Log in

Establishment of a mechanism-based in vitro coculture assay for evaluating the efficacy of immune checkpoint inhibitors

  • Original Article
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

Cancer immunotherapy, which blocks immune checkpoint molecules, is an effective therapeutic strategy for human cancer patients through restoration of tumor-infiltrating (TI) cell function. However, evaluating the efficacy of immune checkpoint inhibitors (ICIs) is difficult because no standard in vitro assay for ICI efficacy evaluation exists. Additionally, blocking a particular immune checkpoint receptor (ICR) is insufficient to restore T cell functionality, because other ICRs still transduce inhibitory signals. Therefore, limiting inhibitory signals transduced via other ICRs is needed to more accurately assess the efficacy of ICIs targeting a particular immune checkpoint. Here, we introduce a newly developed in vitro coculture assay using human peripheral blood mononuclear cells (hPBMCs) and engineered human cancer cell lines. We enriched CD8+ T cells from hPBMCs of healthy donors through low-dose T cell receptor stimulation and cytokine (human IL-2 and IL-7) addition. These enriched CD8+ T cells were functional and expressed multiple ICRs, especially TIM-3 and TIGIT. We also established immune checkpoint ligand (ICL) knockout (KO) cancer cell lines with the CRISPR-Cas9 system. Then, we optimized the in vitro coculture assay conditions to evaluate ICI efficacy. For example, we selected the most effective anti-TIM-3 antibody through coculture of TIM-3+CD8+ T cells with PD-L1-/-PVR-/- cancer cells. In summary, we developed a mechanism-based in vitro coculture assay with hPBMCs and ICL KO cancer cell lines, which could be a useful tool to identify promising ICIs by providing reliable ICI efficacy information.

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

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price includes VAT (Canada)

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data availability

All data generated and analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

CRISPR:

Clustered regularly interspaced short palindromic repeats

Cas9:

CRISPR-associated protein 9

hPBMC:

Human peripheral blood mononuclear cell

ICI:

Immune checkpoint inhibitor

ICL:

Immune checkpoint ligand

ICR:

Immune checkpoint receptor

TI:

Tumor-infiltrating

References

  1. Pauken KE, Wherry EJ (2015) Overcoming T cell exhaustion in infection and cancer. Trends Immunol 36:265–276. https://doi.org/10.1016/j.it.2015.02.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Anderson AC, Joller N, Kuchroo VK (2016) Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 44:989–1004. https://doi.org/10.1016/j.immuni.2016.05.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Fourcade J, Sun Z, Benallaoua M, Guillaume P, Luescher IF, Sander C, Kirkwood JM, Kuchroo V, Zarour HM (2010) Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med 207:2175–2186. https://doi.org/10.1084/jem.20100637

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P et al (2010) Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci 107:7875–7880. https://doi.org/10.1073/pnas.1003345107

    Article  PubMed  PubMed Central  Google Scholar 

  5. Chauvin JM, Pagliano O, Fourcade J et al (2015) TIGIT and PD-1 impair tumor antigen-specific CD8(+) T cells in melanoma patients. J Clin Invest 125:2046–2058. https://doi.org/10.1172/JCI80445

    Article  PubMed  PubMed Central  Google Scholar 

  6. Kamphorst AO, Pillai RN, Yang S et al (2017) Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci USA 114:4993–4998. https://doi.org/10.1073/pnas.1705327114

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Thommen DS, Koelzer VH, Herzig P et al (2018) A transcriptionally and functionally distinct PD-1(+) CD8(+) T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 24:994–1004. https://doi.org/10.1038/s41591-018-0057-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, Redmond WL, Seliger B, Marincola FM (2017) Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer 81:116–129. https://doi.org/10.1016/j.ejca.2017.01.035

    Article  CAS  PubMed  Google Scholar 

  9. Chen L, Han X (2015) Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest 125:3384–3391. https://doi.org/10.1172/JCI80011

    Article  PubMed  PubMed Central  Google Scholar 

  10. Gong J, Chehrazi-Raffle A, Reddi S, Salgia R (2018) Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer 6:8. https://doi.org/10.1186/s40425-018-0316-z

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kim JE, Patel MA, Mangraviti A et al (2017) Combination therapy with anti-PD-1, Anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin Cancer Res 23:124–136. https://doi.org/10.1158/1078-0432.CCR-15-1535

    Article  CAS  PubMed  Google Scholar 

  12. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC (2010) Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 207:2187–2194. https://doi.org/10.1084/jem.20100643

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hung AL, Maxwell R, Theodros D et al (2018) TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM. Oncoimmunology 7:e1466769. https://doi.org/10.1080/2162402X.2018.1466769

    Article  PubMed  PubMed Central  Google Scholar 

  14. Harris-Bookman S, Mathios D, Martin AM et al (2018) Expression of LAG-3 and efficacy of combination treatment with anti-LAG-3 and anti-PD-1 monoclonal antibodies in glioblastoma. Int J Cancer 143:3201–3208. https://doi.org/10.1002/ijc.31661

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yonesaka K, Haratani K, Takamura S et al (2018) B7–H3 negatively modulates CTL-mediated cancer immunity. Clin Cancer Res 24:2653–2664. https://doi.org/10.1158/1078-0432.CCR-17-2852

    Article  CAS  PubMed  Google Scholar 

  16. Lee YH, Martin-Orozco N, Zheng P et al (2017) Inhibition of the B7–H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function. Cell Res 27:1034–1045. https://doi.org/10.1038/cr.2017.90

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rotte A (2019) Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J Exp Clin Cancer Res 38:255. https://doi.org/10.1186/s13046-019-1259-z

    Article  PubMed  PubMed Central  Google Scholar 

  18. Kamphorst AO, Wieland A, Nasti T et al (2017) Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355:1423–1427. https://doi.org/10.1126/science.aaf0683

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hui E, Cheung J, Zhu J et al (2017) T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355:1428–1433. https://doi.org/10.1126/science.aaf1292

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Khan O, Giles JR, McDonald S et al (2019) TOX transcriptionally and epigenetically programs CD8(+) T cell exhaustion. Nature 571:211–218. https://doi.org/10.1038/s41586-019-1325-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Han HS, Jeong S, Kim H et al (2021) TOX-expressing terminally exhausted tumor-infiltrating CD8(+) T cells are reinvigorated by co-blockade of PD-1 and TIGIT in bladder cancer. Cancer Lett 499:137–147. https://doi.org/10.1016/j.canlet.2020.11.035

    Article  CAS  PubMed  Google Scholar 

  22. Kim K, Park S, Park SY et al (2020) Single-cell transcriptome analysis reveals TOX as a promoting factor for T cell exhaustion and a predictor for anti-PD-1 responses in human cancer. Genome Med 12:22. https://doi.org/10.1186/s13073-020-00722-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sharpe AH, Pauken KE (2018) The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18:153–167. https://doi.org/10.1038/nri.2017.108

    Article  CAS  PubMed  Google Scholar 

  24. Kim KH, Cho J, Ku BM et al (2019) The first-week proliferative response of peripheral blood PD-1(+)CD8(+) T cells predicts the response to anti-PD-1 therapy in solid tumors. Clin Cancer Res 25:2144–2154. https://doi.org/10.1158/1078-0432.CCR-18-1449

    Article  CAS  PubMed  Google Scholar 

  25. Kim KH, Hur JY, Koh J et al (2020) Immunological characteristics of hyperprogressive disease in patients with non-small cell lung cancer treated with anti-PD-1/PD-L1 abs. Immune Netw 20:e48. https://doi.org/10.4110/in.2020.20.e48

    Article  PubMed  PubMed Central  Google Scholar 

  26. Abraham RT, Weiss A (2004) Jurkat T cells and development of the T-cell receptor signalling paradigm. Nat Rev Immunol 4:301–308. https://doi.org/10.1038/nri1330

    Article  CAS  PubMed  Google Scholar 

  27. Wang C, Thudium KB, Han M 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 2:846–856. https://doi.org/10.1158/2326-6066.CIR-14-0040

    Article  CAS  PubMed  Google Scholar 

  28. Kwon M, Choi YJ, Sa M, Park SH, Shin EC (2018) Two-round mixed lymphocyte reaction for evaluation of the functional activities of Anti-PD-1 and Immunomodulators. Immune Netw 18:e45. https://doi.org/10.4110/in.2018.18.e45

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mujib S, Jones RB, Lo C, Aidarus N, Clayton K, Sakhdari A, Benko E, Kovacs C, Ostrowski MA (2012) Antigen-independent induction of Tim-3 expression on human T cells by the common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 is associated with proliferation and is dependent on the phosphoinositide 3-kinase pathway. J Immunol 188:3745–3756. https://doi.org/10.4049/jimmunol.1102609

    Article  CAS  PubMed  Google Scholar 

  30. Zhang ZN, Zhu ML, Chen YH, Fu YJ, Zhang TW, Jiang YJ, Chu ZX, Shang H (2015) Elevation of Tim-3 and PD-1 expression on T cells appears early in HIV infection, and differential Tim-3 and PD-1 expression patterns can be induced by common gamma -chain cytokines. Biomed Res Int 2015:916936. https://doi.org/10.1155/2015/916936

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Martin MD, Badovinac VP (2018) Defining memory CD8 T cell. Front Immunol 9:2692. https://doi.org/10.3389/fimmu.2018.02692

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Picarda E, Ohaegbulam KC, Zang X (2016) Molecular pathways: targeting B7–H3 (CD276) for human cancer immunotherapy. Clin Cancer Res 22:3425–3431. https://doi.org/10.1158/1078-0432.CCR-15-2428

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

The authors declare no competing financial interests. This study was supported by grants funded by the Ministry of Food and Drug Safety (18182MFDS408) and the Ministry of Science and ICT (MSIT) (2017R1A5A1014560, 2019M3A9B6065221). This study was also supported by Korean Health Technology R&D Project through the Korean Health Industry Development Institute (KHIDI) funded by the Ministry of Health and Welfare (HV20C0144) and Korea Drug Development Fund funded by Ministry of Science and ICT, Ministry of Trade, Industry, and Energy, and Ministry of Health and Welfare (HN21C1410). The funder had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

MJK and S-JH designed and interpreted the study, wrote the manuscript and edited the manuscript. MJK performed the experiments and analyzed the data. KHH and BRL assisted with experiments. S-JH supervised the study. All authors approved the final version of the article, including the authorship list.

Corresponding author

Correspondence to Sang-Jun Ha.

Ethics declarations

Conflicts of interest

The authors have no conflicts of interest to declare that are relevant to the content of this article.

Consent to participate

Informed consent was obtained from all individual participants included in the study.

Ethical approval

The studies were approved by the Institutional Review Board of Yonsei University Severance Hospital with IRB no. 4-2016-0788 for a single patient with NSCLC and a single patient with HNSCC. All patients who participated in these studies provided written informed consent prior to enrollment and sample collection at Yonsei University Severance Hospital. The research conformed to the principles of the Helsinki Declaration.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

262_2022_3201_MOESM1_ESM.pdf

Supplementary Figure 1 Comparison between the CD8+ T cell enrichments by the existing protocol and the newly developed protocol. hPBMCs were isolated from the peripheral blood of healthy donors. For the existing protocol using αCD3/CD28 dynabeads, isolated hPBMCs were cultured with αCD3/CD28 dynabeads (the ratio of 1:1) and hIL-2 (10 ng/ml). For the newly developed protocol using soluble anti-CD3 antibodies, isolated hPBMCs were cultured with anti-CD3 antibodies (1 μg/ml, soluble), hIL-2 (10 ng/ml), and hIL-7 (10 ng/ml) in non-tissue 24-well culture plates. (a) Representative FACS plot of singlets, lymphocytes, live cells and T cells of the cultured hPBMCs for 15 days according to the indicated protocol. The experiment was performed with hPBMCs from three donors. The data are representative of a single donor. (PDF 436 KB)

262_2022_3201_MOESM2_ESM.pdf

Supplementary Figure 2 Comparison between the CD8+ T cell enrichments by using hPBMCs of healthy donors and cancer patients. hPBMCs were isolated from the peripheral blood of healthy donors, a single patient with NSCLC, and a single patient with HNSCC. Isolated hPBMCs were cultured with anti-CD3 antibodies (1 μg/ml, soluble), hIL-2 (10 ng/ml), and hIL-7 (10 ng/ml) in non-tissue 24-well culture plates. (a) Representative FACS plot of the T cell frequency at the indicated time point after culture initiation. (b) Kinetics of the number of total cells among the cultured hPBMCs. (c) Kinetics of the frequency of T cells, and the number of T cells among the cultured hPBMCs. The experiment was performed with hPBMCs from healthy donors, a single patient with NSCLC, and a single patient with HNSCC. (PDF 332 KB)

262_2022_3201_MOESM3_ESM.pdf

Supplementary Figure 3 Naïve/memory phenotype of enriched CD8+ T cells. hPBMCs were isolated from the peripheral blood of healthy donors. Isolated hPBMCs were cultured with anti-CD3 antibodies (1 μg/ml, soluble), hIL-2 (10 ng/ml), and hIL-7 (10 ng/ml) in non-tissue 24-well culture plates. (a) Representative FACS plot of naïve/memory phenotype of cultured CD8+ T cells at the indicated time point after culture initiation. (b) Quantification of the percentage of effector memory (EM), effector memory expressing CD45RA (EMRA), central memory (CM), and naïve CD8+ T cells at the indicated time point after culture initiation. (c) Representative FACS plot of the ICR expression patterns of CD8+ T cells enriched 15 days according to naïve/memory phenotype. (d) Representative FACS plot of naïve/memory CD8+ T cell frequency in PD-1+ and PD-1-CD8+ T cells at 15 days after culture initiation (left). Quantification of the percentage of naïve/memory CD8+ T cell frequency in PD-1+ and PD-1-CD8+ T cells at 15 days after culture initiation (right). The experiment was performed with hPBMCs from two donors. The data are representative of triplicate samples from a single donor. The data were analyzed by two-tailed unpaired Student’s t-test (b, d). The error bars indicate the means ± SEMs. **p < 0.01; ****p < 0.0001. (PDF 938 KB)

262_2022_3201_MOESM4_ESM.pdf

Supplementary Figure 4 Degranulation in ICR-positive CD8+ T cells was augmented by ICIs. Before coculture, CD8+ T cells enriched for 21 days were preincubated with isotype control or ICR-blocking antibodies for 20 min in a 37°C incubator. Preincubated CD8+ T cells were cocultured with HCC4006 cell lines for 6 hr at a ratio of 1:1 under restimulation with the anti-CD3 antibody (20 μg/ml, plate-coated), the anti-CD28 antibody (5 μg/ml, plate-coated), hIL-2 (10 ng/ml), and hIL-7 (10 ng/ml). (a) Representative FACS plots of cytokine secretion and degranulation in PD-1+CD8+ T cells cultured with and without anti-PD-1 antibodies. (b) Rate of increase in PD-1+CD8+ T cell degranulation induced by anti-PD-1 antibody treatment. (c) Representative FACS plots of cytokine secretion and degranulation in TIGIT+CD8+ T cells cultured with and without anti-TIGIT antibodies. (d) Rate of increase in TIGIT+CD8+ T cell degranulation induced by anti-TIGIT antibody treatment. The data were concatenated in each group (a, c). The data were analyzed by two-tailed unpaired Student’s t-test (b,d). The error bars indicate the means ± SEMs. **p < 0.01; ****p < 0.0001. (PDF 496 KB)

262_2022_3201_MOESM5_ESM.pdf

Supplementary Figure 5 Comparison between the increased cytokine secretion in TIGIT+CD8+ T cells in the ratio of 1:1 and 0.1:1. Before coculture, CD8+ T cells enriched for 21 days were preincubated with isotype control or TIGIT-blocking antibodies for 20 min in a 37°C incubator. Preincubated CD8+ T cells were cocultured with HCC4006 cell lines for 6 hr at a ratio of 1:1 and 0.1:1 under restimulation with the anti-CD3 antibody (20 μg/ml, plate-coated), the anti-CD28 antibody (5 μg/ml, plate-coated), hIL-2 (10 ng/ml), and hIL-7 (10 ng/ml). (a) Representative FACS plot of cytokine secreting-TIGIT+CD8+ T cells cultured with and without anti-TIGIT antibodies in the ratio of 1:1 and 0.1:1. (b) Rate of increase in TIGIT+CD8+ T cell function induced by anti-TIGIT antibodies in the ratio of 1:1 and 0.1:1. (c) Representative FACS plot of degranulation in TIGIT+CD8+ T cells cultured with and without anti-TIGIT antibodies in the ratio of 1:1 and 0.1:1. (d) Rate of increase in TIGIT+CD8+ T cell degranulation induced by anti-TIGIT antibodies in the ratio of 1:1 and 0.1:1. The data were concatenated in each group (a, c). The data were analyzed by two-tailed unpaired Student’s t-test (b,d). The error bars indicate the means ± SEMs. ***p < 0.001; ****p < 0.0001. (PDF 490 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, M.J., Hong, K.H., Lee, B.R. et al. Establishment of a mechanism-based in vitro coculture assay for evaluating the efficacy of immune checkpoint inhibitors. Cancer Immunol Immunother 71, 2777–2789 (2022). https://doi.org/10.1007/s00262-022-03201-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-022-03201-9

Keywords

Navigation