101
|
Insights on Structural Characteristics and Ligand Binding Mechanisms of CDK2. Int J Mol Sci 2015; 16:9314-40. [PMID: 25918937 PMCID: PMC4463590 DOI: 10.3390/ijms16059314] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/01/2015] [Accepted: 04/15/2015] [Indexed: 12/20/2022] Open
Abstract
Cyclin-dependent kinase 2 (CDK2) is a crucial regulator of the eukaryotic cell cycle. However it is well established that monomeric CDK2 lacks regulatory activity, which needs to be aroused by its positive regulators, cyclins E and A, or be phosphorylated on the catalytic segment. Interestingly, these activation steps bring some dynamic changes on the 3D-structure of the kinase, especially the activation segment. Until now, in the monomeric CDK2 structure, three binding sites have been reported, including the adenosine triphosphate (ATP) binding site (Site I) and two non-competitive binding sites (Site II and III). In addition, when the kinase is subjected to the cyclin binding process, the resulting structural changes give rise to a variation of the ATP binding site, thus generating an allosteric binding site (Site IV). All the four sites are demonstrated as being targeted by corresponding inhibitors, as is illustrated by the allosteric binding one which is targeted by inhibitor ANS (fluorophore 8-anilino-1-naphthalene sulfonate). In the present work, the binding mechanisms and their fluctuations during the activation process attract our attention. Therefore, we carry out corresponding studies on the structural characterization of CDK2, which are expected to facilitate the understanding of the molecular mechanisms of kinase proteins. Besides, the binding mechanisms of CDK2 with its relevant inhibitors, as well as the changes of binding mechanisms following conformational variations of CDK2, are summarized and compared. The summary of the conformational characteristics and ligand binding mechanisms of CDK2 in the present work will improve our understanding of the molecular mechanisms regulating the bioactivities of CDK2.
Collapse
|
102
|
He X, Xiang H, Zong X, Yan X, Yu Y, Liu G, Zou D, Yang H. CDK2-AP1 inhibits growth of breast cancer cells by regulating cell cycle and increasing docetaxel sensitivity in vivo and in vitro. Cancer Cell Int 2014; 14:130. [PMID: 25550687 PMCID: PMC4279590 DOI: 10.1186/s12935-014-0130-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 11/17/2014] [Indexed: 11/10/2022] Open
Abstract
Background Cell cycle regulatory pathway is a well-established pathway mainly dependent on cyclin-dependent kinases (CDKs), which are regulated positively by cyclins and negatively by cyclin-dependent kinase inhibitors(CKIs). Cyclin-dependent kinase 2 associate protein 1(CDK2-AP1) is a specific negative regulatory protein for CDK2, is important in the cancer cell cycle. However, the function of CDK2-AP1 in breast cancer remains unclear. We designed therefore explored the effects of CDK2-AP1 on breast cancer growth and its chemo-sensitivity. Methods Expression of CDK2-AP1, CDK2 and CyclinD1 in 209 cases of pathological specimens using IHC staining was measured. Lost-of-function and Gain-of-function assays were used in vivo and in vitro relating to the specific role of CDK2-AP1 in breast cancer. We analyzed in vivo and in vitro the impact of CDK2-AP1 on chemotherapy sensitivity in breast cancer. Results The positive ratio of CDK2-AP1 expression was reduced successively in normal breast tissue, DCIS, invasive breast cancer and relapsed breast cancer, however, with CDK2 and CyclinD1 it was suggested that CDK2-AP1 was correlated closely with the tumorigenesis and progress, and might work as a tumor suppressor. After down-regulating CDK2-AP1 in breast cancer cells, the cell cycle was accelerated and cell proliferation enhanced. The cell cycle was arrested in G0/G1 phase and G2/M phase after up-regulating CDK2-AP1 in breast cancer cells, inhibiting cell proliferation. The expression of CDK2 and CyclinD1 changed accordingly after downregulation or upregulation of CDK2-AP1 by western blot, suggesting a role of the CDK2-AP1/CDK2/CyclinD1 cell cycle pathway in the initiation and progression of breast cancer. Similar results were obtained in animal assays. The data indicates that CDK2-AP1 can induce sensitivity to docetaxel treatment in breast cancer cells. Conclusions CDK2-AP1 affects tumorigenesis, tumor growth and chemo-sensitivity by cell cycle regulation, which can potentially to be a therapeutical agent in breast cancer.
Collapse
Affiliation(s)
- Xiangming He
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Hua Xiang
- Department of Pathology, First Affiliated Hospital of College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 China
| | - Xiangyun Zong
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China ; Department of Breast Surgery, Shanghai Jiao Tong University affiliated Shanghai sixth Hospital, 600 Yishan Road, Shanghai, 200233 China
| | - Xuebing Yan
- Department of Breast Surgery, Shanghai Jiao Tong University affiliated Shanghai sixth Hospital, 600 Yishan Road, Shanghai, 200233 China
| | - Yang Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Guan Liu
- Department of Radiotherapy, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Dehong Zou
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| | - Hongjian Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022 China
| |
Collapse
|
103
|
Chipumuro E, Marco E, Christensen CL, Kwiatkowski N, Zhang T, Hatheway CM, Abraham BJ, Sharma B, Yeung C, Altabef A, Perez-Atayde A, Wong KK, Yuan GC, Gray NS, Young RA, George RE. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 2014; 159:1126-1139. [PMID: 25416950 DOI: 10.1016/j.cell.2014.10.024] [Citation(s) in RCA: 465] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/18/2014] [Accepted: 09/24/2014] [Indexed: 02/06/2023]
Abstract
The MYC oncoproteins are thought to stimulate tumor cell growth and proliferation through amplification of gene transcription, a mechanism that has thwarted most efforts to inhibit MYC function as potential cancer therapy. Using a covalent inhibitor of cyclin-dependent kinase 7 (CDK7) to disrupt the transcription of amplified MYCN in neuroblastoma cells, we demonstrate downregulation of the oncoprotein with consequent massive suppression of MYCN-driven global transcriptional amplification. This response translated to significant tumor regression in a mouse model of high-risk neuroblastoma, without the introduction of systemic toxicity. The striking treatment selectivity of MYCN-overexpressing cells correlated with preferential downregulation of super-enhancer-associated genes, including MYCN and other known oncogenic drivers in neuroblastoma. These results indicate that CDK7 inhibition, by selectively targeting the mechanisms that promote global transcriptional amplification in tumor cells, may be useful therapy for cancers that are driven by MYC family oncoproteins.
Collapse
Affiliation(s)
- Edmond Chipumuro
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Eugenio Marco
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard School of Public Health, Boston, MA 02115, USA
| | | | - Nicholas Kwiatkowski
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Clark M Hatheway
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA
| | - Brian J Abraham
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bandana Sharma
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA
| | - Caleb Yeung
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abigail Altabef
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard School of Public Health, Boston, MA 02115, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Richard A Young
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rani E George
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA 02215, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
104
|
Selective inhibition of CDK7 ameliorates experimental arthritis in mice. Clin Exp Med 2014; 15:269-75. [DOI: 10.1007/s10238-014-0305-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/28/2014] [Indexed: 12/24/2022]
|
105
|
Yik JHN, Hu Z, Kumari R, Christiansen BA, Haudenschild DR. Cyclin-dependent kinase 9 inhibition protects cartilage from the catabolic effects of proinflammatory cytokines. Arthritis Rheumatol 2014; 66:1537-46. [PMID: 24470357 DOI: 10.1002/art.38378] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 01/21/2014] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Cyclin-dependent kinase 9 (CDK-9) controls the activation of primary inflammatory response genes. The purpose of this study was to determine whether CDK-9 inhibition protects cartilage from the catabolic effects of proinflammatory cytokines. METHODS Human chondrocytes were challenged with different proinflammatory stimuli (interleukin-1β [IL-1β], lipopolysaccharides, and tumor necrosis factor α) in the presence or absence of either the CDK-9 inhibitor flavopiridol or small interfering RNA (siRNA). The expression of messenger RNA (mRNA) for inflammatory mediator genes, catabolic genes, and anabolic genes were determined by real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis. Cartilage explants were incubated for 6 days with IL-1β in the presence or absence of flavopiridol. Cartilage matrix degradation was assessed by the release of glycosaminoglycan (GAG) and cleaved type II collagen (COL2A) peptides. RESULTS CDK-9 inhibition by flavopiridol or knockdown by siRNA effectively suppressed the induction of mRNA for inducible nitric oxide synthase by all 3 proinflammatory stimuli. Results from NF-κB-targeted PCR array analysis showed that flavopiridol suppressed IL-1β induction of a broad range of inflammatory mediator genes (59 of 67 tested). CDK-9 inhibition also suppressed the induction of catabolic genes (matrix metalloproteinase 1 [MMP-1], MMP-3, MMP-9, MMP-13, ADAMTS-4, and ADAMTS-5), but did not affect the basal expression of anabolic genes (COL2A, aggrecan, and cartilage oligomeric matrix protein) and housekeeping genes. Flavopiridol had no apparent short-term cytotoxicity, as assessed by G6PDH activity. Finally, in IL-1β-treated cartilage explants, flavopiridol reduced the release of the matrix degradation product GAG and cleaved COL2A peptides, but did not affect long-term chondrocyte viability. CONCLUSION CDK-9 activity is required for the primary inflammatory response in chondrocytes. Flavopiridol suppresses the induction of inflammatory mediator genes and catabolic genes to protect cartilage from the deleterious effects of proinflammatory cytokines, without affecting cell viability and functions.
Collapse
|
106
|
Abstract
Cyclin-dependent kinases (CDKs) are involved in temporal control of the cell cycle and transcription and play central roles in cancer development and metastasis. Recently, Kwiatkowski and colleagues identified a novel CDK7-specific inhibitor, THZ1, that hinders proliferation in cancer cell lines and dampens global transcription in T cell leukemia.
Collapse
Affiliation(s)
- Kaixiang Cao
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Ali Shilatifard
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA.
| |
Collapse
|
107
|
Cyclin-dependent kinase 7 controls mRNA synthesis by affecting stability of preinitiation complexes, leading to altered gene expression, cell cycle progression, and survival of tumor cells. Mol Cell Biol 2014; 34:3675-88. [PMID: 25047832 DOI: 10.1128/mcb.00595-14] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cyclin-dependent kinase 7 (CDK7) activates cell cycle CDKs and is a member of the general transcription factor TFIIH. Although there is substantial evidence for an active role of CDK7 in mRNA synthesis and associated processes, the degree of its influence on global and gene-specific transcription in mammalian species is unclear. In the current study, we utilize two novel inhibitors with high specificity for CDK7 to demonstrate a restricted but robust impact of CDK7 on gene transcription in vivo and in in vitro-reconstituted reactions. We distinguish between relative low- and high-dose responses and relate them to distinct molecular mechanisms and altered physiological responses. Low inhibitor doses cause rapid clearance of paused RNA polymerase II (RNAPII) molecules and sufficed to cause genome-wide alterations in gene expression, delays in cell cycle progression at both the G1/S and G2/M checkpoints, and diminished survival of human tumor cells. Higher doses and prolonged inhibition led to strong reductions in RNAPII carboxyl-terminal domain (CTD) phosphorylation, eventual activation of the p53 program, and increased cell death. Together, our data reason for a quantitative contribution of CDK7 to mRNA synthesis, which is critical for cellular homeostasis.
Collapse
|
108
|
Echalier A, Hole AJ, Lolli G, Endicott JA, Noble MEM. An inhibitor's-eye view of the ATP-binding site of CDKs in different regulatory states. ACS Chem Biol 2014; 9:1251-6. [PMID: 24669831 PMCID: PMC4068217 DOI: 10.1021/cb500135f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
![]()
We have used a chemically diverse
panel of kinase inhibitors to
assess the chemical similarity of the ATP-binding sites of cyclin-dependent
kinase (CDK) subfamily members in a range of activation states. Using
this approach, we find that different activation states of a particular
CDK may differ from each other as much as different CDKs in the same
activation state. We also find that inhibitors discriminate more effectively
among CDK family members in their monomeric state than in their cyclin-bound
state, providing direct evidence for the belief that selective binding
to inactive kinase states might be more readily achieved than selective
binding to active states.
Collapse
Affiliation(s)
- Aude Echalier
- Laboratory
of Molecular Biophysics, Department of Biochemistry, Oxford University, South
Parks Road, Oxford OX1
3QU, United Kingdom
| | - Alison J. Hole
- Laboratory
of Molecular Biophysics, Department of Biochemistry, Oxford University, South
Parks Road, Oxford OX1
3QU, United Kingdom
| | - Graziano Lolli
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
| | - Jane A. Endicott
- Laboratory
of Molecular Biophysics, Department of Biochemistry, Oxford University, South
Parks Road, Oxford OX1
3QU, United Kingdom
| | - Martin E. M. Noble
- Laboratory
of Molecular Biophysics, Department of Biochemistry, Oxford University, South
Parks Road, Oxford OX1
3QU, United Kingdom
| |
Collapse
|
109
|
Li Y, Gao W, Li F, Wang J, Zhang J, Yang Y, Zhang S, Yang L. An in silico exploration of the interaction mechanism of pyrazolo[1,5-a]pyrimidine type CDK2 inhibitors. MOLECULAR BIOSYSTEMS 2014; 9:2266-81. [PMID: 23864105 DOI: 10.1039/c3mb70186g] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CDK2, which interacts with cyclin A and cyclin E, is an important member of the CDK family. Having been proved to be associated with many diseases for its vital role in cell cycle, CDK2 is a promising target of anti-cancer drugs dealing with cell cycle disorders. In the present work, a total of 111 pyrazolo[1,5-a]pyrimidines (PHTPPs) as CDK2/cyclin A inhibitors were studied to conduct three-dimensional quantitative structure-activity (3D-QSAR) analyses. The optimal comparative molecular similarity indices analysis (CoMSIA) model shows that Q(2) = 0.516, Rncv(2) = 0.912, Rpre(2) = 0.914, Rm(2) = 0.843, SEP = 0.812, SEE = 0.347 with 10 components using steric, hydrophobic and H-bond donor field descriptors, indicating its effective internal and external predictive capacity. The contour maps further indicate that (1) bulky substituents in R1 are beneficial while H-bond donor groups at this position are detrimental; (2) hydrophobic contributions in the R2 area are favorable; (3) large and hydrophilic groups are well tolerated at the R3 position (a close H-bond donor moiety is favorable while a distal H-bond donor moiety in this area is disfavored); (4) bulky and hydrophobic features in the R4 region are beneficial for the biological activities and (5) the 7-N-aryl substitution is crucial to boost the inhibitory activities of the PHTPP inhibitors. Finally, docking and MD simulations demostrate that PHTPP derivatives are stabilized in a 'flying bat' conformation mainly through the H-bond interactions and hydrophobic contacts. Comparative studies indicate that PHTPP derivatives fit well within the ATP binding cleft in CDK2, with the core heterocyclic ring overlapping significantly with the adenine group of ATP despite a small deflection. In comparison to numerous other inhibitors binding to the ATP pocket, PHTPP analogues follow the binding fashion of purine inhibitors of this kinase. It is anticipated that the binding mechanism and structural features of PHTPP inhibitors studied in the present work will benefit the discovery of more potent CDK2 inhibitors, and the valid pyrazolo[1,5-a]pyrimidine-7-N-yl inhibitors will soon emerge from the large number of screening programmes to enter in clinical studies.
Collapse
Affiliation(s)
- Yan Li
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Dalian University of Technology, Dalian, 116024, Liaoning, China.
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Kaliszczak M, Patel H, Kroll SHB, Carroll L, Smith G, Delaney S, Heathcote DA, Bondke A, Fuchter MJ, Coombes RC, Barrett AGM, Ali S, Aboagye EO. Development of a cyclin-dependent kinase inhibitor devoid of ABC transporter-dependent drug resistance. Br J Cancer 2013; 109:2356-67. [PMID: 24071597 PMCID: PMC3817326 DOI: 10.1038/bjc.2013.584] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/30/2013] [Accepted: 09/04/2013] [Indexed: 01/15/2023] Open
Abstract
Background: Cyclin-dependent kinases (CDKs) control cell cycle progression, RNA transcription and apoptosis, making them attractive targets for anticancer drug development. Unfortunately, CDK inhibitors developed to date have demonstrated variable efficacy. Methods: We generated drug-resistant cells by continuous low-dose exposure to a model pyrazolo[1,5-a]pyrimidine CDK inhibitor and investigated potential structural alterations for optimal efficacy. Results: We identified induction of the ATP-binding cassette (ABC) transporters, ABCB1 and ABCG2, in resistant cells. Assessment of features involved in the ABC transporter substrate specificity from a compound library revealed high polar surface area (>100 Å2) as a key determinant of transporter interaction. We developed ICEC-0782 that preferentially inhibited CDK2, CDK7 and CDK9 in the nanomolar range. The compound inhibited phosphorylation of CDK substrates and downregulated the short-lived proteins, Mcl-1 and cyclin D1. ICEC-0782 induced G2/M arrest and apoptosis. The permeability and cytotoxicity of ICEC-0782 were unaffected by ABC transporter expression. Following daily oral dosing, the compound inhibited growth of human colon HCT-116 and human breast MCF7 tumour xenografts in vivo by 84% and 94%, respectively. Conclusion: We identified a promising pyrazolo[1,5-a]pyrimidine compound devoid of ABC transporter interaction, highly suitable for further preclinical and clinical evaluation for the treatment of cancer.
Collapse
Affiliation(s)
- M Kaliszczak
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Wu D, Gu QH, Li ZW. Cyclin-dependent kinases, control of cell cycle and hepatic fibrosis. Shijie Huaren Xiaohua Zazhi 2013; 21:2158-2163. [DOI: 10.11569/wcjd.v21.i22.2158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Multiple etiologies of liver disease lead to liver fibrosis by driving the activation of hepatic stellate cells (HSCs) into a myofibroblast-like phenotype that is contractile, proliferative and fibrogenic. Liver fibrosis is associated with the proliferation of HSCs, and the cell cycle of activated HSCs is abnormal. Cyclin-dependent kinases (CDKs) play essential roles in cell proliferation. However, the molecular mechanisms responsible for the abnormal proliferation of activated HSCs during hepatic fibrogenesis remain to be defined. Here we will review recent progress in understanding the associations among CDKs, the control of cell cycle and hepatic fibrosis, with an aim to reveal the potential mechanisms of hepatic fibrosis.
Collapse
|
112
|
Liu H, Liu K, Huang Z, Park CM, Thimmegowda NR, Jang JH, Ryoo IJ, He L, Kim SO, Oi N, Lee KW, Soung NK, Bode AM, Yang Y, Zhou X, Erikson RL, Ahn JS, Hwang J, Kim KE, Dong Z, Kim BY. A chrysin derivative suppresses skin cancer growth by inhibiting cyclin-dependent kinases. J Biol Chem 2013; 288:25924-25937. [PMID: 23888052 DOI: 10.1074/jbc.m113.464669] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chrysin (5,7-dihydroxyflavone), a natural flavonoid widely distributed in plants, reportedly has chemopreventive properties against various cancers. However, the anticancer activity of chrysin observed in in vivo studies has been disappointing. Here, we report that a chrysin derivative, referred to as compound 69407, more strongly inhibited EGF-induced neoplastic transformation of JB6 P(+) cells compared with chrysin. It attenuated cell cycle progression of EGF-stimulated cells at the G1 phase and inhibited the G1/S transition. It caused loss of retinoblastoma phosphorylation at both Ser-795 and Ser-807/811, the preferred sites phosphorylated by Cdk4/6 and Cdk2, respectively. It also suppressed anchorage-dependent and -independent growth of A431 human epidermoid carcinoma cells. Compound 69407 reduced tumor growth in the A431 mouse xenograft model and retinoblastoma phosphorylation at Ser-795 and Ser-807/811. Immunoprecipitation kinase assay results showed that compound 69407 attenuated endogenous Cdk4 and Cdk2 kinase activities in EGF-stimulated JB6 P(+) cells. Pulldown and in vitro kinase assay results indicated that compound 69407 directly binds with Cdk2 and Cdk4 in an ATP-independent manner and inhibited their kinase activities. A binding model between compound 69407 and a crystal structure of Cdk2 predicted that compound 69407 was located inside the Cdk2 allosteric binding site. The binding was further verified by a point mutation binding assay. Overall results indicated that compound 69407 is an ATP-noncompetitive cyclin-dependent kinase inhibitor with anti-tumor effects, which acts by binding inside the Cdk2 allosteric pocket. This study provides new insights for creating a general pharmacophore model to design and develop novel ATP-noncompetitive agents with chemopreventive or chemotherapeutic potency.
Collapse
Affiliation(s)
- Haidan Liu
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea,; the Hormel Institute, University of Minnesota, Austin, Minnesota 55912,; the Department of Cardiothoracic Surgery and; Clinical Center for Gene Diagnosis and Therapy, The Second Xiangya Hospital, Central South University, Renmin Road 139, Changsha, Hunan 410011, China
| | - Kangdong Liu
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea,; the Hormel Institute, University of Minnesota, Austin, Minnesota 55912,; the Basic Medical College, Zhengzhou University, ZhengZhou 450001 China, and
| | - Zunnan Huang
- the Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Chan-Mi Park
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea
| | - N R Thimmegowda
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea
| | - Jae-Hyuk Jang
- the Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Yangcheng-ri, Ochang, Cheongwon, Chungcheongbuk-do 363-883, Republic of Korea
| | - In-Ja Ryoo
- the Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Yangcheng-ri, Ochang, Cheongwon, Chungcheongbuk-do 363-883, Republic of Korea
| | - Long He
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea
| | - Sun-Ok Kim
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea
| | - Naomi Oi
- the Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Ki Won Lee
- the Department of Agricultural Biotechnology, Seoul National University, Seoul 151-921, Republic of Korea
| | - Nak-Kyun Soung
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea
| | - Ann M Bode
- the Hormel Institute, University of Minnesota, Austin, Minnesota 55912
| | - Yifeng Yang
- the Department of Cardiothoracic Surgery and
| | - Xinmin Zhou
- the Department of Cardiothoracic Surgery and
| | - Raymond L Erikson
- the Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Jong-Seog Ahn
- the Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Yangcheng-ri, Ochang, Cheongwon, Chungcheongbuk-do 363-883, Republic of Korea
| | - Joonsung Hwang
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea
| | - Kyoon Eon Kim
- the Department of Biochemistry, College of Natural Science, Chung Nam National University, Yuseong, Daejeon 305-764, Republic of Korea
| | - Zigang Dong
- the Hormel Institute, University of Minnesota, Austin, Minnesota 55912,.
| | - Bo-Yeon Kim
- From the World Class Institute, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea,.
| |
Collapse
|
113
|
Abstract
Cyclin-dependent kinases (CDKs) play essential roles in cell proliferation and gene expression. Although distinct sets of CDKs work in cell division and transcription by RNA polymerase II (Pol II), they share a CDK-activating kinase (CAK), which is itself a CDK-Cdk7-in metazoans. Thus a unitary CDK network controls and may coordinate cycles of cell division and gene expression. Recent work reveals decisive roles for Cdk7 in both pathways. The CAK function of Cdk7 helps determine timing of activation and cyclin-binding preferences of different CDKs during the cell cycle. In the transcription cycle, Cdk7 is both an effector kinase, which phosphorylates Pol II and other proteins and helps establish promoter-proximal pausing; and a CAK for Cdk9 (P-TEFb), which releases Pol II from the pause. By governing the transition from initiation to elongation, Cdk7, Cdk9 and their substrates influence expression of genes important for developmental and cell-cycle decisions, and ensure co-transcriptional maturation of Pol II transcripts. Cdk7 engaged in transcription also appears to be regulated by phosphorylation within its own activation (T) loop. Here I review recent studies of CDK regulation in cell division and gene expression, and propose a model whereby mitogenic signals trigger a cascade of CDK T-loop phosphorylation that drives cells past the restriction (R) point, when continued cell-cycle progression becomes growth factor-independent. Because R-point control is frequently deregulated in cancer, the CAK-CDK pathway is an attractive target for chemical inhibition aimed at impeding the inappropriate commitment to cell division.
Collapse
|
114
|
Wang W, Cao X, Zhu X, Gu Y. Molecular dynamic simulations give insight into the mechanism of binding between 2-aminothiazole inhibitors and CDK5. J Mol Model 2013; 19:2635-45. [PMID: 23525963 DOI: 10.1007/s00894-013-1815-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Accepted: 03/04/2013] [Indexed: 12/20/2022]
Abstract
Molecular docking, molecular dynamics (MD) simulations, and binding free energy analysis were performed to reveal differences in the binding affinities between five 2-aminothiazole inhibitors and CDK5. The hydrogen bonding and hydrophobic interactions between inhibitors and adjacent residues are analyzed and discussed. The rank of calculated binding free energies using the MM-PBSA method is consistent with experimental result. The results illustrate that hydrogen bonds with Cys83 favor inhibitor binding. The van der Waals interactions, especially the important contact with Ile10, dominate in the binding free energy and play a crucial role in distinguishing the different bioactivity of the five inhibitors.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemistry and Chemical Engineering, Nanjing University of Technology, Nanjing 210009, China
| | | | | | | |
Collapse
|
115
|
Hwang JY, Windisch MP, Jo S, Kim K, Kong S, Kim HC, Kim S, Kim H, Lee ME, Kim Y, Choi J, Park DS, Park E, Kwon J, Nam J, Ahn S, Cechetto J, Kim J, Liuzzi M, No Z, Lee J. Discovery and characterization of a novel 7-aminopyrazolo[1,5-a]pyrimidine analog as a potent hepatitis C virus inhibitor. Bioorg Med Chem Lett 2012; 22:7297-301. [PMID: 23159569 DOI: 10.1016/j.bmcl.2012.10.123] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/25/2012] [Accepted: 10/29/2012] [Indexed: 02/01/2023]
Abstract
We describe a novel 7-aminopyrazolo[1,5-a]pyrimidine (7-APP) derivative as a potent hepatitis C virus (HCV) inhibitor. A series of 7-APPs was synthesized and evaluated for inhibitory activity against HCV in different cell culture systems. The synthesis and preliminary structure-activity relationship study of 7-APP are reported.
Collapse
Affiliation(s)
- Jong Yeon Hwang
- Medicinal Chemistry 2, Institut Pasteur Korea, Sampyeong-dong 696, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Uitdehaag JCM, Verkaar F, Alwan H, de Man J, Buijsman RC, Zaman GJR. A guide to picking the most selective kinase inhibitor tool compounds for pharmacological validation of drug targets. Br J Pharmacol 2012; 166:858-76. [PMID: 22250956 DOI: 10.1111/j.1476-5381.2012.01859.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
To establish the druggability of a target, genetic validation needs to be supplemented with pharmacological validation. Pharmacological studies, especially in the kinase field, are hampered by the fact that many reference inhibitors are not fully selective for one target. Fortunately, the initial trickle of selective inhibitors released in the public domain has steadily swelled into a stream. However, rationally picking the most selective tool compound out of the increasing amounts of available inhibitors has become progressively difficult due to the lack of accurate quantitative descriptors of drug selectivity. A recently published approach, termed 'selectivity entropy', is an improved way of expressing selectivity as a single-value parameter and enables rank ordering of inhibitors. We provide a guide to select the best tool compounds for pharmacological validation experiments of candidate drug targets using selectivity entropy. In addition, we recommend which inhibitors to use for studying the biology of the 20 most investigated kinases that are clinically relevant: Abl (ABL1), AKT1, ALK, Aurora A/B, CDKs, MET, CSF1R (FMS), EGFR, FLT3, ERBB2 (HER2), IKBKB (IKK2), JAK2/3, JNK1/2/3 (MAPK8/9/10), MEK1/2, PLK1, PI3Ks, p38α (MAPK14), BRAF, SRC and VEGFR2 (KDR).
Collapse
|
117
|
Merrick KA, Fisher RP. Why minimal is not optimal: driving the mammalian cell cycle--and drug discovery--with a physiologic CDK control network. Cell Cycle 2012; 11:2600-5. [PMID: 22732498 DOI: 10.4161/cc.20758] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Progression through the eukaryotic cell division cycle is governed by the activity of cyclin-dependent kinases (CDKs). For a CDK to become active it must (1) bind a positive regulatory subunit (cyclin) and (2) be phosphorylated on its activation (T) loop. In metazoans, multiple CDK catalytic subunits, each with a distinct set of preferred cyclin partners, regulate the cell cycle, but it has been difficult to assign functions to individual CDKs in vivo. Biochemical analyses and experiments with dominant-negative alleles suggested that specific CDK/cyclin complexes regulate different events, but genetic loss of interphase CDKs (Cdk2, -4 and -6), alone or in combination, did not block proliferation of cells in culture. These knockout and knockdown studies suggested redundancy or plasticity built into the CDK network but did not address whether there was true redundancy in normal cells with a full complement of CDKs. Here, we discuss recent work that took a chemical-genetic approach to reveal that the activity of a genetically non-essential CDK, Cdk2, is required for cell proliferation when normal cyclin pairing is maintained. These results have implications for the systems-level organization of the cell cycle, for regulation of the restriction point and G 1/S transition and for efforts to target Cdk2 therapeutically in human cancers.
Collapse
Affiliation(s)
- Karl A Merrick
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
118
|
The impact of CDK inhibition in human malignancies associated with pronounced defects in apoptosis: advantages of multi-targeting small molecules. Future Med Chem 2012; 4:395-424. [DOI: 10.4155/fmc.12.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Malignant cells in chronic lymphocytic leukemia (CLL) and related diseases are heterogeneous and consist primarily of long-lived resting cells in the periphery and a minor subset of dividing cells in proliferating centers. Both cell populations have different molecular signatures that play a major role in determining their sensitivity to therapy. Contemporary approaches to treating CLL are heavily reliant on cytotoxic chemotherapeutics. However, none of the current treatment regimens can be considered curative. Pharmacological CDK inhibitors have extended the repertoire of potential drugs for CLL. Multi-targeted CDK inhibitors affect CDKs involved in regulating both cell cycle progression and transcription. Their interference with transcriptional elongation represses anti-apoptotic proteins and, thus, promotes the induction of apoptosis. Importantly, there is evidence that treatment with CDK inhibitors can overcome resistance to therapy. The pharmacological CDK inhibitors have great potential for use in combination with other therapeutics and represent promising tools for the development of new curative treatments for CLL.
Collapse
|
119
|
Tolhurst RS, Thomas RS, Kyle FJ, Patel H, Periyasamy M, Photiou A, Thiruchelvam PTR, Lai CF, Al-Sabbagh M, Fisher RA, Barry S, Crnogorac-Jurcevic T, Martin LA, Dowsett M, Charles Coombes R, Kamalati T, Ali S, Buluwela L. Transient over-expression of estrogen receptor-α in breast cancer cells promotes cell survival and estrogen-independent growth. Breast Cancer Res Treat 2011; 128:357-68. [PMID: 20730598 DOI: 10.1007/s10549-010-1122-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 08/09/2010] [Indexed: 01/30/2023]
Abstract
Estrogen receptor-α (ERα) positive breast cancer frequently responds to inhibitors of ERα activity, such as tamoxifen, and/or to aromatase inhibitors that block estrogen biosynthesis. However, many patients become resistant to these agents through mechanisms that remain unclear. Previous studies have shown that expression of ERα in ERα-negative breast cancer cell lines frequently inhibits their growth. In order to determine the consequence of ERα over-expression in ERα-positive breast cancer cells, we over-expressed ERα in the MCF-7 breast cancer cell line using adenovirus gene transduction. ERα over-expression led to ligand-independent expression of the estrogen-regulated genes pS2 and PR and growth in the absence of estrogen. Interestingly, prolonged culturing of these cells in estrogen-free conditions led to the outgrowth of cells capable of growth in cultures from ERα transduced, but not in control cultures. From these cultures a line, MLET5, was established which remained ERα-positive, but grew in an estrogen-independent manner. Moreover, MLET5 cells were inhibited by anti-estrogens showing that ERα remains important for their growth. Gene expression microarray analysis comparing MCF-7 cells with MLET5 highlighted apoptosis as a major functional grouping that is altered in MLET5 cells, such that cell survival would be favoured. This conclusion was further substantiated by the demonstration that MLET5 show resistance to etoposide-induced apoptosis. As the gene expression microarray analysis also shows that the apoptosis gene set differentially expressed in MLET5 is enriched for estrogen-regulated genes, our findings suggest that transient over-expression of ERα could lead to increased cell survival and the development of estrogen-independent growth, thereby contributing to resistance to endocrine therapies in breast cancer patients.
Collapse
Affiliation(s)
- Robert S Tolhurst
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Metal-free imidazolium salts inhibit the growth of hepatocellular carcinoma in a mouse model. J Transl Med 2011; 91:744-51. [PMID: 21339744 DOI: 10.1038/labinvest.2011.4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Imidazolium salts (IMSs) are precursors to N-heterocyclic carbenes (NHCs), which are routinely used as ligands or organo-catalysts in synthetic chemistry. We recently identified several IMSs as anti-fibrotic agents in liver fibrosis, which often has a consequence in the oncogenesis of hepatocellular carcinoma (HCC). Here, we investigate the potential anti-tumor property of three IMSs (named IBN-1, IBN-9, and DPIM) in HCC cell lines and in a xenograft mouse model. Our results showed that both IBN-1 and IBN-9 significantly inhibited the cell proliferation and arrested HCC cells in the G1-phase, whereas DPIM did not have any anti-tumor activity. When tested in a Huh7 HCC xenograft mouse model, IBN-1 reduced the tumor volume by 31% (P<0.05), however accompanied by a 9% loss in body weight (P<0.005), suggesting a general toxicity. In contrast, IBN-9 significantly reduced the tumor volume by 45% (P<0.05) and 60% (P<0.01) at doses of 0.6 and 1.5 g/l in drinking water, respectively, without any loss in body weight. Our in vitro and in vivo data suggested that IBN-1 and IBN-9 inhibited the growth of HCC by suppressing the expression of Survivin and Cyclin-dependent kinases. The current study provides a proof of concept for using the metal-free IMSs to develop novel anti-cancer agents.
Collapse
|
121
|
Liebl J, Krystof V, Vereb G, Takács L, Strnad M, Pechan P, Havlicek L, Zatloukal M, Fürst R, Vollmar AM, Zahler S. Anti-angiogenic effects of purine inhibitors of cyclin dependent kinases. Angiogenesis 2011; 14:281-91. [DOI: 10.1007/s10456-011-9212-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 03/31/2011] [Indexed: 01/23/2023]
|
122
|
Jorda R, Havlíček L, McNae IW, Walkinshaw MD, Voller J, Šturc A, Navrátilová J, Kuzma M, Mistrík M, Bártek J, Strnad M, Kryštof V. Pyrazolo[4,3-d]pyrimidine Bioisostere of Roscovitine: Evaluation of a Novel Selective Inhibitor of Cyclin-Dependent Kinases with Antiproliferative Activity. J Med Chem 2011; 54:2980-93. [DOI: 10.1021/jm200064p] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Radek Jorda
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany ASCR, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| | - Libor Havlíček
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany ASCR, Šlechtitelů 11, 78371 Olomouc, Czech Republic
- Isotope Laboratory, Institute of Experimental Botany ASCR, Videnska 1083, 142 20 Prague, Czech Republic
| | - Iain W. McNae
- Structural Biochemistry Group, University of Edinburgh, Michael Swann Building, King’s Buildings, Edinburgh, EH9 3JR, Scotland
| | - Malcolm D. Walkinshaw
- Structural Biochemistry Group, University of Edinburgh, Michael Swann Building, King’s Buildings, Edinburgh, EH9 3JR, Scotland
| | - Jiří Voller
- Centre of the Region Haná for Biotechnological and Agricultural Research, Faculty of Science, Department of Growth Regulators, Palacký University, Šlechtitelů 11, Olomouc, CZ-783 71, Czech Republic
| | - Antonín Šturc
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany ASCR, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| | - Jana Navrátilová
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany ASCR, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| | - Marek Kuzma
- Laboratory of Molecular Structure Characterization, Institute of Microbiology ASCR, Videnska 1083, 142 20 Prague, Czech Republic
| | - Martin Mistrík
- Laboratory of Genome Integrity and Institute of Molecular and Translational Medicine, Palacký University, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| | - Jiří Bártek
- Laboratory of Genome Integrity and Institute of Molecular and Translational Medicine, Palacký University, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany ASCR, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| | - Vladimír Kryštof
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany ASCR, Šlechtitelů 11, 78371 Olomouc, Czech Republic
| |
Collapse
|
123
|
Heathcote DA, Patel H, Kroll SHB, Hazel P, Periyasamy M, Alikian M, Kanneganti SK, Jogalekar AS, Scheiper B, Barbazanges M, Blum A, Brackow J, Siwicka A, Pace RDM, Fuchter MJ, Snyder JP, Liotta DC, Freemont PS, Aboagye EO, Coombes RC, Barrett AGM, Ali S. A novel pyrazolo[1,5-a]pyrimidine is a potent inhibitor of cyclin-dependent protein kinases 1, 2, and 9, which demonstrates antitumor effects in human tumor xenografts following oral administration. J Med Chem 2010; 53:8508-22. [PMID: 21080703 DOI: 10.1021/jm100732t] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cyclin-dependent protein kinases (CDKs) are central to the appropriate regulation of cell proliferation, apoptosis, and gene expression. Abnormalities in CDK activity and regulation are common features of cancer, making CDK family members attractive targets for the development of anticancer drugs. Here, we report the identification of a pyrazolo[1,5-a]pyrimidine derived compound, 4k (BS-194), as a selective and potent CDK inhibitor, which inhibits CDK2, CDK1, CDK5, CDK7, and CDK9 (IC₅₀= 3, 30, 30, 250, and 90 nmol/L, respectively). Cell-based studies showed inhibition of the phosphorylation of CDK substrates, Rb and the RNA polymerase II C-terminal domain, down-regulation of cyclins A, E, and D1, and cell cycle block in the S and G₂/M phases. Consistent with these findings, 4k demonstrated potent antiproliferative activity in 60 cancer cell lines tested (mean GI₅₀= 280 nmol/L). Pharmacokinetic studies showed that 4k is orally bioavailable, with an elimination half-life of 178 min following oral dosing in mice. When administered at a concentration of 25 mg/kg orally, 4k inhibited human tumor xenografts and suppressed CDK substrate phosphorylation. These findings identify 4k as a novel, potent CDK selective inhibitor with potential for oral delivery in cancer patients.
Collapse
Affiliation(s)
- Dean A Heathcote
- Dept of Oncology, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, England
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Toumi M, Barbazanges M, Kroll SH, Patel H, Ali S, Coombes RC, Barrett AG. Concise, flexible syntheses of 4-(4-imidazolyl)pyrimidine cyclin-dependent kinase 2 (CDK2) inhibitors. Tetrahedron Lett 2010. [DOI: 10.1016/j.tetlet.2010.09.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
125
|
Lamoral-Theys D, Pottier L, Kerff F, Dufrasne F, Proutière F, Wauthoz N, Neven P, Ingrassia L, Van Antwerpen P, Lefranc F, Gelbcke M, Pirotte B, Kraus JL, Nève J, Kornienko A, Kiss R, Dubois J. Simple di- and trivanillates exhibit cytostatic properties toward cancer cells resistant to pro-apoptotic stimuli. Bioorg Med Chem 2010; 18:3823-33. [PMID: 20466556 PMCID: PMC4966668 DOI: 10.1016/j.bmc.2010.04.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 04/08/2010] [Accepted: 04/16/2010] [Indexed: 01/20/2023]
Abstract
A series of 33 novel divanillates and trivanillates were synthesized and found to possess promising cytostatic rather than cytotoxic properties. Several compounds under study decreased by >50% the activity of Aurora A, B, and C, and WEE1 kinase activity at concentrations <10% of their IC(50) growth inhibitory ones, accounting, at least partly, for their cytostatic effects in cancer cells and to a lesser extent in normal cells. Compounds 6b and 13c represent interesting starting points for the development of cytostatic agents to combat cancers, which are naturally resistant to pro-apoptotic stimuli, including metastatic malignancies.
Collapse
Affiliation(s)
- Delphine Lamoral-Theys
- Laboratoire de Chimie Analytique, Toxicologie et Chimie Physique Appliquée, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Laurent Pottier
- Laboratoire de Toxicologie, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frédéric Kerff
- Centre d'Ingéniérie des Protéines, Institut de Chimie B6, Université de Liège, Liège, Belgium
| | - François Dufrasne
- Laboratoire de Chimie Pharmaceutique Organique, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fabien Proutière
- Laboratoire de Toxicologie, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nathalie Wauthoz
- Laboratoire de Pharmacie Galénique et Biopharmacie, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Philippe Neven
- Laboratoire de Chimie Pharmaceutique, Centre Interfacultaire de Recherche du Médicament, Faculté de Médecine et de Pharmacie, Université de Liège, Liège, Belgium
| | - Laurent Ingrassia
- Laboratoire de Toxicologie, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pierre Van Antwerpen
- Laboratoire de Chimie Pharmaceutique Organique, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Florence Lefranc
- Laboratoire de Toxicologie, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Michel Gelbcke
- Laboratoire de Chimie Pharmaceutique Organique, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Bernard Pirotte
- Laboratoire de Chimie Pharmaceutique, Centre Interfacultaire de Recherche du Médicament, Faculté de Médecine et de Pharmacie, Université de Liège, Liège, Belgium
| | - Jean-Louis Kraus
- Laboratoire de Chimie Biomoléculaire, CNRS, IBDML-UMR-6216, Campus de Luminy Case 907 13288, Marseille cedex 09, France
| | - Jean Nève
- Laboratoire de Chimie Pharmaceutique Organique, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alexander Kornienko
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Robert Kiss
- Laboratoire de Toxicologie, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jacques Dubois
- Laboratoire de Chimie Analytique, Toxicologie et Chimie Physique Appliquée, Institut de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
126
|
Galons H, Oumata N, Meijer L. Cyclin-dependent kinase inhibitors: a survey of recent patent literature. Expert Opin Ther Pat 2010; 20:377-404. [PMID: 20180621 DOI: 10.1517/13543770903524284] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IMPORTANCE OF THE FIELD Abnormalities in protein phosphorylation by cyclin-dependent kinases (CDKs) have been observed in numerous major human diseases, which has strongly encouraged the search for pharmacological inhibitors. Almost 10 years after the first compounds entered clinical studies, numerous CDK inhibitors with differing selectivity profiles are now undergoing preclinical and clinical evaluation. Nevertheless, these intensive searches have not yet resulted in drug approvals. AREAS COVERED IN THIS REVIEW This paper reviews patent activity associated with these efforts during the 2005 - 2008 period. WHAT THE READER WILL GAIN Readers will rapidly obtain an overview of the majority of CDK inhibitor scaffolds; they will discover which companies are the main players in the field and acquire information on products that have reached the clinical phases. TAKE HOME MESSAGE In most cases, applications have been claimed in the field of cancer; however, potential applications of CDK inhibitors in other therapeutic areas are regularly reported and could herald therapeutic introduction over the next few years.
Collapse
Affiliation(s)
- Hervé Galons
- INSERM U648, Université Paris Descartes, 4 avenue de l'observatoire, 75006 Paris, France.
| | | | | |
Collapse
|