534
Views
3
CrossRef citations to date
0
Altmetric
Review

Advances in therapeutic targeting of immune checkpoints receptors within the CD96-TIGIT axis: clinical implications and future perspectives

ORCID Icon, , , , &
Pages 1217-1237 | Received 31 Mar 2022, Accepted 20 Sep 2022, Published online: 26 Sep 2022

References

  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries: global cancer statistics 2018. CA Cancer J Clin [Internet]. 2018;68:394–424.
  • Siegel RL, Miller KD, Jemal A. Cancer Stat. 2020;70:7–30.
  • Types of Cancer Treatment. [Internet]. Natl Cancer Inst. 2021. https://www.cancer.gov/about-cancer/treatment/types
  • Khalil DN, Smith EL, Brentjens RJ, et al. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13:273–290.
  • Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069–1086.
  • Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30:507–519.
  • Alteber Z, Kotturi MF, Whelan S, et al. Therapeutic targeting of checkpoint receptors within the DNAM1 axis. Cancer Discov. 2021;11:1040–1051.
  • Quagliariello V, Passariello M, Rea D, et al. Evidences of CTLA-4 and PD-1 blocking agents-induced cardiotoxicity in cellular and preclinical models. J Pers Med [Internet]. 2020;10:179.
  • Quagliariello V, Passariello M, Coppola C, et al. Cardiotoxicity and pro-inflammatory effects of the immune checkpoint inhibitor Pembrolizumab associated to Trastuzumab. Int J Cardiol [Internet]. 2019;292:171–179.
  • Johnson DB, Nebhan CA, Moslehi JJ, et al. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol [Internet]. 2022;19:254–267.
  • Bernhardt G. TACTILE becomes tangible: CD96 discloses its inhibitory peculiarities. Nat Immunol. 2014;15:406–408.
  • Minton K. A TACTILE restraint. Nat Rev Immunol. 2014;14:285.
  • Deuss FA, Watson GM, Fu Z, et al., Structural basis for CD96 immune receptor recognition of nectin-like protein-5, CD155. Structure. 2019;27(219–228.e3):219–228.e3.
  • Dougall WC, Kurtulus S, Smyth MJ, et al. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol Rev. 2017;276:112–120.
  • 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;15:431–438.
  • Wang PL, O’Farrell S, Clayberger C, et al. Identification and molecular cloning of tactile: a novel human T cell activation antigen that is a member of the Ig gene superfamily. J Immunol. 1992;148:2600–2608.
  • Stanko K, Iwert C, Appelt C, et al. CD96 expression determines the inflammatory potential of IL-9-producing Th9 cells. PNAS. 2018;115:2940–2949.
  • Toutirais O, Cabillic F, Le Friec G, et al. DNAX accessory molecule-1 (CD226) promotes human hepatocellular carcinoma cell lysis by Vγ9Vδ2 T cells. Eur J Immunol. 2009;39:1361–1368.
  • Fuchs A, Cella M, Giurisato E, et al. Cutting edge: CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J Immunol. 2004;172:3994–3998.
  • Lepletier A, Lutzky VP, Mittal D, et al. The immune checkpoint CD96 defines a distinct lymphocyte phenotype and is highly expressed on tumor-infiltrating T cells. Immunol Cell Biol. 2019;97:152–164.
  • Hosen N, Park CY, Tatsumi N, et al. CD96 is a leukemic stem cell-specific marker in human acute myeloid leukemia. PNAS. 2007;104:11008–11013.
  • Chávez-González A, Dorantes-Acosta E, Moreno-Lorenzana D, et al. Expression of CD90, CD96, CD117, and CD123 on different hematopoietic cell populations from pediatric patients with acute myeloid leukemia. Arch Med Res. 2014;45:343–350.
  • Gramatzki M, Ludwig WD, Burger R, et al. Antibodies TC-12 (“unique”) and TH-111 (CD96) characterize T-cell acute lymphoblastic leukemia and a subgroup of acute myeloid leukemia. Exp Hematol. 1998;26:1209–1214.
  • Zhang W, Shao Z, Fu R, et al. Expressions of CD96 and CD123 in bone marrow cells of patients with myelodysplastic syndromes. Clin Lab. 2015;61:1429–1434.
  • Meyer D, Seth S, Albrecht J, et al. CD96 interaction with CD155 via its first Ig-like domain is modulated by alternative splicing or mutations in distal Ig-like domains. J Biol Chem. 2009;284:2235–2244.
  • Schneider H, Rudd CE. Diverse mechanisms regulate the surface expression of immunotherapeutic target ctla-4. Front Immunol. 2014;5:619.
  • Yu X, Harden K, C Gonzalez L, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2008;10:48–57.
  • Stanietsky N, Rovis TL, Glasner A, et al. Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR. Eur J Immunol. 2013;43:2138–2150.
  • Meng F, Li L, Lu F, et al. Overexpression of TIGIT in NK and T cells contributes to tumor immune escape in myelodysplastic syndromes. Front Oncol. 2020;10:1595.
  • Yang -Z-Z, Kim HJ, Wu H, et al. TIGIT expression is associated with T-cell suppression and exhaustion and predicts clinical outcome and anti–PD-1 response in follicular lymphoma. Clin Cancer Res. 2020;26:5217–5231.
  • Kurtulus S, Sakuishi K, Ngiow S-F, et al. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest. 2015;125:4053–4062.
  • Jin Z, Lan T, Zhao Y, et al. Higher TIGIT(+)CD226(-) γδ T cells in patients with acute myeloid leukemia. Immunol Invest. 2020;51:1–11.
  • Reches A, Ophir Y, Stein N, et al. Nectin4 is a novel TIGIT ligand which combines checkpoint inhibition and tumor specificity. J Immunother Cancer. 2020;8:e000266.
  • Chauvin J-M, Zarour HM. TIGIT in cancer immunotherapy. J Immunother Cancer. 2020;8:e000957.
  • Liu S, Zhang H, Li M, et al. Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ. 2013;20:456–464.
  • Stanietsky N, Simic H, Arapovic J, et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci. 2009;106:17858L–17863 .
  • Li M, Xia P, Du Y, et al. T-cell immunoglobulin and ITIM domain (TIGIT) receptor/poliovirus receptor (PVR) ligand engagement suppresses interferon-γ production of natural killer cells via β-arrestin 2-mediated negative signaling. J Biol Chem. 2014;289:17647–17657.
  • Shibuya A, Campbell D, Hannum C, et al. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity. 1996;4:573–581.
  • Bottino C, Castriconi R, Pende D, et al. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med. 2003;198:557–567.
  • Pende D, Bottino C, Castriconi R, et al. PVR (CD155) and Nectin-2 (CD112) as ligands of the human DNAM-1 (CD226) activating receptor: involvement in tumor cell lysis. Mol Immunol. 2005;42:463–469.
  • Zhu Y, Paniccia A, Schulick AC, et al. Identification of CD112R as a novel checkpoint for human T cells. J Exp Med. 2016;213:167–176.
  • Zhang Z, Wu N, Lu Y, et al. DNAM-1 controls NK cell activation via an ITT-like motif. J Exp Med. 2015;212:2165–2182.
  • Ralston KJ, Hird SL, Zhang X, et al. The LFA-1-associated molecule PTA-1 (CD226) on T cells forms a dynamic molecular complex with protein 4.1G and human discs large. J Biol Chem. 2004;279:33816–33828.
  • Enqvist M, Ask EH, Forslund E, et al. Coordinated expression of DNAM-1 and LFA-1 in educated NK cells. J Immunol. 2015;194:4518–4527.
  • Shibuya A, Lanier LL, Phillips JH. Protein kinase C is involved in the regulation of both signaling and adhesion mediated by DNAX accessory molecule-1 receptor. J Immunol. 1998;161:1671–1676.
  • Shibuya K, Shirakawa J, Kameyama T, et al. CD226 (DNAM-1) is involved in lymphocyte function-associated antigen 1 costimulatory signal for naive T cell differentiation and proliferation. J Exp Med. 2003;198:1829–1839.
  • Kim HS, Das A, Gross CC, et al. Synergistic signals for natural cytotoxicity are required to overcome inhibition by c-Cbl ubiquitin ligase. Immunity. 2010;32:175–186.
  • Kim HS, Long EO. Complementary phosphorylation sites in the adaptor protein SLP-76 promote synergistic activation of natural killer cells. Sci Signal. 2012;5:ra49.
  • Wu B, Zhong C, Lang Q, et al. Poliovirus receptor (PVR)-like protein cosignaling network: new opportunities for cancer immunotherapy. J Exp Clin Cancer Res. 2021;40:267.
  • Wang N, Yi H, Fang L, et al. CD226 attenuates treg proliferation via Akt and Erk Signaling in an EAE Model. Front Immunol. 2020:11. DOI:10.3389/fimmu.2020.00011.
  • Fourcade J, Sun Z, Chauvin J-M, et al. CD226 opposes TIGIT to disrupt Tregs in melanoma. JCI Insight. 2018;3(14):e121157.
  • Takai Y, Irie K, Shimizu K, et al. Nectins and nectin-like molecules: roles in cell adhesion, migration, and polarization. Cancer Sci. 2003;94:655–667.
  • He Y, Bowman VD, Mueller S, et al. Interaction of the poliovirus receptor with poliovirus. Proc Natl Acad Sci. 2000;97:79L–84 .
  • Mendelsohn CL, Wimmer E, Racaniello VR. Cellular receptor for poliovirus: molecular cloning, nucleotide sequence, and expression of a new member of the immunoglobulin superfamily. Cell. 1989;56:855–865.
  • Sloan KE, Eustace BK, Stewart JK, et al. CD155/PVR plays a key role in cell motility during tumor cell invasion and migration. BMC Cancer. 2004;4:73.
  • Gao J, Zheng Q, Xin N, et al. CD155, an onco-immunologic molecule in human tumors. Cancer Sci. 2017;108:1934–1938.
  • Tahara-Hanaoka S, Shibuya K, Onoda Y, et al. Functional characterization of DNAM-1 (CD226) interaction with its ligands PVR (CD155) and nectin-2 (PRR-2/CD112). Int Immunol. 2004;16:533–538.
  • Holmes VM, Maluquer de Motes C, Richards PT, et al. Interaction between nectin-1 and the human natural killer cell receptor CD96. PLoS One. 2019;14:e0212443.
  • Geraghty RJ, Krummenacher C, Cohen GH, et al. Entry of alphaherpesviruses mediated by poliovirus receptor-related protein 1 and poliovirus receptor. Science. 1998;280:1618–1620.
  • Seth S, Maier MK, Qiu Q, et al. The murine pan T cell marker CD96 is an adhesion receptor for CD155 and nectin-1. Biochem Biophys Res Commun. 2007;364:959–965.
  • Lopez M, Aoubala M, Jordier F, et al. The human poliovirus receptor related 2 protein is a new hematopoietic/endothelial homophilic adhesion molecule. Blood. 1998;92:4602–4611.
  • Sanchez-Correa B, Gayoso I, Bergua JM, et al. Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients. Immunol Cell Biol. 2012;90:109–115.
  • Casado JG, Pawelec G, Morgado S, et al. Expression of adhesion molecules and ligands for activating and costimulatory receptors involved in cell-mediated cytotoxicity in a large panel of human melanoma cell lines. Cancer Immunol Immunother. 2009;58:1517–1526.
  • El-Sherbiny YM, Meade JL, Holmes TD, et al. The requirement for DNAM-1, NKG2D, and NKp46 in the natural killer cell-mediated killing of myeloma cells. Cancer Res. 2007;67:8444L–8449 .
  • Miao X, Yang Z-L, 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;6:179–190.
  • Satoh-Horikawa K, Nakanishi H, Takahashi K, et al. Nectin-3, a new member of immunoglobulin-like cell adhesion molecules that shows homophilic and heterophilic cell-cell adhesion activities. J Biol Chem. 2000;275:10291–10299.
  • Devilard E, Xerri L, Dubreuil P, et al. Nectin-3 (CD113) interacts with Nectin-2 (CD112) to promote lymphocyte transendothelial migration. PLoS One. 2013;8:e77424–e77424.
  • Nishiwada S, Sho M, Yasuda S, et al. Nectin-4 expression contributes to tumor proliferation, angiogenesis and patient prognosis in human pancreatic cancer. J Exp Clin Cancer Res. 2015;34:30.
  • Manes TD, Pober JS. Identification of endothelial cell junctional proteins and lymphocyte receptors involved in transendothelial migration of human effector memory CD4+ T cells. J Immunol. 2011;186:1763–1768.
  • Eriksson EM, Keh CE, Deeks SG, et al. Differential expression of CD96 surface molecule represents CD8+ T cells with dissimilar effector function during HIV-1 infection. PLoS One. 2012;7:e51696–e51696.
  • Sun H, Huang Q, Huang M, et al. Human CD96 Correlates to Natural Killer Cell Exhaustion and Predicts the Prognosis of Human Hepatocellular Carcinoma. Hepatology. 2019;70:168–183.
  • Carlsten M, Björkström NK, Norell H, et al. DNAX accessory molecule-1 mediated recognition of freshly isolated ovarian carcinoma by resting natural killer cells. Cancer Res. 2007;67:1317–1325.
  • 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;7:257–268.
  • Chiang EY, de Almeida PE, de Almeida Nagata DE, et al. CD96 functions as a co-stimulatory receptor to enhance CD8 + T cell activation and effector responses. Eur J Immunol. 2020;50:891–902.
  • Feng M, Wu Z, Zhou Y, et al. BCL9 regulates CD226 and CD96 checkpoints in CD8(+) T cells to improve PD-1 response in cancer. Signal Transduct Target Ther. 2021;6:313.
  • Mittal D, Lepletier A, Madore J, et al. CD96 Is an Immune Checkpoint That Regulates CD8(+) T-cell Antitumor Function. Cancer Immunol Res. 2019;7:559–571.
  • Bi J, Zhang Q, Liang D, et al. T-cell Ig and ITIM domain regulates natural killer cell activation in murine acute viral hepatitis. Hepatology. 2014;59:1715–1725.
  • Joller N, Lozano E, Burkett PR, et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity. 2014;40:569–581.
  • Weulersse M, Asrir A, Pichler AC, et al. Eomes-Dependent Loss of the Co-activating Receptor CD226 Restrains CD8+ T Cell Anti-tumor Functions and Limits the Efficacy of Cancer Immunotherapy. Immunity [Internet]. 2020;53:824–839.e10.
  • Braun M, Aguilera AR, Sundarrajan A, et al. CD155 on Tumor Cells Drives Resistance to Immunotherapy by Inducing the Degradation of the Activating Receptor CD226 in CD8+ T Cells. Immunity. 2020;53:805–823.e15.
  • Paramore A, Frantz S. Bortezomib. Nat Rev Drug Discov. 2003;2:611–612.
  • Yang F, Zhao L, Wei Z, et al. A Cross-Species Reactive TIGIT-Blocking Antibody Fc Dependently Confers Potent Antitumor Effects. J Immunol. 2020;205:ji1901413.
  • Lupo KB, Matosevic S. CD155 immunoregulation as a target for natural killer cell immunotherapy in glioblastoma. J Hematol Oncol. 2020;13:76.
  • Nishiwada S, SHO M, Yasuda S, et al. Clinical Significance of CD155 Expression in Human Pancreatic Cancer. Anticancer Res. 2015;35:2287L–2297 .
  • Gong J, Fang L, Liu R, et al. UPR decreases CD226 ligand CD155 expression and sensitivity to NK cell-mediated cytotoxicity in hepatoma cells. Eur J Immunol. 2014;44:3758–3767.
  • Lepletier A, Madore J, O’Donnell JS, et al. Tumor CD155 Expression Is Associated with Resistance to Anti-PD1 Immunotherapy in Metastatic Melanoma. Clin Cancer Res. 2020;26:3671–3681.
  • Sun Y, Luo J, Chen Y, et al. Combined evaluation of the expression status of CD155 and TIGIT plays an important role in the prognosis of LUAD (lung adenocarcinoma). Int Immunopharmacol. 2020;80:106198.
  • He W, Zhang H, Han F, et al. CD155T/TIGIT Signaling regulates CD8+ T-cell metabolism and promotes tumor progression in human gastric cancer. Cancer Res. 2017;77:6375–6388.
  • Yoshikawa K, Ishida M, Yanai H, et al. Immunohistochemical analysis of CD155 expression in triple-negative breast cancer patients. PLoS One. 2021;16:e0253176.
  • Masson D, Jarry A, Baury B, et al. Overexpression of the CD155 gene in human colorectal carcinoma. Gut. 2001;49:236–240.
  • Papanicolau-Sengos A, Yang Y, Pabla S, et al. Identification of targets for prostate cancer immunotherapy. Prostate. 2019;79:498–505.
  • Smazynski J, Hamilton PT, Thornton S, et al. The immune suppressive factors CD155 and PD-L1 show contrasting expression patterns and immune correlates in ovarian and other cancers. Gynecol Oncol. 2020;158:167–177.
  • Wu L, Mao L, Liu J-F, et al. Blockade of TIGIT/CD155 signaling reverses T-cell exhaustion and enhances antitumor capability in head and neck squamous cell carcinoma. Cancer Immunol Res. 2019;7:1700–1713.
  • Mao L, Xiao Y, Yang Q-C, et al. TIGIT/CD155 blockade enhances anti-PD-L1 therapy in head and neck squamous cell carcinoma by targeting myeloid-derived suppressor cells. Oral Oncol. 2021;121:105472.
  • Liu F, Huang J, He F, et al. CD96, a new immune checkpoint, correlates with immune profile and clinical outcome of glioma. Sci Rep. 2020;10:10768.
  • Zhang Q, Zhong H, Fan Y, et al. Immune and clinical features of CD96 expression in glioma by in silico analysis. Front Bioeng Biotechnol. 2020;8:592.
  • Hung AL, Maxwell R, Theodros D, et al. TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM. Oncoimmunology. 2018;7:e1466769.
  • Iguchi-Manaka A, Okumura G, Kojima H, et al. Increased Soluble CD155 in the Serum of Cancer Patients. PLoS One. 2016;11:e0152982–e0152982.
  • Sloan KE, Stewart JK, Treloar AF, et al. CD155/PVR Enhances Glioma Cell Dispersal by Regulating Adhesion Signaling and Focal Adhesion Dynamics. Cancer Res. 2005;65:10930L–10937 .
  • Sangro B, Sarobe P, Hervás-Stubbs S, et al. Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2021;18:525–543.
  • Chiu DK-C, Yuen VW-H, Cheu JW-S, et al. Hepatocellular Carcinoma Cells Up-regulate PVRL1, Stabilizing PVR and Inhibiting the Cytotoxic T-Cell Response via TIGIT to Mediate Tumor Resistance to PD1 Inhibitors in Mice. Gastroenterology. 2020;159:609–623.
  • Ge Z, Zhou G, Campos Carrascosa L, et al. TIGIT and PD1 Co-blockade Restores ex vivo Functions of Human Tumor-Infiltrating CD8(+) T Cells in Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol. 2021;12:443–464.
  • Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin [Internet]. 2021;71:209–249.
  • Sánchez-Paulete AR, Cueto FJ, Martínez-López M, et al. Cancer Immunotherapy with immunomodulatory anti-CD137 and Anti–PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells. Cancer Discov. 2016;6:71L–79 .
  • Rethacker L, Roelens M, Bejar C, et al. Specific patterns of blood ILCs in metastatic melanoma patients and their modulations in response to immunotherapy. Cancers (Basel). 2021;14:13.
  • Chauvin J-M, Ka M, Pagliano O, et al. IL15 stimulation with TIGIT blockade reverses CD155-mediated NK-cell dysfunction in melanoma. Clin Cancer Res. 2020;26:5520–5533.
  • Ma L, Gai J, Qiao P, et al. A novel bispecific nanobody with PD-L1/TIGIT dual immune checkpoint blockade. Biochem Biophys Res Commun. 2020;531:144–151.
  • Inozume T, Yaguchi T, Furuta J, et al. Melanoma cells control antimelanoma CTL responses via interaction between TIGIT and CD155 in the effector phase. J Invest Dermatol. 2016;136:255–263.
  • Blake SJ, Stannard K, Liu J, et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov. 2016;6:446–459.
  • Zhang H, Liu Q, Lei Y, et al. Direct interaction between CD155 and CD96 promotes immunosuppression in lung adenocarcinoma. Cell Mol Immunol. 2020;18:1575–1577.
  • Rodriguez-Abreu D, Johnson ML, Hussein MA, et al. Primary analysis of a randomized, double-blind, phase II study of the anti-TIGIT antibody tiragolumab (tira) plus atezolizumab (atezo) versus placebo plus atezo as first-line (1L) treatment in patients with PD-L1-selected NSCLC (CITYSCAPE). J Clin Oncol. 2020;38:9503.
  • Peng Y-P, Xi C-H, Zhu Y, et al. Altered expression of CD226 and CD96 on natural killer cells in patients with pancreatic cancer. Oncotarget. 2016;7:66586–66594.
  • Freed-Pastor WA, Lambert LJ, Ely ZA, et al. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer. Cancer Cell. 2021;39(1342–1360.e14). DOI:10.1016/j.ccell.2021.07.007.
  • Siegel RL, Miller KD, Fuchs HE, et al. Cancer Statistics, 2021. CA Cancer J Clin. 2021;71:7–33.
  • Stamm H, Oliveira-Ferrer L, Grossjohann E-M, et al. Targeting the TIGIT-PVR immune checkpoint axis as novel therapeutic option in breast cancer. Oncoimmunology. 2019;8:e1674605.
  • Chen C, Guo Q, Fu H, et al. Asynchronous blockade of PD-L1 and CD155 by polymeric nanoparticles inhibits triple-negative breast cancer progression and metastasis. Biomaterials. 2021;275:120988.
  • Maas RJ, Hoogstad-van Evert JS, Van der Meer JM, et al. TIGIT blockade enhances functionality of peritoneal NK cells with altered expression of DNAM-1/TIGIT/CD96 checkpoint molecules in ovarian cancer. Oncoimmunology. 2020;9:1843247.
  • Johnson DE, Burtness B, Leemans CR, et al. Head and neck squamous cell carcinoma. Nat Rev Dis Prim. 2020;6:92.
  • Nelson MH, Paulos CM. Novel immunotherapies for hematologic malignancies. Immunol Rev. 2015;263:90–105.
  • Kumar SK, Rajkumar V, Kyle RA, et al. Multiple myeloma. Nat Rev Dis Prim [Internet]. 2017;3:17046.
  • Guillerey C, Harjunpää H, Carrié N, et al. TIGIT immune checkpoint blockade restores CD8+ T-cell immunity against multiple myeloma. Blood. 2018;132:1689–1694.
  • Mohammad H, Wahba Y, Gouida M, et al. Cluster of differentiation 96 in children with acute leukemia: a single center cohort study. Indian J Hematol Blood Transfus. 2020;36:178–182.
  • Valhondo I, Hassouneh F, Lopez-Sejas N, et al. Characterization of the DNAM-1, TIGIT and TACTILE axis on circulating NK, NKT-like and T cell subsets in patients with acute myeloid leukemia. Cancers (Basel). 2020;12(8):2171.
  • Xu L, Liu L, Yao D, et al. PD-1 and TIGIT are highly co-expressed on CD8(+) T cells in AML Patient bone marrow. Front Oncol. 2021;11:686156.
  • Soriani A, Zingoni A, Cerboni C, et al. ATM-ATR–dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype. Blood. 2009;113:3503–3511.
  • Ye W, Luo C, Liu F, et al. CD96 correlates with immune infiltration and impacts patient prognosis: a pan-cancer analysis. Front Oncol. 2021;11:634617.
  • Song X, Tang T, Li C, et al. CBX8 and CD96 are important prognostic biomarkers of colorectal cancer. Med Sci Monit. 2018;24:7820–7827.
  • So YK, Byeon S-J, Ku BM, et al. An increase of CD8+ T cell infiltration following recurrence is a good prognosticator in HNSCC. Sci Rep. 2020;10:20059.
  • Zhang J, Fu M, Zhang M, et al. DDX60 is associated with glioma malignancy and serves as a potential immunotherapy biomarker. Front Oncol. 2021;11:665360.
  • Miyashita M, Oshiumi H, Matsumoto M, et al. DDX60, a DEXD/H box helicase, is a novel antiviral factor promoting RIG-I-like receptor-mediated signaling. Mol Cell Biol. 2011;31:3802–3819.
  • Lozano E, Mena M-P, Díaz T, et al. Nectin-2 expression on malignant plasma cells is associated with better response to TIGIT blockade in multiple Myeloma. Clin Cancer Res. 2020;26:4688–4698.
  • Jin H-S, Ko M, Choi D-S, et al. CD226(hi)CD8(+) T cells are a prerequisite for anti-TIGIT immunotherapy. Cancer Immunol Res. 2020;8:912–925.
  • Drake CG. Combination immunotherapy approaches. Ann Oncol. 2012;23:41–46.
  • Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14:561–584.
  • Raphael I, Kumar R, McCarl LH, et al. TIGIT and PD-1 immune checkpoint pathways are associated with patient outcome and anti-tumor immunity in glioblastoma. Front Immunol. 2021;12:637146.
  • Hansen K, Kumar S, Logronio K, et al. COM902, a novel therapeutic antibody targeting TIGIT augments anti-tumor T cell function in combination with PVRIG or PD-1 pathway blockade. Cancer Immunol Immunother. 2021;70:3525–3540.
  • Grapin M, Richard C, Limagne E, et al. Optimized fractionated radiotherapy with anti-PD-L1 and anti-TIGIT: a promising new combination. J Immunother Cancer. 2019;7:160.
  • Li X-Y, Das I, Lepletier A, et al. CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms. J Clin Invest. 2018;128:2613–2625.
  • Chauvin J-M, Pagliano O, Fourcade J, et al. TIGIT and PD-1 impair tumor antigen-specific CD8+ T cells in melanoma patients. J Clin Invest. 2015;125:2046–2058.
  • Han HS, Jeong S, Kim H, et al. TOX-expressing terminally exhausted tumor-infiltrating CD8(+) T cells are reinvigorated by co-blockade of PD-1 and TIGIT in bladder cancer. Cancer Lett. 2021;499:137–147.
  • Xu F, Sunderland A, Zhou Y, et al. Blockade of CD112R and TIGIT signaling sensitizes human natural killer cell functions. Cancer Immunol Immunother. 2017;66:1367–1375.
  • Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell. 2014;26:923–937.
  • Wang B, Zhao Q, Zhang Y, et al. Targeting hypoxia in the tumor microenvironment: a potential strategy to improve cancer immunotherapy. J Exp Clin Cancer Res. 2021;40:24.
  • Fathi M, Bahmanpour S, Barshidi A, et al. Simultaneous blockade of TIGIT and HIF-1α induces synergistic anti-tumor effect and decreases the growth and development of cancer cells. Int Immunopharmacol. 2021;101:108288.
  • Mo Z, Liu D, Rong D, et al. Hypoxic characteristic in the immunosuppressive microenvironment of hepatocellular carcinoma. Front Immunol. 2021;12:611058.
  • Klümper N, Ralser DJ, Bawden EG, et al. LAG3 (LAG-3, CD223) DNA methylation correlates with LAG3 expression by tumor and immune cells, immune cell infiltration, and overall survival in clear cell renal cell carcinoma. J Immunother Cancer. 2020;8:e000552.
  • Yang H, Bueso-Ramos C, DiNardo C, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014;28:1280–1288.
  • Woods DM, Sodré AL, Villagra A, et al. HDAC inhibition upregulates PD-1 ligands in melanoma and augments immunotherapy with PD-1 blockade. Cancer Immunol Res. 2015;3:1375–1385.
  • Li X, Su X, Liu R, et al. HDAC inhibition potentiates anti-tumor activity of macrophages and enhances anti-PD-L1-mediated tumor suppression. Oncogene. 2021;40:1836–1850.
  • Llopiz D, Ruiz M, Villanueva L, et al. Enhanced anti-tumor efficacy of checkpoint inhibitors in combination with the histone deacetylase inhibitor Belinostat in a murine hepatocellular carcinoma model. Cancer Immunol Immunother. 2019;68:379–393.
  • Chen X, Pan X, Zhang W, et al. Epigenetic strategies synergize with PD-L1/PD-1 targeted cancer immunotherapies to enhance antitumor responses. Acta Pharm Sin B. 2020;10:723–733.
  • Lee Y-H, Lee HJ, Kim HC, et al. PD-1 and TIGIT downregulation distinctly affect the effector and early memory phenotypes of CD19-targeting CAR T cells. Mol Ther. 2022;30:579–592.
  • Grosser R, Cherkassky L, Chintala N, et al. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36:471–482.
  • John LB, Devaud C, Duong CPM, et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res. 2013;19:5636–5646.
  • Gargett T, Yu W, Dotti G, et al. GD2-specific CAR T cells undergo potent activation and deletion following antigen encounter but can be protected from activation-induced cell death by PD-1 blockade. Mol Ther. 2016;24:1135–1149.
  • Cherkassky L, Morello A, Villena-Vargas J, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016;126:3130–3144.
  • Jackson Z, Hong C, Schauner R, et al. Sequential single cell transcriptional and protein marker profiling reveals tigit as a marker of CD19 CAR-T cell dysfunction in patients with non-hodgkin’s lymphoma. Blood. 2021;138:164.
  • Ma H, Wang H, Sové RJ, et al. Combination therapy with T cell engager and PD-L1 blockade enhances the antitumor potency of T cells as predicted by a QSP model. J Immunother Cancer. 2020;8:e001141.
  • Zhang Y, Yang Q, Zeng X, et al. MET amplification attenuates lung tumor response to immunotherapy by inhibiting STING. Cancer Discov. 2021;11:2726–2737. candisc.1500.2020.
  • Zhou X, Du J, Wang H, et al. Repositioning liothyronine for cancer immunotherapy by blocking the interaction of immune checkpoint TIGIT/PVR. Cell Commun Signal. 2020;18:142.
  • Zhou X, Jiao L, Qian Y, et al. Repositioning azelnidipine as a dual inhibitor targeting CD47/SIRPα and TIGIT/PVR pathways for cancer immuno-therapy. Biomolecules. 2021;12:11.
  • Harjunpää H, Blake SJ, Ahern E, et al. Deficiency of host CD96 and PD-1 or TIGIT enhances tumor immunity without significantly compromising immune homeostasis. Oncoimmunology [Internet]. 2018;7:e1445949.
  • Waliany S, Lee D, Witteles RM, et al. Immune checkpoint inhibitor cardiotoxicity: understanding basic mechanisms and clinical characteristics and finding a cure. Annu Rev Pharmacol Toxicol [Internet]. 2021;61:113–134.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.