1
|
Zhang F, Ramar S, Wang Y, Xu H, Zhang K, Awadasseid A, Rao G, Zhang W. Advances in cancer immunotherapy using small-molecular inhibitors targeting the PD-1/PD-L1 interaction. Bioorg Med Chem 2025; 127:118238. [PMID: 40367914 DOI: 10.1016/j.bmc.2025.118238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Cancer cells evade immune responses by interacting with PD-1 and its ligand, PD-L1. Although monoclonal antibodies targeting this pathway have revolutionized oncology, their high production costs, poor oral bioavailability, and limited tumor penetration remain significant challenges. Small-molecule inhibitors provide a promising alternative, offering advantages such as improved tumor penetration and cost-effectiveness. This review highlights advancements in small-molecule PD-1/PD-L1 inhibitors, focusing on their mechanisms, structural designs, and therapeutic potential. Key innovations, including biphenyl scaffolds, heterocyclic frameworks, enhance binding efficiency and immune activation. The article effectively integrates fundamental principles of drug chemistry with real-world clinical needs, offering a comprehensive approach to the design of PD-1/PD-L1 small-molecule inhibitors. It systematically classifies various molecular structures, analyzes relevant industrial cases, and incorporates the most recent research findings. By examining these aspects, it uncovers the underlying logic driving the design process and provides a fresh, innovative perspective on advancing the field of immune small-molecule inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Feng Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Sivaramakarthikeyan Ramar
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Moganshan Institute ZJUT, Deqing 313202, China
| | - Yu Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Haoran Xu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Koutian Zhang
- Zhejiang Qingzhenghong Technology Co., Ltd, Hangzhou 311121, China
| | - Annoor Awadasseid
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Zhejiang Qingzhenghong Technology Co., Ltd, Hangzhou 311121, China.
| | - Guowu Rao
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Zhejiang Jieyuan Med-Tech Co., Ltd., Hangzhou 311113, China.
| |
Collapse
|
2
|
Kahvecioglu D. Molecular Pathways in Idiopathic Pulmonary Fibrosis: A Review of Novel Insights for Drug Design. Drug Dev Res 2025; 86:e70094. [PMID: 40293838 DOI: 10.1002/ddr.70094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Idiopathic pulmonary fibrosis is a progressive, irreversible lung disease of unknown cause, characterized by gradual thickening and scarring of lung tissue, impairing oxygen transfer into the bloodstream. As a result, symptoms such as shortness of breath, fatigue, and a persistent dry cough occur. Currently, the FDA-approved antifibrotic agents Pirfenidone and Nintedanib can slow the progression of the disease. However, these treatments cannot completely stop the loss of lung function and do not provide a significant improvement in the quality of life of patients. As fibrosis progresses, lung capacity decreases, shortness of breath increases, and general health deteriorates significantly. Therefore, new more effective, and targeted therapies that can halt the progression of IPF are urgently needed. This review addresses novel strategies to slow or halt the disease-related loss of lung function by targeting key mechanisms involved in the pathogenesis of IPF. The molecular structure-activity relationships (SARs) of synthesized compounds targeting JAK/STAT, TGF-β/Smad, Wnt/β-catenin, PI3K, JNK1, and other critical signaling pathways were examined. These targeted approaches have great potential for the development of more potent and selective therapeutic agents for the treatment of IPF. The insights provided in this review may contribute to the future development of more efficient and selective antifibrotic drugs.
Collapse
Affiliation(s)
- Dilay Kahvecioglu
- Edirne Sultan 1. Murat State Hospital, Republic of Türkiye Ministry of Health, Edirne, Turkey
| |
Collapse
|
3
|
Lu T, Zhang J, Chen Q, Ni M, Zhang J, Wu Y, Jia R, Wang Y. Design, Synthesis, Evaluation, and SAR of 5-Phenylisoindoline Derivatives, a Potent Class of Small-Molecule Inhibitors Targeting the Programmed Cell Death-1/Programmed Cell Death-Ligand 1 (PD-1/PD-L1) Interaction. J Med Chem 2025; 68:7291-7312. [PMID: 40153736 DOI: 10.1021/acs.jmedchem.4c02206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
Abstract
A novel series of 5-phenylisoindoline derivatives were designed, synthesized, and evaluated for their activity to inhibit the interaction of PD-1/PD-L1 through the homogeneous time-resolved fluorescence assay. Meanwhile, structure-activity relationships were discussed according to both experiments and calculations. Several compounds exhibited potent activity with an IC50 value less than 10 nM, especially D6 (4.8 nM). D6 could promote IFN-γ secretion and reduce the proportion of PD-L1 late apoptosis at 100 nM in the coculture model of peripheral blood mononuclear cells and hPD-L1-FC. Beyond this, the in vitro model showed D6 could lead to the weakening of migration caused by the PD-1/PD-L1 axis. Furthermore, D6 also displayed dose-dependent and low-toxic efficacy in the MC38 mouse tumor model with the tumor growth inhibition of 52.8% (20 mg/kg, ip) and 64.4% (160 mg/kg, i.g.). Mechanistic investigations suggested that D6 could activate the immune microenvironment in the tumor. Thus, D6 is a promising small molecule lead for blocking PD-1/PD-L1.
Collapse
Affiliation(s)
- Tian Lu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Key Laboratory of Drug Innovation for Neuro-Oncology, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Jiyi Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Qiyu Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Mengyue Ni
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Jingwen Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yufei Wu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Ruining Jia
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuji Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Key Laboratory of Drug Innovation for Neuro-Oncology, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
4
|
Jiang D, Kwon HK, Kwon OW, Choi Y. A Comparative Molecular Dynamics Study of Food-Derived Compounds as PD-L1 Inhibitors: Insights Across Six Flavonoid Subgroups. Molecules 2025; 30:907. [PMID: 40005217 PMCID: PMC11858612 DOI: 10.3390/molecules30040907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
In this study, we investigated the inhibitory potential of 60 flavonoids from six distinct subgroups on the programmed cell death ligand 1 (PD-L1) dimer through molecular docking and dynamics simulations. Using AutoDock Vina for docking, the binding poses and affinities were evaluated, revealing an average binding affinity of -8.5 kcal/mol for the flavonoids. Among them, ginkgetin exhibited the highest binding free energy of -46.73 kcal/mol, indicating a strong interaction with PD-L1, while diosmin followed closely, with -44.96 kcal/mol. Molecular dynamics simulations were used to further elucidate the dynamic interactions and stability of the flavonoid-PD-L1 complexes, with the analyses showing minimal root mean square deviation (RMSD) and favorable root mean square fluctuation (RMSF) profiles for several compounds, particularly formononetin, idaein, and neohesperidin. Additionally, contact number and hydrogen bond analyses were performed, which highlighted ginkgetin and diosmin as key flavonoids with significant binding interactions, evidenced by their stable conformations and robust molecular interactions throughout the simulations. Ultimately, a cell-based assay confirmed their ability to inhibit the proliferation of cancer cells. These results, validated through cell-based assays, indicate that the strategy of identifying natural compounds with anticancer activity using computational modeling is highly effective.
Collapse
Affiliation(s)
- Dejun Jiang
- Department of Environmental Engineering, Hoseo University, Asan 31499, Republic of Korea; (D.J.); (H.-K.K.)
| | - Hyuk-Ku Kwon
- Department of Environmental Engineering, Hoseo University, Asan 31499, Republic of Korea; (D.J.); (H.-K.K.)
| | - Oh Wook Kwon
- Pet-Loss Center, Hoseo University, Asan 31499, Republic of Korea;
| | - Youngjin Choi
- Department of Food Science & Technology, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
5
|
Sobral PS, Carvalho T, Izadi S, Castilho A, Silva Z, Videira PA, Pereira F. Advancements in drug discovery: integrating CADD tools and drug repurposing for PD-1/PD-L1 axis inhibition. RSC Adv 2025; 15:2298-2316. [PMID: 39867321 PMCID: PMC11755407 DOI: 10.1039/d4ra08245a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025] Open
Abstract
Despite significant strides in improving cancer survival rates, the global cancer burden remains substantial, with an anticipated rise in new cases. Immune checkpoints, key regulators of immune responses, play a crucial role in cancer evasion mechanisms. The discovery of immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 has revolutionized cancer treatment, with monoclonal antibodies (mAbs) becoming widely prescribed. However, challenges with current mAb ICIs, such as limited oral bioavailability, adverse effects, and high costs, underscore the need to explore alternative small-molecule inhibitors. In this work, we aimed to identify new potential ICI among all FDA-approved drugs. We employed QSAR models to predict PD-1/PD-L1 inhibition, utilizing a diverse dataset of 29 197 molecules sourced from ChEMBL, PubChem, and recent literature. Machine learning techniques, including Random Forest, Support Vector Machine, and Convolutional Neural Network, were employed for benchmarking to assess model performance. Additionally, we undertook a drug repurposing strategy, leveraging the best in silico model for a virtual screening campaign involving 1576 off-patent approved drugs. Only two virtual screening hits were proposed based on the criteria established for this approach, including: (1) QSAR probability of being active against PD-L1; (2) QSAR applicability domain; (3) prediction of the affinity between the PD-L1 and ligands through molecular docking. One of the proposed hits was sonidegib, an anticancer drug, featuring a biphenyl system. Sonidegib was subsequently validated for in vitro PD-1/PD-L1 binding modulation using ELISA and flow cytometry. This integrated approach, which combines computer-aided drug design (CADD) tools, QSAR modelling, drug repurposing, and molecular docking, offers a pioneering strategy to expedite drug discovery for PD-1/PD-L1 axis inhibition. The findings underscore the potential to identify a wider range small molecules to contribute to the ongoing efforts to advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Patrícia S Sobral
- LAQV and REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa Caparica Portugal
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa Caparica Portugal
| | - Tiago Carvalho
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa Caparica Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa 2829-516 Caparica Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Shiva Izadi
- University of Natural Resources and Life Sciences, Department of Applied Genetics and Cell Biology Vienna Austria
| | - Alexandra Castilho
- University of Natural Resources and Life Sciences, Department of Applied Genetics and Cell Biology Vienna Austria
| | - Zélia Silva
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa Caparica Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa 2829-516 Caparica Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Paula A Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa Caparica Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa 2829-516 Caparica Portugal
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Florbela Pereira
- LAQV and REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa Caparica Portugal
| |
Collapse
|
6
|
Sasmal P, Prabitha P, Prashantha Kumar BR, Swetha BR, Babasahib SK, Raghavendra NM. Beyond peptides: Unveiling the design strategies, structure activity correlations and protein-ligand interactions of small molecule inhibitors against PD-1/PD-L1. Bioorg Chem 2025; 154:108036. [PMID: 39693923 DOI: 10.1016/j.bioorg.2024.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
The landscape of cancer treatment has been transformed by the emergence of immunotherapy, especially through the use of antibodies that target the PD-1/PD-L1 pathway. Recently, there has been a notable increase in interest surrounding immune checkpoint inhibitors for cancer therapy. While antibody-based approaches have drawbacks like high costs and prolonged activity, the approval of monoclonal antibodies such as pembrolizumab and nivolumab has paved the way for a range of alternative therapies, including peptides, peptidomimetics, and small-molecule inhibitors. These smaller molecules, which target the PD-1/PD-L1 interaction, are seen as potential substitutes or supplements to monoclonal antibodies. Our focus in this article is primarily on exploring small molecules designed for PD-1/PD-L1 checkpoint pathway modulation in cancer immunotherapy, along with highlighting current advances in their structural and preclinical/clinical development. The pursuit of therapeutics based on small-molecule inhibitors of the PD-1/PD-L1 axis offers a promising yet intricate avenue for advancing cancer treatment.
Collapse
Affiliation(s)
- Pujan Sasmal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar - 160 062, Punjab, India; Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy (ABMRCP), Bengaluru 560 107, Karnataka, India.
| | - P Prabitha
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - B R Swetha
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University (DSU), Bengaluru 560 111, Karnataka, India
| | - Sajeev Kumar Babasahib
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University (DSU), Bengaluru 560 111, Karnataka, India
| | - Nulgumnalli Manjunathaiah Raghavendra
- Department of Pharmaceutical Chemistry, College of Pharmaceutical Sciences, Dayananda Sagar University (DSU), Bengaluru 560 111, Karnataka, India; Department of Pharmaceutical Chemistry, R R College of Pharmacy, Bengaluru 560 090, Karnataka, India.
| |
Collapse
|
7
|
Wang K, Cai S, Cheng Y, Qi Z, Ni X, Zhang K, Xiao Y, Zhang X, Wang T. Discovery of Benzo[ d]oxazoles as Novel Dual Small-Molecule Inhibitors Targeting PD-1/PD-L1 and VISTA Pathway. J Med Chem 2024; 67:18526-18548. [PMID: 39389791 DOI: 10.1021/acs.jmedchem.4c01899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The blockers of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway have achieved great clinical success. However, the limited efficacy and low tumor response rate of anti-PD-1/PD-L1 monotherapy limit the clinical application of PD-1/PD-L1 inhibitors. V-domain immunoglobulin suppressor of T-cell activation (VISTA), a novel checkpoint regulator, exhibits potential synergy with PD-1/PD-L1 in enhancing antitumor immunity. Herein, we report the discovery of benzo[d]oxazole B3 as novel dual small-molecule inhibitors targeting PD-1/PD-L1 and VISTA with high PD-1/PD-L1 inhibitory activity and VISTA binding affinity. B3 rescues the immunosuppression of T-cells mediated by PD-L1 and VISTA and activates antitumor immunity effectively. Moreover, B3 could induce degradation of PD-L1 and VISTA in tumor cell. Furthermore, B3 displays significant in vivo antitumor efficacy in a CT26 mouse model. Our results discover B3 as a promising dual PD-1/PD-L1 and VISTA inhibitor, providing a novel therapeutic strategy to overcome the limitations of current anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi Cai
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Innovation Department of the Research Institute, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Yao Cheng
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Qi
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiang Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
8
|
Zhang F, Zhang H, Zhou S, Plewka J, Wang M, Sun S, Wu C, Yu Q, Zhu M, Awadasseid A, Wu Y, Magiera-Mularz K, Zhang W. Design, synthesis, and evaluation of antitumor activity of 2-arylmethoxy-4-(2-fluoromethyl-biphenyl-3-ylmethoxy) benzylamine derivatives as PD-1/PD-l1 inhibitors. Eur J Med Chem 2024; 276:116683. [PMID: 39032403 DOI: 10.1016/j.ejmech.2024.116683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
A series of novel 2-arylmethoxy-4-(2-fluoromethyl-biphenyl-3-ylmethoxy) benzylamine derivatives was designed, synthesized, and evaluated for their antitumor effects as PD-1/PD-L1 inhibitors both in vitro and in vivo. Firstly, the ability of these compounds to block the PD-1/PD-L1 immune checkpoint was assessed using the homogeneous time-resolved fluorescence (HTRF) assay. Two of the compounds can strongly block the PD-1/PD-L1 interaction, with IC50 values of less than 10 nM, notably, compound HD10 exhibited significant clinical potential by inhibiting the PD-1/PD-L1 interaction with an IC50 value of 3.1 nM. Further microscale thermophoresis (MST) analysis demonstrated that HD10 had strong interaction with PD-L1 protein. Co-crystal structure (2.7 Å) analysis of HD10 in complex with the PD-L1 protein revealed a strong affinity between the compound and the target PD-L1 dimer. This provides a solid theoretical basis for further in vitro and in vivo studies. Next, a typical cell-based experiment demonstrated that HD10 could remarkably prevent the interaction of hPD-1 293 T cells from human recombinant PD-L1 protein, effectively restoring T cell function, and promoting IFN-γ secretion in a dose-dependent manner. Moreover, HD10 was effective in suppressing tumor growth (TGI = 57.31 %) in a PD-1/PD-L1 humanized mouse model without obvious toxicity. Flow cytometry, qPCR, and immunohistochemistry data suggested that HD10 inhibits tumor growth by activating the immune system in vivo. Based on these results, it seems likely that HD10 is a promising clinical candidate that should be further investigated.
Collapse
Affiliation(s)
- Feng Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Hua Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China; Department of Pharmacy, Changzhi Medical College, Shanxi, 046012, China
| | - Shijia Zhou
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Jacek Plewka
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Ming Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Shishi Sun
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Caiyun Wu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Qimeng Yu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Mengyu Zhu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China
| | - Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China; Moganshan Institute, Zhejiang University of Technology, Deqing, 313200, China.
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China.
| | - Katarzyna Magiera-Mularz
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing, 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing, 313299, China; Zhejiang Jieyuan Med-Tech Co., Ltd., Hangzhou, 311113, China.
| |
Collapse
|
9
|
Wu X, Li H, Liu H, Ding X, Chen X, Yin C, Gao Y, Ma J. Design, Synthesis, and Evaluation of 8-( o-Tolyl)quinazoline Derivatives as Small-Molecule PD-1/PD-L1 Antagonists. ACS Med Chem Lett 2024; 15:518-523. [PMID: 38628793 PMCID: PMC11017391 DOI: 10.1021/acsmedchemlett.4c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Small-molecule inhibitors targeting programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) interactions can compensate for the shortcomings of antibody-based inhibitors and have attracted considerable attention, some of which have already entered clinical trials. Herein, based on our previous study on small-molecule PD-L1 inhibitors, we reported a series of 8-(o-tolyl)quinazoline derivatives by the skeleton merging strategy. Homogenous time-resolved fluorescence (HTRF) assay against PD-1/PD-L1 interaction identified compound A5, which showed the most potent inhibition with an IC50 value of 23.78 nM. Meanwhile, based on the results of HTRF assay, the structure-activity relationships (SARs) of the tail were focused on. Cell-based PD-1/PD-L1 blockade assay further revealed that A5 significantly blocked the PD-1/PD-L1 interaction at 1.1 μM in the co-culture system of Jurkat-NFAT-PD-1 cells and Hep3B-OS8-hPD-L1 cells with no significant cytotoxicity on Jurkat cells. Moreover, the proposed binding mode of A5 was investigated by a docking analysis. These results indicate that compound A5 is a promising lead compound that deserves further investigation.
Collapse
Affiliation(s)
- Xingye Wu
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| | - He Li
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| | - Han Liu
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| | - Xueyan Ding
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| | - Xinting Chen
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| | - Chenxi Yin
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| | - Yali Gao
- Pharmacy
Department, The Second Affiliated Hospital
of Fujian Medical University, Quanzhou, 362002, China
| | - Junjie Ma
- School
of Medicine, Huaqiao University, Quanzhou, 362000, China
| |
Collapse
|
10
|
Javed SA, Najmi A, Ahsan W, Zoghebi K. Targeting PD-1/PD-L-1 immune checkpoint inhibition for cancer immunotherapy: success and challenges. Front Immunol 2024; 15:1383456. [PMID: 38660299 PMCID: PMC11039846 DOI: 10.3389/fimmu.2024.1383456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The programmed death-1 receptor (PD-1) acts as a T-cell brake, and its interaction with ligand-1 (PD-L-1) interferes with signal transduction of the T-cell receptor. This leads to suppression of T-cell survival, proliferation, and activity in the tumor microenvironment resulting in compromised anticancer immunity. PD-1/PD-L-1 interaction blockade shown remarkable clinical success in various cancer immunotherapies. To date, most PD-1/PD-L-1 blockers approved for clinical use are monoclonal antibodies (mAbs); however, their therapeutic use are limited owing to poor clinical responses in a proportion of patients. mAbs also displayed low tumor penetration, steep production costs, and incidences of immune-related side effects. This strongly indicates the importance of developing novel inhibitors as cancer immunotherapeutic agents. Recently, advancements in the small molecule-based inhibitors (SMIs) that directly block the PD-1/PD-L-1 axis gained attention from the scientific community involved in cancer research. SMIs demonstrated certain advantages over mAbs, including longer half-lives, low cost, greater cell penetration, and possibility of oral administration. Currently, several SMIs are in development pipeline as potential therapeutics for cancer immunotherapy. To develop new SMIs, a wide range of structural scaffolds have been explored with excellent outcomes; biphenyl-based scaffolds are most studied. In this review, we analyzed the development of mAbs and SMIs targeting PD-1/PD-L-1 axis for cancer treatment. Altogether, the present review delves into the problems related to mAbs use and a detailed discussion on the development and current status of SMIs. This article may provide a comprehensive guide to medicinal chemists regarding the potential structural scaffolds required for PD-1/PD-L-1 interaction inhibition.
Collapse
Affiliation(s)
| | - Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | | |
Collapse
|
11
|
Wang K, Zhang X, Cheng Y, Qi Z, Ye K, Zhang K, Jiang S, Liu Y, Xiao Y, Wang T. Discovery of Novel PD-L1 Inhibitors That Induce the Dimerization, Internalization, and Degradation of PD-L1 Based on the Fragment Coupling Strategy. J Med Chem 2023; 66:16807-16827. [PMID: 38109261 DOI: 10.1021/acs.jmedchem.3c01534] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Tumor cells can evade immune surveillance through overexpressing programmed cell death-ligand 1 (PD-L1) to interact with programmed cell death-1 (PD-1). Besides, tumor-intrinsic PD-L1 is involved in tumor progression without interaction with PD-1, which provides more challenges for the discovery of PD-L1 inhibitors. Herein, we report the discovery of novel PD-L1 inhibitors using the fragment coupling strategy. Among them, B9 was found to inhibit the PD-1/PD-L1 interaction with the best IC50 value of 1.8 ± 0.7 nM. Beyond the blockade of the PD-1/PD-L1 axis, B9 promotes the dimerization, internalization, and degradation of PD-L1. Furthermore, B9 displayed high in vivo antitumor efficacy in the CT26 mouse model and activated the immune microenvironment and induced PD-L1 degradation of PD-L1 in the tumor. These results show that B9 is a promising lead PD-L1 inhibitor through the blockade of PD-1/PD-L1 interaction and functional inhibition of the PD-L1 signal pathway.
Collapse
Affiliation(s)
- Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yao Cheng
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Qi
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ke Ye
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
12
|
Zhang H, Zhou S, Plewka J, Wu C, Zhu M, Yu Q, Musielak B, Wang X, Awadasseid A, Magiera-Mularz K, Wu Y, Zhang W. Design, Synthesis, and Antitumor Activity Evaluation of 2-Arylmethoxy-4-(2,2'-dihalogen-substituted biphenyl-3-ylmethoxy) Benzylamine Derivatives as Potent PD-1/PD-L1 Inhibitors. J Med Chem 2023; 66:10579-10603. [PMID: 37496104 DOI: 10.1021/acs.jmedchem.3c00731] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Novel 2-arylmethoxy-4-(2,2'-dihalogen-substituted biphenyl-3-ylmethoxy) benzylamine derivatives were designed, synthesized, and evaluated in vitro and in vivo against cancers as PD-1/PD-L1 inhibitors. Through the computer-aided structural optimization and the homogeneous time-resolved fluorescence (HTRF) assay, compound A56 was found to most strongly block the PD-1/PD-L1 interaction with an IC50 value of 2.4 ± 0.8 nM and showed the most potent activity. 1H NMR titration results indicated that A56 can tightly bind to the PD-L1 protein with KD < 1 μM. The X-ray diffraction data for the cocrystal structure of the A56/PD-L1 complex (3.5 Å) deciphered a novel binding mode in detail, which can account for its most potent inhibitory activity. Cell-based assays further demonstrated the strong ability of A56 as an hPD-1/hPD-L1 blocker. Especially in an hPD-L1 MC38 humanized mouse model, A56 significantly inhibited tumor growth without obvious toxicity, with a TGI rate of 55.20% (50 mg/kg, i.g.). In conclusion, A56 is a promising clinical candidate worthy of further development.
Collapse
Affiliation(s)
- Hua Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shijia Zhou
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jacek Plewka
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Caiyun Wu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Mengyu Zhu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qimeng Yu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Bogdan Musielak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Xiao Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Katarzyna Magiera-Mularz
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
13
|
Cai S, Wang K, Qi Z, Ye K, Zhou X, Jiang S, Zhang K, Zhang X, Wang T. Design, synthesis, and evaluation of PD-1/PD-L1 small-molecule inhibitors bearing a rigid indane scaffold. Eur J Med Chem 2023; 256:115468. [PMID: 37207535 DOI: 10.1016/j.ejmech.2023.115468] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
Discovery of small-molecule inhibitors against programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) axis provides a promising alternative to overcome the inevitable defects of PD-1/PD-L1 monoclonal antibodies (mAbs). Here, we report a series of indanes as novel small-molecule inhibitors of PD-1/PD-L1 interaction. Thirty-one indanes were synthesized and the structure-activity relationships (SARs) demonstrated that conformational restriction with (S)-indane is superior in potency to inhibit the interaction of PD-1 and PD-L1. Compound D3 was found to be the most potent inhibitor with an IC50 value of 2.2 nM against PD-1/PD-L1 interaction. Cell-based assay showed that D3 significantly induced immune activity of peripheral blood mononuclear cells (PBMCs) against MDA-MB-231 cells and could restore the immune function of T cells by promoting secretion of the IFN-γ. The above results indicate that compound D3 is a promising PD-1/PD-L1 inhibitor that deserves further development.
Collapse
Affiliation(s)
- Shi Cai
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kaizhen Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhihao Qi
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ke Ye
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xinyuan Zhou
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiangyu Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Tianyu Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
14
|
Regnault R, Klupsch F, El-Bouazzati H, Magnez R, Le Biannic R, Leleu-Chavain N, Ahouari H, Vezin H, Millet R, Goossens JF, Thuru X, Bailly C. Novel PD-L1-Targeted Phenyl-Pyrazolone Derivatives with Antioxidant Properties. Molecules 2023; 28:molecules28083491. [PMID: 37110727 PMCID: PMC10144346 DOI: 10.3390/molecules28083491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Orally-active anticancer small molecules targeting the PD-1/PD-L1 immune checkpoint are actively searched. Phenyl-pyrazolone derivatives with a high affinity for PD-L1 have been designed and characterized. In addition, the phenyl-pyrazolone unit acts as a scavenger of oxygen free radicals, providing antioxidant effects. The mechanism is known for the drug edaravone (1) which is also an aldehyde-reactive molecule. The present study reports the synthesis and functional characterization of new molecules (2-5) with an improved anti-PD-L1 activity. The leading fluorinated molecule 5 emerges as a potent checkpoint inhibitor, avidly binding to PD-L1, inducing its dimerization, blocking PD-1/PD-L1 signaling mediated by phosphatase SHP-2 and reactivating the proliferation of CTLL-2 cells in the presence of PD-L1. In parallel, the compound maintains a significant antioxidant activity, characterized using electron paramagnetic resonance (EPR)-based free radical scavenging assays with the probes DPPH and DMPO. The aldehyde reactivity of the molecules was investigated using 4-hydroxynonenal (4-HNE), which is a major lipid peroxidation product. The formation of drug-HNE adducts, monitored by high resolution mass spectrometry (HRMS), was clearly identified and compared for each compound. The study leads to the selection of compound 5 and the dichlorophenyl-pyrazolone unit as a scaffold for the design of small molecule PD-L1 inhibitors endowed with antioxidant properties.
Collapse
Affiliation(s)
- Romain Regnault
- ULR 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, CHU Lille, University Lille, F-59000 Lille, France
| | - Frédérique Klupsch
- U1286-INFINITE-Institute for Translational Research in Inflammation, ICPAL, Inserm, University Lille, F-59000 Lille, France
| | - Hassiba El-Bouazzati
- UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Inserm, CNRS, CHU Lille, University Lille, F-59000 Lille, France
| | - Romain Magnez
- UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Inserm, CNRS, CHU Lille, University Lille, F-59000 Lille, France
| | - Raphaël Le Biannic
- U1286-INFINITE-Institute for Translational Research in Inflammation, ICPAL, Inserm, University Lille, F-59000 Lille, France
| | - Natascha Leleu-Chavain
- U1286-INFINITE-Institute for Translational Research in Inflammation, ICPAL, Inserm, University Lille, F-59000 Lille, France
| | - Hania Ahouari
- LASIRE Laboratoire Avancé de Spectroscopie pour les Intéractions la Réactivité et l'Environnement, F-59655 Villeneuve d'Ascq, France
- FR 2638-IMEC-Institut Michel-Eugène Chevreul, University Lille, F-59655 Lille, France
| | - Hervé Vezin
- LASIRE Laboratoire Avancé de Spectroscopie pour les Intéractions la Réactivité et l'Environnement, F-59655 Villeneuve d'Ascq, France
| | - Régis Millet
- U1286-INFINITE-Institute for Translational Research in Inflammation, ICPAL, Inserm, University Lille, F-59000 Lille, France
| | - Jean-François Goossens
- ULR 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, CHU Lille, University Lille, F-59000 Lille, France
| | - Xavier Thuru
- UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Inserm, CNRS, CHU Lille, University Lille, F-59000 Lille, France
| | - Christian Bailly
- UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Inserm, CNRS, CHU Lille, University Lille, F-59000 Lille, France
- Oncowitan, Scientific Consulting Office, Wasquehal, F-59290 Lille, France
| |
Collapse
|
15
|
Sobral PS, Luz VCC, Almeida JMGCF, Videira PA, Pereira F. Computational Approaches Drive Developments in Immune-Oncology Therapies for PD-1/PD-L1 Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24065908. [PMID: 36982981 PMCID: PMC10054797 DOI: 10.3390/ijms24065908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Computational approaches in immune-oncology therapies focus on using data-driven methods to identify potential immune targets and develop novel drug candidates. In particular, the search for PD-1/PD-L1 immune checkpoint inhibitors (ICIs) has enlivened the field, leveraging the use of cheminformatics and bioinformatics tools to analyze large datasets of molecules, gene expression and protein-protein interactions. Up to now, there is still an unmet clinical need for improved ICIs and reliable predictive biomarkers. In this review, we highlight the computational methodologies applied to discovering and developing PD-1/PD-L1 ICIs for improved cancer immunotherapies with a greater focus in the last five years. The use of computer-aided drug design structure- and ligand-based virtual screening processes, molecular docking, homology modeling and molecular dynamics simulations methodologies essential for successful drug discovery campaigns focusing on antibodies, peptides or small-molecule ICIs are addressed. A list of recent databases and web tools used in the context of cancer and immunotherapy has been compilated and made available, namely regarding a general scope, cancer and immunology. In summary, computational approaches have become valuable tools for discovering and developing ICIs. Despite significant progress, there is still a need for improved ICIs and biomarkers, and recent databases and web tools have been compiled to aid in this pursuit.
Collapse
Affiliation(s)
- Patrícia S Sobral
- LAQV and REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Vanessa C C Luz
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - João M G C F Almeida
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Paula A Videira
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Florbela Pereira
- LAQV and REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
16
|
DiFrancesco M, Hofer J, Aradhya A, Rufinus J, Stoddart J, Finocchiaro S, Mani J, Tevis S, Visconti M, Walawender G, DiFlumeri J, Fattakhova E, Patil SP. Discovery of small-molecule PD-1/PD-L1 antagonists through combined virtual screening and experimental validation. Comput Biol Chem 2023; 102:107804. [PMID: 36610303 DOI: 10.1016/j.compbiolchem.2022.107804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 11/17/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Inhibition of the interaction between the PD-1 protein on activated lymphocytes and the PD-L1 protein on tumors represents a novel therapeutic approach for selective activation of the innate immune response against a variety of cancers. Therefore, the present study utilized a combined virtual and experimental screening approach to screen databases of both lead-like and larger molecules for identification of novel inhibitors of PD-1/PD-L1 interaction. First, high-throughput virtual screening of ∼3.7 million lead-like molecules using a rigid-receptor docking approach against both human PD-1 and PD-L1 proteins revealed possible small-molecule tractability of PD-1, but not PD-L1, binding interface. The subsequent work, therefore, involved screening of the National Cancer Institute (NCI) compound database against the PD-1 pocket. Several NCI compounds were identified with potential to bind to the PD-1 pocket and in turn inhibit the PD-1/PD-L1 interaction. The dynamic binding behavior of these molecules was further investigated using long 100 ns molecular dynamics (MD) stimulation revealing NSC631535 to be a potentially stable binder at PD-1 interface pocket. In support of these MD data, the experimental testing of NSC631535 exhibited 50% inhibition at ∼15 μM test concentration. The observed activity of this compound is promising as despite its relatively low molecular weight (415.5 g/mol) it is still capable of inhibiting the PD-1/PD-L1 interaction having a large interface area (∼1970 Å2). In summary, our integrated computational and experimental screening led to identification of a novel PD-1 antagonist that may serve as a starting point for further optimization into more potent small-molecule PD-1/PD-L1 inhibitors for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Jeremy Hofer
- Department of Computer Science, Widener University, Chester, PA, USA
| | - Abhay Aradhya
- Department of Computer Science, Widener University, Chester, PA, USA
| | - Jeffrey Rufinus
- Department of Computer Science, Widener University, Chester, PA, USA
| | - John Stoddart
- Department of Computer Science, Widener University, Chester, PA, USA
| | - Stephen Finocchiaro
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Jabari Mani
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Sean Tevis
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Michael Visconti
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Griffin Walawender
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Juliette DiFlumeri
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Elena Fattakhova
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA
| | - Sachin P Patil
- NanoBio Laboratory, School of Engineering, Widener University, Chester, PA, USA.
| |
Collapse
|
17
|
Immune Checkpoint Inhibitors for Vaccine Improvements: Current Status and New Approaches. Pharmaceutics 2022; 14:pharmaceutics14081721. [PMID: 36015348 PMCID: PMC9415890 DOI: 10.3390/pharmaceutics14081721] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 12/11/2022] Open
Abstract
In recent years, the use of immune checkpoint inhibitors (ICIs) in combination with approved or experimental vaccines has proven to be a promising approach to improve vaccine immunogenicity and efficacy. This strategy seeks to overcome the immunosuppressive mechanisms associated with the vaccine response, thereby achieving increased immunogenicity and efficacy. Most of the information on the use of ICIs combined with vaccines derives from studies on certain anti-tumor vaccines combined with monoclonal antibodies (mAbs) against either cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), or programmed death-ligand 1 (PD-L1). However, over the past few years, emerging strategies to use new-generation ICIs as molecular adjuvants are paving the way for future advances in vaccine research. Here, we review the current state and future directions of the use of ICIs in experimental and clinical settings, including mAbs and alternative new approaches using antisense oligonucleotides (ASOs), small non-coding RNAs, aptamers, peptides, and other small molecules for improving vaccine efficacy. The scope of this review mainly includes the use of ICIs in therapeutic antitumor vaccines, although recent research on anti-infective vaccines will also be addressed.
Collapse
|