239
Views
5
CrossRef citations to date
0
Altmetric
Review

Small molecules as antagonists of co-inhibitory pathways for cancer immunotherapy: a patent review (2018-2019)

, , , &
Pages 677-694 | Received 13 May 2020, Accepted 22 Jul 2020, Published online: 08 Sep 2020

References

  • Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61.
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264.
  • Kruger S, Ilmer M, Kobold S, et al. Advances in cancer immunotherapy 2019 - latest trends. J Exp Clin Cancer Res. 2019;38:268.
  • Sharma P, Allison James P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214.
  • Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–461.
  • Hoos A. Development of immuno-oncology drugs-from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov. 2016;15:235–247.
  • Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015;125:3384–3391.
  • Iwai Y, Hamanishi J, Chamoto K, et al. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci. 2017;24:26.
  • Jiao P, Zhou H, Otto M, et al. Leading neuroblastoma cells to die by multiple premeditated attacks from a multifunctionalized nanoconstruct. J Am Chem Soc. 2011;133:13918–13921.
  • Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492–499.
  • Noy R, Pollard Jeffrey W. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61.
  • Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4:336–347.
  • Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14:561–584.
  • Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8:467–477.
  • Callahan Margaret K, Postow Michael A, Wolchok Jedd D. Targeting T cell co-receptors for cancer therapy. Immunity. 2016;44:1069–1078.
  • Fontana MF, Vance RE. Two signal models in innate immunity. Immunol Rev. 2011;243:26–39.
  • Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13:227–242.
  • Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol. 2009;27:591–619.
  • Paterson AM, Vanguri VK, Sharpe AH. SnapShot: B7/CD28 costimulation. Cell. 2009;137:974–4.e1.
  • Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol. 2002;2:116–126.
  • Salomon B, Bluestone JA. Complexities of CD28/B7: CTLA-4 costimulatory pathways in autoimmunity and transplantation. Annu Rev Immunol. 2001;19:225–252.
  • Murakami N, Riella LV. Co-inhibitory pathways and their importance in immune regulation. Transplantation. 2014;98:3–14.
  • Baumeister SH, Freeman GJ, Dranoff G, et al. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol. 2016;34:539–573.
  • Gettinger S, Horn L, Jackman D, et al. Five-year follow-up of nivolumab in previously treated advanced non-small-cell lung cancer: results from the CA209-003 study. J Clin Oncol. 2018;36:1675–1684.
  • Cho JH. Immunotherapy for non-small-cell lung cancer: current status and future obstacles. Immune Netw. 2017;17:378–391.
  • Tachihara M, Negoro S, Inoue T, et al. Efficacy of anti-PD-1/PD-L1 antibodies after discontinuation due to adverse events in non-small cell lung cancer patients (HANSHIN 0316). BMC Cancer. 2018;18:946.
  • Huang M, Lou Y, Pellissier J, et al. Cost effectiveness of pembrolizumab vs. standard-of-care chemotherapy as first-line treatment for metastatic NSCLC that expresses high levels of PD-L1 in the United States. Pharmacoeconomics. 2017;35:831–844.
  • Bailly C, Vergoten G. Protein homodimer sequestration with small molecules: focus on PD-L1. Biochem Pharmacol. 2020;174:113821.
  • Geng Q, Jiao P, Jin P, et al. PD-1/PD-L1 inhibitors for immuno-oncology: from antibodies to small molecules. Curr Pharm Des. 2017;23:6033–6041.
  • Jiao P, Geng Q, Jin P, et al. Small molecules as PD-1/PD-L1 pathway modulators for cancer immunotherapy. Curr Pharm Des. 2018;24:4911–4920.
  • Tang S, Kim PS. A high-affinity human PD-1/PD-L2 complex informs avenues for small-molecule immune checkpoint drug discovery. Proc Natl Acad Sci U S A. 2019;116:24500–24506.
  • Dalvin LA, Shields CL, Orloff M, et al. Checkpoint inhibitor immune therapy: systemic indications and ophthalmic side effects. RETINA. 2018;38:1063–1078.
  • Chen T, Li Q, Liu Z, et al. Peptide-based and small synthetic molecule inhibitors on PD-1/PD-L1 pathway: A new choice for immunotherapy? Eur J Med Chem. 2019;161:378–398.
  • Udhwani T, Mukherjee S, Sharma K, et al. Design of PD-L1 inhibitors for lung cancer. Bioinformation. 2019;15:139–150.
  • Ashizawa T, Iizuka A, Tanaka E, et al. Antitumor activity of the PD-1/PD-L1 binding inhibitor BMS-202 in the humanized MHC-double knockout NOG mouse. Biomed Res. 2019;40:243–250.
  • Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. Embo J. 1992;11:3887–3895. .
  • Gordon SR, Maute RL, Dulken BW, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545:495–499.
  • Dong H, Zhu G, Tamada K, et al. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5:1365–1369. .
  • Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800.
  • Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the pd-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–1034. .
  • Wang T, Wu X, Guo C, et al. Development of inhibitors of the programmed cell death-1/programmed cell death-ligand 1 signaling pathway. J Med Chem. 2019;62:1715–1730.
  • Li K, Tian H. Development of small-molecule immune checkpoint inhibitors of PD-1/PD-L1 as a new therapeutic strategy for tumour immunotherapy. J Drug Target. 2018;27:244–256.
  • Hirano F, Kaneko K, Tamura H, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65:1089–1096.
  • Zarganes-Tzitzikas T, Konstantinidou M, Gao Y, et al. Inhibitors of programmed cell death 1 (PD-1): A patent review (2010-2015). Expert Opin Ther Pat. 2016;26:973–977.
  • Collin M. Immune checkpoint inhibitors: A patent review (2010-2015). Expert Opin Ther Pat. 2016;26:555–564.
  • Hamanishi J, Mandai M, Matsumura N, et al. PD-1/PD-L1 blockade in cancer treatment: perspectives and issues. Int J Clin Oncol. 2016;21:462–473.
  • Adams JL, Smothers J, Srinivasan R, et al. Big opportunities for small molecules in immuno-oncology. Nat Rev Drug Discov. 2015;14:603–622.
  • Weinmann H. Cancer immunotherapy: selected targets and small-molecule modulators. ChemMedChem. 2016;11:450–466.
  • Zhan M, Hu X, Liu X, et al. From monoclonal antibodies to small molecules: the development of inhibitors targeting the PD-1/PD-L1 pathway. Drug Discov Today. 2016;21:1027–1036.
  • Kerr WG, Chisholm JD. The next generation of immunotherapy for cancer: small molecules could make big waves. J Immunol. 2019;202:11–19.
  • Kopalli SR, Kang T, Lee K, et al. Novel small molecule inhibitors of programmed cell death (PD)-1, and its ligand, PD-L1 in cancer immunotherapy: A review update of patent literature. Recent Pat Anticancer Drug Discov. 2019;14(2):100–112. .
  • Shaabani S, Huizinga HPS, Butera R, et al. A patent review on PD-1/PD-L1 antagonists: small molecules, peptides, and macrocycles (2015-2018). Expert Opin Ther Pat. 2018;28:665–678. .
  • Cheng B, Yuan W, Su J, et al. Recent advances in small molecule based cancer immunotherapy. Eur J Med Chem. 2018;157:582–598.
  • Skalniak L, Kocik J, Guzik K, et al. Small molecules, peptides and antibodies-the comparison of PD-1/PD-L1 blocking potential in an in vitro immune checkpoint blockade assay. ESMO Open. 2018;3:A400–A400.
  • Guzik K, Zak KM, Grudnik P, et al. Small-molecule inhibitors of the programmed cell death-1/programmed death-ligand 1 (PD-1/PD-L1) interaction via transiently induced protein states and dimerization of PD-L1. J Med Chem. 2017;60:5857–5867.
  • Zhu H, Li Y. Small-molecule targets in tumor immunotherapy. Nat Prod Bioprospect. 2018;8(4):297–301.
  • Sharpe AH, Butte MJ, Oyama S. Modulators of immunoinhibitory receptor PD-1, and methods of use thereof. President and Fellows of Harvard College; 2011. (WO2011082400 A1).
  • Chupak LS, Ding M, Martin SW, et al. Compounds useful as immunomodulators. Bristol-Myers Squibb Company; 2015. (WO2015160641 A1).
  • Chupak LS, Zheng X Compounds useful as immunomodulators. Bristol-Myers Squibb Company; 2015. (WO2015034820 A1).
  • Yeung K, GrantYong KA, Zhu J, et al. 1,3-Dihydroxy-phenyl derivatives useful as immunomodulators. Bristol-Myers Squibb Company; 2018. (WO2018009505 A1).
  • Yeung K, GrantYong KA, Zhu J, et al. Bristol-Myers Squibb Company. Biaryl compounds useful as immunomodulators. WO2018044963 A1. 2018.
  • Yeung K, GrantYong KA, Sun L, et al. Compounds useful as immunomodulators. Bristol-Myers Squibb Company; 2018. (WO2018118848 A1).
  • Wang Y, Xu Z, Wu T, et al. Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof. Guangzhou Maxinovel Pharmaceuticals; 2018. (WO2018006795 A1).
  • Wang Y, Zhang N, Wang F, et al. Orally active small molecule PD-L1 inhibitor demonstrating similar efficacy as Durvalumab in human knock-in MC38 model. Cancer Res. 2019;79:LB–018.
  • Wang M. Symmetric or semi-symmetric compounds useful as immunomodulators. Arising International LLC; 2018. (WO2018026971 A1).
  • Webber SE, Almassy R, Immune checkpoint inhibitors, compositions and methods thereof. Polaris Pharmaceuticals; 2018. (WO20180045142 A1).
  • Sasikumar PGN, Ramachandra M, Naremaddepalli SSS Limited. 1,2,4-Oxadiazole derivatives as immunomodulators. Aurigene Discovery Technologies; 2015. (WO2015033299 A1).
  • Sasikumar PGN, Prasad A, Naremaddepalli SSS, et al. Cyclic substituted-1,2,4-oxadiazole compounds as immunomodulators. Aurigene Discovery Technologies Limited; 2018. (WO2018051254 A1).
  • Sasikumar PGN, Prasad A, Naremaddepalli SSS, et al. Aurigene Discovery Technologies Limited. Cyclic substituted-1,3,4-oxadiazole and thiadiazole compounds as immunomodulators. WO2018051255 A1. 2018.
  • Liu PC, Volgina A, Wynn R, et al. Tetrahydro imidazo[4,5-c]pyridine derivatives as PD-L1 internalization inducers. Incyte Corporation; 2018. (WO2018119224 A1).
  • Wu L, Qian D-Q, Lajkiewicz N, et al. Heterocyclic compounds derivatives as pd-l1 internalization inducers. Incyte Corporation; 2018. (WO2018119263 A1).
  • Aktoudianakis E, Appleby T, Cho A, et al. PD-1/PD-L1 inhibitors. Gilead Science; 2018. (WO2018195321 A1).
  • Aktoudianakis E, Cho A, Du Z, et al. PD-1/PD-L1 inhibitors. Gilead Science; 2018. (WO2019160882 A1).
  • Aktoudianakis E, Cho A, Du Z, et al. Gilead Science. PD-1/PD-L1 inhibitors. WO2019160882 A1. 2018.
  • Dömling A. 3-Cyanothiophene derivatives as inhibitors of the PD-1/PD-L1 protein/protein interaction. Rijksuniversiteit Groningen; 2019. (WO2019008152 A1).
  • Dömling A. Rijksuniversiteit Groningen. 3-(Azolylmethoxy)biphenyl derivatives as inhibitors of the PD-1/PD-L1 protein/protein interaction. WO2019008154 A1. 2019.
  • Dömling A. Inhibitors of the PD-1/PD-L1 protein/protein interaction.Rijksuniversiteit Groningen; 2019. (WO2019008156 A1).
  • Venkateshappa C, Duraiswamy AJ, Putta RKVP, et al. Pyrimidine derivatives as inhibitors of PD-1/PD-L1 activation. Jubilant Biosys Limited; 2019. (WO2019087214 A1).
  • Venkateshappa C, Jeyaraj DA, Pendyala M, et al. Bicyclic compounds as inhibitors of PD-1/PD-L1 interaction/activation. Jubilant Biosys Limited; 2019. (WO2019175897 A1).
  • Yang F, Zhang Y, Ying H, et al. Biaryl derivative, preparation method thereof and pharmaceutical application thereof. Abbisko Therapeutics; 2019. (WO2019149183 A1).
  • Lange C, Punna S, Singh R, et al. Indane-amines as PD-L1 antagonists. Chemocentryx; 2019. (WO2019165043 A2).
  • Wang L, Rubinstein R, Lines JL, et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208:577–592.
  • Johnston RJ, Su LJ, Pinckney J, et al. VISTA is an acidic pH-selective ligand for PSGL-1. Nature. 2019;574:565–570.
  • Xu W, Hieu T, Malarkannan S, et al. 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.
  • Flies DB, Wang S, Xu H, et al. 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.
  • Lines JL, Pantazi E, Mak J, et al. VISTA is an immune checkpoint molecule for human T cells. Cancer Res. 2014;74:1924–1932.
  • Li N, Xu W, Yuan Y, et al. Immune-checkpoint protein VISTA critically regulates the IL-23/IL-17 inflammatory axis. Sci Rep. 2017;7:1485.
  • Le Mercier I, Chen W, Lines JL, et al. VISTA regulates the development of protective antitumor immunity. Cancer Res. 2014;74:1933–1944.
  • Flies DB, Higuchi T, Chen L. Mechanistic assessment of PD-1H coinhibitory receptor-induced T cell tolerance to allogeneic antigens. J Immunol. 2015;194:5294–5304.
  • Sasikumar PGN, Ramachandra M, Naremaddepalli SSS Dual inhibitors of VISTA and PD-1 pathways. Aurigene Discovery Technologies Limited; 2018. (WO2018073754 A1).
  • Musielak B, Kocik J, Skalniak L, et al. CA-170 – a potent small-molecule PD-L1 inhibitor or not? Molecules. 2019;24:2804.
  • Guzik K, Tomala M, Muszak D, et al. Development of the inhibitors that target the pd-1/pd-l1 interaction-a brief look at progress on small molecules, peptides and macrocycles. Molecules. 2019;24:2071.
  • Picardo SL, Doi J, Hansen AR. Structure and optimization of checkpoint inhibitors. Cancers (Basel). 2019;12:38.
  • Zauderer M, Brody J, Marron T, et al. Phase 1 study of CA-170: first-in-class small molecule targeting VISTA/PD-L1 in patients with malignant pleural mesothelioma. J Thorac Oncol. 2019;14:S757–S758.
  • Radhakrishnan V, Banavali S, Gupta S, et al. Excellent CBR and prolonged PFS in non-squamous NSCLC with oral CA-170, an inhibitor of VISTA and PD-L1. Ann Oncol. 2019;30:v494.
  • He Y, Cao J, Zhao C, et al. TIM-3, a promising target for cancer immunotherapy. Onco Targets Ther. 2018;11:7005–7009.
  • Monney L, Sabatos CA, Gaglia JL, et al. Th1-specific cell surface protein TIM-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415:536–541.
  • Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6:1245–1252.
  • Meyers JH, Sabatos CA, Chakravarti S, et al. The TIM gene family regulates autoimmune and allergic diseases. Trends Mol Med. 2005;11:362–369.
  • Hastings WD, Anderson DE, Kassam N, et al. TIM-3 is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. Eur J Immunol. 2009;39:2492–2501.
  • Zhu C, Sakuishi K, Xiao S, et al. An IL-27/NFIL3 signalling axis drives Tim-3 and IL-10 expression and T-cell dysfunction. Nat Commun. 2015;6:6072.
  • Jones RB, Ndhlovu LC, Barbour JD, et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J Exp Med. 2008;205:2763–2779.
  • Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486–499.
  • Fourcade J, Sun Z, Benallaoua M, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207:2175–2186.
  • Gao X, Zhu Y, Li G, et al. TIM-3 expression characterizes regulatory T cells in tumor tissues and is associated with lung cancer progression. PLoS One. 2012;7:e30676.
  • Yan W, Liu X, Ma H, et al. Tim-3 fosters HCC development by enhancing TGF-beta-mediated alternative activation of macrophages. Gut. 2015;64:1593–1604.
  • Joller N, Kuchroo VK. TIM-3, LAG-3, and TIGIT. Curr Top Microbiol Immunol. 2017;410:127–156.
  • Du W, Yang M, Turner A, et al. TIM-3 as a target for cancer immunotherapy and mechanisms of action. Int J Mol Sci. 2017;18:645.
  • Das M, Zhu C, Kuchroo VK. TIM-3 and its role in regulating anti-tumor immunity. Immunol Rev. 2017;276:97–111.
  • Meggyes M, Lajko A, Palkovics T, et al. Feto-maternal immune regulation by TIM-3/galectin-9 pathway and PD-1 molecule in mice at day 14.5 of pregnancy. Placenta. 2015;36:1153–1160.
  • Yoneda A, Jinushi M. T cell immunoglobulin domain and mucin domain-3 as an emerging target for immunotherapy in cancer management. Immunotargets Ther. 2013;2:135–141.
  • Xu Y, Jiang X, Huang Y. T-cell immunoglobulin and mucin-domain containing-3 in malignant cancers. Transl Cancer Res. 2017;6:613–619.
  • Kane LP. T cell Ig and mucin domain proteins and immunity. J Immunol. 2010;184:2743–2749.
  • Su EW, Lin JY, Kane LP. TIM-1 and TIM-3 proteins in immune regulation. Cytokine. 2008;44:9–13.
  • Sasikumar PGN, Ramachandra M, Naremaddepalli SSS, et al. Dual inhibitors of TIM-3 and PD-1 pathways. Aurigene Discovery Technologies Limited; 2019. (WO2019087087 A1).
  • Sasikumar PGN, Ramachandra M, RAMACHANDRA RK, et al. Conjoint therapies for immunomodulation. Aurigene Discovery Technologies Limited; 2019. WO2019087092 A1.
  • Solomon BL, Garrido-Laguna I. TIGIT: A novel immunotherapy target moving from bench to bedside. Cancer Immunol Immunother. 2018;67:1659–1667.
  • Harjunpää H, Guillerey C. TIGIT as an emerging immune checkpoint. Clin Exp Immunol. 2019;200:108–119.
  • Sun H, Sun C. The rise of NK cell checkpoints as promising therapeutic targets in cancer immunotherapy. Front Immunol. 2019;10:2354.
  • Yu X, Harden K, LC G, 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.
  • Chauvin JM, 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.
  • Kong Y, Zhu L, Schell TD, et al. T-cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T-cell exhaustion and poor clinical outcome in AML patients. Clin Cancer Res. 2016;22:3057–3066.
  • 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.
  • Kučan Brlić P, Lenac Roviš T, Cinamon G, et al. Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell Mol Immunol. 2019;16:40–52.
  • Kurtulus S, Sakuishi K, Ngiow SF, et al. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest. 2015;125:4053–4062.
  • Sasikumar PGN, Ramachandra M, Naremaddepalli SSS, et al. Aurigene Discovery Technologies Limited. Method of modulating TIGIT and PD-1 signaling pathways using 1,2,4-oxadiazole compounds. WO2019175799 A2. 2019.
  • Lu S, He X, Ni D, et al. Allosteric modulator discovery: from serendipity to structure-based design. J Med Chem. 2019;62:6405–6421.

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.