Discovery of SY-5609: A Selective, Noncovalent Inhibitor of CDK7Click to copy article linkArticle link copied!
- Jason J. Marineau*Jason J. Marineau*Email: [email protected]. Phone: (617)-674-9075.Syros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Jason J. Marineau
- Kristin B. HammanKristin B. HammanSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Kristin B. Hamman
- Shanhu HuShanhu HuSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Shanhu Hu
- Sydney AlnemySydney AlnemySyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Sydney Alnemy
- Janessa MihalichJanessa MihalichSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Janessa Mihalich
- Anzhelika KabroAnzhelika KabroParaza Pharma Inc., 2525 Avenue Marie-Curie, Montreal, Quebec H4S 2E1, CanadaMore by Anzhelika Kabro
- Kenneth Matthew WhitmoreKenneth Matthew WhitmoreParaza Pharma Inc., 2525 Avenue Marie-Curie, Montreal, Quebec H4S 2E1, CanadaMore by Kenneth Matthew Whitmore
- Dana K. WinterDana K. WinterParaza Pharma Inc., 2525 Avenue Marie-Curie, Montreal, Quebec H4S 2E1, CanadaMore by Dana K. Winter
- Stephanie RoyStephanie RoyParaza Pharma Inc., 2525 Avenue Marie-Curie, Montreal, Quebec H4S 2E1, CanadaMore by Stephanie Roy
- Stephane CiblatStephane CiblatParaza Pharma Inc., 2525 Avenue Marie-Curie, Montreal, Quebec H4S 2E1, CanadaMore by Stephane Ciblat
- Nan KeNan KeSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Nan Ke
- Anneli SavinainenAnneli SavinainenSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Anneli Savinainen
- Ashraf WilsilyAshraf WilsilySyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Ashraf Wilsily
- Goran MalojcicGoran MalojcicSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Goran Malojcic
- Robert ZahlerRobert ZahlerSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Robert Zahler
- Darby SchmidtDarby SchmidtSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Darby Schmidt
- Michael J. BradleyMichael J. BradleySyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Michael J. Bradley
- Nigel J. WatersNigel J. WatersSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Nigel J. Waters
- Claudio ChuaquiClaudio ChuaquiSyros Pharmaceuticals Inc., 35 Cambridge Park Drive, Fourth Floor, Cambridge, Massachusetts 02140, United StatesMore by Claudio Chuaqui
Abstract
CDK7 has emerged as an exciting target in oncology due to its roles in two important processes that are misregulated in cancer cells: cell cycle and transcription. This report describes the discovery of SY-5609, a highly potent (sub-nM CDK7 Kd) and selective, orally available inhibitor of CDK7 that entered the clinic in 2020 (ClinicalTrials.gov Identifier: NCT04247126). Structure-based design was leveraged to obtain high selectivity (>4000-times the closest off target) and slow off-rate binding kinetics desirable for potent cellular activity. Finally, incorporation of a phosphine oxide as an atypical hydrogen bond acceptor helped provide the required potency and metabolic stability. The development candidate SY-5609 displays potent inhibition of CDK7 in cells and demonstrates strong efficacy in mouse xenograft models when dosed as low as 2 mg/kg.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
SPECIAL ISSUE
This article is part of the
Introduction
Results and Discussion
Initial Structure Activity Relationship (SAR) Exploration
IC50 values with CDK7/CycH/MAT1 are at or below the limit of detection in the enzymatic activity assay, so SPR Kd values were employed for SAR.
Development of ADME and PK SAR
Identification of SY-5102 and SY-5609
CDK7 SPR Kd (nM) | Enzymatic Activity IC50 (nM) | Anti-Proliferation EC50 (nM) | LogD | Mouse Microsomal Stability, Clint (mL/min/kg) | MDCK Papp A-B (10–6cm/s) | Mouse Unbound IV Cl (mL/min/kg) | Mouse po %F | |||
---|---|---|---|---|---|---|---|---|---|---|
CDK2 | CDK9 | CDK12 | HCC70 | |||||||
11 | 0.08 | 284 | 60 | 33 | 15 | 1.05 | <38 | 1.86 | 368 | 5 |
12 | 0.15 | 1638 | 907 | 215 | 287 | 0.49 | 89 | 1.12 | 394 | 0 |
SY-5102 | 0.03 | 189 | 90 | 75 | 9 | 2.6 | <38 | 4.62 | 712 | 36 |
13 | 0.06a | 771 | 572 | 156 | 1 | 2.7 | 164 | 3.32 | 597 | 28 |
SY-5609 | 0.07a | 5524 | 1919 | 1702 | 1 | 2 | <38 | 3.91 | 239 | 47 |
Long dissociation SPR.
Chemical Synthesis
Computational Modeling
CDK Family and Kinome Selectivity
Cellular Activity of SY-5609
In Vivo Efficacy and Pharmacodynamic Effects of SY-5609
Conclusions
Experimental Section
Chemistry
General Methods and Compound Characterization
Synthesis of 3-(2,5-Dichloropyrimidin-4-yl)-1H-indole (16)
Synthesis of 3-(2,5-Dichloropyrimidin-4-yl)-1-(phenylsulfonyl)-1H-indole (17)
Synthesis of (S)-tert-Butyl 3-((5-chloro-4-(1-(phenylsulfonyl)-1H-indol-3-yl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (19)
Synthesis of 5-Chloro-4-(1H-indol-3-yl)-N-[(3S)-3-piperidyl]pyrimidin-2-amine (1)
Synthesis of (S)-tert-Butyl 3-((4-(1-(phenylsulfonyl)-1H-indol-3-yl)-5-vinylpyrimidin-2-yl)amino)piperidine-1-carboxylate (21)
Synthesis of (S)-tert-Butyl 3-((5-ethyl-4-(1-(phenylsulfonyl)-1H-indol-3-yl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (22)
Synthesis of (S)-5-Ethyl-4-(1H-indol-3-yl)-N-(piperidin-3-yl)pyrimidin-2-amine hydrochloride (3)
Synthesis of 3-(2-(Methylthio)-5-(trifluoromethyl)pyrimidin-4-yl)-1-(phenylsulfonyl)-1H-indole (25)
Synthesis of 3-(2-(Methylsulfonyl)-5-(trifluoromethyl)pyrimidin-4-yl)-1-(phenylsulfonyl)-1H-indole (26)
Synthesis of (S)-1-Benzyl-6,6-dimethylpiperidin-3-amine 2,2,2-trifluoroacetate (27)
Synthesis of (S)-1-Benzyl-5,5-dimethylpiperidin-3-amine hydrochloride (28b)
Synthesis of (S)-Benzyl 5-amino-3,3-dimethylpiperidine-1-carboxylate hydrochloride (28a)
General Method for Synthesis of 29
Synthesis of (S)-4-(1H-Indol-3-yl)-N-(piperidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine hydrochloride (2)
Synthesis of (S)-N-(6,6-Dimethylpiperidin-3-yl)-4-(1H-indol-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine (6)
Synthesis of (S)-N-(5,5-Dimethylpiperidin-3-yl)-4-(1H-indol-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine (8)
Synthesis of 6-Bromo-3-(2-chloro-5-(trifluoromethyl)pyrimidin-4-yl)-1-(phenylsulfonyl)-1H-indole (31)
Synthesis of (S)-tert-Butyl 3-((4-(6-bromo-1-(phenylsulfonyl)-1H-indol-3-yl)-5-(trifluoromethyl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (32)
Synthesis of (S)-tert-Butyl 3-((4-(6-bromo-1H-indol-3-yl)-5-(trifluoromethyl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (33)
Synthesis of (S)-3-(2-(Piperidin-3-ylamino)-5-(trifluoromethyl)pyrimidin-4-yl)-1H-indole-6-carbonitrile (5)
Synthesis of (S)-4-(6-(Methylsulfonyl)-1H-indol-3-yl)-N-(piperidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine (7)
Synthesis of (S)-4-(6-(3,5-Dimethylisoxazol-4-yl)-1H-indol-3-yl)-N-(piperidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine (10)
Synthesis of tert-Butyl 7-bromo-1H-indole-1-carboxylate (35)
Synthesis of tert-Butyl 7-bromo-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (36)
Synthesis of (S)-tert-Butyl 3-((4-chloro-5-(trifluoromethyl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (37)
Synthesis of (S)-tert-Butyl 3-((4-(7-bromo-1H-indol-3-yl)-5-(trifluoromethyl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (38)
Synthesis of (S)-4-(7-(Methylsulfonyl)-1H-indol-3-yl)-N-(piperidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine (4)
Synthesis of (S)-Dimethyl(3-(2-(piperidin-3-ylamino)-5-(trifluoromethyl)pyrimidin-4-yl)-1H-indol-7-yl)phosphine oxide (9)
Synthesis of 3,5-Dimethyl-4-(1H-pyrrolo[2,3-b]pyridin-6-yl)isoxazole (40)
Synthesis of 4-(3-Bromo-1-(phenylsulfonyl)-1H-pyrrolo[2,3-b]pyridin-6-yl)-3,5-dimethylisoxazole (41)
Synthesis of 3,5-Dimethyl-4-(1-(phenylsulfonyl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridin-6-yl)isoxazole (42)
Synthesis of (S)-tert-Butyl 3-((4-(6-(3,5-dimethylisoxazol-4-yl)-1-(phenylsulfonyl)-1H-pyrrolo[2,3-b]pyridin-3-yl)-5-(trifluoromethyl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (43)
Synthesis of (S)-4-(6-(3,5-Dimethylisoxazol-4-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl)-N-(piperidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine hydrochloride (SY-5102)
Synthesis of (S)-6,6-Dimethylpiperidin-3-amine (45)
Synthesis of 7-Bromo-1H-indole-6-carboxylic acid (47)
Synthesis of 7-Bromo-1H-indole-6-carbonitrile (48)
Synthesis of 7-Bromo-3-(2-chloro-5-(trifluoromethyl)pyrimidin-4-yl)-1H-indole-6-carbonitrile (49)
Synthesis of (S)-3-(2-((1-Benzyl-5,5-dimethylpiperidin-3-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)-7-bromo-1H-indole-6-carbonitrile (50)
Synthesis of (S)-7-(Dimethylphosphoryl)-3-(2-((5,5-dimethylpiperidin-3-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)-1H-indole-6-carbonitrile (13)
Synthesis of (S)-7-(Dimethylphosphoryl)-3-(2-((6,6-dimethylpiperidin-3-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)-1H-indole-6-carbonitrile (SY-5609)
Synthesis of (2-Bromo-6-nitrophenyl)(methyl)sulfane (52)
Synthesis of 6-Bromo-7-(methylsulfonyl)-1H-indole (53)
Synthesis of 7-(Methylsulfonyl)-1H-indole-6-carbonitrile (54)
Synthesis of tert-Butyl 3-bromo-6-cyano-7-(methylsulfonyl)-1H-indole-1-carboxylate (55)
Synthesis of tert-Butyl 6-cyano-7-(methylsulfonyl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (56)
Synthesis of (S)-7-(Methylsulfonyl)-3-(2-(piperidin-3-ylamino)-5-(trifluoromethyl)pyrimidin-4-yl)-1H-indole-6-carbonitrile (11)
Synthesis of 6-Bromo-7-(methylthio)-1H-indole (57)
Synthesis of 6-Bromo-3-(2,5-dichloropyrimidin-4-yl)-7-(methylthio)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indole (58)
Synthesis of 3-(2,5-Dichloropyrimidin-4-yl)-7-(methylsulfonyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indole-6-carbonitrile (59)
Synthesis of (S)-tert-Butyl 3-((5-chloro-4-(6-cyano-7-(methylsulfonyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indol-3-yl)pyrimidin-2-yl)amino)piperidine-1-carboxylate (60)
Synthesis of (S)-tert-Butyl 3-((4-(6-cyano-7-(methylsulfonyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indol-3-yl)-5-ethylpyrimidin-2-yl)amino)piperidine-1-carboxylate (61)
Synthesis of (S)-3-(5-Ethyl-2-(piperidin-3-ylamino)pyrimidin-4-yl)-7-(methylsulfonyl)-1H-indole-6-carbonitrile (12)
Computational Chemistry
SPR
Kinase Enzymatic Activity Assay
Kinase Selectivity
Cell Culture
Antiproliferation Assay
Animal Study
ADME Profiling
Pharmacokinetics
Immunoblotting
Apoptosis Assay
Cell Cycle Analysis
PD Sample Analysis
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c01171.
Additional figures illustrating dihedral angles from molecular dynamics simulations, CDK selectivity of SY-5102 and SY-5609, kinase selectivity of SY-5102 and SY-5609, cellular antiproliferation panel for SY-5609, quantification of immunoblotting from Figure 6, immunoblot images from Figure 8, small molecule X-ray structure of SY-5609, protein cocrystal structure of Compound 4 with CDK2, NMR spectra and LC–MS chromatograms for all compounds (PDF)
Molecular formula strings (CSV)
PDB ID Codes: Compound 4 with CDK2 (PDB: 7RA5)
CCDC 2093192 contains the supplementary crystallographic data for this paper. These data can be obtained free of charge via www.ccdc.cam.ac.uk/data_request/cif, or by emailing [email protected], or by contacting The Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, UK; fax: +44 1223 336033.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank Wojciech Dworakowski, John Carulli, and Eric Olson for the helpful discussions and critical reading of the manuscript.
MeCN | acetonitrile |
CAK | cyclin activating kinase |
CDK | cyclin-dependent kinase |
CTD | C-terminal domain |
EtOAc | ethyl acetate |
MCL | myeloid cell leukemia |
MDCK | Madin-Darby canine kidney |
MeTHF | 2-methyl tetrahydrofuran |
OVA | ovarian cancer |
RBP | retinol binding protein |
SNAr | nucleophilic aromatic substitution |
SPR | surface plasmon resonance |
ns | nanoseconds |
TFIIH | transcription factor II H |
TNBC | triple negative breast cancer |
References
This article references 30 other publications.
- 1Hanahan, D.; Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 2011, 144, 646– 674, DOI: 10.1016/j.cell.2011.02.013Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjsFeqtrk%253D&md5=b36f160c417a712e83cb83c577f0018eHallmarks of cancer: the next generationHanahan, Douglas; Weinberg, Robert A.Cell (Cambridge, MA, United States) (2011), 144 (5), 646-674CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. The hallmarks of cancer comprise six biol. capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metab. and evading immune destruction. In addn. to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment.". Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer.
- 2Sava, G. P.; Fan, H.; Coombes, R. C.; Buluwela, L.; Ali, S. CDK7 inhibitors as anticancer drugs. Cancer Metastasis Rev. 2020, 39, 805– 823, DOI: 10.1007/s10555-020-09885-8Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXptVersL4%253D&md5=b61a237dcb6b1caa843baadf74df87ffCDK7 inhibitors as anticancer drugsSava, Georgina P.; Fan, Hailing; Coombes, R. Charles; Buluwela, Lakjaya; Ali, SimakCancer and Metastasis Reviews (2020), 39 (3), 805-823CODEN: CMRED4; ISSN:0167-7659. (Springer)A review. Cyclin-dependent kinase 7 (CDK7), along with cyclin H and MAT1, forms the CDK-activating complex (CAK), which directs progression through the cell cycle via T-loop phosphorylation of cell cycle CDKs. CAK is also a component of the general transcription factor, TFIIH. CDK7-mediated phosphorylation of RNA polymerase II (Pol II) at active gene promoters permits transcription. These findings identify CDK7 as a cancer therapeutic target, and several recent publications report selective CDK7 inhibitors (CDK7i) with activity against diverse cancer types. Preclin. studies have shown that CDK7i cause cell cycle arrest, apoptosis and repression of transcription, particularly of super-enhancer-assocd. genes in cancer, and have demonstrated their potential for overcoming resistance to cancer treatments. Moreover, combinations of CDK7i with other targeted cancer therapies, including BET inhibitors, BCL2 inhibitors and hormone therapies, have shown efficacy in model systems. Four CDK7i, ICEC0942 (CT7001), SY-1365, SY-5609 and LY3405105, have now progressed to Phase I/II clin. trials. Here we describe the work that has led to the development of selective CDK7i, the current status of the most advanced clin. candidates, and discuss their potential importance as cancer therapeutics, both as monotherapies and in combination settings. ClinicalTrials.gov Identifiers: NCT03363893; NCT03134638; NCT04247126; NCT03770494.
- 3Diab, S.; Yu, M.; Wang, S. CDK7 Inhibitors in cancer therapy: the sweet smell of success?. J. Med. Chem. 2020, 63, 7458– 7474, DOI: 10.1021/acs.jmedchem.9b01985Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksFaitLg%253D&md5=bdd5744330515e094612de7153d8cf36CDK7 Inhibitors in Cancer Therapy: The Sweet Smell of Success?Diab, Sarah; Yu, Mingfeng; Wang, ShudongJournal of Medicinal Chemistry (2020), 63 (14), 7458-7474CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Cyclin-dependent kinase (CDK) 7 has a unique functional repertoire by virtue of its dual role in transcription and cell cycle progression. Whereas CDK7 is ubiquitously expressed in various types of cancer, its downregulation leads to reduced cell proliferation. Importantly, it is now agreed that targeting transcription selectively limits the synthesis of mRNAs involved in tumor growth without causing an outage of transcription of housekeeping genes. Thus, CDK7 has been considered as a viable therapeutic target in cancer. Indeed, the development of CDK7 inhibitors has gained huge momentum with two mols., CT7001 and SY-1365, currently under clin. development. Herein, we discuss the latest understanding of the role of CDK7 in cancer cells and provide an overview of the pharmacophores of CDK7 inhibitors, their efficacy in various cancer models, and their clin. development.
- 4Sanchez-Martinez, C.; Lallena, M. J.; Sanfeliciano, S. G.; de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015–2019). Bioorg. Med. Chem. Lett. 2019, 29 (29), 126637, DOI: 10.1016/j.bmcl.2019.126637Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslWjs73N&md5=2808e04d5c09daad462133bcf9b9cb3dCyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015-2019)Sanchez-Martinez, Concepcion; Lallena, Maria Jose; Sanfeliciano, Sonia Gutierrez; de Dios, AlfonsoBioorganic & Medicinal Chemistry Letters (2019), 29 (20), 126637CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)A review. Sustained proliferative capacity and gene dysregulation are hallmarks of cancer. In mammalian cells, cyclin-dependent kinases (CDKs) control crit. cell cycle checkpoints and key transcriptional events in response to extracellular and intracellular signals leading to proliferation. Significant clin. activity for the treatment of hormone receptor pos. metastatic breast cancer has been demonstrated by palbociclib, ribociclib and abemaciclib, dual CDK4/6 inhibitors recently FDA-approved. SY-1365, a CDK7 inhibitor has shown initial encouraging data in phase I for solid tumors treatment. These results have rejuvenated the CDKs research field. This review provides an overview of relevant advances on CDK inhibitor research since 2015 to 2019, with special emphasis on transcriptional CDK inhibitors, new emerging strategies such as target protein degrdn. and compds. under clin. evaluation.
- 5Larochelle, S.; Amat, R.; Glover-Cutter, K.; Sanso, M.; Zhang, C.; Allen, J. J.; Shokat, K. M.; Bentley, D. L.; Fisher, R. P. Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II. Nat. Struct. Mol. Biol. 2012, 19, 1108– 1115, DOI: 10.1038/nsmb.2399Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsV2rsLvI&md5=e483e572df1f8645db9da40800dffe4dCyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase IILarochelle, Stephane; Amat, Ramon; Glover-Cutter, Kira; Sanso, Miriam; Zhang, Chao; Allen, Jasmina J.; Shokat, Kevan M.; Bentley, David L.; Fisher, Robert P.Nature Structural & Molecular Biology (2012), 19 (11), 1108-1115CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Promoter-proximal pausing by RNA polymerase II (Pol II) ensures gene-specific regulation and RNA quality control. Structural considerations suggested a requirement for initiation-factor eviction in elongation-factor engagement and pausing of transcription complexes. Here we show that selective inhibition of Cdk7-part of TFIIH-increases TFIIE retention, prevents DRB sensitivity-inducing factor (DSIF) recruitment and attenuates pausing in human cells. Pause release depends on Cdk9-cyclin T1 (P-TEFb); Cdk7 is also required for Cdk9-activating phosphorylation and Cdk9-dependent downstream events-Pol II C-terminal domain Ser2 phosphorylation and histone H2B ubiquitylation-in vivo. Cdk7 inhibition, moreover, impairs Pol II transcript 3'-end formation. Cdk7 thus acts through TFIIE and DSIF to establish, and through P-TEFb to relieve, barriers to elongation: incoherent feedforward that might create a window to recruit RNA-processing machinery. Therefore, cyclin-dependent kinases govern Pol II handoff from initiation to elongation factors and cotranscriptional RNA maturation.
- 6Rimel, J. K.; Poss, Z. C.; Erickson, B.; Maas, Z. L.; Ebmeier, C. C.; Johnson, J. L.; Decker, T. M.; Yaron, T. M.; Bradley, M. J.; Hamman, K. B.; Hu, S.; Malojcic, G.; Marineau, J. J.; White, P. W.; Brault, M.; Tao, L.; DeRoy, P.; Clavette, C.; Nayak, S.; Damon, L. J.; Kaltheuner, I. H.; Bunch, H.; Cantley, L. C.; Geyer, M.; Iwasa, J.; Dowell, R. D.; Bentley, D. L.; Old, W. M.; Taatjes, D. J. Selective inhibition of CDK7 reveals high-confidence targets and new models for TFIIH function in transcription. Genes Dev. 2020, 34, 1452– 1473, DOI: 10.1101/gad.341545.120Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisV2rt7zF&md5=5f6db2effae88caabdfc963fb2ab3e19Selective inhibition of CDK7 reveals high-confidence targets and new models for TFIIH function in transcriptionRimel, Jenna K.; Poss, Zachary C.; Erickson, Benjamin; Maas, Zachary L.; Ebmeier, Christopher C.; Johnson, Jared L.; Decker, Tim-Michael; Yaron, Tomer M.; Bradley, Michael J.; Hamman, Kristin B.; Hu, Shanhu; Malojcic, Goran; Marineau, Jason J.; White, Peter W.; Brault, Martine; Tao, Limei; DeRoy, Patrick; Clavette, Christian; Nayak, Shraddha; Damon, Leah J.; Kaltheuner, Ines H.; Bunch, Heeyoun; Cantley, Lewis C.; Geyer, Matthias; Iwasa, Janet; Dowell, Robin D.; Bentley, David L.; Old, William M.; Taatjes, Dylan J.Genes & Development (2020), 34 (21-22), 1452-1473CODEN: GEDEEP; ISSN:0890-9369. (Cold Spring Harbor Laboratory Press)CDK7 assocs. with the 10-subunit TFIIH complex and regulates transcription by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (RNAPII). Few addnl. CDK7 substrates are known. Here, using the covalent inhibitor SY-351 and quant. phosphoproteomics, we identified CDK7 kinase substrates in human cells. Among hundreds of high-confidence targets, the vast majority are unique to CDK7 (i.e., distinct from other transcription-assocd. kinases), with a subset that suggest novel cellular functions. Transcription-assocd. factors were predominant CDK7 substrates, including SF3B1, U2AF2, and other splicing components. Accordingly, widespread and diverse splicing defects, such as alternative exon inclusion and intron retention, were characterized in CDK7-inhibited cells. Combined with biochem. assays, we establish that CDK7 directly activates other transcription-assocd. kinases CDK9, CDK12, and CDK13, invoking a "master regulator" role in transcription. We further demonstrate that TFIIH restricts CDK7 kinase function to the RNAPII CTD, whereas other substrates (e.g., SPT5 and SF3B1) are phosphorylated by the three-subunit CDK-activating kinase (CAK; CCNH, MAT1, and CDK7). These results suggest new models for CDK7 function in transcription and implicate CAK dissocn. from TFIIH as essential for kinase activation. This straightforward regulatory strategy ensures CDK7 activation is spatially and temporally linked to transcription, and may apply toward other transcription-assocd. kinases.
- 7Fisher, R. P. Cdk7: a kinase at the core of transcription and in the crosshairs of cancer drug discovery. Transcription 2019, 10, 47– 56, DOI: 10.1080/21541264.2018.1553483Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVKit7jJ&md5=b6344f44b27d777d4fb7aad3a609d087Cdk7: a kinase at the core of transcription and in the crosshairs of cancer drug discoveryFisher, Robert P.Transcription (2019), 10 (2), 47-56CODEN: TRANH2; ISSN:2154-1272. (Taylor & Francis, Inc.)A review. The transcription cycle of RNA polymerase II (Pol II) is regulated by a set of cyclin-dependent kinases (CDKs). Cdk7, assocd. with the transcription initiation factor TFIIH, is both an effector CDK that phosphorylates Pol II and other targets within the transcriptional machinery, and a CDK-activating kinase (CAK) for at least one other essential CDK involved in transcription. Recent studies have illuminated Cdk7 functions that are executed throughout the Pol II transcription cycle, from promoter clearance and promoter-proximal pausing, to co-transcriptional chromatin modification in gene bodies, to mRNA 3'-end formation and termination. Cdk7 has also emerged as a target of small-mol. inhibitors that show promise in the treatment of cancer and inflammation. The challenges now are to identify the relevant targets of Cdk7 at each step of the transcription cycle, and to understand how heightened dependence on an essential CDK emerges in cancer, and might be exploited therapeutically.
- 8Hu, S.; Marineau, J. J.; Rajagopal, N.; Hamman, K. B.; Choi, Y. J.; Schmidt, D. R.; Ke, N.; Johannessen, L.; Bradley, M. J.; Orlando, D. A.; Alnemy, S. R.; Ren, Y.; Ciblat, S.; Winter, D. K.; Kabro, A.; Sprott, K. T.; Hodgson, J. G.; Fritz, C. C.; Carulli, J. P.; di Tomaso, E.; Olson, E. R. Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res. 2019, 79, 3479– 3491, DOI: 10.1158/0008-5472.CAN-19-0119Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M7jtl2iug%253D%253D&md5=4fd24fb5eea04eb6e921f52206f69098Discovery and Characterization of SY-1365, a Selective, Covalent Inhibitor of CDK7Hu Shanhu; Marineau Jason J; Rajagopal Nisha; Hamman Kristin B; Choi Yoon Jong; Schmidt Darby R; Ke Nan; Johannessen Liv; Bradley Michael J; Orlando David A; Alnemy Sydney R; Ren Yixuan; Sprott Kevin T; Hodgson J Graeme; Fritz Christian C; Carulli John P; di Tomaso Emmanuelle; Olson Eric R; Ciblat Stephane; Winter Dana K; Kabro AnzhelikaCancer research (2019), 79 (13), 3479-3491 ISSN:.Recent studies suggest that targeting transcriptional machinery can lead to potent and selective anticancer effects in cancers dependent on high and constant expression of certain transcription factors for growth and survival. Cyclin-dependent kinase 7 (CDK7) is the catalytic subunit of the CDK-activating kinase complex. Its function is required for both cell-cycle regulation and transcriptional control of gene expression. CDK7 has recently emerged as an attractive cancer target because its inhibition leads to decreased transcript levels of oncogenic transcription factors, especially those associated with super-enhancers. Here, we describe a selective CDK7 inhibitor SY-1365, which is currently in clinical trials in populations of patients with ovarian and breast cancer (NCT03134638). In vitro, SY-1365 inhibited cell growth of many different cancer types at nanomolar concentrations. SY-1365 treatment decreased MCL1 protein levels, and cancer cells with low BCL2L1 (BCL-XL) expression were found to be more sensitive to SY-1365. Transcriptional changes in acute myeloid leukemia (AML) cell lines were distinct from those following treatment with other transcriptional inhibitors. SY-1365 demonstrated substantial antitumor effects in multiple AML xenograft models as a single agent; SY-1365-induced growth inhibition was enhanced in combination with the BCL2 inhibitor venetoclax. Antitumor activity was also observed in xenograft models of ovarian cancer, suggesting the potential for exploring SY-1365 in the clinic in both hematologic and solid tumors. Our findings support targeting CDK7 as a new approach for treating transcriptionally addicted cancers. SIGNIFICANCE: These findings demonstrate the molecular mechanism of action and potent antitumor activity of SY-1365, the first selective CDK7 inhibitor to enter clinical investigation.
- 9Hazel, P.; Kroll, S. H.; Bondke, A.; Barbazanges, M.; Patel, H.; Fuchter, M. J.; Coombes, R. C.; Ali, S.; Barrett, A. G.; Freemont, P. S. Inhibitor selectivity for cyclin-dependent kinase 7: A structural, thermodynamic, and modelling study. ChemMedChem 2017, 12, 372– 380, DOI: 10.1002/cmdc.201600535Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitFegsb8%253D&md5=7fb792661b58d7e3d0859c2630dccdaeInhibitor selectivity for cyclin-dependent kinase 7: A structural, thermodynamic, and modeling studyHazel, Pascale; Kroll, Sebastian H. B.; Bondke, Alexander; Barbazanges, Marion; Patel, Hetal; Fuchter, Matthew J.; Coombes, R. Charles; Ali, Simak; Barrett, Anthony G. M.; Freemont, Paul S.ChemMedChem (2017), 12 (5), 372-380CODEN: CHEMGX; ISSN:1860-7179. (Wiley-VCH Verlag GmbH & Co. KGaA)Deregulation of the cell cycle by mechanisms that lead to elevated activities of cyclin-dependent kinases (CDKs) is a feature of many human diseases, cancer in particular. Here, we identified small-mol.-wt. inhibitors that selectively inhibit CDK7, the kinase that phosphorylates cell-cycle CDKs to promote their activities. To investigate the selectivity of these inhibitors, we used a combination of structural, biophys., and modeling approaches. We detd. the crystal structures of CDK7-selective compds. ICEC 0942 and ICEC 0943 bound to CDK2, and used these to build models of inhibitor binding to CDK7. Mol. dynamics (MD) simulations of inhibitors bound to CDK2 and CDK7 generated possible models of inhibitor binding. To exptl. validate these models, we gathered isothermal titrn. calorimetry (ITC) binding data for recombinant wild-type and binding site mutants of CDK7 and CDK2. We identified specific residues of CDK7, notably Asp-155, that were involved in detg. inhibitor selectivity. The MD simulations also showed that the flexibility of the G-rich and activation loops of CDK7 is likely an important determinant of inhibitor specificity similar to CDK2.
- 10Study of XL102 as single-agent and combination therapy in subjects with solid tumors. ClinicalTrials.gov; U.S. National Library of Medicine, 2021. https://ClinicalTrials.gov/show/NCT04726332.Google ScholarThere is no corresponding record for this reference.
- 11Lolli, G.; Lowe, E. D.; Brown, N. R.; Johnson, L. N. The crystal structure of human CDK7 and its protein recognition properties. Structure 2004, 12, 2067– 2079, DOI: 10.1016/j.str.2004.08.013Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpsVOrs7Y%253D&md5=b090d554993e8968c64d28fabaf6a507The Crystal Structure of Human CDK7 and Its Protein Recognition PropertiesLolli, Graziano; Lowe, Edward D.; Brown, Nick R.; Johnson, Louise N.Structure (Cambridge, MA, United States) (2004), 12 (11), 2067-2079CODEN: STRUE6; ISSN:0969-2126. (Cell Press)CDK7, a member of the cyclin-dependent protein kinase family, regulates the activities of other CDKs through phosphorylation on their activation segment and hence contributes to control of the eukaryotic cell cycle. CDK7 also assists in the regulation of transcription as part of the transcription factor TFIIH complex. For max. activity and stability, CDK7 requires phosphorylation, assocn. with cyclin H, and assocn. with a third protein, MAT1. We have detd. the crystal structure of human CDK7 in complex with ATP at 3Å resoln. The kinase is in the inactive conformation, similar to that obsd. for inactive CDK2. The activation segment is phosphorylated at Thr170 and is in a defined conformation that differs from that in phospho-CDK2 and phospho-CDK2/cyclin A. The functional properties of the enzyme against CDK2 and CTD as substrates are characterized through kinase assays. Expts. confirm that CDK7 is not a substrate for kinase-assocd. phosphatase.
- 12Finkbeiner, P.; Hehn, J. P.; Gnamm, C. Phosphine oxides from a medicinal chemist’s perspective: physicochemical and in vitro parameters relevant for drug discovery. J. Med. Chem. 2020, 63, 7081– 7107, DOI: 10.1021/acs.jmedchem.0c00407Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVKnt7rP&md5=da34d045059662fb8fb73a17ee1dfd17Phosphine Oxides from a Medicinal Chemist's Perspective: Physicochemical and in Vitro Parameters Relevant for Drug DiscoveryFinkbeiner, Peter; Hehn, Joerg P.; Gnamm, ChristianJournal of Medicinal Chemistry (2020), 63 (13), 7081-7107CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Phosphine oxides and related phosphorus-contg. functional groups such as phosphonates and phosphinates are established structural motifs that are still underrepresented in today's drug discovery projects, and only few examples can be found among approved drugs. In this account, the physicochem. and in vitro properties of phosphine oxides and related phosphorus-contg. functional groups are reported and compared to more commonly used structural motifs in drug discovery. Furthermore, the impact on the physicochem. properties of a real drug scaffold is exemplified by a series of phosphorus-contg. analogs of imatinib. We demonstrate that phosphine oxides are highly polar functional groups leading to high soly. and metabolic stability but occasionally at the cost of reduced permeability. We conclude that phosphine oxides and related phosphorus-contg. functional groups are valuable polar structural elements and that they deserve to be considered as a routine part of every medicinal chemist's toolbox.
- 13Huang, W. S.; Liu, S.; Zou, D.; Thomas, M.; Wang, Y.; Zhou, T.; Romero, J.; Kohlmann, A.; Li, F.; Qi, J.; Cai, L.; Dwight, T. A.; Xu, Y.; Xu, R.; Dodd, R.; Toms, A.; Parillon, L.; Lu, X.; Anjum, R.; Zhang, S.; Wang, F.; Keats, J.; Wardwell, S. D.; Ning, Y.; Xu, Q.; Moran, L. E.; Mohemmad, Q. K.; Jang, H. G.; Clackson, T.; Narasimhan, N. I.; Rivera, V. M.; Zhu, X.; Dalgarno, D.; Shakespeare, W. C. Discovery of brigatinib (AP26113), a phosphine oxide-containing, potent, orally active inhibitor of anaplastic lymphoma kinase. J. Med. Chem. 2016, 59, 4948– 4964, DOI: 10.1021/acs.jmedchem.6b00306Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntlCgur8%253D&md5=4cb93ff587579c97fda8aff0c7d43317Discovery of Brigatinib (AP26113), a Phosphine Oxide-Containing, Potent, Orally Active Inhibitor of Anaplastic Lymphoma KinaseHuang, Wei-Sheng; Liu, Shuangying; Zou, Dong; Thomas, Mathew; Wang, Yihan; Zhou, Tianjun; Romero, Jan; Kohlmann, Anna; Li, Feng; Qi, Jiwei; Cai, Lisi; Dwight, Timothy A.; Xu, Yongjin; Xu, Rongsong; Dodd, Rory; Toms, Angela; Parillon, Lois; Lu, Xiaohui; Anjum, Rana; Zhang, Sen; Wang, Frank; Keats, Jeffrey; Wardwell, Scott D.; Ning, Yaoyu; Xu, Qihong; Moran, Lauren E.; Mohemmad, Qurish K.; Jang, Hyun Gyung; Clackson, Tim; Narasimhan, Narayana I.; Rivera, Victor M.; Zhu, Xiaotian; Dalgarno, David; Shakespeare, William C.Journal of Medicinal Chemistry (2016), 59 (10), 4948-4964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)In the treatment of echinoderm microtubule-assocd. protein-like 4 (EML4)-anaplastic lymphoma kinase pos. (ALK+) non-small-cell lung cancer (NSCLC), secondary mutations within the ALK kinase domain have emerged as a major resistance mechanism to both first- and second-generation ALK inhibitors. This report describes the design and synthesis of a series of 2,4-diarylaminopyrimidine-based potent and selective ALK inhibitors culminating in identification of the investigational clin. candidate brigatinib. A unique structural feature of brigatinib is a phosphine oxide, an overlooked but novel hydrogen-bond acceptor that drives potency and selectivity in addn. to favorable ADME properties. Brigatinib displayed low nanomolar IC50s against native ALK and all tested clin. relevant ALK mutants in both enzyme-based biochem. and cell-based viability assays and demonstrated efficacy in multiple ALK+ xenografts in mice, including Karpas-299 (anaplastic large-cell lymphomas [ALCL]) and H3122 (NSCLC). Brigatinib represents the most clin. advanced phosphine oxide-contg. drug candidate to date and is currently being evaluated in a global phase 2 registration trial.
- 14Bradley, M.; Ciblat, S.; Kabro, A.; Marineau, J. J.; Chuaqui, C. Inhibitors of cyclin-dependent kinase 7 (CDK7). Patent WO2020093011A1, 2020.Google ScholarThere is no corresponding record for this reference.
- 15Marineau, J. J.; Chuaqui, C.; Ciblat, S.; Kabro, A.; Piras, H.; Whitmore, K. M.; Lund, K.-L. Preparation of substituted aminopyrimidines as inhibitors of cyclin-dependent kinase 7 (CDK7). Patent WO2019143719A1, 2019.Google ScholarThere is no corresponding record for this reference.
- 16Marineau, J. J.; Zahler, R.; Ciblat, S.; Winter, D. K.; Kabro, A.; Roy, S.; Schmidt, D.; Chuaqui, C.; Malojcic, G.; Piras, H.; Whitmore, K. M.; Lund, K.-I.; Sinko, B.; Sprott, K. Preparation of inhibitors of cyclin dependent kinase 7 (CDK7). Patent WO2018013867A1, 2018.Google ScholarThere is no corresponding record for this reference.
- 17Panday, S. K.; Langlois, N. Enantioselective synthesis of (S)-5-aminopiperidin-2-one from (S)-pyroglutaminol. Tetrahedron Lett. 1995, 36, 8205– 8208, DOI: 10.1016/00404-0399(50)17557-Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXptFeitb0%253D&md5=000b9e9296f97404f8f6745698c351e4Enantioselective synthesis of (S)-5-aminopiperidin-2-one from (S)-pyroglutaminolPanday, Sharad Kumar; Langlois, NicoleTetrahedron Letters (1995), 36 (45), 8205-8CODEN: TELEAY; ISSN:0040-4039. (Elsevier)(5S)-5-aminopiperidin-2-one and several derivs. were synthesized from (S)-pyroglutaminol through ring opening and Mitsunobu reaction as the key steps.
- 18Greber, B. J.; Perez-Bertoldi, J. M.; Lim, K.; Iavarone, A. T.; Toso, D. B.; Nogales, E. The cryoelectron microscopy structure of the human CDK-activating kinase. Proc. Natl. Acad. Sci. U. S. A. 2020, 117, 22849– 22857, DOI: 10.1073/pnas.2009627117Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVCjurbJ&md5=ef7d1df2ddb5e988fe89507a4f3cd902The cryoelectron microscopy structure of the human CDK-activating kinaseGreber, Basil J.; Perez-Bertoldi, Juan M.; Lim, Kif; Iavarone, Anthony T.; Toso, Daniel B.; Nogales, EvaProceedings of the National Academy of Sciences of the United States of America (2020), 117 (37), 22849-22857CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The human CDK-activating kinase (CAK), a complex composed of cyclin-dependent kinase (CDK) 7, cyclin H, and MAT1, is a crit. regulator of transcription initiation and the cell cycle. It acts by phosphorylating the C-terminal heptapeptide repeat domain of the RNA polymerase II (Pol II) subunit RPB1, which is an important regulatory event in transcription initiation by Pol II, and it phosphorylates the regulatory T-loop of CDKs that control cell cycle progression. Here, the authors detd. the three-dimensional (3D) structure of the catalytic module of human CAK, revealing the structural basis of its assembly and providing insight into CDK7 activation in this context. The unique third component of the complex, MAT1, substantially extends the interaction interface between CDK7 and cyclin H, explaining its role as a CAK assembly factor, and it forms interactions with the CDK7 T-loop, which may contribute to enhancing CAK activity. The authors also detd. the structure of the CAK in complex with the covalently bound inhibitor THZ1 to provide insight into the binding of inhibitors at the CDK7 active site and to aid in the rational design of therapeutic compds.
- 19Greber, B. J.; Remis, J.; Ali, S.; Nogales, E. 2.5 A-resolution structure of human CDK-activating kinase bound to the clinical inhibitor ICEC0942. Biophys. J. 2021, 120, 677– 686, DOI: 10.1016/j.bpj.2020.12.030Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjtVSrs7s%253D&md5=16cffe07b3fe538abaa36eb66202406eResolution structure 2.5 Å-of human CDK-activating kinase bound to the clinical inhibitor ICEC0942Greber, Basil J.; Remis, Jonathan; Ali, Simak; Nogales, EvaBiophysical Journal (2021), 120 (4), 677-686CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)The human CDK-activating kinase (CAK), composed of CDK7, cyclin H, and MAT1, is involved in the control of transcription initiation and the cell cycle. Because of these activities, it has been identified as a promising target for cancer chemotherapy. A no. of CDK7 inhibitors have entered clin. trials, among them ICEC0942 (also known as CT7001). Structural information can aid in improving the affinity and specificity of such drugs or drug candidates, reducing side effects in patients. Here, we have detd. the structure of the human CAK in complex with ICEC0942 at 2.5 Å-resoln. using cryogenic electron microscopy. Our structure reveals conformational differences of ICEC0942 compared with previous X-ray crystal structures of the CDK2-bound complex, and highlights the crit. ability of cryogenic electron microscopy to resolve structures of drug-bound protein complexes without the need to crystalize the protein target.
- 20Kwiatkowski, N.; Zhang, T.; Rahl, P. B.; Abraham, B. J.; Reddy, J.; Ficarro, S. B.; Dastur, A.; Amzallag, A.; Ramaswamy, S.; Tesar, B.; Jenkins, C. E.; Hannett, N. M.; McMillin, D.; Sanda, T.; Sim, T.; Kim, N. D.; Look, T.; Mitsiades, C. S.; Weng, A. P.; Brown, J. R.; Benes, C. H.; Marto, J. A.; Young, R. A.; Gray, N. S. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014, 511, 616– 620, DOI: 10.1038/nature13393Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1ChurnF&md5=895a28fb536663a31e7558fe79ce628bTargeting transcription regulation in cancer with a covalent CDK7 inhibitorKwiatkowski, Nicholas; Zhang, Tinghu; Rahl, Peter B.; Abraham, Brian J.; Reddy, Jessica; Ficarro, Scott B.; Dastur, Anahita; Amzallag, Arnaud; Ramaswamy, Sridhar; Tesar, Bethany; Jenkins, Catherine E.; Hannett, Nancy M.; McMillin, Douglas; Sanda, Takaomi; Sim, Taebo; Kim, Nam Doo; Look, Thomas; Mitsiades, Constantine S.; Weng, Andrew P.; Brown, Jennifer R.; Benes, Cyril H.; Marto, Jarrod A.; Young, Richard A.; Gray, Nathanael S.Nature (London, United Kingdom) (2014), 511 (7511), 616-620CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Tumor oncogenes include transcription factors that co-opt the general transcriptional machinery to sustain the oncogenic state, but direct pharmacol. inhibition of transcription factors has so far proven difficult. However, the transcriptional machinery contains various enzymic cofactors that can be targeted for the development of new therapeutic candidates, including cyclin-dependent kinases (CDKs). Here the authors present the discovery and characterization of a covalent CDK7 inhibitor, THZ1, which has the unprecedented ability to target a remote cysteine residue located outside of the canonical kinase domain, providing an unanticipated means of achieving selectivity for CDK7. Cancer cell-line profiling indicates that a subset of cancer cell lines, including human T-cell acute lymphoblastic leukemia (T-ALL), have exceptional sensitivity to THZ1. Genome-wide anal. in Jurkat T-ALL cells shows that THZ1 disproportionally affects transcription of RUNX1 and suggests that sensitivity to THZ1 may be due to vulnerability conferred by the RUNX1 super-enhancer and the key role of RUNX1 in the core transcriptional regulatory circuitry of these tumor cells. Pharmacol. modulation of CDK7 kinase activity may thus provide an approach to identify and treat tumor types that are dependent on transcription for maintenance of the oncogenic state.
- 21Schrödinger Release 2020–2; Schrödinger, L.: New York, NY, 2020.Google ScholarThere is no corresponding record for this reference.
- 22Kyte, J.; Doolittle, R. F. A simple method for displaying the hydropathic character of a protein. J. Mol. Biol. 1982, 157, 105– 132, DOI: 10.1016/0022-2836(82)90515-0Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL38Xks1yjtro%253D&md5=ee67eb115939dfe56b2b2cae2c32dbd3A simple method for displaying the hydropathic character of a proteinKyte, Jack; Doolittle, Russell F.Journal of Molecular Biology (1982), 157 (1), 105-32CODEN: JMOBAK; ISSN:0022-2836.A computer program that progressively evaluates the hydrophilicity and hydrophobicity of a protein along its amino acid sequence was devised. A hydropathy scale takes into consideration the hydrophilic and hydrophobic properties of each of the 20 amino acid side chains. Correlation was demonstrated between the plotted values and known structures detd. by crystallog.
- 23Zamyatnin, A. A. Protein volume in solution. Prog. Biophys. Mol. Biol. 1972, 24, 107– 123, DOI: 10.1016/0079-6107(72)90005-3Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE3s%252Fnt1Gluw%253D%253D&md5=1eb0435b9ebb74a00ff9517a292cd5c5Protein volume in solutionZamyatnin A AProgress in biophysics and molecular biology (1972), 24 (), 107-23 ISSN:0079-6107.There is no expanded citation for this reference.
- 24Eid, S.; Turk, S.; Volkamer, A.; Rippmann, F.; Fulle, S. KinMap: a web-based tool for interactive navigation through human kinome data. BMC Bioinf. 2017, 18, 16– 21, DOI: 10.1186/s12859-016-1433-7Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjsFCjtLc%253D&md5=29737c7cda8d9caf75018e0d29e9a459KinMap: a web-based tool for interactive navigation through human kinome dataEid, Sameh; Turk, Samo; Volkamer, Andrea; Rippmann, Friedrich; Fulle, SimoneBMC Bioinformatics (2017), 18 (), 16/1-16/6CODEN: BBMIC4; ISSN:1471-2105. (BioMed Central Ltd.)Background: Annotations of the phylogenetic tree of the human kinome is an intuitive way to visualize compd. profiling data, structural features of kinases or functional relationships within this important class of proteins. The increasing vol. and complexity of kinase-related data underlines the need for a tool that enables complex queries pertaining to kinase disease involvement and potential therapeutic uses of kinase inhibitors. Results: Here, we present KinMap, a user-friendly online tool that facilitates the interactive navigation through kinase knowledge by linking biochem., structural, and disease assocn. data to the human kinome tree. To this end, preprocessed data from freely-available sources, such as ChEMBL, the Protein Data Bank, and the Center for Therapeutic Target Validation platform are integrated into KinMap and can easily be complemented by proprietary data. The value of KinMap will be exemplarily demonstrated for uncovering new therapeutic indications of known kinase inhibitors and for prioritizing kinases for drug development efforts. Conclusions: KinMap represents a new generation of kinome tree viewers which facilitates interactive exploration of the human kinome. KinMap enables generation of high-quality annotated images of the human kinome tree as well as exchange of kinome-related data in scientific communications. Furthermore, KinMap supports multiple input and output formats and recognizes alternative kinase names and links them to a unified naming scheme, which makes it a useful tool across different disciplines and applications. A web-service of KinMap is freely available at http://www.kinhub.org/kinmap/.
- 25Wang, Y.; Zhang, T.; Kwiatkowski, N.; Abraham, B. J.; Lee, T. I.; Xie, S.; Yuzugullu, H.; Von, T.; Li, H.; Lin, Z.; Stover, D. G.; Lim, E.; Wang, Z. C.; Iglehart, J. D.; Young, R. A.; Gray, N. S.; Zhao, J. J. CDK7-dependent transcriptional addiction in triple-negative breast cancer. Cell 2015, 163, 174– 86, DOI: 10.1016/j.cell.2015.08.063Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFKqtrjP&md5=34441125e931816d29e419f3f7dc0346CDK7-dependent transcriptional addiction in triple-negative breast cancerWang, Yubao; Zhang, Tinghu; Kwiatkowski, Nicholas; Abraham, Brian J.; Lee, Tong Ihn; Xie, Shaozhen; Yuzugullu, Haluk; Von, Thanh; Li, Heyuan; Lin, Ziao; Stover, Daniel G.; Lim, Elgene; Wang, Zhigang C.; Iglehart, J. Dirk; Young, Richard A.; Gray, Nathanael S.; Zhao, Jean J.Cell (Cambridge, MA, United States) (2015), 163 (1), 174-186CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Triple-neg. breast cancer (TNBC) is a highly aggressive form of breast cancer that exhibits extremely high levels of genetic complexity and yet a relatively uniform transcriptional program. We postulate that TNBC might be highly dependent on uninterrupted transcription of a key set of genes within this gene expression program and might therefore be exceptionally sensitive to inhibitors of transcription. Utilizing kinase inhibitors and CRISPR/Cas9-mediated gene editing, we show here that triple-neg. but not hormone receptor-pos. breast cancer cells are exceptionally dependent on CDK7, a transcriptional cyclin-dependent kinase. TNBC cells are unique in their dependence on this transcriptional CDK and suffer apoptotic cell death upon CDK7 inhibition. An "Achilles cluster" of TNBC-specific genes is esp. sensitive to CDK7 inhibition and frequently assocd. with super-enhancers. We conclude that CDK7 mediates transcriptional addiction to a vital cluster of genes in TNBC and CDK7 inhibition may be a useful therapy for this challenging cancer.
- 26Zhang, Z.; Peng, H.; Wang, X.; Yin, X.; Ma, P.; Jing, Y.; Cai, M. C.; Liu, J.; Zhang, M.; Zhang, S.; Shi, K.; Gao, W. Q.; Di, W.; Zhuang, G. Preclinical efficacy and molecular mechanism of targeting CDK7-dependent transcriptional addiction in ovarian cancer. Mol. Cancer Ther. 2017, 16, 1739– 1750, DOI: 10.1158/1535-7163.MCT-17-0078Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVCitrzL&md5=49a9f8b77dd02150dabd4fdcfe16622bPreclinical Efficacy and Molecular Mechanism of Targeting CDK7-Dependent Transcriptional Addiction in Ovarian CancerZhang, Zhenfeng; Peng, Huixin; Wang, Xiaojie; Yin, Xia; Ma, Pengfei; Jing, Ying; Cai, Mei-Chun; Liu, Jin; Zhang, Meiying; Zhang, Shengzhe; Shi, Kaixuan; Gao, Wei-Qiang; Di, Wen; Zhuang, GuangleiMolecular Cancer Therapeutics (2017), 16 (9), 1739-1750CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Ovarian cancer remains a significant cause of gynecol. cancer mortality, and novel therapeutic strategies are urgently needed in clinic as new treatment options. We previously showed that BET bromodomain inhibitors displayed promising efficacy for the treatment of epithelial ovarian cancer by downregulating pivot transcription factors. However, the potential antitumor activities and mol. mechanisms of other epigenetic or transcriptional therapies have not been systematically detd. Here, by performing an unbiased high-throughput drug screen to identify candidate compds. with antineoplastic effects, we identified THZ1, a recently developed covalent CDK7 inhibitor, as a new transcription-targeting compd. that exerted broad cytotoxicity against ovarian tumors. Mechanistically, CDK7 represented a previously unappreciated actionable vulnerability in ovarian cancer, and CDK7 inhibition led to a pronounced dysregulation of gene transcription, with a preferential repression of E2F-regulated genes and transcripts assocd. with super-enhancers. Our findings revealed the mol. underpinnings of THZ1 potency and established pharmaceutically targeting transcriptional addiction as a promising therapeutic strategy in aggressive ovarian cancer. Mol Cancer Ther; 16(9); 1739-50. ©2017 AACR.
- 27Holmes, I. P.; Bergman, Y.; Lunniss, G. E.; Nikac, M.; Choi, N.; Hemley, C. F.; Walker, S. R.; Foitzik, R. C.; Ganame, D.; Lessene, R. Preparation of pyrimidine derivatives useful as FAK inhibitors. U.S. Patent US20130017194A1, 2013.Google ScholarThere is no corresponding record for this reference.
- 28Albers, R.; Ayala, L.; Clareen, S. S.; Delgado Mederos, M. M.; Hilgraf, R.; Hedge, S.; Hughes, K.; Kois, A.; Plantevin-Krenitsky, V.; McCarrick, M.; Nadolny, L.; Palanki, M.; Sahasrabudhe, K.; Sapienza, J.; Satoh, Y.; Sloss, M.; Sudbeck, E.; Wright, J. Preparation of haloaryl substituted aminopurines for use as a prodrug in the treatment of cancers, cardiovascular or renal diseases. Patent WO2006076595A1, 2006.Google ScholarThere is no corresponding record for this reference.
- 29Lovitt, B.; Vanderporten, E. C.; Sheng, Z.; Zhu, H.; Drummond, J.; Liu, Y. Differential effects of divalent manganese and magnesium on the kinase activity of leucine-rich repeat kinase 2 (LRRK2). Biochemistry 2010, 49, 3092– 3100, DOI: 10.1021/bi901726cGoogle Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1Ohsrg%253D&md5=022788aecea44e220ad93ec48af21ae3Differential Effects of Divalent Manganese and Magnesium on the Kinase Activity of Leucine-Rich Repeat Kinase 2 (LRRK2)Lovitt, Brian; Vander Porten, Erica C.; Sheng, Zejuan; Zhu, Haitao; Drummond, Jake; Liu, YichinBiochemistry (2010), 49 (14), 3092-3100CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Various mutations in leucine-rich repeat kinase 2 (LRRK2) have been linked to susceptibility for both familial and idiopathic late-onset Parkinson's disease (PD). In this study, it was demonstrated that phosphorylation of MBP and LRRKtide by the LRRK2 G2019S mutant was activated by Mn2+ in vitro. This enhanced G2019S kinase activity was due to the combination of an increase in kinase and a decrease in ATPase activity by Mn2+. Compared to 10 mM Mg2+, 1 mM Mn2+ reduced ATP Km for G2019S from 103 to 1.8 μM and only modestly reduced kcat (2.5-fold); as a result, the Mn2+ increased its kcat/Km by 22-fold. This change in ATP Km was due in large part to an increase in nucleotide affinity. While Mn2+ also increased ATP affinity and had similar effects on kcat/Km for LRRK2 WT and R1441C enzymes, it reduced their kcat values significantly by 13-17-fold. Consequently, the difference in the kinase activity between G2019S and other LRRK2 variants was enhanced from about 2-fold in Mg2+ to 10-fold in Mn2+ at satg. ATP concns. relative to its Km. Furthermore, while Mg2+ yielded optimal Vmax values at Mg2+ concn. greater than 5 mM, the optimal Mn2+ concn. for activating LRRK2 catalysis was in the micromolar range with increasing Mn2+ above 1 mM causing a decrease in enzyme activity. Finally, despite the large but expected differences in IC50 tested at 100 μM ATP, the apparent Ki values of a small set of LRRK2 ATP-competitive inhibitors were within 5-fold between Mg2+- and Mn2+-mediated reactions except AMP-CPP, an ATP analog.
- 30Haq, N.; Niu, D.; Petter, R. C.; Qiao, L.; Singh, J.; Zhu, Z. Pyrimidine derivatives as ERK inhibitors and their preparation. Patent WO2014124230A2, 2014.Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 43 publications.
- Hongfu Lu, Yihong Zhang, Jinxin Liu, Tao Jiang, Xiang Yu, Haoyu Zhang, Tao Liang, Jingjing Peng, Xin Cai, Xiaoling Lan, Jinmin Ren, Mei Ge, Jingyang Zhang, Jingjin Shang, Jiaojiao Yu, Hongcan Ren, Qiang Liu, Jinting Gao, Lili Tang, Xiao Ding, Man Zhang, Alex Aliper, Qiang Lu, Fusheng Zhou, Jiong Lan, Feng Ren, Alex Zhavoronkov. Discovery of a Novel Macrocyclic Noncovalent CDK7 Inhibitor for Cancer Therapy. Journal of Medicinal Chemistry 2024, 67
(22)
, 20580-20594. https://doi.org/10.1021/acs.jmedchem.4c02098
- Funing Lin, Mark F. Mabanglo, Jin Lin Zhou, Gursonika Binepal, Marim M. Barghash, Keith S. Wong, Scott D. Gray-Owen, Robert A. Batey, Walid A. Houry. Structure-Based Design and Development of Phosphine Oxides as a Novel Chemotype for Antibiotics that Dysregulate Bacterial ClpP Proteases. Journal of Medicinal Chemistry 2024, 67
(17)
, 15131-15147. https://doi.org/10.1021/acs.jmedchem.4c00773
- Hualin Zhang, Yutong Tu, Zhaofan Tao, Lixin Gao, Shengjie Huang, Mingshan Gao, Jialuo Mao, Yang Zhou, Yupeng Li, Jia Li, Yubo Zhou, Tianfeng Xu. Design, Synthesis, and Biological Evaluation of 2,4-Diaminopyrimidine Derivatives as Potent CDK7 Inhibitors. ACS Medicinal Chemistry Letters 2024, 15
(8)
, 1213-1220. https://doi.org/10.1021/acsmedchemlett.4c00040
- Pritesh Keshari, Rajan Yadav, Priya Mata, Susanta Hazra. Micelles Enabled Nickel-Catalyzed P-Arylation of Aromatic Phosphine Oxide in Water Under Mild Conditions. ACS Sustainable Chemistry & Engineering 2024, 12
(31)
, 11473-11479. https://doi.org/10.1021/acssuschemeng.4c03527
- Pengpeng Niu, Yanxin Tao, Guohao Lin, Huiqi Xu, Qingyuan Meng, Kang Yang, Weixue Huang, Meiru Song, Ke Ding, Dawei Ma, Mengyang Fan. Design and Synthesis of Novel Macrocyclic Derivatives as Potent and Selective Cyclin-Dependent Kinase 7 Inhibitors. Journal of Medicinal Chemistry 2024, 67
(8)
, 6099-6118. https://doi.org/10.1021/acs.jmedchem.3c01832
- Zhuo Zhang, Mingrui Liu, Min Liu, Chenhong Pan, Zhengtong Mao, Xingxian Zhang. Visible-Light-Induced Highly Site-Selective Direct C–H Phosphorylation of Pyrrolo[2,3-d]pyrimidine Derivatives with H-Phosphine Oxides. The Journal of Organic Chemistry 2024, 89
(5)
, 2996-3009. https://doi.org/10.1021/acs.joc.3c02416
- Zhenfeng Shi, Lei Tian, Taotao Qiang, Jingyi Li, Yue Xing, Xiaodong Ren, Chang Liu, Chengyuan Liang. From Structure Modification to Drug Launch: A Systematic Review of the Ongoing Development of Cyclin-Dependent Kinase Inhibitors for Multiple Cancer Therapy. Journal of Medicinal Chemistry 2022, 65
(9)
, 6390-6418. https://doi.org/10.1021/acs.jmedchem.1c02064
- Zhouling Xie, Shuzeng Hou, Xiaoxiao Yang, Yajun Duan, Jihong Han, Qin Wang, Chenzhong Liao. Lessons Learned from Past Cyclin-Dependent Kinase Drug Discovery Efforts. Journal of Medicinal Chemistry 2022, 65
(9)
, 6356-6389. https://doi.org/10.1021/acs.jmedchem.1c02190
- Jana Škerlová, Veronika Krejčiříková, Miroslav Peřina, Veronika Vojáčková, Milan Fábry, Vladimír Kryštof, Radek Jorda, Pavlína Řezáčová. CDK2-based CDK7 mimic as a tool for structural analysis: Biochemical validation and crystal structure with SY5609. International Journal of Biological Macromolecules 2025, 294 , 139117. https://doi.org/10.1016/j.ijbiomac.2024.139117
- Olivia Luyties, Lynn Sanford, Jessica Rodino, Michael Nagel, Taylor Jones, Jenna K. Rimel, Christopher C. Ebmeier, Grace S. Shelby, Kira Cozzolino, Finn Brennan, Axel Hartzog, Mirzam B. Saucedo, Lotte P. Watts, Sabrina Spencer, Jennifer F. Kugel, Robin D. Dowell, Dylan J. Taatjes. Multi-omics and biochemical reconstitution reveal CDK7-dependent mechanisms controlling RNA polymerase II function at gene 5’- and 3’-ends. 2025https://doi.org/10.1101/2025.01.08.632016
- Victoria I. Cushing, Adrian F. Koh, Junjie Feng, Kaste Jurgaityte, Alexander Bondke, Sebastian H. B. Kroll, Marion Barbazanges, Bodo Scheiper, Ash K. Bahl, Anthony G. M. Barrett, Simak Ali, Abhay Kotecha, Basil J. Greber. High-resolution cryo-EM of the human CDK-activating kinase for structure-based drug design. Nature Communications 2024, 15
(1)
https://doi.org/10.1038/s41467-024-46375-9
- , Izhar Wallach, Denzil Bernard, Kong Nguyen, Gregory Ho, Adrian Morrison, Adrian Stecula, Andreana Rosnik, Ann Marie O’Sullivan, Aram Davtyan, Ben Samudio, Bill Thomas, Brad Worley, Brittany Butler, Christian Laggner, Desiree Thayer, Ehsan Moharreri, Greg Friedland, Ha Truong, Henry van den Bedem, Ho Leung Ng, Kate Stafford, Krishna Sarangapani, Kyle Giesler, Lien Ngo, Michael Mysinger, Mostafa Ahmed, Nicholas J. Anthis, Niel Henriksen, Pawel Gniewek, Sam Eckert, Saulo de Oliveira, Shabbir Suterwala, Srimukh Veccham Krishna PrasadPrasad, Stefani Shek, Stephanie Contreras, Stephanie Hare, Teresa Palazzo, Terrence E. O’Brien, Tessa Van Grack, Tiffany Williams, Ting-Rong Chern, Victor Kenyon, Andreia H. Lee, Andrew B. Cann, Bastiaan Bergman, Brandon M. Anderson, Bryan D. Cox, Jeffrey M. Warrington, Jon M. Sorenson, Joshua M. Goldenberg, Matthew A. Young, Nicholas DeHaan, Ryan P. Pemberton, Stefan Schroedl, Tigran M. Abramyan, Tushita Gupta, Venkatesh Mysore, Adam G. Presser, Adolfo A. Ferrando, Adriano D. Andricopulo, Agnidipta Ghosh, Aicha Gharbi Ayachi, Aisha Mushtaq, Ala M. Shaqra, Alan Kie Leong Toh, Alan V. Smrcka, Alberto Ciccia, Aldo Sena de Oliveira, Aleksandr Sverzhinsky, Alessandra Mara de Sousa, Alexander I. Agoulnik, Alexander Kushnir, Alexander N. Freiberg, Alexander V. Statsyuk, Alexandre R. Gingras, Alexei Degterev, Alexey Tomilov, Alice Vrielink, Alisa A. Garaeva, Amanda Bryant-Friedrich, Amedeo Caflisch, Amit K. Patel, Amith Vikram Rangarajan, An Matheeussen, Andrea Battistoni, Andrea Caporali, Andrea Chini, Andrea Ilari, Andrea Mattevi, Andrea Talbot Foote, Andrea Trabocchi, Andreas Stahl, Andrew B. Herr, Andrew Berti, Andrew Freywald, Andrew G. Reidenbach, Andrew Lam, Andrew R. Cuddihy, Andrew White, Angelo Taglialatela, Anil K. Ojha, Ann M. Cathcart, Anna A. L. Motyl, Anna Borowska, Anna D’Antuono, Anna K. H. Hirsch, Anna Maria Porcelli, Anna Minakova, Anna Montanaro, Anna Müller, Annarita Fiorillo, Anniina Virtanen, Anthony J. O’Donoghue, Antonio Del Rio Flores, Antonio E. Garmendia, Antonio Pineda-Lucena, Antonito T. Panganiban, Ariela Samantha, Arnab K. Chatterjee, Arthur L. Haas, Ashleigh S. Paparella, Ashley L. St. John, Ashutosh Prince, Assmaa ElSheikh, Athena Marie Apfel, Audrey Colomba, Austin O’Dea, Bakary N’tji Diallo, Beatriz Murta Rezende Moraes Ribeiro, Ben A. Bailey-Elkin, Benjamin L. Edelman, Benjamin Liou, Benjamin Perry, Benjamin Soon Kai Chua, Benjámin Kováts, Bernhard Englinger, Bijina Balakrishnan, Bin Gong, Bogos Agianian, Brandon Pressly, Brenda P. Medellin Salas, Brendan M. Duggan, Brian V. Geisbrecht, Brian W. Dymock, Brianna C. Morten, Bruce D. Hammock, Bruno Eduardo Fernandes Mota, Bryan C. Dickinson, Cameron Fraser, Camille Lempicki, Carl D. Novina, Carles Torner, Carlo Ballatore, Carlotta Bon, Carly J. Chapman, Carrie L. Partch, Catherine T. Chaton, Chang Huang, Chao-Yie Yang, Charlene M. Kahler, Charles Karan, Charles Keller, Chelsea L. Dieck, Chen Huimei, Chen Liu, Cheryl Peltier, Chinmay Kumar Mantri, Chinyere Maat Kemet, Christa E. Müller, Christian Weber, Christina M. Zeina, Christine S. Muli, Christophe Morisseau, Cigdem Alkan, Clara Reglero, Cody A. Loy, Cornelia M. Wilson, Courtney Myhr, Cristina Arrigoni, Cristina Paulino, César Santiago, Dahai Luo, Damon J. Tumes, Daniel A. Keedy, Daniel A. Lawrence, Daniel Chen, Danny Manor, Darci J. Trader, David A. Hildeman, David H. Drewry, David J. Dowling, David J. Hosfield, David M. Smith, David Moreira, David P. Siderovski, David Shum, David T. Krist, David W. H. Riches, Davide Maria Ferraris, Deborah H. Anderson, Deirdre R. Coombe, Derek S. Welsbie, Di Hu, Diana Ortiz, Dina Alramadhani, Dingqiang Zhang, Dipayan Chaudhuri, Dirk J. Slotboom, Donald R. Ronning, Donghan Lee, Dorian Dirksen, Douglas A. Shoue, Douglas William Zochodne, Durga Krishnamurthy, Dustin Duncan, Dylan M. Glubb, Edoardo Luigi Maria Gelardi, Edward C. Hsiao, Edward G. Lynn, Elany Barbosa Silva, Elena Aguilera, Elena Lenci, Elena Theres Abraham, Eleonora Lama, Eleonora Mameli, Elisa Leung, Ellie Giles, Emily M. Christensen, Emily R. Mason, Enrico Petretto, Ephraim F. Trakhtenberg, Eric J. Rubin, Erick Strauss, Erik W. Thompson, Erika Cione, Erika Mathes Lisabeth, Erkang Fan, Erna Geessien Kroon, Eunji Jo, Eva M. García-Cuesta, Evgenia Glukhov, Evripidis Gavathiotis, Fang Yu, Fei Xiang, Fenfei Leng, Feng Wang, Filippo Ingoglia, Focco van den Akker, Francesco Borriello, Franco J. Vizeacoumar, Frank Luh, Frederick S. Buckner, Frederick S. Vizeacoumar, Fredj Ben Bdira, Fredrik Svensson, G. Marcela Rodriguez, Gabriella Bognár, Gaia Lembo, Gang Zhang, Garrett Dempsey, Gary Eitzen, Gaétan Mayer, Geoffrey L. Greene, George A. Garcia, Gergely L. Lukacs, Gergely Prikler, Gian Carlo G. Parico, Gianni Colotti, Gilles De Keulenaer, Gino Cortopassi, Giovanni Roti, Giulia Girolimetti, Giuseppe Fiermonte, Giuseppe Gasparre, Giuseppe Leuzzi, Gopal Dahal, Gracjan Michlewski, Graeme L. Conn, Grant David Stuchbury, Gregory R. Bowman, Grzegorz Maria Popowicz, Guido Veit, Guilherme Eduardo de Souza, Gustav Akk, Guy Caljon, Guzmán Alvarez, Gwennan Rucinski, Gyeongeun Lee, Gökhan Cildir, Hai Li, Hairol E. Breton, Hamed Jafar-Nejad, Han Zhou, Hannah P. Moore, Hannah Tilford, Haynes Yuan, Heesung Shim, Heike Wulff, Heinrich Hoppe, Helena Chaytow, Heng-Keat Tam, Holly Van Remmen, Hongyang Xu, Hosana Maria Debonsi, Howard B. Lieberman, Hoyoung Jung, Hua-Ying Fan, Hui Feng, Hui Zhou, Hyeong Jun Kim, Iain R. Greig, Ileana Caliandro, Ileana Corvo, Imanol Arozarena, Imran N. Mungrue, Ingrid M. Verhamme, Insaf Ahmed Qureshi, Irina Lotsaris, Isin Cakir, J. Jefferson P. Perry, Jacek Kwiatkowski, Jacob Boorman, Jacob Ferreira, Jacob Fries, Jadel Müller Kratz, Jaden Miner, Jair L. Siqueira-Neto, James G. Granneman, James Ng, James Shorter, Jan Hendrik Voss, Jan M. Gebauer, Janelle Chuah, Jarrod J. Mousa, Jason T. Maynes, Jay D. Evans, Jeffrey Dickhout, Jeffrey P. MacKeigan, Jennifer N. Jossart, Jia Zhou, Jiabei Lin, Jiake Xu, Jianghai Wang, Jiaqi Zhu, Jiayu Liao, Jingyi Xu, Jinshi Zhao, Jiusheng Lin, Jiyoun Lee, Joana Reis, Joerg Stetefeld, John B. Bruning, John Burt Bruning, John G. Coles, John J. Tanner, John M. Pascal, Jonathan So, Jordan L. Pederick, Jose A. Costoya, Joseph B. Rayman, Joseph J. Maciag, Joshua Alexander Nasburg, Joshua J. Gruber, Joshua M. Finkelstein, Joshua Watkins, José Miguel Rodríguez-Frade, Juan Antonio Sanchez Arias, Juan José Lasarte, Julen Oyarzabal, Julian Milosavljevic, Julie Cools, Julien Lescar, Julijus Bogomolovas, Jun Wang, Jung-Min Kee, Jung-Min Kee, Junzhuo Liao, Jyothi C. Sistla, Jônatas Santos Abrahão, Kamakshi Sishtla, Karol R. Francisco, Kasper B. Hansen, Kathleen A. Molyneaux, Kathryn A. Cunningham, Katie R. Martin, Kavita Gadar, Kayode K. Ojo, Keith S. Wong, Kelly L. Wentworth, Kent Lai, Kevin A. Lobb, Kevin M. Hopkins, Keykavous Parang, Khaled Machaca, Kien Pham, Kim Ghilarducci, Kim S. Sugamori, Kirk James McManus, Kirsikka Musta, Kiterie M. E. Faller, Kiyo Nagamori, Konrad J. Mostert, Konstantin V. Korotkov, Koting Liu, Kristiana S. Smith, Kristopher Sarosiek, Kyle H. Rohde, Kyu Kwang Kim, Kyung Hyeon Lee, Lajos Pusztai, Lari Lehtiö, Larisa M. Haupt, Leah E. Cowen, Lee J. Byrne, Leila Su, Leon Wert-Lamas, Leonor Puchades-Carrasco, Lifeng Chen, Linda H. Malkas, Ling Zhuo, Lizbeth Hedstrom, Lizbeth Hedstrom, Loren D. Walensky, Lorenzo Antonelli, Luisa Iommarini, Luke Whitesell, Lía M. Randall, M. Dahmani Fathallah, Maira Harume Nagai, Mairi Louise Kilkenny, Manu Ben-Johny, Marc P. Lussier, Marc P. Windisch, Marco Lolicato, Marco Lucio Lolli, Margot Vleminckx, Maria Cristina Caroleo, Maria J. Macias, Marilia Valli, Marim M. Barghash, Mario Mellado, Mark A. Tye, Mark A. Wilson, Mark Hannink, Mark R. Ashton, Mark Vincent C.dela Cerna, Marta Giorgis, Martin K. Safo, Martin St. Maurice, Mary Ann McDowell, Marzia Pasquali, Masfique Mehedi, Mateus Sá Magalhães Serafim, Matthew B. Soellner, Matthew G. Alteen, Matthew M. Champion, Maxim Skorodinsky, Megan L. O’Mara, Mel Bedi, Menico Rizzi, Michael Levin, Michael Mowat, Michael R. Jackson, Mikell Paige, Minnatallah Al-Yozbaki, Miriam A. Giardini, Mirko M. Maksimainen, Monica De Luise, Muhammad Saddam Hussain, Myron Christodoulides, Natalia Stec, Natalia Zelinskaya, Natascha Van Pelt, Nathan M. Merrill, Nathanael Singh, Neeltje A. Kootstra, Neeraj Singh, Neha S. Gandhi, Nei-Li Chan, Nguyen Mai Trinh, Nicholas O. Schneider, Nick Matovic, Nicola Horstmann, Nicola Longo, Nikhil Bharambe, Nirvan Rouzbeh, Niusha Mahmoodi, Njabulo Joyfull Gumede, Noelle C. Anastasio, Noureddine Ben Khalaf, Obdulia Rabal, Olga Kandror, Olivier Escaffre, Olli Silvennoinen, Ozlem Tastan Bishop, Pablo Iglesias, Pablo Sobrado, Patrick Chuong, Patrick O’Connell, Pau Martin-Malpartida, Paul Mellor, Paul V. Fish, Paulo Otávio Lourenço Moreira, Pei Zhou, Pengda Liu, Pengda Liu, Pengpeng Wu, Percy Agogo-Mawuli, Peter L. Jones, Peter Ngoi, Peter Toogood, Philbert Ip, Philipp von Hundelshausen, Pil H. Lee, Rachael B. Rowswell-Turner, Rafael Balaña-Fouce, Rafael Eduardo Oliveira Rocha, Rafael V. C. Guido, Rafaela Salgado Ferreira, Rajendra K. Agrawal, Rajesh K. Harijan, Rajesh Ramachandran, Rajkumar Verma, Rakesh K. Singh, Rakesh Kumar Tiwari, Ralph Mazitschek, Rama K. Koppisetti, Remus T. Dame, Renée N. Douville, Richard C. Austin, Richard E. Taylor, Richard G. Moore, Richard H. Ebright, Richard M. Angell, Riqiang Yan, Rishabh Kejriwal, Robert A. Batey, Robert Blelloch, Robert J. Vandenberg, Robert J. Hickey, Robert J. Kelm, Robert J. Lake, Robert K. Bradley, Robert M. Blumenthal, Roberto Solano, Robin Matthias Gierse, Ronald E. Viola, Ronan R. McCarthy, Rosa Maria Reguera, Ruben Vazquez Uribe, Rubens Lima do Monte-Neto, Ruggiero Gorgoglione, Ryan T. Cullinane, Sachin Katyal, Sakib Hossain, Sameer Phadke, Samuel A. Shelburne, Sandra E. Geden, Sandra Johannsen, Sarah Wazir, Scott Legare, Scott M. Landfear, Senthil K. Radhakrishnan, Serena Ammendola, Sergei Dzhumaev, Seung-Yong Seo, Shan Li, Shan Zhou, Shaoyou Chu, Shefali Chauhan, Shinsaku Maruta, Shireen R. Ashkar, Show-Ling Shyng, Silvestro G. Conticello, Silvia Buroni, Silvia Garavaglia, Simon J. White, Siran Zhu, Sofiya Tsimbalyuk, Somaia Haque Chadni, Soo Young Byun, Soonju Park, Sophia Q. Xu, Sourav Banerjee, Stefan Zahler, Stefano Espinoza, Stefano Gustincich, Stefano Sainas, Stephanie L. Celano, Stephen J. Capuzzi, Stephen N. Waggoner, Steve Poirier, Steven H. Olson, Steven O. Marx, Steven R. Van Doren, Suryakala Sarilla, Susann M. Brady-Kalnay, Sydney Dallman, Syeda Maryam Azeem, Tadahisa Teramoto, Tamar Mehlman, Tarryn Swart, Tatjana Abaffy, Tatos Akopian, Teemu Haikarainen, Teresa Lozano Moreda, Tetsuro Ikegami, Thaiz Rodrigues Teixeira, Thilina D. Jayasinghe, Thomas H. Gillingwater, Thomas Kampourakis, Timothy I. Richardson, Timothy J. Herdendorf, Timothy J. Kotzé, Timothy R. O’Meara, Timothy W. Corson, Tobias Hermle, Tomisin Happy Ogunwa, Tong Lan, Tong Su, Toshihiro Banjo, Tracy A. O’Mara, Tristan Chou, Tsui-Fen Chou, Ulrich Baumann, Umesh R. Desai, Vaibhav P. Pai, Van Chi Thai, Vasudha Tandon, Versha Banerji, Victoria L. Robinson, Vignesh Gunasekharan, Vigneshwaran Namasivayam, Vincent F. M. Segers, Vincent Maranda, Vincenza Dolce, Vinícius Gonçalves Maltarollo, Viola Camilla Scoffone, Virgil A. Woods, Virginia Paola Ronchi, Vuong Van Hung Le, W. Brent Clayton, W. Todd Lowther, Walid A. Houry, Wei Li, Weiping Tang, Wenjun Zhang, Wesley C. Van Voorhis, William A. Donaldson, William C. Hahn, William G. Kerr, William H. Gerwick, William J. Bradshaw, Wuen Ee Foong, Xavier Blanchet, Xiaoyang Wu, Xin Lu, Xin Qi, Xin Xu, Xinfang Yu, Xingping Qin, Xingyou Wang, Xinrui Yuan, Xu Zhang, Yan Jessie Zhang, Yanmei Hu, Yasser Ali Aldhamen, Yicheng Chen, Yihe Li, Ying Sun, Yini Zhu, Yogesh K. Gupta, Yolanda Pérez-Pertejo, Yong Li, Young Tang, Yuan He, Yuk-Ching Tse-Dinh, Yulia A. Sidorova, Yun Yen, Yunlong Li, Zachary J. Frangos, Zara Chung, Zhengchen Su, Zhenghe Wang, Zhiguo Zhang, Zhongle Liu, Zintis Inde, Zoraima Artía, Abraham Heifets. AI is a viable alternative to high throughput screening: a 318-target study. Scientific Reports 2024, 14
(1)
https://doi.org/10.1038/s41598-024-54655-z
- Ying Gong, Huiping Li. CDK7 in breast cancer: mechanisms of action and therapeutic potential. Cell Communication and Signaling 2024, 22
(1)
https://doi.org/10.1186/s12964-024-01577-y
- Ping Song, Rongyan Han, Fan Yang. Super enhancer lncRNAs: a novel hallmark in cancer. Cell Communication and Signaling 2024, 22
(1)
https://doi.org/10.1186/s12964-024-01599-6
- Wenzhi Ji, Guangyan Du, Jie Jiang, Wenchao Lu, Caitlin E. Mills, Linjie Yuan, Fen Jiang, Zhixiang He, Gary A. Bradshaw, Mirra Chung, Zixuan Jiang, Woong Sub Byun, Stephen M. Hinshaw, Tinghu Zhang, Nathanael S. Gray. Discovery of bivalent small molecule degraders of cyclin-dependent kinase 7 (CDK7). European Journal of Medicinal Chemistry 2024, 276 , 116613. https://doi.org/10.1016/j.ejmech.2024.116613
- Mahder Dawit Belew, Jingrui Chen, Zhaokang Cheng. Emerging roles of cyclin-dependent kinase 7 in health and diseases. Trends in Molecular Medicine 2024, 15 https://doi.org/10.1016/j.molmed.2024.09.004
- Yaoguang Huang, Wenwu Liu, Changhao Zhao, Xiaoyu Shi, Qingchun Zhao, Jingming Jia, Anhua Wang. Targeting cyclin-dependent kinases: From pocket specificity to drug selectivity. European Journal of Medicinal Chemistry 2024, 275 , 116547. https://doi.org/10.1016/j.ejmech.2024.116547
- Manjeet Mukherjee, Philip J. Day, Duncan Laverty, Juan A. Bueren-Calabuig, Andrew J. Woodhead, Charlotte Griffiths-Jones, Steve Hiscock, Charlotte East, Susan Boyd, Marc O'Reilly. Protein engineering enables a soakable crystal form of human CDK7 primed for high-throughput crystallography and structure-based drug design. Structure 2024, 32
(8)
, 1040-1048.e3. https://doi.org/10.1016/j.str.2024.05.011
- Hongjin Zhang, Guohao Lin, Suyun Jia, Jianbo Wu, Ying Zhang, Yanxin Tao, Weixue Huang, Meiru Song, Ke Ding, Dawei Ma, Mengyang Fan. Design, synthesis and evaluation of thieno[3,2-d]pyrimidine derivatives as novel potent CDK7 inhibitors. Bioorganic Chemistry 2024, 148 , 107456. https://doi.org/10.1016/j.bioorg.2024.107456
- Dominic D.G. Owens, Matthew E.R. Maitland, Cheryl H. Arrowsmith, Dalia Barsyte-Lovejoy. Therapeutic Discovery for Chromatin Complexes: Where Do We Stand?. Annual Review of Cancer Biology 2024, 8
(1)
, 373-393. https://doi.org/10.1146/annurev-cancerbio-062822-110356
- Xue Song, Chen Fang, Yan Dai, Yang Sun, Chang Qiu, Xiaojie Lin, Rui Xu. Cyclin-dependent kinase 7 (CDK7) inhibitors as a novel therapeutic strategy for different molecular types of breast cancer. British Journal of Cancer 2024, 130
(8)
, 1239-1248. https://doi.org/10.1038/s41416-024-02589-8
- Basil J. Greber. High-resolution cryo-EM of a small protein complex: The structure of the human CDK-activating kinase. Structure 2024, 32
(4)
, 380-392. https://doi.org/10.1016/j.str.2024.03.003
- Shuvam M. Chaudhuri, Samuel E. Weinberg, Dongmei Wang, Lenore K. Yalom, Elena Montauti, Radhika Iyer, Amy Y. Tang, Manuel A. Torres Acosta, Jian Shen, Nikita L. Mani, Shengnan Wang, Kun Liu, Weiyuan Lu, Triet M. Bui, Laura D. Manzanares, Zeinab Dehghani, Ching Man Wai, Beixue Gao, Juncheng Wei, Feng Yue, Weiguo Cui, Benjamin D. Singer, Ronen Sumagin, Yana Zhang, Deyu Fang. Mediator complex subunit 1 architects a tumorigenic Treg cell program independent of inflammation. Cell Reports Medicine 2024, 5
(3)
, 101441. https://doi.org/10.1016/j.xcrm.2024.101441
- Hongjin Zhang, Guohao Lin, Suyun Jia, Ying Zhang, Jianbo Wu, Yanxin Tao, Weixue Huang, Meiru Song, Ke Ding, Dawei Ma, Mengyang Fan. Discovery and optimization of thieno[3,2-d]pyrimidine derivatives as highly selective inhibitors of cyclin-dependent kinase 7. European Journal of Medicinal Chemistry 2024, 263 , 115955. https://doi.org/10.1016/j.ejmech.2023.115955
- Ratnakar Reddy Kuchukulla, Injeoung Hwang, Suhn Hyung Kim, Younghyeon Kye, Narae Park, Heary Cha, Sojeong Moon, Hwan Won Chung, Cheolju Lee, Gu Kong, Wooyoung Hur. Identification of a novel potent CDK inhibitor degrading cyclinK with a superb activity to reverse trastuzumab-resistance in HER2-positive breast cancer in vivo. European Journal of Medicinal Chemistry 2024, 264 , 116014. https://doi.org/10.1016/j.ejmech.2023.116014
- Lanshu Xiao, Yi Liu, Hui Chen, Lisong Shen. Targeting CDK9 with selective inhibitors or degraders in tumor therapy: an overview of recent developments. Cancer Biology & Therapy 2023, 24
(1)
https://doi.org/10.1080/15384047.2023.2219470
- Matthias Ocker, Daniel Neureiter. Gene expression inhibitors for the treatment of liver fibrosis: drugs under preclinical and early clinical investigation. Expert Opinion on Investigational Drugs 2023, 32
(12)
, 1133-1141. https://doi.org/10.1080/13543784.2023.2288075
- Rafael Navrátil, Kristýna Kellovská, Ondřej Baszczyňski. Multimetallic Pd- and Ni-catalyzed C(sp
2
)–P cross-coupling under aqueous micellar conditions. Green Chemistry 2023, 25
(23)
, 9779-9794. https://doi.org/10.1039/D3GC02735J
- Liguo Wang, Zhouli Yang, Guangchen Li, Yongbo Liu, Chao Ai, Yu Rao. Discovery of small molecule degraders for modulating cell cycle. Frontiers of Medicine 2023, 17
(5)
, 823-854. https://doi.org/10.1007/s11684-023-1027-5
- Shuai Liu, Fang Xia Pan, Yu Qi, Hai Yun Bai. Hexafluoroisopropanol/trifluoromethanesulfonic acid assists the synthesis of 3-(2-chloropyrimidin-4-yl)-1H-indoles. Tetrahedron 2023, 146 , 133630. https://doi.org/10.1016/j.tet.2023.133630
- Minqiang Jia, Weimin Wang, Gang Chen, Ting Wu, Ting Zhang, Qian Zhou, Junzhao Yin, Jie Li, Xun Li, Yuchang Mao, Jun Feng, Min Hu, Xin Li, Feng He. Discovery of SHR5428 as a selective and noncovalent inhibitor of CDK7. Bioorganic & Medicinal Chemistry Letters 2023, 93 , 129429. https://doi.org/10.1016/j.bmcl.2023.129429
- Reza Emadi, Abbas Bahrami Nekoo, Fatemeh Molaverdi, Zahra Khorsandi, Reza Sheibani, Hojjat Sadeghi-Aliabadi. Applications of palladium-catalyzed C–N cross-coupling reactions in pharmaceutical compounds. RSC Advances 2023, 13
(27)
, 18715-18733. https://doi.org/10.1039/D2RA07412E
- Markéta Kovalová, Libor Havlíček, Stefan Djukic, Jana Škerlová, Miroslav Peřina, Tomáš Pospíšil, Eva Řezníčková, Pavlína Řezáčová, Radek Jorda, Vladimír Kryštof. Characterization of new highly selective pyrazolo[4,3-d]pyrimidine inhibitor of CDK7. Biomedicine & Pharmacotherapy 2023, 161 , 114492. https://doi.org/10.1016/j.biopha.2023.114492
- Victoria I. Cushing, Adrian F. Koh, Junjie Feng, Kaste Jurgaityte, Ash K. Bahl, Simak Ali, Abhay Kotecha, Basil J. Greber. High-resolution cryo-electron microscopy of the human CDK-activating kinase for structure-based drug design. 2023https://doi.org/10.1101/2023.04.07.536029
- Chiara Tarantelli and, Francesco Bertoni. Targeting the Cell Cycle and Cyclin‐dependent Kinases. 2023, 444-456. https://doi.org/10.1002/9781119819950.ch31
- Markéta Kovalová, Libor Havlíček, Stefan Djukic, Jana Škerlová, Miroslav Peřina, Tomáš Pospíšil, Eva Řezníčková, Pavlína Řezáčová, Radek Jorda, Vladimír Kryštof. Characterization of new highly selective pyrazolo[4,3-d]pyrimidine inhibitor of CDK7. 2023https://doi.org/10.1101/2023.02.24.529844
- Feng Ren, Xiao Ding, Min Zheng, Mikhail Korzinkin, Xin Cai, Wei Zhu, Alexey Mantsyzov, Alex Aliper, Vladimir Aladinskiy, Zhongying Cao, Shanshan Kong, Xi Long, Bonnie Hei Man Liu, Yingtao Liu, Vladimir Naumov, Anastasia Shneyderman, Ivan V. Ozerov, Ju Wang, Frank W. Pun, Daniil A. Polykovskiy, Chong Sun, Michael Levitt, Alán Aspuru-Guzik, Alex Zhavoronkov. AlphaFold accelerates artificial intelligence powered drug discovery: efficient discovery of a novel CDK20 small molecule inhibitor. Chemical Science 2023, 14
(6)
, 1443-1452. https://doi.org/10.1039/D2SC05709C
- Markéta Kovalová, Joseph Peter Baraka, Václav Mik, Radek Jorda, Lei Luo, Hao Shao, Vladimír Kryštof. A patent review of cyclin-dependent kinase 7 (CDK7) inhibitors (2018-2022). Expert Opinion on Therapeutic Patents 2023, 33
(2)
, 67-87. https://doi.org/10.1080/13543776.2023.2195547
- Shuai Liu, Fang Xia Pan, Yu Qi, Hai Yun Bai. Hexafluoroisopropanol/Trifluoromethanesulfonic Acid Assists the Synthesis of 3- (4-Pyrimidineyl)-1h-Indole Derivatives. 2023https://doi.org/10.2139/ssrn.4505895
- Zhi-Mei Li, Guan Liu, Ya Gao, Ming-Gao Zhao. Targeting CDK7 in oncology: The avenue forward. Pharmacology & Therapeutics 2022, 240 , 108229. https://doi.org/10.1016/j.pharmthera.2022.108229
- Micah G. Donovan, Matthew D. Galbraith, Joaquin M. Espinosa. Multi-omics investigation reveals functional specialization of transcriptional cyclin dependent kinases in cancer biology. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-26860-1
- Kevin C. Clopper, Dylan J. Taatjes. Chemical inhibitors of transcription-associated kinases. Current Opinion in Chemical Biology 2022, 70 , 102186. https://doi.org/10.1016/j.cbpa.2022.102186
- Theodora A. Constantin, Kyle K. Greenland, Anabel Varela-Carver, Charlotte L. Bevan. Transcription associated cyclin-dependent kinases as therapeutic targets for prostate cancer. Oncogene 2022, 41
(24)
, 3303-3315. https://doi.org/10.1038/s41388-022-02347-1
- Seng Chuan Tang, Udhaya Vijayakumar, Ying Zhang, Melissa Jane Fullwood. Super-Enhancers, Phase-Separated Condensates, and 3D Genome Organization in Cancer. Cancers 2022, 14
(12)
, 2866. https://doi.org/10.3390/cancers14122866
- . Synthesis of CDK7 Inhibitor SY-5609. Synfacts 2022, 0353. https://doi.org/10.1055/s-0041-1737933
- Kai Funke, Robert Düster, Prince De-Graft Wilson, Lena Arévalo, Matthias Geyer, Hubert Schorle. Transcriptional CDK Inhibitors as Potential Treatment Option for Testicular Germ Cell Tumors. Cancers 2022, 14
(7)
, 1690. https://doi.org/10.3390/cancers14071690
- Tobias Ettl, Daniela Schulz, Richard Bauer. The Renaissance of Cyclin Dependent Kinase Inhibitors. Cancers 2022, 14
(2)
, 293. https://doi.org/10.3390/cancers14020293
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 30 other publications.
- 1Hanahan, D.; Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 2011, 144, 646– 674, DOI: 10.1016/j.cell.2011.02.0131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjsFeqtrk%253D&md5=b36f160c417a712e83cb83c577f0018eHallmarks of cancer: the next generationHanahan, Douglas; Weinberg, Robert A.Cell (Cambridge, MA, United States) (2011), 144 (5), 646-674CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. The hallmarks of cancer comprise six biol. capabilities acquired during the multistep development of human tumors. The hallmarks constitute an organizing principle for rationalizing the complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. Underlying these hallmarks are genome instability, which generates the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hallmark functions. Conceptual progress in the last decade has added two emerging hallmarks of potential generality to this list-reprogramming of energy metab. and evading immune destruction. In addn. to cancer cells, tumors exhibit another dimension of complexity: they contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hallmark traits by creating the "tumor microenvironment.". Recognition of the widespread applicability of these concepts will increasingly affect the development of new means to treat human cancer.
- 2Sava, G. P.; Fan, H.; Coombes, R. C.; Buluwela, L.; Ali, S. CDK7 inhibitors as anticancer drugs. Cancer Metastasis Rev. 2020, 39, 805– 823, DOI: 10.1007/s10555-020-09885-82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXptVersL4%253D&md5=b61a237dcb6b1caa843baadf74df87ffCDK7 inhibitors as anticancer drugsSava, Georgina P.; Fan, Hailing; Coombes, R. Charles; Buluwela, Lakjaya; Ali, SimakCancer and Metastasis Reviews (2020), 39 (3), 805-823CODEN: CMRED4; ISSN:0167-7659. (Springer)A review. Cyclin-dependent kinase 7 (CDK7), along with cyclin H and MAT1, forms the CDK-activating complex (CAK), which directs progression through the cell cycle via T-loop phosphorylation of cell cycle CDKs. CAK is also a component of the general transcription factor, TFIIH. CDK7-mediated phosphorylation of RNA polymerase II (Pol II) at active gene promoters permits transcription. These findings identify CDK7 as a cancer therapeutic target, and several recent publications report selective CDK7 inhibitors (CDK7i) with activity against diverse cancer types. Preclin. studies have shown that CDK7i cause cell cycle arrest, apoptosis and repression of transcription, particularly of super-enhancer-assocd. genes in cancer, and have demonstrated their potential for overcoming resistance to cancer treatments. Moreover, combinations of CDK7i with other targeted cancer therapies, including BET inhibitors, BCL2 inhibitors and hormone therapies, have shown efficacy in model systems. Four CDK7i, ICEC0942 (CT7001), SY-1365, SY-5609 and LY3405105, have now progressed to Phase I/II clin. trials. Here we describe the work that has led to the development of selective CDK7i, the current status of the most advanced clin. candidates, and discuss their potential importance as cancer therapeutics, both as monotherapies and in combination settings. ClinicalTrials.gov Identifiers: NCT03363893; NCT03134638; NCT04247126; NCT03770494.
- 3Diab, S.; Yu, M.; Wang, S. CDK7 Inhibitors in cancer therapy: the sweet smell of success?. J. Med. Chem. 2020, 63, 7458– 7474, DOI: 10.1021/acs.jmedchem.9b019853https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksFaitLg%253D&md5=bdd5744330515e094612de7153d8cf36CDK7 Inhibitors in Cancer Therapy: The Sweet Smell of Success?Diab, Sarah; Yu, Mingfeng; Wang, ShudongJournal of Medicinal Chemistry (2020), 63 (14), 7458-7474CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Cyclin-dependent kinase (CDK) 7 has a unique functional repertoire by virtue of its dual role in transcription and cell cycle progression. Whereas CDK7 is ubiquitously expressed in various types of cancer, its downregulation leads to reduced cell proliferation. Importantly, it is now agreed that targeting transcription selectively limits the synthesis of mRNAs involved in tumor growth without causing an outage of transcription of housekeeping genes. Thus, CDK7 has been considered as a viable therapeutic target in cancer. Indeed, the development of CDK7 inhibitors has gained huge momentum with two mols., CT7001 and SY-1365, currently under clin. development. Herein, we discuss the latest understanding of the role of CDK7 in cancer cells and provide an overview of the pharmacophores of CDK7 inhibitors, their efficacy in various cancer models, and their clin. development.
- 4Sanchez-Martinez, C.; Lallena, M. J.; Sanfeliciano, S. G.; de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015–2019). Bioorg. Med. Chem. Lett. 2019, 29 (29), 126637, DOI: 10.1016/j.bmcl.2019.1266374https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslWjs73N&md5=2808e04d5c09daad462133bcf9b9cb3dCyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015-2019)Sanchez-Martinez, Concepcion; Lallena, Maria Jose; Sanfeliciano, Sonia Gutierrez; de Dios, AlfonsoBioorganic & Medicinal Chemistry Letters (2019), 29 (20), 126637CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)A review. Sustained proliferative capacity and gene dysregulation are hallmarks of cancer. In mammalian cells, cyclin-dependent kinases (CDKs) control crit. cell cycle checkpoints and key transcriptional events in response to extracellular and intracellular signals leading to proliferation. Significant clin. activity for the treatment of hormone receptor pos. metastatic breast cancer has been demonstrated by palbociclib, ribociclib and abemaciclib, dual CDK4/6 inhibitors recently FDA-approved. SY-1365, a CDK7 inhibitor has shown initial encouraging data in phase I for solid tumors treatment. These results have rejuvenated the CDKs research field. This review provides an overview of relevant advances on CDK inhibitor research since 2015 to 2019, with special emphasis on transcriptional CDK inhibitors, new emerging strategies such as target protein degrdn. and compds. under clin. evaluation.
- 5Larochelle, S.; Amat, R.; Glover-Cutter, K.; Sanso, M.; Zhang, C.; Allen, J. J.; Shokat, K. M.; Bentley, D. L.; Fisher, R. P. Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II. Nat. Struct. Mol. Biol. 2012, 19, 1108– 1115, DOI: 10.1038/nsmb.23995https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsV2rsLvI&md5=e483e572df1f8645db9da40800dffe4dCyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase IILarochelle, Stephane; Amat, Ramon; Glover-Cutter, Kira; Sanso, Miriam; Zhang, Chao; Allen, Jasmina J.; Shokat, Kevan M.; Bentley, David L.; Fisher, Robert P.Nature Structural & Molecular Biology (2012), 19 (11), 1108-1115CODEN: NSMBCU; ISSN:1545-9993. (Nature Publishing Group)Promoter-proximal pausing by RNA polymerase II (Pol II) ensures gene-specific regulation and RNA quality control. Structural considerations suggested a requirement for initiation-factor eviction in elongation-factor engagement and pausing of transcription complexes. Here we show that selective inhibition of Cdk7-part of TFIIH-increases TFIIE retention, prevents DRB sensitivity-inducing factor (DSIF) recruitment and attenuates pausing in human cells. Pause release depends on Cdk9-cyclin T1 (P-TEFb); Cdk7 is also required for Cdk9-activating phosphorylation and Cdk9-dependent downstream events-Pol II C-terminal domain Ser2 phosphorylation and histone H2B ubiquitylation-in vivo. Cdk7 inhibition, moreover, impairs Pol II transcript 3'-end formation. Cdk7 thus acts through TFIIE and DSIF to establish, and through P-TEFb to relieve, barriers to elongation: incoherent feedforward that might create a window to recruit RNA-processing machinery. Therefore, cyclin-dependent kinases govern Pol II handoff from initiation to elongation factors and cotranscriptional RNA maturation.
- 6Rimel, J. K.; Poss, Z. C.; Erickson, B.; Maas, Z. L.; Ebmeier, C. C.; Johnson, J. L.; Decker, T. M.; Yaron, T. M.; Bradley, M. J.; Hamman, K. B.; Hu, S.; Malojcic, G.; Marineau, J. J.; White, P. W.; Brault, M.; Tao, L.; DeRoy, P.; Clavette, C.; Nayak, S.; Damon, L. J.; Kaltheuner, I. H.; Bunch, H.; Cantley, L. C.; Geyer, M.; Iwasa, J.; Dowell, R. D.; Bentley, D. L.; Old, W. M.; Taatjes, D. J. Selective inhibition of CDK7 reveals high-confidence targets and new models for TFIIH function in transcription. Genes Dev. 2020, 34, 1452– 1473, DOI: 10.1101/gad.341545.1206https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisV2rt7zF&md5=5f6db2effae88caabdfc963fb2ab3e19Selective inhibition of CDK7 reveals high-confidence targets and new models for TFIIH function in transcriptionRimel, Jenna K.; Poss, Zachary C.; Erickson, Benjamin; Maas, Zachary L.; Ebmeier, Christopher C.; Johnson, Jared L.; Decker, Tim-Michael; Yaron, Tomer M.; Bradley, Michael J.; Hamman, Kristin B.; Hu, Shanhu; Malojcic, Goran; Marineau, Jason J.; White, Peter W.; Brault, Martine; Tao, Limei; DeRoy, Patrick; Clavette, Christian; Nayak, Shraddha; Damon, Leah J.; Kaltheuner, Ines H.; Bunch, Heeyoun; Cantley, Lewis C.; Geyer, Matthias; Iwasa, Janet; Dowell, Robin D.; Bentley, David L.; Old, William M.; Taatjes, Dylan J.Genes & Development (2020), 34 (21-22), 1452-1473CODEN: GEDEEP; ISSN:0890-9369. (Cold Spring Harbor Laboratory Press)CDK7 assocs. with the 10-subunit TFIIH complex and regulates transcription by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (RNAPII). Few addnl. CDK7 substrates are known. Here, using the covalent inhibitor SY-351 and quant. phosphoproteomics, we identified CDK7 kinase substrates in human cells. Among hundreds of high-confidence targets, the vast majority are unique to CDK7 (i.e., distinct from other transcription-assocd. kinases), with a subset that suggest novel cellular functions. Transcription-assocd. factors were predominant CDK7 substrates, including SF3B1, U2AF2, and other splicing components. Accordingly, widespread and diverse splicing defects, such as alternative exon inclusion and intron retention, were characterized in CDK7-inhibited cells. Combined with biochem. assays, we establish that CDK7 directly activates other transcription-assocd. kinases CDK9, CDK12, and CDK13, invoking a "master regulator" role in transcription. We further demonstrate that TFIIH restricts CDK7 kinase function to the RNAPII CTD, whereas other substrates (e.g., SPT5 and SF3B1) are phosphorylated by the three-subunit CDK-activating kinase (CAK; CCNH, MAT1, and CDK7). These results suggest new models for CDK7 function in transcription and implicate CAK dissocn. from TFIIH as essential for kinase activation. This straightforward regulatory strategy ensures CDK7 activation is spatially and temporally linked to transcription, and may apply toward other transcription-assocd. kinases.
- 7Fisher, R. P. Cdk7: a kinase at the core of transcription and in the crosshairs of cancer drug discovery. Transcription 2019, 10, 47– 56, DOI: 10.1080/21541264.2018.15534837https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVKit7jJ&md5=b6344f44b27d777d4fb7aad3a609d087Cdk7: a kinase at the core of transcription and in the crosshairs of cancer drug discoveryFisher, Robert P.Transcription (2019), 10 (2), 47-56CODEN: TRANH2; ISSN:2154-1272. (Taylor & Francis, Inc.)A review. The transcription cycle of RNA polymerase II (Pol II) is regulated by a set of cyclin-dependent kinases (CDKs). Cdk7, assocd. with the transcription initiation factor TFIIH, is both an effector CDK that phosphorylates Pol II and other targets within the transcriptional machinery, and a CDK-activating kinase (CAK) for at least one other essential CDK involved in transcription. Recent studies have illuminated Cdk7 functions that are executed throughout the Pol II transcription cycle, from promoter clearance and promoter-proximal pausing, to co-transcriptional chromatin modification in gene bodies, to mRNA 3'-end formation and termination. Cdk7 has also emerged as a target of small-mol. inhibitors that show promise in the treatment of cancer and inflammation. The challenges now are to identify the relevant targets of Cdk7 at each step of the transcription cycle, and to understand how heightened dependence on an essential CDK emerges in cancer, and might be exploited therapeutically.
- 8Hu, S.; Marineau, J. J.; Rajagopal, N.; Hamman, K. B.; Choi, Y. J.; Schmidt, D. R.; Ke, N.; Johannessen, L.; Bradley, M. J.; Orlando, D. A.; Alnemy, S. R.; Ren, Y.; Ciblat, S.; Winter, D. K.; Kabro, A.; Sprott, K. T.; Hodgson, J. G.; Fritz, C. C.; Carulli, J. P.; di Tomaso, E.; Olson, E. R. Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res. 2019, 79, 3479– 3491, DOI: 10.1158/0008-5472.CAN-19-01198https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M7jtl2iug%253D%253D&md5=4fd24fb5eea04eb6e921f52206f69098Discovery and Characterization of SY-1365, a Selective, Covalent Inhibitor of CDK7Hu Shanhu; Marineau Jason J; Rajagopal Nisha; Hamman Kristin B; Choi Yoon Jong; Schmidt Darby R; Ke Nan; Johannessen Liv; Bradley Michael J; Orlando David A; Alnemy Sydney R; Ren Yixuan; Sprott Kevin T; Hodgson J Graeme; Fritz Christian C; Carulli John P; di Tomaso Emmanuelle; Olson Eric R; Ciblat Stephane; Winter Dana K; Kabro AnzhelikaCancer research (2019), 79 (13), 3479-3491 ISSN:.Recent studies suggest that targeting transcriptional machinery can lead to potent and selective anticancer effects in cancers dependent on high and constant expression of certain transcription factors for growth and survival. Cyclin-dependent kinase 7 (CDK7) is the catalytic subunit of the CDK-activating kinase complex. Its function is required for both cell-cycle regulation and transcriptional control of gene expression. CDK7 has recently emerged as an attractive cancer target because its inhibition leads to decreased transcript levels of oncogenic transcription factors, especially those associated with super-enhancers. Here, we describe a selective CDK7 inhibitor SY-1365, which is currently in clinical trials in populations of patients with ovarian and breast cancer (NCT03134638). In vitro, SY-1365 inhibited cell growth of many different cancer types at nanomolar concentrations. SY-1365 treatment decreased MCL1 protein levels, and cancer cells with low BCL2L1 (BCL-XL) expression were found to be more sensitive to SY-1365. Transcriptional changes in acute myeloid leukemia (AML) cell lines were distinct from those following treatment with other transcriptional inhibitors. SY-1365 demonstrated substantial antitumor effects in multiple AML xenograft models as a single agent; SY-1365-induced growth inhibition was enhanced in combination with the BCL2 inhibitor venetoclax. Antitumor activity was also observed in xenograft models of ovarian cancer, suggesting the potential for exploring SY-1365 in the clinic in both hematologic and solid tumors. Our findings support targeting CDK7 as a new approach for treating transcriptionally addicted cancers. SIGNIFICANCE: These findings demonstrate the molecular mechanism of action and potent antitumor activity of SY-1365, the first selective CDK7 inhibitor to enter clinical investigation.
- 9Hazel, P.; Kroll, S. H.; Bondke, A.; Barbazanges, M.; Patel, H.; Fuchter, M. J.; Coombes, R. C.; Ali, S.; Barrett, A. G.; Freemont, P. S. Inhibitor selectivity for cyclin-dependent kinase 7: A structural, thermodynamic, and modelling study. ChemMedChem 2017, 12, 372– 380, DOI: 10.1002/cmdc.2016005359https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXitFegsb8%253D&md5=7fb792661b58d7e3d0859c2630dccdaeInhibitor selectivity for cyclin-dependent kinase 7: A structural, thermodynamic, and modeling studyHazel, Pascale; Kroll, Sebastian H. B.; Bondke, Alexander; Barbazanges, Marion; Patel, Hetal; Fuchter, Matthew J.; Coombes, R. Charles; Ali, Simak; Barrett, Anthony G. M.; Freemont, Paul S.ChemMedChem (2017), 12 (5), 372-380CODEN: CHEMGX; ISSN:1860-7179. (Wiley-VCH Verlag GmbH & Co. KGaA)Deregulation of the cell cycle by mechanisms that lead to elevated activities of cyclin-dependent kinases (CDKs) is a feature of many human diseases, cancer in particular. Here, we identified small-mol.-wt. inhibitors that selectively inhibit CDK7, the kinase that phosphorylates cell-cycle CDKs to promote their activities. To investigate the selectivity of these inhibitors, we used a combination of structural, biophys., and modeling approaches. We detd. the crystal structures of CDK7-selective compds. ICEC 0942 and ICEC 0943 bound to CDK2, and used these to build models of inhibitor binding to CDK7. Mol. dynamics (MD) simulations of inhibitors bound to CDK2 and CDK7 generated possible models of inhibitor binding. To exptl. validate these models, we gathered isothermal titrn. calorimetry (ITC) binding data for recombinant wild-type and binding site mutants of CDK7 and CDK2. We identified specific residues of CDK7, notably Asp-155, that were involved in detg. inhibitor selectivity. The MD simulations also showed that the flexibility of the G-rich and activation loops of CDK7 is likely an important determinant of inhibitor specificity similar to CDK2.
- 10Study of XL102 as single-agent and combination therapy in subjects with solid tumors. ClinicalTrials.gov; U.S. National Library of Medicine, 2021. https://ClinicalTrials.gov/show/NCT04726332.There is no corresponding record for this reference.
- 11Lolli, G.; Lowe, E. D.; Brown, N. R.; Johnson, L. N. The crystal structure of human CDK7 and its protein recognition properties. Structure 2004, 12, 2067– 2079, DOI: 10.1016/j.str.2004.08.01311https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpsVOrs7Y%253D&md5=b090d554993e8968c64d28fabaf6a507The Crystal Structure of Human CDK7 and Its Protein Recognition PropertiesLolli, Graziano; Lowe, Edward D.; Brown, Nick R.; Johnson, Louise N.Structure (Cambridge, MA, United States) (2004), 12 (11), 2067-2079CODEN: STRUE6; ISSN:0969-2126. (Cell Press)CDK7, a member of the cyclin-dependent protein kinase family, regulates the activities of other CDKs through phosphorylation on their activation segment and hence contributes to control of the eukaryotic cell cycle. CDK7 also assists in the regulation of transcription as part of the transcription factor TFIIH complex. For max. activity and stability, CDK7 requires phosphorylation, assocn. with cyclin H, and assocn. with a third protein, MAT1. We have detd. the crystal structure of human CDK7 in complex with ATP at 3Å resoln. The kinase is in the inactive conformation, similar to that obsd. for inactive CDK2. The activation segment is phosphorylated at Thr170 and is in a defined conformation that differs from that in phospho-CDK2 and phospho-CDK2/cyclin A. The functional properties of the enzyme against CDK2 and CTD as substrates are characterized through kinase assays. Expts. confirm that CDK7 is not a substrate for kinase-assocd. phosphatase.
- 12Finkbeiner, P.; Hehn, J. P.; Gnamm, C. Phosphine oxides from a medicinal chemist’s perspective: physicochemical and in vitro parameters relevant for drug discovery. J. Med. Chem. 2020, 63, 7081– 7107, DOI: 10.1021/acs.jmedchem.0c0040712https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVKnt7rP&md5=da34d045059662fb8fb73a17ee1dfd17Phosphine Oxides from a Medicinal Chemist's Perspective: Physicochemical and in Vitro Parameters Relevant for Drug DiscoveryFinkbeiner, Peter; Hehn, Joerg P.; Gnamm, ChristianJournal of Medicinal Chemistry (2020), 63 (13), 7081-7107CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Phosphine oxides and related phosphorus-contg. functional groups such as phosphonates and phosphinates are established structural motifs that are still underrepresented in today's drug discovery projects, and only few examples can be found among approved drugs. In this account, the physicochem. and in vitro properties of phosphine oxides and related phosphorus-contg. functional groups are reported and compared to more commonly used structural motifs in drug discovery. Furthermore, the impact on the physicochem. properties of a real drug scaffold is exemplified by a series of phosphorus-contg. analogs of imatinib. We demonstrate that phosphine oxides are highly polar functional groups leading to high soly. and metabolic stability but occasionally at the cost of reduced permeability. We conclude that phosphine oxides and related phosphorus-contg. functional groups are valuable polar structural elements and that they deserve to be considered as a routine part of every medicinal chemist's toolbox.
- 13Huang, W. S.; Liu, S.; Zou, D.; Thomas, M.; Wang, Y.; Zhou, T.; Romero, J.; Kohlmann, A.; Li, F.; Qi, J.; Cai, L.; Dwight, T. A.; Xu, Y.; Xu, R.; Dodd, R.; Toms, A.; Parillon, L.; Lu, X.; Anjum, R.; Zhang, S.; Wang, F.; Keats, J.; Wardwell, S. D.; Ning, Y.; Xu, Q.; Moran, L. E.; Mohemmad, Q. K.; Jang, H. G.; Clackson, T.; Narasimhan, N. I.; Rivera, V. M.; Zhu, X.; Dalgarno, D.; Shakespeare, W. C. Discovery of brigatinib (AP26113), a phosphine oxide-containing, potent, orally active inhibitor of anaplastic lymphoma kinase. J. Med. Chem. 2016, 59, 4948– 4964, DOI: 10.1021/acs.jmedchem.6b0030613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XntlCgur8%253D&md5=4cb93ff587579c97fda8aff0c7d43317Discovery of Brigatinib (AP26113), a Phosphine Oxide-Containing, Potent, Orally Active Inhibitor of Anaplastic Lymphoma KinaseHuang, Wei-Sheng; Liu, Shuangying; Zou, Dong; Thomas, Mathew; Wang, Yihan; Zhou, Tianjun; Romero, Jan; Kohlmann, Anna; Li, Feng; Qi, Jiwei; Cai, Lisi; Dwight, Timothy A.; Xu, Yongjin; Xu, Rongsong; Dodd, Rory; Toms, Angela; Parillon, Lois; Lu, Xiaohui; Anjum, Rana; Zhang, Sen; Wang, Frank; Keats, Jeffrey; Wardwell, Scott D.; Ning, Yaoyu; Xu, Qihong; Moran, Lauren E.; Mohemmad, Qurish K.; Jang, Hyun Gyung; Clackson, Tim; Narasimhan, Narayana I.; Rivera, Victor M.; Zhu, Xiaotian; Dalgarno, David; Shakespeare, William C.Journal of Medicinal Chemistry (2016), 59 (10), 4948-4964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)In the treatment of echinoderm microtubule-assocd. protein-like 4 (EML4)-anaplastic lymphoma kinase pos. (ALK+) non-small-cell lung cancer (NSCLC), secondary mutations within the ALK kinase domain have emerged as a major resistance mechanism to both first- and second-generation ALK inhibitors. This report describes the design and synthesis of a series of 2,4-diarylaminopyrimidine-based potent and selective ALK inhibitors culminating in identification of the investigational clin. candidate brigatinib. A unique structural feature of brigatinib is a phosphine oxide, an overlooked but novel hydrogen-bond acceptor that drives potency and selectivity in addn. to favorable ADME properties. Brigatinib displayed low nanomolar IC50s against native ALK and all tested clin. relevant ALK mutants in both enzyme-based biochem. and cell-based viability assays and demonstrated efficacy in multiple ALK+ xenografts in mice, including Karpas-299 (anaplastic large-cell lymphomas [ALCL]) and H3122 (NSCLC). Brigatinib represents the most clin. advanced phosphine oxide-contg. drug candidate to date and is currently being evaluated in a global phase 2 registration trial.
- 14Bradley, M.; Ciblat, S.; Kabro, A.; Marineau, J. J.; Chuaqui, C. Inhibitors of cyclin-dependent kinase 7 (CDK7). Patent WO2020093011A1, 2020.There is no corresponding record for this reference.
- 15Marineau, J. J.; Chuaqui, C.; Ciblat, S.; Kabro, A.; Piras, H.; Whitmore, K. M.; Lund, K.-L. Preparation of substituted aminopyrimidines as inhibitors of cyclin-dependent kinase 7 (CDK7). Patent WO2019143719A1, 2019.There is no corresponding record for this reference.
- 16Marineau, J. J.; Zahler, R.; Ciblat, S.; Winter, D. K.; Kabro, A.; Roy, S.; Schmidt, D.; Chuaqui, C.; Malojcic, G.; Piras, H.; Whitmore, K. M.; Lund, K.-I.; Sinko, B.; Sprott, K. Preparation of inhibitors of cyclin dependent kinase 7 (CDK7). Patent WO2018013867A1, 2018.There is no corresponding record for this reference.
- 17Panday, S. K.; Langlois, N. Enantioselective synthesis of (S)-5-aminopiperidin-2-one from (S)-pyroglutaminol. Tetrahedron Lett. 1995, 36, 8205– 8208, DOI: 10.1016/00404-0399(50)17557-17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXptFeitb0%253D&md5=000b9e9296f97404f8f6745698c351e4Enantioselective synthesis of (S)-5-aminopiperidin-2-one from (S)-pyroglutaminolPanday, Sharad Kumar; Langlois, NicoleTetrahedron Letters (1995), 36 (45), 8205-8CODEN: TELEAY; ISSN:0040-4039. (Elsevier)(5S)-5-aminopiperidin-2-one and several derivs. were synthesized from (S)-pyroglutaminol through ring opening and Mitsunobu reaction as the key steps.
- 18Greber, B. J.; Perez-Bertoldi, J. M.; Lim, K.; Iavarone, A. T.; Toso, D. B.; Nogales, E. The cryoelectron microscopy structure of the human CDK-activating kinase. Proc. Natl. Acad. Sci. U. S. A. 2020, 117, 22849– 22857, DOI: 10.1073/pnas.200962711718https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVCjurbJ&md5=ef7d1df2ddb5e988fe89507a4f3cd902The cryoelectron microscopy structure of the human CDK-activating kinaseGreber, Basil J.; Perez-Bertoldi, Juan M.; Lim, Kif; Iavarone, Anthony T.; Toso, Daniel B.; Nogales, EvaProceedings of the National Academy of Sciences of the United States of America (2020), 117 (37), 22849-22857CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The human CDK-activating kinase (CAK), a complex composed of cyclin-dependent kinase (CDK) 7, cyclin H, and MAT1, is a crit. regulator of transcription initiation and the cell cycle. It acts by phosphorylating the C-terminal heptapeptide repeat domain of the RNA polymerase II (Pol II) subunit RPB1, which is an important regulatory event in transcription initiation by Pol II, and it phosphorylates the regulatory T-loop of CDKs that control cell cycle progression. Here, the authors detd. the three-dimensional (3D) structure of the catalytic module of human CAK, revealing the structural basis of its assembly and providing insight into CDK7 activation in this context. The unique third component of the complex, MAT1, substantially extends the interaction interface between CDK7 and cyclin H, explaining its role as a CAK assembly factor, and it forms interactions with the CDK7 T-loop, which may contribute to enhancing CAK activity. The authors also detd. the structure of the CAK in complex with the covalently bound inhibitor THZ1 to provide insight into the binding of inhibitors at the CDK7 active site and to aid in the rational design of therapeutic compds.
- 19Greber, B. J.; Remis, J.; Ali, S.; Nogales, E. 2.5 A-resolution structure of human CDK-activating kinase bound to the clinical inhibitor ICEC0942. Biophys. J. 2021, 120, 677– 686, DOI: 10.1016/j.bpj.2020.12.03019https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjtVSrs7s%253D&md5=16cffe07b3fe538abaa36eb66202406eResolution structure 2.5 Å-of human CDK-activating kinase bound to the clinical inhibitor ICEC0942Greber, Basil J.; Remis, Jonathan; Ali, Simak; Nogales, EvaBiophysical Journal (2021), 120 (4), 677-686CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)The human CDK-activating kinase (CAK), composed of CDK7, cyclin H, and MAT1, is involved in the control of transcription initiation and the cell cycle. Because of these activities, it has been identified as a promising target for cancer chemotherapy. A no. of CDK7 inhibitors have entered clin. trials, among them ICEC0942 (also known as CT7001). Structural information can aid in improving the affinity and specificity of such drugs or drug candidates, reducing side effects in patients. Here, we have detd. the structure of the human CAK in complex with ICEC0942 at 2.5 Å-resoln. using cryogenic electron microscopy. Our structure reveals conformational differences of ICEC0942 compared with previous X-ray crystal structures of the CDK2-bound complex, and highlights the crit. ability of cryogenic electron microscopy to resolve structures of drug-bound protein complexes without the need to crystalize the protein target.
- 20Kwiatkowski, N.; Zhang, T.; Rahl, P. B.; Abraham, B. J.; Reddy, J.; Ficarro, S. B.; Dastur, A.; Amzallag, A.; Ramaswamy, S.; Tesar, B.; Jenkins, C. E.; Hannett, N. M.; McMillin, D.; Sanda, T.; Sim, T.; Kim, N. D.; Look, T.; Mitsiades, C. S.; Weng, A. P.; Brown, J. R.; Benes, C. H.; Marto, J. A.; Young, R. A.; Gray, N. S. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014, 511, 616– 620, DOI: 10.1038/nature1339320https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1ChurnF&md5=895a28fb536663a31e7558fe79ce628bTargeting transcription regulation in cancer with a covalent CDK7 inhibitorKwiatkowski, Nicholas; Zhang, Tinghu; Rahl, Peter B.; Abraham, Brian J.; Reddy, Jessica; Ficarro, Scott B.; Dastur, Anahita; Amzallag, Arnaud; Ramaswamy, Sridhar; Tesar, Bethany; Jenkins, Catherine E.; Hannett, Nancy M.; McMillin, Douglas; Sanda, Takaomi; Sim, Taebo; Kim, Nam Doo; Look, Thomas; Mitsiades, Constantine S.; Weng, Andrew P.; Brown, Jennifer R.; Benes, Cyril H.; Marto, Jarrod A.; Young, Richard A.; Gray, Nathanael S.Nature (London, United Kingdom) (2014), 511 (7511), 616-620CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Tumor oncogenes include transcription factors that co-opt the general transcriptional machinery to sustain the oncogenic state, but direct pharmacol. inhibition of transcription factors has so far proven difficult. However, the transcriptional machinery contains various enzymic cofactors that can be targeted for the development of new therapeutic candidates, including cyclin-dependent kinases (CDKs). Here the authors present the discovery and characterization of a covalent CDK7 inhibitor, THZ1, which has the unprecedented ability to target a remote cysteine residue located outside of the canonical kinase domain, providing an unanticipated means of achieving selectivity for CDK7. Cancer cell-line profiling indicates that a subset of cancer cell lines, including human T-cell acute lymphoblastic leukemia (T-ALL), have exceptional sensitivity to THZ1. Genome-wide anal. in Jurkat T-ALL cells shows that THZ1 disproportionally affects transcription of RUNX1 and suggests that sensitivity to THZ1 may be due to vulnerability conferred by the RUNX1 super-enhancer and the key role of RUNX1 in the core transcriptional regulatory circuitry of these tumor cells. Pharmacol. modulation of CDK7 kinase activity may thus provide an approach to identify and treat tumor types that are dependent on transcription for maintenance of the oncogenic state.
- 21Schrödinger Release 2020–2; Schrödinger, L.: New York, NY, 2020.There is no corresponding record for this reference.
- 22Kyte, J.; Doolittle, R. F. A simple method for displaying the hydropathic character of a protein. J. Mol. Biol. 1982, 157, 105– 132, DOI: 10.1016/0022-2836(82)90515-022https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL38Xks1yjtro%253D&md5=ee67eb115939dfe56b2b2cae2c32dbd3A simple method for displaying the hydropathic character of a proteinKyte, Jack; Doolittle, Russell F.Journal of Molecular Biology (1982), 157 (1), 105-32CODEN: JMOBAK; ISSN:0022-2836.A computer program that progressively evaluates the hydrophilicity and hydrophobicity of a protein along its amino acid sequence was devised. A hydropathy scale takes into consideration the hydrophilic and hydrophobic properties of each of the 20 amino acid side chains. Correlation was demonstrated between the plotted values and known structures detd. by crystallog.
- 23Zamyatnin, A. A. Protein volume in solution. Prog. Biophys. Mol. Biol. 1972, 24, 107– 123, DOI: 10.1016/0079-6107(72)90005-323https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE3s%252Fnt1Gluw%253D%253D&md5=1eb0435b9ebb74a00ff9517a292cd5c5Protein volume in solutionZamyatnin A AProgress in biophysics and molecular biology (1972), 24 (), 107-23 ISSN:0079-6107.There is no expanded citation for this reference.
- 24Eid, S.; Turk, S.; Volkamer, A.; Rippmann, F.; Fulle, S. KinMap: a web-based tool for interactive navigation through human kinome data. BMC Bioinf. 2017, 18, 16– 21, DOI: 10.1186/s12859-016-1433-724https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjsFCjtLc%253D&md5=29737c7cda8d9caf75018e0d29e9a459KinMap: a web-based tool for interactive navigation through human kinome dataEid, Sameh; Turk, Samo; Volkamer, Andrea; Rippmann, Friedrich; Fulle, SimoneBMC Bioinformatics (2017), 18 (), 16/1-16/6CODEN: BBMIC4; ISSN:1471-2105. (BioMed Central Ltd.)Background: Annotations of the phylogenetic tree of the human kinome is an intuitive way to visualize compd. profiling data, structural features of kinases or functional relationships within this important class of proteins. The increasing vol. and complexity of kinase-related data underlines the need for a tool that enables complex queries pertaining to kinase disease involvement and potential therapeutic uses of kinase inhibitors. Results: Here, we present KinMap, a user-friendly online tool that facilitates the interactive navigation through kinase knowledge by linking biochem., structural, and disease assocn. data to the human kinome tree. To this end, preprocessed data from freely-available sources, such as ChEMBL, the Protein Data Bank, and the Center for Therapeutic Target Validation platform are integrated into KinMap and can easily be complemented by proprietary data. The value of KinMap will be exemplarily demonstrated for uncovering new therapeutic indications of known kinase inhibitors and for prioritizing kinases for drug development efforts. Conclusions: KinMap represents a new generation of kinome tree viewers which facilitates interactive exploration of the human kinome. KinMap enables generation of high-quality annotated images of the human kinome tree as well as exchange of kinome-related data in scientific communications. Furthermore, KinMap supports multiple input and output formats and recognizes alternative kinase names and links them to a unified naming scheme, which makes it a useful tool across different disciplines and applications. A web-service of KinMap is freely available at http://www.kinhub.org/kinmap/.
- 25Wang, Y.; Zhang, T.; Kwiatkowski, N.; Abraham, B. J.; Lee, T. I.; Xie, S.; Yuzugullu, H.; Von, T.; Li, H.; Lin, Z.; Stover, D. G.; Lim, E.; Wang, Z. C.; Iglehart, J. D.; Young, R. A.; Gray, N. S.; Zhao, J. J. CDK7-dependent transcriptional addiction in triple-negative breast cancer. Cell 2015, 163, 174– 86, DOI: 10.1016/j.cell.2015.08.06325https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFKqtrjP&md5=34441125e931816d29e419f3f7dc0346CDK7-dependent transcriptional addiction in triple-negative breast cancerWang, Yubao; Zhang, Tinghu; Kwiatkowski, Nicholas; Abraham, Brian J.; Lee, Tong Ihn; Xie, Shaozhen; Yuzugullu, Haluk; Von, Thanh; Li, Heyuan; Lin, Ziao; Stover, Daniel G.; Lim, Elgene; Wang, Zhigang C.; Iglehart, J. Dirk; Young, Richard A.; Gray, Nathanael S.; Zhao, Jean J.Cell (Cambridge, MA, United States) (2015), 163 (1), 174-186CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Triple-neg. breast cancer (TNBC) is a highly aggressive form of breast cancer that exhibits extremely high levels of genetic complexity and yet a relatively uniform transcriptional program. We postulate that TNBC might be highly dependent on uninterrupted transcription of a key set of genes within this gene expression program and might therefore be exceptionally sensitive to inhibitors of transcription. Utilizing kinase inhibitors and CRISPR/Cas9-mediated gene editing, we show here that triple-neg. but not hormone receptor-pos. breast cancer cells are exceptionally dependent on CDK7, a transcriptional cyclin-dependent kinase. TNBC cells are unique in their dependence on this transcriptional CDK and suffer apoptotic cell death upon CDK7 inhibition. An "Achilles cluster" of TNBC-specific genes is esp. sensitive to CDK7 inhibition and frequently assocd. with super-enhancers. We conclude that CDK7 mediates transcriptional addiction to a vital cluster of genes in TNBC and CDK7 inhibition may be a useful therapy for this challenging cancer.
- 26Zhang, Z.; Peng, H.; Wang, X.; Yin, X.; Ma, P.; Jing, Y.; Cai, M. C.; Liu, J.; Zhang, M.; Zhang, S.; Shi, K.; Gao, W. Q.; Di, W.; Zhuang, G. Preclinical efficacy and molecular mechanism of targeting CDK7-dependent transcriptional addiction in ovarian cancer. Mol. Cancer Ther. 2017, 16, 1739– 1750, DOI: 10.1158/1535-7163.MCT-17-007826https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVCitrzL&md5=49a9f8b77dd02150dabd4fdcfe16622bPreclinical Efficacy and Molecular Mechanism of Targeting CDK7-Dependent Transcriptional Addiction in Ovarian CancerZhang, Zhenfeng; Peng, Huixin; Wang, Xiaojie; Yin, Xia; Ma, Pengfei; Jing, Ying; Cai, Mei-Chun; Liu, Jin; Zhang, Meiying; Zhang, Shengzhe; Shi, Kaixuan; Gao, Wei-Qiang; Di, Wen; Zhuang, GuangleiMolecular Cancer Therapeutics (2017), 16 (9), 1739-1750CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Ovarian cancer remains a significant cause of gynecol. cancer mortality, and novel therapeutic strategies are urgently needed in clinic as new treatment options. We previously showed that BET bromodomain inhibitors displayed promising efficacy for the treatment of epithelial ovarian cancer by downregulating pivot transcription factors. However, the potential antitumor activities and mol. mechanisms of other epigenetic or transcriptional therapies have not been systematically detd. Here, by performing an unbiased high-throughput drug screen to identify candidate compds. with antineoplastic effects, we identified THZ1, a recently developed covalent CDK7 inhibitor, as a new transcription-targeting compd. that exerted broad cytotoxicity against ovarian tumors. Mechanistically, CDK7 represented a previously unappreciated actionable vulnerability in ovarian cancer, and CDK7 inhibition led to a pronounced dysregulation of gene transcription, with a preferential repression of E2F-regulated genes and transcripts assocd. with super-enhancers. Our findings revealed the mol. underpinnings of THZ1 potency and established pharmaceutically targeting transcriptional addiction as a promising therapeutic strategy in aggressive ovarian cancer. Mol Cancer Ther; 16(9); 1739-50. ©2017 AACR.
- 27Holmes, I. P.; Bergman, Y.; Lunniss, G. E.; Nikac, M.; Choi, N.; Hemley, C. F.; Walker, S. R.; Foitzik, R. C.; Ganame, D.; Lessene, R. Preparation of pyrimidine derivatives useful as FAK inhibitors. U.S. Patent US20130017194A1, 2013.There is no corresponding record for this reference.
- 28Albers, R.; Ayala, L.; Clareen, S. S.; Delgado Mederos, M. M.; Hilgraf, R.; Hedge, S.; Hughes, K.; Kois, A.; Plantevin-Krenitsky, V.; McCarrick, M.; Nadolny, L.; Palanki, M.; Sahasrabudhe, K.; Sapienza, J.; Satoh, Y.; Sloss, M.; Sudbeck, E.; Wright, J. Preparation of haloaryl substituted aminopurines for use as a prodrug in the treatment of cancers, cardiovascular or renal diseases. Patent WO2006076595A1, 2006.There is no corresponding record for this reference.
- 29Lovitt, B.; Vanderporten, E. C.; Sheng, Z.; Zhu, H.; Drummond, J.; Liu, Y. Differential effects of divalent manganese and magnesium on the kinase activity of leucine-rich repeat kinase 2 (LRRK2). Biochemistry 2010, 49, 3092– 3100, DOI: 10.1021/bi901726c29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1Ohsrg%253D&md5=022788aecea44e220ad93ec48af21ae3Differential Effects of Divalent Manganese and Magnesium on the Kinase Activity of Leucine-Rich Repeat Kinase 2 (LRRK2)Lovitt, Brian; Vander Porten, Erica C.; Sheng, Zejuan; Zhu, Haitao; Drummond, Jake; Liu, YichinBiochemistry (2010), 49 (14), 3092-3100CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Various mutations in leucine-rich repeat kinase 2 (LRRK2) have been linked to susceptibility for both familial and idiopathic late-onset Parkinson's disease (PD). In this study, it was demonstrated that phosphorylation of MBP and LRRKtide by the LRRK2 G2019S mutant was activated by Mn2+ in vitro. This enhanced G2019S kinase activity was due to the combination of an increase in kinase and a decrease in ATPase activity by Mn2+. Compared to 10 mM Mg2+, 1 mM Mn2+ reduced ATP Km for G2019S from 103 to 1.8 μM and only modestly reduced kcat (2.5-fold); as a result, the Mn2+ increased its kcat/Km by 22-fold. This change in ATP Km was due in large part to an increase in nucleotide affinity. While Mn2+ also increased ATP affinity and had similar effects on kcat/Km for LRRK2 WT and R1441C enzymes, it reduced their kcat values significantly by 13-17-fold. Consequently, the difference in the kinase activity between G2019S and other LRRK2 variants was enhanced from about 2-fold in Mg2+ to 10-fold in Mn2+ at satg. ATP concns. relative to its Km. Furthermore, while Mg2+ yielded optimal Vmax values at Mg2+ concn. greater than 5 mM, the optimal Mn2+ concn. for activating LRRK2 catalysis was in the micromolar range with increasing Mn2+ above 1 mM causing a decrease in enzyme activity. Finally, despite the large but expected differences in IC50 tested at 100 μM ATP, the apparent Ki values of a small set of LRRK2 ATP-competitive inhibitors were within 5-fold between Mg2+- and Mn2+-mediated reactions except AMP-CPP, an ATP analog.
- 30Haq, N.; Niu, D.; Petter, R. C.; Qiao, L.; Singh, J.; Zhu, Z. Pyrimidine derivatives as ERK inhibitors and their preparation. Patent WO2014124230A2, 2014.There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c01171.
Additional figures illustrating dihedral angles from molecular dynamics simulations, CDK selectivity of SY-5102 and SY-5609, kinase selectivity of SY-5102 and SY-5609, cellular antiproliferation panel for SY-5609, quantification of immunoblotting from Figure 6, immunoblot images from Figure 8, small molecule X-ray structure of SY-5609, protein cocrystal structure of Compound 4 with CDK2, NMR spectra and LC–MS chromatograms for all compounds (PDF)
Molecular formula strings (CSV)
PDB ID Codes: Compound 4 with CDK2 (PDB: 7RA5)
CCDC 2093192 contains the supplementary crystallographic data for this paper. These data can be obtained free of charge via www.ccdc.cam.ac.uk/data_request/cif, or by emailing [email protected], or by contacting The Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, UK; fax: +44 1223 336033.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.