Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

Tumor organoid–T-cell coculture systems

Abstract

T cells are key players in cancer immunotherapy, but strategies to expand tumor-reactive cells and study their interactions with tumor cells at the level of an individual patient are limited. Here we describe the generation and functional assessment of tumor-reactive T cells based on cocultures of tumor organoids and autologous peripheral blood lymphocytes. The procedure consists of an initial coculture of 2 weeks, in which tumor-reactive T cells are first expanded in the presence of (IFNγ-stimulated) autologous tumor cells. Subsequently, T cells are evaluated for their capacity to carry out effector functions (IFNγ secretion and degranulation) after recognition of tumor cells, and their capacity to kill tumor organoids. This strategy is unique in its use of peripheral blood as a source of tumor-reactive T cells in an antigen-agnostic manner. In 2 weeks, tumor-reactive CD8+ T-cell populations can be obtained from ~33–50% of samples from patients with non-small-cell lung cancer (NSCLC) and microsatellite-instable colorectal cancer (CRC). This enables the establishment of ex vivo test systems for T-cell-based immunotherapy at the level of the individual patient.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of the procedure.
Fig. 2: Examples of flow cytometry plots showing tumor reactivity of CD8+ T cells.
Fig. 3: Killing experiment overview.
Fig. 4: Quantification of the tumor organoid killing assay.
Fig. 5: Example of processing of live-cell imaging data.

Similar content being viewed by others

Data availability

The data that provide examples of the results that can be generated with this protocol are available from the corresponding author on reasonable request.

References

  1. Blank, C. U., Haanen, J. B., Ribas, A. & Schumacher, T. N. The “cancer immunogram”. Science 352, 658–660 (2016).

    Article  CAS  Google Scholar 

  2. Dijkstra, K. K., Voabil, P., Schumacher, T. N. & Voest, E. E. Genomics- and transcriptomics-based patient selection for cancer treatment with immune checkpoint inhibitors: a review. JAMA Oncol. 2, 1490–1495 (2016).

    Article  Google Scholar 

  3. Pitt, J. M. et al. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity 44, 1255–1269 (2016).

    Article  CAS  Google Scholar 

  4. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  Google Scholar 

  5. Turcotte, S. et al. Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy. J. Immunol. 191, 2217–2225 (2013).

    Article  CAS  Google Scholar 

  6. Dijkstra, K. K. et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell 174, 1586–1598 (2018).

    Article  CAS  Google Scholar 

  7. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016).

    Article  CAS  Google Scholar 

  8. Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    Article  CAS  Google Scholar 

  9. Geukes Foppen, M. H., Donia, M., Svane, I. M. & Haanen, J. B. Tumor-infiltrating lymphocytes for the treatment of metastatic cancer. Mol. Oncol. 9, 1918–1935 (2015).

    Article  CAS  Google Scholar 

  10. Scheper, W. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat. Med. 25, 89–94 (2018).

    Article  Google Scholar 

  11. Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018).

    Article  CAS  Google Scholar 

  12. Gros, A. et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J. Clin. Invest. 124, 2246–2259 (2014).

    Article  CAS  Google Scholar 

  13. Strønen, E. et al. Targeting of cancer neoantigens with donor-derived T cell receptor repertoires. Science 352, 1337–1341 (2016).

    Article  Google Scholar 

  14. Yossef, R. et al. Enhanced detection of neoantigen-reactive T cells targeting unique and shared oncogenes for personalized cancer immunotherapy. JCI Insight 3, e122467 (2018).

    Article  Google Scholar 

  15. Ye, Q. et al. CD137 accurately identifies and enriches for naturally occurring tumor-reactive T cells in tumor. Clin. Cancer Res. 20, 44–55 (2014).

    Article  CAS  Google Scholar 

  16. Gros, A. et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 22, 433–438 (2016).

    Article  CAS  Google Scholar 

  17. Cohen, C. J. et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J. Clin. Invest. 125, 3981–3991 (2015).

    Article  Google Scholar 

  18. Schumacher, T. N., Scheper, W. & Kvistborg, P. Cancer neoantigens. Annu. Rev. Immunol. 37, 173–200 (2019).

    Article  CAS  Google Scholar 

  19. Verdegaal, E. M. et al. Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha. Cancer Immunol. Immunother. 60, 953–963 (2011).

    Article  CAS  Google Scholar 

  20. Nozaki, K. et al. Co-culture with intestinal epithelial organoids allows efficient expansion and motility analysis of intraepithelial lymphocytes. J. Gastroenterol. 52, 206–213 (2016).

    Article  Google Scholar 

  21. Tsai, S. et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer 18, 335 (2018).

    Article  Google Scholar 

  22. Jenkins, R. W. et al. Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. 8, 196–215 (2018).

    Article  CAS  Google Scholar 

  23. Neal, J. T. et al. Organoid modeling of the tumor immune microenvironment. Cell 175, 1972–1988 (2018).

    Article  CAS  Google Scholar 

  24. Weeber, F. et al. Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases. Proc. Natl Acad. Sci. USA 112, 13308–13311 (2015).

    Article  CAS  Google Scholar 

  25. van de Wetering, M. et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161, 933–945 (2015).

    Article  Google Scholar 

  26. Sachs, N. et al. Long-term expanding human airway organoids for disease modeling. EMBO J. 38, e100300 (2019).

    Article  Google Scholar 

  27. Schütte, M. et al. Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat. Commun. 8, 14262 (2017).

    Article  Google Scholar 

  28. Drost, J. et al. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 11, 347–358 (2016).

    Article  CAS  Google Scholar 

  29. Broutier, L. et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat. Protoc. 11, 1724–1743 (2016).

    Article  CAS  Google Scholar 

  30. Fujii, M., Matano, M., Nanki, K. & Sato, T. Efficient genetic engineering of human intestinal organoids using electroporation. Nat. Protoc. 10, 1474–1485 (2015).

    Article  CAS  Google Scholar 

  31. Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 26, 332–342 (2003).

    Article  Google Scholar 

Download references

Acknowledgements

We thank M. van Baalen, A. Pfauth and F. van Diepen for assistance with flow cytometry experiments; M. Mertz, L. Brocks and B. van den Broek for help with live-cell imaging experiments; M. van de Wetering, N. Sachs, H. Clevers and Hubrecht Organoid Technology (HUB) for advice on organoid cultures; and Merus for the provision of the anti-PD-1 antibody. This work was supported by the NWO gravitation program (NWO 2012-2022) (to E.E.V. on behalf of CancerGenomics.nl), KWF grant HUBR2014-7006 (to E.E.V.), the KWF Queen Wilhelmina Award (NKI 2013-6122, to T.N.S.) and ERC AdG SENSIT (to T.N.S.).

Author information

Authors and Affiliations

Authors

Contributions

C.M.C. and K.K.D. designed the protocol, performed the experiments and wrote the manuscript. L.F.F., S. Kelderman, S. Kaing and N.v.R. provided critical input in the design of the protocol. S.v.d.B. provided the protocol for Noggin-conditioned medium. T.N.S. and E.E.V. supervised the study.

Corresponding author

Correspondence to Emile E. Voest.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Marco Gerlinger, Michael James and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related link

Key reference using this protocol

Dijkstra, K. K. et al. Cell 174, 1586–1598 (2018): https://doi.org/10.1016/j.cell.2018.07.009

Integrated supplementary information

Supplementary Fig. 1 Quantification of the tumor organoid killing assay using fluorescent dyes.

Example of flow cytometry plots showing labeling of tumor cells with CellTrace Yellow and T cells with CellTrace Far Red.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cattaneo, C.M., Dijkstra, K.K., Fanchi, L.F. et al. Tumor organoid–T-cell coculture systems. Nat Protoc 15, 15–39 (2020). https://doi.org/10.1038/s41596-019-0232-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0232-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer