1
|
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: 1] [Impact Index Per Article: 1.0] [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
|
2
|
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
|
3
|
Fan X, Feng D, Wei D, Li A, Wei F, Deng S, Shen M, Qin C, Yu Y, Liang L. Characterizing tertiary lymphoid structures associated single-cell atlas in breast cancer patients. Cancer Cell Int 2025; 25:12. [PMID: 39806382 PMCID: PMC11727541 DOI: 10.1186/s12935-025-03635-y] [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: 09/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
The tertiary lymphoid structure (TLS) is recognized as a potential prognosis factor for breast cancer and is strongly associated with response to immunotherapy. Inducing TLS neogenesis can enhance the immunogenicity of tumors and improve the efficacy of immunotherapy. However, our understanding of TLS associated region at the single-cell level remains limited. Therefore, we employed high-resolution techniques, including single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST), and a TLS-specific signature to investigate TLS associated regions in breast cancer. We identified eighteen cell types within the TLS associated regions and calculated differential expression genes by comparing TLS associated regions with other areas. Notably, macrophages in the TLS associated regions exhibit lineage transformation, shifting from facilitators of immune activation to supporters of tumor cell growth. In terms of cell-cell communication within the TLS associated regions, KRT86+ CD8+ T cells, HISTIH4C+ cycling CD8+ T cells, IFNG+ CD8+ T cells, and IGKV3-20+ B cells demonstrate strong interactions with other cells. Additionally, we found that APOD+ fibroblast and CCL21+ fibroblast primarily recruit T and B cells through the CXCL12-CXCR4 ligand-receptor signaling pathway. We also validate these findings in four independent breast cancer datasets, which include one cell-level resolution dataset from the 10 × Xenium platform and three spot-level datasets from the 10 × Visium platform.
Collapse
Affiliation(s)
- Xiaokai Fan
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Daqin Feng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Donggui Wei
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Anqi Li
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Fangyi Wei
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Shufang Deng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Muling Shen
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Congzhi Qin
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yongjia Yu
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China.
| | - Lun Liang
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China.
| |
Collapse
|
4
|
Sun M, Lv S, Pan Y, Song Q, Ma C, Yu M, Gao X, Guo X, Wang S, Gao Z, Wang S, Meng Q, Zhang L, Li Y. Discovery of Daclatasvir as a potential PD-L1 inhibitor from drug repurposing. Bioorg Chem 2024; 153:107874. [PMID: 39418845 DOI: 10.1016/j.bioorg.2024.107874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
This study employed a drug repositioning strategy to discover novel PD-L1 small molecule inhibitors. 3D-QSAR pharmacophore models were establishedand subsequently validated through various means to select a robust model, Hypo-1, suitable for virtual screening. Hypo1 was used toscreen a library of 7,475 compounds from the Drugbank database, leading to the identification of 283 molecules following molecular docking with PD-L1.19 compounds underwent HTRF assays, with 15 showing varying degrees of inhibition of the PD-1/PD-L1 interaction. Compounds2202,2204,2207, and2208were further confirmed to bind to PD-L1 using SPR experiments. Among them, compound2204(Daclatasvir, KD = 11.4 μM) showeda higher affinity for human PD-L1 than the control compound BMS-1. In the HepG2/Jurkat cell co-culture model, Daclatasvir effectively activated Jurkat cells to kill HepG2 cells. In the mouse H22 hepatocellular tumor model, Daclatasvir significantly inhibited tumor growth (TGI = 53.4 % at a dose of 100 mg/kg). Its anti-tumor effect was more pronounced when combined with Lenvatinib (TGI = 85.1 %). Flow cytometry analysis of splenocytes and tumor cells indicated that Daclatasvir activated the immune system in both models. In summary, Daclatasvir was identified as a novel PD-L1small molecule inhibitor.
Collapse
Affiliation(s)
- Mengmeng Sun
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024
| | - Shixuan Lv
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024
| | - Yanyan Pan
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning, China, 116023
| | - Qiling Song
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024
| | - Chunyan Ma
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024
| | - Menglin Yu
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024
| | - Xinru Gao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024
| | - Xiuhan Guo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024; Ningbo Institute of Dalian University of Technology, No.26 Yucai Road, Jiangbei District, Ningbo, China, 315016
| | - Shisheng Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024; Ningbo Institute of Dalian University of Technology, No.26 Yucai Road, Jiangbei District, Ningbo, China, 315016
| | - Zhigang Gao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024; Ningbo Institute of Dalian University of Technology, No.26 Yucai Road, Jiangbei District, Ningbo, China, 315016
| | - Shuai Wang
- Ningbo Institute of Dalian University of Technology, No.26 Yucai Road, Jiangbei District, Ningbo, China, 315016
| | - Qingwei Meng
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024; Ningbo Institute of Dalian University of Technology, No.26 Yucai Road, Jiangbei District, Ningbo, China, 315016
| | - Li Zhang
- Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning, China, 116023
| | - Yueqing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, Liaoning, China, 116024; Ningbo Institute of Dalian University of Technology, No.26 Yucai Road, Jiangbei District, Ningbo, China, 315016.
| |
Collapse
|
5
|
Xu J, Kong Y, Zhu P, Du M, Liang X, Tong Y, Li X, Dong C. Progress in small-molecule inhibitors targeting PD-L1. RSC Med Chem 2024; 15:1161-1175. [PMID: 38665838 PMCID: PMC11042164 DOI: 10.1039/d3md00655g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/29/2024] [Indexed: 04/28/2024] Open
Abstract
PD-L1 is a transmembrane protein overexpressed by tumor cells. It binds to PD-1 on the surface of T-cells, suppresses T-cell activity and hinders the immune response against cancer. Clinically, several monoclonal antibodies targeting PD-1/PD-L1 have achieved significant success in cancer immunotherapy. Nevertheless, their disadvantages, such as unchecked immune responses, high cost and long half-life, stimulated pharmacologists to develop small-molecule inhibitors targeting PD-1/PD-L1. After a batch of excellent inhibitors with a biphenyl core structure were firstly reported by BMS, more and more researchers focused on small-molecule inhibitors targeting PD-L1 rather than PD-1. Numerous small-molecule inhibitors were extensively designed and synthesized in the past few years. In this paper, the structural characteristics of PD-L1 and complexes of PD-L1 with its inhibitors are elaborated and small molecule inhibitors developed in the last decade are summarized as well. This paper aims to provide insights into further designing and synthesis of small molecule inhibitors targeting PD-L1.
Collapse
Affiliation(s)
- Jindan Xu
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
- Henan Polysaccharide Research Center Zhengzhou 450046 Henan China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research Zhengzhou 450046 Henan China
| | - Yuanfang Kong
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
| | - Pengbo Zhu
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
- Henan Polysaccharide Research Center Zhengzhou 450046 Henan China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research Zhengzhou 450046 Henan China
| | - Mingyan Du
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
- Henan Polysaccharide Research Center Zhengzhou 450046 Henan China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research Zhengzhou 450046 Henan China
| | - Xuan Liang
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
| | - Yan Tong
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
| | - Xiaofei Li
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
- Henan Polysaccharide Research Center Zhengzhou 450046 Henan China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research Zhengzhou 450046 Henan China
| | - Chunhong Dong
- Henan University of Chinese Medicine Zhengzhou 450046 Henan China
- Henan Polysaccharide Research Center Zhengzhou 450046 Henan China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research Zhengzhou 450046 Henan China
| |
Collapse
|
6
|
Biersack B, Nitzsche B, Höpfner M. Immunomodulatory properties of HDAC6 inhibitors in cancer diseases: New chances for sophisticated drug design and treatment optimization. Semin Cell Dev Biol 2024; 154:286-294. [PMID: 36127263 DOI: 10.1016/j.semcdb.2022.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/03/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Histone deacetylases (HDACs) are promising targets for the design of anticancer drugs. HDAC6 is of particular interest since it is a cytoplasmic HDAC regulating the acetylation state of cancer-relevant cytoplasmic proteins such as tubulin, Hsp90, p53, and others. HDAC6 also influences the immune system, and the combination of HDAC6 inhibitors with immune therapy showed promising anticancer results. In addition, the design of new HDAC6 inhibitors led to potent anticancer drugs with immunomodulatory activities. This review describes the current state of play, and the recent developments in the research on the interactions of HDAC6 inhibitors with the immune system, and the development of new HDAC6 inhibitors with immunomodulatory activities to improve the therapy options for cancer patients.
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany.
| | - Bianca Nitzsche
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Höpfner
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
7
|
Boisgerault N, Bertrand P. Inside PD-1/PD-L1,2 with their inhibitors. Eur J Med Chem 2023; 256:115465. [PMID: 37196547 DOI: 10.1016/j.ejmech.2023.115465] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
This review summarizes current knowledge in the development of immune checkpoint inhibitors, including antibodies and small molecules.
Collapse
Affiliation(s)
- Nicolas Boisgerault
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université D'Angers, CRCI2NA, LabEx IGO, F-44000, Nantes, France
| | - Philippe Bertrand
- University of Poitiers, IC2MP UMR 7285 CNRS, 4 Rue Michel Brunet B27, TSA 51106, 86073 Poitiers Cedex 9, France.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Marjault HB, Yang-Sung S, Zuo K, Carloni P, Mittler R, Nechushtai R. Structure-Based Screening Reveals a Ligand That Stabilizes the [2Fe-2S] Clusters of Human mitoNEET and Reduces Ovarian Cancer Cell Proliferation. J Phys Chem B 2022; 126:9559-9565. [PMID: 36374279 DOI: 10.1021/acs.jpcb.2c05728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human NEET proteins play an important role in a variety of diseases, including cancer. Using the recently published X-ray structure of the human mNT-M1 complex, we screened a commercial chemical compound library and identified a new human mitoNEET (mNT) binding ligand (NTS-01). Biochemical investigations revealed that NTS-01 specifically binds to the human mNT protein and stabilizes its [2Fe-2S] clusters under oxidative conditions in vitro. Treatment of ovarian cancer cells with NTS-01 induces ovarian cancer (SKOV-3) mitochondrial fragmentation (fission) and reduces ovarian cancer cell proliferation in a 2D single-layer cell culture, as well as in a 3D-spheroids culture. The NTS-01 molecule represents therefore a new lead compound for further drug design studies attempting to develop efficient treatment against ovarian cancer.
Collapse
Affiliation(s)
- Henri-Baptiste Marjault
- The Alexander Silberman Institute of Life Science and The Wolfson Centre for Applied Structural Biology, Faculty of Science and Mathematics, The Edmond J. Safra Campus at Givat Ram, The Hebrew University of Jerusalem, Jerusalem91904, Israel
- Department of Physics, RWTH Aachen University, 52074Aachen, Germany
| | - Sohn Yang-Sung
- The Alexander Silberman Institute of Life Science and The Wolfson Centre for Applied Structural Biology, Faculty of Science and Mathematics, The Edmond J. Safra Campus at Givat Ram, The Hebrew University of Jerusalem, Jerusalem91904, Israel
| | - Ke Zuo
- The Alexander Silberman Institute of Life Science and The Wolfson Centre for Applied Structural Biology, Faculty of Science and Mathematics, The Edmond J. Safra Campus at Givat Ram, The Hebrew University of Jerusalem, Jerusalem91904, Israel
- Department of Physics, RWTH Aachen University, 52074Aachen, Germany
| | - Paolo Carloni
- Department of Physics, RWTH Aachen University, 52074Aachen, Germany
- Computational Biomedicine Section, Institute of Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, 52425Jülich, Germany
- Computational Biomedicine, Institute of Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, 52425Jülich, Germany
- JARA Institute: Molecular Neuroscience and Imaging, Institute of Neuroscience and Medicine INM-11, Forschungszentrum Jülich GmbH, 52425Jülich, Germany
| | - Ron Mittler
- Department of Surgery, University of Missouri School of Medicine, and Interdisciplinary Plant Group, Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Street, Columbia, Missouri65211, United States
| | - Rachel Nechushtai
- The Alexander Silberman Institute of Life Science and The Wolfson Centre for Applied Structural Biology, Faculty of Science and Mathematics, The Edmond J. Safra Campus at Givat Ram, The Hebrew University of Jerusalem, Jerusalem91904, Israel
| |
Collapse
|
11
|
Alshammari AM. Screening of Phytochemicals Against Snake Venom Metalloproteinase: Molecular Docking and Simulation Based Computational Approaches. ARCHIVES OF PHARMACY PRACTICE 2022. [DOI: 10.51847/hirdcdpcgl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|