Article Text

Download PDFPDF

511 T cell-instructed inflammation drives immune checkpoint inhibitor therapy resistance
Free
  1. Nam Woo Cho1,
  2. Sophia Guldberg1,
  3. Eun Ji Kim2,
  4. Kamir Hiam-Galvez1,
  5. Katherine Wai1,
  6. Lauren Levine1,
  7. Rachel DeBarge1,
  8. Jacqueline Yee1,
  9. Naa Ashitey1 and
  10. Matthew Spitzer1
  1. 1Helen Diller Family Comprehensive Cancer Center, Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, CA, United States
  2. 2Unaffiliated, San Francisco, CA, United States

Abstract

Background Resistance to immune checkpoint inhibitors (ICIs) is a significant barrier to improving cancer immunotherapy.1 To this end, we interrogated ICI-induced inflammation. One type of inflammation, driven by nuclear factor-kB (NF-kB) activation in the tumor microenvironment (TME), can power tumor-promoting mechanisms including immunosuppression to limit ICI efficacy.2 Conversely, the current paradigm predicts that another type of inflammation, characterized by T cell infiltration, improves ICI responsiveness.3,4 How these two units of inflammation can co-exist in the TME yet direct divergent responses to ICIs requires reconciliation. In particular, it is unknown whether conventional CD4 or CD8 T cells can themselves instruct NF-kB inflammation in the TME, shaping the immunologic setpoint and, unexpectedly, limiting ICI efficacy.

Methods We generated new mouse syngeneic microsatellite instability-high (MSI-H) tumor models to study ICI response and resistance in the context of ample T cell inflammation. High-neoantigen-burden cell lines were implanted into syngeneic mice to assess responses to combination anti-PD-1 and -CTLA4 treatment.

Results Consistent with response in human tumors5, 50% of mouse MSI-H tumors were resistant to combination ICIs. Analysis of the TME in the resistant AT3 breast cancer model and responsive B16F10 melanoma model by mass cytometry revealed that ICIs increased tumor T cell infiltration in each model, but CD8 T cells in AT3 tumors assumed a non-cytotoxic but early-activated phenotype characterized by CD69 expression. T cell expansion in AT3 MSI-H tumors was accompanied by recruitment of T cell-suppressive polymorphonuclear myeloid-derived suppressor cells requiring cytokines G-CSF and CXCL1, absent in B16F10 tumors. Inhibition of the inflammatory NF-kB circuit, which drives expression of these cytokines, via IL-1 receptor neutralization increased ICI efficacy against AT3 MSI-H tumors. Unexpectedly, in vivo depletion of T cells also abolished the NF-kB circuit in AT3 MSI-H tumors, and activated CD8 T cells were sufficient to instruct tumor cells to increase G-CSF and CXCL1 expression. We found that TNFa, increased in T cells and neutrophils upon ICIs, perpetuated the NF-kB circuit and contributed to ICI resistance in AT3 MSI-H tumors. Finally, analysis of single-cell RNA sequencing data from ICI-treated breast cancer patients revealed candidate human T cell correlates.

Conclusions We report a surprising, novel mechanism of ICI resistance whereby treatment-induced T cell infiltration and activation can paradoxically exacerbate a TNFa- and IL-1-dependent resistance circuit in tumors with active NF-kB inflammation. Our findings refine the current models of ICI response and resistance with important therapeutic implications.

Acknowledgements We thank E. Engleman and J. Bluestone for cell lines. We thank A. Marson and T. Roth for CRISPR-Cas9 reagents, protocols and equipment. We thank the UCSF Clinical Cancer Genomics Laboratory, Molecular Oncology Initiative, and D. Raleigh for access to the UCSF cBioPortal dataset.

References

  1. P Sharma, S Hu-Lieskovan, JA Wargo, and A Ribas, Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy, Cell, Feb. 2017;168(4): 707–723, doi: 10.1016/J.CELL.2017.01.017.

  2. AC Betzler, et al. NF-κB and its role in checkpoint control, International Journal of Molecular Sciences May 2020;21(11):3949, doi: 10.3390/IJMS21113949.

  3. J Galon and D Bruni. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies, Nature Reviews Drug Discovery 2018;18(3):197–218, Jan. 2019, doi: 10.1038/s41573-018-0007-y.

  4. DT Le, et al. PD-1 blockade in tumors with mismatch-repair deficiency, New England Journal of Medicine, 2015;372(26):2509–2520, doi: 10.1056/nejmoa1500596.

  5. F Petrelli, M Ghidini, A Ghidini, and G Tomasello. Outcomes following immune checkpoint inhibitor treatment of patients with microsatellite instability-high cancers: a systematic review and meta-analysis, JAMA Oncology, Jul. 2020;6(7):1068–1071, doi: 10.1001/JAMAONCOL.2020.1046.

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.