1
|
Singh A, Lakkaniga NR. Exploring the Conformational Space of MPS1 Kinase Using Metadynamics. Proteins 2025. [PMID: 39786318 DOI: 10.1002/prot.26796] [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: 06/24/2024] [Revised: 11/22/2024] [Accepted: 12/22/2024] [Indexed: 01/12/2025]
Abstract
MPS1 kinase is a dual specificity kinase that plays an important role in the spindle assembly checkpoint mechanism during cell division. Overexpression of MPS1 kinase is reported in several cancers. However, drug discovery and development efforts targeting MPS1 kinase did not result in any clinically successful candidates. All the reported crystal structures of MPS1 kinase adopt the DFG "in" conformation. Knowledge of the other conformations of the kinase would be beneficial in the structure-based drug design of novel inhibitors. This work employs well-tempered metadynamics simulations to explore the conformational space of MPS1 kinase by using its experimentally determined DFG "in" conformation as the starting structure. The simulation could successfully predict the DFG "out" conformation and identify the possible transition states. The key interactions that stabilize the kinase in various conformations were identified, and the effect of phosphorylation of the key residues on the conformation of the kinase was determined.
Collapse
Affiliation(s)
- Anuradha Singh
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| |
Collapse
|
2
|
Ma M. Current scenario of pyrazole hybrids with anti-breast cancer therapeutic applications. Arch Pharm (Weinheim) 2024; 357:e2400344. [PMID: 38943440 DOI: 10.1002/ardp.202400344] [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: 05/04/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/01/2024]
Abstract
Breast cancer stands as the leading cause of cancer-related deaths among women globally, but current therapy is restricted to the serious adverse effects and multidrug resistance, necessitating the exploration of novel, safe, and efficient anti-breast cancer chemotherapeutic agents. Pyrazoles exhibit excellent potential for utilization as effective anti-breast cancer agents due to their ability to act on various biological targets. Particularly, pyrazole hybrids demonstrated the advantage of targeting multiple pathways, and some of them, which are exemplified by larotrectinib (pyrazolo[1,5-a]pyrimidine hybrid), can be applied for breast cancer therapy. Thus, pyrazole hybrids hold great promise as useful therapeutic interventions for breast cancer. The aim of this review is to summarize the current scenario of pyrazole hybrids with in vitro and/or in vivo anti-breast cancer potential, along with the modes of action and structure-activity relationships, covering articles published from 2020 to the present, to streamline the development of rational, effective and safe anti-breast cancer candidates.
Collapse
Affiliation(s)
- Mengyu Ma
- Department of Pharmaceutical Engineering, School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, Henan, People's Republic of China
| |
Collapse
|
3
|
Halder P, Rai A, Talukdar V, Das P, Lakkaniga NR. Pyrazolopyridine-based kinase inhibitors for anti-cancer targeted therapy. RSC Med Chem 2024; 15:1452-1470. [PMID: 38784451 PMCID: PMC11110789 DOI: 10.1039/d4md00003j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/24/2024] [Indexed: 05/25/2024] Open
Abstract
The need for effective cancer treatments continues to be a challenge for the biomedical research community. In this case, the advent of targeted therapy has significantly improved therapeutic outcomes. Drug discovery and development efforts targeting kinases have resulted in the approval of several small-molecule anti-cancer drugs based on ATP-mimicking heterocyclic cores. Pyrazolopyridines are a group of privileged heterocyclic cores in kinase drug discovery, which are present in several inhibitors that have been developed against various cancers. Notably, selpercatinib, glumetinib, camonsertib and olverembatinib have either received approval or are in late-phase clinical studies. This review presents the success stories employing pyrazolopyridine scaffolds as hinge-binding cores to address various challenges in kinase-targeted drug discovery research.
Collapse
Affiliation(s)
- Pallabi Halder
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Anubhav Rai
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Vishal Talukdar
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Parthasarathi Das
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| |
Collapse
|
4
|
Gupta D, Kumar M, Saifi S, Rawat S, Ethayathulla AS, Kaur P. A comprehensive review on role of Aurora kinase inhibitors (AKIs) in cancer therapeutics. Int J Biol Macromol 2024; 265:130913. [PMID: 38508544 DOI: 10.1016/j.ijbiomac.2024.130913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Aurora kinases (AURKs) are a family of serine /threonine protein kinases that have a crucial role in cell cycle process mainly in the event of chromosomal segregation, centrosome maturation and cytokinesis. The family consists of three members including Aurora kinase A (AURK-A), Aurora kinase B (AURK-B) and Aurora kinase C (AURK-C). All AURKs contain a conserved kinase domain for their activity but differ in their cellular localization and functions. AURK-A and AURK-B are expressed mainly in somatic cells while the expression of AURK-C is limited to germ cells. AURK-A promotes G2 to M transition of cell cycle by controlling centrosome maturation and mitotic spindle assembly. AURK-B and AURK-C form the chromosome passenger complex (CPC) that ensures proper chromosomal alignments and segregation. Aberrant expression of AURK-A and AURK-B has been detected in several solid tumours and malignancies. Hence, they have become an attractive therapeutic target against cancer. The first part of this review focuses on AURKs structure, functions, subcellular localization, and their role in tumorigenesis. The review also highlights the functional and clinical impact of selective as well as pan kinase inhibitors. Currently, >60 compounds that target AURKs are in preclinical and clinical studies. The drawbacks of existing inhibitors like selectivity, drug resistance and toxicity have also been addressed. Since, majority of inhibitors are Aurora kinase inhibitor (AKI) type-1 that bind to the active (DFGin and Cin) conformation of the kinase, this information may be utilized to design highly selective kinase inhibitors that can be combined with other therapeutic agents for better clinical outcomes.
Collapse
Affiliation(s)
- Deepali Gupta
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Sana Saifi
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Shivani Rawat
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - A S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, Delhi 110029, India.
| |
Collapse
|
5
|
Lakkaniga NR, Wang Z, Xiao Y, Kharbanda A, Lan L, Li HY. Revisiting Aurora Kinase B: A promising therapeutic target for cancer therapy. Med Res Rev 2024; 44:686-706. [PMID: 37983866 DOI: 10.1002/med.21994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/28/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Cancer continues to be a major health concern globally, although the advent of targeted therapy has revolutionized treatment options. Aurora Kinase B is a serine-threonine kinase that has been explored as an oncology therapeutic target for more than two decades. Aurora Kinase B inhibitors show promising biological results in in-vitro and in-vivo experiments. However, there are no inhibitors approved yet for clinical use, primarily because of the side effects associated with Aurora B inhibitors. Several studies demonstrate that Aurora B inhibitors show excellent synergy with various chemotherapeutic agents, radiation therapy, and targeted therapies. This makes it an excellent choice as an adjuvant therapy to first-line therapies, which greatly improves the therapeutic window and side effect profile. Recent studies indicate the role of Aurora B in some deadly cancers with limited therapeutic options, like triple-negative breast cancer and glioblastoma. Herein, we review the latest developments in Aurora Kinase B targeted research, with emphasis on its potential as an adjuvant therapy and its role in some of the most difficult-to-treat cancers.
Collapse
Affiliation(s)
- Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | - Zhengyu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yao Xiao
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Anupreet Kharbanda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Li Lan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
6
|
Zhang J, Ma J, Li Y, An Y, Du W, Yang Q, Huang M, Cai X. Overexpression of Aurora Kinase B Is Correlated with Diagnosis and Poor Prognosis in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:2199. [PMID: 38396874 PMCID: PMC10889672 DOI: 10.3390/ijms25042199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Aurora kinase B (AURKB) overexpression promotes tumor initiation and development by participating in the cell cycle. In this study, we focused on the mechanism of AURKB in hepatocellular carcinoma (HCC) progression and on AURKB's value as a diagnostic and prognostic biomarker in HCC. We used data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) to analyze AURKB expression in HCC. We found that the expression levels of AURKB in HCC samples were higher than those in the corresponding control group. R packages were used to analyze RNA sequencing data to identify AURKB-related differentially expressed genes (DEGs), and these genes were found to be significantly enriched during the cell cycle. The biological function of AURKB was verified, and the results showed that cell proliferation was slowed down and cells were arrested in the G2/M phase when AURKB was knocked down. AURKB overexpression resulted in significant differences in clinical symptoms, such as the clinical T stage and pathological stage. Kaplan-Meier survival analysis, Cox regression analysis, and Receiver Operating Characteristic (ROC) curve analysis suggested that AURKB overexpression has good diagnostic and prognostic potential in HCC. Therefore, AURKB may be used as a potential target for the diagnosis and cure of HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xuefei Cai
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, 1 Yixue Yuan Road, Chongqing 400016, China; (J.Z.); (J.M.); (Y.L.); (Y.A.); (W.D.); (Q.Y.); (M.H.)
| |
Collapse
|
7
|
Bertran-Alamillo J, Giménez-Capitán A, Román R, Talbot S, Whiteley R, Floc'h N, Martínez-Pérez E, Martin MJ, Smith PD, Sullivan I, Terp MG, Saeh J, Marino-Buslje C, Fabbri G, Guo G, Xu M, Tornador C, Aguilar-Hernández A, Reguart N, Ditzel HJ, Martínez-Bueno A, Nabau-Moretó N, Gascó A, Rosell R, Pease JE, Polanska UM, Travers J, Urosevic J, Molina-Vila MA. BID expression determines the apoptotic fate of cancer cells after abrogation of the spindle assembly checkpoint by AURKB or TTK inhibitors. Mol Cancer 2023; 22:110. [PMID: 37443114 PMCID: PMC10339641 DOI: 10.1186/s12943-023-01815-w] [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: 02/27/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Drugs targeting the spindle assembly checkpoint (SAC), such as inhibitors of Aurora kinase B (AURKB) and dual specific protein kinase TTK, are in different stages of clinical development. However, cell response to SAC abrogation is poorly understood and there are no markers for patient selection. METHODS A panel of 53 tumor cell lines of different origins was used. The effects of drugs were analyzed by MTT and flow cytometry. Copy number status was determined by FISH and Q-PCR; mRNA expression by nCounter and RT-Q-PCR and protein expression by Western blotting. CRISPR-Cas9 technology was used for gene knock-out (KO) and a doxycycline-inducible pTRIPZ vector for ectopic expression. Finally, in vivo experiments were performed by implanting cultured cells or fragments of tumors into immunodeficient mice. RESULTS Tumor cells and patient-derived xenografts (PDXs) sensitive to AURKB and TTK inhibitors consistently showed high expression levels of BH3-interacting domain death agonist (BID), while cell lines and PDXs with low BID were uniformly resistant. Gene silencing rendered BID-overexpressing cells insensitive to SAC abrogation while ectopic BID expression in BID-low cells significantly increased sensitivity. SAC abrogation induced activation of CASP-2, leading to cleavage of CASP-3 and extensive cell death only in presence of high levels of BID. Finally, a prevalence study revealed high BID mRNA in 6% of human solid tumors. CONCLUSIONS The fate of tumor cells after SAC abrogation is driven by an AURKB/ CASP-2 signaling mechanism, regulated by BID levels. Our results pave the way to clinically explore SAC-targeting drugs in tumors with high BID expression.
Collapse
Affiliation(s)
- Jordi Bertran-Alamillo
- Laboratory of Oncology, Pangaea Oncology, Quiron Dexeus University Hospital, C/ Sabino Arana 5-19, 08913, Barcelona, Spain
| | - Ana Giménez-Capitán
- Laboratory of Oncology, Pangaea Oncology, Quiron Dexeus University Hospital, C/ Sabino Arana 5-19, 08913, Barcelona, Spain
| | - Ruth Román
- Laboratory of Oncology, Pangaea Oncology, Quiron Dexeus University Hospital, C/ Sabino Arana 5-19, 08913, Barcelona, Spain
| | - Sara Talbot
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Rebecca Whiteley
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Nicolas Floc'h
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | | | - Matthew J Martin
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Paul D Smith
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Ivana Sullivan
- Servicio de Oncología Médica, Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Barcelona, 08028, Spain
| | - Mikkel G Terp
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, 5000, Denmark
| | - Jamal Saeh
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, 02451, USA
| | | | - Giulia Fabbri
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, 02451, USA
| | - Grace Guo
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, 02451, USA
| | - Man Xu
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, 02451, USA
| | | | | | - Noemí Reguart
- Thoracic Oncology Unit, Department of Medical Oncology, Hospital Clínic, Barcelona, 08036, Spain
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, 5000, Denmark
- Department of Oncology, Odense University Hospital, Odense, 5000, Denmark
| | | | | | - Amaya Gascó
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Rafael Rosell
- Instituto Oncológico Dr. Rosell, Hospital Universitario Dexeus, Barcelona, 08028, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, 08916, Spain
| | - J Elizabeth Pease
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Urszula M Polanska
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Jon Travers
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK
| | - Jelena Urosevic
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, CB21 6GH, UK.
| | - Miguel A Molina-Vila
- Laboratory of Oncology, Pangaea Oncology, Quiron Dexeus University Hospital, C/ Sabino Arana 5-19, 08913, Barcelona, Spain.
| |
Collapse
|
8
|
Kovacs AH, Zhao D, Hou J. Aurora B Inhibitors as Cancer Therapeutics. Molecules 2023; 28:3385. [PMID: 37110619 PMCID: PMC10144992 DOI: 10.3390/molecules28083385] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The Aurora kinases (A, B, and C) are a family of three isoform serine/threonine kinases that regulate mitosis and meiosis. The Chromosomal Passenger Complex (CPC), which contains Aurora B as an enzymatic component, plays a critical role in cell division. Aurora B in the CPC ensures faithful chromosome segregation and promotes the correct biorientation of chromosomes on the mitotic spindle. Aurora B overexpression has been observed in several human cancers and has been associated with a poor prognosis for cancer patients. Targeting Aurora B with inhibitors is a promising therapeutic strategy for cancer treatment. In the past decade, Aurora B inhibitors have been extensively pursued in both academia and industry. This paper presents a comprehensive review of the preclinical and clinical candidates of Aurora B inhibitors as potential anticancer drugs. The recent advances in the field of Aurora B inhibitor development will be highlighted, and the binding interactions between Aurora B and inhibitors based on crystal structures will be presented and discussed to provide insights for the future design of more selective Aurora B inhibitors.
Collapse
Affiliation(s)
- Antal H. Kovacs
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, ON P7B 5E1, Canada
| | - Dong Zhao
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, ON P7B 5E1, Canada
| | - Jinqiang Hou
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, ON P7B 5E1, Canada
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada
| |
Collapse
|
9
|
Pang K, Wang W, Qin J, Shi Z, Hao L, Ma Y, Xu H, Wu Z, Pan D, Chen Z, Han C. Role of protein phosphorylation in cell signaling, disease, and the intervention therapy. MedComm (Beijing) 2022; 3:e175. [PMID: 36349142 PMCID: PMC9632491 DOI: 10.1002/mco2.175] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/06/2022] Open
Abstract
Protein phosphorylation is an important post-transcriptional modification involving an extremely wide range of intracellular signaling transduction pathways, making it an important therapeutic target for disease intervention. At present, numerous drugs targeting protein phosphorylation have been developed for the treatment of various diseases including malignant tumors, neurological diseases, infectious diseases, and immune diseases. In this review article, we analyzed 303 small-molecule protein phosphorylation kinase inhibitors (PKIs) registered and participated in clinical research obtained in a database named Protein Kinase Inhibitor Database (PKIDB), including 68 drugs approved by the Food and Drug Administration of the United States. Based on previous classifications of kinases, we divided these human protein phosphorylation kinases into eight groups and nearly 50 families, and delineated their main regulatory pathways, upstream and downstream targets. These groups include: protein kinase A, G, and C (AGC) and receptor guanylate cyclase (RGC) group, calmodulin-dependent protein kinase (CaMK) group, CMGC [Cyclin-dependent kinases (CDKs), Mitogen-activated protein kinases (MAPKs), Glycogen synthase kinases (GSKs), and Cdc2-like kinases (CLKs)] group, sterile (STE)-MAPKs group, tyrosine kinases (TK) group, tyrosine kinase-like (TKL) group, atypical group, and other groups. Different groups and families of inhibitors stimulate or inhibit others, forming an intricate molecular signaling regulatory network. This review takes newly developed new PKIs as breakthrough point, aiming to clarify the regulatory network and relationship of each pathway, as well as their roles in disease intervention, and provide a direction for future drug development.
Collapse
Affiliation(s)
- Kun Pang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Wei Wang
- Department of Medical CollegeSoutheast UniversityNanjingJiangsuChina
| | - Jia‐Xin Qin
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Zhen‐Duo Shi
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| | - Yu‐Yang Ma
- Graduate SchoolBengbu Medical CollegeBengbuAnhuiChina
| | - Hao Xu
- Graduate SchoolBengbu Medical CollegeBengbuAnhuiChina
| | - Zhuo‐Xun Wu
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's University, QueensNew YorkNew YorkUSA
| | - Deng Pan
- Graduate SchoolBengbu Medical CollegeBengbuAnhuiChina
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's University, QueensNew YorkNew YorkUSA
| | - Cong‐Hui Han
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical CollegeThe Affiliated Xuzhou Hospital of Medical College of Southeast UniversityThe Affiliated Xuzhou Center Hospital of Nanjing University of Chinese MedicineXuzhouJiangsuChina
| |
Collapse
|
10
|
Zhang L, Moccia M, Briggs DC, Bharate JB, Lakkaniga NR, Knowles P, Yan W, Tran P, Kharbanda A, Wang X, Leung YK, Frett B, Santoro M, McDonald NQ, Carlomagno F, Li HY. Discovery of N-Trisubstituted Pyrimidine Derivatives as Type I RET and RET Gatekeeper Mutant Inhibitors with a Novel Kinase Binding Pose. J Med Chem 2022; 65:1536-1551. [PMID: 35081714 PMCID: PMC10536133 DOI: 10.1021/acs.jmedchem.1c01280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mutations of the rearranged during transfection (RET) kinase are frequently reported in cancer, which make it as an attractive therapeutic target. Herein, we discovered a series of N-trisubstituted pyrimidine derivatives as potent inhibitors for both wild-type (wt) RET and RETV804M, which is a resistant mutant for several FDA-approved inhibitors. The X-ray structure of a representative inhibitor with RET revealed that the compound binds in a unique pose that bifurcates beneath the P-loop and confirmed the compound as a type I inhibitor. Through the structure-activity relationship (SAR) study, compound 20 was identified as a lead compound, showing potent inhibition of both RET and RETV804M. Additionally, compound 20 displayed potent antiproliferative activity of CCDC6-RET-driven LC-2/ad cells. Analysis of RET phosphorylation indicated that biological activity was mediated by RET inhibition. Collectively, N-trisubstituted pyrimidine derivatives could serve as scaffolds for the discovery and development of potent inhibitors of type I RET and its gatekeeper mutant for the treatment of RET-driven cancers.
Collapse
Affiliation(s)
- Lingtian Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Marialuisa Moccia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italia
| | - David C Briggs
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, U.K
| | - Jaideep B Bharate
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Naga Rajiv Lakkaniga
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Phillip Knowles
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, U.K
| | - Wei Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Phuc Tran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Anupreet Kharbanda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Xiuqi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Yuet-Kin Leung
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italia
| | - Neil Q McDonald
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, U.K
- Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck College, London WC1E 7HX, U.K
| | - Francesca Carlomagno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italia
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, 80131 Napoli, Italia
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
11
|
Sianglam P, Ngamdee K, Ngeontae W. Simultaneous preconcentration and fluorescence detection of ATP by a hybrid nanocomposite of magnetic nanoparticles incorporated in mixed metal hydroxide. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:188-198. [PMID: 34935797 DOI: 10.1039/d1ay01593a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A new approach for increasing the sensitivity of adenosine triphosphate (ATP) detection was demonstrated. The assay was based on the synergetic function of a hybrid nanocomposite (MNPs@MMH) composed of magnetic nanoparticles (MNPs) incorporated in a mixed metal hydroxide (MMH). MNPs@MMH can be utilized as an efficient green extractant and peroxidase catalyst. The trace level of ATP in the sample solution was first extracted by the MNPs@MMH hybrid nanocomposite through the ion exchange properties of MMH and adsorbed on the surface of the MNPs@MMH. The concentration of ATP was related to the fluorescence intensity of 2,3-diaminophenazine (DAP) generated from peroxidase-like activity of the MNPs in the presence of H2O2 and o-phenylenediamine (OPD). In the presence of ATP, the active surface of the MNPs was diminished, and the amount of DAP generated was reduced. Thus, the concentration of ATP was related to the degree of fluorescence decrease compared to the fluorescence intensity of the system without ATP. Based on the proposed strategy, a highly sensitive assay for ATP was achieved. This assay exhibited good selectivity for detection of ATP over derivatives and other common anions. The proposed assay allowed the detection of ATP in a concentration range of 2.5-20 μM with a detection limit of 0.41 μM.
Collapse
Affiliation(s)
- Pradthana Sianglam
- Department of Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Kessarin Ngamdee
- Department of Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand.
- Department of Chemistry, Center of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wittaya Ngeontae
- Department of Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand.
- Department of Chemistry, Center of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
- Research Center for Environmental and Hazardous Substance Management (EHSM), Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
12
|
Evaluation of Substituted Pyrazole-Based Kinase Inhibitors in One Decade (2011-2020): Current Status and Future Prospects. Molecules 2022; 27:molecules27010330. [PMID: 35011562 PMCID: PMC8747022 DOI: 10.3390/molecules27010330] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 11/17/2022] Open
Abstract
Pyrazole has been recognized as a pharmacologically important privileged scaffold whose derivatives produce almost all types of pharmacological activities and have attracted much attention in the last decades. Of the various pyrazole derivatives reported as potential therapeutic agents, this article focuses on pyrazole-based kinase inhibitors. Pyrazole-possessing kinase inhibitors play a crucial role in various disease areas, especially in many cancer types such as lymphoma, breast cancer, melanoma, cervical cancer, and others in addition to inflammation and neurodegenerative disorders. In this article, we reviewed the structural and biological characteristics of the pyrazole derivatives recently reported as kinase inhibitors and classified them according to their target kinases in a chronological order. We reviewed the reports including pyrazole derivatives as kinase inhibitors published during the past decade (2011-2020).
Collapse
|
13
|
Discovery of imidazo[1,2-a]pyridine-thiophene derivatives as FLT3 and FLT3 mutants inhibitors for acute myeloid leukemia through structure-based optimization of an NEK2 inhibitor. Eur J Med Chem 2021; 225:113776. [PMID: 34479037 DOI: 10.1016/j.ejmech.2021.113776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 11/20/2022]
Abstract
FMS-like tyrosine kinase 3 (FLT3) with an internal tandem duplication (ITD) mutation has been validated as a driver lesion and a therapeutic target for acute myeloid leukemia (AML). Currently, several potent small-molecule FLT3 kinase inhibitors are being evaluated or have completed evaluation in clinical trials. However, many of these inhibitors are challenged by the secondary mutations on kinase domain, especially the point mutations at the activation loop (D835) and gatekeeper residue (F691). To overcome the resistance challenge, we identified a novel series of imidazo[1,2-a]pyridine-thiophene derivatives from a NIMA-related kinase 2 (NEK2) kinase inhibitor CMP3a, which retained inhibitory activities on FTL3-ITDD835V and FLT3-ITDF691L. Through this study, we identified the imidazo[1,2-a]pyridine-thiophene derivatives as type-I inhibitors of FLT3. Moreover, we observed compound 5o as an inhibitor displaying equal anti-proliferative activities against FLT3-ITD, FTL3-ITDD835Y and FLT3-ITDF691L driven acute myeloid leukemia (AML) cell lines. Meanwhile, the apoptotic effects of compound supported its mechanism of anti-proliferative action. These results indicate that the imidazo[1,2-a]pyridine-thiophene scaffold is promising for targeting acquired resistance caused by FLT3 secondary mutations and compound 5o is an interesting lead in this direction.
Collapse
|
14
|
Indolin-2-one derivatives as selective Aurora B kinase inhibitors targeting breast cancer. Bioorg Chem 2021; 117:105451. [PMID: 34736137 DOI: 10.1016/j.bioorg.2021.105451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022]
Abstract
Aurora B is a pivotal cell cycle regulator where errors in its function results in polyploidy, genetic instability, and tumorigenesis. It is overexpressed in many cancers, consequently, targeting Aurora B with small molecule inhibitors constitutes a promising approach for anticancer therapy. Guided by structure-based design and molecular hybridization approach we developed a series of fifteen indolin-2-one derivatives based on a previously reported indolin-2-one-based multikinase inhibitor (1). Seven derivatives, 5g, 6a, 6c-e, 7, and 8a showed preferential antiproliferative activity in NCI-60 cell line screening and out of these, carbamate 6e and cyclopropylurea 8a derivatives showed optimum activity against Aurora B (IC50 = 16.2 and 10.5 nM respectively) and MDA-MB-468 cells (IC50 = 32.6 ± 9.9 and 29.1 ± 7.3 nM respectively). Furthermore, 6e and 8a impaired the clonogenic potential of MDA-MB-468 cells. Mechanistic investigations indicated that 6e and 8a induced G2/M cell cycle arrest, apoptosis, and necrosis of MDA-MB-468 cells and western blot analysis of 8a effect on MDA-MB-468 cells revealed 8a's ability to reduce Aurora B and its downstream target, Histone H3 phosphorylation. 6e and 8a displayed better safety profiles than multikinase inhibitors such as sunitinib, showing no cytotoxic effects on normal rat cardiomyoblasts and murine hepatocytes. Finally, 8a demonstrated a more selective profile than 1 when screened against ten related kinases. Based on these findings, 8a represents a promising candidate for further development to target breast cancer via Aurora B selective inhibition.
Collapse
|
15
|
Saha D, Ryan KR, Lakkaniga NR, Smith EL, Frett B. Pyrazoloadenine Inhibitors of the RET Lung Cancer Oncoprotein Discovered by a Fragment Optimization Approach. ChemMedChem 2021; 16:1605-1608. [PMID: 33559353 PMCID: PMC9969764 DOI: 10.1002/cmdc.202100013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/01/2021] [Indexed: 12/24/2022]
Abstract
A fragment-based drug-discovery approach was used on a pyrazoloadenine fragment library to uncover new molecules that target the RET (REarranged during Transfection) oncoprotein, which is a driver oncoprotein in ∼2 % of non-small-cell lung cancers. The fragment library was screened against the RET kinase and LC-2/ad (RET-driven), KM-12 (TRKA-driven matched control) and A549 (cytotoxic control) cells to identify selective scaffolds that could inhibit RET-driven growth. An unsubstituted pyrazoloadenine fragment was found to be active on RET in a biochemical assay, but reduced cell viability in non-RET-driven cell lines (EC50 =1 and 3 μM, respectively). To increase selectivity for RET, the pyrazoloadenine was modeled in the RET active site, and two domains were identified that were probed with pyrazoloadenine fragment derivatives to improve RET affinity. Scaffolds at each domain were merged to generate a novel lead compound, 8 p, which exhibited improved activity and selectivity for the RET oncoprotein (A549 EC50 =5.92 μM, LC-2/ad EC50 =0.016 μM, RET IC50 =0.000326 μM).
Collapse
Affiliation(s)
- Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharm
acy, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Katie Rose Ryan
- Department of Biochemistry and Molecular Biology, College
of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharm
acy, University of Arkansas for Medical Sciences, Little Rock, AR USA,SmartBio Labs, Chennai, India
| | - Erica Lane Smith
- Department of Pharmaceutical Sciences, College of Pharm
acy, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharm
acy, University of Arkansas for Medical Sciences, Little Rock, AR USA
| |
Collapse
|
16
|
Abstract
Introduction: Aurora kinases are a family of serine/threonine kinases, and promote mitotic spindle assembly by regulating centrosome duplication and separation. Aurora kinases are overexpressed in a variety of tumor cell lines, thus, the use of Aurora kinase small-molecule inhibitors has become a potential treatment option for cancer.Areas covered: As a continuing review of Aurora kinase inhibitors and their patents published in 2009, 2011 and 2014. Herein, we updated the information for Aurora kinase inhibitors in clinical trials and the patents filed from 2014 to 2020. PubMed, Scopus, SciFinder, and www.clinicaltrials.gov databases were used for searching the clinical information and patents of Aurora kinase inhibitors.Expert opinion: Even though Aurora A or B selective as well as pan inhibitors show preclinical and clinical efficacy, so far, no Aurora kinase inhibitor has been approved for clinical use. Preliminary evidence suggested that highly selective Aurora kinase or multi-target inhibitors as a single agent as well as in combination therapy are still the current main development trend of Aurora kinase inhibitors.
Collapse
Affiliation(s)
- Xue-Li Jing
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shi-Wu Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
17
|
Aurora Kinase B Inhibition: A Potential Therapeutic Strategy for Cancer. Molecules 2021; 26:molecules26071981. [PMID: 33915740 PMCID: PMC8037052 DOI: 10.3390/molecules26071981] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Aurora kinase B (AURKB) is a mitotic serine/threonine protein kinase that belongs to the aurora kinase family along with aurora kinase A (AURKA) and aurora kinase C (AURKC). AURKB is a member of the chromosomal passenger protein complex and plays a role in cell cycle progression. Deregulation of AURKB is observed in several tumors and its overexpression is frequently linked to tumor cell invasion, metastasis and drug resistance. AURKB has emerged as an attractive drug target leading to the development of small molecule inhibitors. This review summarizes recent findings pertaining to the role of AURKB in tumor development, therapy related drug resistance, and its inhibition as a potential therapeutic strategy for cancer. We discuss AURKB inhibitors that are in preclinical and clinical development and combination studies of AURKB inhibition with other therapeutic strategies.
Collapse
|
18
|
Yan W, Zhang L, Lv F, Moccia M, Carlomagno F, Landry C, Santoro M, Gosselet F, Frett B, Li HY. Discovery of pyrazolo-thieno[3,2-d]pyrimidinylamino-phenyl acetamides as type-II pan-tropomyosin receptor kinase (TRK) inhibitors: Design, synthesis, and biological evaluation. Eur J Med Chem 2021; 216:113265. [PMID: 33652352 DOI: 10.1016/j.ejmech.2021.113265] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 12/21/2022]
Abstract
Tropomyosin receptor kinase (TRK) represents an attractive oncology target for cancer therapy related to its critical role in cancer formation and progression. NTRK fusions are found to occur in 3.3% of lung cancers, 2.2% of colorectal cancers, 16.7% of thyroid cancers, 2.5% of glioblastomas, and 7.1% of pediatric gliomas. In this paper, we described the discovery of the type-II pan-TRK inhibitor 4c through the structure-based drug design strategy from the original hits 1b and 2b. Compound 4c exhibited excellent in vitro TRKA, TRKB, and TRKC kinase inhibitory activity and anti-proliferative activity against human colorectal carcinoma derived cell line KM12. In the NCI-60 human cancer cell lines screen, compound 4g demonstrated nearly 80% of growth inhibition for KM12, while only minimal inhibitory activity was observed for the remaining 59 cancer cell lines. Western blot analysis demonstrated that 4c and its urea cousin 4k suppressed the TPM3-TRKA autophosphorylation at the concentrations of 100 nM and 10 nM, respectively. The work presented that 2-(4-(thieno[3,2-d]pyrimidin-4-ylamino)phenyl)acetamides could serve as a novel scaffold for the discovery and development of type-II pan-TRK inhibitors for the treatment of TRK driven cancers.
Collapse
Affiliation(s)
- Wei Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Lingtian Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Fengping Lv
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Marialuisa Moccia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131, Naples, Italy
| | - Francesca Carlomagno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131, Naples, Italy; Istituto di Endocrinologia e Oncologia Sperimentale Del CNR, Via S Pansini 5, 80131, Naples, Italy
| | - Christophe Landry
- Blood Brain Barrier Laboratory (LBHE), University of Artois, UR2465, F-62300, Lens, France
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131, Naples, Italy
| | - Fabien Gosselet
- Blood Brain Barrier Laboratory (LBHE), University of Artois, UR2465, F-62300, Lens, France
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
19
|
Juillet C, Ermolenko L, Boyarskaya D, Baratte B, Josselin B, Nedev H, Bach S, Iorga BI, Bignon J, Ruchaud S, Al-Mourabit A. From Synthetic Simplified Marine Metabolite Analogues to New Selective Allosteric Inhibitor of Aurora B Kinase. J Med Chem 2021; 64:1197-1219. [PMID: 33417773 DOI: 10.1021/acs.jmedchem.0c02064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significant inhibition of Aurora B was achieved by the synthesis of simplified fragments of benzosceptrins and oroidin belonging to the marine pyrrole-2-aminoimidazoles metabolites isolated from sponges. Evaluation of kinase inhibition enabled the discovery of a synthetically accessible rigid acetylenic structural analogue EL-228 (1), whose structure could be optimized into the potent CJ2-150 (37). Here we present the synthesis of new inhibitors of Aurora B kinase, which is an important target for cancer therapy through mitosis regulation. The biologically oriented synthesis yielded several nanomolar inhibitors. The optimized compound CJ2-150 (37) showed a non-ATP competitive allosteric mode of action in a mixed-type inhibition for Aurora B kinase. Molecular docking identified a probable binding mode in the allosteric site "F" and highlighted the key interactions with the protein. We describe the improvement of the inhibitory potency and specificity of the novel scaffold as well as the characterization of the mechanism of action.
Collapse
Affiliation(s)
- Charlotte Juillet
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Ludmila Ermolenko
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Dina Boyarskaya
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Blandine Baratte
- Plateforme de Criblage KISSf, Station Biologique de Roscoff, Sorbonne Université, CNRS, FR 2424, Roscoff, 29680, France
| | - Béatrice Josselin
- Plateforme de Criblage KISSf, Station Biologique de Roscoff, Sorbonne Université, CNRS, FR 2424, Roscoff, 29680, France
| | - Hristo Nedev
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Stéphane Bach
- Plateforme de Criblage KISSf, Station Biologique de Roscoff, Sorbonne Université, CNRS, FR 2424, Roscoff, 29680, France.,Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, Sorbonne Université, CNRS, UMR 8227, Roscoff, 29680, France
| | - Bogdan I Iorga
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| | - Sandrine Ruchaud
- Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, Sorbonne Université, CNRS, UMR 8227, Roscoff, 29680, France
| | - Ali Al-Mourabit
- Institut de Chimie des Substances Naturelles, Université Paris-Saclay, CNRS, Gif-sur-Yvette, 91190, France
| |
Collapse
|