1
|
Khalife M, Jia T, Caron P, Shreim A, Genoux A, Cristini A, Pucciarelli A, Leverve M, Lepeltier N, García-Rodríguez N, Dalonneau F, Ramachandran S, Fernandez Martinez L, Marcion G, Lemaitre N, Brambilla E, Garrido C, Hammond E, Huertas P, Gazzeri S, Sordet O, Eymin B. SRSF2 overexpression induces transcription-/replication-dependent DNA double-strand breaks and interferes with DNA repair pathways to promote lung tumor progression. NAR Cancer 2025; 7:zcaf011. [PMID: 40181846 PMCID: PMC11963763 DOI: 10.1093/narcan/zcaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 02/04/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
SRSF2 (serine/arginine-rich splicing factor 2) is a critical regulator of pre-messenger RNA splicing, which also plays noncanonical functions in transcription initiation and elongation. Although elevated levels of SRSF2 are associated with advanced stages of lung adenocarcinoma (LUAD), the mechanisms connecting SRSF2 to lung tumor progression remain unknown. We show that SRSF2 overexpression increases global transcription and replicative stress in LUAD cells, which correlates with the production of DNA damage, notably double-strand breaks (DSBs), likely resulting from conflicts between transcription and replication. Moreover, SRSF2 regulates DNA repair pathways by promoting homologous recombination and inhibiting nonhomologous end joining. Mechanistically, SRSF2 interacts with and enhances MRE11 (meiotic recombination 11) recruitment to chromatin, while downregulating 53BP1 messenger RNA and protein levels. Both events are likely contributing to SRSF2-mediated DNA repair process rerouting. Lastly, we show that SRSF2 and MRE11 expression is commonly elevated in LUAD and predicts poor outcome of patients. Altogether, our results identify a mechanism by which SRSF2 overexpression promotes lung cancer progression through a fine control of both DSB production and repair. Finally, we show that SRSF2 knockdown impairs late repair of ionizing radiation-induced DSBs, suggesting a more global function of SRSF2 in DSB repair by homologous recombination.
Collapse
Affiliation(s)
- Manal Khalife
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Tao Jia
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Pierre Caron
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Amani Shreim
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Aurelie Genoux
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Agnese Cristini
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Amelie Pucciarelli
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Marie Leverve
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Nina Lepeltier
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Néstor García-Rodríguez
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Fabien Dalonneau
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Shaliny Ramachandran
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Lara Fernandez Martinez
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Guillaume Marcion
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Nicolas Lemaitre
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Elisabeth Brambilla
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Carmen Garrido
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Ester M Hammond
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Pablo Huertas
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Sylvie Gazzeri
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Olivier Sordet
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Beatrice Eymin
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| |
Collapse
|
2
|
Hu W, Wang L, Luo J, Zhang J, Li N. The Potent Novel CDK4/6 Inhibitor TQB3616 in Hormone Receptor Positive Breast Cancer: Preclinical Characterization with in vitro and Human Tumor Xenograft Models. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:899-912. [PMID: 38090281 PMCID: PMC10715022 DOI: 10.2147/bctt.s434973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024]
Abstract
PURPOSE Inhibition of the cyclin-dependent kinase (CDK) 4/6-retinoblastoma (RB) pathway exerts a considerable inhibitory effect, preventing the spread and metastasis of breast cancer cells and promoting tumor regression. In this study, we examined the antitumor activity of TQB3616, a novel inhibitor of CDK4/6 activity, which showed a greater efficacy improvement in antitumor effects. METHODS TQB3616 group, abemaciclib group and endocrine or HER-2 targeted combination therapy group were set up respectively. The effects of drugs on cell proliferation activity, cell cycle, apoptosis, downstream protein expression and gene expression of HR positive (T47D, MCF-7) and HER-2 positive (BT474, MDA-MB-361) breast cancer cell lines were studied. The antiproliferative effect of TQB3616 was also measured in vivo. RESULTS TQB3616 showed a remarkable inhibitory effect on the proliferation of hormone receptor-positive breast cancer cells in vitro. In addition, TQB3616 combined with endocrine therapy or Human Epidermal Growth Factor Receptor 2 (HER2) targeted therapy showed significant synergistic antitumor activity in estrogen receptor (ER)-positive/HER2-negative or HER2-positive breast cancer. In contrast to abemaciclib, which targets the CDK4/6 pathway with proven efficacy, the oral agent TQB3616 not only induced G1 stalling, leading to a profound reduction in the level of RB protein phosphorylated at Ser807/811, but also showed enhanced tumor killing effects by promoting cell apoptosis. Oral administration of TQB3616 showed more potent antitumor activity than abemaciclib in an in vitro breast cancer xenograft model, causing significant tumor regression associated with sustained target inhibition in tumor tissue and manageable in vivo toxicity. CONCLUSION The results of this study indicate that TQB3616 is a novel CDK4/6 inhibitor, and its highly effective antitumor activity against breast cancer is expected to yield promising therapeutic effects in clinical studies.
Collapse
Affiliation(s)
- Wenyu Hu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Lei Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - JiaLing Luo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jian Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Nanlin Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
3
|
El-Gazzar MG, El-Gazzar MG, Ghorab MM. Quinazolinone derivatives as new potential CDK4/6 inhibitors, apoptosis inducers and radiosensitizers for breast cancer. Future Med Chem 2023; 15:1133-1147. [PMID: 37529897 DOI: 10.4155/fmc-2023-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023] Open
Abstract
Background: Targeting CDK4/6 has advanced breast cancer treatment. Herein, new quinazolinones were synthesized with acetamide linkers as potential anti-breast cancer agents. Methods & results: In vitro cytotoxic evaluation on human breast cancer cell lines (MCF7 and MDA-MB-231) identified 1,3-benzodioxole (5d) to be of the highest potency. It showed good inhibitory activity on CDK4/6. Compound 5d arrested the cell cycle at the G1-phase, caused induction of early and late apoptosis in an Annexin V-FITC assay, led to an increase in the level of caspase-3 and upregulated Bax expression and downregulated Bcl-2 in MCF7 cells. Compound 5d showed good radiosensitizing activity when combined with a single dose of 8-Gy γ-radiation. Conclusion: This study introduces quinazolinone scaffolds as new CDK4/6 inhibitors for breast cancer.
Collapse
Affiliation(s)
- Mostafa Gm El-Gazzar
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, 11787, Egypt
| | - Marwa G El-Gazzar
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, 11787, Egypt
| | - Mostafa M Ghorab
- Department of Drug Radiation Research, National Center for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, 11787, Egypt
| |
Collapse
|
4
|
Fang G, Chen H, Cheng Z, Tang Z, Wan Y. Azaindole derivatives as potential kinase inhibitors and their SARs elucidation. Eur J Med Chem 2023; 258:115621. [PMID: 37423125 DOI: 10.1016/j.ejmech.2023.115621] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
Currently, heterocycles have occupied an important position in the fields of drug design. Among them, azaindole moiety is regarded as one privileged scaffold to develop therapeutic agents. Since two nitrogen atoms of azaindole increase the possibility to form hydrogen bonds in the adenosine triphosphate (ATP)-binding site, azaindole derivatives are important sources of kinase inhibitors. Moreover, some of them have been on the market or in clinical trials for the treatment of some kinase-related diseases (e.g., vemurafenib, pexidartinib, decernotinib). In this review, we focused on the recent development of azaindole derivatives as potential kinase inhibitors based on kinase targets, such as adaptor-associated kinase 1 (AAK1), anaplastic lymphoma kinase (ALK), AXL, cell division cycle 7 (Cdc7), cyclin-dependent kinases (CDKs), dual-specificity tyrosine (Y)-phosphorylation regulated kinase 1A (DYRK1A), fibroblast growth factor receptor 4 (FGFR4), phosphatidylinositol 3-kinase (PI3K) and proviral insertion site in moloney murine leukemia virus (PIM) kinases. Meanwhile, the structure-activity relationships (SARs) of most azaindole derivatives were also elucidated. In addition, the binding modes of some azaindoles complexed with kinases were also investigated during the SARs elucidation. This review may offer an insight for medicinal chemists to rationally design more potent kinase inhibitors bearing the azaindole scaffold.
Collapse
Affiliation(s)
- Guoqing Fang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, 411201, PR China
| | - Hongjuan Chen
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, 411201, PR China
| | - Zhiyun Cheng
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, 411201, PR China
| | - Zilong Tang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, 411201, PR China
| | - Yichao Wan
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, 411201, PR China.
| |
Collapse
|
5
|
Zhang H, Yan S, Zhan Y, Ma S, Bian Y, Li S, Tian J, Li G, Zhong D, Diao X, Miao L. A mass balance study of [14C]SHR6390 (dalpiciclib), a selective and potent CDK4/6 inhibitor in humans. Front Pharmacol 2023; 14:1116073. [PMID: 37063263 PMCID: PMC10102643 DOI: 10.3389/fphar.2023.1116073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
SHR6390 (dalpiciclib) is a selective and effective cyclin-dependent kinase (CDK) 4/6 inhibitor and an effective cancer therapeutic agent. On 31 December 2021, the new drug application was approved by National Medical Product Administration (NMPA). The metabolism, mass balance, and pharmacokinetics of SHR6390 in 6 healthy Chinese male subjects after a single oral dose of 150 mg [14C]SHR6390 (150 µCi) in this research. The Tmax of SHR6390 was 3.00 h. In plasma, the t1/2 of SHR6390 and its relative components was approximately 17.50 h. The radioactivity B/P (blood-to-plasma) AUC0-t ratio was 1.81, indicating the preferential distribution of drug-related substances in blood cells. At 312 h after administration, the average cumulative excretion of radioactivity was 94.63% of the dose, including 22.69% in urine and 71.93% in stool. Thirteen metabolites were identified. In plasma, because of the low level of radioactivity, only SHR6390 was detected in pooled AUC0-24 h plasma. Stool SHR6390 was the main component in urine and stool. Five metabolites were identified in urine, and 12 metabolites were identified in stool. Overall, faecal clearance is the main method of excretion.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Shu Yan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yan Zhan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Sheng Ma
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Yicong Bian
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Shaorong Li
- Jiangsu Hengrui Medicine Co., Ltd., Lianyungang, Jiangsu, China
| | - Junjun Tian
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Guangze Li
- Jiangsu Hengrui Medicine Co., Ltd., Lianyungang, Jiangsu, China
| | - Dafang Zhong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Dafang Zhong, ; Xingxing Diao, ; Liyan Miao,
| | - Xingxing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Dafang Zhong, ; Xingxing Diao, ; Liyan Miao,
| | - Liyan Miao
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- *Correspondence: Dafang Zhong, ; Xingxing Diao, ; Liyan Miao,
| |
Collapse
|
6
|
Fang L, Chu M, Yan C, Liu Y, Zhao Z. Palbociclib and Michael-acceptor hybrid compounds as CDK4/6 covalent inhibitors: improved potency, broad anticancer spectrum and overcoming drug resistance. Bioorg Med Chem 2023; 84:117263. [PMID: 37011445 DOI: 10.1016/j.bmc.2023.117263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 03/28/2023]
Abstract
To search for potent CDK4/6 covalent inhibitors, total 14 compounds have been designed and synthesized by connecting different Michael-acceptor to the piperazine moiety of palbociclib. All the compounds displayed good antiproliferative activity against human hepatoma cell (HepG2), non-small cell lung cancer (A549), and breast cancer (MDA-MB-231 and MCF-7) cell lines. In particular, compound A4 showed the highest inhibitory activity to MDA-MB-231 and MCF-7 cells with IC50 values of 0.51 μM and 0.48 μM, respectively. More importantly, A4 also showed strong inhibition against MDA-MB-231/palbociclib cells, indicating that A4 could effectively avoid the resistance of palbociclib. In the enzyme test, A4 showed selective inhibitory activity against CDK4/6, with the IC50 value of 18 nM and 13 nM, respectively. It was also found that A4 could efficiently induce apoptosis and arrest the cell cycle at G0/G1 phase. Moreover, A4 could significantly decrease the phosphorylation level of CDK4 and CDK6. HPLC and molecular modeling studies suggested that A4 could form a covalent bond with the target protein.
Collapse
Affiliation(s)
- Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| | - Mengqi Chu
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Changhang Yan
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yilin Liu
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Zimeng Zhao
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
7
|
Xue Z, Zeng J, Yin X, Li Y, Meng B, Zhao Y, Fang X, Gong X, Dai X. Investigation on acquired palbociclib resistance by LC-MS based multi-omics analysis. Front Mol Biosci 2023; 10:1116398. [PMID: 36743215 PMCID: PMC9892630 DOI: 10.3389/fmolb.2023.1116398] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Palbociclib is a specific CDK4/6 inhibitor that has been widely applied in multiple types of tumors. Different from cytotoxic drugs, the anticancer mechanism of palbociclib mainly depends on cell cycle inhibition. Therefore, the resistance mechanism is different. For clinical cancer patients, drug resistance is inevitable for almost all cancer therapies including palbociclib. We have trained palbociclib resistant cells in vitro to simulate the clinical situation and applied LC-MS multi-omics analysis methods including proteomic, metabolomic, and glycoproteomic techniques, to deeply understand the underly mechanism behind the resistance. As a result of proteomic analysis, the resistant cells were found to rely on altered metabolic pathways to keep proliferation. Metabolic processes related to carbohydrates, lipids, DNA, cellular proteins, glucose, and amino acids were observed to be upregulated. Most dramatically, the protein expressions of COX-1 and NDUFB8 have been detected to be significantly overexpressed by proteomic analysis. When a COX-1 inhibitor was hired to combine with palbociclib, a synergistic effect could be obtained, suggesting the altered COX-1 involved metabolic pathway is an important reason for the acquired palbociclib resistance. The KEGG pathway of N-glycan biosynthesis was identified through metabolomics analysis. N-glycoproteomic analysis was therefore included and the global glycosylation was found to be elevated in the palbociclib-resistant cells. Moreover, integration analysis of glycoproteomic data allowed us to detect a lot more proteins that have been glycosylated with low abundances, these proteins were considered to be overwhelmed by those highly abundant proteins during regular proteomic LC-MS detection. These low-abundant proteins are mainly involved in the cellular biology processes of cell migration, the regulation of chemotaxis, as well as the glycoprotein metabolic process which offered us great more details on the roles played by N-glycosylation in drug resistance. Our result also verified that N-glycosylation inhibitors could enhance the cell growth inhibition of palbociclib in resistant cells. The high efficiency of the integrated multi-omics analysis workflow in discovering drug resistance mechanisms paves a new way for drug development. With a clear understanding of the resistance mechanism, new drug targets and drug combinations could be designed to resensitize the resistant tumors.
Collapse
Affiliation(s)
- Zhichao Xue
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Jiaming Zeng
- College of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang, China
| | - Xinchi Yin
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Yongshu Li
- Shenzhen Institute for Technology Innovation, National Institute of Metrology Shenzhen, Shenzhen, China
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Yang Zhao
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Xiang Fang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China,*Correspondence: Xiaoyun Gong, ; Xinhua Dai,
| | - Xinhua Dai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China,*Correspondence: Xiaoyun Gong, ; Xinhua Dai,
| |
Collapse
|
8
|
He H, Liu Q, Chen L, Wang J, Yuan Y, Li H, Qian X, Zhao Z, Chen Z. Design, synthesis and biological evaluation of pteridine-7(8H)-one derivatives as potent and selective CDK4/6 inhibitors. Bioorg Med Chem Lett 2022; 76:128991. [PMID: 36130661 DOI: 10.1016/j.bmcl.2022.128991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 11/02/2022]
Abstract
Cyclin-dependent kinases play an important role in the regulation of cell cycle and transcription. Selective CDK4/6 inhibitors have been demonstrated to be effective in the treatment of cancer. In this article, we described the design and synthesis of a series of pteridine-7(8H)-one derivatives as dual CDK4/6 inhibitors. Among them, the most promising compound L2 exhibited significant inhibitory activity against CDK4 and CDK6 with IC50 values of 16.7 nM and 30.5 nM respectively and showed excellent selectivity to CDK1/2/7/9. Moreover, compound L2 displayed potent antiproliferative activities at low digital micromolar range via inducing apoptosis in breast and colon cancer cells. In all, we developed a new series of pteridine-7(8H)-one derivatives which exhibited promising antitumor activities as selective CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Huan He
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Qi Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Lu Chen
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Jie Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Yuan Yuan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Xuhong Qian
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China
| | - Zhenjiang Zhao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China.
| | - Zhuo Chen
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai 200237, China.
| |
Collapse
|
9
|
Fakhri S, Zachariah Moradi S, DeLiberto LK, Bishayee A. Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies. Biochem Pharmacol 2022; 199:114989. [DOI: 10.1016/j.bcp.2022.114989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022]
|
10
|
Novel Combinatorial Strategy Using Thermal Inkjet Bioprinting, Chemotherapy, and Radiation on Human Breast Cancer Cells; an In-Vitro Cell Viability Assessment. MATERIALS 2021; 14:ma14247864. [PMID: 34947458 PMCID: PMC8704911 DOI: 10.3390/ma14247864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022]
Abstract
Background: Breast cancer (BC) continues to have the second highest mortality amongst women in the United States after lung cancer. For 2021, the American Cancer Association predicted 281,550 new invasive breast cancer cases besides 49,290 new cases of non-invasive breast cancer and 43,600 deaths from the metastatic disease. A treatment modality is radiation therapy, which is given for local control as well as palliation of patient symptoms. The initial step of new drug development is in-vitro cell studies, which help describe new drug properties and toxicities. However, these models are not optimal, and better ones have yet to be determined. This study uses bioprinting technology to elucidate the sensitivity of tumor cells to the combination of palbociclib (PD) and letrozole (Let) treatment. We hypothesize that this technology could serve as a model to predict treatment outcomes more efficiently. Methods: The breast cancer cell lines MCF7 and MDA-MB-231 as well as the normal breast epithelial cell line, MCF-10A, were treated with PD-Let with and without radiotherapy (RT), and cell viability was compared in pairwise fashion for thermally inkjet bioprinted (TIB) and manually seeded (MS) cells. Results: In absence of radiation, the TIB MCF7 cells have 2.5 times higher viability than manually seeded (MS) cells when treated with 100 µM palbociclib and 10 µM letrozole, a 36% higher viability when treated with 50 µM palbociclib and 10 µM letrozole, and an 8% higher viability when treated with 10 µM palbociclib and 10 µM letrozole. With 10 Gy of radiation, TIB cells had a 45% higher survival rate than MS cells at the lowest palbociclib concentration and a 29% higher survival rate at the intermediate palbociclib concentration. Without radiation treatment, at a concentration of 10 μM PD-Let, TIB MDA-MB-231 cells show a 8% higher viability than MS cells when treated with 10 µM PD and 10 µM Let; at higher drug concentrations, the differences disappeared, but some 1.7% of the TIB MDA-MB-231 cells survived exposure to 150 μM of PD + 10 μM letrozole vs. none of the MS cells. These cells are more radiation sensitive than the other cell lines tested and less sensitive to the combo drug treatments. We observed an 18% higher survival of TIB MCF-10A cells without radiation treatment when exposed to 10 μM PD + 10 μM Let but no difference in cell survival between the two groups when radiation was applied. Independent of growth conditions, TIB cells did not show more resistance to radiation treatment than MS cells, but a higher resistance to the combo treatment was observed, which was most pronounced in the MCF-7 cell line. Conclusion: Based on these results, we suggest that TIB used in in-vitro models could be a feasible strategy to develop and/or test new anticancer drugs.
Collapse
|
11
|
Innovations and Patent Trends in the Development of USFDA Approved Protein Kinase Inhibitors in the Last Two Decades. Pharmaceuticals (Basel) 2021; 14:ph14080710. [PMID: 34451807 PMCID: PMC8400070 DOI: 10.3390/ph14080710] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Protein kinase inhibitors (PKIs) are important therapeutic agents. As of 31 May 2021, the United States Food and Drug Administration (USFDA) has approved 70 PKIs. Most of the PKIs are employed to treat cancer and inflammatory diseases. Imatinib was the first PKI approved by USFDA in 2001. This review summarizes the compound patents and the essential polymorph patents of the PKIs approved by the USFDA from 2001 to 31 May 2021. The dates on the generic drug availability of the PKIs in the USA market have also been forecasted. It is expected that 19 and 48 PKIs will be genericized by 2025 and 2030, respectively, due to their compound patent expiry. This may reduce the financial toxicity associated with the existing PKIs. There are nearly 535 reported PKs. However, the USFDA approved PKIs target only about 10-15% of the total said PKs. As a result, there are still a large number of unexplored PKs. As the field advances during the next 20 years, one can anticipate that PKIs with many scaffolds, chemotypes, and pharmacophores will be developed.
Collapse
|
12
|
Palbociclib combined with endocrine therapy in heavily pretreated HR +/HER2 - advanced breast cancer patients: Results from the compassionate use program in Spain (PALBOCOMP). Breast 2020; 54:286-292. [PMID: 33242755 PMCID: PMC7695980 DOI: 10.1016/j.breast.2020.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 01/22/2023] Open
Abstract
Background This study evaluated efficacy and safety of palbociclib, a CDK4/6 inhibitor, in heavily-pretreated hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer (mBC) patients during the compassionate use program in Spain from February 2015 to November 2017. Patients and methods Patient data were collected retrospectively from 35 hospitals in Spain. Patients with HR+/HER2- mBC who had progressed on ≥4 treatments for advanced disease were eligible. Results A total of 219 patients received palbociclib in combination with aromatase inhibitors (110; 50.2%), fulvestrant (87; 39.7%), tamoxifen (8; 3.6%) or as single agent (10; 4.6%). Mean age of the patients was 58 years; 31 patients (16.1%) were premenopausal and 162 (83.9%) were postmenopausal at the beginning of treatment with palbociclib. Patients had received a median of 3 previous lines of endocrine therapy (ET) for advanced disease. Real-world tumor response (rwTR) and clinical benefit rate were 5.9% (n = 13) and 46.2% (n = 101), respectively. The median real world progression-free survival (rwPFS) was 6.0 months (95% CI 5.7–7.0) and the median overall survival was 19.0 months (95% CI 16.4–21.7). Subgroup analysis revealed a significant difference in median rwPFS in patients treated with palbociclib plus fulvestrant depending on the duration of prior treatment with fulvestrant monotherapy (>6 versus ≤6 months; HR 1.93, 95% CI 1.37–2.73, p < 0.001). The most frequently reported toxicities were neutropenia, asthenia, thrombopenia and anemia. Conclusions Palbociclib can be an effective and safe treatment option in patients with heavily pretreated endocrine-sensitive mBC, especially in those with longer PFS to previous ET. CDK4/6 inhibitors combined with endocrine therapy have been widely accepted as a new standard therapy for hormone receptor-positive metastatic breast cancer patients in first or second line. Palbociclib alone or in combination with aromatase inhibitors, fulvestrant, or tamoxifen was effective and safe in heavily pretreated HR+/HER2- metastatic breast cancer patients. Palbociclib could be of higher benefit to patients with endocrine-sensitive disease that had a long duration of response to previous endocrine therapy. Real-world evidence of effectiveness and safety of use of palbociclib in heavily pretreated advanced breast cancer patients complements data from randomized clinical trials.
Collapse
|
13
|
Abstract
Proteolysis-targeting chimera (PROTAC) is a new technology to selectively degrade target proteins via ubiquitin-proteasome system. PROTAC molecules (PROTACs) are a class of heterobifunctional molecules, which contain a ligand targeting the protein of interest, a ligand recruiting an E3 ligase and a linker connecting these two ligands. They provide several advantages over traditional inhibitors in potency, selectivity and drug resistance. Thus, many promising PROTACs have been developed in the recent two decades, especially small-molecule PROTACs. In this review, we briefly introduce the mechanism of PROTACs and focus on the progress of small-molecule PROTACs based on different E3 ligases. In addition, we also introduce the opportunities and challenges of small-molecule PROTACs for cancer therapy.
Collapse
|
14
|
Lin DI, Hemmerich A, Edgerly C, Duncan D, Severson EA, Huang RSP, Ramkissoon SH, Connor YD, Shea M, Hecht JL, Ali SM, Vergilio JA, Ross JS, Elvin JA. Genomic profiling of BCOR-rearranged uterine sarcomas reveals novel gene fusion partners, frequent CDK4 amplification and CDKN2A loss. Gynecol Oncol 2020; 157:357-366. [PMID: 32156473 DOI: 10.1016/j.ygyno.2020.02.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Genomic alterations of BCOR via ZC3H7B-BCOR fusion or BCOR internal tandem duplication (ITD) define a subset of endometrial stromal sarcoma (ESS). The goals of this study were to: 1) determine the molecular landscape of BCOR-rearranged ESS, 2) to identify novel BCOR fusion gene partners in ESS and their associated clinicopathological characteristics, and 3) to potentially unravel targetable genomic alterations in BCOR-mutated ESS. METHODS A retrospective database search of a CLIA-certified molecular laboratory was performed for uterine sarcomas that contained BCOR rearrangements or BCOR ITD. The cases were previously assayed by comprehensive genomic profiling via both DNA- and RNA-based targeted next generation sequencing during the course of clinical care. Clinicopathological and genomic data was centrally re-reviewed. RESULTS We identify largest cohort of BCOR-rearranged ESS to date (n = 40), which included 31 cases with canonical ZC3H7B-BCOR fusion as well as 8 cases with novel BCOR gene rearrangement partners, such as BCOR-L3MBTL2, EP300-BCOR, BCOR-NUTM2G, BCOR-RALGPS1, BCOR-MAP7D2, RGAG1-BCOR, ING3-BCOR, BCOR-NUGGC, KMT2D-BCOR, CREBBP-BCOR and 1 case with BCOR internal rearrangement. Re-review of cases with novel rearrangements demonstrated sarcomas with spindle, epithelioid or small round cell components and frequent myxoid stromal change. Comprehensive genomic profiling revealed high frequency of CDK4 and MDM2 amplification in 38% and 45% of BCOR-rearranged cases, respectively, and homozygous deletion of CDKN2A, which encodes an inhibitor of CDK4 in 28% of cases. Notably, CDK4 and MDM2 amplification was absent in all cases from 15 different ESS cases harboring BCOR ITD. CONCLUSIONS Alterations of CDK4 pathway members, for which targeted therapy is clinically available (i.e. palbociclib), via CDK4 amplification or CDKN2A loss, contributes to the pathogenesis of BCOR-rearranged uterine sarcomas, which may have therapeutic implications.
Collapse
Affiliation(s)
- Douglas I Lin
- Foundation Medicine Inc., Cambridge, MA, United States of America.
| | - Amanda Hemmerich
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | - Claire Edgerly
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | - Daniel Duncan
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | - Eric A Severson
- Foundation Medicine Inc., Morrisville, NC, United States of America
| | | | - Shakti H Ramkissoon
- Foundation Medicine Inc., Morrisville, NC, United States of America; Wake Forest Comprehensive Cancer Center, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Yamicia D Connor
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Meghan Shea
- Department of Internal Medicine, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Siraj M Ali
- Foundation Medicine Inc., Cambridge, MA, United States of America
| | - Jo-Anne Vergilio
- Foundation Medicine Inc., Cambridge, MA, United States of America
| | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, MA, United States of America; Upstate Medical University, Syracuse, NY, United States of America
| | - Julia A Elvin
- Foundation Medicine Inc., Cambridge, MA, United States of America
| |
Collapse
|
15
|
Lee Y, Lahens NF, Zhang S, Bedont J, Field JM, Sehgal A. G1/S cell cycle regulators mediate effects of circadian dysregulation on tumor growth and provide targets for timed anticancer treatment. PLoS Biol 2019; 17:e3000228. [PMID: 31039152 PMCID: PMC6490878 DOI: 10.1371/journal.pbio.3000228] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/27/2019] [Indexed: 12/31/2022] Open
Abstract
Circadian disruption has multiple pathological consequences, but the underlying mechanisms are largely unknown. To address such mechanisms, we subjected transformed cultured cells to chronic circadian desynchrony (CCD), mimicking a chronic jet-lag scheme, and assayed a range of cellular functions. The results indicated a specific circadian clock–dependent increase in cell proliferation. Transcriptome analysis revealed up-regulation of G1/S phase transition genes (myelocytomatosis oncogene cellular homolog [Myc], cyclin D1/3, chromatin licensing and DNA replication factor 1 [Cdt1]), concomitant with increased phosphorylation of the retinoblastoma (RB) protein by cyclin-dependent kinase (CDK) 4/6 and increased G1-S progression. Phospho-RB (Ser807/811) was found to oscillate in a circadian fashion and exhibit phase-shifted rhythms in circadian desynchronized cells. Consistent with circadian regulation, a CDK4/6 inhibitor approved for cancer treatment reduced growth of cultured cells and mouse tumors in a time-of-day–specific manner. Our study identifies a mechanism that underlies effects of circadian disruption on tumor growth and underscores the use of treatment timed to endogenous circadian rhythms. A study of “jet-lagged” cells reveals a specific molecular mechanism regulating cell proliferation that it impacted by circadian disruption, underscoring the importance of administering anti-cancer treatment at a specific time of day. Circadian misalignment caused by altered sleep–wake cycles, shift work, or frequent jet lag increases susceptibility to several disorders, including cancer. However, the mechanisms by which circadian disruption contributes to disease are not well understood, and so we addressed this issue by investigating the molecular, cellular, and biochemical consequences of chronic circadian desynchronization. Our studies using cancer cell or tumor tissue models show that chronic circadian desynchronization induces multiple oncogenic pathways to promote cell proliferation. In particular, chronic circadian desynchronization promotes phosphorylation of the retinoblastoma (RB) protein, thereby favoring G1/S phase cell cycle progression. Consistent with these findings, the antiproliferative activity of a selective inhibitor of the enzyme that phosphorylates RB has time-of-day–specific effects on cancer cells and mouse tumors, but this time dependence is abrogated by chronic jet-lag conditions. These data suggest a circadian regulation of G1/S cell cycle progression and provide an important molecular rationale for time-of-day–specific treatment of cancer patients, also known as chronotherapy.
Collapse
Affiliation(s)
- Yool Lee
- Penn Chronobiology, Howard Hughes Medical Institute, Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nicholas F. Lahens
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Shirley Zhang
- Penn Chronobiology, Howard Hughes Medical Institute, Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joseph Bedont
- Penn Chronobiology, Howard Hughes Medical Institute, Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jeffrey M. Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amita Sehgal
- Penn Chronobiology, Howard Hughes Medical Institute, Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Long F, He Y, Fu H, Li Y, Bao X, Wang Q, Wang Y, Xie C, Lou L. Preclinical characterization of SHR6390, a novel CDK 4/6 inhibitor, in vitro and in human tumor xenograft models. Cancer Sci 2019; 110:1420-1430. [PMID: 30724426 PMCID: PMC6447953 DOI: 10.1111/cas.13957] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
Inhibition of the cyclin‐dependent kinase (CDK) 4/6‐retinoblastoma (RB) pathway is an effective therapeutic strategy against cancer. Here, we performed a preclinical investigation of the antitumor activity of SHR6390, a novel CDK4/6 inhibitor. SHR6390 exhibited potent antiproliferative activity against a wide range of human RB‐positive tumor cells in vitro, and exclusively induced G1 arrest as well as cellular senescence, with a concomitant reduction in the levels of Ser780‐phosphorylated RB protein. Compared with the well‐known CDK4/6 inhibitor palbociclib, orally administered SHR6390 led to equivalent or improved tumor efficacy against a panel of carcinoma xenografts, and produced marked tumor regression in some models, in association with sustained target inhibition in tumor tissues. Furthermore, SHR6390 overcame resistance to endocrine therapy and HER2‐targeting antibody in ER‐positive and HER2‐positive breast cancer, respectively. Moreover, SHR6390 combined with endocrine therapy exerted remarkable synergistic antitumor activity in ER‐positive breast cancer. Taken together, our findings indicate that SHR6390 is a novel CDK4/6 inhibitor with favorable pharmaceutical properties for use as an anticancer agent.
Collapse
Affiliation(s)
- Fei Long
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Zhejiang, China.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ye He
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Haoyu Fu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yun Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xubin Bao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Quanren Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Zhejiang, China
| | - Chengying Xie
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Liguang Lou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Du Y, Yan D, Yuan Y, Xu J, Wang S, Yang Z, Cheng W, Tian X, Kan Q. CDK11 p110 plays a critical role in the tumorigenicity of esophageal squamous cell carcinoma cells and is a potential drug target. Cell Cycle 2019; 18:452-466. [PMID: 30722725 DOI: 10.1080/15384101.2019.1577665] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a serious malignancy with limited options for targeted therapy. The exploration of novel targeted therapies for combating ESCC is urgently needed. Cyclin-dependent kinases (CDKs) play important roles in the progression of cancers; however, the function of CDK11p110 (cyclin-dependent kinase 11p110) in ESCC is still unknown. Here, we investigated the effects and molecular mechanisms of CDK11p110 in the proliferation and growth of ESCC by examining the expression of CDK11p110 in ESCC tissues and by detecting phenotypic changes in ESCC cells after CDK11p110 knockdown or overexpression in vitro and in vivo. According to the tissue microarray analysis, compared with its expression level in normal tissues, the expression level of CDK11p110 was significantly elevated in ESCC tissues; this result was in concordance with the data in TCGA (The Cancer Genome Atlas) datasets. In addition, RNAi-mediated CDK11p110 silencing exerted a substantial inhibitory effect on the proliferation, clonogenicity and migration ability of ESCC cells. Further study indicated that CDK11p110 knockdown arrested ESCC cells in the G2/M phase of the cell cycle and induced cell apoptosis. Moreover, stable shRNA-mediated CDK11p110 knockdown inhibited tumor growth in an ESCC xenograft model, and overexpression of CDK11p110 enhanced tumor growth. In addition, the Ki67 proliferation index was closely associated with the elevation or depletion of CDK11p110 in vivo. In summary, this study provides evidence that CDK11p110 play a critical role in the tumorigenicity of ESCC cells, which suggests that CDK11p110 may be a promising therapeutic target in ESCC. Abbreviations: CDKs: cyclin-dependent kinases; CDK11: Cyclin-dependent kinase 11; CDK11p110: Cyclin-dependent kinase 11p110, the larger isomer of cyclin-dependent kinase 11; ESCC: esophageal squamous cell carcinoma; FACS: fluorescence-activated cell sorting; FDA: the Food and Drug Administration; TCGA: The Cancer Genome Atlas; TMA: tissue microarray.
Collapse
Affiliation(s)
- Yue Du
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Dan Yan
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Yongliang Yuan
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Jian Xu
- c Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Suhua Wang
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Zhiheng Yang
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Weiyan Cheng
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Xin Tian
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Quancheng Kan
- a Department of Pharmacy , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China.,b Henan Key Laboratory of Precision Clinical Pharmacy , The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| |
Collapse
|
18
|
Cheng W, Yang Z, Wang S, Li Y, Wei H, Tian X, Kan Q. Recent development of CDK inhibitors: An overview of CDK/inhibitor co-crystal structures. Eur J Med Chem 2019; 164:615-639. [PMID: 30639897 DOI: 10.1016/j.ejmech.2019.01.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/31/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023]
Abstract
The cyclin-dependent protein kinases (CDKs) are protein-serine/threonine kinases that display crucial effects in regulation of cell cycle and transcription. While the excessive expression of CDKs is intimate related to the development of diseases including cancers, which provides opportunities for disease treatment. A large number of small molecules are explored targeting CDKs. CDK/inhibitor co-crystal structures play an important role during the exploration of inhibitors. So far nine kinds of CDK/inhibitor co-crystals have been determined, they account for the highest proportion among the Protein Data Bank (PDB) deposited crystal structures. Herein, we review main co-crystals of CDKs in complex with corresponding inhibitors reported in recent years, focusing our attention on the binding models and the pharmacological activities of inhibitors.
Collapse
Affiliation(s)
- Weiyan Cheng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhiheng Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Suhua Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ying Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Han Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|