Skip to main content

Induction and Potential Reversal of a T Cell Exhaustion-Like State: In Vitro Potency Assay for Functional Screening of Immune Checkpoint Drug Candidates

  • Protocol
  • First Online:
Immuno-Oncology

Abstract

Tumor infiltrating lymphocytes (TIL) entering the tumor microenvironment (TME) encounter many suppressive factors resulting in a spectrum of possible differentiation paths, many with overlapping characteristics. This differentiation spectrum includes T cell anergy, unresponsiveness, quiescence, tolerance, dysfunction/suppression, and exhaustion, each with many subtypes. Immune checkpoint blockade (ICB) cancer drug therapies have focused on reinvigorating exhausted T cells (TEX) and dysfunctional T cells (herein referred to as TEX). Many factors have been attributed to as causative to this exhausted state including sustained surface expression of multiple co-inhibitory receptors, altered transcription factor expression, epigenetic rewiring, and dysregulated metabolism. Antigen persistence is necessary for driving TEX maintenance in both the chronic viral infection setting and cancer. In addition to persistent antigen exposure, TILs within the TME encounter numerous tumor-mediated immunosuppressive metabolic by-products, suppressive cytokines, hypoxia, and cellular debris which converge to suppress T cell function and uniquely alter its transcription factor profile. These suppressed or dysfunctional T cells are incapable of mounting an optimal anti-tumor response in part due to lack of fitness in competing for glucose and oxygen. This chapter describes two different potency assays: (a) first we describe optimization of a core recall antigen-based in vitro potency assay for screening of immunopotentiating drug candidates; (b) the second assay bundles the core assay with further addition of immunosuppressive agents derived from the TME making a customizable T cell exhaustion-like assay. Our recall antigen assay is being used as a tool for function-based screening of ICB drug candidates. In this assay healthy human Peripheral Blood Mononuclear Cells (PBMCs) are stimulated with peptide(s) and grown in culture for 1 week. Day 4 supernatants are functionally assayed by ELISA for IFN-γ secretion, and cells are assayed on day 7 by flow cytometry for CD8+ or CD4+ T cell expansion by using a single or cocktail of pMHC tetramers. We have shown that in roughly 30% of donor PBMCs, ICB drugs such as pembrolizumab are able to boost both IFN-γ secretion and antigen-specific recall. One potential explanation for this effect is that the observed increase in T cell co-inhibitory receptor expression and presumed co-inhibitory receptor downstream signaling is ameliorated with ICB drugs releasing these T cells from the repressive effects of these co-inhibitory receptors. We have further enhanced our recall assay to more closely resemble the TME by including metabolites and other suppressive agents at concentrations not normally encountered by T cells in a healthy environment. We show one example of this whereby addition of adenosine to the culture results in suppression of antigen-specific T cell expansion without affecting cell viability. Further, we screened several drug candidates on their ability to counteract the negative effects of adenosine and found that one of the drugs was indeed able to restore antigen-specific T cell expansion. Hence, this TME-based recall antigen assay allows for dissection of the effects of TME-based factors on donor-specific T cell response as well as drug candidate screening.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 99.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 129.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Wherry EJ, Kurachi M (2015) Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 15(8):486–499

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. McLane LM, Abdel-Hakeem MS, Wherry EJ (2019) CD8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol 37:457–495

    Article  CAS  PubMed  Google Scholar 

  3. Hashimoto M et al (2018) CD8 T cell exhaustion in chronic infection and cancer: opportunities for interventions. Annu Rev Med 69:301–318

    Article  CAS  PubMed  Google Scholar 

  4. Shin H et al (2007) Viral antigen and extensive division maintain virus-specific CD8 T cells during chronic infection. J Exp Med 204(4):941–949

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schietinger A et al (2016) Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity 45(2):389–401

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yi JS, Cox MA, Zajac AJ (2010) T-cell exhaustion: characteristics, causes and conversion. Immunology 129(4):474–481

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Crawford A et al (2014) Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity 40(2):289–302

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Thommen DS, Schumacher TN (2018) T cell dysfunction in cancer. Cancer Cell 33(4):547–562

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Chang CH et al (2015) Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162(6):1229–1241

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Eil R et al (2016) Ionic immune suppression within the tumour microenvironment limits T cell effector function. Nature 537(7621):539–543

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vodnala SK et al (2019) T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science 363(6434):pii:eaau0135

    Article  Google Scholar 

  12. Ohta A, Metabolic Immune A (2016) Checkpoint: adenosine in tumor microenvironment. Front Immunol 7:109

    Article  PubMed  PubMed Central  Google Scholar 

  13. Anderson KG, Stromnes IM, Greenberg PD (2017) Obstacles posed by the tumor microenvironment to T cell activity: a case for synergistic therapies. Cancer Cell 31(3):311–325

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Revenfeld ALS et al (2017) Induction of a regulatory phenotype in CD3+ CD4+ HLA-DR+ T cells after allogeneic mixed lymphocyte culture; indications of both contact-dependent and -independent activation. Int J Mol Sci 8:7

    Google Scholar 

  15. Ghosh S et al (2019) TSR-033, a novel therapeutic antibody targeting LAG-3, enhances T-cell function and the activity of PD-1 blockade in vitro and in vivo. Mol Cancer Ther 18(3):632–641

    Article  CAS  PubMed  Google Scholar 

  16. Wang C et al (2014) In vitro characterization of the anti-PD-1 antibody nivolumab, BMS-936558, and in vivo toxicology in non-human primates. Cancer Immunol Res 2(9):846–856

    Article  CAS  PubMed  Google Scholar 

  17. Grenga I et al (2016) A fully human IgG1 anti-PD-L1 MAb in an in vitro assay enhances antigen-specific T-cell responses. Clin Transl Immunol 5(5):e83

    Article  Google Scholar 

  18. Li Y et al (2018) Discovery and preclinical characterization of the antagonist anti-PD-L1 monoclonal antibody LY3300054. J Immunother Cancer 6(1):31

    Article  PubMed  PubMed Central  Google Scholar 

  19. European Medicines Agency (2016) Guideline on potency testing of cell based immunotherapy medicinal products for the treatment of cancer. European Medicines Agency, London

    Google Scholar 

  20. Wang Y et al (2017) How an alloreactive T-cell receptor achieves peptide and MHC specificity. Proc Natl Acad Sci U S A 114(24):E4792–E4801

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hundrieser J et al (2019) Role of human and porcine MHC DRB1 alleles in determining the intensity of individual human anti-pig T-cell responses. Xenotransplantation 26:e12523

    Article  PubMed  Google Scholar 

  22. Haley Laken KM, Murtaza A, de Silva Correia J, McNeeley P, Zhang J, Vancutsem P, Wilcoxen K, Jenkins D (2016) Discovery of TSR-022, a novel, potent anti-TIM-3 therapeutic antibody. EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics, Munich

    Google Scholar 

  23. Lamphear JG, Stevens KR, Rich RR (1998) Intercellular adhesion molecule-1 and leukocyte function-associated antigen-3 provide costimulation for superantigen-induced T lymphocyte proliferation in the absence of a specific presenting molecule. J Immunol 160(2):615–623

    CAS  PubMed  Google Scholar 

  24. Gjetting T et al (2019) Sym021, a promising anti-PD1 clinical candidate antibody derived from a new chicken antibody discovery platform. MAbs 11(4):666–680

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Krakauer T (2017) FDA-approved immunosuppressants targeting staphylococcal superantigens: mechanisms and insights. Immunotargets Ther 6:17–29

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jones RB et al (2008) Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J Exp Med 205(12):2763–2779

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang W et al (2009) PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol 21(9):1065–1077

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lichtenegger FS et al (2018) Targeting LAG-3 and PD-1 to enhance T cell activation by antigen-presenting cells. Front Immunol 9:385

    Article  PubMed  PubMed Central  Google Scholar 

  29. Filippis C et al (2017) Nivolumab enhances in vitro effector functions of PD-1(+) T-lymphocytes and leishmania-infected human myeloid cells in a host cell-dependent manner. Front Immunol 8:1880

    Article  PubMed  PubMed Central  Google Scholar 

  30. Li Pira G et al (2008) Evaluation of antigen-specific T-cell responses with a miniaturized and automated method. Clin Vaccine Immunol 15(12):1811–1818

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Duffy D (2018) Standardized Immunomonitoring: separating the signals from the noise. Trends Biotechnol 36(11):1107–1115

    Article  CAS  PubMed  Google Scholar 

  32. Bucks CM et al (2009) Chronic antigen stimulation alone is sufficient to drive CD8+ T cell exhaustion. J Immunol 182(11):6697–6708

    Article  CAS  PubMed  Google Scholar 

  33. Eikawa S, Mizukami S, Udono H (2014) Monitoring multifunctionality of immune-exhausted CD8 T cells in cancer patients. Methods Mol Biol 1142:11–17

    Article  CAS  PubMed  Google Scholar 

  34. Balkhi MY et al (2018) YY1 upregulates checkpoint receptors and downregulates type I cytokines in exhausted, chronically stimulated human T cells. iScience 2:105–122

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hefti FF (2008) Requirements for a lead compound to become a clinical candidate. BMC Neurosci 9(Suppl 3):S7

    Article  PubMed  PubMed Central  Google Scholar 

  36. Smith SG et al (2017) Assay optimisation and technology transfer for multi-site immuno-monitoring in vaccine trials. PLoS One 12(10):e0184391

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

All assays and data were generated at JSR Life Sciences and MBL International. M.C.T. trained as an intern at JSR Life Sciences when she contributed to some studies.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Eden Kleiman or Pirouz M. Daftarian .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2020 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Kleiman, E. et al. (2020). Induction and Potential Reversal of a T Cell Exhaustion-Like State: In Vitro Potency Assay for Functional Screening of Immune Checkpoint Drug Candidates. In: Tan, SL. (eds) Immuno-Oncology. Methods in Pharmacology and Toxicology. Humana, New York, NY. https://doi.org/10.1007/978-1-0716-0171-6_5

Download citation

  • DOI: https://doi.org/10.1007/978-1-0716-0171-6_5

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-0716-0170-9

  • Online ISBN: 978-1-0716-0171-6

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics