Next Article in Journal
miR-449a Repression Leads to Enhanced NOTCH Signaling in TMPRSS2:ERG Fusion Positive Prostate Cancer Cells
Next Article in Special Issue
Osteoblast-Derived Paracrine and Juxtacrine Signals Protect Disseminated Breast Cancer Cells from Stress
Previous Article in Journal
Peptide Receptor Radionuclide Therapy (PRRT) with 177Lu-DOTATATE; Differences in Tumor Dosimetry, Vascularity and Lesion Metrics in Pancreatic and Small Intestinal Neuroendocrine Neoplasms
Previous Article in Special Issue
High-Plex and High-Throughput Digital Spatial Profiling of Non-Small-Cell Lung Cancer (NSCLC)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mouse Models of Peritoneal Carcinomatosis to Develop Clinical Applications

by
Ángela Bella
1,2,
Claudia Augusta Di Trani
1,2,
Myriam Fernández-Sendin
1,2,
Leire Arrizabalaga
1,2,
Assunta Cirella
1,2,
Álvaro Teijeira
1,2,
José Medina-Echeverz
3,
Ignacio Melero
1,2,4,5,6,
Pedro Berraondo
1,2,4,* and
Fernando Aranda
1,2,*
1
Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
2
Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
3
Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Planegg, Germany
4
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
5
Department of Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
6
Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(5), 963; https://doi.org/10.3390/cancers13050963
Submission received: 2 February 2021 / Revised: 19 February 2021 / Accepted: 20 February 2021 / Published: 25 February 2021

Abstract

:

Simple Summary

Peritoneal carcinomatosis mouse models as a platform to test, improve and/or predict the appropriate therapeutic interventions in patients are crucial to providing medical advances. Here, we overview reported mouse models to explore peritoneal carcinomatosis in translational biomedical research.

Abstract

Peritoneal carcinomatosis of primary tumors originating in gastrointestinal (e.g., colorectal cancer, gastric cancer) or gynecologic (e.g., ovarian cancer) malignancies is a widespread type of tumor dissemination in the peritoneal cavity for which few therapeutic options are available. Therefore, reliable preclinical models are crucial for research and development of efficacious treatments for this condition. To date, a number of animal models have attempted to reproduce as accurately as possible the complexity of the tumor microenvironment of human peritoneal carcinomatosis. These include: Syngeneic tumor cell lines, human xenografts, patient-derived xenografts, genetically induced tumors, and 3D scaffold biomimetics. Each experimental model has its own strengths and limitations, all of which can influence the subsequent translational results concerning anticancer and immunomodulatory drugs under exploration. This review highlights the current status of peritoneal carcinomatosis mouse models for preclinical development of anticancer drugs or immunotherapeutic agents.

1. Introduction

Peritoneal Carcinomatosis (PCa) refers to the metastatic involvement of the peritoneum, characteristic of advanced-stage cancer [1]. PCa in gastrointestinal (e.g., colorectal cancer, gastric cancer) and gynecological (e.g., ovarian cancer) malignancies currently has a poor prognosis with a median survival of under six months [2]. At present, cytoreductive surgery in combination with hyperthermic intraperitoneal chemotherapy (HIPEC) has become widely accepted as an effective option to treat peritoneal metastasis [3,4]. However, locoregional HIPEC or pressurized intraperitoneal aerosol chemotherapy (PIPAC) treatments are only feasible in a small number of patients, and severe side-effects have been reported after peritoneal chemotherapy for PCa [5,6,7]. New therapeutic opportunities and experimental strategies in PCa are needed, and preclinical studies may help provide essential information about future successful clinical treatments.
Approaches based on mouse models allow researchers to learn about the tissue microenvironment in highly complex and dynamic physiological and pathological systems, such as cancer [8,9]. Clinical advances in cancer research as made over recent decades are linked to the efficient use of preclinical models of tumorigenesis. These constitute a critical tool to understand tumor growth and the interactions among different stromal and malignant cells in the tumor microenvironment. Indeed, mouse models of cancer are key to evaluating new therapeutic alternatives. In general, heterotopic syngeneic mouse models are subcutaneous grafts of same-strain tumor cells in mice due to the easy follow up of tumor growth. They represent a simple and accessible model, but suffer from the main disadvantage of not adequately reproducing the natural tumor microenvironment. This limitation is overcome by orthotopic engraftment (seeding tumor cells into the corresponding tissue), which offers a more realistic approach and provides a more relevant tissue microenvironment for assessing tumor development and treatment efficacy [10]. While syngeneic orthotopic models mimic more efficiently the disease biology of human disease, their main drawback relates to the monitoring of tumor progression, which requires the use of reporter genes, invasive surgical procedures or imaging methods such as real-time in-life fluorescence and bioluminescence imaging. Despite these limitations, syngeneic mouse models offer clues and proofs of concept in immunocompetent hosts and thereby represent an excellent preclinical platform to test compounds based on immuno-oncology targets to treat cancer [11].
Human tumor xenografted mouse models are also often used in preclinical cancer studies. Xenograft models constitute the cornerstone of the study of antineoplastic targeted compounds and tyrosine kinase inhibitors [12]. However, the lack of a fully functional immune system in these models is an obstacle in the study of the efficacy of most immunotherapy agents [13]. Furthermore, these models do not take into account cancer immunoediting, thus ignoring the role of the immune tumor microenvironment for cancer progression. Similar to traditional cell line-derived xenograft models, a more personalized option is the direct transfer of tumor specimens from individual patients, so-called patient-derived xenografts (PDXs) [14]. This model could also be engrafted with matched patient-derived peripheral blood mononuclear cells (PBMCs), making it a suitable translational research model for evaluating the efficacy of immunotherapeutic agents. Thus, results originating from PDX models are considered relevant for decision-making in drug development. However, the main drawbacks of the so-called humanized mice models are the limited source of tumor-initiating material, the long latency period for tumor development, and the high cost [13]. Moreover, some features such as the myeloid and vascular stromal compartments are not well reproduced. Furthermore, in addition to all the models on tumor cell inoculation into syngeneic, immunocompetent mice (suspension, tumor fragments, seeded 3D scaffolds, etc.), genetically engineered mouse models (GEMMs) also constitute useful experimental tools in cancer research [15,16]. GEMMs develop de novo tumors in a natural cancer immunoediting context. Oncogenes and tumor suppressor genes are modified in the somatic cells of mice leading to a classification based on the genetic intervention: Knock-out, knock-in, or transgenic mouse models [17,18,19,20]. Tumor onset in these models is sometimes accelerated by the administration of carcinogens or gene transfer of recombinases to activate the transforming tumorogenic and surrogate tumor antigens [21].
Despite all these models, establishing standardized, relevant, feasible, and reproducible PCa mice models remains a challenge. In general, peritoneal tumor burden or cancer progression in PCa mouse models can be quantified by (i) bioluminescence or biofluorescence: With the use of a tumor cell line engineered to express a reporter gene [22,23]; (ii) number and weight of peritoneal implants and/or omentum; iii) quantification of tumor burden by RT-PCR, immunohistochemistry, in vivo fluorescence microscopy, and flow cytometry, among other techniques [10,24]; (iv) ascites: Volume collected, hemorrhagic status; (v) survival: Mouse life span is evaluated by surveillance of spontaneous death or by euthanasia of animals with signs of pain or suffering according to established ethical protocol criteria [25,26]. Here, we provide a brief overview of reported mouse models to explore PCa in translational biomedical research (Figure 1).

2. PCa Mouse Models

2.1. PCa Syngeneic Models

A wide repertoire of syngeneic rodent cell lines has been studied in different PCa model research contexts. Among the most common cell lines to develop peritoneal metastasis in syngeneic mouse models are MC38 and CT26 (colon adenocarcinoma cell lines from the inbred C57BL/6 and Balb/c strains, respectively) [27,28,29,30,31,32], ID8 (an epithelial ovarian cancer cell line from C57BL/6) [33,34,35,36], YTN16 (a gastric cancer cell line from C57BL/6) [37,38], and Panc02 (a pancreatic adenocarcinoma epithelial cell line from C57BL/6) [25,39,40,41]. Some studies have also used the melanoma cell line B16.F10 for peritoneal carcinomatosis as an alternative albeit unreal model, in part as it offers the advantages of the implants being highly detectable due to melanin secretion and the aggressive progression of such tumors [42,43,44].
The classic PCa model consists of the inoculation of tumor cells directly into the peritoneal cavity. Depending on the tumor cell line and the degree of aggressiveness desired for the model (i.e., MC38: 2–5 × 105 cells, and ID8: 5–10 × 106 cells), the number of cells is critical, as well as a broad spread of volumes where cells are suspended for peritoneal inoculation (range from 400 to 700 µL) to disseminate cells throughout the cavity [27,28,29,33]. The development of observable peritoneal implants takes time. For ID8 PCa, this occurs around day 30–40 after injection of 5 × 106 cells [10]. Several strategies have been used to increase the aggressiveness of the respective cell lines. Overexpression of vascular endothelial growth factor (VEGF) mediates increased tumor vascularization and correlates with poor prognosis. The ID8 cell line engineered to overexpress VEGF and, in some cases, the dendritic cells chemoattractant beta-defensin-29 (Defb29) has yielded dramatic increases in peritoneal implants and has reduced survival of the animals [35,45,46,47]. In addition, the overexpression of immunosuppressive molecules such as PD-L1 can promote PCa progression by inhibiting peritoneal cytotoxic lymphocytes [48].
Nevertheless, these kinds of peritoneal metastasis models do not reflect the biology underlying the onset of PCa since intraperitoneal (i.p.) inoculation spreads over the entire peritoneum. To address this, different studies have been carried out with a more realistic but laborious and often irreproducible model. This consists of generating orthotopic primary tumors (in the colon, ovary bursa, pancreas, etc.) and selecting cancer cells that have migrated to the secondary peritoneal organs localized as tumor implants or to the omentum after two or three weeks of primary tumor challenge. Then, consecutive passages are made in mice until a highly metastatic line is achieved. Using this selected aggressive cell line, PCa latency for experimental studies can be shortened [33,49,50,51,52]. The selection of these clones could also be useful for models such as ID8, where the development of peritoneal implants is very slow, even if 5 × 106 cells are inoculated into the peritoneal cavity. By selecting the proper clone, this time can be significantly reduced to 20–30 days to obtain peritoneal implants after i.p. challenge.
Altogether, syngeneic PCa models are feasible and convenient models to test immunotherapeutic agents delivered into the peritoneal cavity. A major drawback is the significant differences between the peritoneum of rodents and humans. While the omentum, a highly vascularized organ critical in the development of PCa is a large organ in humans, it does not play the same role in mice [53] (Figure 2).
Despite this fact, the omentum of mice is considered an interesting organ for immune studies of peritoneal carcinomatosis, especially to understand the role of resident macrophages that promote peritoneal carcinomatosis [54,55,56]. However, on the whole, syngeneic PCa models are not entirely a bona fide model for PCa in patients.

2.2. PCa Human Xenograft and PDX Models

PCa xenograft models are based on the transplantation of human cancer cells or tissue into the peritoneal cavity of immunodeficient mice, such as athymic nude, non-obese diabetic (NOD), severe combined immunodeficiency (SCID), or NOD SCID gamma mice (NSG). These mice are unable to generate an immune response against human cells and tumor engraftment is thus promoted in the peritoneal cavity. The xenografted PCa models have been studied using colon cancer (HCT-116, LS174T, HT29, SW480, SW620, LoVo, RKO, Caco-2, KM12, MDST8, HUTU80) [57,58,59,60,61], pancreatic cancer (Panc-1, TCC-Pan2, BxPC3, AsPC-1) [62,63,64], gastric cancer (60As6, HSC-44PE, HSC-58, MKN45-P) [56,57,58,59,60,61,62,63,64,65,66,67,68,69], and ovarian cancer (OVCAR3, OVCAR4, OVCAR5, OVCAR8, CAOV3, OVSAHO, OV2944-HM-1, SKOV-3) [59,70,71,72] cell lines. These models have been used to attain proofs of concept and explore treatments that determine in vivo tumor cell cytotoxicity of drugs, such as chemotherapeutic agents. However, the main drawback of these standard PCa models is the lack of an immunocompetent environment [15]. In fact, xenograft models have been controversial due to their poor accuracy in predicting clinical response. For instance, they do not consider the effect of anti-tumor immune responses or the tumor immune microenvironment (tumor-infiltrating lymphocytes, macrophages, myeloid-derived immuno-suppressor cells, and regulatory T cells, among others) in the progression of cancer. Therefore, the use of PCa xenograft models is not useless to test immunotherapeutic agents but may lack an important component if adaptive immunity is involved in the mechanism of action.
To overcome the limitations of PCa xenograft models, PCa PDX models are currently under development. PDX models are ideally suited for testing potential, promising, and “personalized” cancer therapeutics [73]. However, although PDX models have been well-studied in different primary tumors (e.g., melanoma, lung cancer, breast cancer), peritoneal carcinomatosis remains to be an unexploited personalized platform to test cancer treatments. In line with this, Elien De Thaye and colleagues recently established and characterized PDX from peritoneal metastasis of ovarian cancer for the first time [74]. They used fresh peritoneal tissue specimens from 10 patients with metastatic ovarian cancer. These tumor fragments were processed to tumor slurry and inoculated by orthotopic engraftment i.p. into SCID/Beige mice. Then, tumors were harvested from the peritoneal implant established in the mouse by passages until they became more prominent. Overall, De Thaye et al. demonstrated a feasible orthotopic PDX model from a peritoneal metastasis of ovarian cancer and a sophisticated translational research platform. Furthermore, the generation of NSG mice has facilitated the co-engraftment of human hematopoietic cells, peripheral blood mononuclear cells (PBMCs), or bone marrow precursors [75]. The standard method to repopulate a mouse with human immune cells is to intravenously engraft PBMCs (autologous or allogenic) in immunodeficient mice. However, in the case of a PCa model, it would be more appropriate to isolate immune cells from peritoneum lavage or ascites from PCa patients and co-engraft these into the peritoneum of PDX mice with already established tumors. With this model, the local effects and activation of peritoneal resident immune cells could be studied with immunotherapeutic agents [76].

2.3. PCa Genetically Induced Models

Genetically engineered mouse models (GEMM) (transgenic, knock-out, knock-in mice) have been used to study peritoneal metastasis. GEMM can faithfully recapitulate some human cancers genetically with very similar tumor microenvironment phenotype [77,78]. Some mice models expressing human tumor endogenous antigens such as carcinoembryonic antigen (CEA) as a transgene have shown better engraftment of mice tumor cells expressing this antigen in particular, as is the case of the MC38-CEA adenocarcinoma cell line [79]. However, more elaborate and realistic models which more faithfully represent clinical peritoneal metastasis are achieved through gene modification of primary aggressive peritoneal tumors (i.e., ovary, colon, stomach, pancreas) induced to study early peritoneal metastasis. In line with, some studies have used triple-mutant (TKO) mice: p53LSL-R172H/+ Dicer1flox/flox Ptenflox/flox Amhr2cre/+ [80,81]. This mouse p53R172H is equivalent to human p53R175H and is one of the most common p53 mutations in ovarian high-grade serous carcinoma (HGSCs) [82]. In TKO mouse, the tumors begin to form in the fallopian tube 1–2 months after birth. All mice develop PCa, with severe hemorrhagic ascites, leading to death (six months after birth).
Another recent and interesting work published by SSu-Hsueh Tseng, et al. reported on the development of a PCa model where the histological morphology and immune microenvironment are the same as peritoneal metastasis HGSCs in humans [83]. First, they observed that in immunocompetent mice the combination of shRNA-p53 (p53 suppression) with AKT and c-Myc oncogene overexpression in the peritoneum resulted in aggressive PCa with the presence of macroscopic peritoneal implants in only 21 days. This model consists of integrating the combination of shRNA-p53, AKT, and c-Myc via a sleeping beauty transposase system by i.p. administration, followed by electroporation in the abdominal cavity. Interestingly, this suppression of p53 and overexpression of AKT and c-Myc are able to overcome immunosurveillance and induce peritoneal tumors in immunocompetent mice too. Similarly, Sonia Iyer and colleagues constructed cell lines combining loss of Trp53 and overexpression of Ccne1, Akt2, and Trp53R172H, and driven by KRASG12V (KPCA) or Brd4 (BPCA) or Smarca4 (SPCA) overexpression [84]. Thus, this model represents an excellent and reliable platform for preclinical and translational PCa research and, more interestingly, the testing of immunotherapeutic agents. Additionally, this GEMM model can be used to investigate the initiation and progression of PCa, to identify potential biomarkers, and to predict the origin of peritoneal cancer spreading.

2.4. 3D-Biomimetics Peritoneal Implants

Advances in biotechnology and tissue engineering are increasingly being used in cancer research, as in other medical fields. One of these is the use of scaffolds as tools to test drugs or tumor implants with a structure that resembles the natural tumor [85]. Several studies have used these scaffolds to make an ex vivo tumor recapitulating the complexity of the tumor microenvironment. This has been achieved by including cancer cells, immune cells, fibroblasts, growth factors, and stromal components [86,87,88]. In PCa, this methodology has been used for the development of high-complexity bona fide peritoneal implants, although it is debatable whether this should be defined as an in vivo model per se. Therefore, this preclinical approach mimics the complexity (truly reflect in 3D models) of tumors developed in the peritoneum that may lead to potential therapies to improve the current PCa treatment options.
Accordingly, some groups are working on these 3D biomimetic peritoneal tumors. Daniela Loessner and colleagues observed that kallikrein-related peptidase 4, 5, 6, and 7 (KLK4-7) overexpression in ovarian cancer cells spheroids with integrin activation produces PCa after nine weeks of the inoculation in the peritoneal cavity in mice [88]. Moreover, in 2019, the same group recreated ex vivo ovarian tumor peritoneal metastasis. They used “3D co-cultured” hydrogel-encapsulated ovarian cancer cells with mesothelial cell-layered melt electrospun written scaffolds. These ex vivo tumor implants engrafted well and mimicked the same progression, invasion, and microenvironment as peritoneal implants of natural origin in mice [87].
Peritoneal implants present a complex structure, and the challenge of 3D culture is to mimic these ex vivo as realistically as possible so as to improve engraftment and the clinical biology in animal models. In order to achieve this, Emiel De Jaeghere et al. took into account the heterocellularity of 3D scaffolds in order to generate scaffolds which could then be implanted in the peritoneal cavity more successfully [86]. For that purpose, this group used polylactic acid (PLA) scaffolds with collagen type I hydrogel, co-seeding SKOV-3 cells (ovarian cancer), and cancer-associated fibroblasts (CAFs) not only to enhance the paracrine factor to improve spheroid formation in vitro, but also to enhance cancer cell survival, and angiogenesis. This model also seeks to improve the cellular composition and architecture of the tumor microenvironment and the organization of host cells in vivo. In fact, it was observed that CAFs, endothelial cells, macrophages, and cancer cells showed similar features when compared with patient-derived peritoneal metastasis. The authors concluded that this scaffold model represents a promising platform for the preclinical study of drug penetration and efficacy following delivery of intraperitoneal chemotherapy, among other therapeutic agents [86].

3. Concluding Remarks

Overall, PCa models have both advantages and drawbacks (Figure 3). Therefore, the read-out of the results obtained in mice should always be interpreted with caution. The complexity of cancer development and all the biological factors involved make it even more difficult to construct an ideal preclinical syngeneic model. Even PDX models have sometimes been shown not to correlate with the therapeutic outcome observed in the same patient from whom the model was derived.
In general terms, the human peritoneum environment where PCa develops is highly dynamic and is, at least currently, unmatched in mice with the same tissue structure and biology. However, syngeneic, humanized, personalized PDX, GEMM models, or approaches using biotechnology for 3D tumors have offered proofs of concept enabling the preclinical study of promising therapeutic alternatives in patients with PCa. Unfortunately, the currently dismal survival figures for PCa will only become worse in the following years due to the collateral COVID-19 impact associated with the cancellation of multiple routine medical exams to detect early gastrointestinal or gynecologic primary tumors. For this reason, exploring new PCa animal models as a platform to test, improve and/or predict the appropriate therapeutic interventions in patients should be a cancer research priority in the coming years.

Author Contributions

Conceptualization, F.A.; Writing—original draft preparation, Á.B., P.B. and F.A.; Writing—review and editing, Á.B., L.A., C.A.D.T., M.F.-S., A.C., J.M.-E., Á.T., I.M., P.B. and F.A.; Supervision, I.M., P.B. and F.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Instituto de Salud Carlos III (PI20/00002, PI19/01128,) cofinanced by Fondos FEDER “A way to make Europe” and Joint Translational Call for Proposals 2015 (JTC 2015), TRANSCAN456 2 (code: TRS-2016-00000371), Gobierno de Navarra Proyecto LINTERNA Ref.: 0011-1411-2020-000075. This project has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 765394. F.A. receives a Miguel Servet I (CP19/00114) contract from ISCIII (Instituto de Salud Carlos III) co-financed by FSE (Fondo Social Europeo) "Investing in your future". Á.B. is recipients of PFIS fellowship from ISCIII (FI20/00058), and M.F.-S. is recipients for a fellowship of the Aid Program Assigned to Projects from the University of Navarra. Á.T. has received funding from La Caixa research foundation LCF/BQ/LR18/11640014.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to Paul Miller for English editing. The figures were created by BioRender website platform.

Conflicts of Interest

J.M.-E. is an employee of Bavarian Nordic. I.M. reports receiving commercial research grants from BMS, Bioncotech, Alligator, Pfizer, Leadartis and Roche; has received speakers bureau honoraria from MSD; and is a consultant or advisory board member for BMS, Roche, Genmab, F-Star, Bioncotech, Bayer, Numab, Pieris, Alligator, and Merck Serono.

References

  1. Coccolini, F.; Gheza, F.; Lotti, M.; Virzi, S.; Iusco, D.; Ghermandi, C.; Melotti, R.; Baiocchi, G.; Giulini, S.M.; Ansaloni, L.; et al. Peritoneal carcinomatosis. World J. Gastroenterol. 2013, 19, 6979–6994. [Google Scholar] [CrossRef] [PubMed]
  2. McMullen, J.R.W.; Selleck, M.; Wall, N.R.; Senthil, M. Peritoneal carcinomatosis: Limits of diagnosis and the case for liquid biopsy. Oncotarget 2017, 8, 43481–43490. [Google Scholar] [CrossRef] [Green Version]
  3. Gamboa, A.C.; Winer, J.H. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for gastric cancer. Cancers 2019, 11, 1662. [Google Scholar] [CrossRef] [Green Version]
  4. Chia, C.S.; You, B.; Decullier, E.; Vaudoyer, D.; Lorimier, G.; Abboud, K.; Bereder, J.M.; Arvieux, C.; Boschetti, G.; Glehen, O.; et al. Patients with peritoneal carcinomatosis from gastric cancer treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy: Is cure a possibility? Ann. Surg. Oncol. 2016, 23, 1971–1979. [Google Scholar] [CrossRef] [PubMed]
  5. Teixeira Farinha, H.; Grass, F.; Labgaa, I.; Pache, B.; Demartines, N.; Hubner, M. Inflammatory response and toxicity after pressurized intraperitoneal aerosol chemotherapy. J. Cancer 2018, 9, 13–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Malfroy, S.; Wallet, F.; Maucort-Boulch, D.; Chardonnal, L.; Sens, N.; Friggeri, A.; Passot, G.; Glehen, O.; Piriou, V. Complications after cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for treatment of peritoneal carcinomatosis: Risk factors for ICU admission and morbidity prognostic score. Surg. Oncol. 2016, 25, 6–15. [Google Scholar] [CrossRef]
  7. Canda, A.E.; Sokmen, S.; Terzi, C.; Arslan, C.; Oztop, I.; Karabulut, B.; Ozzeybek, D.; Sarioglu, S.; Fuzun, M. Complications and toxicities after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann. Surg. Oncol. 2013, 20, 1082–1087. [Google Scholar] [CrossRef]
  8. Ireson, C.R.; Alavijeh, M.S.; Palmer, A.M.; Fowler, E.R.; Jones, H.J. The role of mouse tumour models in the discovery and development of anticancer drugs. Br. J. Cancer 2019, 121, 101–108. [Google Scholar] [CrossRef] [PubMed]
  9. Buque, A.; Galluzzi, L. Modeling tumor immunology and immunotherapy in mice. Trends Cancer 2018, 4, 599–601. [Google Scholar] [CrossRef]
  10. Zhang, L.; Yang, N.; Garcia, J.R.; Mohamed, A.; Benencia, F.; Rubin, S.C.; Allman, D.; Coukos, G. Generation of a syngeneic mouse model to study the effects of vascular endothelial growth factor in ovarian carcinoma. Am. J. Pathol. 2002, 161, 2295–2309. [Google Scholar] [CrossRef] [Green Version]
  11. Taylor, M.A.; Hughes, A.M.; Walton, J.; Coenen-Stass, A.M.L.; Magiera, L.; Mooney, L.; Bell, S.; Staniszewska, A.D.; Sandin, L.C.; Barry, S.T.; et al. Longitudinal immune characterization of syngeneic tumor models to enable model selection for immune oncology drug discovery. J. Immunother. Cancer 2019, 7, 328. [Google Scholar] [CrossRef] [PubMed]
  12. Gebreyohannes, Y.K.; Burton, E.A.; Wozniak, A.; Matusow, B.; Habets, G.; Wellens, J.; Cornillie, J.; Lin, J.; Nespi, M.; Wu, G.; et al. PLX9486 shows anti-tumor efficacy in patient-derived, tyrosine kinase inhibitor-resistant KIT-mutant xenograft models of gastrointestinal stromal tumors. Clin. Exp. Med. 2019, 19, 201–210. [Google Scholar] [CrossRef] [PubMed]
  13. Sanmamed, M.F.; Chester, C.; Melero, I.; Kohrt, H. Defining the optimal murine models to investigate immune checkpoint blockers and their combination with other immunotherapies. Ann. Oncol. 2016, 27, 1190–1198. [Google Scholar] [CrossRef]
  14. Xu, C.; Li, X.; Liu, P.; Li, M.; Luo, F. Patient-derived xenograft mouse models: A high fidelity tool for individualized medicine. Oncol. Lett. 2019, 17, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Walrath, J.C.; Hawes, J.J.; Van Dyke, T.; Reilly, K.M. Genetically engineered mouse models in cancer research. Adv. Cancer Res. 2010, 106, 113–164. [Google Scholar] [PubMed] [Green Version]
  16. DuPage, M.; Jacks, T. Genetically engineered mouse models of cancer reveal new insights about the antitumor immune response. Curr. Opin. Immunol. 2013, 25, 192–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Smith, H.W.; Muller, W.J. Transgenic mouse models-a seminal breakthrough in oncogene research. Cold Spring Harb. Protoc. 2013, 2013, 1099–1108. [Google Scholar] [CrossRef] [Green Version]
  18. Hanahan, D.; Wagner, E.F.; Palmiter, R.D. The origins of oncomice: A history of the first transgenic mice genetically engineered to develop cancer. Genes Dev. 2007, 21, 2258–2270. [Google Scholar] [CrossRef] [Green Version]
  19. Kim, H.; Kim, M.; Im, S.K.; Fang, S. Mouse Cre-LoxP system: General principles to determine tissue-specific roles of target genes. Lab. Anim. Res. 2018, 34, 147–159. [Google Scholar] [CrossRef] [Green Version]
  20. Ng, S.R.; Rideout, W.M., 3rd; Akama-Garren, E.H.; Bhutkar, A.; Mercer, K.L.; Schenkel, J.M.; Bronson, R.T.; Jacks, T. CRISPR-mediated modeling and functional validation of candidate tumor suppressor genes in small cell lung cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 513–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Kemp, C.J. Animal models of chemical carcinogenesis: Driving breakthroughs in cancer research for 100 Years. Cold Spring Harb. Protoc. 2015, 2015, 865–874. [Google Scholar] [CrossRef] [Green Version]
  22. Kishimoto, H.; Zhao, M.; Hayashi, K.; Urata, Y.; Tanaka, N.; Fujiwara, T.; Penman, S.; Hoffman, R.M. In vivo internal tumor illumination by telomerase-dependent adenoviral GFP for precise surgical navigation. Proc. Natl. Acad. Sci. USA 2009, 106, 14514–14517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chambers, L.M.; Esakov, E.; Braley, C.; AlHilli, M.; Michener, C.; Reizes, O. Use of transabdominal ultrasound for the detection of intra-peritoneal tumor engraftment and growth in mouse xenografts of epithelial ovarian cancer. PLoS ONE 2020, 15, e0228511. [Google Scholar] [CrossRef] [PubMed]
  24. Chiriva-Internati, M.; Yu, Y.; Mirandola, L.; Jenkins, M.R.; Chapman, C.; Cannon, M.; Cobos, E.; Kast, W.M. Cancer testis antigen vaccination affords long-term protection in a murine model of ovarian cancer. PLoS ONE 2010, 5, e10471. [Google Scholar] [CrossRef] [PubMed]
  25. Miller, A.M.; Lemke-Miltner, C.D.; Blackwell, S.; Tomanek-Chalkley, A.; Gibson-Corely, K.N.; Coleman, K.L.; Weiner, G.J.; Chan, C.H.F. Intraperitoneal CMP-001: A novel immunotherapy for treating peritoneal carcinomatosis of gastrointestinal and pancreaticobiliary cancer. Ann. Surg. Oncol. 2021, 28, 1187–1197. [Google Scholar] [CrossRef] [PubMed]
  26. Takahashi, Y.; Saga, Y.; Koyanagi, T.; Takei, Y.; Machida, S.; Taneichi, A.; Mizukami, H.; Sato, Y.; Matsubara, S.; Fujiwara, H. The angiogenesis regulator vasohibin-1 inhibits ovarian cancer growth and peritoneal dissemination and prolongs host survival. Int. J. Oncol. 2015, 47, 2057–2063. [Google Scholar] [CrossRef] [Green Version]
  27. Abou-Elkacem, L.; Arns, S.; Brix, G.; Gremse, F.; Zopf, D.; Kiessling, F.; Lederle, W. Regorafenib inhibits growth, angiogenesis, and metastasis in a highly aggressive, orthotopic colon cancer model. Mol. Cancer Ther. 2013, 12, 1322–1331. [Google Scholar] [CrossRef] [Green Version]
  28. Lee, Y.S.; Lee, W.S.; Kim, C.W.; Lee, S.J.; Yang, H.; Kong, S.J.; Ning, J.; Yang, K.M.; Kang, B.; Kim, W.R.; et al. Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer. J. Immunother. Cancer 2020, 8, e000857. [Google Scholar] [CrossRef] [PubMed]
  29. Sedlacek, A.L.; Gerber, S.A.; Randall, T.D.; Van Rooijen, N.; Frelinger, J.G.; Lord, E.M. Generation of a dual-functioning antitumor immune response in the peritoneal cavity. Am. J. Pathol. 2013, 183, 1318–1328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Taibi, A.; Albouys, J.; Jacques, J.; Perrin, M.L.; Yardin, C.; Durand Fontanier, S.; Bardet, S.M. Comparison of implantation sites for the development of peritoneal metastasis in a colorectal cancer mouse model using non-invasive bioluminescence imaging. PLoS ONE 2019, 14, e0220360. [Google Scholar] [CrossRef]
  31. Chen, C.H.; Kuo, C.Y.; Chen, S.H.; Mao, S.H.; Chang, C.Y.; Shalumon, K.T.; Chen, J.P. Thermosensitive injectable hydrogel for simultaneous intraperitoneal delivery of doxorubicin and prevention of peritoneal adhesion. Int. J. Mol. Sci. 2018, 19, 1373. [Google Scholar] [CrossRef] [Green Version]
  32. Yoshimura, M.; Fujiwara, H.; Kubota, T.; Amaike, H.; Takashima, K.; Inada, S.; Atsuji, K.; Araki, Y.; Matsumoto, K.; Nakamura, T.; et al. Possible inhibition of cancer cell adhesion to the extracellular matrix in NK4-induced suppression of peritoneal implantation. Anticancer Res. 2005, 25, 3847–3854. [Google Scholar] [PubMed]
  33. Cai, Q.; Yan, L.; Xu, Y. Anoikis resistance is a critical feature of highly aggressive ovarian cancer cells. Oncogene 2015, 34, 3315–3324. [Google Scholar] [CrossRef] [Green Version]
  34. Flies, D.B.; Higuchi, T.; Harris, J.C.; Jha, V.; Gimotty, P.A.; Adams, S.F. Immune checkpoint blockade reveals the stimulatory capacity of tumor-associated CD103(+) dendritic cells in late-stage ovarian cancer. Oncoimmunology 2016, 5, e1185583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Janat-Amsbury, M.M.; Yockman, J.W.; Anderson, M.L.; Kieback, D.G.; Kim, S.W. Comparison of ID8 MOSE and VEGF-modified ID8 cell lines in an immunocompetent animal model for human ovarian cancer. Anticancer Res. 2006, 26, 2785–2789. [Google Scholar]
  36. Wilkinson-Ryan, I.; Pham, M.M.; Sergent, P.; Tafe, L.J.; Berwin, B.L. A syngeneic mouse model of epithelial ovarian cancer Port site metastases. Transl. Oncol. 2019, 12, 62–68. [Google Scholar] [CrossRef]
  37. Fujimori, D.; Kinoshita, J.; Yamaguchi, T.; Nakamura, Y.; Gunjigake, K.; Ohama, T.; Sato, K.; Yamamoto, M.; Tsukamoto, T.; Nomura, S.; et al. Established fibrous peritoneal metastasis in an immunocompetent mouse model similar to clinical immune microenvironment of gastric cancer. BMC Cancer 2020, 20, 1014. [Google Scholar] [CrossRef] [PubMed]
  38. Banan, B.; Beckstead, J.A.; Dunavant, L.E.; Sohn, Y.; Adcock, J.M.; Nomura, S.; Abumrad, N.; Goldenring, J.R.; Fingleton, B. Development of a novel murine model of lymphatic metastasis. Clin. Exp. Metastasis. 2020, 37, 247–255. [Google Scholar] [CrossRef]
  39. Greco, S.H.; Tomkotter, L.; Vahle, A.K.; Rokosh, R.; Avanzi, A.; Mahmood, S.K.; Deutsch, M.; Alothman, S.; Alqunaibit, D.; Ochi, A.; et al. TGF-beta blockade reduces mortality and metabolic changes in a validated murine model of pancreatic cancer cachexia. PLoS ONE 2015, 10, e0132786. [Google Scholar] [CrossRef]
  40. Akimoto, M.; Iizuka, M.; Kanematsu, R.; Yoshida, M.; Takenaga, K. Anticancer effect of ginger extract against pancreatic cancer cells mainly through reactive oxygen species-mediated autotic cell death. PLoS ONE 2015, 10, e0126605. [Google Scholar] [CrossRef]
  41. Yu, Z.; Zhao, S.; Ren, L.; Wang, L.; Chen, Z.; Hoffman, R.M.; Zhou, J. Pancreatic cancer-derived exosomes promote tumor metastasis and liver pre-metastatic niche formation. Oncotarget 2017, 8, 63461–63483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nowacki, M.; Wisniewski, M.; Werengowska-Ciecwierz, K.; Roszek, K.; Czarnecka, J.; Lakomska, I.; Kloskowski, T.; Tyloch, D.; Debski, R.; Pietkun, K.; et al. Nanovehicles as a novel target strategy for hyperthermic intraperitoneal chemotherapy: A multidisciplinary study of peritoneal carcinomatosis. Oncotarget 2015, 6, 22776–22798. [Google Scholar] [CrossRef] [PubMed]
  43. Taniguchi, S.; Takeoka, M.; Ehara, T.; Hashimoto, S.; Shibuki, H.; Yoshimura, N.; Shigematsu, H.; Takahashi, K.; Katsuki, M. Structural fragility of blood vessels and peritoneum in calponin h1-deficient mice, resulting in an increase in hematogenous metastasis and peritoneal dissemination of malignant tumor cells. Cancer Res. 2001, 61, 7627–7634. [Google Scholar] [PubMed]
  44. Weiss, J.M.; Davies, L.C.; Karwan, M.; Ileva, L.; Ozaki, M.K.; Cheng, R.Y.; Ridnour, L.A.; Annunziata, C.M.; Wink, D.A.; McVicar, D.W. Itaconic acid mediates crosstalk between macrophage metabolism and peritoneal tumors. J. Clin. Investig. 2018, 128, 3794–3805. [Google Scholar] [CrossRef] [PubMed]
  45. Conejo-Garcia, J.R.; Benencia, F.; Courreges, M.C.; Kang, E.; Mohamed-Hadley, A.; Buckanovich, R.J.; Holtz, D.O.; Jenkins, A.; Na, H.; Zhang, L.; et al. Tumor-infiltrating dendritic cell precursors recruited by a beta-defensin contribute to vasculogenesis under the influence of Vegf-A. Nat Med. 2004, 10, 950–958. [Google Scholar] [CrossRef] [PubMed]
  46. Shukla, S.; Wang, C.; Beiss, V.; Steinmetz, N.F. Antibody response against cowpea mosaic viral nanoparticles improves in situ vaccine efficacy in ovarian cancer. ACS Nano 2020, 14, 2994–3003. [Google Scholar] [CrossRef] [PubMed]
  47. Cubillos-Ruiz, J.R.; Engle, X.; Scarlett, U.K.; Martinez, D.; Barber, A.; Elgueta, R.; Wang, L.; Nesbeth, Y.; Durant, Y.; Gewirtz, A.T.; et al. Polyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunity. J. Clin. Investig. 2009, 119, 2231–2244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Abiko, K.; Mandai, M.; Hamanishi, J.; Yoshioka, Y.; Matsumura, N.; Baba, T.; Yamaguchi, K.; Murakami, R.; Yamamoto, A.; Kharma, B.; et al. PD-L1 on tumor cells is induced in ascites and promotes peritoneal dissemination of ovarian cancer through CTL dysfunction. Clin. Cancer Res. 2013, 19, 1363–1374. [Google Scholar] [CrossRef] [Green Version]
  49. Ward, K.K.; Tancioni, I.; Lawson, C.; Miller, N.L.; Jean, C.; Chen, X.L.; Uryu, S.; Kim, J.; Tarin, D.; Stupack, D.G.; et al. Inhibition of focal adhesion kinase (FAK) activity prevents anchorage-independent ovarian carcinoma cell growth and tumor progression. Clin. Exp. Metastasis. 2013, 30, 579–594. [Google Scholar] [CrossRef] [Green Version]
  50. Yang, T.; Wall, E.M.; Milne, K.; Theiss, P.; Watson, P.; Nelson, B.H. CD8+ T cells induce complete regression of advanced ovarian cancers by an interleukin (IL)-2/IL-15 dependent mechanism. Clin. Cancer Res. 2007, 13, 7172–7180. [Google Scholar] [CrossRef] [Green Version]
  51. Martin, S.D.; Brown, S.D.; Wick, D.A.; Nielsen, J.S.; Kroeger, D.R.; Twumasi-Boateng, K.; Holt, R.A.; Nelson, B.H. Low mutation burden in ovarian cancer may limit the utility of neoantigen-targeted vaccines. PLoS ONE 2016, 11, e0155189. [Google Scholar] [CrossRef]
  52. Sekiya, A.; Suzuki, S.; Tanaka, A.; Hattori, S.; Shimizu, Y.; Yoshikawa, N.; Koya, Y.; Kajiyama, H.; Kikkawa, F. Interleukin33 expression in ovarian cancer and its possible suppression of peritoneal carcinomatosis. Int. J. Oncol. 2019, 55, 755–765. [Google Scholar]
  53. Wilkosz, S.; Ireland, G.; Khwaja, N.; Walker, M.; Butt, R.; De Giorgio-Miller, A.; Herrick, S.E. A comparative study of the structure of human and murine greater omentum. Anat. Embryol. (Berl.) 2005, 209, 251–261. [Google Scholar] [CrossRef] [PubMed]
  54. Etzerodt, A.; Moulin, M.; Doktor, T.K.; Delfini, M.; Mossadegh-Keller, N.; Bajenoff, M.; Sieweke, M.H.; Moestrup, S.K.; Auphan-Anezin, N.; Lawrence, T. Tissue-resident macrophages in omentum promote metastatic spread of ovarian cancer. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef]
  55. Krishnan, V.; Tallapragada, S.; Schaar, B.; Kamat, K.; Chanana, A.M.; Zhang, Y.; Patel, S.; Parkash, V.; Rinker-Schaeffer, C.; Folkins, A.K.; et al. Omental macrophages secrete chemokine ligands that promote ovarian cancer colonization of the omentum via CCR1. Commun. Biol. 2020, 3, 524. [Google Scholar] [CrossRef] [PubMed]
  56. Van der Bij, G.J.; Bogels, M.; Oosterling, S.J.; Kroon, J.; Schuckmann, D.T.; De Vries, H.E.; Meijer, S.; Beelen, R.H.; Van Egmond, M. Tumor infiltrating macrophages reduce development of peritoneal colorectal carcinoma metastases. Cancer Lett. 2008, 262, 77–86. [Google Scholar] [CrossRef]
  57. Miyoshi, J.; Toden, S.; Yoshida, K.; Toiyama, Y.; Alberts, S.R.; Kusunoki, M.; Sinicrope, F.A.; Goel, A. MiR-139-5p as a novel serum biomarker for recurrence and metastasis in colorectal cancer. Sci. Rep. 2017, 7, 43393. [Google Scholar] [CrossRef]
  58. Bastiaenen, V.P.; Klaver, C.E.L.; Van Der Heijden, M.C.S.; Nijman, L.E.; Lecca, M.C.; Tanis, P.J.; Lenos, P.J.; Vermeulen, L. A mouse model for peritoneal metastases of colorectal origin recapitulates patient heterogeneity. Lab. Investig. 2020, 100, 1465–1474. [Google Scholar] [CrossRef]
  59. Yao, Y.; Zhou, Y.; Su, X.; Dai, L.; Yu, L.; Deng, H.; Gou, L.; Yang, J. Establishment and characterization of intraperitoneal xenograft models by co-injection of human tumor cells and extracellular matrix gel. Oncol. Lett. 2015, 10, 3450–3456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Qiu, C.; Li, Y.; Liang, X.; Qi, Y.; Chen, Y.; Meng, X.; Zheng, H.; Xu, Y.; Cai, S.; Cai, G.; et al. A study of peritoneal metastatic xenograft model of colorectal cancer in the treatment of hyperthermic intraperitoneal chemotherapy with Raltitrexed. Biomed. Pharm. 2017, 92, 149–156. [Google Scholar] [CrossRef]
  61. Mikula-Pietrasik, J.; Sosinska, P.; Maksin, K.; Kucinska, M.G.; Piotrowska, H.; Murias, M.; Wozniak, A.; Szpurek, D.; Ksiazek, K. Colorectal cancer-promoting activity of the senescent peritoneal mesothelium. Oncotarget 2015, 6, 29178–29195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Menen, R.S.; Hassanein, M.K.; Momiyama, M.; Suetsugu, A.; Moossa, A.R.; Hoffman, R.M.; Bouvet, M. Tumor-educated macrophages promote tumor growth and peritoneal metastasis in an orthotopic nude mouse model of human pancreatic cancer. Vivo 2012, 26, 565–569. [Google Scholar]
  63. Saimura, M.; Nagai, E.; Mizumoto, K.; Maehara, N.; Okino, H.; Katano, M.; Matsumoto, K.; Nakamura, T.; Narumi, K.; Nukiwa, T.; et al. Intraperitoneal injection of adenovirus-mediated NK4 gene suppresses peritoneal dissemination of pancreatic cancer cell line AsPC-1 in nude mice. Cancer Gene Ther. 2002, 9, 799–806. [Google Scholar] [CrossRef] [Green Version]
  64. Yanagihara, K.; Kubo, T.; Mihara, K.; Kuwata, T.; Ochiai, A.; Seyama, T.; Yokozaki, H. Development and biological analysis of a novel orthotopic peritoneal dissemination mouse model generated using a pancreatic ductal adenocarcinoma cell line. Pancreas 2019, 48, 315–322. [Google Scholar] [CrossRef]
  65. Okazaki, M.; Fushida, S.; Harada, S.; Tsukada, T.; Kinoshita, J.; Oyama, K.; Miyashita, T.; Ninomiya, I.; Ohta, T. Establishing a xenograft mouse model of peritoneal dissemination of gastric cancer with organ invasion and fibrosis. BMC Cancer 2017, 17, 23. [Google Scholar] [CrossRef] [Green Version]
  66. Yanagihara, K.; Takigahira, M.; Tanaka, H.; Komatsu, T.; Fukumoto, H.; Koizumi, F.; Nishio, K.; Ochiya, T.; Ino, Y.; Hirohashi, S. Development and biological analysis of peritoneal metastasis mouse models for human scirrhous stomach cancer. Cancer Sci. 2005, 96, 323–332. [Google Scholar] [CrossRef]
  67. Fujita, T.; Yanagihara, K.; Takeshita, F.; Aoyagi, K.; Nishimura, T.; Takigahira, M.; Chiwaki, F.; Fukagawa, T.; Katai, H.; Ochiya, T.; et al. Intraperitoneal delivery of a small interfering RNA targeting NEDD1 prolongs the survival of scirrhous gastric cancer model mice. Cancer Sci. 2013, 104, 214–222. [Google Scholar] [CrossRef] [PubMed]
  68. Yasumoto, K.; Koizumi, K.; Kawashima, A.; Saitoh, Y.; Arita, Y.; Shinohara, K.; Minami, T.; Nakayama, T.; Sakurai, H.; Takahashi, Y.; et al. Role of the CXCL12/CXCR4 axis in peritoneal carcinomatosis of gastric cancer. Cancer Res. 2006, 66, 2181–2187. [Google Scholar] [CrossRef] [Green Version]
  69. Mori, T.; Fujiwara, Y.; Yano, M.; Yasuda, T.; Takiguchi, S.; Miyata, H.; Viliotou, V.; Monden, M. A mouse model of early-stage peritoneal metastasis: Optimal RT-PCR-based method for detection of peritoneal micrometastases. Oncol. Rep. 2005, 13, 801–805. [Google Scholar] [CrossRef] [PubMed]
  70. Rezniczek, G.A.; Buggisch, J.; Sobilo, J.; Launay, A.; Lerondel, S.; Le Pape, A.; Ouaissi, M.; Gohler, D.; Senkal, M.; Giger-Pabst, U.; et al. Establishment of a mouse ovarian cancer and peritoneal metastasis model to study intraperitoneal chemotherapy. Cancers 2020, 12, 3818. [Google Scholar] [CrossRef]
  71. Mitra, A.K.; Davis, D.A.; Tomar, S.; Roy, L.; Gurler, H.; Xie, J.; Lantvit, D.D.; Cardenas, H.; Fang, F.; Liu, Y.; et al. In vivo tumor growth of high-grade serous ovarian cancer cell lines. Gynecol. Oncol. 2015, 138, 372–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Tanizaki, Y.; Kobayashi, A.; Toujima, S.; Shiro, M.; Mizoguchi, M.; Mabuchi, Y.; Yagi, S.; Minami, S.; Takikawa, O.; Ino, K. Indoleamine 2,3-dioxygenase promotes peritoneal metastasis of ovarian cancer by inducing an immunosuppressive environment. Cancer Sci. 2014, 105, 966–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Bhimani, J.; Ball, K.; Stebbing, J. Patient-derived xenograft models-the future of personalised cancer treatment. Br. J. Cancer 2020, 122, 601–602. [Google Scholar] [CrossRef] [PubMed]
  74. De Thaye, E.; Van De Vijver, K.; Van Der Meulen, J.; Taminau, J.; Wagemans, G.; Denys, H.; Van Dorpe, J.; Berx, G.; Ceelen, W.; Van Bocxlaer, J.; et al. Establishment and characterization of a cell line and patient-derived xenograft (PDX) from peritoneal metastasis of low-grade serous ovarian carcinoma. Sci. Rep. 2020, 10, 6688. [Google Scholar] [CrossRef] [Green Version]
  75. Shultz, L.D.; Brehm, M.A.; Garcia-Martinez, J.V.; Greiner, D.L. Humanized mice for immune system investigation: Progress, promise and challenges. Nat. Rev. Immunol. 2012, 12, 786–798. [Google Scholar] [CrossRef] [PubMed]
  76. Lin, S.; Huang, G.; Cheng, L.; Li, Z.; Xiao, Y.; Deng, Q.; Jiang, Y.; Li, B.; Lin, S.; Wang, S.; et al. Establishment of peripheral blood mononuclear cell-derived humanized lung cancer mouse models for studying efficacy of PD-L1/PD-1 targeted immunotherapy. MAbs 2018, 10, 1301–1311. [Google Scholar] [CrossRef] [Green Version]
  77. Gopinathan, A.; Tuveson, D.A. The use of GEM models for experimental cancer therapeutics. Dis Model Mech. 2008, 1, 83–86. [Google Scholar] [CrossRef] [Green Version]
  78. Kersten, K.; De Visser, K.E.; Van Miltenburg, M.H.; Jonkers, J. Genetically engineered mouse models in oncology research and cancer medicine. EMBO Mol. Med. 2017, 9, 137–153. [Google Scholar] [CrossRef]
  79. Abdul-Wahid, A.; Huang, E.H.; Lu, H.; Flanagan, J.; Mallick, A.I.; Gariepy, J. A focused immune response targeting the homotypic binding domain of the carcinoembryonic antigen blocks the establishment of tumor foci in vivo. Int. J. Cancer 2012, 131, 2839–2851. [Google Scholar] [CrossRef]
  80. Kim, J.; Coffey, D.M.; Creighton, C.J.; Yu, Z.; Hawkins, S.M.; Matzuk, M.M. High-grade serous ovarian cancer arises from fallopian tube in a mouse model. Proc. Natl. Acad. Sci. USA 2012, 109, 3921–3926. [Google Scholar] [CrossRef] [Green Version]
  81. Kim, O.; Park, E.Y.; Klinkebiel, D.L.; Pack, S.D.; Shin, Y.H.; Abdullaev, Z.; Emerson, R.E.; Coffey, D.M.; Kwon, S.Y.; Creighton, C.J.; et al. In vivo modeling of metastatic human high-grade serous ovarian cancer in mice. PLoS Genet. 2020, 16, e1008808. [Google Scholar] [CrossRef] [PubMed]
  82. Brachova, P.; Thiel, K.W.; Leslie, K.K. The consequence of oncomorphic TP53 mutations in ovarian cancer. Int. J. Mol. Sci. 2013, 14, 19257–19275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tseng, S.H.; Park, S.T.; Lam, B.; Tsai, Y.C.; Cheng, M.A.; Farmer, E.; Xing, D.; Hung, C.F. Novel, genetically induced mouse model that recapitulates the histological morphology and immunosuppressive tumor microenvironment of metastatic peritoneal carcinomatosis. J. Immunother. Cancer 2020, 8, e000480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Iyer, S.; Zhang, S.; Yucel, S.; Horn, H.; Smith, S.G.; Reinhardt, F.; Hoefsmit, E.; Assatova, B.; Casado, J.; Meinsohn, M.C.; et al. Genetically defined syngeneic mouse models of ovarian cancer as tools for the discovery of combination immunotherapy. Cancer Discov. 2020, 11, 384–407. [Google Scholar] [CrossRef]
  85. Turetta, M.; Ben, F.D.; Brisotto, G.; Biscontin, E.; Bulfoni, M.; Cesselli, D.; Colombatti, A.; Scoles, G.; Gigli, G.; Del Mercato, L.L. Emerging technologies for cancer research: Towards personalized medicine with microfluidic platforms and 3D tumor models. Curr. Med. Chem. 2018, 25, 4616–4637. [Google Scholar] [CrossRef] [PubMed]
  86. De Jaeghere, E.; De Vlieghere, E.; Van Hoorick, J.; Van Vlierberghe, S.; Wagemans, G.; Pieters, L.; Melsens, E.; Praet, M.; Van Dorpe, J.; Boone, M.N.; et al. Heterocellular 3D scaffolds as biomimetic to recapitulate the tumor microenvironment of peritoneal metastases in vitro and in vivo. Biomaterials 2018, 158, 95–105. [Google Scholar] [CrossRef]
  87. Loessner, D.; Rockstroh, A.; Shokoohmand, A.; Holzapfel, B.M.; Wagner, F.; Baldwin, J.; Boxberg, M.; Schmalfeldt, B.; Lengyel, E.; Clements, J.A.; et al. A 3D tumor microenvironment regulates cell proliferation, peritoneal growth and expression patterns. Biomaterials 2019, 190–191, 63–75. [Google Scholar] [CrossRef] [PubMed]
  88. Loessner, D.; Rizzi, S.C.; Stok, K.S.; Fuehrmann, T.; Hollier, B.; Magdolen, V.; Hutmacher, D.W.; Clements, J.A. A bioengineered 3D ovarian cancer model for the assessment of peptidase-mediated enhancement of spheroid growth and intraperitoneal spread. Biomaterials 2013, 34, 7389–7400. [Google Scholar] [CrossRef]
Figure 1. Peritoneal carcinomatosis mouse models. A repertoire of preclinical platforms to test new therapeutic opportunities and transfer the results to patients with peritoneal carcinomatosis. CAFs, cancer-associated fibroblasts; EP, electroporation; i.p., intraperitoneally; PBMCs, peripheral blood mononuclear cells; PCa, peritoneal carcinomatosis; PDX, patient-derived xenograft; PLA, polylactic acid; SB, sleeping beauty. Created with BioRender.com, accessed on 19 February 2021.
Figure 1. Peritoneal carcinomatosis mouse models. A repertoire of preclinical platforms to test new therapeutic opportunities and transfer the results to patients with peritoneal carcinomatosis. CAFs, cancer-associated fibroblasts; EP, electroporation; i.p., intraperitoneally; PBMCs, peripheral blood mononuclear cells; PCa, peritoneal carcinomatosis; PDX, patient-derived xenograft; PLA, polylactic acid; SB, sleeping beauty. Created with BioRender.com, accessed on 19 February 2021.
Cancers 13 00963 g001
Figure 2. Anatomy and localization of human and mouse omentum. The mouse omentum is a thin, slightly elongated, and vascularized tissue located under the stomach and between the spleen and pancreas. Created with BioRender.com, accessed on 19 February 2021.
Figure 2. Anatomy and localization of human and mouse omentum. The mouse omentum is a thin, slightly elongated, and vascularized tissue located under the stomach and between the spleen and pancreas. Created with BioRender.com, accessed on 19 February 2021.
Cancers 13 00963 g002
Figure 3. Benefits and limitations of PCa mouse models. Assessment of the current platforms for preclinical development of anticancer drugs or immunotherapeutic agents in PCa. Relative scores are represented as being suitable (green checkmark), partially suitable (yellow checkmark), and not suitable (red cross). Created with BioRender.com, accessed on 19 February 2021.
Figure 3. Benefits and limitations of PCa mouse models. Assessment of the current platforms for preclinical development of anticancer drugs or immunotherapeutic agents in PCa. Relative scores are represented as being suitable (green checkmark), partially suitable (yellow checkmark), and not suitable (red cross). Created with BioRender.com, accessed on 19 February 2021.
Cancers 13 00963 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bella, Á.; Di Trani, C.A.; Fernández-Sendin, M.; Arrizabalaga, L.; Cirella, A.; Teijeira, Á.; Medina-Echeverz, J.; Melero, I.; Berraondo, P.; Aranda, F. Mouse Models of Peritoneal Carcinomatosis to Develop Clinical Applications. Cancers 2021, 13, 963. https://doi.org/10.3390/cancers13050963

AMA Style

Bella Á, Di Trani CA, Fernández-Sendin M, Arrizabalaga L, Cirella A, Teijeira Á, Medina-Echeverz J, Melero I, Berraondo P, Aranda F. Mouse Models of Peritoneal Carcinomatosis to Develop Clinical Applications. Cancers. 2021; 13(5):963. https://doi.org/10.3390/cancers13050963

Chicago/Turabian Style

Bella, Ángela, Claudia Augusta Di Trani, Myriam Fernández-Sendin, Leire Arrizabalaga, Assunta Cirella, Álvaro Teijeira, José Medina-Echeverz, Ignacio Melero, Pedro Berraondo, and Fernando Aranda. 2021. "Mouse Models of Peritoneal Carcinomatosis to Develop Clinical Applications" Cancers 13, no. 5: 963. https://doi.org/10.3390/cancers13050963

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop