Summary

Ovarian Cancer Patient-Derived Organoid Models for Pre-Clinical Drug Testing

Published: September 15, 2023
doi:

Summary

We present a protocol that can be used to conduct therapeutic drug testing with patient-derived ovarian cancer organoids.

Abstract

Ovarian cancer is a fatal gynecologic cancer and the fifth leading cause of cancer death among women in the United States. Developing new drug treatments is crucial to advancing healthcare and improving patient outcomes. Organoids are in-vitro three-dimensional multicellular miniature organs. Patient-derived organoid (PDO) models of ovarian cancer may be optimal for drug screening because they more accurately recapitulate tissues of interest than two-dimensional cell culture models and are inexpensive compared to patient-derived xenografts. In addition, ovarian cancer PDOs mimic the variable tumor microenvironment and genetic background typically observed in ovarian cancer. Here, a method is described that can be used to test conventional and novel drugs on PDOs derived from ovarian cancer tissue and ascites. A luminescence-based adenosine triphosphate (ATP) assay is used to measure viability, growth rate, and drug sensitivity. Drug screens in PDOs can be completed in 7-10 days, depending on the rate of organoid formation and drug treatments.

Introduction

Although rare, ovarian cancer is one of the most lethal gynecological cancers1,2. A challenge in developing new treatments is that ovarian cancer is heterogeneous, and the tumor microenvironment differs greatly among patients. Additionally, many ovarian cancers develop resistance to platinum-based chemotherapy and poly (ADP-ribose) polymerase inhibitors, highlighting the need for greater therapeutic options3,4,5.

One approach that may be useful in identifying new therapeutics is using patient-derived organoids (PDOs). Organoids are three-dimensional clusters of multiple cell types that self-organize and form in vitro "mini-organs"6,7,8,9,10. Organoids can recapitulate important tissue morphology and gene expression profiles11,12. Some of the first organoids were derived from intestinal, gastric, and colon cancer cells from both mice and humans8,9,13. Long-lived organoid cultures have been established from a wide range of benign and malignant tissues, including the bladder, colon, stomach, pancreas, brain, retina, and liver14,15,16. We previously demonstrated methods to establish PDOs from ovarian cancer tumors and ascites samples17. PDOs can be used to study molecular characteristics, cellular mechanisms, and novel drug treatments18,19,20. PDOs have several advantages over traditional two-dimensional primary cell cultures for drug screening. Although primary two-dimensional cultures are a low-cost method for drug screens, primary cell cultures are single-cell types and lack the three-dimensional architecture of tumors21,22,23. Nevertheless, PDOs are a precious resource, and cost-effective protocols are needed to optimize their use in therapeutic drug screening.

This article describes an in vitro method to use ovarian cancer PDOs to test the effects of known or candidate drugs. Whereas current medium- and high-throughput drug screens using PDOs require expensive automated dispensing instruments24,25,26, this cost-effective method uses readily available basic lab supplies and an ATP-based cell viability assay in a standard 96-well plate format (Figure 1A). This method will facilitate preliminary tests of novel ovarian cancer drugs prior to scaling up to larger screens27,28. Although ovarian cancer PDOs are used here, this method can be applied to other cancer organoid models.

Protocol

The collection of human specimens for this research was approved by the Washington University School of Medicine Institutional Review Board. All eligible patients over the age of 18 years had a diagnosis or presumed diagnosis of high-grade serous ovarian cancer and were willing and able to provide informed consent. The tumor tissue from either primary or metastatic sites, in addition to ascites and pleural fluid, were obtained from consented patients at the time of care. 1. Selection of …

Representative Results

These results illustrate the response of two PDOs to the chemotherapy drug carboplatin, which is used to treat ovarian cancer. Organoids were derived from tumor biopsy (PDO #1) and from ascites (PDO #2). These organoids were selected based on their perceived doubling time (1-2 days) and morphological appearance (formation of many large organoids). Both PDO #1 and PDO #2 were plated on Day -2, at passage two, and carboplatin was added on Day 0. We tested the following carboplatin concentrations diluted in Advance Organoid…

Discussion

This article describes a method that can be used to assess the therapeutic effects of conventional or novel drugs on ovarian cancer PDOs. Researchers must consider several issues before conducting the viability assay in the PDO model.

First, when selecting a PDO to use in the viability assay, one must determine the ideal organoid type (tumor vs. ascites) and passage number for their needs. In our experience, ascites-derived PDOs grow more rapidly and are easier to generate than tumor-…

Disclosures

The authors have nothing to disclose.

Acknowledgements

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under Award Number R01CA243511. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The authors thank Deborah Frank for her editorial comments.

Materials

1.5 mL Plastic Tubes
15 mL Plastic Tubes
96 well Flat Black Plates MidSci 781968
Advance Organoid Media  see Graham et al 2022 (Jove)
Advanced DMEM/F12 Thermo Fisher 12634028
Automated Cell Counter Thermo Fisher AMQAX1000
Brightfield Microscope
Carboplatin  Teva Pharmaceuticals USA NDC 00703-4246-01
CellTiter-Glo 3D Viability  Promega G9681
Cultrex R & D Systems 3533-010-02
DMSO Sigma Aldrich D2650-100ML
Glutamax Life Technologies 35050061
GR Calculator  http://www.grcalculator.org Online calculator
GraphPad Prism GraphPad Software, Inc.
HEPES Life Technologies 15630080
Matrigel Corning 354230
Microsoft Excel Microsoft
Penicillin-Streptomycin Thermo Fisher 15140122
Plate Rocker
Sterile P10, P200, and P1000 Barrier Sterile Pipette Tips
Sterile P10, P200, and P1000 Pipettes
Tecan Infinte 200Pro Plate Reader; i-Control Software Tecan
TrypLE Thermo Fisher 12605010 Organoid dissociation reagent

References

  1. Matulonis, U. A., Sood, A. K., Fallowfield, L., Howitt, B. E., Sehouli, J. m., Karlan, B. Y. Ovarian cancer. Nature Reviews Disease Primers. 2 (1), 16061 (2016).
  2. Siegel, R. L., Miller, K. D., Jemal, A. Cancer statistics, 2019. CA: A Cancer Journal for Clinicians. 69 (1), 7-34 (2019).
  3. Christie, E. L., Bowtell, D. D. L. Acquired chemotherapy resistance in ovarian cancer. Annals of Oncology. 28 (suppl_8), viii13-viii15 (2017).
  4. Wang, L., Wang, Q., Xu, Y., Cui, M., Han, L. Advances in the treatment of ovarian cancer using PARP inhibitors and the underlying mechanism of resistance. Current Drug Targets. 21 (2), 167-178 (2020).
  5. Wang, Q., Peng, H., Qi, X., Wu, M., Zhao, X. Targeted therapies in gynecological cancers: a comprehensive review of clinical evidence. Signal Transduction and Targeted Therapy. 5 (1), 137 (2020).
  6. Hofer, M., Lutolf, M. P. Engineering organoids. Nature Reviews Materials. 6 (5), 402-420 (2021).
  7. Lancaster, M. A., Knoblich, J. A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science. 345 (6194), 1247125 (2014).
  8. Sato, T., et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and barrett’s epithelium. Gastroenterology. 141 (5), 1762-1772 (2011).
  9. Sato, T., et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 459 (7244), 262-265 (2009).
  10. Spence, J. R., et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 470 (7332), 105-109 (2011).
  11. Schutgens, F., Clevers, H. Human organoids: Tools for understanding biology and treating diseases. Annual Review of Pathology: Mechanisms of Disease. 15 (1), 211-234 (2020).
  12. Zhao, Z., et al. Organoids. Nature Reviews Methods Primers. 2 (1), 94 (2022).
  13. Barker, N., et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 6 (1), 25-36 (2010).
  14. Corrò, C., Novellasdemunt, L., Li, V. S. W. A brief history of organoids. American Journal of Physiology-Cell Physiology. 319 (1), C151-C165 (2020).
  15. Kim, J., Koo, B. K., Knoblich, J. A. Human organoids: model systems for human biology and medicine. Nature Reviews Molecular Cell Biology. 21 (10), 571-584 (2020).
  16. Simian, M., Bissell, M. J. Organoids: A historical perspective of thinking in three dimensions. Journal of Cell Biology. 216 (1), 31-40 (2017).
  17. Graham, O., et al. Generation and culturing of high-grade serous ovarian cancer patient-derived organoids. Journal of Visualized Experiments. 191, e64878 (2023).
  18. Liu, C., Qin, T., Huang, Y., Li, Y., Chen, G., Sun, C. Drug screening model meets cancer organoid technology. Translational Oncology. 13 (11), 100840 (2020).
  19. Driehuis, E., Kretzschmar, K., Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nature Protocols. 15 (10), 3380-3409 (2020).
  20. Zhou, Z., Cong, L., Cong, X. Patient-derived organoids in precision medicine: drug screening, organoid-on-a-chip and living organoid biobank. Frontiers in Oncology. 11, 762184 (2021).
  21. Costa, E. C., Moreira, A. F., de Melo-Diogo, D., Gaspar, V. M., Carvalho, M. P., Correia, I. J. 3D tumor spheroids: an overview on the tools and techniques used for their analysis. Biotechnology Advances. 34 (8), 1427-1441 (2016).
  22. Foo, M. A., et al. Clinical translation of patient-derived tumour organoids- bottlenecks and strategies. Biomarker Research. 10 (1), 10 (2022).
  23. Kapałczyńska, M., et al. 2D and 3D cell cultures – a comparison of different types of cancer cell cultures. Archives of Medical Science. 14 (4), 910-919 (2018).
  24. Bergdorf, K., et al. High-throughput drug screening of fine-needle aspiration-derived cancer organoids. STAR Protocols. 1 (3), 100212 (2020).
  25. Phan, N., et al. A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Communications Biology. 2 (1), 78 (2019).
  26. Putker, M., et al. Medium-throughput drug- and radiotherapy screening assay using patient-derived organoids. Journal of Visualized Experiments. 170, e62495 (2021).
  27. Dahlin, J. L., Inglese, J., Walters, M. A. Mitigating risk in academic preclinical drug discovery. Nature Reviews Drug Discovery. 14 (4), 279-294 (2015).
  28. Honkala, A., Malhotra, S. V., Kummar, S., Junttila, M. R. Harnessing the predictive power of preclinical models for oncology drug development. Nature Reviews Drug Discovery. 21 (2), 99-114 (2022).
  29. de Witte, C. J., et al. Patient-derived ovarian cancer organoids mimic clinical response and exhibit heterogeneous inter- and intrapatient drug responses. Cell Reports. 31 (11), 107762 (2020).
  30. Hill, S. J., et al. Prediction of DNA repair inhibitor response in short-term patient-derived ovarian cancer organoids. Cancer Discovery. 8 (11), 1404-1421 (2018).
  31. Kopper, O., et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nature Medicine. 25 (5), 838-849 (2019).
  32. Maenhoudt, N., Vankelecom, H. Protocol for establishing organoids from human ovarian cancer biopsies. STAR Protocols. 2 (2), 100429 (2021).
  33. Nanki, Y., et al. Patient-derived ovarian cancer organoids capture the genomic profiles of primary tumours applicable for drug sensitivity and resistance testing. Scientific Reports. 10 (1), 12581 (2020).
  34. Hafner, M., Niepel, M., Chung, M., Sorger, P. K. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat Methods. 13 (6), 521-527 (2016).
  35. Clark, N. A., et al. GRcalculator: an online tool for calculating and mining dose-response data. BMC Cancer. 17 (1), 698 (2017).
  36. Senkowski, W., et al. A platform for efficient establishment and drug-response profiling of high-grade serous ovarian cancer organoids. Developmental Cell. 58 (12), 1106.e7-1121.e7 (2023).
check_url/65068?article_type=t

Play Video

Cite This Article
Fashemi, B. E., van Biljon, L., Rodriguez, J., Graham, O., Mullen, M., Khabele, D. Ovarian Cancer Patient-Derived Organoid Models for Pre-Clinical Drug Testing. J. Vis. Exp. (199), e65068, doi:10.3791/65068 (2023).

View Video