Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors
Abstract
:Simple Summary
Abstract
1. Introduction
2. Current Role of Immunotherapy in Lung Cancer
PD-L1 Expression Levels and Outcome Related
3. Newly Immune Checkpoints
3.1. Lymphocyte-Activation Gene 3
3.2. T Cell Immunoreceptor with Ig and ITIM Domains
3.3. T Cell Immunoglobulin and Mucin-Domain Containing-3 (TIM-3)
3.4. V-Domain Ig Suppressor of T Cell Activation
3.5. Inducible T-Cell COStimulator (ICOS)
3.6. B7 Homolog 3 Protein, (B7-H3)
3.7. B- and T-Lymphocyte Attenuator (BTLA)
3.8. Other Immuno-Target Molecules
4. Future Perspective
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Rabinovich, G.A.; Gabrilovich, D.; Sotomayor, E.M. Immunosuppressive strategies that are mediated by tumor cells. Annu. Rev. Immunol. 2007, 25, 267–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wellenstein, M.D.; de Visser, K.E. Cancer-Cell-Intrinsic Mechanisms Shaping the Tumor Immune Landscape. Immunity 2018, 48, 399–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, A.A.; Drake, V.; Huang, H.S.; Chiu, S.C.; Zheng, L. Reprogramming the tumor microenvironment: Tumor-induced immunosuppressive factors paralyze T cells. Oncoimmunology 2015, 4, e1016700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duan, Q.; Zhang, H.; Zheng, J.; Zhang, L. Turning Cold into Hot: Firing up the Tumor Microenvironment. Trends Cancer 2020, 6, 605–618. [Google Scholar] [CrossRef] [PubMed]
- Croci, D.O.; Zacarías Fluck, M.F.; Rico, M.J.; Matar, P.; Rabinovich, G.A.; Scharovsky, O.G. Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment. Cancer Immunol. Immunother. 2007, 56, 1687–1700. [Google Scholar] [CrossRef]
- Parayath, N.; Padmakumar, S.; Nair, S.V.; Menon, D.; Amiji, M.M. Strategies for Targeting Cancer Immunotherapy Through Modulation of the Tumor Microenvironment. Regen. Eng. Transl. Med. 2020, 6, 29–49. [Google Scholar] [CrossRef]
- Seidel, J.A.; Otsuka, A.; Kabashima, K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: Mechanisms of action, efficacy, and limitations. Front. Oncol. 2018, 8, 86. [Google Scholar] [CrossRef]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Marin-Acevedo, J.A.; Dholaria, B.; Soyano, A.E.; Knutson, K.L.; Chumsri, S.; Lou, Y. Next generation of immune checkpoint therapy in cancer: New developments and challenges. J. Hematol. Oncol. 2018, 11, 8. [Google Scholar] [CrossRef]
- Kruger, S.; Ilmer, M.; Kobold, S.; Cadilha, B.L.; Endres, S.; Ormanns, S.; Schuebbe, G.; Renz, B.W.; D’Haese, J.G.; Schloesser, H.; et al. Advances in cancer immunotherapy 2019-Latest trends. J. Exp. Clin. Cancer Res. 2019, 38, 268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smyth, M.J.; Ngiow, S.F.; Ribas, A.; Teng, M.W.L. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 2016, 13, 143–158. [Google Scholar] [CrossRef]
- Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 2019, 16, 563–580. [Google Scholar] [CrossRef]
- Chu, X.; Niu, L.; Xiao, G.; Peng, H.; Deng, F.; Liu, Z.; Wu, H.; Yang, L.; Tan, Z.; Li, Z.; et al. The Long-Term and Short-Term Efficacy of Immunotherapy in Non-Small Cell Lung Cancer Patients With Brain Metastases: A Systematic Review and Meta-Analysis. Front. Immunol. 2022, 13, 2297. [Google Scholar] [CrossRef] [PubMed]
- Singh, N.; Temin, S.; Baker, S.; Blanchard, E.; Brahmer, J.R.; Celano, P.; Duma, N.; Ellis, P.M.; Elkins, I.B.; Haddad, R.Y.; et al. Therapy for Stage IV Non-Small-Cell Lung Cancer Without Driver Alterations: ASCO Living Guideline. J. Clin. Oncol. 2022, 40, 3323–3343. [Google Scholar] [CrossRef] [PubMed]
- Zugazagoitia, J.; Paz-Ares, L. Extensive-Stage Small-Cell Lung Cancer: First-Line and Second-Line Treatment Options. J. Clin. Oncol. 2022, 40, 671–680. [Google Scholar] [CrossRef]
- Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Trebeschi, S.; Drago, S.G.; Birkbak, N.J.; Kurilova, I.; Cǎlin, A.M.; Delli Pizzi, A.; Lalezari, F.; Lambregts, D.M.J.; Rohaan, M.W.; Parmar, C.; et al. Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann. Oncol. 2019, 30, 998–1004. [Google Scholar] [CrossRef] [Green Version]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [Green Version]
- Boutros, C.; Tarhini, A.; Routier, E.; Lambotte, O.; Ladurie, F.L.; Carbonnel, F.; Izzeddine, H.; Marabelle, A.; Champiat, S.; Berdelou, A.; et al. Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat. Rev. Clin. Oncol. 2016, 13, 473–486. [Google Scholar] [CrossRef]
- Altmann, D.M. A Nobel Prize-worthy pursuit: Cancer immunology and harnessing immunity to tumour neoantigens. Immunology 2018, 155, 283–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Zhou, S.; Yang, F.; Qi, X.; Wang, X.; Guan, X.; Shen, C.; Duma, N.; Vera Aguilera, J.; Chintakuntlawar, A.; et al. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol. 2019, 5, 1008–1019. [Google Scholar] [CrossRef] [PubMed]
- Yi, M.; Yu, S.; Qin, S.; Liu, Q.; Xu, H.; Zhao, W.; Chu, Q.; Wu, K. Gut microbiome modulates efficacy of immune checkpoint inhibitors. J. Hematol. Oncol. 2018, 11, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Donnell, J.S.; Long, G.V.; Scolyer, R.A.; Teng, M.W.L.; Smyth, M.J. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat. Rev. 2017, 52, 71–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
- Andrews, L.P.; Marciscano, A.E.; Drake, C.G.; Vignali, D.A.A. LAG3 (CD223) as a cancer immunotherapy target. Immunol. Rev. 2017, 276, 80–96. [Google Scholar] [CrossRef] [Green Version]
- Monney, L.; Sabatos, C.A.; Gaglia, J.L.; Ryu, A.; Waldner, H.; Chernova, T.; Manning, S.; Greenfield, E.A.; Coyle, A.J.; Sobel, R.A.; et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 2002, 415, 536–541. [Google Scholar] [CrossRef]
- Yu, X.; Harden, K.; Gonzalez, L.C.; Francesco, M.; Chiang, E.; Irving, B.; Tom, I.; Ivelja, S.; Refino, C.J.; Clark, H.; et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat. Immunol. 2009, 10, 48–57. [Google Scholar] [CrossRef]
- Wang, L.; Rubinstein, R.; Lines, J.L.; Wasiuk, A.; Ahonen, C.; Guo, Y.; Lu, L.F.; Gondek, D.; Wang, Y.; Fava, R.A.; et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 2011, 208, 577–592. [Google Scholar] [CrossRef]
- Yang, S.; Wei, W.; Zhao, Q. B7-H3, a checkpoint molecule, as a target for cancer immunotherapy. Int. J. Biol. Sci. 2020, 16, 1767–1773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marinelli, O.; Nabissi, M.; Morelli, M.B.; Torquati, L.; Amantini, C.; Santoni, G. ICOS-L as a Potential Therapeutic Target for Cancer Immunotherapy. Curr. Protein Pept. Sci. 2018, 19, 1107–1113. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Zheng, Y.; Mao, R.; Su, Z.; Zhang, J. BTLA/HVEM Signaling: Milestones in Research and Role in Chronic Hepatitis B Virus Infection. Front. Immunol. 2019, 10, 617. [Google Scholar] [CrossRef] [PubMed]
- Forde, P.M.; Spicer, J.; Lu, S.; Provencio, M.; Mitsudomi, T.; Awad, M.M.; Felip, E.; Broderick, S.R.; Brahmer, J.R.; Swanson, S.J.; et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N. Engl. J. Med. 2022, 386, 1973–1985. [Google Scholar] [CrossRef]
- Felip, E.; Altorki, N.; Zhou, C.; Csőszi, T.; Vynnychenko, I.; Goloborodko, O.; Luft, A.; Akopov, A.; Martinez-Marti, A.; Kenmotsu, H.; et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB–IIIA non-small-cell lung cancer (IMpower010): A randomised, multicentre, open-label, phase 3 trial. Lancet 2021, 398, 1344–1357. [Google Scholar] [CrossRef]
- FDA Approves Atezolizumab as Adjuvant Treatment for Non-Small Cell Lung Cancer|FDA. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-atezolizumab-adjuvant-treatment-non-small-cell-lung-cancer (accessed on 16 May 2022).
- Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef] [Green Version]
- Spigel, D.R.; Faivre-Finn, C.; Gray, J.E.; Vicente, D.; Planchard, D.; Paz-Ares, L.; Vansteenkiste, J.F.; Garassino, M.C.; Hui, R.; Quantin, X.; et al. Five-Year Survival Outcomes From the PACIFIC Trial: Durvalumab After Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2022, 40, 1301–1311. [Google Scholar] [CrossRef]
- Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef]
- Borghaei, H.; Gettinger, S.; Vokes, E.E.; Chow, L.Q.M.; Burgio, M.A.; de Castro Carpeno, J.; Pluzanski, A.; Arrietac, O.; Frontera, O.A.; Chiari, R.; et al. Five-year outcomes from the randomized, phase iii trials checkmate 017 and 057: Nivolumab versus docetaxel in previously treated non-small-cell lung cancer. J. Clin. Oncol. 2021, 39, 723–733. [Google Scholar] [CrossRef]
- Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicentre randomised controlled trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
- Herbst, R.S.; Baas, P.; Kim, D.W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.Y.; Molina, J.; Kim, J.H.; Arvis, C.D.; Ahn, M.J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
- Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herbst, R.S.; Giaccone, G.; de Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for First-Line Treatment of PD-L1–Selected Patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339. [Google Scholar] [CrossRef] [PubMed]
- Sezer, A.; Kilickap, S.; Gümüş, M.; Bondarenko, I.; Özgüroğlu, M.; Gogishvili, M.; Turk, H.M.; Cicin, I.; Bentsion, D.; Gladkov, O.; et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: A multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet 2021, 397, 592–604. [Google Scholar] [CrossRef] [PubMed]
- Awad, M.M.; Gadgeel, S.M.; Borghaei, H.; Patnaik, A.; Yang, J.C.H.; Powell, S.F.; Gentzler, R.D.; Martins, R.G.; Stevenson, J.P.; Altan, M.; et al. Long-Term Overall Survival From KEYNOTE-021 Cohort G: Pemetrexed and Carboplatin With or Without Pembrolizumab as First-Line Therapy for Advanced Nonsquamous NSCLC. J. Thorac. Oncol. 2021, 16, 162–168. [Google Scholar] [CrossRef]
- Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef]
- Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef]
- West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef]
- Gogishvili, M.; Melkadze, T.; Makharadze, T.; Giorgadze, D.; Dvorkin, M.; Penkov, K.; Laktionov, K.; Nemsadze, G.; Nechaeva, M.; Rozhkova, I.; et al. Cemiplimab plus chemotherapy versus chemotherapy alone in non-small cell lung cancer: A randomized, controlled, double-blind phase 3 trial. Nat. Med. 2022, 28, 2374–2380. [Google Scholar] [CrossRef]
- Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef]
- FDA Approves Nivolumab Plus Ipilimumab for First-Line mNSCLC (PD-L1 Tumor Expression ≥1%)|FDA. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-nivolumab-plus-ipilimumab-first-line-mnsclc-pd-l1-tumor-expression-1 (accessed on 16 May 2022).
- Paz-Ares, L.; Ciuleanu, T.E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 198–211. [Google Scholar] [CrossRef] [PubMed]
- FDA Approves Nivolumab Plus Ipilimumab and Chemotherapy for First-Line Treatment of Metastatic NSCLC|FDA. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-nivolumab-plus-ipilimumab-and-chemotherapy-first-line-treatment-metastatic-nsclc (accessed on 16 May 2022).
- Johnson, M.L.; Cho, B.C.; Luft, A.; Alatorre-Alexander, J.; Geater, S.L.; Laktionov, K.; Kim, S.-W.; Ursol, G.; Hussein, M.; Lim, F.L.; et al. Durvalumab With or Without Tremelimumab in Combination With Chemotherapy as First-Line Therapy for Metastatic Non–Small-Cell Lung Cancer: The Phase III POSEIDON Study. J. Clin. Oncol. 2022. [Google Scholar] [CrossRef] [PubMed]
- Horn, L.; Mansfield, A.S.; Szczęsna, A.; Havel, L.; Krzakowski, M.; Hochmair, M.J.; Huemer, F.; Losonczy, G.; Johnson, M.L.; Nishio, M.; et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2220–2229. [Google Scholar] [CrossRef] [PubMed]
- Giunchi, F.; Degiovanni, A.; Daddi, N.; Trisolini, R.; Dell’Amore, A.; Agostinelli, C.; Ardizzoni, A.; Fiorentino, M. Fading with time of PD-L1 immunoreactivity in non-small cells lung cancer tissues: A methodological study. Appl. Immunohistochem. Mol. Morphol. 2018, 26, 489–494. [Google Scholar] [CrossRef] [PubMed]
- Ameratunga, M.; Asadi, K.; Lin, X.; Walkiewicz, M.; Murone, C.; Knight, S.; Mitchell, P.; Boutros, P.; John, T. PD-L1 and Tumor infiltrating lymphocytes as prognostic markers in resected NSCLC. PLoS ONE 2016, 11, e0153954. [Google Scholar] [CrossRef] [Green Version]
- Casadevall, D.; Clavé, S.; Taus, Á.; Hardy-Werbin, M.; Rocha, P.; Lorenzo, M.; Menéndez, S.; Salido, M.; Albanell, J.; Pijuan, L.; et al. Heterogeneity of Tumor and Immune Cell PD-L1 Expression and Lymphocyte Counts in Surgical NSCLC Samples. Clin. Lung Cancer 2017, 18, 682–691.e5. [Google Scholar] [CrossRef]
- Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the Treatment of Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef]
- Jaafar, J.; Fernandez, E.; Alwan, H.; Philippe, J. Programmed cell death-1 and programmed cell death ligand-1 antibodies-induced dysthyroidism. Endocr. Connect. 2018, 7, R196–R211. [Google Scholar] [CrossRef] [Green Version]
- Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
- Spira, A.I.; Park, K.; Mazières, J.; Vansteenkiste, J.F.; Rittmeyer, A.; Ballinger, M.; Waterkamp, D.; Kowanetz, M.; Mokatrin, A.; Fehrenbacher, L. Efficacy, safety and predictive biomarker results from a randomized phase II study comparing MPDL3280A vs docetaxel in 2L/3L NSCLC (POPLAR). J. Clin. Oncol. 2015, 33, 8010. [Google Scholar] [CrossRef]
- Dietel, M.; Savelov, N.; Salanova, R.; Micke, P.; Bigras, G.; Hida, T.; Antunez, J.; Guldhammer Skov, B.; Hutarew, G.; Sua, L.F.; et al. Real-world prevalence of programmed death ligand 1 expression in locally advanced or metastatic non–small-cell lung cancer: The global, multicenter EXPRESS study. Lung Cancer 2019, 134, 174–179. [Google Scholar] [CrossRef] [PubMed]
- Skov, B.G.; Rørvig, S.B.; Jensen, T.H.L.; Skov, T. The prevalence of programmed death ligand-1 (PD-L1) expression in non-small cell lung cancer in an unselected, consecutive population. Mod. Pathol. 2020, 33, 109–117. [Google Scholar] [CrossRef] [PubMed]
- Holmes, M.; Mahar, A.; Lum, T.; Boyer, M.; Kao, S.; Cooper, W. P1.09-26 Prevalence of PD-L1 Expression Rates in Different NSCLC Specimens. J. Thorac. Oncol. 2019, 14, S506. [Google Scholar] [CrossRef]
- Ullah, A.; Pulliam, S.; Karki, N.R.; Khan, J.; Jogezai, S.; Sultan, S.; Muhammad, L.; Khan, M.; Jamil, N.; Waheed, A.; et al. PD-L1 Over-Expression Varies in Different Subtypes of Lung Cancer: Will This Affect Future Therapies? Clin. Pract. 2022, 12, 653–671. [Google Scholar] [CrossRef]
- Zhang, P.P.; Wang, J.; Ding, D.Z.; Zhang, L.; Cheng, C.; Chen, D.K. Efficacy and safety of PD-1/PD-L1 inhibitors combined with CTLA-4 inhibitor versus chemotherapy for advanced lung cancer: A meta-analysis. Medicine 2021, 100, e27121. [Google Scholar] [CrossRef]
- Marin-Acevedo, J.A.; Kimbrough, E.M.O.; Lou, Y. Next generation of immune checkpoint inhibitors and beyond. J. Hematol. Oncol. 2021, 14, 45. [Google Scholar] [CrossRef]
- Huard, B.; Mastrangeli, R.; Prigent, P.; Bruniquel, D.; Donini, S.; El-Tayar, N.; Maigret, B.; Dréano, M.; Triebel, F. Characterization of the major histocompatibility complex class II binding site on LAG-3 protein. Proc. Natl. Acad. Sci. USA 1997, 94, 5744–5749. [Google Scholar] [CrossRef] [Green Version]
- Triebel, F.; Jitsukawa, S.; Baixeras, E.; Roman-Roman, S.; Genevee, C.; Viegas-Pequignot, E.; Hercend, T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J. Exp. Med. 1990, 171, 1393–1405. [Google Scholar] [CrossRef] [Green Version]
- Huard, B.; Tournier, M.; Triebel, F. LAG-3 does not define a specific mode of natural killing in human. Immunol. Lett. 1998, 61, 109–112. [Google Scholar] [CrossRef]
- Kisielow, M.; Kisielow, J.; Capoferri-Sollami, G.; Karjalainen, K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur. J. Immunol. 2005, 35, 2081–2088. [Google Scholar] [CrossRef]
- Andreae, S.; Piras, F.; Burdin, N.; Triebel, F. Maturation and Activation of Dendritic Cells Induced by Lymphocyte Activation Gene-3 (CD223). J. Immunol. 2002, 168, 3874–3880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mao, X.; Ou, M.T.; Karuppagounder, S.S.; Kam, T.I.; Yin, X.; Xiong, Y.; Ge, P.; Umanah, G.E.; Brahmachari, S.; Shin, J.H.; et al. Pathological α-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science 2016, 353, aah3374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maçon-Lemaître, L.; Triebel, F. The negative regulatory function of the lymphocyte-activation gene-3 co-receptor (CD223) on human T cells. Immunology 2005, 115, 170–178. [Google Scholar] [CrossRef] [PubMed]
- Huard, B.; Tournier, M.; Hercend, T.; Triebel, F.; Faure, F. Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes. Eur. J. Immunol. 1994, 24, 3216–3221. [Google Scholar] [CrossRef]
- Workman, C.J.; Dugger, K.J.; Vignali, D.A.A. Cutting Edge: Molecular Analysis of the Negative Regulatory Function of Lymphocyte Activation Gene-3. J. Immunol. 2002, 169, 5392–5395. [Google Scholar] [CrossRef] [Green Version]
- Maruhashi, T.; Sugiura, D.; Okazaki, I.M.; Okazaki, T. LAG-3: From molecular functions to clinical applications. J. Immunother. cancer 2020, 8, e001014. [Google Scholar] [CrossRef]
- Baumeister, S.H.; Freeman, G.J.; Dranoff, G.; Sharpe, A.H. Coinhibitory Pathways in Immunotherapy for Cancer. Annu. Rev. Immunol. 2016, 34, 539–573. [Google Scholar] [CrossRef]
- Woo, S.R.; Turnis, M.E.; Goldberg, M.V.; Bankoti, J.; Selby, M.; Nirschl, C.J.; Bettini, M.L.; Gravano, D.M.; Vogel, P.; Liu, C.L.; et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012, 72, 917–927. [Google Scholar] [CrossRef] [Green Version]
- Initial results from a phase II study (TACTI-002) in metastatic non-small cell lung or head and neck carcinoma patients receiving eftilagimod alpha (soluble LAG-3 protein) and pembrolizumab. J. Clin. Oncol. 2020, 38, 3100. [CrossRef]
- Lipson, E.J.; Tawbi, H.A.-H.; Schadendorf, D.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.C.; Rutkowski, P.; Gogas, H.; Lao, C.D.; Janoski de Menezes, J.; et al. Relatlimab (RELA) plus nivolumab (NIVO) versus NIVO in first-line advanced melanoma: Primary phase III results from RELATIVITY-047 (CA224-047). J. Clin. Oncol. 2021, 39, 9503. [Google Scholar] [CrossRef]
- Uboha, N.V.; Milhem, M.M.; Kovacs, C.; Amin, A.; Magley, A.; Purkayastha, D.D.; Piha-Paul, S.A. Phase II study of spartalizumab (PDR001) and LAG525 in advanced solid tumors and hematologic malignancies. J. Clin. Oncol. 2019, 37, 2553. [Google Scholar] [CrossRef]
- Schöffski, P.; Tan, D.S.W.; Martín, M.; Ochoa-De-Olza, M.; Sarantopoulos, J.; Carvajal, R.D.; Kyi, C.; Esaki, T.; Prawira, A.; Akerley, W.; et al. Phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) ± anti-PD-1 spartalizumab (PDR001) in patients with advanced malignancies. J. Immunother. Cancer 2022, 10, 3776. [Google Scholar] [CrossRef] [PubMed]
- Papadopoulos, K.P.; Lakhani, N.J.; Johnson, M.L.; Park, H.; Wang, D.; Yap, T.A.; Dowlati, A.; Maki, R.G.; Lynce, F.; Ulahannan, S.V.; et al. First-in-human study of REGN3767 (R3767), a human LAG-3 monoclonal antibody (mAb), ± cemiplimab in patients (pts) with advanced malignancies. J. Clin. Oncol. 2019, 37, 2508. [Google Scholar] [CrossRef]
- Johnson, M.L.; Patel, M.R.; Cherry, M.; Kang, Y.-K.; Yamaguchi, K.; Oh, D.-Y.; Hussein, M.A.; Kitano, S.; Kondo, S.; Hansen, A.R.; et al. Safety of BI 754111, an anti-LAG-3 monoclonal antibody (mAb), in combination with BI 754091, an anti-PD-1 mAb, in patients with advanced solid tumors. J. Clin. Oncol. 2020, 38, 3063. [Google Scholar] [CrossRef]
- Phase I Studies of Sym021, an anti-PD-1 Antibody, Alone and in Combination with Sym022 (anti-LAG-3) or Sym023 (Anti-TIM-3)|OncologyPRO. Available online: https://oncologypro.esmo.org/meeting-resources/esmo-virtual-congress-2020/phase-i-studies-of-sym021-an-anti-pd-1-antibody-alone-and-in-combination-with-sym022-anti-lag-3-or-sym023-anti-tim-3 (accessed on 16 May 2022).
- Luke, J.J.; Patel, M.R.; Hamilton, E.P.; Chmielowski, B.; Ulahannan, S.V.; Kindler, H.L.; Bahadur, S.W.; Clingan, P.R.; Mallesara, G.; Weickhardt, A.J.; et al. A phase I, first-in-human, open-label, dose-escalation study of MGD013, a bispecific DART molecule binding PD-1 and LAG-3, in patients with unresectable or metastatic neoplasms. J. Clin. Oncol. 2020, 38, 3004. [Google Scholar] [CrossRef]
- Long, L.; Zhang, X.; Chen, F.; Pan, Q.; Phiphatwatchara, P.; Zeng, Y.; Chen, H. The promising immune checkpoint LAG-3: From tumor microenvironment to cancer immunotherapy. Genes Cancer 2018, 9, 176–189. [Google Scholar] [CrossRef] [Green Version]
- Paz-Ares, L.; Dvorkin, M.; Chen, Y.; Reinmuth, N.; Hotta, K.; Trukhin, D.; Statsenko, G.; Hochmair, M.J.; Özgüroğlu, M.; Ji, J.H.; et al. Durvalumab plus platinum–etoposide versus platinum–etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): A randomised, controlled, open-label, phase 3 trial. Lancet 2019, 394, 1929–1939. [Google Scholar] [CrossRef]
- Levin, S.D.; Taft, D.W.; Brandt, C.S.; Bucher, C.; Howard, E.D.; Chadwick, E.M.; Johnston, J.; Hammond, A.; Bontadelli, K.; Ardourel, D.; et al. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. Eur. J. Immunol. 2011, 41, 902–915. [Google Scholar] [CrossRef]
- Boles, K.S.; Vermi, W.; Facchetti, F.; Fuchs, A.; Wilson, T.J.; Diacovo, T.G.; Cella, M.; Colonna, M. A novel molecular interaction for the adhesion of follicular CD4 T cells to follicular DC. Eur. J. Immunol. 2009, 39, 695–703. [Google Scholar] [CrossRef]
- Stanietsky, N.; Simic, H.; Arapovic, J.; Toporik, A.; Levy, O.; Novik, A.; Levine, Z.; Beiman, M.; Dassa, L.; Achdout, H.; et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 17858–17863. [Google Scholar] [CrossRef] [Green Version]
- Dougall, W.C.; Kurtulus, S.; Smyth, M.J.; Anderson, A.C. TIGIT and CD96: New checkpoint receptor targets for cancer immunotherapy. Immunol. Rev. 2017, 276, 112–120. [Google Scholar] [CrossRef]
- Le Mercier, I.; Lines, J.L.; Noelle, R.J. Beyond CTLA-4 and PD-1, the generation Z of negative checkpoint regulators. Front. Immunol. 2015, 6, 418. [Google Scholar] [CrossRef]
- Lozano, E.; Dominguez-Villar, M.; Kuchroo, V.; Hafler, D.A. The TIGIT/CD226 Axis Regulates Human T Cell Function. J. Immunol. 2012, 188, 3869–3875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stanietsky, N.; Rovis, T.L.; Glasner, A.; Seidel, E.; Tsukerman, P.; Yamin, R.; Enk, J.; Jonjic, S.; Mandelboim, O. Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR. Eur. J. Immunol. 2013, 43, 2138–2150. [Google Scholar] [CrossRef]
- Liu, S.; Zhang, H.; Li, M.; Hu, D.; Li, C.; Ge, B.; Jin, B.; Fan, Z. 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. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Xia, P.; Du, Y.; Liu, S.; Huang, G.; Chen, J.; Zhang, H.; Hou, N.; Cheng, X.; Zhou, L.; 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. [Google Scholar] [CrossRef] [Green Version]
- Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 protein of fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef] [Green Version]
- Manieri, N.A.; Chiang, E.Y.; Grogan, J.L. TIGIT: A Key Inhibitor of the Cancer Immunity Cycle. Trends Immunol. 2017, 38, 20–28. [Google Scholar] [CrossRef]
- Chauvin, J.M.; Zarour, H.M. TIGIT in cancer immunotherapy. J. Immunother. Cancer 2020, 8, 957. [Google Scholar] [CrossRef]
- Hosseinkhani, N.; Shadbad, M.A.; Asghari Jafarabadi, M.; Karim Ahangar, N.; Asadzadeh, Z.; Mohammadi, S.M.; Lotfinejad, P.; Alizadeh, N.; Brunetti, O.; Fasano, R.; et al. A Systematic Review and Meta-Analysis on the Significance of TIGIT in Solid Cancers: Dual TIGIT/PD-1 Blockade to Overcome Immune-Resistance in Solid Cancers. Int. J. Mol. Sci. 2021, 22, 10389. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.; Chen, F.; Liu, D.; Gu, F.; Chen, Z.; Wang, Y. Prognostic value of immune checkpoint molecules in breast cancer. Biosci. Rep. 2020, 40, BSR20201054. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Abreu, D.; Johnson, M.L.; Hussein, M.A.; Cobo, M.; Patel, A.J.; Secen, N.M.; Lee, K.H.; Massuti, B.; Hiret, S.; Yang, J.C.-H.; 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. [Google Scholar] [CrossRef]
- Rudin, C.M.; Liu, S.V.; Lu, S.; Soo, R.A.; Hong, M.H.; Lee, J.-S.; Bryl, M.; Dumoulin, D.W.; Rittmeyer, A.; Chiu, C.-H.; et al. SKYSCRAPER-02: Primary results of a phase III, randomized, double-blind, placebo-controlled study of atezolizumab (atezo) + carboplatin + etoposide (CE) with or without tiragolumab (tira) in patients (pts) with untreated extensive-stage small cell lung cancer (ES-SCLC). J. Clin. Oncol. 2022, 40, LBA8507. [Google Scholar] [CrossRef]
- Ahn, M.-J.; Niu, J.; Kim, D.-W.; Rasco, D.; Mileham, K.F.; Chung, H.C.; Vaishampayan, U.N.; Maurice-Dror, C.; Lo Russo, P.; Golan, T.; et al. 1400P Vibostolimab, an anti-TIGIT antibody, as monotherapy and in combination with pembrolizumab in anti-PD-1/PD-L1-refractory NSCLC. Ann. Oncol. 2020, 31, S887. [Google Scholar] [CrossRef]
- Nagahara, K.; Arikawa, T.; Oomizu, S.; Kontani, K.; Nobumoto, A.; Tateno, H.; Watanabe, K.; Niki, T.; Katoh, S.; Miyake, M.; et al. Galectin-9 Increases Tim-3 + Dendritic Cells and CD8 + T Cells and Enhances Antitumor Immunity via Galectin-9-Tim-3 Interactions. J. Immunol. 2008, 181, 7660–7669. [Google Scholar] [CrossRef] [Green Version]
- Van de Weyer, P.S.; Muehlfeit, M.; Klose, C.; Bonventre, J.V.; Walz, G.; Kuehn, E.W. A highly conserved tyrosine of Tim-3 is phosphorylated upon stimulation by its ligand galectin-9. Biochem. Biophys. Res. Commun. 2006, 351, 571–576. [Google Scholar] [CrossRef]
- Anderson, A.C. Tim-3, a negative regulator of anti-tumor immunity. Curr. Opin. Immunol. 2012, 24, 213–216. [Google Scholar] [CrossRef]
- Liu, F.; Liu, Y.; Chen, Z. Tim-3 expression and its role in hepatocellular carcinoma. J. Hematol. Oncol. 2018, 11, 126. [Google Scholar] [CrossRef]
- Huang, Y.-H.; Zhu, C.; Kondo, Y.; Anderson, A.C.; Gandhi, A.; Russell, A.; Dougan, S.K.; Petersen, B.-S.; Melum, E.; Pertel, T.; et al. Corrigendum: CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 2016, 536, 359. [Google Scholar] [CrossRef]
- Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeKruyff, R.H.; Bu, X.; Ballesteros, A.; Santiago, C.; Chim, Y.-L.E.; Lee, H.-H.; Karisola, P.; Pichavant, M.; Kaplan, G.G.; Umetsu, D.T.; et al. T Cell/Transmembrane, Ig, and Mucin-3 Allelic Variants Differentially Recognize Phosphatidylserine and Mediate Phagocytosis of Apoptotic Cells. J. Immunol. 2010, 184, 1918–1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saleh, R.; Toor, S.M.; Elkord, E. Targeting TIM-3 in solid tumors: Innovations in the preclinical and translational realm and therapeutic potential. Expert Opin. Ther. Targets 2020, 24, 1251–1262. [Google Scholar] [CrossRef] [PubMed]
- Zang, K.; Hui, L.; Wang, M.; Huang, Y.; Zhu, X.; Yao, B. TIM-3 as a Prognostic Marker and a Potential Immunotherapy Target in Human Malignant Tumors: A Meta-Analysis and Bioinformatics Validation. Front. Oncol. 2021, 11, 273. [Google Scholar] [CrossRef]
- Harding, J.J.; Patnaik, A.; Moreno, V.; Stein, M.; Jankowska, A.M.; Velez de Mendizabal, N.; Tina Liu, Z.; Koneru, M.; Calvo, E. A phase Ia/Ib study of an anti-TIM-3 antibody (LY3321367) monotherapy or in combination with an anti-PD-L1 antibody (LY3300054): Interim safety, efficacy, and pharmacokinetic findings in advanced cancers. J. Clin. Oncol. 2019, 37, 12. [Google Scholar] [CrossRef]
- Curigliano, G.; Gelderblom, H.; Mach, N.; Doi, T.; Tai, D.; Forde, P.M.; Sarantopoulos, J.; Bedard, P.L.; Lin, C.C.; Stephen Hodi, F.; et al. Phase I/Ib clinical trial of sabatolimab, an anti–TIM-3 antibody, alone and in combination with spartalizumab, an anti–PD-1 antibody, in advanced solid tumors. Clin. Cancer Res. 2021, 27, 3620–3629. [Google Scholar] [CrossRef]
- Hosseinkhani, N.; Derakhshani, A.; Shadbad, M.A.; Argentiero, A.; Racanelli, V.; Kazemi, T.; Mokhtarzadeh, A.; Brunetti, O.; Silvestris, N.; Baradaran, B. The Role of V-Domain Ig Suppressor of T Cell Activation (VISTA) in Cancer Therapy: Lessons Learned and the Road Ahead. Front. Immunol. 2021, 12, 676181. [Google Scholar] [CrossRef]
- Flies, D.B.; Wang, S.; Xu, H.; Chen, L. Cutting Edge: A Monoclonal Antibody Specific for the Programmed Death-1 Homolog Prevents Graft-versus-Host Disease in Mouse Models. J. Immunol. 2011, 187, 1537–1541. [Google Scholar] [CrossRef] [Green Version]
- Lines, J.L.; Sempere, L.F.; Broughton, T.; Wang, L.; Noelle, R. VISTA Is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol. Res. 2014, 2, 510–517. [Google Scholar] [CrossRef] [Green Version]
- Le Mercier, I.; Chen, W.; Lines, J.L.; Day, M.; Li, J.; Sergent, P.; Noelle, R.J.; Wang, L. VISTA regulates the development of protective antitumor immunity. Cancer Res. 2014, 74, 1933–1944. [Google Scholar] [CrossRef]
- Zhang, M.; Pang, H.J.; Zhao, W.; Li, Y.F.; Yan, L.X.; Dong, Z.Y.; He, X.F. VISTA expression associated with CD8 confers a favorable immune microenvironment and better overall survival in hepatocellular carcinoma. BMC Cancer 2018, 18, 511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loeser, H.; Kraemer, M.; Gebauer, F.; Bruns, C.; Schröder, W.; Zander, T.; Persa, O.D.; Alakus, H.; Hoelscher, A.; Buettner, R.; et al. The expression of the immune checkpoint regulator VISTA correlates with improved overall survival in pT1/2 tumor stages in esophageal adenocarcinoma. Oncoimmunology 2019, 8, e1581546. [Google Scholar] [CrossRef] [PubMed]
- Xu, W.; Hiếu, T.M.; Malarkannan, S.; Wang, L. The structure, expression, and multifaceted role of immune-checkpoint protein VISTA as a critical regulator of anti-tumor immunity, autoimmunity, and inflammation. Cell. Mol. Immunol. 2018, 15, 438–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zong, L.; Zhou, Y.; Zhang, M.; Chen, J.; Xiang, Y. VISTA expression is associated with a favorable prognosis in patients with high-grade serous ovarian cancer. Cancer Immunol. Immunother. 2020, 69, 33–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuklinski, L.F.; Yan, S.; Li, Z.; Fisher, J.L.; Cheng, C.; Noelle, R.J.; Angeles, C.V.; Turk, M.J.; Ernstoff, M.S. VISTA expression on tumor-infiltrating inflammatory cells in primary cutaneous melanoma correlates with poor disease-specific survival. Cancer Immunol. Immunother. 2018, 67, 1113–1121. [Google Scholar] [CrossRef]
- Böger, C.; Behrens, H.M.; Krüger, S.; Röcken, C. The novel negative checkpoint regulator VISTA is expressed in gastric carcinoma and associated with PD-L1/PD-1: A future perspective for a combined gastric cancer therapy? Oncoimmunology 2017, 6, e1293215. [Google Scholar] [CrossRef]
- Wu, L.; Deng, W.W.; Huang, C.F.; Bu, L.L.; Yu, G.T.; Mao, L.; Zhang, W.F.; Liu, B.; Sun, Z.J. Expression of VISTA correlated with immunosuppression and synergized with CD8 to predict survival in human oral squamous cell carcinoma. Cancer Immunol. Immunother. 2017, 66, 627–636. [Google Scholar] [CrossRef]
- Liu, J.; Yuan, Y.; Chen, W.; Putra, J.; Suriawinata, A.A.; Schenk, A.D.; Miller, H.E.; Guleria, I.; Barth, R.J.; Huang, Y.H.; et al. Immune-checkpoint proteins VISTA and PD-1 nonredundantly regulate murine T-cell responses. Proc. Natl. Acad. Sci. USA 2015, 112, 6682–6687. [Google Scholar] [CrossRef] [Green Version]
- Powderly, J.; Patel, M.R.; Lee, J.J.; Brody, J.; Meric-Bernstam, F.; Hamilton, E.; Ponce Aix, S.; Garcia-Corbacho, J.; Bang, Y.-J.; Ahn, M.-J.; et al. CA-170, a first in class oral small molecule dual inhibitor of immune checkpoints PD-L1 and VISTA, demonstrates tumor growth inhibition in pre-clinical models and promotes T cell activation in Phase 1 study. Ann. Oncol. 2017, 28, v405–v406. [Google Scholar] [CrossRef]
- Excellent CBR and Prolonged PFS in Non-Squamous NSCLC with Oral CA-170, an Inhibitor of VISTA and PD-L1 | OncologyPRO. Available online: https://oncologypro.esmo.org/meeting-resources/esmo-2019-congress/excellent-cbr-and-prolonged-pfs-in-non-squamous-nsclc-with-oral-ca-170-an-inhibitor-of-vista-and-pd-l1 (accessed on 16 May 2022).
- Hutloff, A.; Dittrich, A.M.; Beier, K.C.; Eljaschewitsch, B.; Kraft, R.; Anagnostopoulos, I.; Kroczek, R.A. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 1999, 397, 263–266. [Google Scholar] [CrossRef]
- Solinas, C.; Gu-Trantien, C.; Willard-Gallo, K. The rationale behind targeting the ICOS-ICOS ligand costimulatory pathway in cancer immunotherapy. ESMO Open 2020, 5, e000544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoshinaga, S.K.; Whorlskey, J.S.; Khare, S.D.; Sarmiento, U.; Guo, J.; Horan, T.; Shih, G.; Zhang, M.; Coccia, M.A.; Kohno, T.; et al. T-cell co-stimulation through B7RP-1 and ICOS. Nature 1999, 402, 827–830. [Google Scholar] [CrossRef] [PubMed]
- Amatore, F.; Gorvel, L.; Olive, D. Inducible co-stimulator (ICOS) as a potential therapeutic target for anti-cancer therapy. Expert Opin. Ther. Targets 2018, 22, 343–351. [Google Scholar] [CrossRef] [PubMed]
- Le, K.S.; Thibult, M.L.; Just-Landi, S.; Pastor, S.; Gondois-Rey, F.; Granjeaud, S.; Broussais, F.; Bouabdallah, R.; Colisson, R.; Caux, C.; et al. Follicular B lymphomas generate regulatory T cells via the ICOS/ICOSL pathway and are susceptible to treatment by anti-ICOS/ICOSL therapy. Cancer Res. 2016, 76, 4648–4660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, X.; Quezada, S.A.; Sepulveda, M.A.; Sharma, P.; Allison, J.P. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J. Exp. Med. 2014, 211, 715–725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Massarelli, E.; Balmanoukian, A.S.; Vieito, M.; Le Tourneau, C.; Hernandez-Guerrero, T.; Trigo, J.M.; Aljumaily, R.; Chisamore, M.J.; Rogan, D.; Sung, R.; et al. INDUCE-1: Report on safety run-in cohorts combining Inducible T-cell co-stimulatory receptor (ICOS) agonist GSK3359609 (GSK609) with platinum+5-FU chemotherapy (5-FU/plat), with or without pembrolizumab (PE), for the treatment of advanced solid tumors. J. Clin. Oncol. 2020, 38, 6544. [Google Scholar] [CrossRef]
- Sanmamed, M.F.; Pastor, F.; Rodriguez, A.; Perez-Gracia, J.L.; Rodriguez-Ruiz, M.E.; Jure-Kunkel, M.; Melero, I. Agonists of Co-stimulation in Cancer Immunorapy Directed Against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin. Oncol. 2015, 42, 640–655. [Google Scholar] [CrossRef]
- Patel, M.R.; Naing, A.; Burris III, H.A.; Lin, C.-C.; Curigliano, G.; Thistlethwaite, F.; Minchom, A.R.; Ascierto, P.A.; De Braud, F.G.; Cecchini, M.; et al. A phase 1/2 open-label study of KY1044, an anti-ICOS antibody with dual mechanism of action, as single agent and in combination with atezolizumab, in adult patients with advanced malignancies. J. Clin. Oncol. 2021, 39, 2624. [Google Scholar] [CrossRef]
- Yap, T.A.; Burris, H.A.; Kummar, S.; Falchook, G.S.; Pachynski, R.K.; LoRusso, P.; Tykodi, S.S.; Gibney, G.T.; Gainor, J.F.; Rahma, O.E.; et al. ICONIC: Biologic and clinical activity of first in class ICOS agonist antibody JTX-2011 +/- nivolumab (nivo) in patients (pts) with advanced cancers. J. Clin. Oncol. 2018, 36, 3000. [Google Scholar] [CrossRef]
- Sun, M.; Richards, S.; Prasad, D.V.R.; Mai, X.M.; Rudensky, A.; Dong, C. Characterization of Mouse and Human B7-H3 Genes. J. Immunol. 2002, 168, 6294–6297. [Google Scholar] [CrossRef]
- Steinberger, P.; Majdic, O.; Derdak, S.V.; Pfistershammer, K.; Kirchberger, S.; Klauser, C.; Zlabinger, G.; Pickl, W.F.; Stöckl, J.; Knapp, W. Molecular Characterization of Human 4Ig-B7-H3, a Member of the B7 Family with Four Ig-Like Domains. J. Immunol. 2004, 172, 2352–2359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, G.; Hou, J.; Shi, J.; Yu, G.; Lu, B.; Zhang, X. Soluble CD276 (B7-H3) is released from monocytes, dendritic cells and activated T cells and is detectable in normal human serum. Immunology 2008, 123, 538–546. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Liu, P.; Wang, Y.; Nie, W.; Li, Z.; Xu, W.; Li, F.; Zhou, Z.; Zhao, M.; Liu, H. Characterization of a Soluble B7-H3 (sB7-H3) Spliced from the Intron and Analysis of sB7-H3 in the Sera of Patients with Hepatocellular Carcinoma. PLoS ONE 2013, 8, e76965. [Google Scholar] [CrossRef] [PubMed]
- Prasad, D.V.R.; Nguyen, T.; Li, Z.; Yang, Y.; Duong, J.; Wang, Y.; Dong, C. Murine B7-H3 Is a Negative Regulator of T Cells. J. Immunol. 2004, 173, 2500–2506. [Google Scholar] [CrossRef] [Green Version]
- Dong, P.; Xiong, Y.; Yue, J.; Hanley, S.J.B.; Watari, H. B7H3 As a promoter of metastasis and promising therapeutic target. Front. Oncol. 2018, 8, 264. [Google Scholar] [CrossRef] [Green Version]
- Janakiram, M.; Shah, U.A.; Liu, W.; Zhao, A.; Schoenberg, M.P.; Zang, X. The third group of the B7-CD28 immune checkpoint family: HHLA2, TMIGD2, B7x, and B7-H3. Immunol. Rev. 2017, 276, 26–39. [Google Scholar] [CrossRef] [Green Version]
- Hashiguchi, M.; Kobori, H.; Ritprajak, P.; Kamimura, Y.; Kozono, H.; Azuma, M. Triggering receptor expressed on myeloid cell-like transcript 2 (TLT-2) is a counter-receptor for B7-H3 and enhances T cell responses. Proc. Natl. Acad. Sci. USA 2008, 105, 10495–10500. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Zhang, G.; Zhang, X.; Lv, G.; Wei, X.; Yuan, H.; Hou, J. Overexpression of B7-H3 in CD14+ monocytes is associated with renal cell carcinoma progression. Med. Oncol. 2014, 31, 349. [Google Scholar] [CrossRef]
- Jin, Y.; Zhang, P.; Li, J.; Zhao, J.; Liu, C.; Yang, F.; Yang, D.; Gao, A.; Lin, W.; Ma, X.; et al. B7-H3 in combination with regulatory T cell is associated with tumor progression in primary human non-small cell lung cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 13987–13995. [Google Scholar]
- Benzon, B.; Zhao, S.G.; Haffner, M.C.; Takhar, M.; Erho, N.; Yousefi, K.; Hurley, P.; Bishop, J.L.; Tosoian, J.; Ghabili, K.; et al. Correlation of B7-H3 with androgen receptor, immune pathways and poor outcome in prostate cancer: An expression-based analysis. Prostate Cancer Prostatic Dis. 2017, 20, 28–35. [Google Scholar] [CrossRef]
- Mao, Y.; Chen, L.; Wang, F.; Zhu, D.; Ge, X.; Hua, D.; Sun, J. Cancer cell-expressed B7-H3 regulates the differentiation of tumor-associated macrophages in human colorectal carcinoma. Oncol. Lett. 2017, 14, 6177–6183. [Google Scholar] [CrossRef] [PubMed]
- Powderly, J.; Cote, G.; Flaherty, K.; Szmulewitz, R.Z.; Ribas, A.; Weber, J.; Loo, D.; Baughman, J.; Chen, F.; Moore, P.; et al. Interim results of an ongoing Phase I, dose escalation study of MGA271 (Fc-optimized humanized anti-B7-H3 monoclonal antibody) in patients with refractory B7-H3-expressing neoplasms or neoplasms whose vasculature expresses B7-H3. J. Immunother. Cancer 2015, 3, O8. [Google Scholar] [CrossRef] [Green Version]
- Ceeraz, S.; Nowak, E.C.; Noelle, R.J. B7 family checkpoint regulators in immune regulation and disease. Trends Immunol. 2013, 34, 556–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paulos, C.M.; June, C.H. Putting the brakes on BTLA in T cell-mediated cancer immunotherapy. J. Clin. Investig. 2010, 120, 76–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gavrieli, M.; Watanabe, N.; Loftin, S.K.; Murphy, T.L.; Murphy, K.M. Characterization of phosphotyrosine binding motifs in the cytoplasmic domain of B and T lymphocyte attenuator required for association with protein tyrosine phosphatases SHP-1 and SHP-2. Biochem. Biophys. Res. Commun. 2003, 312, 1236–1243. [Google Scholar] [CrossRef]
- Murphy, T.L.; Murphy, K.M. Slow Down and Survive: Enigmatic Immunoregulation by BTLA and HVEM. Annu. Rev. Immunol. 2010, 28, 389–411. [Google Scholar] [CrossRef]
- Sedy, J.R.; Gavrieli, M.; Potter, K.G.; Hurchla, M.A.; Lindsley, R.C.; Hildner, K.; Scheu, S.; Pfeffer, K.; Ware, C.F.; Murphy, T.L.; et al. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nat. Immunol. 2005, 6, 90–98. [Google Scholar] [CrossRef]
- Cai, G.; Anumanthan, A.; Brown, J.A.; Greenfield, E.A.; Zhu, B.; Freeman, G.J. CD160 inhibits activation of human CD4+ T cells through interaction with herpesvirus entry mediator. Nat. Immunol. 2008, 9, 176–185. [Google Scholar] [CrossRef]
- Rodriguez-Barbosa, J.I.; Schneider, P.; Weigert, A.; Lee, K.-M.; Kim, T.-J.; Perez-Simon, J.-A.; del Rio, M.-L. HVEM, a cosignaling molecular switch, and its interactions with BTLA, CD160 and LIGHT. Cell. Mol. Immunol. 2019, 16, 679–682. [Google Scholar] [CrossRef]
- Murphy, K.M.; Nelson, C.A.; Šedý, J.R. Balancing co-stimulation and inhibition with BTLA and HVEM. Nat. Rev. Immunol. 2006, 6, 671–681. [Google Scholar] [CrossRef]
- Song, J.; Wu, L. Friend or Foe: Prognostic and Immunotherapy Roles of BTLA in Colorectal Cancer. Front. Mol. Biosci. 2020, 7, 148. [Google Scholar] [CrossRef]
- Mo, L.; Chen, Q.; Zhang, X.; Shi, X.; Wei, L.; Zheng, D.; Li, H.; Gao, J.; Li, J.; Hu, Z. Depletion of regulatory T cells by anti-ICOS antibody enhances anti-tumor immunity of tumor cell vaccine in prostate cancer. Vaccine 2017, 35, 5932–5938. [Google Scholar] [CrossRef] [PubMed]
- Moon, Y.W.; Hajjar, J.; Hwu, P.; Naing, A. Targeting the indoleamine 2,3-dioxygenase pathway in cancer. J. Immunother. Cancer 2015, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theate, I.; Van Baren, N.; Pilotte, L.; Moulin, P.; Larrieu, P.; Renauld, J.C.; Herve, C.; Gutierrez-Roelens, I.; Marbaix, E.; Sempoux, C.; et al. Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues. Cancer Immunol. Res. 2015, 3, 161–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munn, D.H. Indoleamine 2,3-dioxygenase, tumor-induced tolerance and counter-regulation. Curr. Opin. Immunol. 2006, 18, 220–225. [Google Scholar] [CrossRef]
- Mondal, A.; Smith, C.; DuHadaway, J.B.; Sutanto-Ward, E.; Prendergast, G.C.; Bravo-Nuevo, A.; Muller, A.J. IDO1 is an Integral Mediator of Inflammatory Neovascularization. EBioMedicine 2016, 14, 74–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prendergast, G.C. Immune escape as a fundamental trait of cancer: Focus on IDO. Oncogene 2008, 27, 3889–3900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spranger, S.; Koblish, H.K.; Horton, B.; Scherle, P.A.; Newton, R.; Gajewski, T.F. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8+ T cells directly within the tumor microenvironment. J. Immunother. Cancer 2014, 2, 3. [Google Scholar] [CrossRef] [Green Version]
- Gibney, G.T.; Hamid, O.; Lutzky, J.; Olszanski, A.J.; Mitchell, T.C.; Gajewski, T.F.; Chmielowski, B.; Hanks, B.A.; Zhao, Y.; Newton, R.C.; et al. Phase 1/2 study of epacadostat in combination with ipilimumab in patients with unresectable or metastatic melanoma. J. Immunother. Cancer 2019, 7, 80. [Google Scholar] [CrossRef] [Green Version]
- Beatty, G.L.; O’Dwyer, P.J.; Clark, J.; Shi, J.G.; Bowman, K.J.; Scherle, P.A.; Newton, R.C.; Schaub, R.; Maleski, J.; Leopold, L.; et al. First-in-human phase I study of the oral inhibitor of indoleamine 2,3-dioxygenase-1 epacadostat (INCB024360) in patients with advanced solid malignancies. Clin. Cancer Res. 2017, 23, 3269–3276. [Google Scholar] [CrossRef] [Green Version]
- Mitchell, T.C.; Hamid, O.; Smith, D.C.; Bauer, T.M.; Wasser, J.S.; Olszanski, A.J.; Luke, J.J.; Balmanoukian, A.S.; Schmidt, E.V.; Zhao, Y.; et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: Phase I results from a multicenter, open-label phase I/II trial (ECHO-202/KEYNOTE-037). J. Clin. Oncol. 2018, 36, 3223–3230. [Google Scholar] [CrossRef] [PubMed]
- Long, G.V.; Dummer, R.; Hamid, O.; Gajewski, T.F.; Caglevic, C.; Dalle, S.; Arance, A.; Carlino, M.S.; Grob, J.J.; Kim, T.M.; et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): A phase 3, randomised, double-blind study. Lancet Oncol. 2019, 20, 1083–1097. [Google Scholar] [CrossRef] [PubMed]
- Jung, K.H.; LoRusso, P.; Burris, H.; Gordon, M.; Bang, Y.J.; Hellmann, M.D.; Cervantes, A.; Ochoa de Olza, M.; Marabelle, A.; Stephen Hodi, F.; et al. Phase I study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) administered with PD-L1 inhibitor (atezolizumab) in advanced solid tumors. Clin. Cancer Res. 2019, 25, 3220–3228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prendergast, G.C.; Mondal, A.; Dey, S.; Laury-Kleintop, L.D.; Muller, A.J. Inflammatory Reprogramming with IDO1 Inhibitors: Turning Immunologically Unresponsive ‘Cold’ Tumors ‘Hot’. Trends Cancer 2018, 4, 38–58. [Google Scholar] [CrossRef] [PubMed]
- Haanen, J.B.; Cerundolo, V. NKG2A, a New Kid on the Immune Checkpoint Block. Cell 2018, 175, 1720–1722. [Google Scholar] [CrossRef] [Green Version]
- Kochan, G.; Escors, D.; Breckpot, K.; Guerrero-Setas, D. Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology 2013, 2, e26491. [Google Scholar] [CrossRef] [Green Version]
- Levy, E.M.; Bianchini, M.; Von Euw, E.M.; Barrio, M.M.; Bravo, A.I.; Furman, D.; Domenichini, E.; Macagno, C.; Pinsky, V.; Zucchini, C.; et al. Human leukocyte antigen-E protein is overexpressed in primary human colorectal cancer. Int. J. Oncol. 2008, 32, 633–641. [Google Scholar] [CrossRef] [Green Version]
- Herbst, R.S.; Majem, M.; Barlesi, F.; Carcereny, E.; Chu, Q.; Monnet, I.; Sanchez-Hernandez, A.; Dakhil, S.; Camidge, D.R.; Winzer, L.; et al. COAST: An Open-Label, Phase II, Multidrug Platform Study of Durvalumab Alone or in Combination with Oleclumab or Monalizumab in Patients with Unresectable, Stage III Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2022, 3, 3383–3393. [Google Scholar] [CrossRef]
- Yu, M.; Guo, G.; Huang, L.; Deng, L.; Chang, C.S.; Achyut, B.R.; Canning, M.; Xu, N.; Arbab, A.S.; Bollag, R.J.; et al. CD73 on cancer-associated fibroblasts enhanced by the A2B-mediated feedforward circuit enforces an immune checkpoint. Nat. Commun. 2020, 11, 515. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B. CD73: A novel target for cancer immunotherapy. Cancer Res. 2010, 70, 6407–6411. [Google Scholar] [CrossRef] [Green Version]
- Leclerc, B.G.; Charlebois, R.; Chouinard, G.; Allard, B.; Pommey, S.; Saad, F.; Stagg, J. CD73 expression is an independent prognostic factor in prostate cancer. Clin. Cancer Res. 2016, 22, 158–166. [Google Scholar] [CrossRef] [PubMed]
- Turcotte, M.; Spring, K.; Pommey, S.; Chouinard, G.; Cousineau, I.; George, J.; Chen, G.M.; Gendoo, D.M.A.; Haibe-Kains, B.; Karn, T.; et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015, 75, 4494–4503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allard, B.; Pommey, S.; Smyth, M.J.; Stagg, J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin. Cancer Res. 2013, 19, 5626–5635. [Google Scholar] [CrossRef] [Green Version]
- Hay, C.M.; Sult, E.; Huang, Q.; Mulgrew, K.; Fuhrmann, S.R.; McGlinchey, K.A.; Hammond, S.A.; Rothstein, R.; Rios-Doria, J.; Poon, E.; et al. Targeting CD73 in the tumor microenvironment with MEDI9447. Oncoimmunology 2016, 5, e1208875. [Google Scholar] [CrossRef] [PubMed]
- Martinez-Marti, A.; Majem, M.; Barlesi, F.; Carcereny Costa, E.; Chu, Q.; Monnet, I.; Sanchez, A.; Dahkil, S.; Camidge, D.R.; He, P.; et al. COAST: An Open-Label, Randomized, Phase 2 Platform Study of Durvalumab Alone or in Combination with Novel Agents in Patients with Locally Advanced, Unresectable, Stage III NSCLC. Ann. Oncol. 2021, 32, S1283–S1346. [Google Scholar] [CrossRef]
- Cascone, T.; García-Campelo, R.; Spicer, J.; Weder, W.; Daniel, D.; Spigel, D.; Hussein, M.; Mazieres, J.; Oliveira, J.; Yau, E.; et al. Abstract CT011: NeoCOAST: Open-label, randomized, phase 2, multidrug platform study of neoadjuvant durvalumab alone or combined with novel agents in patients (pts) with resectable, early-stage non-small-cell lung cancer (NSCLC). Cancer Res. 2022, 82, CT011. [Google Scholar] [CrossRef]
- Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am. Soc. Clin. Oncol. Educ. B. 2019, 39, 147–164. [Google Scholar] [CrossRef]
- Nicoś, M.; Krawczyk, P.; Crosetto, N.; Milanowski, J. The Role of Intratumor Heterogeneity in the Response of Metastatic Non-Small Cell Lung Cancer to Immune Checkpoint Inhibitors. Front. Oncol. 2020, 10, 2773. [Google Scholar] [CrossRef]
- Saito, Y.; Horiuchi, S.; Morooka, H.; Ibi, T.; Takahashi, N.; Ikeya, T.; Shimizu, Y.; Hoshi, E. Inter-tumor heterogeneity of PD-L1 expression in non-small cell lung cancer. J. Thorac. Dis. 2019, 11, 4982–4991. [Google Scholar] [CrossRef]
- Suda, K.; Mitsudomi, T. Inter-tumor heterogeneity of PD-L1 status: Is it important in clinical decision making? J. Thorac. Dis. 2020, 12, 1770–1775. [Google Scholar] [CrossRef]
- Gagné, A.; Wang, E.; Bastien, N.; Orain, M.; Desmeules, P.; Pagé, S.; Trahan, S.; Couture, C.; Joubert, D.; Joubert, P. Impact of Specimen Characteristics on PD-L1 Testing in Non–Small Cell Lung Cancer: Validation of the IASLC PD-L1 Testing Recommendations. J. Thorac. Oncol. 2019, 14, 2062–2070. [Google Scholar] [CrossRef] [PubMed]
- Sha, D.; Jin, Z.; Budczies, J.; Kluck, K.; Stenzinger, A.; Sinicrope, F.A. Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov. 2020, 10, 1808–1825. [Google Scholar] [CrossRef] [PubMed]
- Roviello, G.; Bersanelli, M.; Catalano, M. The latest developments in biomarkers predicting response to immunotherapy. Immunotherapy 2022, 14, 1085–1088. [Google Scholar] [CrossRef] [PubMed]
- Robertson, J.; Salm, M.; Dangl, M. Adoptive cell therapy with tumour-infiltrating lymphocytes: The emerging importance of clonal neoantigen targets for next-generation products in non-small cell lung cancer. Immuno-Oncology Technol. 2019, 3, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sobhani, N.; Scaggiante, B.; Morris, R.; Chai, D.; Catalano, M.; Tardiel-Cyril, D.R.; Neeli, P.; Roviello, G.; Mondani, G.; Li, Y. Therapeutic cancer vaccines: From biological mechanisms and engineering to ongoing clinical trials. Cancer Treat. Rev. 2022, 109, 102429. [Google Scholar] [CrossRef] [PubMed]
- Bispecific Antibodies and BiTEs Under Continued Evaluation in Lung Cancer. Available online: https://www.onclive.com/view/bispecific-antibodies-and-bites-under-continued-evaluation-in-lung-cancer (accessed on 16 May 2022).
- Galon, J.; Fridman, W.H.; Pages, F. The adaptive immunologic microenvironment in colorectal cancer: A novel perspective. Cancer Res. 2007, 67, 1883–1886. [Google Scholar] [CrossRef]
Protein | Gene Location (Human) | Expression Cell | Ligand and Presenting Cell | Immune Effect | Ref. | |
---|---|---|---|---|---|---|
LAG-3 | Chromosome 12p13.32 | T cells NK B cells DCs | MHC II | APC | Reduction in T helper 1 (Th1) cell activation, proliferation, and cytokine secretion | [70,71,72,73,74,75,76,77,78,79] |
Galactine-3 | Soluble | |||||
LSECtin | Tumor cells | |||||
FGL-1 | Soluble | |||||
TIGIT | Chromosome 3q13.31 | T cells NK | CD 155 CD 112 Nec3 | APC | Suppression of T cell and NK activation | [93,94,95,96,97,98] |
Fap2 | Fusobacterium nucleatum | |||||
TIM-3 | Chromosome 5q33.2 | T cells B cells DCs NK Monocyte Macrophages | Galectine-9 | Soluble | Negative regulation of T cell responses | |
HMGB1 | Soluble | [28,110,111,112,113,114,115,116,117] | ||||
Ceacam | Unknown | |||||
PtdSer | Unknown | |||||
VISTA | Chromosome 10q22.1 | DCs Macrophage Monocytes T cells Tumor cells | VISTA-L | APC | Suppression of T cell activation, proliferation, and cytokine production Activation of anti-cancer immune responses. | [121,122,123,124,125,126,127] |
ICOS | Chromosome 2q33.2 | Activated memory T cells | ICOS-L | APC Somatic cells | Suppression of antitumor T cell response | [135,136,137,138,167] |
B7-H3 | Chromosome 15q24.1 | APC NK T cells Monocytes Tumor cells | TLT2 (receptor) | Unknown | Co-stimulation of T cells activations Inhibition of CD4+ and CD8+ T cells | [145,146,147,148,149,150,151,152] |
BTLA | Chromosome 3q13.2 | B cells T cells DCs Macrophages | HVEM | Unknown | Inhibition of T cell proliferation and cytokine production | [158,159,160,161,162,163,164,165] |
Immune Check Point | No. of Trial | Status | Phase | Estimated Enrollment | Tumor Types | Setting | Investigated Agents | Primary End Points |
---|---|---|---|---|---|---|---|---|
LAG-3 | ||||||||
NCT03625323 | Active not recruiting | II | 183 | NSCLC, HNSCC | Untreated, unresectable or metastatic | Eftilagimod Alpha (anti-LAG3 mAb) + Pembrolizumab | ORR | |
NCT04618393 | Recruiting | I/II | 43 | Solid Tumors | Advanced | EMB-02 (anti-PD-1/LAG-3 bispecific mAb) | AEs and, AEs, ORR | |
NCT03459222 | Recruiting | I/II | 184 | Malignant Tumors | Advanced | Relatlimab (anti-LAG-3 mAb) + Nivolumab and BMS-986205 (IDO1 inhibitor) or Ipilimumab | AEs and AEs, DLT, ORR, DCR, mDOR | |
NCT04140500 | Recruiting | I | 320 | Solid Tumors, Melanoma, NSCLC, ESCC | Advanced and/or metastatic | RO7247669 (anti-PD-1/LAG3 bispecific Ab) | DLTs, ORR, DCR, DOR, PFS | |
NCT04374877 | Recruiting | I/Ib | 220 | RCC, HCC, NSCLC | Advanced | SRF388 (anti-IL-27 mAb) | DLT, ORR, AEs, ORR | |
NCT03625323 | Active, not ecruiting | II | 189 | NSCLC, HNSCC | Untreated unresectable or metastatic | Eftilagimod Alpha (soluble LAG-3 fusion protein) + Pembrolizumab | ORR | |
NCT03250832 | Active, not recruiting | I | 111 | Solid Tumors | Advanced | TSR-033 (anti-LAG-3 mAb) ± anti-PD-1 | Safety, ORR | |
TIGIT | ||||||||
NCT04995523 | Recruiting | II | 147 | NSCLC | Advanced or metastatic | AZD2936 (anti-TIGIT/PD-1 bispecific Ab) | AEs, ORR | |
NCT04952597 | Recruiting | II | 120 | SCLC | Untreated limited stage | Ociperlimab (anti-TIGIT) + Tislelizumab + CT | PFS | |
NCT04746924 | Recruiting | III | 605 | NSCLC | Untreated locally advanced, unresectable, or metastatic | Ociperlimab (anti-TIGIT) + Tislelizumab | PFS, OS | |
NCT04294810 | Recruiting | III | 560 | NSCLC | Untreated locally advanced, unresectable, or metastatic | Tiragolumab (anti-TIGIT) + Atezolizumab | PFS, OS | |
NCT04791839 | Recruiting | II | 30 | NSCLC | Untreated advanced | Zimberelimab (anti-PD-1) + Domvanalimab (anti-TIGIT) + Etrumadenant (anti-A2R) | ORR, | |
NCT04262856 | Recruiting | II | 150 | NSCLC | Metastatic | Zimberelimab (anti-PD-1) ± Domvanalimab (anti-TIGIT) ± Etrumadenant (anti-A2R) | ORR, PFS | |
NCT04761198 | Recruiting | I/II | 125 | Solid tumors | Locally advanced or metastatic | Etigilimab (anti-TIGIT) + Nivolumab | ORR | |
NCT04736173 | Recruiting | III | 625 | NSCLC | Locally advanced or metastatic | Zimberelimab (anti-PD-1) ± Domvanalimab (anti-TIGIT) | OS, PFS | |
NCT03739710 | Recruiting | II | 140 | NSCLC | Relapsed/refractory advanced | Feladilimab, Ipilimumab (anti-CTLA-4), GSK4428859A, Dostarlimab (anti-PD-1) (various combination versus SoC) | AEs, DLT, OS | |
NCT04995523 | Recruiting | I/II | 147 | NSCLC | Advanced, or metastatic | AZD2936 (anti-TIGIT/anti-PD-1 bispecific Ab) | AEs, ORR | |
NCT04746924 | recr | 3 | 605 | NSCLC | Untreated PD-L1-selected, and locally advanced, unresectable, or metastatic | BGB-A1217 (anti-TIGIT Ab) + Tislelizumab | PFS, OS | |
NCT04585815 | Recruiting | I and II | 375 | NSCLC | Advanced or metastatic | Sasanlimab (anti-PD-1) + Encorafenib and Binimetinib or Axitinib and SEA-TGT (anti-TIGIT) | DLT, ORR | |
NCT04866017 | Recruiting | III | 900 | NSCLC | Locally advanced, unresectable | Tislelizumab (anti-PD-1) ± Ociperlimab (anti-TIGIT) + CRT | PFS, CRR | |
NCT04294810 | Recruiting | III | 635 | NSCLC | Untreated locally advanced, unresectable, or metastatic PD-L1-selected | Tiragolumab + Atezolizumab | PFS, OS | |
NCT05102214 | Recruiting | I/II | 150 | Solid Tumors NSCLC | Locally Advanced or Metastatic | HLX301 (PDL1/TIGIT bispecific Ab) | ||
NCT04791839 | Recruiting | II | 30 | NSCLC | Previously treated | Zimberelimab + Domvanalimab (anti-TIGIT) and Etrumadenant | ORR, PR | |
NCT05014815 | Recruiting | 2 | 270 | NSCLC | Untreated locally advanced, unresectable, or metastatic | Ociperlimab (anti-TIGIT) and Tislelizumab + CT | PFS | |
NCT05060432 | Recruiting | I/II | 376 | Lung Cancer, Head and Neck cancer, Melanoma | Advanced | EOS-448 (anti-TIGIT) + SoC or Investigational Therapies | DLT, AE, ORR, RP2D | |
NCT04952597 | Active, not recruiting | II | 126 | SCLC | Limited Stage | Ociperlimab + Tislelizumab + CRT | PFS | |
NCT03563716 | Active, not recruiting | II | 135 | NSCLC | Chemotherapy-naïve patients with locally advanced or metastatic | Tiragolumab, (anti-TIGIT) + Atezolizumab | ORR, PFS | |
NCT04256421 | Active, not recruiting | III | 490 | SCLC | Untreated Extensive Stage | Atezolizumab + Carboplatin and Etoposide ± Tiragolumab (anti TIGIT) | PFS, OS | |
NCT04672356 | Active, not recruiting | 1 | 20 | Lung Cancer | Advanced | IBI939 (anti-TIGIT Ab) + Sintilimab | AE, RP2D | |
NCT04672369 | Active, not recruiting | 1 | 42 | Lung Cancer | Advanced | IBI939 (anti-TIGIT Ab) + Sintilimab | ORR | |
TIM-3 | ||||||||
NCT04931654 | Recruiting | II | 81 | NSCLC | Advanced or metastatic | AZD7789 (PD-1/TIM-3 bispecific Ab) | AE, DLT, ORR | |
NCT03744468 | Recruiting | II | 162 | HNSCC, NSCLC, RCC | Advanced | BGB-A425 (anti-TIM-3) and LBL-007 (anti-LAG-3) + tislelizumab | MTD, ORR | |
NCT03708328 | Active, not recruiting | I | 134 | Solid Tumors, Melanoma, NSCLC, SCLC, ESCC | Advanced and/or Metastatic | RO7121661 (anti-PD-1/TIM-3 bispecific Ab) | Dose Escalation, ORR, DCR, DOR, PFS | |
NCT02817633 | Recruiting | I | 396 | Solid Tumors | Advanced | TSR-022, (anti-TIM-3 Ab) | DLTs, AEs, ORR | |
B7-H3 | ||||||||
NCT04432649 | Recruiting | I/II | 100 | Solid Tumor | Refractory and/or recurrent | 4SCAR-276 (anti-B7-H3) | AE | |
NCT05280470 | Recruiting | II | 80 | SCLC | Pretreated Extensive Stage | DS-7300a (anti-B7-H3 ADC) | ORR | |
NCT03729596 | Recruiting | I/II | 182 | Solid Tumor, SCCHN, TNBC, Melanoma, mCRPC, NSCLC | Advanced | MGC018 (anti-B7-H ADC) ± MGA012 (anti-PD-1) | AE and SAE, DLT | |
VISTA | NCT05082610 | Recruiting | I | 240 | Solid Tumor, NSCLC, TNBC | Advanced | HMBD-002-V4C26 (anti-VISTA) ± Pembrolizumab | DLT, Safety |
BTLA | ||||||||
NCT04137900 | Recruiting | I | 499 | Solid Tumors | Advanced | TAB004 (anti-BTLA) ± Toripalimab | TRAE | |
NCT03758001 | Active, not recruiting | I | 38 | Solid Tumor | Advanced | Cudarolimab (anti-OX40) + Sintilimab (anti-PD-1) | AEs | |
NCT05000684 | Recruiting | I/II | 66 | Lung Cancer | Advanced | JS004 (anti-BTLA) injection + Toripalimab | AE | |
ICOs | NCT03829501 | Recruiting | I/II | 208 | Solid Tumors | Advanced | KY1044 (anti-ICOS) ± Atezolizumab | AEs, ORR, DLTs |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Catalano, M.; Shabani, S.; Venturini, J.; Ottanelli, C.; Voltolini, L.; Roviello, G. Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors. Cancers 2022, 14, 6145. https://doi.org/10.3390/cancers14246145
Catalano M, Shabani S, Venturini J, Ottanelli C, Voltolini L, Roviello G. Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors. Cancers. 2022; 14(24):6145. https://doi.org/10.3390/cancers14246145
Chicago/Turabian StyleCatalano, Martina, Sonia Shabani, Jacopo Venturini, Carlotta Ottanelli, Luca Voltolini, and Giandomenico Roviello. 2022. "Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors" Cancers 14, no. 24: 6145. https://doi.org/10.3390/cancers14246145
APA StyleCatalano, M., Shabani, S., Venturini, J., Ottanelli, C., Voltolini, L., & Roviello, G. (2022). Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors. Cancers, 14(24), 6145. https://doi.org/10.3390/cancers14246145