Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2021

Open Access 01-12-2021 | Colon Cancer | Research

Blockade of checkpoint receptor PVRIG unleashes anti-tumor immunity of NK cells in murine and human solid tumors

Authors: Yangyang Li, Yu Zhang, Guoshuai Cao, Xiaodong Zheng, Cheng Sun, Haiming Wei, Zhigang Tian, Weihua Xiao, Rui Sun, Haoyu Sun

Published in: Journal of Hematology & Oncology | Issue 1/2021

Login to get access

Abstract

Background

Although checkpoint-based immunotherapy has shown exciting results in the treatment of tumors, around 70% of patients have experienced unresponsiveness. PVRIG is a recently identified immune checkpoint receptor and blockade of which could reverse T cell exhaustion to treat murine tumor; however, its therapeutic potential via NK cells in mice and human remains seldom reported.

Methods

In this study, we used patient paraffin-embedded colon adenocarcinoma sections, various murine tumor models (MC38 colon cancer, MCA205 fibrosarcoma and LLC lung cancer), and human NK cell- or PBMC-reconstituted xenograft models (SW620 colon cancer) to investigate the effect of PVRIG on tumor progression.

Results

We found that PVRIG was highly expressed on tumor-infiltrating NK cells with exhausted phenotype. Furthermore, either PVRIG deficiency, early blockade or late blockade of PVRIG slowed tumor growth and prolonged survival of tumor-bearing mice by inhibiting exhaustion of NK cells as well as CD8+ T cells. Combined blockade of PVRIG and PD-L1 showed better effect in controlling tumor growth than using either one alone. Depletion of NK or/and CD8+ T cells in vivo showed that both cell types contributed to the anti-tumor efficacy of PVRIG blockade. By using Rag1−/− mice, we demonstrated that PVRIG blockade could provide therapeutic effect in the absence of adaptive immunity. Further, blockade of human PVRIG with monoclonal antibody enhanced human NK cell function and inhibited human tumor growth in NK cell- or PBMC-reconstituted xenograft mice.

Conclusions

Our results reveal the importance of NK cells and provide novel knowledge for clinical application of PVRIG-targeted drugs in future.
Appendix
Available only for authorised users
Literature
2.
go back to reference Ledford H, Else H, Warren M. Cancer immunologists scoop medicine Nobel prize. Nature. 2018;562(7725):20–1.PubMedCrossRef Ledford H, Else H, Warren M. Cancer immunologists scoop medicine Nobel prize. Nature. 2018;562(7725):20–1.PubMedCrossRef
3.
go back to reference Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311–9.PubMedCrossRef Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311–9.PubMedCrossRef
4.
go back to reference Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–56.PubMedPubMedCentralCrossRef Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–56.PubMedPubMedCentralCrossRef
5.
6.
go back to reference Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856–67.PubMedPubMedCentralCrossRef Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856–67.PubMedPubMedCentralCrossRef
7.
8.
go back to reference Eggermont AMM, Chiarion-Sileni V, Grob J-J, et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N Engl J Med. 2016;375(19):1845–55.PubMedPubMedCentralCrossRef Eggermont AMM, Chiarion-Sileni V, Grob J-J, et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N Engl J Med. 2016;375(19):1845–55.PubMedPubMedCentralCrossRef
9.
go back to reference Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.PubMedPubMedCentralCrossRef Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.PubMedPubMedCentralCrossRef
11.
go back to reference Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nat Rev Immunol. 2015;15(4):243–54.PubMedCrossRef Martinet L, Smyth MJ. Balancing natural killer cell activation through paired receptors. Nat Rev Immunol. 2015;15(4):243–54.PubMedCrossRef
12.
go back to reference Alteber Z, Kotturi MF, Whelan S, et al. Therapeutic targeting of checkpoint receptors within the DNAM1 axis. Cancer Discov. 2021. Alteber Z, Kotturi MF, Whelan S, et al. Therapeutic targeting of checkpoint receptors within the DNAM1 axis. Cancer Discov. 2021.
13.
go back to reference Kucan Brlic P, Lenac Rovis T, Cinamon G, et al. Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell Mol Immunol. 2019;16(1):40–52.PubMedCrossRef Kucan Brlic P, Lenac Rovis T, Cinamon G, et al. Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell Mol Immunol. 2019;16(1):40–52.PubMedCrossRef
14.
go back to reference Sanchez-Correa B, Valhondo I, Hassouneh F, et al. DNAM-1 and the TIGIT/PVRIG/TACTILE axis: novel immune checkpoints for natural killer cell-based cancer immunotherapy. Cancers (Basel). 2019;11(6):877.CrossRef Sanchez-Correa B, Valhondo I, Hassouneh F, et al. DNAM-1 and the TIGIT/PVRIG/TACTILE axis: novel immune checkpoints for natural killer cell-based cancer immunotherapy. Cancers (Basel). 2019;11(6):877.CrossRef
15.
go back to reference Manieri NA, Chiang EY, Grogan JL. TIGIT: a key inhibitor of the cancer immunity cycle. Trends Immunol. 2017;38(1):20–8.PubMedCrossRef Manieri NA, Chiang EY, Grogan JL. TIGIT: a key inhibitor of the cancer immunity cycle. Trends Immunol. 2017;38(1):20–8.PubMedCrossRef
16.
go back to reference Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. 2014;26(6):923–37.PubMedCrossRef Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. 2014;26(6):923–37.PubMedCrossRef
17.
go back to reference Zhang Q, Bi J, Zheng X, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018;19(7):723–32.PubMedCrossRef Zhang Q, Bi J, Zheng X, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018;19(7):723–32.PubMedCrossRef
18.
go back to reference Kong Y, Zhu L, Schell TD, et al. T-cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T-cell exhaustion and poor clinical outcome in AML patients. Clin Cancer Res. 2016;22(12):3057–66.PubMedCrossRef Kong Y, Zhu L, Schell TD, et al. T-cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T-cell exhaustion and poor clinical outcome in AML patients. Clin Cancer Res. 2016;22(12):3057–66.PubMedCrossRef
19.
20.
go back to reference Minnie SA, Kuns RD, Gartlan KH, et al. Myeloma escape after stem cell transplantation is a consequence of T-cell exhaustion and is prevented by TIGIT blockade. Blood. 2018;132(16):1675–88.PubMedCrossRef Minnie SA, Kuns RD, Gartlan KH, et al. Myeloma escape after stem cell transplantation is a consequence of T-cell exhaustion and is prevented by TIGIT blockade. Blood. 2018;132(16):1675–88.PubMedCrossRef
21.
go back to reference Blake SJ, Stannard K, Liu J, et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov. 2016;6(4):446–59.PubMedCrossRef Blake SJ, Stannard K, Liu J, et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov. 2016;6(4):446–59.PubMedCrossRef
22.
go back to reference Mittal D, Lepletier A, Madore J, et al. CD96 is an immune checkpoint that regulates CD8(+) T-cell antitumor function. Cancer Immunol Res. 2019;7(4):559–71.PubMedPubMedCentralCrossRef Mittal D, Lepletier A, Madore J, et al. CD96 is an immune checkpoint that regulates CD8(+) T-cell antitumor function. Cancer Immunol Res. 2019;7(4):559–71.PubMedPubMedCentralCrossRef
23.
go back to reference Sun HY, Huang Q, Huang M, et al. Human CD96 correlates to natural killer cell exhaustion and predicts the prognosis of human hepatocellular carcinoma. Hepatology. 2019;70(1):168–83.PubMedCrossRef Sun HY, Huang Q, Huang M, et al. Human CD96 correlates to natural killer cell exhaustion and predicts the prognosis of human hepatocellular carcinoma. Hepatology. 2019;70(1):168–83.PubMedCrossRef
25.
go back to reference Hudson WH, Gensheimer J, Hashimoto M, et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1(+) stem-like CD8(+) T cells during chronic infection. Immunity. 2019;51(6):1043–58.PubMedPubMedCentralCrossRef Hudson WH, Gensheimer J, Hashimoto M, et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1(+) stem-like CD8(+) T cells during chronic infection. Immunity. 2019;51(6):1043–58.PubMedPubMedCentralCrossRef
26.
go back to reference Murter B, Pan X, Ophir E, et al. Mouse PVRIG has CD8(+) T cell-specific coinhibitory functions and dampens antitumor immunity. Cancer Immunol Res. 2019;7(2):244–56.PubMedPubMedCentralCrossRef Murter B, Pan X, Ophir E, et al. Mouse PVRIG has CD8(+) T cell-specific coinhibitory functions and dampens antitumor immunity. Cancer Immunol Res. 2019;7(2):244–56.PubMedPubMedCentralCrossRef
27.
go back to reference Sivori S, Vacca P, Del Zotto G, et al. Human NK cells: surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol. 2019;16(5):430–41.PubMedPubMedCentralCrossRef Sivori S, Vacca P, Del Zotto G, et al. Human NK cells: surface receptors, inhibitory checkpoints, and translational applications. Cell Mol Immunol. 2019;16(5):430–41.PubMedPubMedCentralCrossRef
29.
go back to reference Liu Z, Han C, Fu YX. Targeting innate sensing in the tumor microenvironment to improve immunotherapy. Cell Mol Immunol. 2020;17(1):13–26.PubMedCrossRef Liu Z, Han C, Fu YX. Targeting innate sensing in the tumor microenvironment to improve immunotherapy. Cell Mol Immunol. 2020;17(1):13–26.PubMedCrossRef
30.
go back to reference Xu F, Sunderland A, Zhou Y, et al. Blockade of CD112R and TIGIT signaling sensitizes human natural killer cell functions. Cancer Immunol Immunother. 2017;66(10):1367–75.PubMedPubMedCentralCrossRef Xu F, Sunderland A, Zhou Y, et al. Blockade of CD112R and TIGIT signaling sensitizes human natural killer cell functions. Cancer Immunol Immunother. 2017;66(10):1367–75.PubMedPubMedCentralCrossRef
31.
go back to reference Li J, Whelan S, Kotturi MF, et al. PVRIG is a novel NK cell immune checkpoint receptor in acute myeloid leukemia. Haematologica. 2020. (Online ahead of print). Li J, Whelan S, Kotturi MF, et al. PVRIG is a novel NK cell immune checkpoint receptor in acute myeloid leukemia. Haematologica. 2020. (Online ahead of print).
32.
go back to reference Cheng M, Ma J, Chen Y, et al. Establishment, characterization, and successful adaptive therapy against human tumors of NKG cell, a new human NK cell line. Cell Transpl. 2011;20(11–12):1731–46.CrossRef Cheng M, Ma J, Chen Y, et al. Establishment, characterization, and successful adaptive therapy against human tumors of NKG cell, a new human NK cell line. Cell Transpl. 2011;20(11–12):1731–46.CrossRef
33.
34.
go back to reference Miao X, Yang ZL, Xiong L, et al. Nectin-2 and DDX3 are biomarkers for metastasis and poor prognosis of squamous cell/adenosquamous carcinomas and adenocarcinoma of gallbladder. Int J Clin Exp Pathol. 2013;6(2):179–90.PubMedPubMedCentral Miao X, Yang ZL, Xiong L, et al. Nectin-2 and DDX3 are biomarkers for metastasis and poor prognosis of squamous cell/adenosquamous carcinomas and adenocarcinoma of gallbladder. Int J Clin Exp Pathol. 2013;6(2):179–90.PubMedPubMedCentral
35.
go back to reference O’Donnell JS, Madore J, Li XY, et al. Tumor intrinsic and extrinsic immune functions of CD155. Semin Cancer Biol. 2019;65:189–96.PubMedCrossRef O’Donnell JS, Madore J, Li XY, et al. Tumor intrinsic and extrinsic immune functions of CD155. Semin Cancer Biol. 2019;65:189–96.PubMedCrossRef
36.
go back to reference Whelan S, Ophir E, Kotturi MF, et al. PVRIG and PVRL2 are induced in cancer and inhibit CD8(+) T-cell function. Cancer Immunol Res. 2019;7(2):257–68.PubMedPubMedCentralCrossRef Whelan S, Ophir E, Kotturi MF, et al. PVRIG and PVRL2 are induced in cancer and inhibit CD8(+) T-cell function. Cancer Immunol Res. 2019;7(2):257–68.PubMedPubMedCentralCrossRef
37.
go back to reference Pitt MJ, Vétizou M, Daillère R, et al. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity. 2016;44(6):1255–69.CrossRefPubMed Pitt MJ, Vétizou M, Daillère R, et al. Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity. 2016;44(6):1255–69.CrossRefPubMed
38.
go back to reference Takai Y, Irie K, Shimizu K, et al. Nectins and nectin-like molecules: roles in cell adhesion, migration, and polarization. Cancer Sci. 2003;94(8):655–67.PubMedCrossRef Takai Y, Irie K, Shimizu K, et al. Nectins and nectin-like molecules: roles in cell adhesion, migration, and polarization. Cancer Sci. 2003;94(8):655–67.PubMedCrossRef
39.
go back to reference Tahara-Hanaoka S, Shibuya K, Kai H, et al. Tumor rejection by the poliovirus receptor family ligands of the DNAM-1 (CD226) receptor. Blood. 2006;107(4):1491–6.PubMedCrossRef Tahara-Hanaoka S, Shibuya K, Kai H, et al. Tumor rejection by the poliovirus receptor family ligands of the DNAM-1 (CD226) receptor. Blood. 2006;107(4):1491–6.PubMedCrossRef
40.
41.
go back to reference Chan CJ, Martinet L, Gilfillan S, et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol. 2014;15(5):431–8.PubMedCrossRef Chan CJ, Martinet L, Gilfillan S, et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol. 2014;15(5):431–8.PubMedCrossRef
42.
go back to reference Pauken KE, Wherry EJ. TIGIT and CD226: tipping the balance between costimulatory and coinhibitory molecules to augment the cancer immunotherapy toolkit. Cancer Cell. 2014;26(6):785–7.PubMedCrossRef Pauken KE, Wherry EJ. TIGIT and CD226: tipping the balance between costimulatory and coinhibitory molecules to augment the cancer immunotherapy toolkit. Cancer Cell. 2014;26(6):785–7.PubMedCrossRef
43.
go back to reference Yu X, Harden K, Gonzalez LC, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009;10(1):48–57.PubMedCrossRef Yu X, Harden K, Gonzalez LC, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009;10(1):48–57.PubMedCrossRef
45.
go back to reference Huang X, Zhang X, Bai X, et al. Blocking PD-L1 for anti-liver cancer immunity: USP22 represents a critical cotarget. Cell Mol Immunol. 2020;17(7):677–9.PubMedCrossRef Huang X, Zhang X, Bai X, et al. Blocking PD-L1 for anti-liver cancer immunity: USP22 represents a critical cotarget. Cell Mol Immunol. 2020;17(7):677–9.PubMedCrossRef
46.
go back to reference COM701 shows antitumor activity, +/- nivolumab. Cancer Discov. 2020; 10(6):752. COM701 shows antitumor activity, +/- nivolumab. Cancer Discov. 2020; 10(6):752.
Metadata
Title
Blockade of checkpoint receptor PVRIG unleashes anti-tumor immunity of NK cells in murine and human solid tumors
Authors
Yangyang Li
Yu Zhang
Guoshuai Cao
Xiaodong Zheng
Cheng Sun
Haiming Wei
Zhigang Tian
Weihua Xiao
Rui Sun
Haoyu Sun
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2021
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-021-01112-3

Other articles of this Issue 1/2021

Journal of Hematology & Oncology 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine