1
|
Smaldone G, Di Matteo F, Castelluccio R, Napolitano V, Miranda MR, Manfra M, Campiglia P, Vestuto V. Targeting the CXCR4/CXCL12 Axis in Cancer Therapy: Analysis of Recent Advances in the Development of Potential Anticancer Agents. Molecules 2025; 30:1380. [PMID: 40142155 PMCID: PMC11945090 DOI: 10.3390/molecules30061380] [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: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer, a leading cause of premature death, arises from genetic and epigenetic mutations that transform normal cells into tumor cells, enabling them to proliferate, evade cell death, and stimulate angiogenesis. Recent evidence indicates that chemokines are essential in tumor development, activating receptors that promote proliferation, invasion, and metastasis. The CXCR4/CXCL12 signaling pathway is gaining attention as a promising target for cancer therapy. CXCR4, a chemokine receptor, is often overexpressed in various types of cancer, including kidney, lung, brain, prostate, breast, pancreas, ovarian, and melanomas. When it binds to its endogenous ligand, CXCL12, it promotes cell survival, proliferation, and migration, crucial mechanisms for the retention of hematopoietic stem cells in the bone marrow and the movement of lymphocytes. The extensive expression of CXCR4 in cancer, coupled with the constant presence of CXCL12 in various organs, drives the activation of this axis, which in turn facilitates angiogenesis, tumor progression, and metastasis. Given the detrimental role of the CXCR4/CXCL12 axis, the search for drugs acting selectively against this protein represents an open challenge. This review aims to summarize the recent advancements in the design and development of CXCR4 antagonists as potential anticancer agents.
Collapse
Affiliation(s)
- Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Francesca Di Matteo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Roberta Castelluccio
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Valeria Napolitano
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Maria Rosaria Miranda
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy;
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy;
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| |
Collapse
|
2
|
Wang F, Ma J, Yang L, Hu P, Tang S, Wang J, Li Z. Discovery of novel CXCR4 inhibitors for the treatment of inflammation by virtual screening and biological evaluation. Eur J Med Chem 2024; 275:116605. [PMID: 38885550 DOI: 10.1016/j.ejmech.2024.116605] [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: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
C-X-C chemokine receptor type 4 (CXCR4) exerts considerable influence on the pathogenesis of inflammatory disorders and offers a potent avenue for drug intervention. This research utilizes a hybrid virtual screening methodology constructed using computer-aided drug design to discover novel CXCR4 inhibitors for the treatment of inflammation. First, a compound library was screened by Lipinski's five rules and adsorption, distribution, metabolism, excretion and toxicity properties. Second, the HypoGen algorithm was used in constructing a 3D-QSAR pharmacophore model and verify it layer by layer, and the obtained optimal pharmacophore 1 (Hypo 1) was used as a 3D query for compound screening. Then, hit compounds were obtained through molecular docking (Libdock and CDOCKER). The toxicity of the compounds to MDA-MB-231 cells was evaluated in vitro, and their binding affinity to the target was evaluated according to how they compete with 12G5 antibody for CXCR4 on the surfaces of the MDA-MB-231 cells. Compound Hit14 showed the strongest binding affinity among the hit compounds and inhibited cell migration and invasion in Matrigel invasion and wound healing assay at a concentration of 100 nM, demonstrating a better effect than AMD3100. Western Blot experiments further showed that Hit14 blocked the CXCR4/CXCL12-mediated phosphorylation of Akt. Meanwhile, cellular thermal displacement assay analysis showed that CXCR4 protein bound to Hit14 had high thermal stability. Finally, through in vivo experiments, we found that Hit14 inhibited mouse ear inflammation and reduced ear swelling and damage. Therefore, Hit14 is a promising drug for the further development of CXCR4 inhibitors for inflammation treatment.
Collapse
Affiliation(s)
- Fang Wang
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jie Ma
- The Central Hospital of Wuhan, Tongji Medical College of HUST, Wuhan, China
| | - Lili Yang
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ping Hu
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Siming Tang
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jing Wang
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Zeng Li
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Yang J, Tian E, Chen L, Liu Z, Ren Y, Mao W, Zhang Y, Zhang J. Development and therapeutic perspectives of CXCR4 antagonists for disease therapy. Eur J Med Chem 2024; 275:116594. [PMID: 38879970 DOI: 10.1016/j.ejmech.2024.116594] [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: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Chemokine receptor 4 (CXCR4) is a subtype receptor protein of the GPCR family with a seven-transmembrane structure widely distributed in human tissues. CXCR4 is involved in diseases (e.g., HIV-1 infection), cancer proliferation and metastasis, inflammation signaling pathways, and leukemia, making it a promising drug target. Clinical trials on CXCR4 antagonists mainly focused on peptides and antibodies, with a few small molecule compounds, such as AMD11070 (2) and MSX-122 (3), showing promise in cancer treatment. This perspective discusses the structure-activity relationship (SAR) of CXCR4 and its role in diseases, mainly focusing on the SAR of CXCR4 antagonists. It also explores the standard structural features and target interactions of CXCR4 binding in different disease categories. Furthermore, it investigates various modification strategies to propose potential improvements in the effectiveness of CXCR4 drugs.
Collapse
Affiliation(s)
- Jun Yang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Erkang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zihang Liu
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Wuyu Mao
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yiwen Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
4
|
Luo D, Bai Z, Bai H, Liu N, Han J, Ma C, Wu D, Bai L, Li Z. A first-in-class dimethyl 2-acetamido terephthalate inhibitor targeting Conyza canadensis SHMT1 with a novel herbicidal mode-of-action. J Adv Res 2024; 62:59-70. [PMID: 37820886 PMCID: PMC11331707 DOI: 10.1016/j.jare.2023.10.003] [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: 08/12/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023] Open
Abstract
INTRODUCTION Herbicide application is a highly efficiency method of weed control that boots agricultural output and assures food security. The development of novel herbicides focuses on improved bioactivity and new modes of action. The amino acid biosynthesis was validated as a promising novel mode of action for herbicidal compounds. However, the amino acid biosynthesis enzyme remains largely unexplored for herbicidal targets. OBJECTIVES Serine hydroxymethyl transferase (SHMT) is an essentialenzyme in the photorespiratory cycle. The study aims to explore Conyza canadensis SHMT1 (CcSHMT1) as a promising target for herbicide discovery. METHODS Structure determination of CcSHMT1 was resolved by X-ray crystallography. Virtual screening docking experiments were performed with Glide version 5.5. Novel derivatives of dimethyl 2-acetamido terephthalate were further designed, synthesized, and bioassay. The druggability of the inhibitor was evidenced by ultrastructural changes in mitochondria, in vivo and vitro enzyme activity assays, and genetics analysis. RESULTS CcSHMT1 has a typical PLP-dependent enzyme 3D structure. The dimethyl 2-acetamido terephthalate-containing compounds had herbicidal activity. Dimethyl 2-(2-(4-(2-(4-bromo-2-chlorophenoxy) acetyl)piperazin-1-yl)acetamido) terephthalate (Compound 9ay, EC50 = 193.8 g a.i./ ha) exhibited the highest herbicidal activity on tested weed among the synthesized compounds. Compound 9ay had no obvious adverse effect on the growth of maize and honeybees. Compound 9ay was verified to target CcSHMT1 as an herbicide candidate. CONCLUSION A first-in-class CcSHMT1 inhibitor that could be developed as a potent herbicide with a new mode of action and provide an avenue for discovering novel inhibitors of pyridoxal-5-phosphate-dependent enzymes.
Collapse
Affiliation(s)
- Dingfeng Luo
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China; Longping Branch, College of Biology, Hunan University, Changsha 410125, China
| | - Zhendong Bai
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Haodong Bai
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Na Liu
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China; Longping Branch, College of Biology, Hunan University, Changsha 410125, China
| | - Jincai Han
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China; Longping Branch, College of Biology, Hunan University, Changsha 410125, China
| | - Changsheng Ma
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China; Longping Branch, College of Biology, Hunan University, Changsha 410125, China
| | - Di Wu
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Lianyang Bai
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China; Longping Branch, College of Biology, Hunan University, Changsha 410125, China.
| | - Zuren Li
- Hunan Provincial Key Laboratory for Biology and Control of Weeds, Hunan Academy of Agricultural Sciences, Changsha 410125, China; Longping Branch, College of Biology, Hunan University, Changsha 410125, China.
| |
Collapse
|
5
|
Jiang X, Lu L, Li J, Jiang J, Zhang J, Zhou S, Wen H, Cai H, Luo X, Li Z, Wang J, Ju B, Bai R. Synthetically Feasible De Novo Molecular Design of Leads Based on a Reinforcement Learning Model: AI-Assisted Discovery of an Anti-IBD Lead Targeting CXCR4. J Med Chem 2024; 67:10057-10075. [PMID: 38863440 DOI: 10.1021/acs.jmedchem.4c00184] [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: 06/13/2024]
Abstract
Artificial intelligence (AI) de novo molecular generation provides leads with novel structures for drug discovery. However, the target affinity and synthesizability of the generated molecules present critical challenges for the successful application of AI technology. Therefore, we developed an advanced reinforcement learning model to bridge the gap between the theory of de novo molecular generation and the practical aspects of drug discovery. This model utilizes chemical reaction templates and commercially available building blocks as a starting point and employs forward reaction prediction to generate molecules, while real-time docking and drug-likeness predictions are conducted to ensure synthesizability and drug-likeness. We applied this model to design active molecules targeting the inflammation-related receptor CXCR4 and successfully prepared them according to the AI-proposed synthetic routes. Several molecules exhibited potent anti-CXCR4 and anti-inflammatory activity in subsequent in vitro and in vivo assays. The top-performing compound XVI alleviated symptoms related to inflammatory bowel disease and showed reasonable pharmacokinetic properties.
Collapse
Affiliation(s)
- Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Liuxin Lu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Junjie Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Jing Jiang
- SanOmics AI Co. Ltd., Hangzhou 311103, PR China
| | - Jiapeng Zhang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
| | - Shengbin Zhou
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, PR China
| | - Hao Wen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Hong Cai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Xinyu Luo
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Zhen Li
- SanOmics AI Co. Ltd., Hangzhou 311103, PR China
| | - Jiahui Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Bin Ju
- SanOmics AI Co. Ltd., Hangzhou 311103, PR China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| |
Collapse
|
6
|
Bai R, Jiang X, Hui Z, Yoon Y, Ge J, longZhu J, Shim H. Bisamide CXCR4 Modulators: Novel Anti‐IBD Agents Acting on the Chemotaxis of Inflammatory Cells. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Renren Bai
- School of Pharmacy Hangzhou Normal University Hangzhou Zhejiang 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Xiaoying Jiang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education Hangzhou Normal University Hangzhou 311121 P.R. China
| | - Zi Hui
- School of Pharmacy Hangzhou Normal University Hangzhou Zhejiang 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Younghyoun Yoon
- Department of Radiation Oncology, School of Medicine Emory University Atlanta GA 30322 USA
| | - Jiamin Ge
- School of Pharmacy Hangzhou Normal University Hangzhou Zhejiang 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Jun longZhu
- School of Pharmacy Hangzhou Normal University Hangzhou Zhejiang 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Hyunsuk Shim
- Department of Radiation Oncology, School of Medicine Emory University Atlanta GA 30322 USA
- Winship Cancer Institute Emory University Atlanta Georgia 30322 USA
| |
Collapse
|
7
|
Daoud S, Taha M. Ligand-based Modeling of CXC Chemokine Receptor 4 and Identification of Inhibitors of Novel Chemotypes as Potential Leads towards New Anti- COVID-19 Treatments. Med Chem 2022; 18:871-883. [PMID: 35040417 DOI: 10.2174/1573406418666220118153541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chemokines are involved in several human diseases and different stages of COVID-19 infection. They play a critical role in the pathophysiology of the associated acute respiratory disease syndrome, a major complication leading to death among COVID-19 patients. In particular, CXC chemokine receptor 4 (CXCR4) was found to be highly expressed in COVID-19 patients. METHODS We herein describe a computational workflow based on combining pharmacophore modeling and QSAR analysis towards the discovery of novel CXCR4 inhibitors. Subsequent virtual screening identified two promising CXCR4 inhibitors from the National Cancer Institute (NCI) list of compounds. The most active hit showed in vitro IC50 value of 24.4 μM. CONCLUSION These results proved the validity of the QSAR model and associated pharmacophore models as means to screen virtual databases for new CXCR4 inhibitors as leads for the development of new COVID-19 therapies.
Collapse
Affiliation(s)
- Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Sciences Private University, Amman, Jordan
| | - Mutasem Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| |
Collapse
|
8
|
Adlere I, Caspar B, Arimont M, Dekkers S, Visser K, Stuijt J, de Graaf C, Stocks M, Kellam B, Briddon S, Wijtmans M, de Esch I, Hill S, Leurs R. Modulators of CXCR4 and CXCR7/ACKR3 Function. Mol Pharmacol 2019; 96:737-752. [PMID: 31548340 DOI: 10.1124/mol.119.117663] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/14/2019] [Indexed: 02/14/2025] Open
Abstract
The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.
Collapse
Affiliation(s)
- Ilze Adlere
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Birgit Caspar
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Marta Arimont
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Sebastian Dekkers
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Kirsten Visser
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Jeffrey Stuijt
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Chris de Graaf
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Michael Stocks
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Barrie Kellam
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Stephen Briddon
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Maikel Wijtmans
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Iwan de Esch
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Stephen Hill
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Rob Leurs
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| |
Collapse
|
9
|
Guo Z, Jie X, Zhu P, Sun J, Gu J, Su F, Bai R, Xie Y. Fragmentation pathways of deprotonated amide-sulfonamide CXCR4 inhibitors investigated by ESI-IT-MS n , ESI-Q-TOF-MS/MS and DFT calculations. JOURNAL OF MASS SPECTROMETRY : JMS 2019; 54:869-877. [PMID: 31749257 DOI: 10.1002/jms.4439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 06/10/2023]
Abstract
Amide-sulfonamides provide a potent anti-inflammatory scaffold targeting the CXCR4 receptor. A series of novel amide-sulfonamide derivatives were investigated for their gas-phase fragmentation behaviors using electrospray ionization ion trap mass spectrometry and quadrupole time-of-flight mass spectrometry in negative ion mode. Upon collision-induced dissociation (CID), deprotonated amide-sulfonamides mainly underwent either an elimination of the amine to form the sulfonyl anion and amide anion or a benzoylamide derivative to provide sulfonamide anion bearing respective substituent groups. Based on the characteristic fragment ions and the deuterium-hydrogen exchange experiments, three possible fragmentation mechanisms corresponding to ion-neutral complexes including [sulfonyl anion/amine] complex (INC-1), [sulfonamide anion/benzoylamide derivative] complex (INC-2) and [amide anion/sulfonamide] complex (INC-3), respectively, were proposed. These three ion-neutral complexes might be produced by the cleavages of S-N and C-N bond from the amide-sulfonamides, which generated the sulfonyl anion (Route 1), sulfonamide anion (Route 2) and the amide anion (Route 3). DFT calculations suggested that Route 1, which generated the sulfonyl anion (ion c) is more favorable. In addition, the elimination of SO2 through a three-membered-ring transition state followed by the formation of C-N was observed for all the amide-sulfonamides.
Collapse
Affiliation(s)
- Zili Guo
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
| | - Xiaokang Jie
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Peixi Zhu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jian Sun
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jinping Gu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Feng Su
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Renren Bai
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yuanyuan Xie
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Zhejiang University of Technology, Hangzhou, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
10
|
Wu R, Yu W, Yao C, Liang Z, Yoon Y, Xie Y, Shim H, Bai R. Amide-sulfamide modulators as effective anti-tumor metastatic agents targeting CXCR4/CXCL12 axis. Eur J Med Chem 2019; 185:111823. [PMID: 31698158 DOI: 10.1016/j.ejmech.2019.111823] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/31/2022]
Abstract
Breast cancer is the most frequently diagnosed malignancy and the second common cause of death in women worldwide. High mortality in breast cancer is frequently associated with metastatic progression rather than the primary tumor itself. It has been recently identified that the CXCR4/CXCL12 axis plays a pivotal role in breast cancer metastasis, especially in directing metastatic cancer cells to CXCL12-riched organs and tissues. Herein, taking the amide-sulfamide as the lead structure, the second-round structural modifications to the sulfamide structure were performed to obtain more active CXCR4 modulators against tumor metastasis. Both in vivo and in vitro experiments illustrated that compound IIIe possessed potent CXCR4 binding affinity, excellent anti-metastatic and anti-angiogenetic activity against breast cancer. More importantly, in a mouse breast cancer lung metastasis model, compound IIIe exerted a significant inhibitory effect on breast cancer metastasis. Taken together, all these positive results demonstrated that developing of CXCR4 modulators is a promising strategy to mediate breast cancer metastasis.
Collapse
Affiliation(s)
- Rui Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Wenyan Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Chuansheng Yao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Zhongxing Liang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Younghyoun Yoon
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuanyuan Xie
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA; Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| | - Renren Bai
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
11
|
Development of CXCR4 modulators based on the lead compound RB-108. Eur J Med Chem 2019; 173:32-43. [PMID: 30981691 DOI: 10.1016/j.ejmech.2019.03.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/28/2019] [Accepted: 03/31/2019] [Indexed: 12/21/2022]
Abstract
The CXCR4/CXCL12 axis plays prominent roles in tumor metastasis and inflammation. CXCR4 has been shown to be involved in a variety of inflammation-related diseases. Therefore, CXCR4 is a promising potential target to develop novel anti-inflammatory agents. Taking our previously discovered CXCR4 modulator RB-108 as the lead compound, a series of derivatives were synthesized structurally modifying and optimizing the amide and sulfamide side chains. The derivatives successfully maintained potent CXCR4 binding affinity. Furthermore, compounds IIb, IIc, IIIg, IIIj, and IIIm were all efficacious in inhibiting the invasion of CXCR4-positive cells, displaying a much more potent effect than the lead compound RB-108. Notably, compound IIIm significantly decreased carrageenan-induced swollen volume and paw thickness in a mouse paw edema model. More importantly, IIIm exhibited satisfying PK profiles with a half-life of 4.77 h in an SD rat model. In summary, we have developed compound IIIm as a new candidate for further investigation based on the lead compound RB-108.
Collapse
|
12
|
Ilya E, Kulikova L, Van der Eycken EV, Voskressensky L. Recent Advances in Phthalan and Coumaran Chemistry. ChemistryOpen 2018; 7:914-929. [PMID: 30498677 PMCID: PMC6250979 DOI: 10.1002/open.201800184] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Indexed: 12/12/2022] Open
Abstract
Oxygen-containing heterocycles are common in biologically active compounds. In particular, phthalan and coumaran cores are found in pharmaceuticals, organic electronics, and other useful medical and technological applications. Recent research has expanded the methods available for their synthesis. This Minireview presents recent advances in the chemistry of phthalans and coumarans, with the goal of overcoming synthetic challenges and facilitating the applications of phthalans and coumarans.
Collapse
Affiliation(s)
- Efimov Ilya
- Peoples' Friendship University of Russia (RUDN University)6 Miklukho-Maklaya StreetMoscow117198Russia
| | - Larisa Kulikova
- Peoples' Friendship University of Russia (RUDN University)6 Miklukho-Maklaya StreetMoscow117198Russia
| | - Erik V. Van der Eycken
- Peoples' Friendship University of Russia (RUDN University)6 Miklukho-Maklaya StreetMoscow117198Russia
- Laboratory for Organic & Microwave-Assisted Chemistry (LOMAC)Department of ChemistryKU Leuven Celestijnenlaan 200F3001LeuvenBelgium
| | - Leonid Voskressensky
- Peoples' Friendship University of Russia (RUDN University)6 Miklukho-Maklaya StreetMoscow117198Russia
| |
Collapse
|
13
|
Abstract
The CXCR4/CXCL12 chemokine axis can chemotactically accumulate inflammatory cells to local tissues and regulate the release of inflammatory factors. Developing novel CXCR4 modulators may provide a desirable strategy to control the development of inflammation. A series of novel hybrids were designed by integrating the key pharmacophores of three CXCR4 modulators. The majority of compounds displayed potent CXCR4 binding affinity. Compound 7a exhibited 1000-fold greater affinity than AMD3100 and significantly inhibited invasion of CXCR4-positive tumor cells. Additionally, compound 7a blocked mice ear inflammation by 67% and suppressed the accumulation of inflammatory cells in an in vivo mouse ear edema evaluation. Western blot analyses revealed that 7a inhibited the CXCR4/CXCL12-mediated phosphorylation of Akt and p44 in a dose-dependent manner. Moreover, compound 7a had no observable cytotoxicity and displayed a favorable plasma stability in our preliminary pharmacokinetic study. These results confirmed that this is a feasible method to develop CXCR4 modulators for the regulation and reduction of inflammation.
Collapse
|
14
|
Peng D, Cao B, Zhou YJ, Long YQ. The chemical diversity and structure-based evolution of non-peptide CXCR4 antagonists with diverse therapeutic potential. Eur J Med Chem 2018; 149:148-169. [PMID: 29500940 DOI: 10.1016/j.ejmech.2018.02.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/11/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022]
Abstract
The CXC chemokine receptor 4 (CXCR4) is a highly reserved G-protein coupled 7-transmembrane (TM) chemokine receptor which consists of 352 amino acids. CXCR4 has only one endogenous chemokine ligand of CXCL12, besides several other natural nonchemokine ligands such as extracellular ubiquitin and noncognate ligand of MIF. CXCR4 strongly binds to CXCL12 and the resulting CXCLl2/CXCR4 axis is the molecular basis of their various biological functions, which include: (1) mediating immune and inflammatory response; (2) regulation of hematopoietic stem cell migration and homing; (3) an essential co-receptor for HIV entry into host cells; (4) participation in the process of embryonic development; (5) malignant tumor invasion and metastasis; (6) myocardial infarction, ischemic stroke and acute kidney injury. Correspondingly, CXCR4 antagonists find potential therapeutic applications in HIV infection, as well as hematopoietic stem cell migration, inflammation, immune-related diseases, tumor and ischemic diseases. Recently, great achievements have been made and a number of non-peptide CXCR4 antagonists with diversity scaffolds have been discovered. In this review, the discovery of small molecule CXCR4 antagonists focused on the structures, activities, evolution and development of representative CXCR4 antagonists is comprehensively described. The central role of CXCR4 in diverse cellular signaling pathways and its involvement in several diseases progressions are discussed as well.
Collapse
Affiliation(s)
- Dian Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bin Cao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying-Jun Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Ya-Qiu Long
- College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou 215123, China.
| |
Collapse
|
15
|
Abdelhamid A, Jouini M, Bel Haj Amor H, Mzoughi Z, Dridi M, Ben Said R, Bouraoui A. Phytochemical Analysis and Evaluation of the Antioxidant, Anti-Inflammatory, and Antinociceptive Potential of Phlorotannin-Rich Fractions from Three Mediterranean Brown Seaweeds. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2018; 20:60-74. [PMID: 29344826 DOI: 10.1007/s10126-017-9787-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/27/2017] [Indexed: 06/07/2023]
Abstract
Phlorotannins, phenolic compounds produced exclusively by seaweeds, have been reported to possess various pharmacological properties. However, there have been few works on these compounds from Mediterranean seaweeds. In this study, we investigated the phytochemical analysis and pharmacological potential of phlorotannin-rich fractions from three brown seaweeds collected along the Tunisia coast: Cystoseira sedoides (PHT-SED), Cladostephus spongeosis (PHT-CLAD), and Padina pavonica (PHT-PAD). Phytochemical determinations showed considerable differences in total phenolic content (TPC) and phlorotannin content (PHT). The highest TPC level (26.45 mg PGE/g dry material (Dm)) and PHT level (873.14 μg PGE/g Dm) were observed in C. sedoides. The antioxidant properties of these three fractions assessed by three different methods indicated that C. sedoides displayed the highest total antioxidant activity among the three species (71.30 mg GAE/g Dm), as well as the free radical scavenging activity with the lowest IC50 value in both DPPH (27.7 μg/mL) and ABTS (19.1 μg/mL) assays. Furthermore, the pharmacological screening of the anti-inflammatory potential of these fractions using in vivo models, in comparison to reference drugs, established a remarkable activity of PHT-SED at the dose of 100 mg/kg; the inhibition percentages of ear edema in mice model and paw edema in rats model were of 82.55 and 81.08%, respectively. The content of malondialdehyde (MDA) in liver tissues has been quantified, and PHT-SED was found to remarkably increase the lipid peroxidation in rat liver tissues. In addition, in two pain mice models, PHT-SED displayed a profound antinociceptive activity at 100 mg/kg and has proved a better analgesic activity when used in combination with the opioid drug, tramadol.
Collapse
Affiliation(s)
- Amal Abdelhamid
- Laboratory of Chemical, Galenic and Pharmacological Development of Drugs, Faculty of Pharmacy of Monastir, University of Monastir, 5000, Monastir, Tunisia.
| | - Meriem Jouini
- Laboratory of Heterocyclic Chemistry, Natural Products and Reactivity, Medicinal Chemistry and Natural Products Team, Faculty of Sciences of Monastir, University of Monastir, 5019, Monastir, Tunisia
| | - Haifa Bel Haj Amor
- Laboratory of Chemical, Galenic and Pharmacological Development of Drugs, Faculty of Pharmacy of Monastir, University of Monastir, 5000, Monastir, Tunisia
| | - Zeineb Mzoughi
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences of Monastir, University of Monastir, 5000, Monastir, Tunisia
| | - Mehdi Dridi
- Laboratory of Chemical, Galenic and Pharmacological Development of Drugs, Faculty of Pharmacy of Monastir, University of Monastir, 5000, Monastir, Tunisia
| | - Rafik Ben Said
- National Institute of Marine Sciences and Technologies, Salambôo, Tunis, Tunisia
| | - Abderrahman Bouraoui
- Laboratory of Chemical, Galenic and Pharmacological Development of Drugs, Faculty of Pharmacy of Monastir, University of Monastir, 5000, Monastir, Tunisia
| |
Collapse
|
16
|
The benefits of in silico modeling to identify possible small-molecule drugs and their off-target interactions. Future Med Chem 2018; 10:423-432. [PMID: 29380627 DOI: 10.4155/fmc-2017-0151] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The research into the use of small molecules as drugs continues to be a key driver in the development of molecular databases, computer-aided drug design software and collaborative platforms. The evolution of computational approaches is driven by the essential criteria that a drug molecule has to fulfill, from the affinity to targets to minimal side effects while having adequate absorption, distribution, metabolism, and excretion (ADME) properties. A combination of ligand- and structure-based drug development approaches is already used to obtain consensus predictions of small molecule activities and their off-target interactions. Further integration of these methods into easy-to-use workflows informed by systems biology could realize the full potential of available data in the drug discovery and reduce the attrition of drug candidates.
Collapse
|
17
|
Grande F, Giancotti G, Ioele G, Occhiuzzi MA, Garofalo A. An update on small molecules targeting CXCR4 as starting points for the development of anti-cancer therapeutics. Eur J Med Chem 2017; 139:519-530. [PMID: 28826086 DOI: 10.1016/j.ejmech.2017.08.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/19/2022]
Abstract
CXCR4 (C-X-C Chemokine Receptor type 4) and its natural ligand SDF-1α (Stromal-Derived-Factor-1α) are involved in a number of physiological and pathological processes including cancer spread and progression. Over the past few years, numerous CXCR4 antagonists have been identified and currently are in different development stages as potential agents for the treatment of several diseases involving the CXCR4/SDF-1α axis. Herein, we focus on small molecules reported in literature between 2013 and 2017, claimed as CXCR4 antagonists and potentially useful in the treatment of cancer and other diseases where this receptor is involved. Most of the compounds resulted from a chemical optimization of previously identified molecules and some of them could represent suitable candidates for the development of advanced anticancer agents.
Collapse
Affiliation(s)
- Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Gilda Giancotti
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Giuseppina Ioele
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy
| | - Maria A Occhiuzzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy
| | - Antonio Garofalo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy
| |
Collapse
|
18
|
Bai R, Liang Z, Yoon Y, Salgado E, Feng A, Gurbani S, Shim H. Novel anti-inflammatory agents targeting CXCR4: Design, synthesis, biological evaluation and preliminary pharmacokinetic study. Eur J Med Chem 2017; 136:360-371. [PMID: 28521261 DOI: 10.1016/j.ejmech.2017.05.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/25/2022]
Abstract
CXCR4 plays a crucial role in the inflammatory disease process, providing an attractive means for drug targeting. A series of novel amide-sulfamide derivatives were designed, synthesized and comprehensively evaluated. This new scaffold exhibited much more potent CXCR4 inhibitory activity, with more than 70% of the compounds showed notably better binding affinity than the reference drug AMD3100 in the binding assay. Additionally, in the Matrigel invasion assay, most of our compounds significantly blocked the tumor cell invasion, demonstrating superior efficacy compared to AMD3100. Furthermore, compound IIj blocked mice ear inflammation by 75% and attenuated ear edema and damage substantially in an in vivo model of inflammation. Western blot analyses revealed that CXCR4 modulator IIj significantly blocked CXCR4/CXCL12-mediated phosphorylation of Akt. Moreover, compound IIj had no observable cytotoxicity and displayed a favourable plasma stability in our preliminary pharmacokinetic study. The preliminary structure-activity relationships were also summarized. In short, this novel amide-sulfamide scaffold exhibited potent CXCR4 inhibitory activity both in vitro and in vivo. These results also confirmed that developing modulators targeting CXCR4 provides an exciting avenue for treatment of inflammation.
Collapse
Affiliation(s)
- Renren Bai
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Zhongxing Liang
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Younghyoun Yoon
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Eric Salgado
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Amber Feng
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Saumya Gurbani
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, School of Medicine, Emory University, Atlanta, GA, USA; Winship Cancer Institute, Emory University, Atlanta, GA, USA; Department of Radiology and Imaging Science, School of Medicine, Emory University, Atlanta, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA.
| |
Collapse
|