceal mucinous neoplasms: a clinicopathologic analysis of 107 cases. Am J Surg Pathol, 2003. 27(8): p. 1089-103.

190. Ronnett, B.M., et al., Disseminated peritoneal adenomucinosis and peritoneal mucinous

carcinomatosis. 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(12): p. 1390-408.

191. Bradley, R.F., et al., Pseudomyxoma peritonei of appendiceal origin: a clinicopathologic analysis of 101 patients uniformly treated at a single institution, with literature review. Am J Surg Pathol, 2006.

30(5): p. 551-9.

192. Misdraji, J., Appendiceal mucinous neoplasms: controversial issues. Arch Pathol Lab Med,

2010. 134(6): p. 864-70.

193. Guerrieri, C., et al., Mucinous tumors of the vermiform appendix and ovary, and pseudomyxoma

peritonei: histogenetic implications of cytokeratin 7 expression. Hum Pathol, 1997. 28(9): p. 1039-45. 194. Szych, C., et al., Molecular genetic evidence supporting the clonality and appendiceal origin of

Pseudomyxoma peritonei in women. Am J Pathol, 1999. 154(6): p. 1849-55.

195. Esquivel, J. and P.H. Sugarbaker, Clinical presentation of the Pseudomyxoma peritonei syndrome. Br J Surg, 2000. 87(10): p. 1414-8.

196. Gough, D.B., et al., Pseudomyxoma peritonei. Long-term patient survival with an aggressive

regional approach. Ann Surg, 1994. 219(2): p. 112-9. 197. Jarvinen, P., H.J. Jarvinen, and A. Lepisto, Survival of patients with pseudomyxoma peritonei

treated by serial debulking. Colorectal Dis, 2010. 12(9): p. 868-72. 198. Sugarbaker, P.H. and D. Chang, Results of treatment of 385 patients with peritoneal surface

spread of appendiceal malignancy. Ann Surg Oncol, 1999. 6(8): p. 727-31.

199. Sorensen, O., et al., Evaluation of complete cytoreductive surgery and two intraperitoneal chemotherapy techniques in Pseudomyxoma peritonei. Eur J Surg Oncol, 2012. 38(10): p. 969-76.

200. Arjona-Sanchez, A., et al., Outcome of patients with aggressive pseudomyxoma peritonei treated

by cytoreductive surgery and intraperitoneal chemotherapy. World J Surg, 2013. 37(6): p. 1263-70. 201. Arjona-Sanchez, A., et al., Pseudomyxoma peritonei treated by cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy: results from a single centre. Clin Transl Oncol, 2011. 13(4): p.

261-7. 202. Guner, Z., et al., Cytoreductive surgery and intraperitoneal chemotherapy for pseudomyxoma

peritonei. Int J Colorectal Dis, 2005. 20(2): p. 155-60.

203. Smeenk, R.M., et al., Survival analysis of pseudomyxoma peritonei patients treated by cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg, 2007. 245(1): p. 104-9.

204. Murphy, E.M., R. Sexton, and B.J. Moran, Early results of surgery in 123 patients with

pseudomyxoma peritonei from a perforated appendiceal neoplasm. Dis Colon Rectum, 2007. 50(1): p. 37-42.

205. Youssef, H., et al., Operative findings, early complications, and long-term survival in 456

patients with pseudomyxoma peritonei syndrome of appendiceal origin. Dis Colon Rectum, 2011. 54(3): p.

293-9.

206. Attanoos, R.L. and A.R. Gibbs, The pathology associated with therapeutic procedures in

malignant mesothelioma. Histopathology, 2004. 45(4): p. 393-7. 207. Antman, K.H., et al., Peritoneal mesothelioma: natural history and response to chemotherapy. J

Clin Oncol, 1983. 1(6): p. 386-91.

208. Bridda, A., et al., Peritoneal mesothelioma: a review. MedGenMed, 2007. 9(2): p. 32. 209. Acherman, Y.I., et al., Clinical presentation of peritoneal mesothelioma. Tumori, 2003. 89(3): p.

269-73.

210. Baratti, D., et al., Circulating CA125 in patients with peritoneal mesothelioma treated with cytoreductive surgery and intraperitoneal hyperthermic perfusion. Ann Surg Oncol, 2007. 14(2): p. 500-8.

211. de Pangher Manzini, V., Malignant peritoneal mesothelioma. Tumori, 2005. 91(1): p. 1-5.

212. Chua, T.C., T.D. Yan, and D.L. Morris, Surgical biology for the clinician: peritoneal mesothelioma: current understanding and management. Can J Surg, 2009. 52(1): p. 59-64.

213. Deraco, M., et al., The role of perioperative systemic chemotherapy in diffuse malignant

peritoneal mesothelioma patients treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg Oncol, 2013. 20(4): p. 1093-100.

214. Yan, T.D., et al., A novel tumor-node-metastasis (TNM) staging system of diffuse malignant

peritoneal mesothelioma using outcome analysis of a multi-institutional database*. Cancer, 2011. 117(9): p. 1855-63.

215. Alexander, H.R., Jr., et al., Treatment factors associated with long-term survival after

cytoreductive surgery and regional chemotherapy for patients with malignant peritoneal mesothelioma. Surgery, 2013. 153(6): p. 779-86.

216. Baratti, D., et al., Cytoreductive surgery with selective versus complete parietal peritonectomy

followed by hyperthermic intraperitoneal chemotherapy in patients with diffuse malignant peritoneal mesothelioma: a controlled study. Ann Surg Oncol, 2012. 19(5): p. 1416-24.

217. Baratti, D., et al., Lymph node metastases in diffuse malignant peritoneal mesothelioma. Ann

Surg Oncol, 2010. 17(1): p. 45-53.

218. Passot, G., et al., [Peritoneal mesothelioma: treatment with cytoreductive surgery combined with

hyperthermic intraperitoneal chemotherapy]. J Chir (Paris), 2008. 145(5): p. 447-53.

219. Chua, T.C., T.D. Yan, and D.L. Morris, Outcomes of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for peritoneal mesothelioma: the Australian experience. J Surg Oncol, 2009.

99(2): p. 109-13.

220. Brigand, C., et al., Peritoneal mesothelioma treated by cytoreductive surgery and intraperitoneal hyperthermic chemotherapy: results of a prospective study. Ann Surg Oncol, 2006. 13(3): p. 405-12.

221. Schaub, N.P., et al., A novel nomogram for peritoneal mesothelioma predicts survival. Ann Surg

Oncol, 2013. 20(2): p. 555-61. 222. Esquivel, J., et al., Consensus statement on the loco regional treatment of colorectal cancer with

peritoneal dissemination. J Surg Oncol, 2008. 98(4): p. 263-7. 223. Smeenk, R.M., V.J. Verwaal, and F.A. Zoetmulder, Learning curve of combined modality

treatment in peritoneal surface disease. Br J Surg, 2007. 94(11): p. 1408-14.

224. Piso, P., et al., Patient selection for a curative approach to carcinomatosis. Cancer J, 2009. 15(3): p. 236-42.

225. Bozzetti, F., et al., Locoregional treatment of peritoneal carcinomatosis from gastric cancer. J

Surg Oncol, 2008. 98(4): p. 273-6. 226. Deraco, M., et al., Cytoreductive surgery followed by intraperitoneal hyperthermic perfusion in

the treatment of recurrent epithelial ovarian cancer: a phase II clinical study. Tumori, 2001. 87(3): p. 120-6.

227. Helm, C.W., et al., Hyperthermic intraperitoneal chemotherapy with and without cytoreductive surgery for epithelial ovarian cancer. J Surg Oncol, 2008. 98(4): p. 283-90.

228. Chua, T.C., et al., Should the treatment of peritoneal carcinomatosis by cytoreductive surgery

and hyperthermic intraperitoneal chemotherapy still be regarded as a highly morbid procedure?: a systematic review of morbidity and mortality. Ann Surg, 2009. 249(6): p. 900-7.

229. Younan, R., et al., Morbidity, toxicity, and mortality classification systems in the local regional

treatment of peritoneal surface malignancy. J Surg Oncol, 2008. 98(4): p. 253-7. 230. Trotti, A., et al., CTCAE v3.0: development of a comprehensive grading system for the adverse

effects of cancer treatment. Semin Radiat Oncol, 2003. 13(3): p. 176-81.

231. Gill, R.S., et al., Treatment of gastric cancer with peritoneal carcinomatosis by cytoreductive

surgery and HIPEC: a systematic review of survival, mortality, and morbidity. J Surg Oncol, 2011. 104(6):

p. 692-8.

232. Deraco, M., D. Baratti, and S. Kusamura, Morbidity and quality of life following cytoreduction and HIPEC. Cancer Treat Res, 2007. 134: p. 403-18.

233. Kerscher, A.G., et al., Morbidity and mortality of 109 consecutive cytoreductive procedures with

hyperthermic intraperitoneal chemotherapy (HIPEC) performed at a community hospital. World J Surg, 2010. 34(1): p. 62-9.

234. Votanopoulos, K., et al., A comparison of hematologic toxicity profiles after heated

intraperitoneal chemotherapy with oxaliplatin and mitomycin C. J Surg Res, 2013. 179(1): p. e133-9.

235. Lambert, L.A., et al., Incidence, risk factors, and impact of severe neutropenia after

hyperthermic intraperitoneal mitomycin C. Ann Surg Oncol, 2009. 16(8): p. 2181-7. 236. Kusamura, S., et al., Impact of cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy on systemic toxicity. Ann Surg Oncol, 2007. 14(9): p. 2550-8.

237. Elias, D., et al., [Pseudomyxoma peritonei treated with complete resection and immediate intraperitoneal chemotherapy]. Gastroenterol Clin Biol, 2003. 27(4): p. 407-12.

238. Glehen, O., et al., Intraperitoneal chemohyperthermia and attempted cytoreductive surgery in

patients with peritoneal carcinomatosis of colorectal origin. Br J Surg, 2004. 91(6): p. 747-54. 239. Hyman, N., et al., Anastomotic leaks after intestinal anastomosis: it's later than you think. Ann

Surg, 2007. 245(2): p. 254-8.

240. Sugarbaker, P.H., et al., Prospective morbidity and mortality assessment of cytoreductive surgery plus perioperative intraperitoneal chemotherapy to treat peritoneal dissemination of appendiceal

mucinous malignancy. Ann Surg Oncol, 2006. 13(5): p. 635-44.

241. Glehen, O., et al., Intraperitoneal chemohyperthermia using a closed abdominal procedure and cytoreductive surgery for the treatment of peritoneal carcinomatosis: morbidity and mortality analysis of

216 consecutive procedures. Ann Surg Oncol, 2003. 10(8): p. 863-9.

242. Glockzin, G., et al., Results of cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy for peritoneal carcinomatosis from colorectal cancer. J Surg Oncol, 2009. 100(4): p. 306-10.

243. Kusamura, S., et al., Cytoreductive surgery followed by intraperitoneal hyperthermic perfusion -

Analysis of morbidity and mortality in 209 peritoneal surface malignancies treated with closed abdomen technique. Cancer, 2006. 106(5): p. 1144-1153.

244. Kusamura, S., et al., The importance of the learning curve and surveillance of surgical

performance in peritoneal surface malignancy programs. Surg Oncol Clin N Am, 2012. 21(4): p. 559-76. 245. Study protocol for the World Health Organization project to develop a Quality of Life

assessment instrument (WHOQOL). Qual Life Res, 1993. 2(2): p. 153-9.

246. Moinpour, C.M., et al., Quality of life end points in cancer clinical trials: review and recommendations. J Natl Cancer Inst, 1989. 81(7): p. 485-95.

247. von Gruenigen, V.E. and B.J. Daly, Futility: clinical decisions at the end-of-life in women with ovarian cancer. Gynecol Oncol, 2005. 97(2): p. 638-44.

248. Barnett, M.M., Effect of breaking bad news on patients' perceptions of doctors. J R Soc Med,

2002. 95(7): p. 343-7. 249. Averbach, A.M. and P.H. Sugarbaker, Recurrent intraabdominal cancer with intestinal

obstruction. Int Surg, 1995. 80(2): p. 141-6.

250. Sloan, J.A., et al., The clinical significance of quality of life assessments in oncology: a summary for clinicians. Support Care Cancer, 2006. 14(10): p. 988-98.

251. Fayers, P. and A. Bottomley, Quality of life research within the EORTC-the EORTC QLQ-C30.

European Organisation for Research and Treatment of Cancer. Eur J Cancer, 2002. 38 Suppl 4: p. S125-33. 252. Lim, C., et al., Prospective study of quality of life after cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy using oxaliplatin for peritoneal carcinomatosis. Bulletin Du Cancer, 2010.

97(9): p. 1053-1060. 253. McQuellon, R.P., et al., Quality of life after intraperitoneal hyperthermic chemotherapy (IPHC)

for peritoneal carcinomatosis. Eur J Surg Oncol, 2001. 27(1): p. 65-73.

254. Bonastre, J., et al., [Cost of an intraperitoneal chemohyperthermia (IPCH) related to cytoreductive surgery]. Ann Chir, 2005. 130(9): p. 553-61.

255. Baratti, D., et al., Cost analysis of the combined procedure of cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy (HIPEC). Eur J Surg Oncol, 2010. 36(5): p. 463-9.

256. Kilian, M., et al., [Hyperthermic intraperitoneal chemotherapy in the German DRG system.

Analysis of case cost calculations of a maximum care university]. Chirurg, 2010. 81(11): p. 1005-12.

257. Parikh, D., et al., D2 gastrectomy: lessons from a prospective audit of the learning curve. Br J Surg, 1996. 83(11): p. 1595-9.

258. Renzulli, P. and U.T. Laffer, Learning curve: the surgeon as a prognostic factor in colorectal

cancer surgery. Recent Results Cancer Res, 2005. 165: p. 86-104. 259. Tseng, J.F., et al., The learning curve in pancreatic surgery. Surgery, 2007. 141(4): p. 456-63.

260. Sutton, D.N., J. Wayman, and S.M. Griffin, Learning curve for oesophageal cancer surgery. Br J

Surg, 1998. 85(10): p. 1399-402.

261. Jimenez-Rodriguez, R.M., et al., Learning curve for robotic-assisted laparoscopic rectal cancer

surgery. Int J Colorectal Dis, 2012. 262. Kim, Y.W., et al., The learning curve for robot-assisted total mesorectal excision for rectal

cancer. Surg Laparosc Endosc Percutan Tech, 2012. 22(5): p. 400-5.

263. Chen, G., et al., The Learning Curve for the Laparoscopic Approach for Colorectal Cancer: A Single Institution's Experience. J Laparoendosc Adv Surg Tech A, 2013. 23(1): p. 17-21.

264. Artibani, W. and G. Novara, Cancer-related outcome and learning curve in retropubic radical

prostatectomy: "if you need an operation, the most important step is to choose the right surgeon". Eur Urol, 2008. 53(5): p. 874-6.

265. Trimbos, J.B., et al., The long learning curve of gynaecological cancer surgery: an argument for

centralisation. BJOG, 2000. 107(1): p. 19-23. 266. Chong, G.O., et al., Learning curve of laparoscopic radical hysterectomy with pelvic and/or

para-aortic lymphadenectomy in the early and locally advanced cervical cancer: comparison of the first 50

and second 50 cases. Int J Gynecol Cancer, 2009. 19(8): p. 1459-64. 267. Wibe, A., et al., Effect of hospital caseload on long-term outcome after standardization of rectal

cancer surgery at a national level. Br J Surg, 2005. 92(2): p. 217-24.

268. Iversen, L.H., et al., Influence of caseload and surgical speciality on outcome following surgery

for colorectal cancer: a review of evidence. Part 2: long-term outcome. Colorectal Dis, 2007. 9(1): p. 38-46.

269. Marusch, F., et al., Effect of caseload on the short-term outcome of colon surgery: results of a

multicenter study. Int J Colorectal Dis, 2001. 16(6): p. 362-9. 270. Iversen, L.H., et al., Influence of caseload and surgical speciality on outcome following surgery

for colorectal cancer: a review of evidence. Part 1: short-term outcome. Colorectal Dis, 2007. 9(1): p. 28-37.

271. Yan, T.D., et al., Learning curve for cytoreductive surgery and perioperative intraperitoneal chemotherapy for peritoneal surface malignancy--a journey to becoming a Nationally Funded

Peritonectomy Center. Ann Surg Oncol, 2007. 14(8): p. 2270-80.

272. Cavaliere, F., et al., [Peritonectomy and chemohyperthermia in the treatment of peritoneal carcinomatosis: learning curve]. Suppl Tumori, 2005. 4(3): p. S119-21.

273. Mohamed, F. and B.J. Moran, Morbidity and mortality with cytoreductive surgery and intraperitoneal chemotherapy: the importance of a learning curve. Cancer J, 2009. 15(3): p. 196-9.

274. Kusamura, S., D. Baratti, and M. Deraco, Multidimensional analysis of the learning curve for

cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in peritoneal surface malignancies. Ann Surg, 2012. 255(2): p. 348-56.

275. Matheny, M.E., et al., Evaluation of an automated safety surveillance system using risk adjusted

sequential probability ratio testing. BMC Med Inform Decis Mak, 2011. 11: p. 75. 276. Matheny, M.E., L. Ohno-Machado, and F.S. Resnic, Risk-adjusted sequential probability ratio

test control chart methods for monitoring operator and institutional mortality rates in interventional

cardiology. Am Heart J, 2008. 155(1): p. 114-20. 277. Spiegelhalter, D., et al., Risk-adjusted sequential probability ratio tests: applications to Bristol,

Shipman and adult cardiac surgery. Int J Qual Health Care, 2003. 15(1): p. 7-13.

278. Benneyan, J.C. and A.D. Borgman, Risk-adjusted sequential probability ratio tests and longitudinal surveillance methods. Int J Qual Health Care, 2003. 15(1): p. 5-6.

279. Boorjian, S.A., et al., Comparative Performance of Comorbidity Indices for Estimating

Perioperative and 5-Year All-Cause Mortality Following Radical Cystectomy for Bladder Cancer. J Urol, 2013.

280. Clark, E.J., et al., Preoperative lymphocyte count as a prognostic factor in resected pancreatic

ductal adenocarcinoma. HPB (Oxford), 2007. 9(6): p. 456-60.

281. Kobayashi, N., et al., Preoperative lymphocyte count is an independent prognostic factor in

node-negative non-small cell lung cancer. Lung Cancer, 2012. 75(2): p. 223-7.

282. Zemanova, M., et al., Outcomes of patients with oesophageal cancer treated with preoperative chemoradiotherapy, followed by tumor resection: influence of nutritional factors. J BUON, 2012. 17(2): p.

310-6.

283. Liu, N., et al., [Prognostic significance of preoperative serum albumin level in patients with gastric cancer]. Zhonghua Wei Chang Wai Ke Za Zhi, 2011. 14(2): p. 100-3.

284. Okada, I., et al., [Significance of Onodera's prognostic nutritional index for treating unresectable

or recurrent colorectal cancer with chemotherapy]. Gan To Kagaku Ryoho, 2012. 39(2): p. 231-5.

285. Sagawa, M., et al., [The significance of Onodera's prognostic nutritional index for the treatment

of gastrointestinal cancer]. Gan To Kagaku Ryoho, 2008. 35(12): p. 2253-5. 286. Sugarbaker, P.H., Peritonectomy procedures. Ann Surg, 1995. 221(1): p. 29-42.

287. Peat, J.K. and B. Barton, Medical statistics : a guide to data analysis and critical appraisal. 1st ed.

2005, Malden, Mass.: Blackwell Pub. xii, 324 p. 288. Livingston, E.H., Who was student and why do we care so much about his t-test? J Surg Res,

2004. 118(1): p. 58-65.

289. Denne, J.S. and C. Jennison, Estimating the sample size for a t-test using an internal pilot. Stat Med, 1999. 18(13): p. 1575-85.

290. Hiller, E., [Statistical comparison of multiple dependent samples--the Friedman test and the

Wilcoxon and Wilcox Dij test]. Z Arztl Fortbild (Jena), 1988. 82(18): p. 885-6. 291. Wong, K.C., Chi squared test versus Fisher's exact test. Hong Kong Med J, 2011. 17(5): p. 427.

292. Turner, N., Chi-squared test. J Clin Nurs, 2000. 9(1): p. 93.

293. Buyse, M.E., M.J. Staquet, and R.J. Sylvester, Cancer clinical trials : methods and practice. Oxford medical publications. 1984, Oxford ; New York: Oxford University Press. xiv, 481 p.

294. Cai, T. and C.S. Moskowitz, Semi-parametric estimation of the binormal ROC curve for a

continuous diagnostic test. Biostatistics, 2004. 5(4): p. 573-86.

295. Kim, M.J., et al., [Optimal cut-off value of PIVKA-II for diagnosis of hepatocellular carcinoma--

using ROC curve]. Korean J Hepatol, 2006. 12(3): p. 404-11.

296. Lemeshow, S. and D.W. Hosmer, Jr., A review of goodness of fit statistics for use in the development of logistic regression models. Am J Epidemiol, 1982. 115(1): p. 92-106.

297. Dinse, G.E. and S.W. Lagakos, Nonparametric estimation of lifetime and disease onset

distributions from incomplete observations. Biometrics, 1982. 38(4): p. 921-32. 298. Bland, J.M. and D.G. Altman, Survival probabilities (the Kaplan-Meier method). BMJ, 1998.

317(7172): p. 1572.

299. Mantel, N., Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother Rep, 1966. 50(3): p. 163-70.

300. Gill, R.D., Multistate life-tables and regression models. Math Popul Stud, 1992. 3(4): p. 259-76. 301. Deraco, M., et al., Morbidity, toxicity, and mortality classification systems in the local regional

treatment of peritoneal surface malignancy. Journal of Surgical Oncology, 2008. 98(4): p. 253-257.

302. Cavaliere, F., et al., 120 peritoneal carcinomatoses from colorectal cancer treated with peritonectomy and intra-abdominal chemohyperthermia: a S.I.T.I.L.O. multicentric study. In Vivo, 2006.

20(6A): p. 747-50.

303. Deraco, M., et al., An Italian Multicentric Phase II study on peritonectomy and intra peritoneal hyperthermic perfusion (IPHP) to treat patients with peritoneal mesothelioma. J Exp Clin Cancer Res, 2003.

22(4 Suppl): p. 41-5.

304. Honore, C., et al., HIPEC for peritoneal carcinomatosis: does an associated urologic procedure increase morbidity? Ann Surg Oncol, 2012. 19(1): p. 104-9.

305. Targarona, J., et al., [Morbidity and mortality rates in relation to the "surgeon factor" after

duodenopancreatectomy]. Cir Esp, 2007. 82(4): p. 219-23. 306. Schlitt, H.J., et al., Morbidity and mortality associated with pancreatogastrostomy and

pancreatojejunostomy following partial pancreatoduodenectomy. Br J Surg, 2002. 89(10): p. 1245-51.

307. Swisher, S.G., et al., Effect of operative volume on morbidity, mortality, and hospital use after esophagectomy for cancer. J Thorac Cardiovasc Surg, 2000. 119(6): p. 1126-32.

308. Chuaqui, R.F., et al., Genetic analysis of synchronous mucinous tumors of the ovary and

appendix. Hum Pathol, 1996. 27(2): p. 165-71.

309. Elias, D., et al., Peritoneal pseudomyxoma: results of a systematic policy of complete

cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Br J Surg, 2008. 95(9): p. 1164-71.

310. Florkowski, C.M., Sensitivity, specificity, receiver-operating characteristic (ROC) curves and likelihood ratios: communicating the performance of diagnostic tests. Clin Biochem Rev, 2008. 29 Suppl 1:

p. S83-7.

311. Asher, V., J. Lee, and A. Bali, Preoperative serum albumin is an independent prognostic predictor of survival in ovarian cancer. Med Oncol, 2012. 29(3): p. 2005-9.

312. Lin, M.Y., et al., Preoperative serum albumin but not prealbumin is an excellent predictor of

postoperative complications and mortality in patients with gastrointestinal cancer. Am Surg, 2011. 77(10): p. 1286-9.

313. Carr, N.J., et al., Pathology and prognosis in pseudomyxoma peritonei: a review of 274 cases. J

Clin Pathol, 2012. 65(10): p. 919-23. 314. Alexander-Sefre, F., et al., Elevated tumour markers prior to complete tumour removal in

patients with pseudomyxoma peritonei predict early recurrence. Colorectal Dis, 2005. 7(4): p. 382-6.

315. Carmignani, C.P., et al., Utility of CEA and CA 19-9 tumor markers in diagnosis and prognostic assessment of mucinous epithelial cancers of the appendix. J Surg Oncol, 2004. 87(4): p. 162-6.

316. Kyung, M.H., et al., [Prognostic impact of Charlson comorbidity index obtained from medical

records and claims data on 1-year mortality and length of stay in gastric cancer patients]. J Prev Med Public Health, 2009. 42(2): p. 117-22.

317. Singh, B., et al., Validation of the Charlson comorbidity index in patients with head and neck

cancer: a multi-institutional study. Laryngoscope, 1997. 107(11 Pt 1): p. 1469-75. 318. Wahlgren, T., et al., Use of the Charlson combined comorbidity index to predict

postradiotherapy quality of life for prostate cancer patients. Int J Radiat Oncol Biol Phys, 2011. 81(4): p.

997-1004. 319. Chua, T.C., et al., Determining the association between preoperative computed tomography

findings and postoperative outcomes after cytoreductive surgery and perioperative intraperitoneal

chemotherapy for pseudomyxoma peritonei. Ann Surg Oncol, 2011. 18(6): p. 1582-9.

320. Nissan, A., et al., Evidence-based medicine in the treatment of peritoneal carcinomatosis: Past,

present, and future. J Surg Oncol, 2009. 100(4): p. 335-44.

321. Yamamoto, M., et al., Prognostic significance of tumor markers in peritoneal lavage in advanced gastric cancer. Oncology, 2004. 67(1): p. 19-26.

322. de Bree, E., et al., Peritoneal carcinomatosis from colorectal or appendiceal origin: correlation of

preoperative CT with intraoperative findings and evaluation of interobserver agreement. J Surg Oncol, 2004. 86(2): p. 64-73.

323. Berthelot, C., et al., Use of FDG-PET/CT for peritoneal carcinomatosis before hyperthermic

intraperitoneal chemotherapy. Nucl Med Commun, 2011. 32(1): p. 23-9. 324. Strohlein, M.A., et al., Immunotherapy of peritoneal carcinomatosis with the antibody

catumaxomab in colon, gastric, or pancreatic cancer: an open-label, multicenter, phase I/II trial. Onkologie, 2011. 34(3): p. 101-8.

325. Spiliotis, J., et al., Morbidity and mortality of cytoreductive surgery and hyperthermic

intraperitoneal chemotherapy in the management of peritoneal carcinomatosis. J BUON, 2009. 14(2): p. 259-64.

326. Minicozzi, A., et al., [Treatment of the peritoneal carcinomatosis by cytoreductive surgery and

intraperitoneal hyperthermic chemotherapy (IHPC): postoperative morbidity and mortality and short-term follow-up]. Ann Ital Chir, 2008. 79(4): p. 231-9.

327. Loggie, B.W., et al., Prospective trial for the treatment of malignant peritoneal mesothelioma.

Am Surg, 2001. 67(10): p. 999-1003. 328. Alexander, H.R., Jr., Surgical treatment of malignant peritoneal mesothelioma: past, present, and

future. Ann Surg Oncol, 2010. 17(1): p. 21-2.

329. Mohamed, F., et al., A new standard of care for the management of peritoneal surface malignancy. Curr Oncol, 2011. 18(2): p. e84-96.

330. Chua, T.C., et al., Morbidity and mortality outcomes of cytoreductive surgery and perioperative

intraperitoneal chemotherapy at a single tertiary institution: towards a new perspective of this treatment. Ann Surg, 2010. 251(1): p. 101-6.

331. Baratti, D., et al., Multicystic peritoneal mesothelioma: outcomes and patho-biological features

in a multi-institutional series treated by cytoreductive surgery and Hyperthermic Intraperitoneal

Chemotherapy (HIPEC). Eur J Surg Oncol, 2010. 36(11): p. 1047-53.

332. Yan, T.D., et al., Sex difference in diffuse malignant peritoneal mesothelioma. Br J Surg, 2006.

93(12): p. 1536-42. 333. Munkholm-Larsen, S., C.Q. Cao, and T.D. Yan, Malignant peritoneal mesothelioma. World J

Gastrointest Surg, 2009. 1(1): p. 38-48.

334. Carteni, G., et al., Malignant peritoneal mesothelioma-Results from the International Expanded Access Program using pemetrexed alone or in combination with a platinum agent. Lung Cancer, 2009.

64(2): p. 211-8.

335. Simon, G.R., et al., Pemetrexed plus gemcitabine as first-line chemotherapy for patients with peritoneal mesothelioma: final report of a phase II trial. J Clin Oncol, 2008. 26(21): p. 3567-72.

336. Isobe, H., et al., Doxorubicin retention and chemoresistance in human mesothelioma cell lines.

Int J Cancer, 1994. 57(4): p. 581-5. 337. Stone, R.L., et al., Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med, 2012. 366(7):

p. 610-8.

338. Brookman-May, S., et al., Does preoperative platelet count and thrombocytosis play a prognostic role in patients undergoing nephrectomy for renal cell carcinoma? Results of a comprehensive retrospective

series. World J Urol, 2012.

339. Cravioto-Villanueva, A., et al., Thrombocytosis as a predictor of distant recurrence in patients with rectal cancer. Arch Med Res, 2012. 43(4): p. 305-11.

340. Villa, R., et al., Multiple mechanisms of telomere maintenance exist and differentially affect

clinical outcome in diffuse malignant peritoneal mesothelioma. Clin Cancer Res, 2008. 14(13): p. 4134-40. 341. Onnis, A. and T. Maggino, Repetitive debulking surgery as adjuvant to chemotherapy in

advanced epithelial ovarian cancer. Clin Exp Obstet Gynecol, 1984. 11(1-2): p. 21-6.

342. Chua, T.C., et al., Intraoperative hyperthermic intraperitoneal chemotherapy after cytoreductive surgery in ovarian cancer peritoneal carcinomatosis: systematic review of current results. J Cancer Res Clin

Oncol, 2009. 135(12): p. 1637-45.

343. Hutanu, I., et al., [Treatment of peritoneal carcinomatosis of gastro-intestinal origin--a

retrospective study of 203 cases]. Rev Med Chir Soc Med Nat Iasi, 2012. 116(1): p. 150-6.

344. Kusamura, S., et al., Circulating tumor markers: Predictors of incomplete cytoreduction and

powerful determinants of outcome in pseudomyxoma peritonei. J Surg Oncol, 2013. 345. Baratti, D., et al., Prognostic value of circulating tumor markers in patients with pseudomyxoma

peritonei treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg

Oncol, 2007. 14(8): p. 2300-8. 346. Ihemelandu, C.U., et al., Predicting Postoperative Morbidity Following Cytoreductive Surgery

with Hyperthermic Intraperitoneal Chemotherapy (CS+HIPEC) with Preoperative FACT-C (Functional

Assessment of Cancer Therapy) and Patient-Rated Performance Status. Ann Surg Oncol, 2013. 347. Glockzin, G., et al., Treatment-related morbidity and toxicity of CRS and oxaliplatin-based

HIPEC compared to a mitomycin and doxorubicin-based HIPEC protocol in patients with peritoneal carcinomatosis: a matched-pair analysis. J Surg Oncol, 2013. 107(6): p. 574-8.

348. Pelz, J.O., et al., Hyperthermic intraperitoneal chemoperfusion (HIPEC) decrease wound

strength of colonic anastomosis in a rat model. Int J Colorectal Dis, 2007. 22(8): p. 941-7. 349. Steiner, S.H., et al., Monitoring surgical performance using risk-adjusted cumulative sum charts.

Biostatistics, 2000. 1(4): p. 441-52.

350. Kusamura, S., et al., Learning curve for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in peritoneal surface malignancies: Analysis of two centres. J Surg Oncol, 2013. 107(4): p.

312-9.

351. Moran, B.J., Establishment of a peritoneal malignancy treatment centre in the United Kingdom. Eur J Surg Oncol, 2006. 32(6): p. 614-8.

352. Gonzalez Bayon, L., et al., Initiation of a program in peritoneal surface malignancy. Surg Oncol

Clin N Am, 2003. 12(3): p. 741-53.

List of scientific papers issued during PhD studies

1. Tratamentul carcinomatozei peritoneale de etiologie digestivă - studiu retrospectiv pe un lot de 203

pacienţi.

I. Huţanu, D. Timofte, V. Scripcari , C. Diaconu Rev Med Chir Soc Med Nat Iasi. 2012 Jan-Mar;116(1):150-6.

http://www.revmedchir.ro/uploads/1/5/7/2/15722076/22_hutanu.pdf

2. Learning curve for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in peritoneal

surface malignancies: Analysis of two centres†

Shigeki Kusamura, Dario Baratti, Salvatore Virzì, Serena Bonomi, Domenico Rosario Iusco ,Antonio Grassi, Ionut Hutanu ,Marcello Deraco

J Surg Oncol., Online first

http://onlinelibrary.wiley.com/doi/10.1002/jso.23231/abstract

3.The importance of the learning curve and surveillance of surgical performance in peritoneal surface

malignancy programs.

Kusamura S, Baratti D, Hutanu I, Rossi P, Deraco M.

Surg Oncol Clin N Am. 2012 Oct;21(4):559-76. doi: 10.1016/j.soc.2012.07.011

http://www.surgonc.theclinics.com/article/S1055-3207(12)00060-9/abstract

4. Circulating tumor markers: predictors of incomplete cytoreduction and powerful determinants of outcome in pseudomyxoma peritonei

Kusamura S, Hutanu I, Baratti D, Deraco M.

J Surg Oncol. 2013 May 29. doi: 10.1002/jso.23329. [Epub ahead of print]

5. The role of perioperative systemic chemotherapy in diffusemalignant peritoneal mesothelioma patients treated withcytoreductive surgery and hyperthermic intraperitoneal Chemotherapy

Marcello Deraco, Dario Baratt, Ionut Hutanu, Rossella Bertuli, Shigeki Kusamura MD PhD.1

Ann Surg Oncol. 2013 Apr;20(4):1093-100. doi: 10.1245/s10434-012-2845-x. Epub 2013 Mar 2.

6. Chimioterapia hipertermică asociată citoreducţiei tumorale (CHIP) - opţiune în tratamentul

carcinomatozei peritoneale de etiologie colorectală I. Huțanu, R.Iulian, B. Filip, Mihaela Buna, Marcello Deraco, V.Scripcariu Jurnalul de Chirurgie NR. 1, VOL. 9, 2013

7. Diffuse malignant peritoneal mesothelioma: Long-term survival with complete cytoreductive surgery followed by hyperthermic intraperitoneal chemotherapy (HIPEC).

Baratti D, Kusamura S, Cabras AD, Bertulli R, Hutanu I, Deraco M.

Eur J Cancer. 2013 Jul 4. pii: S0959-8049(13)00435-8. doi: 10.1016/j.ejca.2013.05.027. [Epub ahead of print]

List of submitted papers

1.Neutrophil-lymphocyte ratio and Onodera index: new prognostic markers in pseudomyxoma peritonei

Shigeki Kusamura, Dario Baratti, Ionut Hutanu, Cecilia Gavazzi, Alessandro Sironi, Haeusler Edward,

Daniele Morelli, Marcello Deraco Annals of Surgical Oncology

2. Diffuse malignant peritoneal mesothelioma: seven-year actual survival after surgical cytoreduction and

hyperthermic intraperitoneal chemotherapy (HIPEC) defines cure D. Baratti, S. Kusamura, A. D. Cabras, R. Bertulli, I. Hutanu, M. Deraco Annals of Surgical Oncology

List of papers presented in national and international congresses National congresses

1. Conferinţa Naţională de Chirurgie Sibiu 19-21 Mai 2011

Carcinomatoza peritoneala cu punct de plecare digestiv . studiuretrospectiv pe un lot de 64 de pacienti ai

Clinicii III Chirurgie Iasi I.Hutanu, G. Anitei, R. lulian, C. Diaconu, V. Scripcariu, D. Timofte

http://chirurgie2011.eventernet.ro/images/program_final.pdf

2.Simpozionul Doctoranzilor 8 septembrie 2011, Timişoara

Experienta clinicii III Chirurgie Iaşi în tratamentul carcinomatozei peritoneale de etiologie digestiva -

studiu retrospectiv pe o perioadă de 5ani.

I.Hutanu

3.Simpozionul National"Contribuţii ale tinerilor cercetatori la dezvoltarea cercetării in domeniul medico-farmaceutic",sectiunea studentilor - doctoranzi.

Iaşi, 7 - 9 Noiembrie 2012

The evaluation of risk factors for complications after cytoreductive surgery associated with hipec for colorectal and appendicular carcinomatosis patients

Hutanu Ionut , ScripcariuViorel, Shigeki Kusamura , Dario Baratti, Marcello Deraco

4.Confer 2012 Conferinţele institutului regional de oncologie iaşi zilele oncologiei ieşene, ediţia a VIII-A

22‐24 NOIEMBRIE 2012, IAŞI Chimioterapia hipertermică intraperitoneală asociată citoreducţiei tumorale. Experienţa Institutului

Naţional pentru Tumori din Milano, Italia I. Huţanu, V. Scripcariu, Shigeki Kusamura, D. Baratti, M. Deraco

http://oncologieiasi.ro/atdoc/Program_detaliat.pdf

5. Congresul Naţional al Societăţii Române de Coloproctologie

ediţia a III-a 18 – 20 aprilie 2013 Iaşi, Centrul de Conferinţe Palas Carcinomatoza peritoneală de etiologie colorectală - prezent şi viitor

I. Huţanu

International congresses

The role of preoperative albumin and lymphocytes in predicting severe complications and survival in patients with appendiceal neoplasms submitted to cytoreduction and hyperthermic intraperitoneal

chemotherapy

Kusamura S, Baratti D, Gavazzi C, Hutanu I, Sironi A, Gil Gomes E, Deraco M Eighth International Symposium on Regional Cancer Therapies February 16—18, 2013

Hyatt Grand Champion Resort & Spa - Indian Wells, California

http://www.regionaltherapies.com/pdf/2013_brochure.pdf

The role of perioperative systemic chemotherapy in diffuse malignant peritoneal mesothelioma patients

treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy Kusamura S, Baratti D, Hutanu I, Deraco M

11th International Conference of the International Mesothelioma Interest Group (iMig 2012), September 11

– 14, 2012, Seaport Hotel and World Trade Center in Boston, Massachusetts, USA. http://www.imig2012.org/scientific-program/documents/iMig2012_OnsiteProgram_vFF_lowres.pdf

O F