1
|
Wang C, Lu X. Targeting MET: Discovery of Small Molecule Inhibitors as Non-Small Cell Lung Cancer Therapy. J Med Chem 2023. [PMID: 37262349 DOI: 10.1021/acs.jmedchem.3c00028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
MET has been considered as a promising drug target for the treatment of MET-dependent diseases, particularly non-small cell lung cancer (NSCLC). Small molecule MET inhibitors with mainly three types of binding modes (Ia/Ib, II, and III) have been developed. In this Review, we provide an overview of the structural features, activation mechanism, and dysregulation pathway of MET and summarize progress on the development and discovery strategies utilized for MET inhibitors as well as mechanisms of acquired resistance to current approved inhibitors. The insights will accelerate discovery of new generation MET inhibitors to overcome clinical acquired resistance.
Collapse
Affiliation(s)
- Chaofan Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| | - Xiaoyun Lu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450001, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou, 510632, China
| |
Collapse
|
2
|
Charoensutthivarakul S, Thomas SE, Curran A, Brown KP, Belardinelli JM, Whitehouse AJ, Acebrón-García-de-Eulate M, Sangan J, Gramani SG, Jackson M, Mendes V, Floto RA, Blundell TL, Coyne AG, Abell C. Development of Inhibitors of SAICAR Synthetase (PurC) from Mycobacterium abscessus Using a Fragment-Based Approach. ACS Infect Dis 2022; 8:296-309. [PMID: 35037462 PMCID: PMC7614835 DOI: 10.1021/acsinfecdis.1c00432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mycobacterium abscessus (Mab) has emerged as a challenging threat to individuals with cystic fibrosis. Infections caused by this pathogen are often impossible to treat due to the intrinsic antibiotic resistance leading to lung malfunction and eventually death. Therefore, there is an urgent need to develop new drugs against novel targets in Mab to overcome drug resistance and subsequent treatment failure. In this study, SAICAR synthetase (PurC) from Mab was identified as a promising target for novel antibiotics. An in-house fragment library screen and a high-throughput X-ray crystallographic screen of diverse fragment libraries were explored to provide crucial starting points for fragment elaboration. A series of compounds developed from fragment growing and merging strategies, guided by crystallographic information and careful hit-to-lead optimization, have achieved potent nanomolar binding affinity against the enzyme. Some compounds also show a promising inhibitory effect against Mab and Mtb. This work utilizes a fragment-based design and demonstrates for the first time the potential to develop inhibitors against PurC from Mab.
Collapse
Affiliation(s)
- Sitthivut Charoensutthivarakul
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Sherine E Thomas
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Amy Curran
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Karen P Brown
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge CB23 3RE, United Kingdom
| | - Juan M Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
| | - Andrew J Whitehouse
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | | | - Jaspar Sangan
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge CB23 3RE, United Kingdom
| | - Subramanian G Gramani
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
| | - Vitor Mendes
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - R Andres Floto
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge CB23 3RE, United Kingdom
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Anthony G Coyne
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Chris Abell
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
3
|
Meng W, Chen T. Association between the HGF/c‑MET signaling pathway and tumorigenesis, progression and prognosis of hepatocellular carcinoma (Review). Oncol Rep 2021; 46:191. [PMID: 34278495 DOI: 10.3892/or.2021.8142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/10/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive and lethal malignancies with a rising incidence, and is characterized by rapid progression, frequent metastasis, late diagnosis, high postoperative recurrence and poor prognosis. Therefore, novel treatment strategies for HCC, particularly advanced HCC, are urgently required. The hepatocyte growth factor (HGF)/c‑mesenchymal‑epithelial transition receptor (c‑MET) axis is a key signaling pathway in HCC and is strongly associated with its highly malignant features. Available treatments based on HGF/c‑MET inhibition may prolong the lifespan of patients with HCC; however, they do not achieve the desired therapeutic effects. The aim of the present article was to review the basic knowledge regarding the role of the HGF/c‑MET signaling pathway in HCC, and examine the association between the HGF/c‑MET signaling pathway and the tumorigenesis, progression and prognosis of HCC.
Collapse
Affiliation(s)
- Wei Meng
- School of Medicine, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| | - Tao Chen
- School of Medicine, China Three Gorges University, Yichang, Hubei 443002, P.R. China
| |
Collapse
|
4
|
Robert‐Scott G, St‐Gelais J, Giguère D. Annulative Dimerization of Carbohydrates: Synthesis of Complex
C
2
‐Symmetrical 1,4‐Dioxane‐Sugar Hybrids. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Gabrielle Robert‐Scott
- Département de Chimie Université Laval 1045 av. De la Médecine Québec City, Qc G1V 0A6 Canada
| | - Jacob St‐Gelais
- Département de Chimie Université Laval 1045 av. De la Médecine Québec City, Qc G1V 0A6 Canada
| | - Denis Giguère
- Département de Chimie Université Laval 1045 av. De la Médecine Québec City, Qc G1V 0A6 Canada
| |
Collapse
|
5
|
Zhou MM, Chen G, Dang L. Enantioselective hydrosilylation of unsaturated carbon-heteroatom bonds (C[double bond, length as m-dash]N, C[double bond, length as m-dash]O) catalyzed by [Ru-S] complexes: a theoretical study. RSC Adv 2020; 10:9431-9437. [PMID: 35497244 PMCID: PMC9050042 DOI: 10.1039/c9ra10760f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/14/2020] [Indexed: 11/21/2022] Open
Abstract
A detailed theoretical study on the mechanism of enanthioselective hydrosilylation of imines and ketones catalyzed by the ruthenium(ii) thiolate catalyst [Ru–S] ([L*-Ru(SDmp)]+[BAr4F]−) with a chiral monodentate phosphine ligand is carried out in this work. We elucidate all the pathways leading to the main products or by products mediated by the [Ru–S] complex in order to have deep understanding of the chemoselectivity and enantioselectivity. The DFT (Density Functional Theory) calculations show that the reaction mechanism including: (1) Si–H bond cleavage by the dual activity of Ru–S bond; (2) the generation of a sulfur-stabilized silane cation; (3) the electrophilic attack of silane cation to NC/OC; (4) hydrogen transfer from Ru to carbon cation. The hydrosilylation products are found to be the final products rather than the dehydrogenative ones, which is consistent with the experimental results. The dehydrogenative silylation reaction pathways which give N- or O-silylated enamine/enol ether are reversible according to our calculations. The computational results also show that the electrophilic attack of silicon to NC/OC is the rate-determining step and the ee value can be improved significantly with more bulky model phosphine ligand based on the same calculation methods. A detailed theoretical study on the mechanism of enanthioselective hydrosilylation of imines and ketones catalyzed by the ruthenium(ii) thiolate catalyst with a chiral monodentate phosphine ligand is carried out in this work.![]()
Collapse
Affiliation(s)
- Miao-Miao Zhou
- Department of Chemistry, Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University Guangdong 515063 P. R. China
| | - Guanghui Chen
- Department of Chemistry, Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University Guangdong 515063 P. R. China
| | - Li Dang
- Department of Chemistry, Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University Guangdong 515063 P. R. China
| |
Collapse
|
6
|
Saha D, Kharbanda A, Yan W, Lakkaniga NR, Frett B, Li HY. The Exploration of Chirality for Improved Druggability within the Human Kinome. J Med Chem 2020; 63:441-469. [PMID: 31550151 PMCID: PMC10536157 DOI: 10.1021/acs.jmedchem.9b00640] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chirality is important in drug discovery because stereoselective drugs can ameliorate therapeutic difficulties including adverse toxicity and poor pharmacokinetic profiles. The human kinome, a major druggable enzyme class has been exploited to treat a wide range of diseases. However, many kinase inhibitors are planar and overlap in chemical space, which leads to selectivity and toxicity issues. By exploring chirality within the kinome, a new iteration of kinase inhibitors is being developed to better utilize the three-dimensional nature of the kinase active site. Exploration into novel chemical space, in turn, will also improve drug solubility and pharmacokinetic profiles. This perspective explores the role of chirality to improve kinome druggability and will serve as a resource for pioneering kinase inhibitor development to address current therapeutic needs.
Collapse
Affiliation(s)
- Debasmita Saha
- 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
| | - Wei Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharmacy, 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
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
7
|
Lv PC, Yang YS, Wang ZC. Recent Progress in the Development of Small Molecule c-Met Inhibitors. Curr Top Med Chem 2019; 19:1276-1288. [PMID: 31526339 DOI: 10.2174/1568026619666190712205353] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/24/2019] [Accepted: 06/28/2019] [Indexed: 02/08/2023]
Abstract
C-Met, also referred to as Hepatocyte Growth Factor Receptor (HGFR), is a heterodimeric
receptor tyrosine kinase. It has been determined that c-Met gene mutations, overexpression, and amplification
also occur in a variety of human tumor types, and these events are closely related to the aberrant
activation of the HGF/c-Met signaling pathway. Meanwhile, high c-Met expression is closely associated
with poor prognosis in cancer patients. The c-Met kinase has emerged as an attractive target for developing
antitumor agents. In this review, we cover the recent advances on the small molecule c-Met inhibitors
discovered from 2018 until now, with a main focus on the rational design, synthesis and structureactivity
relationship analysis.
Collapse
Affiliation(s)
- Peng-Cheng Lv
- Department of Chemistry, Purdue University, West Lafayette, Indiana, IN 47907, United States
| | - Yu-Shun Yang
- Department of Chemistry, Purdue University, West Lafayette, Indiana, IN 47907, United States
| | - Zhong-Chang Wang
- Department of Chemistry, Purdue University, West Lafayette, Indiana, IN 47907, United States
| |
Collapse
|
8
|
Discovery of N-(4-((7-(3-(4-ethylpiperazin-1-yl)propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)-N-(2-(2,6-difluorophenyl)-4-oxothiazolidin-3-yl)urea as a multi-tyrosine kinase inhibitor for drug-sensitive and drug-resistant cancers treatment. Eur J Med Chem 2019; 163:10-27. [DOI: 10.1016/j.ejmech.2018.11.057] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 11/23/2022]
|
9
|
Mane KD, Mukherjee A, Vanka K, Suryavanshi G. Metal-Free Regioselective Cross Dehydrogenative Coupling of Cyclic Ethers and Aryl Carbonyls. J Org Chem 2019; 84:2039-2047. [DOI: 10.1021/acs.joc.8b03048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Kishor D. Mane
- Chemical Engineering & Process Development Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110 025, India
| | - Anagh Mukherjee
- Physical and Material Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110 025, India
| | - Kumar Vanka
- Physical and Material Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110 025, India
| | - Gurunath Suryavanshi
- Chemical Engineering & Process Development Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110 025, India
| |
Collapse
|
10
|
Won SY, Kim SE, Kwon YJ, Shin I, Ham J, Kim WS. Chan–Lam coupling reaction of sulfamoyl azides with arylboronic acids for synthesis of unsymmetrical N-arylsulfamides. RSC Adv 2019; 9:2493-2497. [PMID: 35520509 PMCID: PMC9059883 DOI: 10.1039/c8ra09219b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/14/2019] [Indexed: 11/27/2022] Open
Abstract
An efficient method was developed for the synthesis of unsymmetrical N-arylsulfamides using sulfamoyl azides and arylboronic acids in the presence of 10 mol% of copper chloride as the catalyst. The reaction was facilitated in MeOH in an open flask at room temperature. Unlike the coupling of sulfamides and boronic acids, the use of sulfamoyl azides was found to be beneficial with respect to the yield and reaction time. An efficient method was developed for the synthesis of unsymmetrical N-arylsulfamides using sulfamoyl azides and arylboronic acids in the presence of 10 mol% of copper chloride as the catalyst.![]()
Collapse
Affiliation(s)
- Suk-Young Won
- Department of Chemistry and Nanoscience
- Ewha Womans University
- Seoul 03760
- South Korea
| | - Seo-Eun Kim
- Department of Chemistry and Nanoscience
- Ewha Womans University
- Seoul 03760
- South Korea
| | - Yong-Ju Kwon
- Department of Chemistry and Nanoscience
- Ewha Womans University
- Seoul 03760
- South Korea
| | - Inji Shin
- Therapeutics & Biotechnology Division/Innovative Therapeutics Research Center
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon 34602
- South Korea
- Department of Medicinal and Pharmaceutical Chemistry
| | - Jungyeob Ham
- Natural Product Research Institute
- Korea Institute of Science and Technology (KIST)
- Gangneung 25451
- South Korea
| | - Won-Suk Kim
- Department of Chemistry and Nanoscience
- Ewha Womans University
- Seoul 03760
- South Korea
| |
Collapse
|
11
|
Dehydrogenative reagent-free annulation of alkenes with diols for the synthesis of saturated O-heterocycles. Nat Commun 2018; 9:3551. [PMID: 30177691 PMCID: PMC6120897 DOI: 10.1038/s41467-018-06020-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/10/2018] [Indexed: 11/18/2022] Open
Abstract
Dehydrogenative annulation reactions are among the most straightforward and efficient approach for the preparation of cyclic structures. However, the applications of this strategy for the synthesis of saturated heterocycles have been rare. In addition, reported dehydrogenative bond-forming reactions commonly employ stoichiometric chemical oxidants, the use of which reduces the sustainability of the synthesis and brings safety and environmental issues. Herein, we report an organocatalyzed electrochemical dehydrogenative annulation reaction of alkenes with 1,2- and 1,3-diols for the synthesis of 1,4-dioxane and 1,4-dioxepane derivatives. The combination of electrochemistry and redox catalysis using an organic catalyst allows the electrosynthesis to proceed under transition metal- and oxidizing reagent-free conditions. In addition, the electrolytic method has a broad substrate scope and is compatible with many common functional groups, providing an efficient and straightforward access to functionalized 1,4-dioxane and 1,4-dioxepane products with diverse substitution patterns. Dehydrogenative annulation is a valuable approach to heterocycles, however, stoichiometric oxidants are often required. Here, the authors describe the electrochemical dehydrogenative annulation of diols and alkenes to generate dioxanes and dioxepanes under metal- and oxidant-free conditions.
Collapse
|
12
|
Blasius CK, Vasilenko V, Gade LH. Ultrafast Iron-Catalyzed Reduction of Functionalized Ketones: Highly Enantioselective Synthesis of Halohydrines, Oxaheterocycles, and Aminoalcohols. Angew Chem Int Ed Engl 2018; 57:10231-10235. [PMID: 29939466 DOI: 10.1002/anie.201806196] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Indexed: 11/08/2022]
Abstract
A molecularly defined chiral boxmi iron alkyl complex catalyzes the hydroboration of various functionalized ketones and provides the corresponding chiral halohydrines, oxaheterocycles (oxiranes, oxetanes, tetrahydrofurans, and dioxanes) and amino alcohols with excellent enantioselectivities (up to >99 %ee) and conversion efficiencies at low catalyst loadings (as low as 0.5 mol %). Turnover frequencies of greater than 40000 h-1 at -30 °C highlight the activity of this earth-abundant metal catalyst which tolerates a large number of functional groups.
Collapse
Affiliation(s)
- Clemens K Blasius
- Anorganisch-Chemisches Institut, Universität Heidelberg, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Vladislav Vasilenko
- Anorganisch-Chemisches Institut, Universität Heidelberg, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Lutz H Gade
- Anorganisch-Chemisches Institut, Universität Heidelberg, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| |
Collapse
|
13
|
Blasius CK, Vasilenko V, Gade LH. Ultrafast Iron-Catalyzed Reduction of Functionalized Ketones: Highly Enantioselective Synthesis of Halohydrines, Oxaheterocycles, and Aminoalcohols. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201806196] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Clemens K. Blasius
- Anorganisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Vladislav Vasilenko
- Anorganisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| | - Lutz H. Gade
- Anorganisch-Chemisches Institut; Universität Heidelberg; Im Neuenheimer Feld 270 69120 Heidelberg Germany
| |
Collapse
|
14
|
Parikh PK, Ghate MD. Recent advances in the discovery of small molecule c-Met Kinase inhibitors. Eur J Med Chem 2018; 143:1103-1138. [DOI: 10.1016/j.ejmech.2017.08.044] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/03/2017] [Accepted: 08/21/2017] [Indexed: 12/17/2022]
|
15
|
Jun JH, Kumar V, Dexheimer TS, Wedlich I, Nicklaus MC, Pommier Y, Malhotra SV. Synthesis, anti-cancer screening and tyrosyl-DNA phosphodiesterase 1 (Tdp1) inhibition activity of novel piperidinyl sulfamides. Eur J Pharm Sci 2018; 111:337-348. [PMID: 29037996 PMCID: PMC8087116 DOI: 10.1016/j.ejps.2017.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/06/2017] [Accepted: 10/11/2017] [Indexed: 10/18/2022]
Abstract
Novel piperidinyl-based sulfamide derivatives were designed and synthesized through various synthetic routes. Anticancer activities of these sulfamides were evaluated by phenotypic screening on National Cancer Institute's 60 human tumor cell lines (NCI-60). Preliminary screening at 10μM concentration showed that piperidinyl sulfamide aminoester 26 (NSC 749204) was sensitive to most of the cell lines in the panel. Further dose-response studies showed that 26 was highly selective for inhibition of colon cancer cell lines with minimum GI50=1.88μM for COLO-205 and maximum GI50=11.1μM for SW-620 cells. These newly synthesized sulfamides were also screening for their Tdp1 inhibition activity. Compound 18 (NSC 750706) showed significant inhibition of Tdp1 with IC50=23.7μM. Molecular-docking studies showed that 18 bind to Tdp1 in its binding pocket similar to a known Tdp1 inhibitor.
Collapse
Affiliation(s)
- Jung Ho Jun
- Department of Chemistry, University of Kansas, KS 66045, USA
| | - Vineet Kumar
- Department of Radiation Oncology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA
| | - Thomas S Dexheimer
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Iwona Wedlich
- Chemical Biology Laboratory, Frederick National Laboratory for Cancer Research, 376 Boyles Street, Frederick, MD 21702, USA
| | - Marc C Nicklaus
- Chemical Biology Laboratory, Frederick National Laboratory for Cancer Research, 376 Boyles Street, Frederick, MD 21702, USA
| | - Yves Pommier
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sanjay V Malhotra
- Department of Radiation Oncology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
16
|
Xu Y, Li L, Wang Y, Xing J, Zhou L, Zhong D, Luo X, Jiang H, Chen K, Zheng M, Deng P, Chen X. Aldehyde Oxidase Mediated Metabolism in Drug-like Molecules: A Combined Computational and Experimental Study. J Med Chem 2017; 60:2973-2982. [DOI: 10.1021/acs.jmedchem.7b00019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yuan Xu
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Liang Li
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yulan Wang
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Jing Xing
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Lei Zhou
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Dafang Zhong
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaomin Luo
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kaixian Chen
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mingyue Zheng
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pan Deng
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoyan Chen
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
17
|
Yuan C, Tu G, Zhao Y. Rhodium(III)-Catalyzed Selective Monoarylation of β or γ C(sp3)–H Bonds Assisted by a Trimethylpyrazole Group. Org Lett 2016; 19:356-359. [DOI: 10.1021/acs.orglett.6b03522] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Chunchen Yuan
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, China
| | - Guangliang Tu
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, China
| | - Yingsheng Zhao
- Key Laboratory of Organic
Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering
and Materials Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
18
|
Xiong X, Yeung YY. Highlyortho-Selective Chlorination of Anilines Using a Secondary Ammonium Salt Organocatalyst. Angew Chem Int Ed Engl 2016; 55:16101-16105. [DOI: 10.1002/anie.201607388] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/11/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Xiaodong Xiong
- Department of Chemistry; The Chinese University of Hong Kong, Shatin; N.T. Hong Kong China
| | - Ying-Yeung Yeung
- Department of Chemistry; The Chinese University of Hong Kong, Shatin; N.T. Hong Kong China
| |
Collapse
|
19
|
Xiong X, Yeung YY. Highlyortho-Selective Chlorination of Anilines Using a Secondary Ammonium Salt Organocatalyst. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201607388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Xiaodong Xiong
- Department of Chemistry; The Chinese University of Hong Kong, Shatin; N.T. Hong Kong China
| | - Ying-Yeung Yeung
- Department of Chemistry; The Chinese University of Hong Kong, Shatin; N.T. Hong Kong China
| |
Collapse
|
20
|
Abstract
![]()
Pd-catalyzed
cross-coupling reactions that form C–N bonds
have become useful methods to synthesize anilines and aniline derivatives,
an important class of compounds throughout chemical research. A key
factor in the widespread adoption of these methods has been the continued
development of reliable and versatile catalysts that function under
operationally simple, user-friendly conditions. This review provides
an overview of Pd-catalyzed N-arylation reactions found in both basic
and applied chemical research from 2008 to the present. Selected examples
of C–N cross-coupling reactions between nine classes of nitrogen-based
coupling partners and (pseudo)aryl halides are described for the synthesis
of heterocycles, medicinally relevant compounds, natural products,
organic materials, and catalysts.
Collapse
Affiliation(s)
- Paula Ruiz-Castillo
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Stephen L Buchwald
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| |
Collapse
|
21
|
Chang K, Karnad A, Zhao S, Freeman JW. Roles of c-Met and RON kinases in tumor progression and their potential as therapeutic targets. Oncotarget 2016; 6:3507-18. [PMID: 25784650 PMCID: PMC4414132 DOI: 10.18632/oncotarget.3420] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/21/2015] [Indexed: 02/06/2023] Open
Abstract
c-Met and receptor originated from nantes (RON) are structurally related transmembrane phosphotyrosine kinase receptors. c-Met and RON show increased expression or activity in a variety of tumors leading to tumor progression and may play a role in acquired resistance to therapy. Although often co-expressed, the distinct functional roles of c-Met and RON are not fully understood. c-Met and RON form both activated homodimers and heterodimers with themselves and other families of phosphotyrosine kinase receptors. Inhibitors for c-Met and RON including small molecular weigh kinase inhibitors and neutralizing antibodies are in pre-clinical investigation and clinical trials. Several of the tyrosine kinase inhibitors have activity against both c-Met and RON kinases whereas the antibodies generally are target specific. As with many targeted agents used to treat solid tumors, it is likely that c-Met/RON inhibitors will have greater benefit when used in combination with chemotherapy or other targeted agents. A careful analysis of c-Met/RON expression or activity and a better elucidation of how they influence cell signaling will be useful in predicting which tumors respond best to these inhibitors as well as determining which agents can be used with these inhibitors for combined therapy.
Collapse
Affiliation(s)
- Katherine Chang
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA
| | - Anand Karnad
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA
| | - Shujie Zhao
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - James W Freeman
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, TX, USA.,Research and Development, Audie Murphy Veterans Administration Hospital, San Antonio, TX, USA
| |
Collapse
|
22
|
Liu Y, Jin S, Peng X, Lu D, Zeng L, Sun Y, Ai J, Geng M, Hu Y. Pyridazinone derivatives displaying highly potent and selective inhibitory activities against c-Met tyrosine kinase. Eur J Med Chem 2016; 108:322-333. [DOI: 10.1016/j.ejmech.2015.11.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 02/02/2023]
|
23
|
Ruminski PG, Massa M, Strohbach J, Hanau CE, Schmidt M, Scholten JA, Fletcher TR, Hamper BC, Carroll JN, Shieh HS, Caspers N, Collins B, Grapperhaus M, Palmquist KE, Collins J, Baldus JE, Hitchcock J, Kleine HP, Rogers MD, McDonald J, Munie GE, Messing DM, Portolan S, Whiteley LO, Sunyer T, Schnute ME. Discovery of N-(4-Fluoro-3-methoxybenzyl)-6-(2-(((2S,5R)-5-(hydroxymethyl)-1,4-dioxan-2-yl)methyl)-2H-tetrazol-5-yl)-2-methylpyrimidine-4-carboxamide. A Highly Selective and Orally Bioavailable Matrix Metalloproteinase-13 Inhibitor for the Potential Treatment of Osteoarthritis. J Med Chem 2015; 59:313-27. [PMID: 26653735 DOI: 10.1021/acs.jmedchem.5b01434] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Matrix metalloproteinase-13 (MMP-13) is a zinc-dependent protease responsible for the cleavage of type II collagen, the major structural protein of articular cartilage. Degradation of this cartilage matrix leads to the development of osteoarthritis. We previously have described highly potent and selective carboxylic acid containing MMP-13 inhibitors; however, nephrotoxicity in preclinical toxicology species precluded development. The accumulation of compound in the kidneys mediated by human organic anion transporter 3 (hOAT3) was hypothesized as a contributing factor for the finding. Herein we report our efforts to optimize the MMP-13 potency and pharmacokinetic properties of non-carboxylic acid leads resulting in the identification of compound 43a lacking the previously observed preclinical toxicology at comparable exposures.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Dean M Messing
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer , 700 North Main Street, Cambridge, Massachusetts 02139, United States
| | - Silvia Portolan
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer , 700 North Main Street, Cambridge, Massachusetts 02139, United States
| | - Laurence O Whiteley
- Drug Safety, Pfizer , 1 Burtt Road, Andover, Massachusetts 01810, United States
| | | | | |
Collapse
|
24
|
Yang W, Sun J. Organocatalytic Enantioselective Synthesis of 1,4-Dioxanes and Other Oxa-Heterocycles by Oxetane Desymmetrization. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201509888] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Wen Yang
- Department of Chemistry; The Hong Kong University of Science and Technology; Clear Water Bay Kowloon Hong Kong SAR China
| | - Jianwei Sun
- Department of Chemistry; The Hong Kong University of Science and Technology; Clear Water Bay Kowloon Hong Kong SAR China
| |
Collapse
|
25
|
Yang W, Sun J. Organocatalytic Enantioselective Synthesis of 1,4-Dioxanes and Other Oxa-Heterocycles by Oxetane Desymmetrization. Angew Chem Int Ed Engl 2015; 55:1868-71. [DOI: 10.1002/anie.201509888] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Wen Yang
- Department of Chemistry; The Hong Kong University of Science and Technology; Clear Water Bay Kowloon Hong Kong SAR China
| | - Jianwei Sun
- Department of Chemistry; The Hong Kong University of Science and Technology; Clear Water Bay Kowloon Hong Kong SAR China
| |
Collapse
|
26
|
Baladi T, Abet V, Piguel S. State-of-the-art of small molecule inhibitors of the TAM family: the point of view of the chemist. Eur J Med Chem 2015; 105:220-37. [PMID: 26498569 DOI: 10.1016/j.ejmech.2015.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/01/2015] [Accepted: 10/03/2015] [Indexed: 01/04/2023]
Abstract
The TAM family of tyrosine kinases receptors (Tyro3, Axl and Mer) is implicated in cancer development, autoimmune reactions and viral infection and is therefore emerging as an effective and attractive therapeutic target. To date, only a few small molecules have been intentionally designed to block the TAM kinases, while most of the inhibitors were developed for blocking different protein kinases and then identified through selectivity profile studies. This minireview will examine in terms of chemical structure the different compounds able to act on either one, two or three TAM kinases with details about structure-activity relationships, drug-metabolism and pharmacokinetics properties where they exist.
Collapse
Affiliation(s)
- Tom Baladi
- Institut Curie/UMR9187-U1196, 91405 Orsay cedex, France; Univ Paris Sud, 91405 Orsay cedex, France
| | | | - Sandrine Piguel
- Institut Curie/UMR9187-U1196, 91405 Orsay cedex, France; Univ Paris Sud, 91405 Orsay cedex, France.
| |
Collapse
|
27
|
Wu JF, Liu MM, Huang SX, Wang Y. Design and synthesis of novel substituted naphthyridines as potential c-Met kinase inhibitors based on MK-2461. Bioorg Med Chem Lett 2015; 25:3251-5. [DOI: 10.1016/j.bmcl.2015.05.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 12/14/2022]
|
28
|
Xing W, Ai J, Jin S, Shi Z, Peng X, Wang L, Ji Y, Lu D, Liu Y, Geng M, Hu Y. Enhancing the cellular anti-proliferation activity of pyridazinones as c-met inhibitors using docking analysis. Eur J Med Chem 2015; 95:302-12. [DOI: 10.1016/j.ejmech.2015.03.041] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/20/2023]
|
29
|
Padda S, Neal JW, Wakelee HA. MET inhibitors in combination with other therapies in non-small cell lung cancer. Transl Lung Cancer Res 2015; 1:238-53. [PMID: 25806189 DOI: 10.3978/j.issn.2218-6751.2012.10.08] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 10/25/2012] [Indexed: 12/21/2022]
Abstract
MET and its ligand hepatocyte growth factor/scatter factor (HGF) influence cell motility and lead to tumor growth, invasion, and angiogenesis. Alterations in MET have been observed in non-small cell lung cancer (NSCLC) tumors, with increased expression associated with more aggressive cancer, as well as acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI). MET inhibitors act via two basic mechanisms. Small molecule inhibitors antagonize ATP in the intracellular tyrosine kinase domain of MET, with studies on the following agents reviewed here: tivantinib (ARQ-197), cabozantinib (XL-184), crizotinib (PF-02341066), amuvatinib (MP470), MGCD265, foretinib (EXEL-2880), MK2461, SGX523, PHA665752, JNJ-38877605, SU11274, and K252A. The monoclonal monovalent antibody fragment onartuzumab (MetMAb) is also discussed here, which binds to and prevents the extracellular activation of the receptor by ligand. MET inhibition may both overcome the negative prognostic effect of MET tumor expression as well as antagonize MET-dependent acquired resistance to EGFR inhibitors. Here we discuss MET inhibitors in combination with other therapies in lung cancer.
Collapse
Affiliation(s)
- Sukhmani Padda
- Stanford University/Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA 94305-5826, USA
| | - Joel W Neal
- Stanford University/Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA 94305-5826, USA
| | - Heather A Wakelee
- Stanford University/Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA 94305-5826, USA
| |
Collapse
|
30
|
Molander GA, Traister KM, O’Neill BT. Engaging Nonaromatic, Heterocyclic Tosylates in Reductive Cross-Coupling with Aryl and Heteroaryl Bromides. J Org Chem 2015; 80:2907-11. [DOI: 10.1021/acs.joc.5b00135] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Gary A. Molander
- Roy
and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Kaitlin M. Traister
- Roy
and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Brian T. O’Neill
- Pfizer Worldwide
Medicinal Chemistry, Eastern Point
Road, Groton, Connecticut 06340, United States
| |
Collapse
|
31
|
Ma Y, Sun G, Chen D, Peng X, Chen YL, Su Y, Ji Y, Liang J, Wang X, Chen L, Ding J, Xiong B, Ai J, Geng M, Shen J. Design and optimization of a series of 1-sulfonylpyrazolo[4,3-b]pyridines as selective c-Met inhibitors. J Med Chem 2015; 58:2513-29. [PMID: 25668160 DOI: 10.1021/jm502018y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
c-Met has emerged as an attractive target for targeted cancer therapy because of its abnormal activation in many cancer cells. To identify high potent and selective c-Met inhibitors, we started with profiling the potency and in vitro metabolic stability of a reported hit 7. By rational design, a novel sulfonylpyrazolo[4,3-b]pyridine 9 with improved DMPK properties was discovered. Further elaboration of π-π stacking interactions and solvent accessible polar moieties led to a series of highly potent and selective type I c-Met inhibitors. On the basis of in vitro and in vivo pharmacological and pharmacokinetics studies, compound 46 was selected as a preclinical candidate for further anticancer drug development.
Collapse
Affiliation(s)
- Yuchi Ma
- Department of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kodolins O, Lusis V, Muceniece D. Synthesis and Reactions of Methyl 8-Aryl-6-Methyl-9-Oxo-8,9-Dihydro-5H-Dibenzo[4,5:6,7]Cyclohepta[1,2-b]Pyridine-7-Carboxylates. Chem Heterocycl Compd (N Y) 2014. [DOI: 10.1007/s10593-014-1589-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
33
|
Zhang W, DeRyckere D, Hunter D, Liu J, Stashko M, Minson KA, Cummings CT, Lee M, Glaros TG, Newton DL, Sather S, Zhang D, Kireev D, Janzen W, Earp HS, Graham DK, Frye SV, Wang X. UNC2025, a potent and orally bioavailable MER/FLT3 dual inhibitor. J Med Chem 2014; 57:7031-41. [PMID: 25068800 PMCID: PMC4148167 DOI: 10.1021/jm500749d] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Indexed: 12/14/2022]
Abstract
We previously reported a potent small molecule Mer tyrosine kinase inhibitor UNC1062. However, its poor PK properties prevented further assessment in vivo. We report here the sequential modification of UNC1062 to address DMPK properties and yield a new potent and highly orally bioavailable Mer inhibitor, 11, capable of inhibiting Mer phosphorylation in vivo, following oral dosing as demonstrated by pharmaco-dynamic (PD) studies examining phospho-Mer in leukemic blasts from mouse bone marrow. Kinome profiling versus more than 300 kinases in vitro and cellular selectivity assessments demonstrate that 11 has similar subnanomolar activity against Flt3, an additional important target in acute myelogenous leukemia (AML), with pharmacologically useful selectivity versus other kinases examined.
Collapse
Affiliation(s)
- Weihe Zhang
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Deborah DeRyckere
- Department
of Pediatrics, School of Medicine, University
of Colorado Denver, Aurora, Colorado 80045, United States
| | - Debra Hunter
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jing Liu
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Michael
A. Stashko
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Katherine A. Minson
- Department
of Pediatrics, School of Medicine, University
of Colorado Denver, Aurora, Colorado 80045, United States
| | - Christopher T. Cummings
- Department
of Pediatrics, School of Medicine, University
of Colorado Denver, Aurora, Colorado 80045, United States
| | - Minjung Lee
- Department
of Pediatrics, School of Medicine, University
of Colorado Denver, Aurora, Colorado 80045, United States
| | - Trevor G. Glaros
- Biological
Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for
Cancer Research, Frederick, Maryland 21702, United States
| | - Dianne L. Newton
- Biological
Testing Branch, Developmental Therapeutics Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for
Cancer Research, Frederick, Maryland 21702, United States
| | - Susan Sather
- Department
of Pediatrics, School of Medicine, University
of Colorado Denver, Aurora, Colorado 80045, United States
| | - Dehui Zhang
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Dmitri Kireev
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - William
P. Janzen
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - H. Shelton Earp
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Douglas K. Graham
- Department
of Pediatrics, School of Medicine, University
of Colorado Denver, Aurora, Colorado 80045, United States
| | - Stephen V. Frye
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Department of Pharmacology,
School of Medicine, and Lineberger Comprehensive Cancer Center, Department
of Medicine, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
34
|
Mathur A, Ware C, Davis L, Gazdar A, Pan BS, Lutterbach B. FGFR2 is amplified in the NCI-H716 colorectal cancer cell line and is required for growth and survival. PLoS One 2014; 9:e98515. [PMID: 24968263 PMCID: PMC4072591 DOI: 10.1371/journal.pone.0098515] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/02/2014] [Indexed: 12/21/2022] Open
Abstract
Aberrant kinase activation resulting from mutation, amplification, or translocation can drive growth and survival in a subset of human cancer. FGFR2 is amplified in breast and gastric cancer, and we report here the first characterization of FGFR2 gene amplification in colorectal cancer in the NCI-H716 colorectal cancer cell line. FGFR2 is highly expressed and activated in NCI-H716 cells, and FGFR selective small molecule inhibitors or FGFR2 shRNA strongly inhibited cell viability in vitro, indicating “addiction” of NCI-H716 cells to FGFR2. NCI-H716 growth in a xenograft model was also inhibited by an FGFR small molecule inhibitor. FGFR2 was required for activation of multiple downstream signaling proteins including AKT, ERK, S6RP and NFKB. Inhibition of downstream kinases such as AKT or ERK alone had modest effects on proliferation, whereas combined inhibition of AKT and ERK signaling resulted in a loss of viability similar to FGFR2 inhibition. We identified elevated FGFR2 expression in a small subset of primary colorectal cancer, however FGFR2 amplification was not observed. Although FGFR2 amplification is not common in primary colon cancer or lymph node and liver metastases, other subsets of colorectal cancer such as ascites, from which the NCI-H716 cell line was derived, have yet to be tested. These results suggest that emerging FGFR inhibitor therapeutics may have efficacy in a subset of colon cancer driven by FGFR2 amplification.
Collapse
Affiliation(s)
- Anjili Mathur
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Christopher Ware
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Lenora Davis
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Adi Gazdar
- Hamon Center for Therapeutic Oncology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Bo-Sheng Pan
- Merck Research Labs, Boston, Massachusetts, United States of America
| | - Bart Lutterbach
- Merck Research Labs, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
35
|
Gao W, Xing X, Li Y, Lan S. A novel construction of quino-fused tropone skeleton: first synthesis of 12H-benzo[4,5]cyclohepta[1,2-b]quinolin-12-one derivatives. Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.01.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
36
|
Aoki T, Hyohdoh I, Furuichi N, Ozawa S, Watanabe F, Matsushita M, Sakaitani M, Ori K, Takanashi K, Harada N, Tomii Y, Tabo M, Yoshinari K, Aoki Y, Shimma N, Iikura H. The sulfamide moiety affords higher inhibitory activity and oral bioavailability to a series of coumarin dual selective RAF/MEK inhibitors. Bioorg Med Chem Lett 2013; 23:6223-7. [DOI: 10.1016/j.bmcl.2013.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 10/26/2022]
|
37
|
Zhang D, Zhang X, Ai J, Zhai Y, Liang Z, Wang Y, Chen Y, Li C, Zhao F, Jiang H, Geng M, Luo C, Liu H. Synthesis and biological evaluation of 2-amino-5-aryl-3-benzylthiopyridine scaffold based potent c-Met inhibitors. Bioorg Med Chem 2013; 21:6804-20. [DOI: 10.1016/j.bmc.2013.07.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 07/14/2013] [Accepted: 07/15/2013] [Indexed: 11/28/2022]
|
38
|
Yuan H, Tai W, Hu S, Liu H, Zhang Y, Yao S, Ran T, Lu S, Ke Z, Xiong X, Xu J, Chen Y, Lu T. Fragment-based strategy for structural optimization in combination with 3D-QSAR. J Comput Aided Mol Des 2013; 27:897-915. [DOI: 10.1007/s10822-013-9687-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/24/2013] [Indexed: 12/14/2022]
|
39
|
Li Y, Chang M, Liu R, Lin G, Gao W. A facile synthesis of (E)-3-styryl-1-phenyl cyclohepta[c]pyrazol-8(1H)-ones. RESEARCH ON CHEMICAL INTERMEDIATES 2013. [DOI: 10.1007/s11164-012-0785-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
40
|
Abstract
Since the discovery of MSP (macrophage-stimulating protein; also known as MST1 and hepatocyte growth factor-like (HGFL)) as the ligand for the receptor tyrosine kinase RON (also known as MST1R) in the early 1990s, the roles of this signalling axis in cancer pathogenesis has been extensively studied in various model systems. Both in vitro and in vivo evidence has revealed that MSP-RON signalling is important for the invasive growth of different types of cancers. Currently, small-molecule inhibitors and antibodies blocking RON signalling are under investigation. Substantial responses have been achieved in human tumour xenograft models, laying the foundation for clinical validation. In this Review, we discuss recent advances that demonstrate the importance of MSP-RON signalling in cancer and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hang-Ping Yao
- Viral Oncogenesis Section in State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P. R. China
| | | | | | | |
Collapse
|
41
|
Northrup AB, Katcher MH, Altman MD, Chenard M, Daniels MH, Deshmukh SV, Falcone D, Guerin DJ, Hatch H, Li C, Lu W, Lutterbach B, Allison TJ, Patel SB, Reilly JF, Reutershan M, Rickert KW, Rosenstein C, Soisson SM, Szewczak AA, Walker D, Wilson K, Young JR, Pan BS, Dinsmore CJ. Discovery of 1-[3-(1-methyl-1H-pyrazol-4-yl)-5-oxo-5H-benzo[4,5]cyclohepta[1,2-b]pyridin-7-yl]-N-(pyridin-2-ylmethyl)methanesulfonamide (MK-8033): A Specific c-Met/Ron dual kinase inhibitor with preferential affinity for the activated state of c-Met. J Med Chem 2013; 56:2294-310. [PMID: 23379595 DOI: 10.1021/jm301619u] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This report documents the first example of a specific inhibitor of protein kinases with preferential binding to the activated kinase conformation: 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one 11r (MK-8033), a dual c-Met/Ron inhibitor under investigation as a treatment for cancer. The design of 11r was based on the desire to reduce time-dependent inhibition of CYP3A4 (TDI) by members of this structural class. A novel two-step protocol for the synthesis of benzylic sulfonamides was developed to access 11r and analogues. We provide a rationale for the observed selectivity based on X-ray crystallographic evidence and discuss selectivity trends with additional examples. Importantly, 11r provides full inhibition of tumor growth in a c-Met amplified (GTL-16) subcutaneous tumor xenograft model and may have an advantage over inactive form kinase inhibitors due to equal potency against a panel of oncogenic activating mutations of c-Met in contrast to c-Met inhibitors without preferential binding to the active kinase conformation.
Collapse
Affiliation(s)
- Alan B Northrup
- Department of Chemistry, Merck & Co., Inc. , 33 Avenue Louis Pasteur, BMB-3, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Abstract
Several phenylethyl-substituted pyridinecarboxaldehydes were prepared from 2-bromo-3-pyridinecarboxaldehyde and these substances are found to undergo cyclization reactions in acidic media. In the absence of added nucleophile, acid promoted cyclization and oxidation (MnO2) provides an efficient route to 10,11-dihydro-5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-ones. Arene nucleophiles may also be added to the acidic mixture to provide good yields of triarylmethane products. Mechanisms are proposed involving dicationic superelectrophilic intermediates.
Collapse
Affiliation(s)
| | - Douglas A. Klumpp
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, Illinois 60115
| |
Collapse
|
44
|
Cui JJ, McTigue M, Nambu M, Tran-Dubé M, Pairish M, Shen H, Jia L, Cheng H, Hoffman J, Le P, Jalaie M, Goetz GH, Ryan K, Grodsky N, Deng YL, Parker M, Timofeevski S, Murray BW, Yamazaki S, Aguirre S, Li Q, Zou H, Christensen J. Discovery of a Novel Class of Exquisitely Selective Mesenchymal-Epithelial Transition Factor (c-MET) Protein Kinase Inhibitors and Identification of the Clinical Candidate 2-(4-(1-(Quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (PF-04217903) for the Treatment of Cancer. J Med Chem 2012; 55:8091-109. [DOI: 10.1021/jm300967g] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- J. Jean Cui
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Michele McTigue
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Mitchell Nambu
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Michelle Tran-Dubé
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Mason Pairish
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Hong Shen
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Lei Jia
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Hengmiao Cheng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Jacqui Hoffman
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Phuong Le
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Mehran Jalaie
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Gilles H. Goetz
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Kevin Ryan
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Neil Grodsky
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Ya-li Deng
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Max Parker
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Sergei Timofeevski
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Brion W. Murray
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Shinji Yamazaki
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Shirley Aguirre
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Qiuhua Li
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - Helen Zou
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| | - James Christensen
- La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive,
San Diego, California 92121, United States
| |
Collapse
|
45
|
Discovery of novel 2-aminopyridine-3-carboxamides as c-Met kinase inhibitors. Bioorg Med Chem 2012; 20:5169-80. [DOI: 10.1016/j.bmc.2012.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 07/03/2012] [Accepted: 07/04/2012] [Indexed: 12/21/2022]
|
46
|
Exploring a sulfone linker utilizing trimethyl aluminum as a cleavage reagent: solid-phase synthesis of sulfonamides and ureas. Mol Divers 2012; 16:463-76. [DOI: 10.1007/s11030-012-9380-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 06/05/2012] [Indexed: 01/04/2023]
|