Skip to main content

Advertisement

Log in

PD-1 blockade therapy augments the antitumor effects of lymphodepletion and adoptive T cell transfer

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

Abstract

Lymphodepleting cytotoxic regimens enhance the antitumor effects of adoptively transferred effector and naïve T cells. Although the mechanisms of antitumor immunity augmentation by lymphodepletion have been intensively investigated, the effects of lymphodepletion followed by T cell transfer on immune checkpoints in the tumor microenvironment remain unclear. The current study demonstrated that the expression of immune checkpoint molecules on transferred donor CD4+ and CD8+ T cells was significantly decreased in lymphodepleted tumor-bearing mice. In contrast, lymphodepletion did not reduce immune checkpoint molecule levels on recipient CD4+ and CD8+ T cells. Administration of anti-PD-1 antibodies after lymphodepletion and adoptive transfer of T cells significantly inhibited tumor progression. Further analysis revealed that transfer of both donor CD4+ and CD8+ T cells was responsible for the antitumor effects of a combination therapy consisting of lymphodepletion, T cell transfer and anti-PD-1 treatment. Our findings indicate that a possible mechanism underlying the antitumor effects of lymphodepletion followed by T cell transfer is the prevention of donor T cell exhaustion and dysfunction. PD-1 blockade may reinvigorate exhausted recipient T cells and augment the antitumor effects of lymphodepletion and adoptive T cell transfer.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

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

Similar content being viewed by others

Availability of data and material

All the data are available under reasonable request. Material requests should be addressed to satoshi7@med.niigata-u.ac.jp.

Abbreviations

CAR-T:

Chimeric antigen receptor T

DCs:

Dendritic cells

Foxp3:

Forkhead box P3

i.v:

Intravenous

i.p:

Intraperitoneally

ICIs:

Immune checkpoint inhibitors

mAbs:

Monoclonal antibodies

MDSCs:

Myeloid-derived suppressor cells

PD-1:

Programmed-cell death-1

PD-L1:

PD-ligand 1

s.c:

Subcutaneous

TDLNs:

Tumor-draining lymph nodes

TILs:

Tumor-infiltrating lymphocytes

References

  1. Wang LX, Shu S, Plautz GE (2005) Host lymphodepletion augments T cell adoptive immunotherapy through enhanced intratumoral proliferation of effector cells. Cancer Res 65:9547–9554. https://doi.org/10.1158/0008-5472.Can-05-1175

    Article  CAS  PubMed  Google Scholar 

  2. North RJ (1982) Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J Exp Med 155:1063–1074

    Article  CAS  PubMed  Google Scholar 

  3. North RJ (1984) Gamma-irradiation facilitates the expression of adoptive immunity against established tumors by eliminating suppressor T cells. Cancer Immunol Immunother: CII 16:175–181

    Article  CAS  PubMed  Google Scholar 

  4. Restifo NP, Dudley ME, Rosenberg SA (2012) Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol 12:269–281. https://doi.org/10.1038/nri3191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Rosenberg SA, Restifo NP (2015) Adoptive cell transfer as personalized immunotherapy for human cancer. Science (New York, N.Y.) 348:62–8. https://doi.org/10.1126/science.aaa4967

    Article  CAS  Google Scholar 

  6. Gauthier J, Bezerra ED, Hirayama AV et al (2020) Factors associated with outcomes after a second CD19-targeted CAR T-cell infusion for refractory B cell malignancies. Blood. https://doi.org/10.1182/blood.2020006770

    Article  PubMed  PubMed Central  Google Scholar 

  7. Baba J, Watanabe S, Saida Y et al (2012) Depletion of radio-resistant regulatory T cells enhances antitumor immunity during recovery from lymphopenia. Blood 120:2417–2427. https://doi.org/10.1182/blood-2012-02-411124

    Article  CAS  PubMed  Google Scholar 

  8. Dummer W, Niethammer AG, Baccala R, Lawson BR, Wagner N, Reisfeld RA, Theofilopoulos AN (2002) T cell homeostatic proliferation elicits effective antitumor autoimmunity. J Clin Investig 110:185–192. https://doi.org/10.1172/jci15175

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Watanabe S, Arita M, Takahashi M, Saida Y, Koya T, Kikuchi T (2017) Effect of lymphodepletion on donor T Cells and the role of recipient cells persisting after cytotoxic treatments in cancer immunotherapies. Crit Rev Immunol 37:59–73. https://doi.org/10.1615/CritRevImmunol.2018019497

    Article  PubMed  Google Scholar 

  10. Gattinoni L, Finkelstein SE, Klebanoff CA et al (2005) Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+T cells. J Exp Med 202:907–912. https://doi.org/10.1084/jem.20050732

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Klebanoff C, Khong H, Antony P, Palmer D, Restifo N (2005) Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol 26:111–117. https://doi.org/10.1016/j.it.2004.12.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kato K, Cho BC, Takahashi M et al (2019) Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 20:1506–1517. https://doi.org/10.1016/s1470-2045(19)30626-6

    Article  CAS  PubMed  Google Scholar 

  13. Kang YK, Boku N, Satoh T et al (2017) Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet (London, England) 390:2461–2471. https://doi.org/10.1016/s0140-6736(17)31827-5

    Article  CAS  Google Scholar 

  14. Ferris RL, Blumenschein G Jr, Fayette J et al (2016) Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med 375:1856–1867. https://doi.org/10.1056/NEJMoa1602252

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Borghaei H, Paz-Ares L, Horn L et al (2015) Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 373:1627–1639. https://doi.org/10.1056/NEJMoa1507643

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Brahmer J, Reckamp KL, Baas P et al (2015) Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 373:123–135. https://doi.org/10.1056/NEJMoa1504627

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Robert C, Long GV, Brady B et al (2015) Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 372:320–330. https://doi.org/10.1056/NEJMoa1412082

    Article  CAS  PubMed  Google Scholar 

  18. Motzer RJ, Escudier B, McDermott DF et al (2015) Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med 373:1803–1813. https://doi.org/10.1056/NEJMoa1510665

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gandhi L, Rodriguez-Abreu D, Gadgeel S et al (2018) Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 378:2078–2092. https://doi.org/10.1056/NEJMoa1801005

    Article  CAS  PubMed  Google Scholar 

  20. Sato H, Okonogi N, Nakano T (2020) Rationale of combination of anti-PD-1/PD-L1 antibody therapy and radiotherapy for cancer treatment. Int J Clin Oncol 25:801–809. https://doi.org/10.1007/s10147-020-01666-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kawazoe A, Fukuoka S, Nakamura Y et al (2020) Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol 21:1057–1065. https://doi.org/10.1016/s1470-2045(20)30271-0

    Article  CAS  PubMed  Google Scholar 

  22. McLane LM, Abdel-Hakeem MS, Wherry EJ (2019) CD8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol 37:457–495. https://doi.org/10.1146/annurev-immunol-041015-055318

    Article  CAS  PubMed  Google Scholar 

  23. Shu SY, Rosenberg SA (1985) Adoptive immunotherapy of newly induced murine sarcomas. Cancer Res 45:1657–1662

    CAS  PubMed  Google Scholar 

  24. 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 

  25. Jiang Y, Li Y, Zhu B (2015) T-cell exhaustion in the tumor microenvironment. Cell Death Dis 6:e1792. https://doi.org/10.1038/cddis.2015.162

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Crespo J, Sun H, Welling TH, Tian Z, Zou W (2013) T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr Opin Immunol 25:214–221. https://doi.org/10.1016/j.coi.2012.12.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zarour HM (2016) Reversing T-cell dysfunction and exhaustion in cancer. Clin Cancer Res : An Off J Am Assoc Cancer Res 22:1856–1864. https://doi.org/10.1158/1078-0432.Ccr-15-1849

    Article  CAS  Google Scholar 

  28. Saida Y, Watanabe S, Tanaka T et al (2015) Critical roles of chemoresistant effector and regulatory T cells in antitumor immunity after lymphodepleting chemotherapy. J Immunol 195:726–735. https://doi.org/10.4049/jimmunol.1401468

    Article  CAS  PubMed  Google Scholar 

  29. Kansy BA, Concha-Benavente F, Srivastava RM et al (2017) PD-1 status in CD8(+) T cells associates with survival and Anti-PD-1 therapeutic outcomes in head and neck cancer. Cancer Res 77:6353–6364. https://doi.org/10.1158/0008-5472.Can-16-3167

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kurtulus S, Madi A, Escobar G et al (2019) Checkpoint blockade immunotherapy induces dynamic changes in PD-1(-)CD8(+) tumor-infiltrating T cells. Immunity 50:181–94.e6. https://doi.org/10.1016/j.immuni.2018.11.014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ueha S, Yokochi S, Ishiwata Y et al (2015) Robust antitumor effects of combined anti-CD4-depleting antibody and anti-PD-1/PD-L1 immune checkpoint antibody treatment in mice. Cancer Immunol Res 3:631–640. https://doi.org/10.1158/2326-6066.Cir-14-0190

    Article  CAS  PubMed  Google Scholar 

  32. Hsu J, Hodgins JJ, Marathe M et al (2018) Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Investig 128:4654–4668. https://doi.org/10.1172/jci99317

    Article  PubMed  PubMed Central  Google Scholar 

  33. Gabrilovich DI (2017) Myeloid-derived suppressor cells. Cancer Immunol Res 5:3–8. https://doi.org/10.1158/2326-6066.Cir-16-0297

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Watanabe S, Deguchi K, Zheng R, Tamai H, Wang LX, Cohen PA, Shu S (2008) Tumor-induced CD11b+Gr-1+ myeloid cells suppress T cell sensitization in tumor-draining lymph nodes. J Immunol 181:3291–3300. https://doi.org/10.4049/jimmunol.181.5.3291

    Article  CAS  PubMed  Google Scholar 

  35. Tada K, Kitano S, Shoji H et al (2016) Pretreatment immune status correlates with progression-free survival in chemotherapy-treated metastatic colorectal cancer patients. Cancer Immunol Res 4:592–599. https://doi.org/10.1158/2326-6066.CIR-15-0298

    Article  CAS  PubMed  Google Scholar 

  36. Wang LX (2005) Host lymphodepletion augments T cell adoptive immunotherapy through enhanced intratumoral proliferation of effector cells. Cancer Res 65:9547–9554. https://doi.org/10.1158/0008-5472.can-05-1175

    Article  CAS  PubMed  Google Scholar 

  37. Galluzzi L, Senovilla L, Zitvogel L, Kroemer G (2012) The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 11:215–233. https://doi.org/10.1038/nrd3626

    Article  CAS  PubMed  Google Scholar 

  38. Chen G, Emens LA (2013) Chemoimmunotherapy: reengineering tumor immunity. Cancer Immunol Immunother: CII 62:203–216. https://doi.org/10.1007/s00262-012-1388-0

    Article  CAS  PubMed  Google Scholar 

  39. Thommen DS, Schumacher TN (2018) T Cell Dysfunction in cancer. Cancer Cell 33:547–562. https://doi.org/10.1016/j.ccell.2018.03.012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kamada T, Togashi Y, Tay C et al (2019) PD-1(+) regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc Natl Acad Sci U S A 116:9999–10008. https://doi.org/10.1073/pnas.1822001116

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Serpico AF, Visconti R, Grieco D (2020) Exploiting immune-dependent effects of microtubule-targeting agents to improve efficacy and tolerability of cancer treatment. Cell Death Dis 11:361. https://doi.org/10.1038/s41419-020-2567-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G (2013) Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 39:74–88. https://doi.org/10.1016/j.immuni.2013.06.014

    Article  CAS  PubMed  Google Scholar 

  43. Jure-Kunkel M, Masters G, Girit E, Dito G, Lee F, Hunt JT, Humphrey R (2013) Synergy between chemotherapeutic agents and CTLA-4 blockade in preclinical tumor models. Cancer Immunol Immunother : CII 62:1533–1545. https://doi.org/10.1007/s00262-013-1451-5

    Article  CAS  PubMed  Google Scholar 

  44. Zhang R, Lyu C, Lu W, Pu Y, Jiang Y, Deng Q (2020) Synergistic effect of programmed death-1 inhibitor and programmed death-1 ligand-1 inhibitor combined with chemotherapeutic drugs on DLBCL cell lines in vitro and in vivo. Am J Cancer Res 10:2800–2812

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Tanaka H, Matsushima H, Nishibu A, Clausen BE, Takashima A (2009) Dual therapeutic efficacy of vinblastine as a unique chemotherapeutic agent capable of inducing dendritic cell maturation. Cancer Res 69:6987–6994. https://doi.org/10.1158/0008-5472.Can-09-1106

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by the Japan Society for Promotion of Science KAKENHI Grant Number 24591157 (to S. Watanabe).

Author information

Authors and Affiliations

Authors

Contributions

SW, QZ, BH and LD designed experiments. MT, RS, MA, MS, YS, YA, FS, and SH performed experiments. MT, SW, and KS acquired data. SW and YS analyzed data and generated figures. MT and SW wrote the manuscript. AO, SS, and KN supervised the analysis. KI, RK, NA, YO, MH, TK, and TK discussed the results and conclusions and commented on the manuscript at all stages. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Satoshi Watanabe.

Ethics declarations

Conflict of interest

The authors have no financial conflicts of interest.

Ethical approval

All animal studies were approved by the Niigata University Institutional Animal Care and Use Committee.

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_2021_3078_MOESM1_ESM.pdf

Expression of checkpoint receptors after lymphodepletion and T cell transfer.B6 mice were inoculated s.c. with B16F10 tumor cells. These mice were irradiated and reconstituted with spleen cells from Ly5.1 mice 7 days after tumor inoculation or injected i.p. with anti-PD-1 mAb (250 μg per mouse) on days 7 and 14. Tumor tissues were harvested and prepared for FACS analysis on day 21. A-L, The percentage of PD-L1+, PD-1+, TIGIT+, ICOS+, TIM-3+, LAG-3+cells among CD4+T cells are shown. B-L, The percentage of PD-L1+, PD-1+, TIGIT+, ICOS+, TIM-3+, LAG-3+cells among CD8+T cells are presented. Data are shown as mean ± SE (n = 3/group). P values were estimated with the Student’s t test and are shown as *p < 0.05, **p < 0.01 or ***p < 0.001 (PDF 264 KB)

262_2021_3078_MOESM2_ESM.pdf

Expression of checkpoint receptors after lymphodepletion and the transfer of T cells from irradiated hosts.B6 mice were inoculated s.c. with MCA205 tumor cells. These mice were irradiated and reconstituted with spleen cells from Ly5.1 mice 7 days after tumor inoculation or injected i.p. with anti-PD-1 mAb (250 μg per mouse) on days 7 and 14. Donor Ly5.1 mice were irradiated 14 days before the harvest of spleens. Tumor tissues were harvested and prepared for FACS analysis on day 21. A-L, The percentage of PD-L1+, PD-1+, TIGIT+, ICOS+, TIM-3+, LAG-3+cells among CD4+T cells are shown. B-L, The percentage of PD-L1+, PD-1+, TIGIT+, ICOS+, TIM-3+, LAG-3+cells among CD8+T cells are presented. Data are shown as mean ± SE (n = 3/group). P values were estimated with the Student’s t test and are shown as *p < 0.05, **p < 0.01 or ***p < 0.001 (PDF 324 KB)

262_2021_3078_MOESM3_ESM.pdf

Expression of checkpoint receptors in tumor-draining lymph node cells after lymphodepletion and the transfer of T cells.B6 mice were inoculated s.c. with MCA205 tumor cells. These mice were irradiated and reconstituted with spleen cells from Ly5.1 mice 7 days after tumor inoculation or injected i.p. with anti-PD-1 mAb (250 μg per mouse) on days 7 and 14. Tumor-draining lymph nodes were harvested and prepared for FACS analysis on day 21. A-L,The percentage of PD-L1+, PD-1+, TIGIT+, ICOS+, TIM-3+, LAG-3+cells among CD4+T cells are shown. B-L, The percentage of PD-L1+, PD-1+, TIGIT+, ICOS+, TIM-3+, LAG-3+cells among CD8+T cells are presented. Data are shown as mean ± SE (n = 3/group). P values were estimated with the Student’s t test and are shown as *p < 0.05 or **p < 0.01 (PDF 302 KB)

262_2021_3078_MOESM4_ESM.pdf

The combination of lymphodepletion, T cell transfer, anti-PD-1 Abs, and anti-CD25 Abs for advanced skin tumor model. MCA205 tumor-bearing mice were irradiated and adoptively transferred naïve T cells 20 days after tumor inoculation. The mice were treated with anti-CD25 mAbs to deplete Tregs on day 20 and received anti-PD-1 treatment on days 20, 26, and 32. P value was estimated with the Student’s t test (PDF 113 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Takahashi, M., Watanabe, S., Suzuki, R. et al. PD-1 blockade therapy augments the antitumor effects of lymphodepletion and adoptive T cell transfer. Cancer Immunol Immunother 71, 1357–1369 (2022). https://doi.org/10.1007/s00262-021-03078-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-021-03078-0

Keywords

Navigation