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:

Bilateral murine tumor models for characterizing the response to immune checkpoint blockade

Abstract

The therapeutic response to immune checkpoint blockade (ICB) is highly variable, not only between different cancers but also between patients with the same cancer type. The biological mechanisms underlying these differences in response are incompletely understood. Identifying correlates in patient tumor samples is challenging because of genetic and environmental variability. Murine studies usually compare different tumor models or treatments, introducing potential confounding variables. This protocol describes bilateral murine tumor models, derived from syngeneic cancer cell lines, that display a symmetrical yet dichotomous response to ICB. These models enable detailed analysis of whole tumors in a highly homogeneous background, combined with knowledge of the therapeutic outcome within a few weeks, and could potentially be used for mechanistic studies using other (immuno-)therapies. We discuss key considerations and describe how to use two cell lines as fully optimized models. We discuss experimental details, including proper inoculation technique to achieve symmetry and one-sided surgical tumor removal, which takes only 5 min per mouse. Furthermore, we outline the preparation of bulk tissue or single-cell suspensions for downstream analyses such as bulk RNA-seq, immunohistochemistry, single-cell RNA-seq and flow cytometry.

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

Access options

Buy this article

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

Fig. 1: Schematic of experimental design.
Fig. 2: Tumor models with symmetrical, dichotomous responses to ICB.
Fig. 3: Intermediate responses are not consistently symmetrical.
Fig. 4: Response rates are influenced by day of dosing.
Fig. 5: Analysis of surgically removed tumor.
Fig. 6: Analysis of bulk RNA-seq of responders versus non-responders.
Fig. 7: Flow cytometric analysis of responders versus non-responders.
Fig. 8: Mouse handling for symmetric tumor inoculation.
Fig. 9: Removal of the right-side tumor.

Similar content being viewed by others

Data availability

The raw RNA-seq data used to generate Fig. 6 are available from the Gene Expression Omnibus data repository (accession no. GSE117358).

References

  1. Galluzzi, L., Chan, T. A., Kroemer, G., Wolchok, J. D. & López-Soto, A. The hallmarks of successful anticancer immunotherapy. Sci. Transl. Med. 10, eaat7807 (2018).

    Article  PubMed  CAS  Google Scholar 

  2. Lesterhuis, W. J. et al. Dynamic versus static biomarkers in cancer immune checkpoint blockade: unravelling complexity. Nat. Rev. Drug Discov. 16, 264 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Topalian, S. L. et al. Safety, activity, and immune correlates of anti–PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. 107, 4275 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wolchok, J. D. et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369, 122–133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Das, R. et al. Combination therapy with anti–CTLA-4 and anti–PD-1 leads to distinct immunologic changes in vivo. J. Immunol. 194, 950–959 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ayers, M. et al. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest 127, 2930–2940 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Prat, A. et al. Immune-related gene expression profiling after PD-1 blockade in non–small cell lung carcinoma, head and neck squamous cell carcinoma, and melanoma. Cancer Res. 77, 3540–3550 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Sanmamed, M., Chester, C., Melero, I. & Kohrt, H. Defining the optimal murine models to investigate immune checkpoint blockers and their combination with other immunotherapies. Ann. Oncol. 27, 1190–1198 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Gustafsson, C. & Vallverdú, J. The best model of a cat is several cats. Trends Biotechnol. 34, 207–213 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Gubin, M. M. et al. High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell 175, 1014–1030.e1019 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Moynihan, K. D. et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat. Med. 22, 1402 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133.e1117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mosely, S. I. S. et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol. Res. 5, 29–41 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Lesterhuis, W. J. et al. Network analysis of immunotherapy-induced regressing tumours identifies novel synergistic drug combinations. Sci. Rep. 5, 12298 (2015).

  20. Chen, S. et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell 160, 1246–1260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sutmuller, R. P. et al. Synergism of cytotoxic T lymphocyte–associated antigen 4 blockade and depletion of CD25+ regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J. Exp. Med. 194, 823–832 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zemek, R. M. et al. Sensitization to immune checkpoint blockade through activation of a STAT1/NK axis in the tumor microenvironment. Sci. Transl. Med. 11, eaav7816 (2019).

    Article  PubMed  CAS  Google Scholar 

  23. Ock, C.-Y. et al. Genomic landscape associated with potential response to anti-CTLA-4 treatment in cancers. Nat. Commun. 8, 1050 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Lee, H.-S. et al. Comprehensive immunoproteogenomic analyses of malignant pleural mesothelioma. JCI Insight 3, e98575 (2018).

    Article  PubMed Central  Google Scholar 

  25. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997 (2018). e924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. van Elsas, A., Hurwitz, A. A. & Allison, J. P. Combination immunotherapy of B16 melanoma using anti–cytotoxic T lymphocyte–associated antigen 4 (Ctla-4) and granulocyte/macrophage colony-stimulating factor (Gm-Csf)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 190, 355–366 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ribas, A. et al. Association of response to programmed death receptor 1 (PD-1) blockade with pembrolizumab (MK-3475) with an interferon-inflammatory immune gene signature. J. Clin. Oncol. 33 (Suppl.), 3001 (2015).

    Article  Google Scholar 

  29. Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377, 2500–2501 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sneddon, S. et al. Whole exome sequencing of an asbestos-induced wild-type murine model of malignant mesothelioma. BMC Cancer 17, 396–396 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Ben-David, U. et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature 560, 325–330 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gibney, G. T., Weiner, L. M. & Atkins, M. B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 17, e542–e551 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hegde, P. S., Karanikas, V. & Evers, S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin. Cancer Res. 22, 1865–1874 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Devaud, C., John, L. B., Westwood, J. A., Darcy, P. K. & Kershaw, M. H. Immune modulation of the tumor microenvironment for enhancing cancer immunotherapy. OncoImmunology 2, e25961 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Santegoets, S. J. et al. The anatomical location shapes the immune infiltrate in tumors of same etiology and affects survival. Clin. Cancer Res. 25, 240–252 (2019).

    Article  PubMed  CAS  Google Scholar 

  38. Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Kovacs, E. J. et al. Aging and innate immunity in the mouse: impact of intrinsic and extrinsic factors. Trends Immunol. 30, 319–324 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Macpherson, A. J. & Harris, N. L. Interactions between commensal intestinal bacteria and the immune system. Nat. Rev. Immunol. 4, 478–485 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Tsukamoto, A., Serizawa, K., Sato, R., Yamazaki, J. & Inomata, T. Vital signs monitoring during injectable and inhalant anesthesia in mice. Exp. Anim. 64, 57–64 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Workman, P. et al. Guidelines for the welfare and use of animals in cancer research. Br. J. Cancer 102, 1555–1577 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge the facilities, as well as the scientific and technical assistance of the Australian Microscopy & Microanalysis Research Facility at the Centre for Microscopy, Characterisation & Analysis, The University of Western Australia, a facility funded by the University and State and Commonwealth governments. We thank P. Deng and C. Rinaldi for technical assistance. This work was funded by grant 1103980 from the National Health and Medical Research Council (NHMRC). W.J.L. was funded by an NHMRC RD Wright Fellowship and a Fellowship from the Cancer Council of Western Australia.

Author information

Authors and Affiliations

Authors

Contributions

R.M.Z. optimized the mouse models, performed and interpreted all mouse experiments and flow cytometry experiments, prepared samples for RNA-seq analysis, and wrote the paper. V.S.F., C.F., and T.H.C. provided technical assistance with surgery and in vivo treatment experiments. E.d.J., T.L. and A.B. interpreted the bulk RNA-seq analyses. L.B. provided critical reagents. R.A.L., M.J.M., and A.K.N. were involved in experimental design and interpretation of all in vivo studies. R.A.L. and V.S.F provided critical revision of the manuscript. W.J.L. designed and developed the models, interpreted the data and supervised the project. R.M.Z and W.J.L. wrote the manuscript with input from all authors. All authors gave final approval of the version to be published.

Corresponding authors

Correspondence to Rachael M. Zemek or W. Joost Lesterhuis.

Ethics declarations

Competing interests

A patent application pertaining to aspects of this work has been filed by R.M.Z., W.J.L., R.A.L., and A.B. (PCT/AU2019/050259, “Method for immunotherapy drug treatment”). A.K.N., M.J.M., A.B., R.A.L., and W.J.L. have received research funding and consultancy fees from Douglas Pharmaceuticals. W.J.L. has received research funding from AstraZeneca and consultancy fees from Merck Sharp & Dohme. A.K.N. declares consultancy or advisory board membership for Boehringer Ingelheim, Bayer Pharmaceuticals, Roche Pharmaceutics, Merck Sharp Dohme, Pharmabcine, Atara Biotherapeutics and Trizell, and research funding from AstraZeneca. M.J.M. has served on advisory boards for Merck Sharp & Dohme, Bristol-Myers Squibb, Roche, and AstraZeneca.

Additional information

Peer review information Nature Protocols thanks Gottfried Baier 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 links

Key references using this protocol

Zemek, R. M. et al. Sci. Transl. Med. 11, eaav7816 (2019): https://doi.org/10.1126/scitranslmed.aav7816

Lesterhuis, W. J. et al. Sci. Rep. 5, 12298 (2015): https://doi.org/10.1038/srep12298

Integrated supplementary information

Supplementary Figure 1 Response to ICB in the B16-F10 tumor model.

a) Growth curves of C57BL/6 mice bearing subcutaneous B16-F10 tumours, treated with i.p. PBS or (b) with ICB (anti-CTLA4 day 5 and anti-PD-L1 on days 5,7 and 9 post-inoculation) when tumours were at least 9mm2 (n=15 per arm, combined data from 3 experiments).

Supplementary Figure 2 Flow cytometry gating strategy.

Gating strategy used to delineate immune cell subsets in mouse tumour and lymph nodes.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2 and Supplementary Table 1.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zemek, R.M., Fear, V.S., Forbes, C. et al. Bilateral murine tumor models for characterizing the response to immune checkpoint blockade. Nat Protoc 15, 1628–1648 (2020). https://doi.org/10.1038/s41596-020-0299-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-0299-3

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