Antitumor activity of the PD-1/PD-L1 binding inhibitor BMS-202 in the humanized MHC-double knockout NOG mouse.

Recently, the first series of small molecule inhibitors of PD-1/PD-L1 were reported by Bristol-Myers Squibb (BMS), which were developed using a homogeneous time-resolved fluorescence (HTRF)-based screening investigation of the PD-1/PD-L1 interaction. Additional crystallographic and biophysical studies showed that these compounds inhibited the interaction of PD-1/PD-L1 by inducing the dimerization of PD-L1, in which each dimer binds one molecule of the stabilizer at its interface. However, the immunological mechanism of the antitumor effect of these compounds remains to be elucidated. In the present study, we focused on BMS-202 (a representative of the BMS compounds) and investigated its antitumor activity using in vitro and in vivo experiments. BMS-202 inhibited the proliferation of strongly PD-L1-positive SCC-3 cells (IC50 15 μM) and anti-CD3 antibody-activated Jurkat cells (IC50 10 μM) in vitro. Additionally, BMS-202 had no regulatory effect on the PD-1 or PD-L1 expression level on the cell surface of these cells. In an in vivo study using humanized MHC-double knockout (dKO) NOG mice, BMS-202 showed a clear antitumor effect compared with the controls; however, a direct cytotoxic effect was revealed to be involved in the antitumor mechanism, as there was no lymphocyte accumulation in the tumor site. These results suggest that the antitumor effect of BMS-202 might be partly mediated by a direct off-target cytotoxic effect in addition to the immune response-based mechanism. Also, the humanized dKO NOG mouse model used in this study was shown to be a useful tool for the screening of small molecule inhibitors of PD-1/PD-L1 binding that can inhibit tumor growth via an immune-response-mediated mechanism.

[1]  D. Y. Lin,et al.  The PD-1/PD-L1 complex resembles the antigen-binding Fv domains of antibodies and T cell receptors , 2008, Proceedings of the National Academy of Sciences.

[2]  D. Schadendorf,et al.  Improved survival with ipilimumab in patients with metastatic melanoma. , 2010, The New England journal of medicine.

[3]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[4]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

[5]  C. Horak,et al.  Nivolumab plus ipilimumab in advanced melanoma. , 2013, The New England journal of medicine.

[6]  T. Komori,et al.  Tumor associated macrophage expressing CD204 is associated with tumor aggressiveness of esophageal squamous cell carcinoma , 2013, Cancer science.

[7]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[8]  P. Sharma,et al.  The future of immune checkpoint therapy , 2015, Science.

[9]  A. Abdel-Magid Inhibitors of the PD-1/PD-L1 Pathway Can Mobilize the Immune System: An Innovative Potential Therapy for Cancer and Chronic Infections. , 2015, ACS medicinal chemistry letters.

[10]  C. Drake,et al.  Immune checkpoint blockade: a common denominator approach to cancer therapy. , 2015, Cancer cell.

[11]  G. Freeman,et al.  Combination cancer immunotherapy and new immunomodulatory targets , 2015, Nature Reviews Drug Discovery.

[12]  Lei Liu,et al.  Blocking of the PD-1/PD-L1 Interaction by a D-Peptide Antagonist for Cancer Immunotherapy. , 2015, Angewandte Chemie.

[13]  Jedd D. Wolchok,et al.  The future of cancer treatment: immunomodulation, CARs and combination immunotherapy , 2016, Nature Reviews Clinical Oncology.

[14]  K. Zak,et al.  Structural basis for small molecule targeting of the programmed death ligand 1 (PD-L1) , 2016, Oncotarget.

[15]  T. Ashizawa,et al.  Antitumor Effect of Programmed Death-1 (PD-1) Blockade in Humanized the NOG-MHC Double Knockout Mouse , 2016, Clinical Cancer Research.

[16]  Axel Hoos,et al.  Non-alcoholic steatohepatitis: emerging molecular targets and therapeutic strategies , 2016, Nature Reviews Drug Discovery.

[17]  K. Zak,et al.  Inhibitors of programmed cell death 1 (PD-1): a patent review (2010-2015) , 2016, Expert opinion on therapeutic patents.

[18]  T. Ashizawa,et al.  Immunological effects of the anti-programmed death-1 antibody on human peripheral blood mononuclear cells. , 2016, International journal of oncology.

[19]  Chenzhong Liao,et al.  From monoclonal antibodies to small molecules: the development of inhibitors targeting the PD-1/PD-L1 pathway. , 2016, Drug discovery today.

[20]  T. Holak,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. , 2017, Journal of medicinal chemistry.

[21]  L. Skalniak,et al.  Bioactive Macrocyclic Inhibitors of the PD-1/PD-L1 Immune Checkpoint. , 2017, Angewandte Chemie.

[22]  T. Ashizawa,et al.  The anti-tumor activity of the STAT3 inhibitor STX-0119 occurs via promotion of tumor-infiltrating lymphocyte accumulation in temozolomide-resistant glioblastoma cell line. , 2017, Immunology letters.

[23]  L. Skalniak,et al.  Small-molecule inhibitors of PD-1/PD-L1 immune checkpoint alleviate the PD-L1-induced exhaustion of T-cells , 2017, Oncotarget.

[24]  K. Zak,et al.  Structural Biology of the Immune Checkpoint Receptor PD-1 and Its Ligands PD-L1/PD-L2. , 2017, Structure.

[25]  P. Buchwald,et al.  Toward Small-Molecule Inhibition of Protein-Protein Interactions: General Aspects and Recent Progress in Targeting Costimulatory and Coinhibitory (Immune Checkpoint) Interactions. , 2018, Current topics in medicinal chemistry.

[26]  Katarzyna Guzik,et al.  A patent review on PD-1/PD-L1 antagonists: small molecules, peptides, and macrocycles (2015-2018) , 2018, Expert opinion on therapeutic patents.

[27]  PD-1/PD-L1 Inhibitors for Immuno-oncology: From Antibodies to Small Molecules. , 2018, Current pharmaceutical design.

[28]  S. Patil,et al.  Macrocyclic Compounds from Ansamycin Antibiotic Class as Inhibitors of PD1-PDL1 Protein-Protein Interaction. , 2018, Chemical & pharmaceutical bulletin.

[29]  M. Ramachandra,et al.  Small-Molecule Immune Checkpoint Inhibitors Targeting PD-1/PD-L1 and Other Emerging Checkpoint Pathways , 2018, BioDrugs.

[30]  Mamoru Ito,et al.  Human PBMC-transferred murine MHC class I/II-deficient NOG mice enable long-term evaluation of human immune responses , 2017, Cellular & Molecular Immunology.

[31]  F. Feng,et al.  Peptide-based and small synthetic molecule inhibitors on PD-1/PD-L1 pathway: A new choice for immunotherapy? , 2019, European journal of medicinal chemistry.

[32]  M. Ramachandra,et al.  A Rationally Designed Peptide Antagonist of the PD-1 Signaling Pathway as an Immunomodulatory Agent for Cancer Therapy , 2019, Molecular Cancer Therapeutics.

[33]  T. Holak,et al.  Design, synthesis, evaluation and structural studies of C2-symmetric small molecule inhibitors of the programmed cell death-1/programmed death-ligand 1 (PD-1/PD-L1) protein-protein interaction. , 2019, Journal of medicinal chemistry.