You are on page 1of 7

+Model

JVS-750; No. of Pages 7 ARTICLE IN PRESS


Journal of Visceral Surgery (2017) xxx, xxx—xxx

Available online at

ScienceDirect
www.sciencedirect.com

ORIGINAL ARTICLE

A new animal model for hyperthermic


intraperitoneal chemotherapy (HIPEC) in
tumor-bearing mice in the treatment of
peritoneal carcinomatosis of ovarian origin
G. Miailhe a,c,1, A. Arfi a,c,1, M. Mirshahi c, C. Eveno b,c,
M. Pocard b,c, C. Touboul a,c,∗

a
Service de gynécologie obstétrique, hôpital intercommunal de Créteil, université Paris Est,
Paris XII, 40, avenue de Verdun, 94000 Créteil, France
b
Service de chirurgie digestive et cancérologique, hôpital Lariboisière Fernand-Widal,
université Paris VII, 49, boulevard de la chapelle, 75010 Paris, France
c
UMR Inserm U965, angiogenèse et recherche translationnelle, hôpital Lariboisière, 49,
boulevard de la chapelle, 75010 Paris, France

KEYWORDS Summary
Hyperthermic Aim of the study: We set out to develop and evaluate the morbidity of a non-invasive hyper-
intraperitoneal thermic intraperitoneal chemotherapy (HIPEC) procedure in mice. HIPEC has been shown to
chemotherapy; improve overall survival in treating ovarian cancer with peritoneal carcinomatosis. However,
Peritoneal related complications, toxicity and the lack of randomized trials limits its widespread use. To
carcinomatosis; improve the surgical technique, there is a need for animal models that allow teams to work on
Ovarian cancer; large groups without burdensome logistics.
Murine model; Materials and methods: To develop the model, we first determined optimal HIPEC conditions
Oxaliplatin in 20 Black Six mice without carcinomatosis. To evaluate HIPEC morbidity, peritoneal carcino-
matosis cells of ovarian origin were injected into the peritoneum of 10 pathogen-free Nude
mice. The mice underwent HIPEC 21 days later under general anesthesia. An inflow catheter
was introduced into the left hypochondria and an outflow catheter was introduced into the left
iliac fossa. Bath infusion was oxaliplatin (920 mg/m2 ) at 43 ◦ C for 12 minutes. The mice were
monitored and sacrificed two weeks after the procedure.
Results: No deaths were observed during the procedure and infusion was well tolerated
throughout the HIPEC. One mouse died the day after the procedure. No major dehydration,
hemoperitoneum or evisceration was observed.
Conclusion: This mouse model of closed abdomen HIPEC has limited morbidity and could be a
useful model to study HIPEC regimens and its effects on peritoneal carcinomatosis.
© 2017 Elsevier Masson SAS. All rights reserved.

∗ Corresponding author.
E-mail address: cyril.touboul@gmail.com (C. Touboul).
1 These authors contributed equally to this work.

https://doi.org/10.1016/j.jviscsurg.2017.10.008
1878-7886/© 2017 Elsevier Masson SAS. All rights reserved.

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008
+Model
JVS-750; No. of Pages 7 ARTICLE IN PRESS
2 G. Miailhe et al.

Introduction Maintenance and care of all experimental animals were


carried out according to the guidelines of the local Ani-
Epithelial ovarian cancer (EOC) is the leading cause of death mal Protection Commission and in compliance with national
from gynecological cancer in northwestern Europe and in guidelines (Saisine n◦ 02095, Animal Protection Committee,
northern America [1]. Most women are diagnosed only after French Ministry of Agriculture).
peritoneal dissemination has occurred. EOC tends to be
chemosensitive and confine itself to the surface of the Cells and cell culture
peritoneal cavity for much of its natural history. Though
these features make it an obvious target for intraperitoneal We used the human epithelial ovarian cancer cell (OCC) line
chemotherapy, standard treatment is complete cytoreduc- OVCAR-3. OCCs were transfected with a plasmide expressing
tive surgery followed by five to eight cycles of intravenous luciferase. The OCCs were cultivated in an incubator at 37 ◦ C
taxane chemotherapy [2]. Even though initially effective, and 5% CO2 in monolayer cultures in 20 mL complete medium
drug-resistant cancer or relapse from residual disease of Dulbecco’s Modified Eagle’s Medium (DMEM), 10% heat-
reduces the 5-year survival rate to about 20% [3]. inactivated fetal bovine serum, 1% Penicillin/Streptomycin,
Cisplatin is a DNA damaging chemotherapeutic used and 1% L-glutamate (Gibco, Saint-Aubin, France).
to treat solid tumors including EOC. However, resistance
to cisplatin in this setting limits clinical success. Three Establishment of intraperitoneal metastatic
large randomized clinical trials have shown the benefit ovarian tumors in mice
of hyperthermia when administering chemotherapy agents
intraperitoneally to target small remnant tumor load and The OVCAR-3 cell suspension (6.0 × 106 cells in 500 ␮L of
putative malignant cells in the abdominal cavity, a tech- serum-free DMEM media) was injected into the peritoneum
nique known as hyperthermic intraperitoneal chemotherapy in the left iliac fossa of mice using an 18-gauge needle.
(HIPEC) [4—6]. Hyperthermia is tumoricidal alone [7] and
has been shown to enhance cisplatin inhibition of peritoneal Evaluation of carcinomatosis
tumor growth [8]. A recent Cochrane database [9] has shown
that HIPEC increases overall survival and progression-free To assess the peritoneal carcinomatosis before surgery, the
survival in women with advanced ovarian cancer. However, mice were monitored twice a week by bioluminescence
the potential for catheter-related complications and toxic- after intraperitoneal injection of 0.2 mL of Luciferine (150
ity needs to be taken into account. Efforts to define the ug/mL), 10 minutes before acquisition. Almost 3 weeks after
role of HIPEC in a large randomized trial failed to attain the injection (by day 19), the mice had developed disseminated
required number of patients because of patient dissatisfac- intraperitoneal tumors.
tion with randomization [10]. Moreover, optimal treatment
modalities remain to be defined and targeted therapies Chemicals
developed in order to increase efficacy and decrease tox-
icity. Animal models are therefore needed. Oxaliplatine (Onco-TainTM , Hospira), was chosen as
While animal models have already been developed in rats chemotherapy for its peritoneous absorption qualities. The
and pigs to study HIPEC procedures [11], these are burden- administered dose was 920 mg/m2 according to the results
some in terms of logistics and limit the possibility of working of Piché et al. [15].
on large groups. Mice models are more accessible but pub-
lished data are scarce and existing reports fail to include Experimental design
animal survival or postoperative complications because the
mice were sacrificed immediately after the HIPEC procedure To develop the model, we first tested the procedure HIPEC
[12—14]. in the 20 Black Six mice without carcinomatosis and deter-
The objective of our study was to develop a new non- mined optimal conditions. The following parameters were
invasive HIPEC procedure using mice as a miniaturized tested without chemotherapy in the bath liquid:
• safety and operative morbidity of open or closed abdomen
animal model, and to evaluate the morbidity of the pro-
cedure. intraperitoneal infusion;
• thermic control using a heat mattress: optimal tempera-
ture of the mattress and length of the procedure.
Materials and methods Before surgery, the mice were administered a subcuta-
neous injection of 0.5 mL buprenophine. They were then
Animals anesthetized with 4% isoflurane (2L/mn) in an inhalation
chamber (Fig. 1A). The body temperature was monitored
Twenty female C57BL/6 mice between 6—8 weeks old using a rectal temperature probe (Fig. 1B). The use of a
(Charles River, Arbesle, France) were used to build the warmed mattress to limit body heat loss was evaluated in
experimental procedure and determine optimal conditions. four mice:
Ten female pathogen-free Nude mice (without thymus, • mouse No. 1: no warmed mattress;
T-lymphocyte free), weighing 20 to 27 g (average: 25.3 g) • mouse No. 2: warmed mattress at 35◦ C;
and between 7—9 weeks old (Charles River, Arbesle, France) • mouse No. 3: HIPEC procedure without warmed mattress;
were used to perform the HIPEC procedure on a peritoneal • mouse No. 4: HIPEC procedure with warmed mattress at
carcinomatosis model. 35 ◦ C:
The animals were housed in filter-topped cages (5 mice ◦ Inflow catheter: thermic control and optimal flow;
per cage) under clean, non-sterile, standardized conditions ◦ Outflow catheter: optimal flow.
(temperature 20—24 ◦ C, relative humidity 50—60 percent,
12 h light/12 h dark cycle). They were fed with standard In the second study, we tested HIPEC using oxaliplatin on
laboratory diet and tap water ad libitum. mice with ovarian peritoneal carcinomatosis to evaluate the

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008
+Model
JVS-750; No. of Pages 7 ARTICLE IN PRESS
A new murine model for HIPEC 3

Figure 1. Pictures of the HIPEC procedure. A. Anesthesia. B. Introduction of the rectal temperature probe. C. Introduction of the inflow
catheter. D. Introduction of the outflow catheter. E. Final aspect of the installation. F. Skin closure.

feasibility and morbidity of the procedure. Ten mice were


injected with 6 × 106 OVCAR-3 cells 21 days before HIPEC.
Only one HIPEC procedure was performed per mouse and the
mice were monitored for 14 days before sacrifice. Engraft-
ment with OVCAR-3 was checked by bioluminescence 19 days
after the cell injection, 2 days before HIPEC and each week
until sacrifice.

Evaluation of the mice


The mice were inspected daily for side effects and were
weighed on days 2 and 7 after the HIPEC procedure or if
signs of dehydration were observed. Mice displaying major
dehydration with a weight loss of more than 20% of the
initial weight were sacrificed. Mice with moderate dehy- Figure 2. Graph showing tests to determine the optimal body
dration were hydrated with subcutaneous saline (1 mL) temperature of the mice.
daily until disappearance of the skin fold. The mice were
given water-soaked food in a container and kept under
catheter its evacuation. The main problem encountered was
standard conditions before sacrifice after general anesthesia
intraabdominal hyperpressure responsible for intestinal loop
by isoflurane 2% 14 days after the HIPEC procedure.
collapse and catheter kinking. To fix this problem, a 14G mul-
tiperforated outflow catheter was introduced tangentially to
the abdominal wall. The outflow catheter operated in free
Results flow to ensure good flow quality. To avoid pre-peritoneal
infusion an 18G inflow catheter was inserted perpendicu-
Choice of the technique of the intraperitoneal larly into the abdomen wall. On the final model, the inflow
infusion catheter was positioned into the left hypochondria and
outflow catheter into the right iliac fossa, thus allowing
Open abdomen homogenous diffusion and then aspiration of the liquid. No
We first tested open-abdomen laparotomy on five mice. This complication was observed postoperatively.
model quickly revealed limitations in terms of heat loss,
bath liquid leakage and bleeding. Furthermore, two mice Stabilization of body temperature
(40%) displayed evisceration after the procedure. We then
tested closed abdomen infusion. We tested four conditions to maintain the body tempera-
ture of the mice stable. The temperature of mouse No. 1
(no warmed mattress) rapidly decreased leading to lethal
Closed abdomen hypothermia several minutes after general anesthesia began
This technique was achieved in four mice under general (Fig. 2). The temperature of mouse No. 4 (HIPEC procedure
anesthesia. Two catheters were introduced through two dis- with warmed mattress at 35 ◦ C) rapidly increased leading to
tinct dials on the abdomen. The inflow catheter allowed lethal hyperthermia several minutes after general anesthe-
infusion of the hyperthermic solution and the outflow sia began (Fig. 2). For mouse No. 3 (HIPEC procedure without

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008
+Model
JVS-750; No. of Pages 7 ARTICLE IN PRESS
4 G. Miailhe et al.

Figure 3. Diagram of the experimental HIPEC circuit.

Figure 4. Graph of the temperature course during HIPEC in the abdomen and in the rectal cavity.

warmed mattress), the temperature rapidly decreased dur- the target temperature for the perfusate was 43 ◦ C, an ini-
ing the first 3 minutes during installation of the catheters tial temperature of 53 ◦ C was required because of a heat
and the temperature reached 36 ◦ C only 8 minutes after infu- loss of 10 ◦ C during administration. Once the perfusate had
sion. We therefore decided to use a warmed mattress during reached 53 ◦ C a tube was connected to the inflow catheter
the preparation and the first 3 minutes of hyperthermic infu- and infusion was performed at a rate of 0.08 mL/sec for
sion. 12 min using a Masterflex pump (Cole-Palmer Instrument Co,
Cat # 07524-50). After a few seconds the abdomen had
HIPEC procedure bulged enough to facilitate the introduction of the outflow
catheter in the left iliac fossa. This 14G multi-perforated
Two catheters were introduced into the abdominal cav- catheter was positioned so that the perfusate was drained
ity through the flanks as represented in Fig. 3. The inflow by declivity in free flow without negative pressure
catheter (18 Gauge) was inserted into the left hypochondria (Fig. 1E).
perpendicularly to the abdominal wall with the help of a During the HIPEC procedure, the abdomen was mas-
prehension clamp (Fig. 1C and D). The perfusate, consisting saged gently to achieve a uniform heat distribution. After
of 100 mL of normal saline solution with added oxaliplatin 12 minutes of infusion, the inflow catheter was removed.
at a concentration of 920 mg/m2 , was heated to 53 ◦ C by A light pressure on the abdomen was applied to extract
emerging the saline bag into a heated water bath. Although the surplus solution before removing the perfusate outflow

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008
+Model
JVS-750; No. of Pages 7 ARTICLE IN PRESS
A new murine model for HIPEC 5

Figure 5. Course of mice weight after HIPEC procedure.

®
catheter. The skin was closed using Monocryl 5-0 (Ethicon) to platinum. The combination of cytoreductive surgery
(Fig. 1F). and HIPEC has been introduced as a means to overcome
this issue in the treatment of patients with peritoneal
Infusion characteristics carcinomatosis [20,21]. HIPEC represents a considerable
pharmacokinetic advantage over systemic treatment as it
The mean volume of the chemotherapy bath per mouse was achieves high concentrations of cytotoxic drugs locally and
45 mL. During the HIPEC procedure, the mean intraabdom- minimizes systemic absorption, thus limiting potential side
inal temperature was 43 ◦ C. The mean rectal temperature effects.
at the start of the procedure was 37.0 ◦ C. It increased to The Gynecologic Oncology Group (GOG) [22—24] assessed
38.5 ◦ C after 3 minutes at which point the warmed mattress the efficacy and safety of intraperitoneal chemotherapy
was taken away and the mice were maintained at 38.5 ◦ C in advanced ovarian cancer in three randomized trials.
until the end of the procedure. The course of the rectal and Improved survival data were observed in the women under-
intraabdominal temperatures during the HIPEC procedure is going surgery plus HIPEC compared to those undergoing
shown in Fig. 4. conventional systemic chemotherapy alone. However, the
patient cohorts were heterogeneous, differing in age, pre-
Morbidity and mortality vious systemic therapy, tumor size and timing (primary OC,
consolidation treatment and recurrent/persistent disease)
No death was observed during the procedure and infusion [25]. Furthermore, there was a lack of homogeneity in
was well tolerated until the end of the HIPEC. The 10 mice the way HIPEC was administered, such as variation in the
were monitored for 14 days after which they were sacrificed. perfusate temperature, chemotherapy protocols, surgical
One mouse died the day after the procedure for unknown approach (open or closed technique) and HIPEC devices,
reasons. We observed no major dehydration and one mouse which further influences outcomes. Finally, the nature and
suffered from moderate dehydration on day 1. No cases of small number of the patients reported in these studies are
hemoperitoneum or evisceration were observed. The aver- not enough to increase the level of evidence for HIPEC in OC
age weight loss per mouse was 1.6 g; the mice regained their treatment.
original weight on day 14 just before the sacrifice (Fig. 5). Therefore, a reproducible, non-morbid animal model is
needed to improve HIPEC and test new therapies on human
cell lines. Although an HIPEC mouse model already exists,
Discussion the various studies involved in developing them [12—14]
did not report on post-treatment effects as the mice were
Our work is the first experimental animal study to evaluate sacrificed immediately after the procedure. As seen above,
the effectiveness of HIPEC and its complications in a repro- HIPEC complications are a major limiting factor that must
ducible model of peritoneal carcinomatosis of ovarian origin be improved.
that resembles the clinical situation. The closed abdomen Hyperthermia and the drugs used during HIPEC proce-
HIPEC mouse model we developed results in limited mor- dures have a limited penetration depth [26]. Therefore,
bidity and would facilitate the study of different HIPEC HIPEC can only be effective in patients with minimal residual
regimens and its effects. disease after extensive cytoreductive surgery.
The standard front-line therapy for ovarian cancer is In our study, HIPEC could be conducted without difficulty
cytoreductive surgery followed by intravenous platinum- in all cases and none of the animals died during infusion. The
taxane chemotherapy. However, this approach is not abdominal cavity was closed during the procedure and the
curative and approximately 60% of all patients with ovar- abdomen massaged to ensure stirring of the perfusate and a
ian cancer experience recurrence [16—19]. A major limiting more homogeneous distribution of the heated drug solution
factor of clinical success is acquired or inherent resistance throughout the entire abdominal cavity.

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008
+Model
JVS-750; No. of Pages 7 ARTICLE IN PRESS
6 G. Miailhe et al.

Our model consists of a closed abdomen procedure due to [6] Dovern E, de Hingh IHJT, Verwaal VJ, et al. Hyperthermic
the difficulties encountered during open abdomen surgery: intraperitoneal chemotherapy added to the treatment of ovar-
heat loss, bath liquid leakage and bleeding. Other advan- ian cancer. A review of achieved results and complications. Eur
tages of a closed procedure include less risk of exposure J Gynaecol Oncol 2010;31(3):256—61.
of the chemotherapy for the operating staff, a homogenous [7] Giovanella BC, Stehlin JS, Morgan AC. Selective lethal effect of
distribution of the drug (better diffusion on the diaphrag- supranormal temperatures on human neoplastic cells. Cancer
Res 1976;36(11):3944—50.
matic dome, and a constant intra-abdominal temperature
[8] Los G, van Vugt MJ, Pinedo HM. Response of peritoneal solid
(no loss). A limitation of this model, however, is that the tumours after intraperitoneal chemohyperthermia treatment
abdominal peritoneal perfusate cannot be manually stirred with cisplatin or carboplatin. Br J Cancer 1994;69(2):235—41.
as in open infusion. [9] Jaaback K, Johnson N, Lawrie TA. Intraperitoneal chemother-
In Muenyi’s mice model [12], HIPEC infusion lasted apy for the initial management of primary epithelial ovarian
60 minutes as opposed to 12 minutes in our model beyond cancer. Cochrane Database Syst Rev 2016;1:CD005340.
which time signs of poor tolerance appeared. We there- [10] Elias D, Delperro J-R, Sideris L, et al. Treatment of periton-
fore chose a high-dose chemotherapy with a high peritoneal eal carcinomatosis from colorectal cancer: impact of complete
absorption potential over a shorter duration. cytoreductive surgery and difficulties in conducting random-
We used oxaliplatin in our HIPEC model for several rea- ized trials. Ann Surg Oncol 2004;11(5):518—21.
[11] Gremonprez F, Willaert W, Ceelen W. Intraperitoneal
sons [15]. Oxaliplatin is a platinum compound that acts as
chemotherapy (IPC) for peritoneal carcinomatosis: review of
an alkylating agent known to create DNA adducts leading to animal models. J Surg Oncol 2014;109(2):110—6.
apoptosis and impaired cell replication. Intravenous admin- [12] Muenyi CS, States VA, Masters JH, et al. Sodium arsenite
istration of oxaliplatin has shown success in the treatment of and hyperthermia modulate cisplatin-DNA damage responses
colorectal cancer [27,28]. This has led many surgical teams and enhance platinum accumulation in murine metastatic
to use oxaliplatin by the intra-peritoneal route to treat ovarian cancer xenograft after hyperthermic intraperitoneal
peritoneal carcinomatosis [20]. Intraperitoneal oxaliplatin chemotherapy (HIPEC). J Ovarian Res 2011;4:9.
leads to high concentrations of the drug in peritoneal tis- [13] Lehmann K, Rickenbacher A, Jang J-H, et al. New insight
sues in proportion to the dose administered. Furthermore, into hyperthermic intraperitoneal chemotherapy: induction of
heat enhances peritoneal tissue concentration of oxaliplatin oxidative stress dramatically enhanced tumor killing in in vitro
and in vivo models. Ann Surg 2012;256(5):730-7 [Discussion
while reducing its systemic absorption. This latter effect
737-8].
may possibly lead to lower systemic toxicity. [14] Graziosi L, Mencarelli A, Renga B, et al. Gene expression
No major complications were observed after HIPEC in our changes induced by HIPEC in a murine model of gastric cancer.
experimental mouse model. Although data on side effects Vivo Athens Greece 2012;26(1):39—45.
remain limited, available studies have reported acceptable [15] Piché N, Leblond FA, Sidéris L, et al. Rationale for heating
morbidity and mortality rates [29,30]. These data, together oxaliplatin for the intraperitoneal treatment of periton-
with the results of the present study, show that HIPEC could eal carcinomatosis: a study of the effect of heat on
be a safe therapy for peritoneal carcinomatosis. intraperitoneal oxaliplatin using a murine model. Ann Surg
2011;254(1):138—44.
[16] McGuire WP, Hoskins WJ, Brady MF, et al. Cyclophosphamide
and cisplatin versus paclitaxel and cisplatin: a phase III ran-
Conclusion domized trial in patients with suboptimal stage III/IV ovarian
cancer (from the Gynecologic Oncology Group). Semin Oncol
The mouse model of closed-abdomen HIPEC we developed 1996;23(5):40—7.
in this study shows limited morbidity and could represent [17] Muggia FM, Braly PS, Brady MF, et al. Phase III randomized study
a useful model to study different HIPEC regimens and its of cisplatin versus paclitaxel versus cisplatin and paclitaxel in
effects on peritoneal carcinomatosis and survival. Further patients with suboptimal stage III or IV ovarian cancer: a gyne-
animal studies are now mandatory to increase the effec- cologic oncology group study. J Clin Oncol 2000;18(1):106—15.
tiveness and safety of HIPEC in women with ovarian cancer. [18] Ozols RF, Bundy BN, Greer BE, et al. Phase III trial of
carboplatin and paclitaxel compared with cisplatin and pacli-
taxel in patients with optimally resected stage III ovarian
cancer: a Gynecologic Oncology Group study. J Clin Oncol
Disclosure of interest 2003;21(17):3194—200.
[19] Alberts DS, Green S, Hannigan EV, et al. Improved therapeu-
The authors declare that they have no competing interest. tic index of carboplatin plus cyclophosphamide versus cisplatin
plus cyclophosphamide: final report by the Southwest Oncology
Group of a phase III randomized trial in stages III and IV ovarian
References cancer. J Clin Oncol 1992;10(5):706—17.
[20] Glehen O, Mohamed F, Gilly FN. Peritoneal carcinomatosis
[1] Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA from digestive tract cancer: new management by cytoreduc-
Cancer J Clin 2011;61(2):69—90. tive surgery and intraperitoneal chemohyperthermia. Lancet
[2] Armstrong DK, Bundy B, Wenzel L, et al. Intraperitoneal Oncol 2004;5(4):219—28.
cisplatin and paclitaxel in ovarian cancer. N Engl J Med [21] Sugarbaker PH, Building on a consensus. J Surg Oncol
2006;354(1):34—43. 2008;98(4):215—6.
[3] Vergote I, Tropé CG, Amant F, et al. Neoadjuvant chemotherapy [22] Alberts DS, Liu PY, Hannigan EV, et al. Intraperitoneal cis-
or primary surgery in stage IIIC or IV ovarian cancer. N Engl J platin plus intravenous cyclophosphamide versus intravenous
Med 2010;363(10):943—53. cisplatin plus intravenous cyclophosphamide for stage III ovar-
[4] Helm CW. The role of hyperthermic intraperitoneal chemother- ian cancer. N Engl J Med 1996;335(26):1950—5.
apy (HIPEC) in ovarian cancer. Oncologist 2009;14(7):683—94. [23] Markman M, Bundy BN, Alberts DS, et al. trial of standard-
[5] Yang X-J, Li Y, Yonemura Y. Cytoreductive surgery plus hyper- dose intravenous cisplatin plus paclitaxel versus moderately
thermic intraperitoneal chemotherapy to treat gastric cancer high-dose carboplatin followed by intravenous paclitaxel and
with ascites and/or peritoneal carcinomatosis: results from a intraperitoneal cisplatin in small-volume stage III ovarian carci-
Chinese center. J Surg Oncol 2010;101(6):457—64. noma: an intergroup study of the Gynecologic Oncology Group,

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008
+Model
JVS-750; No. of Pages 7 ARTICLE IN PRESS
A new murine model for HIPEC 7

Southwestern Oncology Group, and Eastern Cooperative Oncol- [27] Wolpin BM, Mayer RJ. Systemic treatment of colorectal cancer.
ogy Group. J Clin Oncol 2001;19(4):1001—7. Gastroenterology 2008;134(5):1296—310.
[24] Armstrong DK, Bundy B, Wenzel L, et al. Intraperitoneal [28] Kelland L. The resurgence of platinum-based cancer
cisplatin and paclitaxel in ovarian cancer. N Engl J Med chemotherapy. Nat Rev Cancer 2007;7(8):573—84.
2006;354(1):34—43. [29] Verwaal VJ, van Tinteren H, Ruth SV, et al. Toxicity
[25] Chua TC, Robertson G, Liauw W, et al. Intraoperative hyper- of cytoreductive surgery and hyperthermic intra-peritoneal
thermic intraperitoneal chemotherapy after cytoreductive chemotherapy. J Surg Oncol 2004;85(2):61—7.
surgery in ovarian cancer peritoneal carcinomatosis: sys- [30] Stephens AD, Alderman R, Chang D, et al. Morbidity and mor-
tematic review of current results. J Cancer Res Clin Oncol tality analysis of 200 treatments with cytoreductive surgery
2009;135(12):1637—45. and hyperthermic intraoperative intraperitoneal chemother-
[26] Los G, Sminia P, Wondergem J, et al. Optimisation of intraperi- apy using the coliseum technique. Ann Surg Oncol 1999;6(8):
toneal cisplatin therapy with regional hyperthermia in rats. Eur 790—6.
J Cancer Oxf Engl 1990;27(4):472—7.

Please cite this article in press as: Miailhe G, et al. A new animal model for hyperthermic intraperitoneal chemotherapy
(HIPEC) in tumor-bearing mice in the treatment of peritoneal carcinomatosis of ovarian origin. Journal of Visceral Surgery
(2017), https://doi.org/10.1016/j.jviscsurg.2017.10.008

You might also like