1
|
Li Y, Zhang Y, Zhang J, Zhan Z, Mao W. Development of novel focal adhesion kinase (FAK) inhibitors for targeting cancer: Structural insights and therapeutic potential. Eur J Med Chem 2024; 279:116913. [PMID: 39357313 DOI: 10.1016/j.ejmech.2024.116913] [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: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase frequently overexpressed in various cancer cells, facilitating tumor growth through the regulation of cell adhesion, migration, and proliferation. Consequently, targeting FAK is considered a promising anti-tumor strategy, particularly for invasive cancers. Numerous potent small-molecule inhibitors have progressed to clinical trials. Among these, Defactinib is under evaluation for regulatory approval as a treatment for ovarian serous tumors. Furthermore, novel FAK inhibitors, including PROTACs, have emerged as key research focuses, anticipated to overcome the limitations of traditional inhibitors. In this Perspective, we highlight the protein structure, biological functions, relevant signaling pathways, and associations of FAK with cancer development. We also analyze the clinical status of FAK inhibitors, paying special attention to the various classes of FAK inhibitors, with detailed analyses of their chemical structures, structure-activity relationships (SARs), bioactivity profiles, selectivity profiles, and therapeutic potentials.
Collapse
Affiliation(s)
- Yingnan Li
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China
| | - Yuming Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China; West China College of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China
| | - Zixuan Zhan
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China.
| | - Wuyu Mao
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Neuro-system and Multimorbidity Laboratory, State Key Laboratory of Biotherapy and Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, 610041, Sichuan, China.
| |
Collapse
|
2
|
Chen MT, Huang TH, Yang FA, Chen BH. Structural tuning enhanced catalytic activity of amido aluminum complexes for the ring-opening polymerization of ε-caprolactone. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2022.122493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
3
|
Yang F, Xu K, Zhang S, Zhang J, Qiu Y, Luo J, Tan G, Zou Z, Wang W, Kang F. Discovery of novel chloropyramine-cinnamic acid hybrids as potential FAK inhibitors for intervention of metastatic triple-negative breast cancer. Bioorg Med Chem 2022; 66:116809. [PMID: 35569251 DOI: 10.1016/j.bmc.2022.116809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/28/2022]
Abstract
To search for novel focal adhesion kinase (FAK) inhibitors for intervention of metastatic triple-negative breast cancer (TNBC), a series of hybrids 7a-s from chloropyramine and cinnamic acid analogs were designed, synthesized and biologically evaluated. The most active compound 7d could potently inhibit the proliferation, invasion and migration of TNBC cells in vitro. The docking analysis of 7d was performed to elucidate its possible binding modes to focal adhesion targeting (FAT) domain of FAK scaffold. Further mechanism studies indicated the ability of 7d in disrupting Y925 autophosphorylation of FAK, reducing formation of focal adhesions (FAs) and stress fibers (SFs) as well as inducing apoptosis of TNBC cells. Together, 7d is a novel FAK inhibitor to inhibit the essential nonkinase scaffolding function of FAK via binding FAT domain and may be worth studying further for intervention of TNBC.
Collapse
Affiliation(s)
- Fei Yang
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Kangping Xu
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Sha Zhang
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Jinlin Zhang
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Yaoren Qiu
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Jin Luo
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Guishan Tan
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Zhenxing Zou
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China
| | - Wenxuan Wang
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China.
| | - Fenghua Kang
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Central South University, Changsha, PR China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, PR China.
| |
Collapse
|
4
|
Wu X, Wang J, Liang Q, Tong R, Huang J, Yang X, Xu Y, Wang W, Sun M, Shi J. Recent progress on FAK inhibitors with dual targeting capabilities for cancer treatment. Biomed Pharmacother 2022; 151:113116. [PMID: 35598365 DOI: 10.1016/j.biopha.2022.113116] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/30/2022] [Accepted: 05/10/2022] [Indexed: 02/08/2023] Open
Abstract
Focal adhesion kinase (FAK, also known as PTK2) is a tyrosine kinase that regulates integrin and growth factor signaling pathways and is involved in the migration, proliferation and survival of cancer cells. FAK is a promising target for cancer treatment. Many small molecule FAK inhibitors have been identified and proven in both preclinical and clinical studies to be effective inhibitors of tumor growth and metastasis. There are many signaling pathways, such as those involving FAK, Src, AKT, MAPK, PI3K, and EGFR/HER-2, that provide survival signals in cancer cells. Dual inhibitors that simultaneously block FAK and another factor can significantly improve efficacy and overcome some of the shortcomings of single-target inhibitors, including drug resistance. In this review, the antitumor mechanisms and research status of dual inhibitors of FAK and other targets, such as Pyk2, IGF-IR, ALK, VEGFR-3, JAK2, EGFR, S6K1, and HDAC2, are summarized, providing new ideas for the development of effective FAK dual-target preparations.
Collapse
Affiliation(s)
- Xianbo Wu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 610041, China
| | - Jie Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550002, China
| | - Qi Liang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Jianli Huang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550002, China
| | - Xinwei Yang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 610041, China
| | - Yihua Xu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Wenjing Wang
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Minghan Sun
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
5
|
Shen D, Zeng Y, Zhang W, Li Y, Zhu J, Liu Z, Yan Z, Huang JA. Chenodeoxycholic acid inhibits lung adenocarcinoma progression via the integrin α5β1/FAK/p53 signaling pathway. Eur J Pharmacol 2022; 923:174925. [PMID: 35364069 DOI: 10.1016/j.ejphar.2022.174925] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-associated death worldwide and is classified into non-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). NSCLC accounts for approximately 80%-85% of all lung cancer cases. Chenodeoxycholic acid (CDCA), a primary bile acid, has been reported to inhibit carcinoma cell proliferation. Here, we aimed to determine the effects and mechanism of action of CDCA against lung adenocarcinoma (LUAD). METHODS Western blotting and quantitative real-time polymerase chain reaction were used to evaluate the protein and mRNA expression levels in LUAD cell lines, respectively. Cell Counting Kit-8 and clone formation assays were performed to evaluate the proliferation ability of different cell types in vitro. Tumor cell motility was evaluated using Transwell assays. The transcriptional profile of A549 cells treated with CDCA was determined through RNA sequencing analysis. A xenograft model was established to evaluate the effects of CDCA on LUAD progression in vivo. RESULTS CDCA inhibited LUAD cell proliferation, migration, and invasion. Furthermore, it promoted apoptosis in LUAD cells. Mechanistically, CDCA inhibited the integrin α5β1 signaling pathway in LUAD cells by inhibiting the expression of the α5 and β1 subunits of integrin and phosphorylated FAK. Moreover, CDCA induced an increase in the levels of p53, a downstream gene of the integrin α5β1/FAK pathway. In addition, CDCA significantly decreased tumor volume in mice without inducing significant toxicity. CONCLUSIONS Our findings indicate that CDCA attenuates LUAD pathogenesis in vitro and in vivo via the integrin α5β1/FAK/p53 axis.
Collapse
Affiliation(s)
- Dan Shen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Weijie Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Yue Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China.
| | - Zhaowei Yan
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Jian-An Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China.
| |
Collapse
|
6
|
Wu X, Zhang Q, Guo Y, Zhang H, Guo X, You Q, Wang L. Methods for the Discovery and Identification of Small Molecules Targeting Oxidative Stress-Related Protein–Protein Interactions: An Update. Antioxidants (Basel) 2022; 11:antiox11040619. [PMID: 35453304 PMCID: PMC9025695 DOI: 10.3390/antiox11040619] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
The oxidative stress response pathway is one of the hotspots of current pharmaceutical research. Many proteins involved in these pathways work through protein–protein interactions (PPIs). Hence, targeting PPI to develop drugs for an oxidative stress response is a promising strategy. In recent years, small molecules targeting protein–protein interactions (PPIs), which provide efficient methods for drug discovery, are being investigated by an increasing number of studies. However, unlike the enzyme–ligand binding mode, PPIs usually exhibit large and dynamic binding interfaces, which raise additional challenges for the discovery and optimization of small molecules and for the biochemical techniques used to screen compounds and study structure–activity relationships (SARs). Currently, multiple types of PPIs have been clustered into different classes, which make it difficult to design stationary methods for small molecules. Deficient experimental methods are plaguing medicinal chemists and are becoming a major challenge in the discovery of PPI inhibitors. In this review, we present current methods that are specifically used in the discovery and identification of small molecules that target oxidative stress-related PPIs, including proximity-based, affinity-based, competition-based, structure-guided, and function-based methods. Our aim is to introduce feasible methods and their characteristics that are implemented in the discovery of small molecules for different types of PPIs. For each of these methods, we highlight successful examples of PPI inhibitors associated with oxidative stress to illustrate the strategies and provide insights for further design.
Collapse
Affiliation(s)
- Xuexuan Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuqi Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hengheng Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; (X.W.); (Q.Z.); (Y.G.); (H.Z.)
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (X.G.); (Q.Y.); (L.W.); Tel.: +86-025-83271351 (Q.Y.); +86-15261483858 (L.W.)
| |
Collapse
|
7
|
Wang Z, Huang W, Zhou K, Ren X, Ding K. Targeting the Non-Catalytic Functions: a New Paradigm for Kinase Drug Discovery? J Med Chem 2022; 65:1735-1748. [PMID: 35000385 DOI: 10.1021/acs.jmedchem.1c01978] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein kinases have been highly fruitful targets for cancer drug discovery in the past two decades, while most of these drugs bind to the "adenosine triphosphate (ATP)-site" and inhibit kinase catalytic activity. Recently, accumulated evidence suggests that kinases possess functions beyond catalysis through their scaffolds, and the scaffolding functions could play critical roles in multiple cellular signaling and cell fate controls. Small molecules modulating the noncatalytic functions of kinases are rarely reported but emerge as new promising therapeutic strategies for various diseases. Herein, we summarize the characterized noncatalytic functions of kinases, and highlight the recent progress on developing small-molecule modulators of the noncatalytic functions of kinases. Mechanisms and characteristics of different kinds of modulators are also discussed. It is also speculated that targeting the noncatalytic functions would represent a new direction for kinase-based drug discovery.
Collapse
Affiliation(s)
- Zhen Wang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Ling Ling Road, Shanghai 200032, People's Republic of China
| | - Weixue Huang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Ling Ling Road, Shanghai 200032, People's Republic of China
| | - Kaijie Zhou
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Ling Ling Road, Shanghai 200032, People's Republic of China
| | - Xiaomei Ren
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of People's Republic of China, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, People's Republic of China
| | - Ke Ding
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Ling Ling Road, Shanghai 200032, People's Republic of China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of People's Republic of China, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, People's Republic of China.,The First Affiliated Hospital (Huaqiao Hospital), Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, People's Republic of China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, People's Republic of China
| |
Collapse
|
8
|
Abstract
FAK, a nonreceptor tyrosine kinase, has been recognized as a novel target class for the development of targeted anticancer agents. Overexpression of FAK is a common occurrence in several solid tumors, in which the kinase has been implicated in promoting metastases. Consequently, designing and developing potent FAK inhibitors is becoming an attractive goal, and FAK inhibitors are being recognized as a promising tool in our armamentarium for treating diverse cancers. This review comprehensively summarizes the different classes of synthetically derived compounds that have been reported as potent FAK inhibitors in the last three decades. Finally, the future of FAK-targeting smart drugs that are designed to slow down the emergence of drug resistance is discussed.
Collapse
|
9
|
Mousson A, Legrand M, Steffan T, Vauchelles R, Carl P, Gies JP, Lehmann M, Zuber G, De Mey J, Dujardin D, Sick E, Rondé P. Inhibiting FAK-Paxillin Interaction Reduces Migration and Invadopodia-Mediated Matrix Degradation in Metastatic Melanoma Cells. Cancers (Basel) 2021; 13:cancers13081871. [PMID: 33919725 PMCID: PMC8070677 DOI: 10.3390/cancers13081871] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 01/09/2023] Open
Abstract
Simple Summary The focal adhesion kinase (FAK) is over-expressed in a variety of human tumors and is involved in many aspects of the metastatic process. This has led to the development of small inhibitors of FAK kinase function which are currently evaluated in clinical trials. We demonstrate here that this class of inhibitors, while decreasing melanoma cell migration, increases invadopodia activity in metastatic melanoma cells. Searching for an alternative strategy to inhibit the oncogenic activity of FAK, we show that inhibiting FAK scaffolding function using a small peptide altering FAK–paxillin interactions reduces both migration and invadopodia-mediated matrix degradation in metastatic melanoma cells. Abstract The nonreceptor tyrosine kinase FAK is a promising target for solid tumor treatment because it promotes invasion, tumor progression, and drug resistance when overexpressed. Investigating the role of FAK in human melanoma cells, we found that both in situ and metastatic melanoma cells strongly express FAK, where it controls tumor cells’ invasiveness by regulating focal adhesion-mediated cell motility. Inhibiting FAK in human metastatic melanoma cells with either siRNA or a small inhibitor targeting the kinase domain impaired migration but led to increased invadopodia formation and extracellular matrix degradation. Using FAK mutated at Y397, we found that this unexpected increase in invadopodia activity is due to the lack of phosphorylation at this residue. To preserve FAK–Src interaction while inhibiting pro-migratory functions of FAK, we found that altering FAK–paxillin interaction, with either FAK mutation in the focal adhesion targeting (FAT) domain or a competitive inhibitor peptide mimicking paxillin LD domains drastically reduces cell migration and matrix degradation by preserving FAK activity in the cytoplasm. In conclusion, our data show that targeting FAK–paxillin interactions could be a potential therapeutic strategy to prevent metastasis formation, and molecules targeting this interface could be alternative to inhibitors of FAK kinase activity which display unexpected effects.
Collapse
Affiliation(s)
- Antoine Mousson
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Marlène Legrand
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Tania Steffan
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Romain Vauchelles
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Plateforme PIQ, Faculté de Pharmacie, 67401 Illkirch, France;
| | - Philippe Carl
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Jean-Pierre Gies
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Maxime Lehmann
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Guy Zuber
- Université de Strasbourg, CNRS UMR7242, Intervention Chémobiologique, ESBS, 67412 Illkirch, France;
| | - Jan De Mey
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Denis Dujardin
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Emilie Sick
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
| | - Philippe Rondé
- Université de Strasbourg, CNRS UMR7021, Laboratoire de Bioimagerie et Pathologies, Migration, Invasion et Microenvironnement, Faculté de Pharmacie, 67401 Illkirch, France; (A.M.); (M.L.); (T.S.); (P.C.); (J.-P.G.); (M.L.); (J.D.M.); (D.D.); (E.S.)
- Correspondence: ; Tel.: +33-3-6885-4184
| |
Collapse
|
10
|
Lu Y, Sun H. Progress in the Development of Small Molecular Inhibitors of Focal Adhesion Kinase (FAK). J Med Chem 2020; 63:14382-14403. [PMID: 33058670 DOI: 10.1021/acs.jmedchem.0c01248] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Focal adhesion kinase (FAK) is a nonreceptor intracellular tyrosine kinase that plays an essential role in cancer cell adhesion, survival, proliferation, and migration through both its enzymatic activities and scaffolding functions. Overexpression of FAK has been found in many human cancer cells from different origins, which promotes tumor progression and influences clinical outcomes in different classes of human tumors. Therefore, FAK has been considered as a promising target for small molecule anticancer drug development. Many FAK inhibitors targeting different domains of FAK with various mechanisms of functions have been reported, including kinase domain inhibitors, FERM domain inhibitors, and FAT domain inhibitors. In addition, FAK-targeting PROTACs, which can induce the degradation of FAK, have also been developed. In this Perspective, we summarized the progress in the development of small molecular FAK inhibitors and proposed the perspectives for the future development of agents targeting FAK.
Collapse
Affiliation(s)
- Yang Lu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Haiying Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| |
Collapse
|
11
|
Kandil SB, Jones SR, Smith S, Hiscox SE, Westwell AD. Structure-Based Virtual Screening, Synthesis and Biological Evaluation of Potential FAK-FAT Domain Inhibitors for Treatment of Metastatic Cancer. Molecules 2020; 25:molecules25153488. [PMID: 32751931 PMCID: PMC7435868 DOI: 10.3390/molecules25153488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 11/16/2022] Open
Abstract
Focal adhesion kinase (FAK) is a tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. In cancer cells, FAK promotes the progression and metastasis of tumours. In this study, we used structure-based virtual screening to filter a library of more than 210K compounds against the focal adhesion targeting FAK-focal adhesion targeting (FAT) domain to identify 25 virtual hit compounds which were screened in the invasive breast cancer line (MDA-MB-231). Most notably, compound I showed low micromolar antiproliferative activity, as well as antimigratory activity. Moreover, examination in a model of triple negative breast cancer (TNBC), revealed that, despite not effecting FAK phosphorylation, compound I significantly impairs proliferation whilst impairing focal adhesion growth and turnover leading to reduced migration. Further optimisation and synthesis of analogues of the lead compound I using a four-step synthetic procedure was performed, and analogues were assessed for their antiproliferative activity against three breast cancer (MDA-MB-231, T47D, BT474) cell lines and one pancreatic cancer (MIAPaCa2) cell line. Compound 5f was identified as a promising lead compound with IC50 values in the range of 4.59–5.28 μM in MDA-MB-231, T47D, BT474, and MIAPaCa2. Molecular modelling and pharmacokinetic studies provided more insight into the therapeutic features of this new series.
Collapse
|
12
|
Cavin S, Riedel T, Rosskopfova P, Gonzalez M, Baldini G, Zellweger M, Wagnières G, Dyson PJ, Ris H, Krueger T, Perentes JY. Vascular‐targeted low dose photodynamic therapy stabilizes tumor vessels by modulating pericyte contractility. Lasers Surg Med 2019; 51:550-561. [DOI: 10.1002/lsm.23069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Sabrina Cavin
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Tina Riedel
- Institute of Chemical Sciences and EngineeringSwiss Federal Institute of Technology (EPFL)LausanneSwitzerland
| | - Petra Rosskopfova
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Michel Gonzalez
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Greg Baldini
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Matthieu Zellweger
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Georges Wagnières
- Institute of PhysicsSwiss Federal Institute of Technology (EPFL)LausanneSwitzerland
| | - Paul J. Dyson
- Institute of Chemical Sciences and EngineeringSwiss Federal Institute of Technology (EPFL)LausanneSwitzerland
| | - Hans‐Beat Ris
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Thorsten Krueger
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Jean Y. Perentes
- Department of Thoracic SurgeryCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| |
Collapse
|
13
|
Chi Q, Wang L, Xie D, Wang X. Characterization of in vitro metabolism of focal adhesion kinase inhibitors by LC/MS/MS. J Pharm Biomed Anal 2019; 168:163-173. [PMID: 30807921 DOI: 10.1016/j.jpba.2019.02.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/14/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is critically involved in cell migration, spreading and proliferation at the early step of various cancers. Small molecule inhibitors of FAK are effective to inhibit its activation in the process of tumor formation in cell. To better understand biotransformation of FAK inhibitors, this work has investigated in vitro phase I metabolism of inhibitors (namely PF-573228, PF-562271 and PF-03814735) by rat liver microsomes model. Using liquid chromatography - quadrupole time of flight mass spectrometry and tandem mass spectrometry (LC/Q-TOF/MS and MS/MS), three metabolites of PF-573228 and PF-562271 were observed and characterized, respectively. These in vitro metabolites were reported for the first time. The structures and fragmentation patterns of these metabolites were elucidated, and phase I metabolic pathways for FAK inhibitors were proposed. The main metabolic pathways of PF-573228 were hydroxylation, dehydrogenation and N-dealkylation. For PF-562271, they were hydroxylation and dehydrogenation. Hydroxylation was observed as the primary metabolism for PF-0381473.
Collapse
Affiliation(s)
- Quan Chi
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China
| | - Ling Wang
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China
| | - Dong Xie
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China
| | - Xian Wang
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan 430074, PR China.
| |
Collapse
|
14
|
Chen MT, Chen YY, Li GL, Chen CT. Diverse Coordinative Zinc Complexes Containing Amido-Pyridinate Ligands: Structural and Catalytic Studies. Front Chem 2019; 6:615. [PMID: 30662891 PMCID: PMC6328485 DOI: 10.3389/fchem.2018.00615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/28/2018] [Indexed: 11/25/2022] Open
Abstract
In this work, zinc complexes containing amidopyridinate ligands substituted with different pendant arms have been described. Treatment of ligand precursors with ZnEt2 at a 1:1 ratio in THF yields zinc ethyl complexes (PyNC1Py)2(ZnEt)2 (1) and (PyNC2NMe2)2(ZnEt)2 (2), respectively. Complexes 1 and 2 show the same geometry as a distorted tetrahedron, but adopt different coordination behaviors supported by the ligands. Complex 1 represents a rare and a non-centrosymmetric mode, which the amido group bridges two zinc centers to form a six-membered ring. However, complex 2 shows a centrosymmetric mode, which the pyridine group links to the zinc centers to form an eight-membered ring. Recrystallization of complex 2 gives an additional complex (PyNC2NMe2)4Zn3(μ3-O) (3). We attempted to prepare zinc benzyl oxide complexes but afforded only a self-assembly cubane complex Zn7Et6(OBn)8(4). All molecular structures 1–4 are characterized depending on both single-crystal X-ray and spectroscopic data. Furthermore, their catalytic properties toward the ring opening polymerization of ε-caprolactone and L-lactide, using benzyl alcohol as the initiation reagent, are under investigation.
Collapse
Affiliation(s)
- Ming-Tsz Chen
- Department of Applied Chemistry, Providence University, Taichung, Taiwan
| | - Yu-Yang Chen
- Department of Applied Chemistry, Providence University, Taichung, Taiwan
| | - Guan-Lin Li
- Department of Applied Chemistry, Providence University, Taichung, Taiwan
| | - Chi-Tien Chen
- Department of Chemistry, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
15
|
Targeting Focal Adhesion Kinase Using Inhibitors of Protein-Protein Interactions. Cancers (Basel) 2018; 10:cancers10090278. [PMID: 30134553 PMCID: PMC6162372 DOI: 10.3390/cancers10090278] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/19/2022] Open
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic non-receptor protein tyrosine kinase that is overexpressed and activated in many human cancers. FAK transmits signals to a wide range of targets through both kinase-dependant and independent mechanism thereby playing essential roles in cell survival, proliferation, migration and invasion. In the past years, small molecules that inhibit FAK kinase function have been developed and show reduced cancer progression and metastasis in several preclinical models. Clinical trials have been conducted and these molecules display limited adverse effect in patients. FAK contain multiple functional domains and thus exhibit both important scaffolding functions. In this review, we describe the major FAK interactions relevant in cancer signalling and discuss how such knowledge provide rational for the development of Protein-Protein Interactions (PPI) inhibitors.
Collapse
|
16
|
Kandil S, Prencipe F, Jones S, Hiscox S, Westwell AD. The discovery of new and more potent chloropyramine (C4) analogues for the potential treatment of invasive breast cancer. Chem Biol Drug Des 2017; 91:314-321. [PMID: 28816016 DOI: 10.1111/cbdd.13083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 12/13/2022]
Abstract
Breast cancer is the second most common cancer worldwide, accounting for 25% of all female cancers. Although the survival rate has increased significantly in the past few decades, patients who develop secondary site metastasis as well as those diagnosed with triple negative breast cancer still represent a real unmet medical challenge. Previous studies have shown that chloropyramine (C4) inhibits FAK-VEGFR3 signalling. More recently, C4 is reported to have SASH1 inducing properties. However, C4 exerts its antitumour and antiangiogenic effects at high micromolar concentrations (>100 μm) that would not be compatible with further drug development against invasive breast cancer driven by FAK signalling. In this study, molecular modelling guided structural modifications have been introduced to the chloropyramine C4 scaffold to improve its activity in breast cancer cell lines. Seventeen compounds were designed and synthesized, and their antiproliferative activity was evaluated against three human breast cancer lines (MDA-MB-231, BT474 and T47D). Compound 5c was identified to display an average activity of IC50 = 23.5-31.3 μm, which represents a significant improvement of C4 activity in the same assay model. Molecular modelling and pharmacokinetic studies provided more promising insights into the mechanistic features of this new series.
Collapse
Affiliation(s)
- Sahar Kandil
- School of Pharmacy & Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, UK
| | - Filippo Prencipe
- School of Pharmacy & Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, UK
| | - Samuel Jones
- School of Pharmacy & Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, UK
| | - Stephen Hiscox
- School of Pharmacy & Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, UK
| | - Andrew D Westwell
- School of Pharmacy & Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
17
|
Zarei O, Hamzeh-Mivehroud M, Benvenuti S, Ustun-Alkan F, Dastmalchi S. Characterizing the Hot Spots Involved in RON-MSPβ Complex Formation Using In Silico Alanine Scanning Mutagenesis and Molecular Dynamics Simulation. Adv Pharm Bull 2017; 7:141-150. [PMID: 28507948 PMCID: PMC5426727 DOI: 10.15171/apb.2017.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 12/30/2022] Open
Abstract
Purpose: Implication of protein-protein interactions (PPIs) in development of many diseases such as cancer makes them attractive for therapeutic intervention and rational drug design. RON (Recepteur d'Origine Nantais) tyrosine kinase receptor has gained considerable attention as promising target in cancer therapy. The activation of RON via its ligand, macrophage stimulation protein (MSP) is the most common mechanism of activation for this receptor. The aim of the current study was to perform in silico alanine scanning mutagenesis and to calculate binding energy for prediction of hot spots in protein-protein interface between RON and MSPβ chain (MSPβ). Methods: In this work the residues at the interface of RON-MSPβ complex were mutated to alanine and then molecular dynamics simulation was used to calculate binding free energy. Results: The results revealed that Gln193, Arg220, Glu287, Pro288, Glu289, and His424 residues from RON and Arg521, His528, Ser565, Glu658, and Arg683 from MSPβ may play important roles in protein-protein interaction between RON and MSP. Conclusion: Identification of these RON hot spots is important in designing anti-RON drugs when the aim is to disrupt RON-MSP interaction. In the same way, the acquired information regarding the critical amino acids of MSPβ can be used in the process of rational drug design for developing MSP antagonizing agents, the development of novel MSP mimicking peptides where inhibition of RON activation is required, and the design of experimental site directed mutagenesis studies.
Collapse
Affiliation(s)
- Omid Zarei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Benvenuti
- Molecular Therapeutics and Exploratory Research Laboratory, Candiolo Cancer Institute-FPO-IRCCS, Candiolo, Turin, Italy
| | - Fulya Ustun-Alkan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, Turkey
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Burgess JT, Bolderson E, Saunus JM, Zhang SD, Reid LE, McNicol AM, Lakhani SR, Cuff K, Richard K, Richard DJ, O'Byrne KJ. SASH1 mediates sensitivity of breast cancer cells to chloropyramine and is associated with prognosis in breast cancer. Oncotarget 2016; 7:72807-72818. [PMID: 27637080 PMCID: PMC5341945 DOI: 10.18632/oncotarget.12020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/02/2016] [Indexed: 11/25/2022] Open
Abstract
Expression of the SASH1 protein is reduced in a range of human cancers and has been implicated in apoptotic cancer cell death. This study investigated whether increasing SASH1 expression could be a useful therapeutic strategy in breast cancer. Ectopic SASH1 expression increased apoptosis in 7/8 breast cancer cell lines. Subsequent in silico connectivity screening demonstrated that the clinically approved antihistamine drug, chloropyramine, increased SASH1 mRNA levels. Chloropyramine has previously been shown to have anti-tumour activity in breast cancer in part through modulation of FAK signalling, a pathway also regulated by SASH1. This study demonstrated that chloropyramine increased SASH1 protein levels in breast cancer cells. Consistent with this the agent reduced cell confluency in 7/8 cell lines treated irrespective of their ER status but not apoptosis incompetent MCF7 cells. In contrast SASH1 siRNA-transfected breast cancer cells exhibited reduced chloropyramine sensitivity. The prognostic significance of SASH1 expression was also investigated in two breast cancer cohorts. Expression was associated with favourable outcome in ER-positive cases, but only those of low histological grade/proliferative status. Conversely, we found a very strong inverse association in HER2+ disease irrespective of ER status, and in triple-negative, basal-like cases. Overall, the data suggest that SASH1 is prognostic in breast cancer and could have subtype-dependent effects on breast cancer progression. Pharmacologic induction of SASH1 by chloropyramine treatment of breast cancer warrants further preclinical and clinical investigation.
Collapse
Affiliation(s)
- Joshua T. Burgess
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology, Brisbane, Australia
| | - Emma Bolderson
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology, Brisbane, Australia
- Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland, Australia
| | - Jodi M. Saunus
- The University of Queensland (UQ), UQ Centre for Clinical Research, Herston, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, University of Ulster, Altnagelvin Hospital Campus, Londonderry, UK
- Center for Cancer Research and Cell Biology, Queen's University Belfast, United Kingdom
| | - Lynne E. Reid
- The University of Queensland (UQ), UQ Centre for Clinical Research, Herston, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Anne Marie McNicol
- The University of Queensland (UQ), UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Sunil R. Lakhani
- The University of Queensland (UQ), UQ Centre for Clinical Research, Herston, Queensland, Australia
- Pathology Queensland, Royal Brisbane Women's Hospital, Herston, Queensland, Australia
- UQ School of Medicine, Herston, Queensland, Australia
| | - Katharine Cuff
- Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland, Australia
| | - Kerry Richard
- UQ School of Medicine, Herston, Queensland, Australia
- Conjoint Endocrine Laboratory, Pathology Queensland, Queensland Health, Herston, Australia
| | - Derek J. Richard
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology, Brisbane, Australia
- Translational Cell Imaging Queensland, Translational Research Institute, Queensland, Australia
| | - Kenneth J. O'Byrne
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), Queensland University of Technology, Brisbane, Australia
- Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland, Australia
- Translational Cell Imaging Queensland, Translational Research Institute, Queensland, Australia
| |
Collapse
|
19
|
Wronska MA, O'Connor IB, Tilbury MA, Srivastava A, Wall JG. Adding Functions to Biomaterial Surfaces through Protein Incorporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5485-5508. [PMID: 27164952 DOI: 10.1002/adma.201504310] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/16/2016] [Indexed: 06/05/2023]
Abstract
The concept of biomaterials has evolved from one of inert mechanical supports with a long-term, biologically inactive role in the body into complex matrices that exhibit selective cell binding, promote proliferation and matrix production, and may ultimately become replaced by newly generated tissues in vivo. Functionalization of material surfaces with biomolecules is critical to their ability to evade immunorecognition, interact productively with surrounding tissues and extracellular matrix, and avoid bacterial colonization. Antibody molecules and their derived fragments are commonly immobilized on materials to mediate coating with specific cell types in fields such as stent endothelialization and drug delivery. The incorporation of growth factors into biomaterials has found application in promoting and accelerating bone formation in osteogenerative and related applications. Peptides and extracellular matrix proteins can impart biomolecule- and cell-specificities to materials while antimicrobial peptides have found roles in preventing biofilm formation on devices and implants. In this progress report, we detail developments in the use of diverse proteins and peptides to modify the surfaces of hard biomaterials in vivo and in vitro. Chemical approaches to immobilizing active biomolecules are presented, as well as platform technologies for isolation or generation of natural or synthetic molecules suitable for biomaterial functionalization.
Collapse
Affiliation(s)
- Małgorzata A Wronska
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Iain B O'Connor
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Maura A Tilbury
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Akshay Srivastava
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - J Gerard Wall
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| |
Collapse
|
20
|
Thompson MG, Larson M, Vidrine A, Barrios K, Navarro F, Meyers K, Simms P, Prajapati K, Chitsike L, Hellman LM, Baker BM, Watkins SK. FOXO3-NF-κB RelA Protein Complexes Reduce Proinflammatory Cell Signaling and Function. THE JOURNAL OF IMMUNOLOGY 2015; 195:5637-47. [PMID: 26561547 DOI: 10.4049/jimmunol.1501758] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/16/2015] [Indexed: 11/19/2022]
Abstract
Tumor-associated myeloid cells, including dendritic cells (DCs) and macrophages, are immune suppressive. This study demonstrates a novel mechanism involving FOXO3 and NF-κB RelA that controls myeloid cell signaling and impacts their immune-suppressive nature. We find that FOXO3 binds NF-κB RelA in the cytosol, impacting both proteins by preventing FOXO3 degradation and preventing NF-κB RelA nuclear translocation. The location of protein-protein interaction was determined to be near the FOXO3 transactivation domain. In turn, NF-κB RelA activation was restored upon deletion of the same sequence in FOXO3 containing the DNA binding domain. We have identified for the first time, to our knowledge, a direct protein-protein interaction between FOXO3 and NF-κB RelA in tumor-associated DCs. These detailed biochemical interactions provide the foundation for future studies to use the FOXO3-NF-κB RelA interaction as a target to enhance tumor-associated DC function to support or enhance antitumor immunity.
Collapse
Affiliation(s)
- Matthew G Thompson
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Michelle Larson
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Amy Vidrine
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Kelly Barrios
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Flor Navarro
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Kaitlyn Meyers
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Patricia Simms
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Kushal Prajapati
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Lennox Chitsike
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| | - Lance M Hellman
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556
| | - Brian M Baker
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556
| | - Stephanie K Watkins
- Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153; and
| |
Collapse
|
21
|
Moen I, Gebre M, Alonso-Camino V, Chen D, Epstein D, McDonald DM. Anti-metastatic action of FAK inhibitor OXA-11 in combination with VEGFR-2 signaling blockade in pancreatic neuroendocrine tumors. Clin Exp Metastasis 2015; 32:799-817. [PMID: 26445848 DOI: 10.1007/s10585-015-9752-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/08/2015] [Indexed: 02/08/2023]
Abstract
The present study sought to determine the anti-tumor effects of OXA-11, a potent, novel small-molecule amino pyrimidine inhibitor (1.2 pM biochemical IC(50)) of focal adhesion kinase (FAK). In studies of cancer cell lines, OXA-11 inhibited FAK phosphorylation at phospho-tyrosine 397 with a mechanistic IC(50) of 1 nM in TOV21G tumor cells, which translated into functional suppression of proliferation in 3-dimensional culture with an EC(50) of 9 nM. Studies of OXA-11 activity in TOV21G tumor-cell xenografts in mice revealed a pharmacodynamic EC(50) of 1.8 nM, indicative of mechanistic inhibition of pFAK [Y397] in these tumors. OXA-11 inhibited TOV21G tumor growth in a dose-dependent manner and also potentiated effects of cisplatin on tumor cell proliferation and apoptosis in vitro and on tumor growth in mice. Studies of pancreatic neuroendocrine tumors in RIP-Tag2 transgenic mice revealed OXA-11 suppression of pFAK [Y397] and pFAK [Y861] in tumors and liver. OXA-11 given daily from age 14 to 17 weeks reduced tumor vascularity, invasion, and when given together with the anti-VEGFR-2 antibody DC101 reduced the incidence, abundance, and size of liver metastases. Liver micrometastases were found in 100 % of mice treated with vehicle, 84 % of mice treated with OXA-11, and 79 % of mice treated with DC101 (19-24 mice per group). In contrast, liver micrometastases were found in only 52 % of 21 mice treated with OXA-11 plus DC101, and those present were significantly smaller and less numerous. Together, these findings indicate that OXA-11 is a potent and selective inhibitor of FAK phosphorylation in vitro and in vivo. OXA-11 slows tumor growth, potentiates the anti-tumor actions of cisplatin and--when combined with VEGFR-2 blockade--reduces metastasis of pancreatic neuroendocrine tumors in RIP-Tag2 mice.
Collapse
Affiliation(s)
- Ingrid Moen
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California - San Francisco, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA.,Department of Biomedicine, University of Bergen, Bergen, Norway.,Oxy Solutions, Parkveien 33B, Oslo, Norway
| | - Matthew Gebre
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California - San Francisco, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA.,School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Vanesa Alonso-Camino
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California - San Francisco, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Debbie Chen
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California - San Francisco, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA.,School of Medicine, University of California - Davis, Sacramento, CA, USA
| | - David Epstein
- Cancer & Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Donald M McDonald
- UCSF Helen Diller Family Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California - San Francisco, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA.
| |
Collapse
|
22
|
Targeting the C-terminal focal adhesion kinase scaffold in pancreatic cancer. Cancer Lett 2014; 353:281-9. [PMID: 25067788 DOI: 10.1016/j.canlet.2014.07.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/16/2014] [Accepted: 07/19/2014] [Indexed: 12/20/2022]
Abstract
Preliminary studies in our laboratory have demonstrated the importance of both the NH2 and COOH terminus scaffolding functions of focal adhesion kinase (FAK). Here, we describe a new small molecule inhibitor, C10, that targets the FAK C-terminus scaffold. C10 showed marked selectivity for cells overexpressing VEGFR3 when tested in isogenic cell lines, MCF7 and MCF7-VEGFR3. C10 preferentially inhibited pancreatic tumor growth in vivo in cells with high FAK-Y925 and VEGFR3 expression. Treatment with C10 led to a significant inhibition in endothelial cell proliferation and tumor endothelial and lymphatic vessel density and decrease in interstitial fluid pressure. These results highlight the underlying importance of targeting the FAK scaffold to treat human cancers.
Collapse
|