1
|
Dunker C, Schlegel K, Junker A. Phenol (bio)isosteres in drug design and development. Arch Pharm (Weinheim) 2025; 358:e2400700. [PMID: 39580699 PMCID: PMC11726161 DOI: 10.1002/ardp.202400700] [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: 08/30/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
Due to their versatile properties, phenolic compounds are integral to various biologically active molecules, including many pharmaceuticals. However, their application in drug design is often hindered by issues such as poor oral bioavailability, rapid metabolism, and potential toxicity. This review explores the use of phenol bioisosteres-structurally similar compounds that can mimic the biological activity of phenols while potentially offering improved drug-like properties. We provide an extensive analysis of various phenol bioisosteres, including benzimidazolones, benzoxazolones, indoles, quinolinones, and pyridones, highlighting their impact on the pharmacokinetic and pharmacodynamic profiles of drugs. Case studies illustrate the successful application of these bioisosteres in enhancing metabolic stability, receptor selectivity, and overall therapeutic efficacy. Additionally, the review addresses the challenges associated with phenol bioisosterism, such as maintaining potency and avoiding undesirable side effects. By offering a detailed examination of current strategies and potential future directions, this review serves as a valuable resource for medicinal chemists seeking to optimize phenolic scaffolds in drug development. The insights provided herein aim to facilitate the design of more effective and safer therapeutic agents through strategic bioisosteric modifications.
Collapse
Affiliation(s)
- Calvin Dunker
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and RadiopharmacyUniversity of TübingenTübingenGermany
| | - Katja Schlegel
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and RadiopharmacyUniversity of TübingenTübingenGermany
| |
Collapse
|
2
|
Kurasawa O, Miyazaki T, Homma M, Oguro Y, Imada T, Uchiyama N, Iwai K, Yamamoto Y, Ohori M, Hara H, Sugimoto H, Iwata K, Skene R, Hoffman I, Ohashi A, Nomura T, Cho N. Discovery of a Novel, Highly Potent, and Selective Thieno[3,2- d]pyrimidinone-Based Cdc7 Inhibitor with a Quinuclidine Moiety (TAK-931) as an Orally Active Investigational Antitumor Agent. J Med Chem 2020; 63:1084-1104. [PMID: 31895562 DOI: 10.1021/acs.jmedchem.9b01427] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In our pursuit of developing a novel, potent, and selective cell division cycle 7 (Cdc7) inhibitor, we optimized the previously reported thieno[3,2-d]pyrimidinone analogue I showing time-dependent Cdc7 kinase inhibition and slow dissociation kinetics. These medicinal chemistry efforts led to the identification of compound 3d, which exhibited potent cellular activity, excellent kinase selectivity, and antitumor efficacy in a COLO205 xenograft mouse model. However, the issue of formaldehyde adduct formation emerged during a detailed study of 3d, which was deemed an obstacle to further development. A structure-based approach to circumvent the adduct formation culminated in the discovery of compound 11b (TAK-931) possessing a quinuclidine moiety as a preclinical candidate. In this paper, the design, synthesis, and biological evaluation of this series of compounds will be presented.
Collapse
Affiliation(s)
- Osamu Kurasawa
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Tohru Miyazaki
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Misaki Homma
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Yuya Oguro
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Takashi Imada
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Noriko Uchiyama
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Kenichi Iwai
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Yukiko Yamamoto
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Momoko Ohori
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Hideto Hara
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Hiroshi Sugimoto
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Kentaro Iwata
- Pharmaceutical Sciences , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Robert Skene
- Takeda California, Inc. , 10410 Science Center Drive , San Diego , California 92121 , United States
| | - Isaac Hoffman
- Takeda California, Inc. , 10410 Science Center Drive , San Diego , California 92121 , United States
| | - Akihiro Ohashi
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Toshiyuki Nomura
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Nobuo Cho
- Pharmaceutical Research Division , Takeda Pharmaceutical Company, Ltd. , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| |
Collapse
|
3
|
Davies RD, Pink JH, Scott JS, Bailey A. Synthesis of 8-substituted-6-phenyl-6,7,8,9-tetrahydro-3H-pyrazolo[4,3-f]isoquinolines using Pictet-Spengler and Bischler-Napieralski cyclisation methods. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.06.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
4
|
Kurasawa O, Oguro Y, Miyazaki T, Homma M, Mori K, Iwai K, Hara H, Skene R, Hoffman I, Ohashi A, Yoshida S, Ishikawa T, Cho N. Identification of a new class of potent Cdc7 inhibitors designed by putative pharmacophore model: Synthesis and biological evaluation of 2,3-dihydrothieno[3,2-d]pyrimidin-4(1H)-ones. Bioorg Med Chem 2017; 25:2133-2147. [PMID: 28284870 DOI: 10.1016/j.bmc.2017.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 11/17/2022]
Abstract
Cell division cycle 7 (Cdc7) is a serine/threonine kinase that plays important roles in the regulation of DNA replication process. A genetic study indicates that Cdc7 inhibition can induce selective tumor-cell death in a p53-dependent manner, suggesting that Cdc7 is an attractive target for the treatment of cancers. In order to identify a new class of potent Cdc7 inhibitors, we generated a putative pharmacophore model based on in silico docking analysis of a known inhibitor with Cdc7 homology model. The pharmacophore model provided a minimum structural motif of Cdc7 inhibitor, by which preliminary medicinal chemistry efforts identified a dihydrothieno[3,2-d]-pyrimidin-4(1H)-one scaffold having a heteroaromatic hinge-binding moiety. The structure-activity relationship (SAR) studies resulted in the discovery of new, potent, and selective Cdc7 inhibitors 14a, c, e. Furthermore, the high selectivity of 14c, e for Cdc7 over Rho-associated protein kinase 1 (ROCK1) is discussed by utilizing a docking study with Cdc7 and ROCK2 crystal structures.
Collapse
Affiliation(s)
- Osamu Kurasawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Yuya Oguro
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tohru Miyazaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Misaki Homma
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kouji Mori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kenichi Iwai
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hideto Hara
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Robert Skene
- Takeda California, Inc., 10410 Science Center Drive, San Diego 92121, CA, USA
| | - Isaac Hoffman
- Takeda California, Inc., 10410 Science Center Drive, San Diego 92121, CA, USA
| | - Akihiro Ohashi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Sei Yoshida
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomoyasu Ishikawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Nobuo Cho
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
5
|
Ishchenko A, Zhang L, Le Brazidec JY, Fan J, Chong JH, Hingway A, Raditsis A, Singh L, Elenbaas B, Hong VS, Marcotte D, Silvian L, Enyedy I, Chao J. Structure-based design of low-nanomolar PIM kinase inhibitors. Bioorg Med Chem Lett 2015; 25:474-80. [DOI: 10.1016/j.bmcl.2014.12.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/09/2014] [Accepted: 12/12/2014] [Indexed: 10/24/2022]
|
6
|
Reichelt A, Bailis JM, Bartberger MD, Yao G, Shu H, Kaller MR, Allen JG, Weidner MF, Keegan KS, Dao JH. Synthesis and structure-activity relationship of trisubstituted thiazoles as Cdc7 kinase inhibitors. Eur J Med Chem 2014; 80:364-82. [PMID: 24793884 DOI: 10.1016/j.ejmech.2014.04.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/01/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
The Cell division cycle 7 (Cdc7) protein kinase is essential for DNA replication and maintenance of genome stability. We systematically explored thiazole-based compounds as inhibitors of Cdc7 kinase activity in cancer cells. Our studies resulted in the identification of a potent, selective Cdc7 inhibitor that decreased phosphorylation of the direct substrate MCM2 in vitro and in vivo, and inhibited DNA synthesis and cell viability in vitro.
Collapse
Affiliation(s)
- Andreas Reichelt
- Medicinal Chemistry, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Julie M Bailis
- Oncology Research, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98119, USA
| | - Michael D Bartberger
- Molecular Structure and Characterization, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Guomin Yao
- Medicinal Chemistry, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Hong Shu
- Medicinal Chemistry, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Matthew R Kaller
- Medicinal Chemistry, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - John G Allen
- Medicinal Chemistry, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Margaret F Weidner
- Oncology Research, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98119, USA
| | - Kathleen S Keegan
- Oncology Research, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98119, USA
| | - Jennifer H Dao
- Molecular Structure and Characterization, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| |
Collapse
|
7
|
Harrington PE, Bourbeau MP, Fotsch C, Frohn M, Pickrell AJ, Reichelt A, Sham K, Siegmund AC, Bailis JM, Bush T, Escobar S, Hickman D, Heller S, Hsieh F, Orf JN, Rong M, San Miguel T, Tan H, Zalameda L, Allen JG. The optimization of aminooxadiazoles as orally active inhibitors of Cdc7. Bioorg Med Chem Lett 2013; 23:6396-400. [PMID: 24120542 DOI: 10.1016/j.bmcl.2013.09.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 10/26/2022]
Abstract
A series of aminooxadiazoles was optimized for inhibition of Cdc7. Early lead isoquinoline 1 suffered from modest cell potency (cellular IC50=0.71 μM measuring pMCM2), low selectivity against structurally related kinases, and high IV clearance in rats (CL=18 L/h/kg). Extensive optimization resulted in azaindole 26 (Cdc7 IC50=1.1 nM, pMCM2 IC50=32 nM) that demonstrated robust lowering of pMCM2 in a mouse pharmacodynamic (PD) model when dosed orally. Modifications to improve the pharmacokinetic profile of this series were guided by trapping experiments with glutathione in rat hepatocytes.
Collapse
Affiliation(s)
- Paul E Harrington
- Medicinal Chemistry, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
van Linden OPJ, Kooistra AJ, Leurs R, de Esch IJP, de Graaf C. KLIFS: a knowledge-based structural database to navigate kinase-ligand interaction space. J Med Chem 2013; 57:249-77. [PMID: 23941661 DOI: 10.1021/jm400378w] [Citation(s) in RCA: 212] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protein kinases regulate the majority of signal transduction pathways in cells and have become important targets for the development of designer drugs. We present a systematic analysis of kinase-ligand interactions in all regions of the catalytic cleft of all 1252 human kinase-ligand cocrystal structures present in the Protein Data Bank (PDB). The kinase-ligand interaction fingerprints and structure database (KLIFS) contains a consistent alignment of 85 kinase ligand binding site residues that enables the identification of family specific interaction features and classification of ligands according to their binding modes. We illustrate how systematic mining of kinase-ligand interaction space gives new insights into how conserved and selective kinase interaction hot spots can accommodate the large diversity of chemical scaffolds in kinase ligands. These analyses lead to an improved understanding of the structural requirements of kinase binding that will be useful in ligand discovery and design studies.
Collapse
Affiliation(s)
- Oscar P J van Linden
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), VU University Amsterdam , De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
9
|
Lee SJ, Han BG, Cho JW, Choi JS, Lee J, Song HJ, Koh JS, Lee BI. Crystal structure of pim1 kinase in complex with a pyrido[4,3-d]pyrimidine derivative suggests a unique binding mode. PLoS One 2013; 8:e70358. [PMID: 23936194 PMCID: PMC3729456 DOI: 10.1371/journal.pone.0070358] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 06/18/2013] [Indexed: 02/07/2023] Open
Abstract
Human Pim1 kinase is a serine/threonine protein kinase that plays important biological roles in cell survival, apoptosis, proliferation, and differentiation. Moreover, Pim1 is up-regulated in various hematopoietic malignancies and solid tumors. Thus, Pim1 is an attractive target for cancer therapeutics, and there has been growing interest in developing small molecule inhibitors for Pim1. Here, we describe the crystal structure of Pim1 in complex with a newly developed pyrido[4,3-d]pyrimidine-derivative inhibitor (SKI-O-068). Our inhibitor exhibits a half maximum inhibitory concentration (IC50) of 123 (±14) nM and has an unusual binding mode in complex with Pim1 kinase. The interactions between SKI-O-068 and the Pim1 active site pocket residue are different from those of other scaffold inhibitor-bound structures. The binding mode analysis suggests that the SKI-O-068 inhibitor can be improved by introducing functional groups that facilitate direct interaction with Lys67, which aid in the design of an optimized inhibitor.
Collapse
Affiliation(s)
- Sang Jae Lee
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Byeong-Gu Han
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | - Jea-Won Cho
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | | | - Jaekyoo Lee
- Genosco, Cambridge, Massachusetts, United States of America
| | - Ho-Juhn Song
- Genosco, Cambridge, Massachusetts, United States of America
| | - Jong Sung Koh
- Oscotec Inc., Seongnam, Gyeonggi, Republic of Korea
- Genosco, Cambridge, Massachusetts, United States of America
- * E-mail: (JSK); (BIL)
| | - Byung Il Lee
- Biomolecular Function Research Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
- * E-mail: (JSK); (BIL)
| |
Collapse
|
10
|
Liachko NF, McMillan PJ, Guthrie CR, Bird TD, Leverenz JB, Kraemer BC. CDC7 inhibition blocks pathological TDP-43 phosphorylation and neurodegeneration. Ann Neurol 2013; 74:39-52. [PMID: 23424178 DOI: 10.1002/ana.23870] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/14/2012] [Accepted: 01/25/2013] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Kinase hyperactivity occurs in both neurodegenerative disease and cancer. Lesions containing hyperphosphorylated aggregated TDP-43 characterize amyotrophic lateral sclerosis and frontotemporal lobar degeneration with TDP-43 inclusions. Dual phosphorylation of TDP-43 at serines 409/410 (S409/410) drives neurotoxicity in disease models; therefore, TDP-43-specific kinases are candidate targets for intervention. METHODS To find therapeutic targets for the prevention of TDP-43 phosphorylation, we assembled and screened a comprehensive RNA interference library targeting kinases in TDP-43 transgenic Caenorhabditis elegans. RESULTS We show CDC7 robustly phosphorylates TDP-43 at pathological residues S409/410 in C. elegans, in vitro, and in human cell culture. In frontotemporal lobar degeneration (FTLD)-TDP cases, CDC7 immunostaining overlaps with the phospho-TDP-43 pathology found in frontal cortex. Furthermore, PHA767491, a small molecule inhibitor of CDC7, reduces TDP-43 phosphorylation and prevents TDP-43-dependent neurodegeneration in TDP-43-transgenic animals. INTERPRETATION Taken together, these data support CDC7 as a novel therapeutic target for TDP-43 proteinopathies, including FTLD-TDP and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Nicole F Liachko
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA; Department of Medicine, University of Washington, Seattle, WA
| | | | | | | | | | | |
Collapse
|
11
|
Bryan MC, Falsey JR, Frohn M, Reichelt A, Yao G, Bartberger MD, Bailis JM, Zalameda L, Miguel TS, Doherty EM, Allen JG. N-substituted azaindoles as potent inhibitors of Cdc7 kinase. Bioorg Med Chem Lett 2013; 23:2056-60. [DOI: 10.1016/j.bmcl.2013.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 01/28/2013] [Accepted: 02/01/2013] [Indexed: 01/30/2023]
|
12
|
|
13
|
Tong Y, Stewart KD, Florjancic AS, Harlan JE, Merta PJ, Przytulinska M, Soni N, Swinger KK, Zhu H, Johnson EF, Shoemaker AR, Penning TD. Azaindole-Based Inhibitors of Cdc7 Kinase: Impact of the Pre-DFG Residue, Val 195. ACS Med Chem Lett 2013; 4:211-5. [PMID: 24900653 DOI: 10.1021/ml300348c] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 01/15/2013] [Indexed: 12/24/2022] Open
Abstract
To investigate the role played by the unique pre-DFG residue Val 195 of Cdc7 kinase on the potency of azaindole-chloropyridines (1), a series of novel analogues with various chloro replacements were synthesized and evaluated for their inhibitory activity against Cdc7. X-ray cocrystallization using a surrogate protein, GSK3β, and modeling studies confirmed the azaindole motif as the hinge binder. Weaker hydrophobic interactions with Met 134 and Val 195 by certain chloro replacements (e.g., H, methyl) led to reduced Cdc7 inhibition. Meanwhile, data from other replacements (e.g., F, O) indicated that loss of such hydrophobic interaction could be compensated by enhanced hydrogen bonding to Lys 90. Our findings not only provide an in-depth understanding of the pre-DFG residue as another viable position impacting kinase inhibition, they also expand the existing knowledge of ligand-Cdc7 binding.
Collapse
Affiliation(s)
- Yunsong Tong
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Kent D. Stewart
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Alan S. Florjancic
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - John E. Harlan
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Philip J. Merta
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Magdalena Przytulinska
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Nirupama Soni
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Kerren K. Swinger
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Haizhong Zhu
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Eric F. Johnson
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Alexander R. Shoemaker
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| | - Thomas D. Penning
- Cancer
Research, ‡Structural Biology, §Lead Discovery, and ∥Protein Biochemistry, Global Pharmaceutical Research
and Development, Abbott Laboratories, 100
Abbott Park Road, Abbott Park, Illinois 60064, United States
| |
Collapse
|
14
|
Ogawa N, Yuki H, Tanaka A. Insights from Pim1 structure for anti-cancer drug design. Expert Opin Drug Discov 2012; 7:1177-92. [DOI: 10.1517/17460441.2012.727394] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
15
|
Parker LJ, Watanabe H, Tsuganezawa K, Tomabechi Y, Handa N, Shirouzu M, Yuki H, Honma T, Ogawa N, Nagano T, Yokoyama S, Tanaka A. Flexibility of the P-loop of Pim-1 kinase: observation of a novel conformation induced by interaction with an inhibitor. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:860-6. [PMID: 22869110 PMCID: PMC3412761 DOI: 10.1107/s1744309112027108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/15/2012] [Indexed: 12/27/2022]
Abstract
The serine/threonine kinase Pim-1 is emerging as a promising target for cancer therapeutics. Much attention has recently been focused on identifying potential Pim-1 inhibitor candidates for the treatment of haematopoietic malignancies. The outcome of a rational drug-design project has recently been reported [Nakano et al. (2012), J. Med. Chem. 55, 5151-5156]. The report described the process of optimization of the structure-activity relationship and detailed from a medicinal chemistry perspective the development of a low-potency and nonselective compound initially identified from in silico screening into a potent, selective and metabolically stable Pim-1 inhibitor. Here, the structures of the initial in silico hits are reported and the noteworthy features of the Pim-1 complex structures are described. A particular focus was placed on the rearrangement of the glycine-rich P-loop region that was observed for one of the initial compounds, (Z)-7-(azepan-1-ylmethyl)-2-[(1H-indol-3-yl)methylidene]-6-hydroxy-1-benzofuran-3(2H)-one (compound 1), and was also found in all further derivatives. This novel P-loop conformation, which appears to be stabilized by an additional interaction with the β3 strand located above the binding site, is not usually observed in Pim-1 structures.
Collapse
Affiliation(s)
- Lorien J. Parker
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Hisami Watanabe
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Keiko Tsuganezawa
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Yuri Tomabechi
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Noriko Handa
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Mikako Shirouzu
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Hitomi Yuki
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Teruki Honma
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Naoko Ogawa
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
| | - Tetsuo Nagano
- Open Innovation Center for Drug Discovery, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shigeyuki Yokoyama
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
- Laboratory of Structural Biology, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Akiko Tanaka
- RIKEN Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan
- Open Innovation Center for Drug Discovery, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
16
|
Lee J, Han SY, Jung H, Yang J, Choi JW, Chae CH, Park CH, Choi SU, Lee K, Ha JD, Lee CO, Ryu JW, Kim HR, Koh JS, Cho SY. Synthesis and structure–activity relationship of aminopyridines with substituted benzoxazoles as c-Met kinase inhibitors. Bioorg Med Chem Lett 2012; 22:4044-8. [DOI: 10.1016/j.bmcl.2012.04.083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 04/10/2012] [Accepted: 04/17/2012] [Indexed: 01/03/2023]
|
17
|
Discovery of XL413, a potent and selective CDC7 inhibitor. Bioorg Med Chem Lett 2012; 22:3727-31. [PMID: 22560567 DOI: 10.1016/j.bmcl.2012.04.024] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 03/29/2012] [Accepted: 04/03/2012] [Indexed: 11/22/2022]
Abstract
CDC7 is a serine/threonine kinase that has been shown to be required for the initiation and maintenance of DNA replication. Up-regulation of CDC7 is detected in multiple tumor cell lines, with inhibition of CDC7 resulting in cell cycle arrest. In this paper, we disclose the discovery of a potent and selective CDC7 inhibitor, XL413 (14), which was advanced into Phase 1 clinical trials. Starting from advanced lead 3, described in a preceding communication, we optimized the CDC7 potency and selectivity to demonstrate in vitro CDC7 dependent cell cycle arrest and in vivo tumor growth inhibition in a Colo-205 xenograft model.
Collapse
|
18
|
Woods KW, Lai C, Miyashiro JM, Tong Y, Florjancic AS, Han EK, Soni N, Shi Y, Lasko L, Leverson JD, Johnson EF, Shoemaker AR, Penning TD. Aminopyrimidinone Cdc7 Kinase Inhibitors. Bioorg Med Chem Lett 2012; 22:1940-3. [DOI: 10.1016/j.bmcl.2012.01.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 01/12/2012] [Accepted: 01/13/2012] [Indexed: 10/14/2022]
|
19
|
Lindvall M, McBride C, McKenna M, Gesner TG, Yabannavar A, Wong K, Lin S, Walter A, Shafer CM. 3D Pharmacophore Model-Assisted Discovery of Novel CDC7 Inhibitors. ACS Med Chem Lett 2011; 2:720-3. [PMID: 24900258 DOI: 10.1021/ml200029w] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 06/15/2011] [Indexed: 01/22/2023] Open
Abstract
A ligand-based 3D pharmacophore model for serine/threonine kinase CDC7 inhibition was created and successfully applied in the discovery of novel 2-(heteroaryl)-6,7-dihydrothieno[3,2-c]pyridin-4(5H)-ones. The pharmacophore model provided a hypothesis for lead generation missed by docking to a homology model. Medicinal chemistry exploration of the series revealed clear structure-activity relationships consistent with the pharmacophore model and pointed to further optimization opportunities.
Collapse
Affiliation(s)
- Mika Lindvall
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Christopher McBride
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Maureen McKenna
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Thomas G. Gesner
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Asha Yabannavar
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Kent Wong
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Song Lin
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Annette Walter
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| | - Cynthia M. Shafer
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, California 94608, United States
| |
Collapse
|
20
|
Dhuguru J, Gheewala C, Kumar NSS, Wilson JN. Highly Chromic, Proton-Responsive Phenyl Pyrimidones. Org Lett 2011; 13:4188-91. [DOI: 10.1021/ol2014945] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jyothi Dhuguru
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33124, United States
| | - Chirag Gheewala
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33124, United States
| | - N. S. Saleesh Kumar
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33124, United States
| | - James N. Wilson
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33124, United States
| |
Collapse
|
21
|
Montagnoli A, Moll J, Colotta F. Targeting cell division cycle 7 kinase: a new approach for cancer therapy. Clin Cancer Res 2010; 16:4503-8. [PMID: 20647475 DOI: 10.1158/1078-0432.ccr-10-0185] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cell division cycle 7 (Cdc7) is a serine-threonine kinase, originally discovered in budding yeast, required to initiate DNA replication. Human Cdc7 phosphorylates the minichromosome maintenance protein 2 (Mcm2), a component of the DNA replicative helicase needed for genome duplication. Inhibition of Cdc7 in cancer cells impairs progression through S phase, inducing a p53-independent apoptotic cell death, whereas in normal cells, it does not affect cell viability. Small molecule compounds able to interfere with Cdc7 activity have been identified and shown to be effective in controlling tumor growth in animal models. Two Cdc7 inhibitors are currently in phase I clinical development. Inhibition of Cdc7 kinase activity in cancer cells restricts DNA replication and induces apoptotic cell death by an unprecedented molecular mechanism of action.
Collapse
|
22
|
|
23
|
Swords R, Mahalingam D, O’Dwyer M, Santocanale C, Kelly K, Carew J, Giles F. Cdc7 kinase – A new target for drug development. Eur J Cancer 2010; 46:33-40. [DOI: 10.1016/j.ejca.2009.09.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 08/14/2009] [Accepted: 09/17/2009] [Indexed: 02/03/2023]
|
24
|
Ermoli A, Bargiotti A, Brasca MG, Ciavolella A, Colombo N, Fachin G, Isacchi A, Menichincheri M, Molinari A, Montagnoli A, Pillan A, Rainoldi S, Sirtori FR, Sola F, Thieffine S, Tibolla M, Valsasina B, Volpi D, Santocanale C, Vanotti E. Cell division cycle 7 kinase inhibitors: 1H-pyrrolo[2,3-b]pyridines, synthesis and structure-activity relationships. J Med Chem 2009; 52:4380-90. [PMID: 19555113 DOI: 10.1021/jm900248g] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cdc7 kinase has recently emerged as an attractive target for cancer therapy and low-molecular-weight inhibitors of Cdc7 kinase have been found to be effective in the inhibition of tumor growth in animal models. In this paper, we describe synthesis and structure-activity relationships of new 1H-pyrrolo[2,3-b]pyridine derivatives identified as inhibitors of Cdc7 kinase. Progress from (Z)-2-phenyl-5-(1H-pyrrolo[2,3-b]pyridin-3-ylmethylene)-3,5-dihydro-4H-imidazol-4-one (1) to [(Z)-2-(benzylamino)-5-(1H-pyrrolo[2,3-b]pyridin-3-ylmethylene)-1,3-thiazol-4(5H)-one] (42), a potent ATP mimetic inhibitor of Cdc7 kinase with IC(50) value of 7 nM, is also reported.
Collapse
|
25
|
Sawa M, Masai H. Drug design with Cdc7 kinase: a potential novel cancer therapy target. DRUG DESIGN DEVELOPMENT AND THERAPY 2009; 2:255-64. [PMID: 19920912 PMCID: PMC2761190 DOI: 10.2147/dddt.s4303] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Identification of novel molecular targets is critical in development of new and efficient cancer therapies. Kinases are one of the most common drug targets with a potential for cancer therapy. Cell cycle progression is regulated by a number of kinases, some of which are being developed to treat cancer. Cdc7 is a serine-threonine kinase originally discovered in budding yeast, which has been shown to be necessary to initiate the S phase. Inhibition of Cdc7 in cancer cells retards the progression of the S phase, accumulates DNA damage, and induces p53-independent cell death, but the same treatment in normal cells does not significantly affect of less than viability. Low-molecular-weight compounds that inhibit Cdc7 kinase with an IC50 10 nM have been identified, and shown to be effective in the inhibition of tumor growth in animal models. Thus Cdc7 kinase can be recognized as a novel molecular target for cancer therapy.
Collapse
|
26
|
Synthesis and evaluation of pyrido-thieno-pyrimidines as potent and selective Cdc7 kinase inhibitors. Bioorg Med Chem Lett 2009; 19:319-23. [DOI: 10.1016/j.bmcl.2008.11.093] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 11/20/2008] [Accepted: 11/24/2008] [Indexed: 11/23/2022]
|