Research Paper
Organoids as preclinical models to improve intraperitoneal chemotherapy effectiveness for colorectal cancer patients with peritoneal metastases: Preclinical models to improve HIPEC

https://doi.org/10.1016/j.ijpharm.2017.07.084Get rights and content

Abstract

Background

Peritoneal metastases (PM), corresponding to tumor implants into the peritoneal cavity, are associated with impaired prognosis and low responsiveness to systemic chemotherapy. A new therapeutic approach has dramatically changed the prognosis of patients with PM from colorectal cancer (CRC), consisting in the association of a complete cytoreductive surgery followed by intraperitoneal chemotherapy associated to hyperthermia (HIPEC). Many drugs have been administered intraperitoneally, but no clear consensus has been approved. Therefore, relevant preclinical models are essentials for the efficient translation of treatments option into affected patients.

Method

Organoids, the last generation of preclinical models, were used to rationalize and improve intraperitoneal chemotherapy. We tested several cytotoxics, combination, effect of hyperthermia, exposure duration and frequency.

Results

Organoids were a representative model of response to chemotherapies used for the treatment of PM from CRC; 460 mg/m2 of oxaliplatin being the most efficient cytotoxic treatment. Repeated incubations with oxaliplatin; mimicking cycles of intraperitoneal treatment, resulted in an increased efficacy.

Conclusion & discussion

Organoids are relevant models to study the chemosensitivity of peritoneal metastases from CRCs. These models could be used for large scale drug screening strategies or personalized medicine, for colorectal carcinoma but also for PM from other origins.

Introduction

Relevant preclinical models are essentials for the efficient translation of treatments option into affected patients. Cancer cells lines and mice xenograft have proven to be valuable in anti-cancer drug discovery, but they both comprise caveats. A third generation of preclinical models, named organoids, holds great appeal. Organoids grow ex vivo from normal or cancer stem cells in tridimensional matrices based-gels to develop into fully differentiated “mini-organs” recapitulating the architecture and function of mature normal or pathological organs (Clevers, 2016, Sato et al., 2011a, Sato et al., 2011b). This technology is already applied to the personalized treatment of patients with cystic fibrosis and proof-of-concept have been made for gene therapy and regenerative medicine (Dekkers et al., 2013, Dekkers et al., 2016, Schwank et al., 2013). As organoids generated from primary cancer explants represent inter and intra-tumour heterogeneity, they show great promises for translational research and clinical applications (van de Wetering et al., 2015, Sachs and Clevers, 2014). To date, all studies use organoids to investigate the effect of systemic chemotherapy delivered intravenously. In this study, we used organoids generated from colorectal carcinomas to identify optimal cytotoxic-based protocols for the contact chemotherapy administered intraperitoneally to patients with peritoneal metastases.

Peritoneal metastases (PM) correspond to the tumour implants into the peritoneal cavity from different solid organs, mostly from colorectal, gastric and ovarian carcinomas. The diagnosis of PM represent in most of cases a fatal issue and treatment based on systemic chemotherapy still offers poor survival (Franko et al., 2016). Since nearly three decades, a new therapeutic approach has dramatically changed the prognosis of patients with PM from colorectal cancers and from primary peritoneal malignancies (i.e. pseudomyxoma and mesothelioma) (Sugarbaker, 1996, Elias et al., 2009, Verwaal et al., 2003). This treatment consists in the association of a complete cytoreductive surgery (CRS) of all the visible peritoneal implants (maximum residual tumour size of 1 mm), followed by intraperitoneal chemotherapy associated to hyperthermia (HIPEC) to treat the remaining non visible tumour implants. Direct intraperitoneal chemotherapy allows to higher local concentration, increases local biodisponibility, with the aim of improving the effect of the cytotoxic drugs. Beside CRS and HIPEC, local administration of chemotherapy has also been used either in patients with unresectable PM to enhance the response to chemotherapy by the local effect (Chan et al., 2017, Ishigami et al., 2010, Canbay et al., 2014, Yonemura et al., 2016), or after surgical resection of PM, in the adjuvant setting (Le Brun et al., 2014, Morgan et al., 2016)

Pharmacokinetics studies have been performed to determine the peritoneal and plasmatic concentrations of the cytotoxic administered directly into the peritoneum aiming to define the optimal cytotoxic and its optimal dosing. Indeed, the behavior of a drug administered into the peritoneal cavity is conveniently described by a two-compartment model: the first compartment represents the peritoneal cavity and the second is characterized by the central and peripheral compartments including the other tissues of the body that are in direct contact with the bloodstream. The intraperitoneal concentration of the cytotoxic can be defined as:CperitoneumCplasma=(plasmaclearance)+(cavityclearance)(cavityclearance)

Pharmacokinetic studies have shown that intravenous injections of 5-Fluorouracyl (5-FU) lead to a peritoneal concentration 100 fold lower than that obtained after direct intraperitoneal (IP) administration of 5-FU (Van der Speeten et al., 2010, Shimizu et al., 2014). These results have been confirmed with Irinotecan and Mitomycin-C. However, since the introduction of this new approach, many drugs, at different concentrations and in diverse combination have been described worldwide; but actually, no clear consensus has been approved (Ceelen and Flessner, 2010, Elias et al., 2000, Kusamura et al., 2008, Emoto et al., 2014, Braam et al., 2015). Therefore, the definition of the optimal cytotoxic drug, dosing, and administration route are still required.

Section snippets

Organoids retrieval and preparation from patient-derived xenograft

Four human colorectal tumour (IC-0010P (CRC 1), IGR-0012P (CRC 2), LRB-0008M(CRC 3), IRG-0014P (CRC 4)) from the CReMEC tumour collection were maintained in immunocompromised mice as previously described by Julien et al. (Elias et al., 2000). Briefly, small tumour fragments were subcutaneously engrafted on the flank of anesthetized mice (2.5% isoflurane). Tumour growth was measured at least once a week. When the volume reach 1500 mm3, mice were sacrificed and tumour were used for ex vivo

Assessing CRC cell viability using ATP-bioluminescence

We prepared organoids from patient-derived-xenografts (PDX) established from CRC tumours (Julien et al., 2012) as previously described (Sato et al., 2011b). In order to assess the chemosensitivity of CRC organoids to cytotoxic drugs, we opted for an ATP-bioluminescence assay (van de Wetering et al., 2015). Organoids were treated with 35 mg/m2 of Mitomycin-C (Mito-C), 4 or 400 mg/m2 of 5-Fluorouracil (5-FU) for 72 h. The number of viable cells was measured by quantifying the level of ATP for each

Discussion

The use of cancer cell lines and murine models to improve the chemotherapeutic treatment of peritoneal carcinomatosis has so far been limited for several reasons. First, mice models of peritoneal carcinomatosis have been described in the literature (Francescutti et al., 2013). However, in these small animals the low abdominal volume and the high peritoneal absorption, combined with the rapid tumour growth, weight loss, ascites formation and death are not representative of the human pathology.

Conclusions

We demonstrated that repeated contact chemotherapy allowed to maintain a low rate of viable cells. These results are in accordance with the use of repeated intraperitoneal chemotherapy cycles in patients with unresectable peritoneal disease We opted for repeated cycles of platinum derived drug exposure, not heated, as done in patients mostly from ovarian and gastric carcinomas (Sugarbaker et al., 1989, Tsubamoto et al., 2015, Fushida et al., 2008). Using this approach we could obtain a much

Acknowledgments

We thank the members of the Jaulin lab and Departments of Surgical and medical Oncology for discussion; Joel Raingeaud and Fotine Libanje for critical reading of the manuscript. This work was supported by the Gustave Roussy fund raising on colorectal cancer (Roulons pour le colon/mars bleu) and the ATIP-AVENIR program.

References (48)

  • N. Sachs et al.

    Organoid cultures for the analysis of cancer phenotypes

    Curr. Opin. Genet. Dev.

    (2014)
  • T. Sato et al.

    Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium

    Gastroenterology

    (2011)
  • G. Schwank et al.

    Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients

    Cell Stem Cell

    (2013)
  • A. Shimizu et al.

    ABL2/ARG tyrosine kinase mediates SEMA3F-induced RhoA inactivation and cytoskeleton collapse in human glioma cells

    J. Biol. Chem.

    (2008)
  • T. Shimizu et al.

    Hyperthermic intraperitoneal chemotherapy using a combination of mitomycin C,5-fluorouracil, and oxaliplatin in patients at high risk of colorectal peritoneal metastasis: a Phase I clinical study

    Eur. J. Surg. Oncol.

    (2014)
  • B.H.M. Williams et al.

    Repeat cytoreductive surgery (CRS) for recurrent colorectal peritoneal metastases: yes or no?

    Eur. J. Surg. Oncol.

    (2014)
  • Y. Yonemura et al.

    A comprehensive treatment for peritoneal metastases from gastric cancer with curative intent

    Eur. J. Surg. Oncol.

    (2016)
  • M. van de Wetering et al.

    Prospective derivation of a living organoid biobank of colorectal cancer patients

    Cell

    (2015)
  • D. Atallah et al.

    Thermal enhancement of oxaliplatin-induced inhibition of cell proliferation and cell cycle progression in human carcinoma cell lines

    Int. J. Hyperthermia

    (2004)
  • A. Brouquet et al.

    The second procedure combining complete cytoreductive surgery and intraperitoneal chemotherapy for isolated peritoneal recurrence: postoperative course and long-term outcome

    Ann. Surg. Oncol.

    (2009)
  • E. Canbay et al.

    Outcome data of patients with peritoneal carcinomatosis from gastric origin treated by a strategy of bidirectional chemotherapy prior to cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in a single specialized center in Japan

    Ann. Surg. Oncol.

    (2014)
  • W.P. Ceelen et al.

    Intraperitoneal therapy for peritoneal tumors: biophysics and clinical evidence

    Nat. Rev. Clin. Oncol.

    (2010)
  • J.F. Dekkers et al.

    A functional CFTR assay using primary cystic fibrosis intestinal organoids

    Nat. Med.

    (2013)
  • J.F. Dekkers et al.

    Characterizing responses to CFTR-modulating drugs using rectal organoids derived from subjects with cystic fibrosis

    Sci. Transl. Med.

    (2016)
  • Cited by (19)

    • The cross-talk between tumor cells and activated fibroblasts mediated by lactate/BDNF/TrkB signaling promotes acquired resistance to anlotinib in human gastric cancer

      2021, Redox Biology
      Citation Excerpt :

      In general, patient-derived organoids (PDOs) are 3D cultures composed of multiple organ-specific cell types that can recapitulate the architecture and gene expression profiles as well as some key features and functions of their corresponding organs. Multiple studies have now identified that PDOs are robust tools to mimic the drug responsiveness in gastrointestinal cancers [28–30]. In order to further confirm above protection role of CAFs in human GC, we established PDO from GC tumor tissue.

    • Survival after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for colorectal peritoneal metastases: A systematic review and discussion of latest controversies

      2021, Surgeon
      Citation Excerpt :

      Ubink and colleagues have shown that colorectal peritoneal organoids can be used to explore the efficacy of both novel and current HIPEC regimens. In future, such technology may drive patient-tailored HIPEC.67,68 Cytoreductive surgery plus HIPEC in combination with systemic modern chemotherapy regimens is safe and feasible for the management of CRPM.

    • Use of hyperthermia versus normothermia during intraperitoneal chemoperfusion with oxaliplatin for colorectal peritoneal carcinomatosis: A propensity score matched analysis

      2019, European Journal of Surgical Oncology
      Citation Excerpt :

      Animal studies have generated conflicting results regarding the anticancer benefit of intraperitoneal hyperthermia, with some authors reporting improved and others a similar or even a decreased tumor control or animal survival. In a recent experimental study using patient derived CRC organoids, hyperthermic administration did not enhance the cytotoxicity of OX [26]. Finally, when compared to normothemia, clinical hyperthermic chemoperfusion increases systemic OX levels.

    View all citing articles on Scopus
    View full text