1
|
Braghini MR, De Stefanis C, Tiano F, Castellano A, Cicolani N, Pezzullo M, Tocco V, Spada M, Alaggio R, Alisi A, Francalanci P. Focal adhesion kinase and its epigenetic interactors as diagnostic and therapeutic hints for pediatric hepatoblastoma. Front Oncol 2024; 14:1397647. [PMID: 38947885 PMCID: PMC11211568 DOI: 10.3389/fonc.2024.1397647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024] Open
Abstract
Background Hepatoblastoma (HB) is the most common pediatric hepatic malignancy. Despite the progress in HB treatment, investigating HB pathomechanisms to optimize stratification and therapies remains a focal point to improve the outcome for high-risk patients. Methods Here, we pointed to explore the impact of these mechanisms in HB. An observational study was performed on liver samples from a cohort of 17 patients with a diagnosis of HB and two normal liver samples. The in vitro experiments were executed on the Huh6 human HB cell line treated with the FAK inhibitor TAE226. Results Our results highlight a significant up-regulation of mRNA and protein expression of FAK in livers from HB with respect to normal livers. The increased protein expression of total and Tyr397 phosphorylated FAK (pTyr397FAK) was significantly correlated with the expression of some epigenetic regulators of histone H3 methylation and acetylation. Of note, the expression of pTyr397FAK, N-methyltransferase enzyme (EZH2) and tri-methylation of the H3K27 residue correlated with tumor size and alpha-fetoprotein (AFP) levels. Finally, TAE226 caused a significant reduction of pTyr397FAK, epigenetic regulators, AFP, EPCAM, OCT4, and SOX2, in association with anti-proliferative and pro-apoptotic effects on HB cells. Conclusion Our results suggest a role of FAK in HB that requires further investigations mainly focused on the exploration of its effective diagnostic and therapeutic translatability.
Collapse
Affiliation(s)
- Maria Rita Braghini
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | - Francesca Tiano
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Aurora Castellano
- Division of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicolo’ Cicolani
- Core Facilities, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marco Pezzullo
- Core Facilities, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Valeria Tocco
- Core Facilities, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marco Spada
- Research Unit of Clinical Hepatogastroenterology and Transplantation; Division of Hepatobiliopancreatic Surgery, Liver and Kidney Transplantation, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Francalanci
- Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
2
|
Liu Y, Tong JB, Gao P, Fan XL, Xiao XC, Xing YC. Combining QSAR techniques, molecular docking, and molecular dynamics simulations to explore anti-tumor inhibitors targeting Focal Adhesion Kinase. J Biomol Struct Dyn 2024:1-17. [PMID: 38173145 DOI: 10.1080/07391102.2023.2301055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
Focal Adhesion Kinase (FAK) is an important target for tumor therapy and is closely related to tumor cell genesis and progression. In this paper, we selected 46 FAK inhibitors with anticancer activity in the pyrrolo pyrimidine backbone to establish 3D/2D-QSAR models to explore the relationship between inhibitory activity and molecular structure. We have established two ideal models, namely, the Topomer CoMFA model (q 2 = 0.715, r 2 = 0.984) and the Holographic Quantitative Structure-Activity Relationship (HQSAR) model (q 2 = 0.707, r 2 = 0.899). Both models demonstrate excellent external prediction capabilities.Based on the QSAR results, we designed 20 structurally modified novel compounds, which were subjected to molecular docking and molecular dynamics studies, and the results showed that the new compounds formed many robust interactions with residues within the active pocket and could maintain stable binding to the receptor proteins. This study not only provides a powerful screening tool for designing novel FAK inhibitors, but also presents a series of novel FAK inhibitors with high micromolar activity that can be used for further characterization. It provides a reference for addressing the shortcomings of drug metabolism and drug resistance of traditional FAK inhibitors, as well as the development of novel clinically applicable FAK inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yuan Liu
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Shaanxi University of Science and Technology, Xi'an, China
| | - Jian-Bo Tong
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Shaanxi University of Science and Technology, Xi'an, China
| | - Peng Gao
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Shaanxi University of Science and Technology, Xi'an, China
| | - Xuan-Lu Fan
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Shaanxi University of Science and Technology, Xi'an, China
| | - Xue-Chun Xiao
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Shaanxi University of Science and Technology, Xi'an, China
| | - Yi-Chaung Xing
- College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Key Laboratory of Chemical Additives for Industry, Shaanxi University of Science and Technology, Xi'an, China
| |
Collapse
|
3
|
Zeng S, Yuan S, Zhang Y, Du J, Wu Y, Chen Y, Zhu P, Huang W. Discovery of novel pyrrolo [2,3-d] pyrimidine derivatives as potent FAK inhibitors based on cyclization strategy. Bioorg Chem 2023; 139:106713. [PMID: 37459823 DOI: 10.1016/j.bioorg.2023.106713] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 08/13/2023]
Abstract
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, plays a pivotal role in tumor invasion and metastasis. Many FAK inhibitors had been reported, but the development of FAK inhibitors in clinical studies are still limited. To facilitate the discovery of FAK modulators and further elucidate the role of FAK in cancer metastasis, it is necessary to discover a novel, potent and selective FAK inhibitor. In this study, a series of FAK inhibitors with novel scaffold were designed and synthesized based on cyclization strategy. Here, we reported compound 10b (HMC-18NH) with excellent inhibition of FAK (IC50 = 9.9 nM) and anticancer activity against several cancer cell lines including BxPC-3, PANC-1, MCF-7, MDA-MB-231, U-87MG, HepG2, HCT-15 and A549. Extraordinary, compound 10b showed the best cytotoxic effects against A549 with the IC50 value of 0.8 μM. In addition, 10b exhibited effective invasion and migration suppression in A549 cells. Further investigations revealed that compound 10b potently induced and promoted apoptosis in a dose-dependent manner and arrested A549 cells in the G2/M phase. Collectively, these results suggest that 10b is a promising FAK inhibitor and serve as a lead compound which deserve for further optimization.
Collapse
Affiliation(s)
- Shenxin Zeng
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Shuai Yuan
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Yu Zhang
- School of Publish Health, Hangzhou Medical College, Hangzhou, Zhejiang 311399 China
| | - Jinbei Du
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Yuhao Wu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Yinqiao Chen
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Peizhen Zhu
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China.
| | - Wenhai Huang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China.
| |
Collapse
|
4
|
Guo C, Li Q, Xiao J, Ma F, Xia X, Shi M. Identification of defactinib derivatives targeting focal adhesion kinase using ensemble docking, molecular dynamics simulations and binding free energy calculations. J Biomol Struct Dyn 2023; 41:8654-8670. [PMID: 36281703 DOI: 10.1080/07391102.2022.2135601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/08/2022] [Indexed: 10/31/2022]
Abstract
Focal adhesion kinase (FAK) belongs to the nonreceptor tyrosine kinases, which selectively phosphorylate tyrosine residues on substrate proteins. FAK is associated with bladder, esophageal, gastric, neck, breast, ovarian and lung cancers. Thus, FAK has been considered as a potential target for tumor treatment. Currently, there are six adenosine triphosphate (ATP)-competitive FAK inhibitors tested in clinical trials but no approved inhibitors targeting FAK. Defactinib (VS-6063) is a second-generation FAK inhibitor with an IC50 of 0.6 nM. The binding model of VS-6063 with FAK may provide a reference model for developing new antitumor FAK-targeting drugs. In this study, the VS-6063/FAK binding model was constructed using ensemble docking and molecular dynamics simulations. Furthermore, the molecular mechanics/generalized Born (GB) surface area (MM/GBSA) method was employed to estimate the binding free energy between VS-6063 and FAK. The key residues involved in VS-6063/FAK binding were also determined using per-residue energy decomposition analysis. Based on the binding model, VS-6063 could be separated into seven regions to enhance its binding affinity with FAK. Meanwhile, 60 novel defactinib-based compounds were designed and verified using ensemble docking. Overall, the present study improves our understanding of the binding mechanism of human FAK with VS-6063 and provides new insights into future drug designs targeting FAK.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chuan Guo
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Qinxuan Li
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Jiujia Xiao
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Feng Ma
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xun Xia
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Anderson B, Rosston P, Ong HW, Hossain MA, Davis-Gilbert ZW, Drewry DH. How many kinases are druggable? A review of our current understanding. Biochem J 2023; 480:1331-1363. [PMID: 37642371 PMCID: PMC10586788 DOI: 10.1042/bcj20220217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
There are over 500 human kinases ranging from very well-studied to almost completely ignored. Kinases are tractable and implicated in many diseases, making them ideal targets for medicinal chemistry campaigns, but is it possible to discover a drug for each individual kinase? For every human kinase, we gathered data on their citation count, availability of chemical probes, approved and investigational drugs, PDB structures, and biochemical and cellular assays. Analysis of these factors highlights which kinase groups have a wealth of information available, and which groups still have room for progress. The data suggest a disproportionate focus on the more well characterized kinases while much of the kinome remains comparatively understudied. It is noteworthy that tool compounds for understudied kinases have already been developed, and there is still untapped potential for further development in this chemical space. Finally, this review discusses many of the different strategies employed to generate selectivity between kinases. Given the large volume of information available and the progress made over the past 20 years when it comes to drugging kinases, we believe it is possible to develop a tool compound for every human kinase. We hope this review will prove to be both a useful resource as well as inspire the discovery of a tool for every kinase.
Collapse
Affiliation(s)
- Brian Anderson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - Peter Rosston
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - Han Wee Ong
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - Zachary W. Davis-Gilbert
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| |
Collapse
|
6
|
Chawengrum P, Luepongpatthana N, Thongnest S, Sirirak J, Boonsombat J, Lirdprapamongkol K, Keeratichamroen S, Kongwaen P, Montatip P, Kittakoop P, Svasti J, Ruchirawat S. The amide derivative of anticopalic acid induces non-apoptotic cell death in triple-negative breast cancer cells by inhibiting FAK activation. Sci Rep 2023; 13:13456. [PMID: 37596365 PMCID: PMC10439230 DOI: 10.1038/s41598-023-40669-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 08/16/2023] [Indexed: 08/20/2023] Open
Abstract
Anticopalic acid (ACP), a labdane type diterpenoid obtained from Kaempferia elegans rhizomes, together with 21 semi-synthetic derivatives, were evaluated for their cancer cytotoxic activity. Most derivatives displayed higher cytotoxic activity than the parent compound ACP in a panel of nine cancer cell lines. Among the tested compounds, the amide 4p showed the highest cytotoxic activity toward leukemia cell lines, HL-60 and MOLT-3, with IC50 values of 6.81 ± 1.99 and 3.72 ± 0.26 µM, respectively. More interestingly, the amide derivative 4l exhibited cytotoxic activity with an IC50 of 13.73 ± 0.04 µM against the MDA-MB-231 triple-negative breast cancer cell line, which is the most aggressive type of breast cancer. Mechanistic studies revealed that 4l induced cell death in MDA-MB-231 cells through non-apoptotic regulated cell death. In addition, western blot analysis showed that compound 4l decreased the phosphorylation of FAK protein in a concentration-dependent manner. Molecular docking simulations elucidated that compound 4l could potentially inhibit FAK activation by binding to a pocket of FAK kinase domain. The data suggested that compound 4l could be a potential FAK inhibitor for treating triple-negative breast cancer and worth being further investigated.
Collapse
Affiliation(s)
- Pornsuda Chawengrum
- Chemical Biology Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Natthaorn Luepongpatthana
- Applied Biological Sciences Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Sanit Thongnest
- Laboratory of Natural Products, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | - Jitnapa Sirirak
- Department of Chemistry, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Jutatip Boonsombat
- Laboratory of Natural Products, Chulabhorn Research Institute, Bangkok, Thailand.
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand.
| | - Kriengsak Lirdprapamongkol
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand.
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand.
| | | | - Patcharin Kongwaen
- Laboratory of Natural Products, Chulabhorn Research Institute, Bangkok, Thailand
| | - Phreeranat Montatip
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| | - Prasat Kittakoop
- Chemical Biology Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Natural Products, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | - Jisnuson Svasti
- Applied Biological Sciences Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| | - Somsak Ruchirawat
- Chemical Biology Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Natural Products, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| |
Collapse
|
7
|
Design, synthesis and evaluation of nitric oxide releasing derivatives of 2,4-diaminopyrimidine as novel FAK inhibitors for intervention of metastatic triple-negative breast cancer. Eur J Med Chem 2023; 250:115192. [PMID: 36801517 DOI: 10.1016/j.ejmech.2023.115192] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
To search for novel medicines for intervention of triple-negative breast cancer (TNBC), a series of phenylsulfonyl furoxan-based 2,4-diaminopyrimidine derivatives (8a-t) were designed and synthesized based on blocking FAK-mediated signaling pathways through both kinase-dependent and -independent manners. The most active compound 8f not only significantly inhibited FAK kinase activity (IC50 = 27.44 nM), displayed potent inhibitory effects on the proliferation (IC50 = 0.126 μM), invasion and migration of MDA-MB-231 cells, superior to the most widely studied FAK inhibitor, TAE226, bearing 2,4-diaminopyrimidine, but also released high levels of NO, contributing to blockage of FAK mediated-signaling pathways by upregulating of p53 as well as suppressing the Y397 phosphorylation and its downstream effectors, including p-Akt, MMP-2, and MMP-9 via kinase-independent manner, leading to apoptosis induction and decrease of FAs and SFs in TNBC cells. Importantly, 8f inhibited the lung metastasis of TNBC in vivo. Together, 8f may serve as a promising candidate for the treatment of metastatic TNBC.
Collapse
|
8
|
Three-Dimensional-QSAR and Relative Binding Affinity Estimation of Focal Adhesion Kinase Inhibitors. Molecules 2023; 28:molecules28031464. [PMID: 36771129 PMCID: PMC9919860 DOI: 10.3390/molecules28031464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Precise binding affinity predictions are essential for structure-based drug discovery (SBDD). Focal adhesion kinase (FAK) is a member of the tyrosine kinase protein family and is overexpressed in a variety of human malignancies. Inhibition of FAK using small molecules is a promising therapeutic option for several types of cancer. Here, we conducted computational modeling of FAK-targeting inhibitors using three-dimensional structure-activity relationship (3D-QSAR), molecular dynamics (MD), and hybrid topology-based free energy perturbation (FEP) methods. The structure-activity relationship (SAR) studies between the physicochemical descriptors and inhibitory activities of the chemical compounds were performed with reasonable statistical accuracy using CoMFA and CoMSIA. These are two well-known 3D-QSAR methods based on the principle of supervised machine learning (ML). Essential information regarding residue-specific binding interactions was determined using MD and MM-PB/GBSA methods. Finally, physics-based relative binding free energy (ΔΔGRBFEA→B) terms of analogous ligands were estimated using alchemical FEP simulation. An acceptable agreement was observed between the experimental and computed relative binding free energies. Overall, the results suggested that using ML and physics-based hybrid approaches could be useful in synergy for the rational optimization of accessible lead compounds with similar scaffolds targeting the FAK receptor.
Collapse
|
9
|
Kumar R, Tiwari V, Dey S. Role of proline-rich tyrosine kinase 2 (Pyk2) in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5442-5452. [PMID: 34905657 DOI: 10.1111/ejn.15569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is the most common form of dementia in the elderly. Two major pathological hallmarks have been identified for AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT). Recently, proline-rich tyrosine kinase 2 (Pyk2), which belongs to the focal adhesion kinase (FAK) non-receptor tyrosine kinase family, was recognized to contribute significantly towards the pathogenesis of AD. Pyk2 can influence the formation of amyloid plaques as well as NFTs. The kinase can directly phosphorylate tau, which is a significant component of NFTs and enhances tau pathology. Several competitive inhibitors have been developed for Pyk2, tested in several cancer models, as Pyk2 is known to be overexpressed under those conditions. The current review article discusses the possible mechanistic pathways by which Pyk2 can influence the pathogenesis of AD. Besides, it describes various inhibitors for Pyk2 and their potential role as therapeutics for AD in the future.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM University, Visakhapatnam, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
10
|
Shi M, Chen T, Wei S, Zhao C, Zhang X, Li X, Tang X, Liu Y, Yang Z, Chen L. Molecular Docking, Molecular Dynamics Simulations, and Free Energy Calculation Insights into the Binding Mechanism between VS-4718 and Focal Adhesion Kinase. ACS OMEGA 2022; 7:32442-32456. [PMID: 36119979 PMCID: PMC9476166 DOI: 10.1021/acsomega.2c03951] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/24/2022] [Indexed: 05/17/2023]
Abstract
Focal adhesion kinase (FAK) is a 125 kDa nonreceptor tyrosine kinase that plays an important role in many carcinomas. Thus, the targeting of FAK by small molecules is considered to be promising for cancer therapy. Some FAK inhibitors have been reported as potential anticancer drugs and have entered into clinical development; for example, VS-4718 is currently undergoing clinical trials. However, the lack of crystal structural data for the binding of VS-4718 with FAK has hindered the optimization of this anticancer agent. In this work, the VS-4718/FAK interaction model was obtained by molecular docking and molecular dynamics simulations. The binding free energies of VS-4718/FAK were also calculated using the molecular mechanics generalized Born surface area method. It was found that the aminopyrimidine group formed hydrogen bonds with the C502 residue of the hinge loop, while the D564 residue of the T-loop interacted with the amide group. In addition, I428, A452, V484, M499, G505, and L553 residues formed hydrophobic interactions with VS-4718. The obtained results therefore provide an improved understanding of the interaction between human FAK and VS-4718. Based on the obtained binding mechanism, 47 novel compounds were designed to target the adenosine 5'-triphosphate-binding pocket of human FAK, and ensemble docking was performed to assess the effects of these modifications on the inhibitor binding affinity. This work is also expected to provide additional insights into potential future target design strategies based on VS-4718.
Collapse
Affiliation(s)
- Mingsong Shi
- State
Key Laboratory of Biotherapy, West China
Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Tao Chen
- State
Key Laboratory of Biotherapy, West China
Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Siping Wei
- Key
Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, China
- Department
of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Chenyu Zhao
- West
China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xinyu Zhang
- West
China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xinghui Li
- West
China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xinyi Tang
- West
China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Liu
- State
Key Laboratory of Biotherapy, West China
Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhuang Yang
- State
Key Laboratory of Biotherapy, West China
Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Lijuan Chen
- State
Key Laboratory of Biotherapy, West China
Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
11
|
Prospect of ULK1 modulators in targeting regulatory T cells. Bioorg Chem 2022; 129:106141. [PMID: 36137312 DOI: 10.1016/j.bioorg.2022.106141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 11/20/2022]
Abstract
Regulatory T (Treg) cells play an instrumental role in coordinating immune homeostasis via potent inhibitory effects. Defects in Treg cells lead to autoimmunity, but an overwhelming proportion of Treg cells encourages cancer progression. Hence, targeting Treg cells has emerged as a promising approach for mitigating disease severity. Recent studies have revealed that kinases are a critical component for tuning the fate of Treg cells, but the entire network of Treg-modulating kinases is still unclear. Here, we propose that the autophagy-activating UNC-51-like kinase 1 (ULK1) is a candidate for Treg cell modulation. While accumulating evidence has highlighted the role of autophagy-related kinases in Treg cells, the ULK1-Treg cell axis is yet to be examined. In this review, we predicted the potential role of ULK1 in Treg cell modulation. Furthermore, we summarized current ULK1 activators and inhibitors that can be investigated as Treg-targeting strategies, which might have beneficial outcomes in autoimmunity and cancer.
Collapse
|
12
|
Design, synthesis and biological evaluation of novel FAK inhibitors with better selectivity over IR than TAE226. Bioorg Chem 2022; 124:105790. [DOI: 10.1016/j.bioorg.2022.105790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/27/2022] [Accepted: 04/01/2022] [Indexed: 12/21/2022]
|
13
|
Zhang Y, Zhang Y, Li H, Cao Y, Han S, Zhang K, He W. Covalent Biosensing Polymer Chain Reaction Enabling Periphery Blood Testing to Predict Tumor Invasiveness with a Platelet Procancerous Protein. Anal Chem 2022; 94:1983-1989. [PMID: 35038868 DOI: 10.1021/acs.analchem.1c03349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Periphery blood testing is an attractive and relatively less invasive way of early cancer screening. In this work, based on the latest understanding of the pivotal role of platelets in promoting cancer invasion, a method for detecting a procancerous protein overexpressed both on platelets and in cancer cells is developed. As a kinase, the enzymatic activity, abundance, and self-phosphorylation of this protein are all important factors influencing its procancerous activity. To simultaneously determine these three important biochemical parameters, electrochemical control is called upon to connect or disconnect a polymer chain reaction (PCR) primer with a small-molecule synthetic probe, and with the target protein, in a target-specific manner. The resulting PCR signal amplification greatly improves the sensitivity of the design and also enables direct detection of the protein and its catalytic activity as well as its self-phosphorylation in clinical periphery blood samples from hepatocellular carcinoma (HCC) patients. This may point to future application of the proposed method in the early screening of HCC to assist its diagnosis and treatment.
Collapse
Affiliation(s)
- Yaxin Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China.,Information Science and Engineering College, University of Jinan, Jinan 250024, China
| | - Yunyan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China.,Information Science and Engineering College, University of Jinan, Jinan 250024, China
| | - Hao Li
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China
| | - Yi Cao
- Information Science and Engineering College, University of Jinan, Jinan 250024, China
| | - Shiyuan Han
- Information Science and Engineering College, University of Jinan, Jinan 250024, China
| | - Kai Zhang
- Department of Geriatric Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wenxing He
- School of Biological Science and Technology, University of Jinan, Jinan 250024, China
| |
Collapse
|
14
|
Chen T, Liu Y, Shi M, Tang M, Si W, Yuan X, Wen Y, Chen L. Design, synthesis, and biological evaluation of novel covalent inhibitors targeting focal adhesion kinase. Bioorg Med Chem Lett 2021; 54:128433. [PMID: 34757216 DOI: 10.1016/j.bmcl.2021.128433] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 02/08/2023]
Abstract
Forty-one new focal adhesion kinase (FAK) covalent inhibitors were designed and synthesized based on FAK inhibitor TAE226. Compound 11w displayed the highest inhibition of FAK with an IC50 value of 35 nM and exhibited potent anticancer activity against Hela, HCT116 and MDA-MB-231 cell lines with IC50 values of 0.41, 0.01 and 0.11 μM respectively, compared to TAE226 (2.68, 0.64 and 4.19 μM respectively). 11w also inhibited the clone formation and migration of HCT-116 cells and stimulated cell cycle arrest in the G2/M phase, inducing tumor cell apoptosis. Compound 11w formed a covalent bond with the Cys427 residue of FAK in a docking model, inhibiting the autophosphorylation of FAK and downstream proteins in a dose-dependent manner. Moreover, 11w showed adequate oral bioavailability of 21.02%. A 74.20% inhibition of tumor growth in the HCT116 xenograft model was also observed. These data indicate that 11w is a promising covalent inhibitor of FAK.
Collapse
Affiliation(s)
- Tao Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Yan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Wenting Si
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Xue Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Yi Wen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China.
| |
Collapse
|
15
|
Quispe PA, Lavecchia MJ, León IE. Focal adhesion kinase inhibitors in the treatment of solid tumors: Preclinical and clinical evidence. Drug Discov Today 2021; 27:664-674. [PMID: 34856395 DOI: 10.1016/j.drudis.2021.11.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/21/2021] [Accepted: 11/23/2021] [Indexed: 01/25/2023]
Abstract
Focal Adhesion Kinase (FAK) is a 125-kDa cytoplasmic protein kinase that is implicated in several cellular functions. This protein is an attractive molecular target for cancer therapy because a wide variety of studies have demonstrated associations between the activation or elevated expression of FAK and tumor progression, invasion, and drug resistance in malignant tumors. Here, we review the strategies used to inhibit FAK activity in solid tumors. We also include an overview of the preclinical (in vitro and in vivo) and clinical studies on FAK inhibitors.
Collapse
Affiliation(s)
- Patricia A Quispe
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina
| | - Martin J Lavecchia
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| | - Ignacio E León
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| |
Collapse
|
16
|
Campylobacter jejuni Triggers Signaling through Host Cell Focal Adhesions To Inhibit Cell Motility. mBio 2021; 12:e0149421. [PMID: 34425711 PMCID: PMC8406305 DOI: 10.1128/mbio.01494-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Campylobacter jejuni is a major foodborne pathogen that exploits the focal adhesions of intestinal cells to promote invasion and cause severe gastritis. Focal adhesions are multiprotein complexes involved in bidirectional signaling between the actin cytoskeleton and the extracellular matrix. We investigated the dynamics of focal adhesion structure and function in C. jejuni-infected cells using a comprehensive set of approaches, including confocal microscopy of live and fixed cells, immunoblotting, and superresolution interferometric photoactivated localization microscopy (iPALM). We found that C. jejuni infection of epithelial cells results in increased focal adhesion size and altered topology. These changes resulted in a persistent modulatory effect on the host cell focal adhesion, evidenced by an increase in cell adhesion strength, a decrease in individual cell motility, and a reduction in collective cell migration. We discovered that C. jejuni infection causes an increase in phosphorylation of paxillin and an alteration of paxillin turnover at the focal adhesion, which together represent a potential mechanistic basis for altered cell motility. Finally, we observed that infection of epithelial cells with the C. jejuni wild-type strain in the presence of a protein synthesis inhibitor, a C. jejuni CadF and FlpA fibronectin-binding protein mutant, or a C. jejuni flagellar export mutant blunts paxillin phosphorylation and partially reestablishes individual host cell motility and collective cell migration. These findings provide a potential mechanism for the restricted intestinal repair observed in C. jejuni-infected animals and raise the possibility that bacteria targeting extracellular matrix components can alter cell behavior after binding and internalization by manipulating focal adhesions.
Collapse
|
17
|
Cho H, Shin I, Yoon H, Jeon E, Lee J, Kim Y, Ryu S, Song C, Kwon NH, Moon Y, Kim S, Kim ND, Choi HG, Sim T. Identification of Thieno[3,2- d]pyrimidine Derivatives as Dual Inhibitors of Focal Adhesion Kinase and FMS-like Tyrosine Kinase 3. J Med Chem 2021; 64:11934-11957. [PMID: 34324343 DOI: 10.1021/acs.jmedchem.1c00459] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Focal adhesion kinase (FAK) is overexpressed in highly invasive and metastatic cancers. To identify novel FAK inhibitors, we designed and synthesized various thieno[3,2-d]pyrimidine derivatives. An intensive structure-activity relationship (SAR) study led to the identification of 26 as a lead. Moreover, 26, a multitargeted kinase inhibitor, possesses excellent potencies against FLT3 mutants as well as FAK. Gratifyingly, 26 remarkably inhibits recalcitrant FLT3 mutants, including F691L, that cause drug resistance. Importantly, 26 is superior to PF-562271 in terms of apoptosis induction, anchorage-independent growth inhibition, and tumor burden reduction in the MDA-MB-231 xenograft mouse model. Also, 26 causes regression of tumor growth in the MV4-11 xenograft mouse model, indicating that it could be effective against acute myeloid leukemia (AML). Finally, in an orthotopic mouse model using MDA-MB-231, 26 remarkably prevents metastasis of orthotopic tumors to lymph nodes. Taken together, the results indicate that 26 possesses potential therapeutic value against highly invasive cancers and relapsed AML.
Collapse
Affiliation(s)
- Hanna Cho
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Injae Shin
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hojong Yoon
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Eunhye Jeon
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jiwon Lee
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Younghoon Kim
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - SeongShick Ryu
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Chiman Song
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Nam Hoon Kwon
- Medicinal Bioconvergence Research Center, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Institute for Artificial Intelligence and Biomedical Research, College of Pharmacy & College of Medicine, Gangnam Severance Hospital, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Youngji Moon
- Medicinal Bioconvergence Research Center, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Institute for Artificial Intelligence and Biomedical Research, College of Pharmacy & College of Medicine, Gangnam Severance Hospital, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Nam Doo Kim
- Voronoibio Inc., 32 Songdogwahak-ro, Yeonsu-gu, Incheon 21984, Republic of Korea
| | - Hwan Geun Choi
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- B2Sbio Inc., 32 Songdogwahak-ro, Yeonsu-gu, Incheon 21984, Republic of Korea
| | - Taebo Sim
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, 5 Hwarangro14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
18
|
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
|
19
|
Pang XJ, Liu XJ, Liu Y, Liu WB, Li YR, Yu GX, Tian XY, Zhang YB, Song J, Jin CY, Zhang SY. Drug Discovery Targeting Focal Adhesion Kinase (FAK) as a Promising Cancer Therapy. Molecules 2021; 26:molecules26144250. [PMID: 34299525 PMCID: PMC8308130 DOI: 10.3390/molecules26144250] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/30/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
FAK is a nonreceptor intracellular tyrosine kinase which plays an important biological function. Many studies have found that FAK is overexpressed in many human cancer cell lines, which promotes tumor cell growth by controlling cell adhesion, migration, proliferation, and survival. Therefore, targeting FAK is considered to be a promising cancer therapy with small molecules. Many FAK inhibitors have been reported as anticancer agents with various mechanisms. Currently, six FAK inhibitors, including GSK-2256098 (Phase I), VS-6063 (Phase II), CEP-37440 (Phase I), VS-6062 (Phase I), VS-4718 (Phase I), and BI-853520 (Phase I) are undergoing clinical trials in different phases. Up to now, there have been many novel FAK inhibitors with anticancer activity reported by different research groups. In addition, FAK degraders have been successfully developed through “proteolysis targeting chimera” (PROTAC) technology, opening up a new way for FAK-targeted therapy. In this paper, the structure and biological function of FAK are reviewed, and we summarize the design, chemical types, and activity of FAK inhibitors according to the development of FAK drugs, which provided the reference for the discovery of new anticancer agents.
Collapse
Affiliation(s)
- Xiao-Jing Pang
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
| | - Xiu-Juan Liu
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
| | - Yuan Liu
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
| | - Wen-Bo Liu
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
| | - Yin-Ru Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.-R.L.); (G.-X.Y.); (X.-Y.T.)
| | - Guang-Xi Yu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.-R.L.); (G.-X.Y.); (X.-Y.T.)
| | - Xin-Yi Tian
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.-R.L.); (G.-X.Y.); (X.-Y.T.)
| | - Yan-Bing Zhang
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
| | - Jian Song
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.-R.L.); (G.-X.Y.); (X.-Y.T.)
- Correspondence: (J.S.); (C.-Y.J.); (S.-Y.Z.)
| | - Cheng-Yun Jin
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.-R.L.); (G.-X.Y.); (X.-Y.T.)
- Correspondence: (J.S.); (C.-Y.J.); (S.-Y.Z.)
| | - Sai-Yang Zhang
- Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (X.-J.P.); (X.-J.L.); (Y.L.); (W.-B.L.); (Y.-B.Z.)
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (Y.-R.L.); (G.-X.Y.); (X.-Y.T.)
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
- Correspondence: (J.S.); (C.-Y.J.); (S.-Y.Z.)
| |
Collapse
|
20
|
Han C, Shen K, Wang S, Wang Z, Su F, Wu X, Hu X, Li M, Han J, Wu L. Discovery of Novel 2,4-Dianilinopyrimidine Derivatives Containing 4-(Morpholinomethyl)phenyl and N-Substituted Benzamides as Potential FAK Inhibitors and Anticancer Agents. Molecules 2021; 26:molecules26144187. [PMID: 34299462 PMCID: PMC8304610 DOI: 10.3390/molecules26144187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/21/2022] Open
Abstract
Focal adhesion kinase (FAK) is responsible for the development and progression of various malignancies. With the aim to explore novel FAK inhibitors as anticancer agents, a series of 2,4-dianilinopyrimidine derivatives 8a–8i and 9a–9g containing 4-(morpholinomethyl)phenyl and N-substituted benzamides have been designed and synthesized. Among them, compound 8a displayed potent anti-FAK activity (IC50 = 0.047 ± 0.006 μM) and selective antiproliferative effects against H1975 (IC50 = 0.044 ± 0.011 μM) and A431 cells (IC50 = 0.119 ± 0.036 μM). Furthermore, compound 8a also induced apoptosis in a dose-dependent manner, arresting the cells in S/G2 phase and inhibiting the migration of H1975 cells, all of which were superior to those of TAE226. The docking analysis of compound 8a was performed to elucidate its possible binding modes with FAK. These results established 8a as our lead compound to be further investigated as a potential FAK inhibitor and anticancer agent.
Collapse
Affiliation(s)
- Chun Han
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Kemin Shen
- Department of Public Health and Preventive Medicine, Changzhi Medical College, Changzhi 046011, China;
| | - Shijun Wang
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Zhijun Wang
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Feng Su
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Xi Wu
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Xiaoqin Hu
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Mengyao Li
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
| | - Jing Han
- School of Chemistry and Materials Science, Jiangsu Normal University, Xuzhou 221116, China
- Correspondence: (J.H.); (L.W.); Tel.: +86-516-8340-3166 (J.H.); +86-355-217-8113 (L.W.)
| | - Lintao Wu
- Department of Chemistry, Changzhi University, Changzhi 046011, China; (C.H.); (S.W.); (Z.W.); (F.S.); (X.W.); (X.H.); (M.L.)
- Correspondence: (J.H.); (L.W.); Tel.: +86-516-8340-3166 (J.H.); +86-355-217-8113 (L.W.)
| |
Collapse
|
21
|
Brullo C, Tasso B. New Insights on Fak and Fak Inhibitors. Curr Med Chem 2021; 28:3318-3338. [PMID: 33143618 DOI: 10.2174/0929867327666201103162239] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/08/2020] [Accepted: 09/19/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Focal adhesion kinase (Fak) is a cytoplasmic protein tyrosine kinase overexpressed and activated in different solid cancers; it has shown an important role in metastasis formation, cell migration, invasion and angiogenesis and consequently it has been proposed as a potential target in cancer therapy, particularly in a metastatic phase. In recent years, different investigations have highlighted the importance of new Fak inhibitors as potential anti-cancer drugs, but other studies evidenced its role in different pathologies related to the cardiac function or viral infection. METHODS An extensive bibliographic research (104 references) has been done concerning the structure of Fak, its importance in tumor development, but also in other pathologies currently under study. The compounds currently subjected to clinical studies were therefore treated using the appropriate databases. Finally, the main chemical scaffolds currently under preclinical investigation were analyzed, focusing on their molecular structures and on the activity structure relationships (SAR). RESULTS At the moment, only a few reversible ATP-competitive inhibitors are under investigation in pre-clinical studies and clinical trials. Other compounds, with different chemical scaffolds, are investigated to obtain more active and selective Fak inhibitors. This mini-review is a summary of different Fak functions in cancer and other pathologies; the compounds today in clinical trials and the recent chemical scaffolds (also included in patents) giving the most interesting results are investigated. In addition, PROTAC molecules are reported. CONCLUSION All reported results evidenced that additional studies are necessary to design and synthesize new selective and more active compounds, although promising information has been obtained from associations between Fak inhibitors and other different anti- cancer drugs. In addition, the other important roles evidenced, both at the nuclear level and in non-cancerous cells, make this protein an increasingly important target in pharmaceutical chemistry.
Collapse
Affiliation(s)
- Chiara Brullo
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-I16132 Genova, Italy
| | - Bruno Tasso
- Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-I16132 Genova, Italy
| |
Collapse
|
22
|
Wang F, Yang W, Li R, Sui Z, Cheng G, Zhou B. Molecular description of pyrimidine-based inhibitors with activity against FAK combining 3D-QSAR analysis, molecular docking and molecular dynamics. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
23
|
Mustafa M, Abd El-Hafeez AA, Abdelhamid D, Katkar GD, Mostafa YA, Ghosh P, Hayallah AM, Abuo-Rahma GEDA. A first-in-class anticancer dual HDAC2/FAK inhibitors bearing hydroxamates/benzamides capped by pyridinyl-1,2,4-triazoles. Eur J Med Chem 2021; 222:113569. [PMID: 34111829 DOI: 10.1016/j.ejmech.2021.113569] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 01/06/2023]
Abstract
Novel 5-pyridinyl-1,2,4-triazoles were designed as dual inhibitors of histone deacetylase 2 (HDAC2) and focal adhesion kinase (FAK). Compounds 5d, 6a, 7c, and 11c were determined as potential inhibitors of both HDAC2 (IC50 = 0.09-1.40 μM) and FAK (IC50 = 12.59-36.11 nM); 6a revealed the highest activity with IC50 values of 0.09 μM and 12.59 nM for HDAC2 and FAK, respectively. Compound 6a was superior to reference drugs vorinostat and valproic acid in its ability to inhibit growth/proliferation of A-498 and Caki-1 renal cancer cells. Further investigation proved that 6a strongly arrests the cell cycle at the G2/M phase and triggers apoptosis in both A-498 and Caki-1 cells. Moreover, the enhanced Akt activity that is observed upon chronic application of HDAC inhibitors was effectively suppressed by the dual HDAC2/FAK inhibitor. Finally, the high potency and selectivity of 6a towards HDAC2 and FAK proteins were rationalized by molecular docking. Taken together, these findings highlight the potential of 6a as a promising dual-acting HDAC2/FAK inhibitor that could benefit from further optimization.
Collapse
Affiliation(s)
- Muhamad Mustafa
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Amer Ali Abd El-Hafeez
- Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt; Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Dalia Abdelhamid
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yaser A Mostafa
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, 71526, Egypt
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA; Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, CA, USA; Veterans Affairs Medical Center, La Jolla, CA, USA
| | - Alaa M Hayallah
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt; Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, 71526, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sphinx University, New Assiut, Egypt
| | - Gamal El-Din A Abuo-Rahma
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt.
| |
Collapse
|
24
|
Qi Y, Li Y, Fang Y, Gao H, Qiang B, Wang S, Zhang H. Design, Synthesis, Biological Evaluation, and Molecular Docking of 2,4-Diaminopyrimidine Derivatives Targeting Focal Adhesion Kinase as Tumor Radiotracers. Mol Pharm 2021; 18:1634-1642. [PMID: 33739836 DOI: 10.1021/acs.molpharmaceut.0c01088] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
There are two important topics in the field of cancer research: one is targeted molecular therapy and the other is tumor molecular imaging. Focal adhesion kinase (FAK) is considered as an attractive target for oncologic diagnosis and therapy. A series of 2,4-diaminopyrimidine derivatives were labeled with 18F to study their biological properties and their potential as positron emission tomography tumor imaging agents. They inhibited the activity of FAK with IC50 values in the wide range of 0.6-2164 nM, among which the IC50 of Q6 was 3.2 nM. For the biodistribution in S180-bearing mice, the corresponding [18F]Q6 was relatively good, with the highest uptake of 3.35 ± 0.32 % ID/g at 30 min postinjection, with a tumor/muscle ratio of 2.08 and a tumor/bone ratio of 2.48. Accordingly, [18F]Q6 was considered as a potential PET imaging agent for tumor diagnosis.
Collapse
Affiliation(s)
- Yueheng Qi
- College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| | - Ye Li
- College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| | - Yu Fang
- College of Chemistry and Chemical Engineering, Anyang Normal University, No. 436 Xian'ge Road, Anyang 455000, Henan Province, China
| | - Hang Gao
- College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| | - Bingchao Qiang
- College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| | - Shuxia Wang
- College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| | - Huabei Zhang
- College of Chemistry, Beijing Normal University, No. 19 Xinjiekouwai Street, Haidian District, Beijing 100875, China
| |
Collapse
|
25
|
Mustafa M, Abuo-Rahma GEDA, Abd El-Hafeez AA, Ahmed ER, Abdelhamid D, Ghosh P, Hayallah AM. Discovery of antiproliferative and anti-FAK inhibitory activity of 1,2,4-triazole derivatives containing acetamido carboxylic acid skeleton. Bioorg Med Chem Lett 2021; 40:127965. [PMID: 33744442 DOI: 10.1016/j.bmcl.2021.127965] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 01/18/2023]
Abstract
Small molecule inhibitors of the focal adhesion kinase are regarded as promising tools in our armamentarium for treating cancer. Here, we identified four 1,2,4-triazole derivatives that inhibit FAK kinase significantly and evaluated their therapeutic potential. Most tested compounds revealed potent antiproliferative activity in HepG2 and Hep3B liver cancer cells, in which 3c and 3d were the most potent (IC50 range; 2.88 ~ 4.83 µM). Compound 3d possessed significant FAK inhibitory activity with IC50 value of 18.10 nM better than the reference GSK-2256098 (IC50 = 22.14 nM). The preliminary mechanism investigation by Western blot analysis showed that both 3c and 3d repressed FAK phosphorylation comparable to GSK-2256098 in HepG2 cells. As a result of FAK inhibition, 3c and 3d inhibited the pro-survival pathways by decreasing the phosphorylation levels of PI3K, Akt, JNK, and STAT3 proteins. This effect led to apoptosis induction and cell cycle arrest. Taken together, these results indicate that 3d could serve as a potent preclinical candidate for the treatment of cancers.
Collapse
Affiliation(s)
- Muhamad Mustafa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Gamal El-Din A Abuo-Rahma
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | - Amer Ali Abd El-Hafeez
- Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt; Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Dalia Abdelhamid
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA; Rebecca and John Moore Comprehensive Cancer Center, University of California San Diego, La Jolla, CA, USA; Veterans Affairs Medical Center, La Jolla, CA, USA
| | - Alaa M Hayallah
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt; Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, 71526, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sphinx University, New Assiut, Egypt
| |
Collapse
|
26
|
Berger BT, Amaral M, Kokh DB, Nunes-Alves A, Musil D, Heinrich T, Schröder M, Neil R, Wang J, Navratilova I, Bomke J, Elkins JM, Müller S, Frech M, Wade RC, Knapp S. Structure-kinetic relationship reveals the mechanism of selectivity of FAK inhibitors over PYK2. Cell Chem Biol 2021; 28:686-698.e7. [PMID: 33497606 DOI: 10.1016/j.chembiol.2021.01.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 01/13/2023]
Abstract
There is increasing evidence of a significant correlation between prolonged drug-target residence time and increased drug efficacy. Here, we report a structural rationale for kinetic selectivity between two closely related kinases: focal adhesion kinase (FAK) and proline-rich tyrosine kinase 2 (PYK2). We found that slowly dissociating FAK inhibitors induce helical structure at the DFG motif of FAK but not PYK2. Binding kinetic data, high-resolution structures and mutagenesis data support the role of hydrophobic interactions of inhibitors with the DFG-helical region, providing a structural rationale for slow dissociation rates from FAK and kinetic selectivity over PYK2. Our experimental data correlate well with computed relative residence times from molecular simulations, supporting a feasible strategy for rationally optimizing ligand residence times. We suggest that the interplay between the protein structural mobility and ligand-induced effects is a key regulator of the kinetic selectivity of inhibitors of FAK versus PYK2.
Collapse
Affiliation(s)
- Benedict-Tilman Berger
- Structural Genomics Consortium, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Marta Amaral
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany; Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica Nacional, 2780-157 Oeiras, Portugal
| | - Daria B Kokh
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Ariane Nunes-Alves
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany; Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Djordje Musil
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Timo Heinrich
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Martin Schröder
- Structural Genomics Consortium, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Rebecca Neil
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Jing Wang
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Iva Navratilova
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Joerg Bomke
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Jonathan M Elkins
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Susanne Müller
- Structural Genomics Consortium, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Matthias Frech
- Discovery Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany; Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Buchmann Institute for Molecular Life Sciences, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany; German Cancer network DKTK and Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
27
|
Shirvani P, Fassihi A. In silico design of novel FAK inhibitors using integrated molecular docking, 3D-QSAR and molecular dynamics simulation studies. J Biomol Struct Dyn 2021; 40:5965-5982. [PMID: 33475043 DOI: 10.1080/07391102.2021.1875880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that plays a crucial role in integrin signaling that regulates essential cellular functions including growth, motility, proliferation and survival in different types of cells. Interestingly, it has also shown to be up-regulated in various types of tumors, hence it has emerged as a significant therapeutic target for the development of selective inhibitors. In present work, with the aim of achieving further insight into the structural characteristics required for the FAK inhibitory activity, a combined approach of molecular modeling studies including molecular docking, three-dimensional quantitative structure activity relationship (3D-QSAR) and molecular dynamics (MD) simulation were carried out on a series of 7H-pyrrolo[2,3-d]pyrimidine and thieno[3,2-d]pyrimidine FAK inhibitors. The probable binding modes and interactions of inhibitors into the FAK active site were predicted by molecular docking. The 3D-QSAR models were developed using the comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) methods, with three ligand-based, docking-based and receptor-based alignment techniques. Both CoMFA and CoMSIA models obtained from receptor-based alignment were superior to the ones obtained by other alignment methods. However, the CoMSIA model (q2 = 0.679, r2 = 0.954 and r2pred = 0.888) depicted almost better predictive ability than the CoMFA model (q2 = 0.617, r2 = 0.932 and r2pred = 0.856). The contour map analysis revealed the relationship between the structural features and inhibitory activity. The docking results and CoMFA and CoMSIA contour maps were in good accordance. Based on the information obtained from the molecular docking and contour map analysis, a series of novel FAK inhibitors were designed that showed better predicted inhibitory activity than the most potent compound 31 in the data set. Finally, the stability of the reference molecule 31 and the designed compounds D15 and D27 were evaluated through a 30 ns of MD simulation and their binding free energies were calculated using the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) method. The result of MD simulation and binding free energy decomposition demonstrated the important role of van der Waals interactions alongside H-bond ones that were in consistent with the docking and contour maps analysis results. In sum, the results from this study may provide a significant insight for developing more effective novel FAK inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pouria Shirvani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran
| | - Afshin Fassihi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran.,Bioinformatics Research Center, Isfahan university of Medical Science, Isfahan, Iran
| |
Collapse
|
28
|
Xie H, Lin X, Zhang Y, Tan F, Chi B, Peng Z, Dong W, An D. Design, synthesis and biological evaluation of ring-fused pyrazoloamino pyridine/pyrimidine derivatives as potential FAK inhibitors. Bioorg Med Chem Lett 2020; 30:127459. [PMID: 32784087 DOI: 10.1016/j.bmcl.2020.127459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/04/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022]
Abstract
We report herein the synthesis of novel ring-fused pyrazoloamino pyridine/pyrimidine derivatives as potential FAK inhibitors and the evaluation of pharmaceutical activity against five cancer cell lines (MDA-MB-231, BXPC-3, NCI-H1975, DU145 and 786O). Generally, the majority of compounds displayed strong anti-FAK enzymatic potencies (IC50 < 1 nM) and could effectively inhibit several class of cancer cell lines within the concentration of 3 μM in comparison with GSK2256098 as a reference. Among them, compound 4o is considered to be the most effective due to high sensitivity in antiproliferation. In culture, 4o could not only inhibit FAK Y397 phosphorylation in MDA-MB-231 cell line, but also trigger apoptosis in a dose-dependent manner. Furthermore, computational docking analysis also suggested that 4o and TAE-226 displayed the similar interaction with FAK kinase domain.
Collapse
Affiliation(s)
- Hongming Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Xinglong Lin
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co. Ltd, Dongguan 523871, PR China
| | - Yingjun Zhang
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co. Ltd, Dongguan 523871, PR China.
| | - Fuxing Tan
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co. Ltd, Dongguan 523871, PR China
| | - Bo Chi
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co. Ltd, Dongguan 523871, PR China
| | - Zhihong Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China.
| | - Wanrong Dong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Delie An
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China.
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Sun C, Feng L, Sun X, Yu R, Kang C. Design and screening of FAK, CDK 4/6 dual inhibitors by pharmacophore model, molecular docking, and molecular dynamics simulation. J Biomol Struct Dyn 2020; 39:5358-5367. [PMID: 32627678 DOI: 10.1080/07391102.2020.1786458] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Focal adhesion kinase (FAK) is one kind of tyrosine kinases that modulates integrin and growth factor signaling pathways, which is a promising therapeutic target because of involving in the migration, proliferation and survival of cancer cell. Overexpression and amplification of cyclin-dependent kinase 4/6 (CDK4/6) occur in many cancers and may be the cause of resistance to CDK4/6 inhibitors in preclinical models. The latest research shows that the combination of FAK and CDK4/6 can be dually targeted to enhance the antitumor effects. In this study, FAK and CDK4/6 dual target inhibitors were designed by computer-aided drug design. Seven million molecules were screened by the pharmacophore model and molecular docking. Finally, 6 compounds were obtained. Molecular dynamics simulation of compound 1, 2 and 3 showed that it has good binding stability to both receptors. According to the binding modes of compound 1 with two receptors, corresponding modifications were made, and 7 novel designed compounds were obtained. The docking energy of these novel designed compounds were lower than that of compound 1, and they can be tested in future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chuance Sun
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Lijun Feng
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xiaohua Sun
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Congmin Kang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
32
|
Zou XY, Zeng Q, Liu P, Nie MH. [Effect of the focal adhesion kinase inhibitor TAE226 on the epithelial-mesenchymal transition in human oral squamous cell carcinoma cell line]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:17-22. [PMID: 32037761 PMCID: PMC7184310 DOI: 10.7518/hxkq.2020.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/19/2019] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To study the effect of the focal adhesion kinase inhibitor TAE226 on epithelial-mesenchymal transition (EMT) in human oral squamous cell carcinoma (OSCC) cell line. METHODS HSC-3 and HSC-4 cells were cultured with TAE226 under different concentrations (0, 1, 5, and 10 μmol·L⁻¹) for 24, 48, and 72 h. Real-time quantitative polymerase chain reaction was performed to detect the mRNA expressions of E-cadherin and Vimentin. The protein expressions of E-cadherin and Vimentin were determined by Western blot assay after 48 h of TAE226 treatment. RESULTS Real-time quantitative polymerase chain reaction showed that increasing the TAE226 dose and reaction time resulted in increased and decreased E-cadherin and Vimentin mRNA expressions, respectively (P<0.05). Western blot assays showed that increasing the TAE226 dose resulted in increased and decreased E-cadherin and Vimentin protein expressions, respectively (P<0.05). CONCLUSIONS TAE226, which is expected to be an effective drug for OSCC treatment, can effectively inhibit the EMT of the OSCC cell line.
Collapse
Affiliation(s)
- Xiang-Yu Zou
- Dept. of Periodontics and Oral Medicine, The Affiliated Hospital of Stomatology, Southwest Medical University, Luzhou 646000, China;Laboratory for Reconstraction and Regeneration of Oral and Maxillofacial Region, Southwest Medical University, Luzhou 646000, China
| | - Qin Zeng
- Dept. of Periodontics and Oral Medicine, The Affiliated Hospital of Stomatology, Southwest Medical University, Luzhou 646000, China;Laboratory for Reconstraction and Regeneration of Oral and Maxillofacial Region, Southwest Medical University, Luzhou 646000, China
| | - Ping Liu
- Dept. of Stomatology, Luohu People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Min-Hai Nie
- Dept. of Periodontics and Oral Medicine, The Affiliated Hospital of Stomatology, Southwest Medical University, Luzhou 646000, China;Laboratory for Reconstraction and Regeneration of Oral and Maxillofacial Region, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
33
|
Fusani L, Palmer DS, Somers DO, Wall ID. Exploring Ligand Stability in Protein Crystal Structures Using Binding Pose Metadynamics. J Chem Inf Model 2020; 60:1528-1539. [PMID: 31910338 PMCID: PMC7145342 DOI: 10.1021/acs.jcim.9b00843] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Identification of
correct protein–ligand binding poses is
important in structure-based drug design and crucial for the evaluation
of protein–ligand binding affinity. Protein–ligand coordinates are commonly obtained from
crystallography experiments that provide a static model of an ensemble
of conformations. Binding pose metadynamics (BPMD) is an enhanced
sampling method that allows for an efficient assessment of ligand
stability in solution. Ligand poses that are unstable under the bias
of the metadynamics simulation are expected to be infrequently occupied
in the energy landscape, thus making minimal contributions to the
binding affinity. Here, the robustness of the method is studied using
crystal structures with ligands known to be incorrectly modeled, as
well as 63 structurally diverse crystal structures with ligand fit
to electron density from the Twilight database. Results show that
BPMD can successfully differentiate compounds whose binding pose is
not supported by the electron density from those with well-defined
electron density.
Collapse
Affiliation(s)
- Lucia Fusani
- Molecular Design UK, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K.,Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G11XL, U.K
| | - David S Palmer
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G11XL, U.K
| | - Don O Somers
- Protein, Cellular and Structural Sciences, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Ian D Wall
- Molecular Design UK, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
34
|
Balsa LM, Quispe P, Baran EJ, Lavecchia MJ, León IE. In silico and in vitro analysis of FAK/MMP signaling axis inhibition by VO-clioquinol in 2D and 3D human osteosarcoma cancer cells. Metallomics 2020; 12:1931-1940. [PMID: 33107537 DOI: 10.1039/d0mt00176g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The study of novel mechanisms of action of vanadium compounds is critical to elucidating the role and importance of these kinds of compounds as antitumor and antimetastatic agents. This work deals with in silico and in vitro studies of one clioquinol oxidovanadium(iv) complex [VO(clioquinol)2], VO(CQ)2, and its regulation of FAK. In particular, we focus on elucidating the relationship of the FAK inhibition, MMP activity and antimetastatic effects of the complex in human bone cancer cells.
Collapse
Affiliation(s)
- Lucia M Balsa
- Centro de Química Inorgánica (CEQUINOR, CONICET-UNLP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bv 120 1465, 1900 La Plata, Argentina.
| | | | | | | | | |
Collapse
|
35
|
Abou-Zied HA, Youssif BGM, Mohamed MFA, Hayallah AM, Abdel-Aziz M. EGFR inhibitors and apoptotic inducers: Design, synthesis, anticancer activity and docking studies of novel xanthine derivatives carrying chalcone moiety as hybrid molecules. Bioorg Chem 2019; 89:102997. [PMID: 31136902 DOI: 10.1016/j.bioorg.2019.102997] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/21/2019] [Accepted: 05/17/2019] [Indexed: 12/22/2022]
Abstract
One of the helpful ways to improve the effectiveness of anticancer agents and weaken drug resistance is to use hybrid molecules. therefore, the current study intended to introduce 20 novel xanthine/chalcone hybrids 9-28 of promising anticancer activity. Compounds 10, 11, 13, 14, 16, 20 and 23 exhibited potent inhibition of cancer cells growth with IC50 ranging from 1.0 ± 0.1 to 3.5 ± 0.4 μM compared to doxorubicin with IC50 ranging from 0.90 ± 0.62 to 1.41 ± 0.58 μM and that compounds 11 and 16 were the best. To verify the mechanism of their anticancer activity, compounds 10, 11, 13, 14, 16, 20 and 23 were evaluated for their EGFR inhibitory effect. The study results revealed that compound 11 showed IC50 = 0.3 µM on the target enzyme which is more potent than staurosporine reference drug (IC50 = 0.4 µM). Accordingly, the apoptotic effect of the most potent compounds 11 was extensively investigated and showed a marked increase in Bax level up to 29 folds, and down-regulation in Bcl2 to 0.28 fold, in comparison to the control. Furthermore, the effect of compound 11 on Caspases 3 and 8 was evaluated and was found to increase their levels by 8 and 14 folds, respectively. Also, the effect of compound 11 on the cell cycle and its cytotoxic effect were examined. Moreover, a molecular docking study was adopted to confirm mechanism of action.
Collapse
Affiliation(s)
- Hesham A Abou-Zied
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Aljouf, Sakaka 2014, Saudi Arabia.
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524 Sohag, Egypt
| | - Alaa M Hayallah
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Deraya University, Minia, Egypt; Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Mohamed Abdel-Aziz
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| |
Collapse
|
36
|
Wang L, Ai M, Yu J, Jin L, Wang C, Liu Z, Shu X, Tang Z, Liu K, Luo H, Guan W, Sun X, Ma X. Structure-based modification of carbonyl-diphenylpyrimidines (Car-DPPYs) as a novel focal adhesion kinase (FAK) inhibitor against various stubborn cancer cells. Eur J Med Chem 2019; 172:154-162. [PMID: 30978560 DOI: 10.1016/j.ejmech.2019.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022]
Abstract
A family of carbonyl-substituted diphenylpyrimidine derivatives (Car-DPPYs) with strong activity against focal adhesion kinase (FAK), were described in this manuscript. Among them, compounds 7a (IC50 = 5.17 nM) and 7f (IC50 = 2.58 nM) displayed equal anti-FAK enzymatic activity to the lead compound TAE226 (6.79 nM). In particular, compound 7a also exhibited strong antiproliferative activity against several stubborn cancer cells, including AsPC-1 cells (IC50 = 0.105 μM), BxPC-3 cells (IC50 = 0.090 μM), and MCF-7/ADR cells (IC50 = 0.59 μM). Additionally, compound 7a also showed great antitumor efficacy in vivo via aAsPC-1 cancer Xenograft mouse model. The preliminary mechanism study by Western blot analysis revealed that 7a repressed FAK phosphorylation in AsPC cancer cells. Taken together, the results indicate that compound 7a may serve as a promising preclinical candidate for treatment of stubborn cancers.
Collapse
Affiliation(s)
- Luhong Wang
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, PR China; College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Min Ai
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Jiawen Yu
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, PR China
| | - Lingling Jin
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Changyuan Wang
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Zhihao Liu
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Xiaohong Shu
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Zeyao Tang
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Kexin Liu
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Hui Luo
- Liaoning Ben-healthy Natural Technology and Dalian Buyun Biotechnology Co., Ltds., 116085, PR China
| | - Wenshun Guan
- Liaoning Ben-healthy Natural Technology and Dalian Buyun Biotechnology Co., Ltds., 116085, PR China
| | - Xiuli Sun
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, PR China.
| | - Xiaodong Ma
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China.
| |
Collapse
|
37
|
Reis IMA, Conceição RS, Ferreira RS, Dos Santos CC, da Silva GR, de Mattos Oliveira L, Cassiano DSA, Dos Santos Junior MC, Botura MB, da Silva VDA, Costa SL, da Silva TMS, Vieira IJC, Braz-Filho R, Branco A. Alkene lactones from Persea fulva (Lauraceae): Evaluation of their effects on tumor cell growth in vitro and molecular docking studies. Bioorg Chem 2019; 86:665-673. [PMID: 30826627 DOI: 10.1016/j.bioorg.2019.02.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/14/2019] [Accepted: 02/09/2019] [Indexed: 12/20/2022]
Abstract
The new alkene lactone, (3E)-5,6-dihydro-5-(hydroxymethyl)-3-docdecylidenefuran-3(4H)-one (1), named majoranolide B, and three alkene lactones known as majorenolide (2), majoranolide (3) and majorynolide (4) were obtained from the aerial parts of Persea fulva (Lauraceae). The structures were elucidated in light of extensive spectroscopic analysis, including 1D, 2D NMR (1H, 13C, 1H-1H-COSY, HMBC and HSQC) and HR-ESI-MS. These compounds were screened for their in vitro antiproliferative activity in rat C6 glioma and astrocyte cells using MTT assay and in silico by molecular docking against targets that play a central role in controlling glioma cell cycle progression. Majoranolide (3) is the most active compound with IC50 6.69 µM against C6 glioma cells, followed by the compounds 1 (IC50 9.06 µM), 2 (IC50 12.04 µM) and 4 (IC50 41.90 µM). The alkene lactones 1-3 exhibited lower toxicity in non-tumor cells when compared to glioma cells. Molecular docking results showed that majoranolide establishes hydrogen bonds with all targets through its α,β-unsaturated-γ-lactone moiety, whereas the long-chain alkyl group binds by means of several hydrophobic bonds. In the present study, it can be concluded from the anti-proliferative activity of isolates against C6 glioma cells that lactone constituents from P. fulva could have a great potential for the control of C6 glioma cells.
Collapse
Affiliation(s)
- Isabella Mary Alves Reis
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Av. Transnordestina s/n, 44036-900 Feira de Santana, BA, Brazil
| | - Rodrigo Souza Conceição
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Av. Transnordestina s/n, 44036-900 Feira de Santana, BA, Brazil
| | - Rafael Short Ferreira
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, 41100-100 Salvador, BA, Brazil
| | - Cleonice Creusa Dos Santos
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, 41100-100 Salvador, BA, Brazil
| | - Girliane Regina da Silva
- Programa de Pós-Graduação em Desenvolvimento e Inovação Tecnológica em Medicamentos, Departamento de Ciências Molecular, Universidade Federal Rural de Pernambuco, Campus Dois Irmãos, 52171-900 Recife, PE, Brazil
| | - Larissa de Mattos Oliveira
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Av. Transnordestina s/n, 44036-900 Feira de Santana, BA, Brazil
| | - Dayse Santos Almeida Cassiano
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Av. Transnordestina s/n, 44036-900 Feira de Santana, BA, Brazil
| | | | - Mariana Borges Botura
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Av. Transnordestina s/n, 44036-900 Feira de Santana, BA, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, 41100-100 Salvador, BA, Brazil
| | - Silvia Lima Costa
- Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia - UFBA, Av. Reitor Miguel Calmon s/n, Vale do Canela, 41100-100 Salvador, BA, Brazil
| | - Tania Maria Sarmento da Silva
- Programa de Pós-Graduação em Desenvolvimento e Inovação Tecnológica em Medicamentos, Departamento de Ciências Molecular, Universidade Federal Rural de Pernambuco, Campus Dois Irmãos, 52171-900 Recife, PE, Brazil
| | - Ivo José Curcino Vieira
- Laboratório de Ciências Químicas, Centro de Ciências e Tecnologia, Universidade Estadual do Norte Fluminense-Darcy Ribeiro, Av. Alberto Lamego, 2000-Parque Califórnia, 28013-602 Campos dos Goytacazes, RJ, Brazil
| | - Raimundo Braz-Filho
- PVE-FAPERJ/DEQUIM-ICE-Universidade Federal Rural do Rio de Janeiro (UFRRJ), CP 74541, 23894-374 Seropédica, RJ, Brazil; LCQUI-CCT-Universidade Estadual do Norte Fluminense Darcy Ribeiro, 28013-600 Campos dos Goytacazes, RJ, Brazil
| | - Alexsandro Branco
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Av. Transnordestina s/n, 44036-900 Feira de Santana, BA, Brazil.
| |
Collapse
|
38
|
Sang CY, Qin WW, Zhang XJ, Xu Y, Ma YZ, Wang XR, Hui L, Chen SW. Synthesis and identification of 2,4-bisanilinopyrimidines bearing 2,2,6,6-tetramethylpiperidine-N-oxyl as potential Aurora A inhibitors. Bioorg Med Chem 2019; 27:65-78. [DOI: 10.1016/j.bmc.2018.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/16/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022]
|
39
|
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
|
40
|
Yen-Pon E, Li B, Acebrón-Garcia-de-Eulate M, Tomkiewicz-Raulet C, Dawson J, Lietha D, Frame MC, Coumoul X, Garbay C, Etheve-Quelquejeu M, Chen H. Structure-Based Design, Synthesis, and Characterization of the First Irreversible Inhibitor of Focal Adhesion Kinase. ACS Chem Biol 2018; 13:2067-2073. [PMID: 29897729 DOI: 10.1021/acschembio.8b00250] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Focal Adhesion Kinase signaling pathway and its functions have been involved in the development and aggressiveness of tumor malignancy, it then presents a promising cancer therapeutic target. Several reversible FAK inhibitors have been developed and are being conducted in clinical trials. On the other hand, irreversible covalent inhibitors would bring many desirable pharmacological features including high potency and increased duration of action. Herein we report the structure-guided development of the first highly potent and irreversible inhibitor of the FAK kinase. This inhibitor showed a very potent decrease of autophosphorylation of FAK in squamous cell carcinoma. A cocrystal structure of the FAK kinase domain in complex with this compound revealed the inhibitor binding mode within the ATP binding site and confirmed the covalent linkage between the targeted Cys427 of the protein and the inhibitor.
Collapse
Affiliation(s)
- Expédite Yen-Pon
- Chemistry of RNA, Nucleosides, Peptides and Heterocycles, CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Bo Li
- Chemistry of RNA, Nucleosides, Peptides and Heterocycles, CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Marta Acebrón-Garcia-de-Eulate
- Cell Signalling and Adhesion Group, Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Céline Tomkiewicz-Raulet
- Toxicologie, Pharmacologie et Signalisation Cellulaire, INSERM, UMR S 1124, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - John Dawson
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, United Kingdom
| | - Daniel Lietha
- Cell Signalling and Adhesion Group, Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Margaret C. Frame
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, United Kingdom
| | - Xavier Coumoul
- Toxicologie, Pharmacologie et Signalisation Cellulaire, INSERM, UMR S 1124, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Christiane Garbay
- Chemistry of RNA, Nucleosides, Peptides and Heterocycles, CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Mélanie Etheve-Quelquejeu
- Chemistry of RNA, Nucleosides, Peptides and Heterocycles, CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Huixiong Chen
- Chemistry of RNA, Nucleosides, Peptides and Heterocycles, CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| |
Collapse
|
41
|
Lederer PA, Zhou T, Chen W, Epshtein Y, Wang H, Mathew B, Jacobson JR. Attenuation of murine acute lung injury by PF-573,228, an inhibitor of focal adhesion kinase. Vascul Pharmacol 2018; 110:16-23. [PMID: 29969688 DOI: 10.1016/j.vph.2018.06.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 05/01/2018] [Accepted: 06/29/2018] [Indexed: 01/07/2023]
Abstract
Acute lung injury (ALI) is characterized by endothelial barrier disruption resulting in increased vascular permeability. As focal adhesion kinase (FAK), a non-receptor protein tyrosine kinase, is involved in endothelial cell (EC) barrier regulation, we hypothesized that FAK inhibition could attenuate agonist-induced EC barrier disruption relevant to ALI. Human lung EC were pretreated with one of three pharmacologic FAK inhibitors, PF-573,228 (PF-228, 10 μM), PF-562,271 (PF-271, 5 μM) or NVP-TAE226 (TAE226, 5 μM) for 30 min prior to treatment with thrombin (1 U/ml, 30 min). Western blotting confirmed attenuated thrombin-induced FAK phosphorylation associated with all three inhibitors. Subsequently, EC were pretreated with either PF-228 (10 μM), TAE226 (5 μM) or PF-271 (5 μM) for 30 min prior to thrombin stimulation (1 U/ml) followed by measurements of barrier integrity by transendothelial electrical resistance (TER). Separately, EC grown in transwell inserts prior to thrombin (1 U/ml) with measurements of FITC-dextran flux after 30 min confirmed a significant attenuation of thrombin-induced EC barrier disruption by PF-228 alone. Finally, in a murine ALI model induced by LPS (1.25 mg/ml, IT), rescue treatment with PF-228 was associated with significantly reduced lung injury. Our findings PF-228, currently being studied in clinical trials, may serve as a novel and effective therapeutic agent for ALI.
Collapse
Affiliation(s)
- Paul A Lederer
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Tingting Zhou
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Weiguo Chen
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Yulia Epshtein
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Huashan Wang
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Biji Mathew
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Jeffrey R Jacobson
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
42
|
Lampi MC, Reinhart-King CA. Targeting extracellular matrix stiffness to attenuate disease: From molecular mechanisms to clinical trials. Sci Transl Med 2018; 10:10/422/eaao0475. [DOI: 10.1126/scitranslmed.aao0475] [Citation(s) in RCA: 274] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/08/2017] [Indexed: 12/11/2022]
Abstract
Tissues stiffen during aging and during the pathological progression of cancer, fibrosis, and cardiovascular disease. Extracellular matrix stiffness is emerging as a prominent mechanical cue that precedes disease and drives its progression by altering cellular behaviors. Targeting extracellular matrix mechanics, by preventing or reversing tissue stiffening or interrupting the cellular response, is a therapeutic approach with clinical potential. Major drivers of changes to the mechanical properties of the extracellular matrix include phenotypically converted myofibroblasts, transforming growth factor β (TGFβ), and matrix cross-linking. Potential pharmacological interventions to overcome extracellular matrix stiffening are emerging clinically. Aside from targeting stiffening directly, alternative approaches to mitigate the effects of increased matrix stiffness aim to identify and inhibit the downstream cellular response to matrix stiffness. Therapeutic interventions that target tissue stiffening are discussed in the context of their limitations, preclinical drug development efforts, and clinical trials.
Collapse
|
43
|
Fang Y, Wang D, Xu X, Dava G, Liu J, Li X, Xue Q, Wang H, Zhang J, Zhang H. Preparation, in vitro and in vivo evaluation, and molecular dynamics (MD) simulation studies of novel F-18 labeled tumor imaging agents targeting focal adhesion kinase (FAK). RSC Adv 2018; 8:10333-10345. [PMID: 35540451 PMCID: PMC9078890 DOI: 10.1039/c8ra00652k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/06/2018] [Indexed: 11/21/2022] Open
Abstract
Coronal micro-PET images of mice bearing S180 tumor at 30 min post-injection of [18F]2.
Collapse
|
44
|
Liu H, Wu B, Ge Y, Huang J, Song S, Wang C, Yao J, Liu K, Li Y, Li Y, Ma X. Phosphamide-containing diphenylpyrimidine analogues (PA-DPPYs) as potent focal adhesion kinase (FAK) inhibitors with enhanced activity against pancreatic cancer cell lines. Bioorg Med Chem 2017; 25:6313-6321. [PMID: 29102081 DOI: 10.1016/j.bmc.2017.09.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
A family of phosphamide-containing diphenylpyrimidine analogues (PA-DPPYs) were synthesized as potent focal adhesion kinase (FAK) inhibitors. The PA-DPPY derivatives could significantly inhibit the FAK enzymatic activity at concentrations lower than 10.69 nM. Among them, compounds 7a and 7e were two of the most active FAK inhibitors, possessing IC50 values of 4.25 nM and 4.65 nM, respectively. In particular, compound 7e also displayed strong activity against AsPC cell line, with an IC50 of 1.66 μM, but show low activity against the normal HPDE6-C7 cells (IC50 > 20 μM), indicating its low cell cytotoxicity. Additionally, flow cytometry analysis showed that after treatment with 7e (8 μM, 72 h), both AsPC and Panc cells growth were almost totally inhibited, with a cell viability rate of 16.8% and 18.1%, respectively. Overall, compound 7e may be served as a valuable FAK inhibitor for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- He Liu
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Bin Wu
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Yang Ge
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Jiaxin Huang
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Shijie Song
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Changyuan Wang
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Jihong Yao
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Kexin Liu
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China
| | - Yanxia Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, PR China
| | - Yongming Li
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China.
| | - Xiaodong Ma
- College of Pharmacy, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
45
|
Zha GF, Qin HL, Youssif BG, Amjad MW, Raja MAG, Abdelazeem AH, Bukhari SNA. Discovery of potential anticancer multi-targeted ligustrazine based cyclohexanone and oxime analogs overcoming the cancer multidrug resistance. Eur J Med Chem 2017; 135:34-48. [DOI: 10.1016/j.ejmech.2017.04.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/06/2017] [Accepted: 04/11/2017] [Indexed: 01/09/2023]
|
46
|
Qu M, Liu Z, Zhao D, Wang C, Zhang J, Tang Z, Liu K, Shu X, Yuan H, Ma X. Design, synthesis and biological evaluation of sulfonamide-substituted diphenylpyrimidine derivatives (Sul-DPPYs) as potent focal adhesion kinase (FAK) inhibitors with antitumor activity. Bioorg Med Chem 2017; 25:3989-3996. [PMID: 28576633 DOI: 10.1016/j.bmc.2017.05.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/17/2017] [Accepted: 05/20/2017] [Indexed: 10/19/2022]
Abstract
A class of sulfonamide-substituted diphenylpyrimidines (Sul-DPPYs) were synthesized to improve activity against the focal adhesion kinase (FAK). Most of these new Sul-DPPYs displayed moderate activity against the FAK enzyme with IC50 values of less than 100nM; regardless, they could effectively inhibit several classes of refractory cancer cell lines with IC50 values of less than 10µM, including the pancreatic cancer cell lines (AsPC-1, Panc-1 and BxPC-3), the NSCLC-resistant H1975 cell line, and the B lymphocyte cell line (Ramos cells). Results of flow cytometry indicated that inhibitor 7e promoted apoptosis of pancreatic cancer cells in a dose-dependent manner. In addition, it almost completely induced the apoptosis at a concentration of 10µM. Compound 7e may be selected as a potent FAK inhibitor for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Menghua Qu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Zhihao Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Dan Zhao
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Changyuan Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Jianbin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Zeyao Tang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China.
| | - Hong Yuan
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, PR China.
| | - Xiaodong Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
47
|
Dao P, Lietha D, Etheve-Quelquejeu M, Garbay C, Chen H. Synthesis of novel 1,2,4-triazine scaffold as FAK inhibitors with antitumor activity. Bioorg Med Chem Lett 2017; 27:1727-1730. [PMID: 28284808 DOI: 10.1016/j.bmcl.2017.02.072] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
A series of 1,3,5-triazinic inhibitors of focal adhesion kinase (FAK) has recently been shown to exert antiangiogenic activity against HUVEC cells and anticancer efficacy against several cancer cell lines. In this report, we designed and synthesized a series of new compounds containing a 1,2,4-triazine core as novel scaffold for FAK inhibitors. These compounds displayed 10-7M IC50 values, and the best one showed IC50 value of 0.23μM against FAK enzymatic activity. Among them, several inhibitors potently inhibited the proliferation of glioblastoma (U-87MG) and colon (HCT-116) cancer cell lines. Docking of compound 10 into the active site of the FAK kinase was performed to explore its potential binding mode.
Collapse
Affiliation(s)
- Pascal Dao
- CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Daniel Lietha
- Cell Signalling and Adhesion Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Mélanie Etheve-Quelquejeu
- CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Christiane Garbay
- CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Huixiong Chen
- CNRS UMR8601, Université Paris Descartes, PRES Sorbonne Paris Cité, UFR Biomédicale, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France.
| |
Collapse
|
48
|
Fang Y, Wang D, Xu X, Liu J, Wu A, Li X, Xue Q, Wang H, Wang H, Zhang H. Synthesis, biological evaluation, and molecular dynamics (MD) simulation studies of three novel F-18 labeled and focal adhesion kinase (FAK) targeted 5-bromo pyrimidines as radiotracers for tumor. Eur J Med Chem 2017; 127:493-508. [DOI: 10.1016/j.ejmech.2017.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
49
|
Zheng X, Bao W, Yang J, Zhang T, Sun D, Liang Y, Li S, Wang Y, Feng X, Hao H, Wang Z. Focal Adhesion Kinase Directly Interacts with TSC2 Through Its FAT Domain and Regulates Cell Proliferation in Cashmere Goat Fetal Fibroblasts. DNA Cell Biol 2016; 35:480-8. [PMID: 27380318 DOI: 10.1089/dna.2015.3033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic nonreceptor tyrosine kinase that senses a variety of extracellular signals, such as growth factors and integrins, to control the process of cell proliferation and metabolism. We cloned three goat FAK transcript variants (KM655805, KM658268, and KM658269) that encode 1052, 1006, and 962 amino-acid residue proteins. Bioinformatics analysis indicated that the putative FAK protein contains an FERM domain, a PTK domain, two Proline-rich regions, and a focal adhesion-targeting (FAT) domain. All the three transcript variants of FAK were detected in seven different goat tissues, and variant 1 had the most accumulation whereas variant 2 and variant 3 had lower accumulation. Treatment of goat fetal fibroblasts (GFbs) with a specific FAK inhibitor, TAE226, inhibited cell proliferation (p < 0.05) and induced damage to the cell morphology in a dose- and time-dependent manner. Further research demonstrated that FAK directly interacted with TSC2 (Tuberous sclerosis 2) tuberin domain through its C-terminus, which contains the complete FAT domain. In conclusion, our results indicated that FAK may be widely expressed in Cashmere goat tissues and its products participate in the mammalian target of rapamycin signaling pathway and cell proliferation through a direct interaction with TSC2 in GFBs.
Collapse
Affiliation(s)
- Xu Zheng
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China .,2 Hulunbeier Municipal People's Hospital , Hailaer, People's Republic of China
| | - Wenlei Bao
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Jiaofu Yang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Tao Zhang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Dongsheng Sun
- 3 Department of Oncology, Kailuan General Hospital , Tangshan, People's Republic of China
| | - Yan Liang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Shuyu Li
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Yanfeng Wang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Xue Feng
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Huifang Hao
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Zhigang Wang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| |
Collapse
|
50
|
Zhan JY, Zhang JL, Wang Y, Li Y, Zhang HX, Zheng QC. Exploring the interaction between human focal adhesion kinase and inhibitors: a molecular dynamic simulation and free energy calculations. J Biomol Struct Dyn 2016; 34:2351-66. [PMID: 26549408 DOI: 10.1080/07391102.2015.1115780] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Focal adhesion kinase is an important target for the treatment of many kinds of cancers. Inhibitors of FAK are proposed to be the anticancer agents for multiple tumors. The interaction characteristic between FAK and its inhibitors is crucial to develop new inhibitors. In the present article, we used Molecular Dynamic (MD) simulation method to explore the characteristic of interaction between FAK and three inhibitors (PHM16, TAE226, and ligand3). The MD simulation results together with MM-GB/SA calculations show that the combinations are enthalpy-driven process. Cys502 and Asp564 are both essential residues due to the hydrogen bond interactions with inhibitors, which was in good agreement with experimental data. Glu500 can form a non-classical hydrogen bond with each inhibitor. Arg426 can form electrostatic interactions with PHM16 and ligand3, while weaker with TAE226. The electronic static potential was employed, and we found that the ortho-position methoxy of TAE226 has a weaker negative charge than the meta-position one in PHM16 or ligand3. Ile428, Val436, Ala452, Val484, Leu501, Glu505, Glu506, Leu553, Gly563 Leu567, Ser568 are all crucial residues in hydrophobic interactions. The key residues in this work will be available for further inhibitor design of FAK and also give assistance to further research of cancer.
Collapse
Affiliation(s)
- Jiu-Yu Zhan
- a State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , Changchun 130023 , P.R. China
| | - Ji-Long Zhang
- a State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , Changchun 130023 , P.R. China
| | - Yan Wang
- a State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , Changchun 130023 , P.R. China
| | - Ye Li
- c Changchun Institute of Biological Products Co. Ltd , Changchun 130012 , P.R. China
| | - Hong-Xing Zhang
- a State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , Changchun 130023 , P.R. China
| | - Qing-Chuan Zheng
- a State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry , Jilin University , Changchun 130023 , P.R. China.,b Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education , Jilin University , Changchun 130023 , P.R. China
| |
Collapse
|