Preparation of Biphenyl-Conjugated Bromotyrosine for Inhibition of PD-1/PD-L1 Immune Checkpoint Interactions

Cancer immunotherapy has been revolutionized by the development of monoclonal antibodies (mAbs) that inhibit interactions between immune checkpoint molecules, such as programmed cell-death 1 (PD-1), and its ligand PD-L1. However, mAb-based drugs have some drawbacks, including poor tumor penetration and high production costs, which could potentially be overcome by small molecule drugs. BMS-8, one of the potent small molecule drugs, induces homodimerization of PD-L1, thereby inhibiting its binding to PD-1. Our assay system revealed that BMS-8 inhibited the PD-1/PD-L1 interaction with IC50 of 7.2 μM. To improve the IC50 value, we designed and synthesized a small molecule based on the molecular structure of BMS-8 by in silico simulation. As a result, we successfully prepared a biphenyl-conjugated bromotyrosine (X) with IC50 of 1.5 μM, which was about five times improved from BMS-8. We further prepared amino acid conjugates of X (amino-X), to elucidate a correlation between the docking modes of the amino-Xs and IC50 values. The results suggested that the displacement of amino-Xs from the BMS-8 in the pocket of PD-L1 homodimer correlated with IC50 values. This observation provides us a further insight how to derivatize X for better inhibitory effect.

[1]  T. Honjo,et al.  Induced expression of PD‐1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. , 1992, The EMBO journal.

[2]  R. Abagyan,et al.  Biased probability Monte Carlo conformational searches and electrostatic calculations for peptides and proteins. , 1994, Journal of molecular biology.

[3]  Ruben Abagyan,et al.  ICM—A new method for protein modeling and design: Applications to docking and structure prediction from the distorted native conformation , 1994, J. Comput. Chem..

[4]  J. Allison,et al.  Enhancement of Antitumor Immunity by CTLA-4 Blockade , 1996, Science.

[5]  R Abagyan,et al.  Flexible protein–ligand docking by global energy optimization in internal coordinates , 1997, Proteins.

[6]  J. Bluestone,et al.  Molecular basis of T cell inactivation by CTLA-4. , 1998, Science.

[7]  G. Zhu,et al.  B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion , 1999, Nature Medicine.

[8]  Ruben Abagyan,et al.  Prediction of the binding energy for small molecules, peptides and proteins , 1999, Journal of molecular recognition : JMR.

[9]  G. Freeman,et al.  Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation , 2000, The Journal of experimental medicine.

[10]  Yan Zhang,et al.  Crystal structure of the B7-1/CTLA-4 complex that inhibits human immune responses , 2001, Nature.

[11]  Yoshimasa Tanaka,et al.  Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[12]  Haidong Dong,et al.  Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion , 2002, Nature Medicine.

[13]  C. Drake Basic overview of current immunotherapy approaches in urologic malignancy. , 2006, Urologic oncology.

[14]  R. Eglen,et al.  The Use of AlphaScreen Technology in HTS: Current Status , 2008, Current chemical genomics.

[15]  Janice M Reichert,et al.  Monoclonal antibodies as innovative therapeutics. , 2008, Current pharmaceutical biotechnology.

[16]  Ruben Abagyan,et al.  A new method for ligand docking to flexible receptors by dual alanine scanning and refinement (SCARE) , 2008, J. Comput. Aided Mol. Des..

[17]  Etienne Weiss,et al.  Therapeutic antibodies: successes, limitations and hopes for the future , 2009, British journal of pharmacology.

[18]  Ruben Abagyan,et al.  Four-dimensional docking: a fast and accurate account of discrete receptor flexibility in ligand docking. , 2009, Journal of medicinal chemistry.

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

[20]  Abhinav A Shukla,et al.  Recent advances in large-scale production of monoclonal antibodies and related proteins. , 2010, Trends in biotechnology.

[21]  Louis M. Weiner,et al.  Monoclonal antibodies: versatile platforms for cancer immunotherapy , 2010, Nature Reviews Immunology.

[22]  Nan Wang,et al.  Monoclonal antibodies in cancer therapy , 2011 .

[23]  George Coukos,et al.  Cancer immunotherapy comes of age , 2011, Nature.

[24]  Ruben Abagyan,et al.  Docking and scoring with ICM: the benchmarking results and strategies for improvement , 2012, Journal of Computer-Aided Molecular Design.

[25]  F. Lyko,et al.  Epigenetic cancer therapy: rationales, targets and drugs , 2012, Oncogene.

[26]  A. Korman,et al.  In Vitro Characterization of the Anti-PD-1 Antibody Nivolumab, BMS-936558, and In Vivo Toxicology in Non-Human Primates , 2014, Cancer Immunology Research.

[27]  Yoshihiro Ito,et al.  In vitro selection of a peptide aptamer that potentiates inhibition of cyclin-dependent kinase 2 by purvalanol , 2014 .

[28]  J. Wolchok,et al.  Immune Checkpoint Blockade in Cancer Therapy. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

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

[31]  Peptide-Assisted Enhancement of Inhibitory Effects of Small Molecular Inhibitors for Kinases , 2016 .

[32]  C. Dann,et al.  Tumor Targeting with Novel 6-Substituted Pyrrolo [2,3-d] Pyrimidine Antifolates with Heteroatom Bridge Substitutions via Cellular Uptake by Folate Receptor α and the Proton-Coupled Folate Transporter and Inhibition of de Novo Purine Nucleotide Biosynthesis. , 2016, Journal of medicinal chemistry.

[33]  P. Peddi,et al.  Immune checkpoint inhibitors: the new frontier in non-small-cell lung cancer treatment , 2016, OncoTargets and therapy.

[34]  S. Sahasranaman,et al.  Immune Checkpoint inhibitors: An introduction to the next‐generation cancer immunotherapy , 2016, Journal of clinical pharmacology.

[35]  R. Ueda,et al.  Current status of immunotherapy. , 2016, Japanese journal of clinical oncology.

[36]  J. Pento Monoclonal Antibodies for the Treatment of Cancer. , 2017, Anticancer research.

[37]  S. Almo,et al.  Structure-guided development of a high-affinity human Programmed Cell Death-1: Implications for tumor immunotherapy , 2017, EBioMedicine.

[38]  G. Gao,et al.  An unexpected N-terminal loop in PD-1 dominates binding by nivolumab , 2017, Nature Communications.

[39]  J. Wargo,et al.  Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy , 2017, Cell.

[40]  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.

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

[42]  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.

[43]  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.

[44]  Sultan Gulce-Iz,et al.  Monoclonal antibodies in cancer immunotherapy , 2018, Molecular Biology Reports.

[45]  Hongming Zhang,et al.  Current status and future directions of cancer immunotherapy , 2018, Journal of Cancer.

[46]  T. Holak,et al.  CA-170 – A Potent Small-Molecule PD-L1 Inhibitor or Not? , 2019, bioRxiv.

[47]  M. Yamamura,et al.  Enhancement of Binding Affinity of Folate to Its Receptor by Peptide Conjugation , 2019, International journal of molecular sciences.