Journal of Pharmaceutical Care & Health Systems

Journal of Pharmaceutical Care & Health Systems
Open Access

ISSN: 2376-0419

+44 1300 500008

Editorial - (2014) Volume 1, Issue 4

Do We Need Small Molecule Inhibitors for the Immune Checkpoints?

Khaled Barakat*
Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
*Corresponding Author: Khaled Barakat, Research Assistant Professor, University of Alberta, Faculty Of Pharmacy And Pharmaceutical Sciences, 02-20G Katz Centre, Edmonton, AB, T6G 2E1, Canada

T lymphocytes preserve the immunological balance between defending against cancer and preventing continual activated immune responses. The balance between the two extremes requires an intricate network of protein-protein interactions (PPIs) that can either inhibit T cell-mediated immune responses targeted against self-antigens or stimulate them to defend against cancer [1]. While T cells’ specificity against cancer is determined by the interaction between the T-cell receptor complex (TCR) and antigenic peptides bound in surface major histocompatibility complex (MHC) molecules, the full activation of T cells requires a second signal obtained by the binding of the co-receptor CD28 on T cells to CD80/86molecules on activated antigen presenting cells (APCs). Once mobilized, T cells also express other receptors that inhibit their proliferation and cytokine production. Among these receptors are three immune inhibitory checkpoints: Cytotoxic T Lymphocyte Antigen-4 (CTLA-4), programmed death-1 (PD-1) and T cell immunoglobulin mucin-3 (TIM-3). Tumors can evade the immune system via inhibitory checkpoints to attenuate T cells’ signaling, leading to a state of immune tolerance [2]. Blocking the immune inhibitory checkpoints pathways recently emerged as a ‘game changer’ approach in cancer immunotherapy [3-5], with antibodies directed toward PD-1, for example, being selected as ‘drug of the year’ for 2013 by Science6. These antibodies proved the concept of restoring exhausted T cells’ functions and reactivating the immune system to recognize and kill tumor cells [6,7]. More importantly, combination blockage of multiple co-inhibitory pathways has a greater efficacy by preventing accumulation of the unblocked negative co-receptor, allowing T cells to continue to survive, proliferate, and carry out effector functions within the tumor [8-16]. However, despite their outstanding success, they still have numerous disadvantages. These agents are monoclonal antibodies and are very expensive to manufacture and administer, making them financially inaccessible to many. For example, under current market conditions, the treatment cost per quality-adjusted life year (QALY) for a patient with metastatic melanoma exceeds $500 000 (USD). These antibodies require bolus intravenous injections every 3 weeks, are administered in high dose and have a long half-life. They cannot be quickly withdrawn from the body if adverse events occur (such as autoimmunity) [17,18]. For example, treatment with antibodies targeting CTLA-4 results in life-threatening autoimmune colitis in 5-10% of patients46 and treatment-related fatalities, while rare, continue to be reported. These limitations suggest that the full potential of the immune checkpoint blockade has yet to be fulfilled and raise an urgent need to explore new modalities that can target the same pathways, but be safer and more effective.

Small molecules can provide this safe therapeutic alternative. They can avoid the problems associated with antibodies while retaining their functionality. Small molecules can provide increased oral bioavailability, increased bio-efficiency and shortened half-life activity for a more controllable treatment, particularly, in the case of autoimmune or other adverse events [17,18]. Small molecules can also offer greater distribution within the tumor tissue, can cross the bloodbrain- barrier to treat brain tumors, have higher stability at ambient temperature facilitating purification during production.

Despite all these advantages over current immune checkpoint’s antibodies, very limited efforts have been directed to development of small molecules toward these targets. This is mainly due to the limited structural information on these proteins. For example, current PD-1 crystal structures describe PPIs of mouse PD-1 and human PD-L1 [19] or with mouse PD-L2 [20]. Similarly, there is only one structure for the mouse TIM-3 [21]. Lacking the detailed human protein-protein interactions is a barrier to rationally design small molecule inhibitors for these targets. With the current shortage in understanding these interactions experimentally, Molecular modelling [22-29] can come into play an important role to develop these models and use them to develop small molecules for the immune checkpoints. The last decade has witnessed many successful examples of designing smallmolecule modulators for many protein-protein interactions, including orthosteric inhibitors, allosteric regulators, and interfacial binders. Recent examples include discovering small molecule inhibitors for menin and mixed lineage leukemia (MLL) interactions [30]. Some of these molecules can inhibit the menin–MLL interaction at a nanomolar concentration [31]. A more challenging example involves small molecule blockers for the shallow interaction between Interleukin-2 (IL-2) and the IL-2 receptor (IL-2Rα) [32,33]. The lead compound, SP4206, traps IL-2 in an unusual conformation and binds with 60nm concentration [34]. Other successful stories include, the discovery of small molecule inhibitors for the EWS-FLI1/RNA helicase [35], the Ras/SOS1 interaction, the xIAP/caspase interaction [36,37], BRaf/CRaf heterodimerization and activation [38,39], urokinasetype plasminogen activator (uPA), urokinase receptor (uPAR) [40], interaction of Interferon-α with Interferon-α receptors (IFNAR2) [41], interaction of ERCC1 with XPF [42], interaction of ERCC1 with XPA [43,44] interaction of p53 with MDM245 and interaction of p53 with MDM4 [45]. All these successful stories call for a similar application of the same methods toward designing small molecules abrogators for the immune checkpoints’ protein-protein interactions to provide a safer and more effective alternative for current antibodies (work in progress) [46].

References

  1. Greenwald RJ, Freeman GJ, Sharpe AH (2005) The B7 family revisitedAnnu Rev Immunol 23: 515-548.
  2. Freeman GJ, Wherry EJ, Ahmed R, Sharpe AH (2006) Reinvigorating exhausted HIV-specific T cells via PD-1-PD-1 ligand blockadeJ Exp Med 203: 2223-2227.
  3. Merelli B, Massi D, Cattaneo L, Mandala M (2014) Targeting the PD1/PD-L1 axis in melanoma: biological rationale, clinical challenges and opportunities. Critical reviews in oncology/hematology 89: 140-165.
  4. Dömling A, Holak TA (2014) Programmed death-1: therapeutic success after more than 100 years of cancer immunotherapyAngew Chem Int Ed Engl 53: 2286-2288.
  5. Intlekofer AM, Thompson CB (2013) At the bench: preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy. Journal of leukocyte biology 94: 25-39.
  6. Couzin-Frankel J (2013) Breakthrough of the year 2013. Cancer immunotherapyScience 342: 1432-1433.
  7. Zhang M (2012) A new approach to simultaneously quantify both TCR alpha- and beta-chain diversity after adoptive immunotherapy. Clinical cancer research: an official journal of the American Association for Cancer Research 18: 4733-4742.
  8. Baghdadi M, Nagao H, Yoshiyama H, Akiba H, Yagita H, et al. (2013) Combined blockade of TIM-3 and TIM-4 augments cancer vaccine efficacy against established melanomasCancer Immunol Immunother 62: 629-637.
  9. Fong L, Kwek SS, O'Brien S, Kavanagh B, McNeel DG, et al. (2009) Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSFCancer Res 69: 609-615.
  10. Hurwitz AA, Foster BA, Kwon ED, Truong T, Choi EM, et al. (2000) Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockadeCancer Res 60: 2444-2448.
  11. Youlin K, Li Z, Xiaodong W, Xiuheng L, Hengchen Z (2012) Combination immunotherapy with 4-1BBL and CTLA-4 blockade for the treatment of prostate cancerClin Dev Immunol 2012: 439235.
  12. Perez-Gracia JL, Labiano S1, Rodriguez-Ruiz ME1, Sanmamed MF1, Melero I2 (2014) Orchestrating immune check-point blockade for cancer immunotherapy in combinationsCurr Opin Immunol 27: 89-97.
  13. Pilon-Thomas S, Mackay A, Vohra N, Mulé JJ (2010) Blockade of programmed death ligand 1 enhances the therapeutic efficacy of combination immunotherapy against melanomaJ Immunol 184: 3442-3449.
  14. Quezada SA, Peggs KS, Curran MA, Allison JP (2006) CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cellsJ Clin Invest 116: 1935-1945.
  15. Riley JL (2013) Combination checkpoint blockade--taking melanoma immunotherapy to the next levelN Engl J Med 369: 187-189.
  16. Youlin K, Li Z, Xiaodong W, Xiuheng L, Hengchen Z (2012) Combination immunotherapy with 4-1BBL and CTLA-4 blockade for the treatment of prostate cancerClin Dev Immunol 2012: 439235.
  17. Caspi RR (2008) Immunotherapy of autoimmunity and cancer: the penalty for successNat Rev Immunol 8: 970-976.
  18. Amos SM, Duong CP, Westwood JA, Ritchie DS, Junghans RP, et al. (2011) Autoimmunity associated with immunotherapy of cancerBlood 118: 499-509.
  19. Lin DY (2008) The PD-1/PD-L1 complex resembles the antigen-binding Fv domains of antibodies and T cell receptors. Proceedings of the National Academy of Sciences of the United States of America 105: 3011-3016.
  20. Lázár-Molnár E, Yan Q, Cao E, Ramagopal U, Nathenson SG, et al. (2008) Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2Proc Natl Acad Sci U S A 105: 10483-10488.
  21. DeKruyff RH, Bu X, Ballesteros A, Santiago C, Chim YL, et al. (2010) T cell/transmembrane, Ig, and mucin-3 allelic variants differentially recognize phosphatidylserine and mediate phagocytosis of apoptotic cellsJ Immunol 184: 1918-1930.
  22. Barakat K, Issack BB, Stepanova M, Tuszynski J (2011) Effects of temperature on the p53-DNA binding interactions and their dynamical behavior: comparing the wild type to the R248Q mutantPLoS One 6: e27651.
  23. Barakat K, Tuszynski J (2011) Relaxed complex scheme suggests novel inhibitors for the lyase activity of DNA polymerase betaJ Mol Graph Model 29: 702-716.
  24. Friesen DE, Barakat KH, Semenchenko V, Perez-Pineiro R, Fenske BW, et al. (2012) Discovery of small molecule inhibitors that interact with γ-tubulinChem Biol Drug Des 79: 639-652.
  25. Barakat K (2013) Hepatitis C Virus NS5A Monomer Inhibitors and Methods of Use.
  26. Barakat KH, Anwar-Mohamed A, Tuszynski JA, Robins MJ, Tyrrell DL, et al. (2014) A Refined Model of the HCV NS5A Protein Bound to Daclatasvir Explains Drug-Resistant Mutations and Activity against Divergent GenotypesJ Chem Inf Model .
  27. Barakat KH, Law J, Prunotto A, Magee WC, Evans DH, et al. (2013) Detailed computational study of the active site of the hepatitis C viral RNA polymerase to aid novel drug designJ Chem Inf Model 53: 3031-3043.
  28. Anwar-Mohamed A (2014) A human ether-a-go-go-related (hERG) ion channel atomistic model generated by long supercomputer molecular dynamics simulations and its use in predicting drug cardiotoxicity.
  29. Hu G, Wang K1, Groenendyk J1, Barakat K1, Mizianty MJ1, et al. (2014) Human structural proteome-wide characterization of Cyclosporine A targetsBioinformatics 30: 3561-3566.
  30. Grembecka J, Belcher AM, Hartley T, Cierpicki T (2010) Molecular basis of the mixed lineage leukemia-menin interaction: implications for targeting mixed lineage leukemiasJ Biol Chem 285: 40690-40698.
  31. He S, Senter TJ, Pollock J, Han C, Upadhyay SK, et al. (2014) High-affinity small-molecule inhibitors of the menin-mixed lineage leukemia (MLL) interaction closely mimic a natural protein-protein interactionJ Med Chem 57: 1543-1556.
  32. Geng X, Zhang R, Yang G, Jiang W, Xu C (2012) Interleukin-2 and autoimmune disease occurrence and therapyEur Rev Med Pharmacol Sci 16: 1462-1467.
  33. Tilley JW (1997) Identification of a Small Molecule Inhibitor of the IL-2/IL-2R? Receptor Interaction Which Binds to IL-2. J Am Chem Soc 119: 7589-7590.
  34. Wells JA, McClendon CL (2007) Reaching for high-hanging fruit in drug discovery at protein-protein interfacesNature 450: 1001-1009.
  35. May WA, Lessnick SL, Braun BS, Klemsz M, Lewis BC, et al. (1993) The Ewing's sarcoma EWS/FLI-1 fusion gene encodes a more potent transcriptional activator and is a more powerful transforming gene than FLI-1Mol Cell Biol 13: 7393-7398.
  36. Flygare JA, Beresini M, Budha N, Chan H, Chan IT, et al. (2012) Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152)J Med Chem 55: 4101-4113.
  37. Cohen F, Koehler MF, Bergeron P, Elliott LO, Flygare JA, et al. (2010) Antagonists of inhibitor of apoptosis proteins based on thiazole amide isosteresBioorg Med Chem Lett 20: 2229-2233.
  38. Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, et al. (2010) Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanomaNature 467: 596-599.
  39. Hatzivassiliou G1, Song K, Yen I, Brandhuber BJ, Anderson DJ, et al. (2010) RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growthNature 464: 431-435.
  40. Khanna M, Wang F, Jo I, Knabe WE, Wilson SM, et al. (2011) Targeting multiple conformations leads to small molecule inhibitors of the uPAR·uPA protein-protein interaction that block cancer cell invasionACS Chem Biol 6: 1232-1243.
  41. Geppert T, Bauer S, Hiss JA, Conrad E, Reutlinger M, et al. (2012) Immunosuppressive small molecule discovered by structure-based virtual screening for inhibitors of protein-protein interactionsAngew Chem Int Ed Engl 51: 258-261.
  42. Jordheim LP (2013) Small molecule inhibitors of ERCC1-XPF protein-protein interaction synergize alkylating agents in cancer cells. Molecular pharmacology 84: 12-24.
  43. Barakat KH, Jordheim LP, Perez-Pineiro R, Wishart D, Dumontet C, et al. (2012) Virtual screening and biological evaluation of inhibitors targeting the XPA-ERCC1 interactionPLoS One 7: e51329.
  44. Barakat KH (2009) Characterization of an inhibitory dynamic pharmacophore for the ERCC1–XPA interaction using a combined molecular dynamics and virtual screening approach. Journal of Molecular Graphics and Modelling 28: 113-130.
  45. Barakat K1, Mane J, Friesen D, Tuszynski J (2010) Ensemble-based virtual screening reveals dual-inhibitors for the p53-MDM2/MDMX interactionsJ Mol Graph Model 28: 555-568.
  46. Viricel C, Ahmed M, Barakat K (2014) Knowledge-Guided Flexible Docking Of PD-1 To Its Two Ligands Reveals The Reasons Behind Their Variant Affinities.
Citation: Barakat K (2014) Do We Need Small Molecule Inhibitors for the Immune Checkpoints? J Pharma Care Health Sys 1: e119.

Copyright: © 2014 Barakat K. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top