1
|
Rawling MJ, Ray-Sinha A, Bestwick M, Carter MK, Chahal SK, Chalmers AJ, Elsey DJ, Giddings A, Gold J, Henderson SH, MacGregor C, Malcolm A, Ortega F, Phelan A, Savory ED, Schwartz AC, Stevenson NG, Turner EL, Vass M, Wright JA, Watson C. Discovery of a Potent, Selective, and Brain-Penetrant Checkpoint Kinase 1 Inhibitor, BEN-28010, for the Treatment of Glioblastoma. J Med Chem 2025; 68:9101-9125. [PMID: 40273287 DOI: 10.1021/acs.jmedchem.5c00279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Glioblastoma (GBM) patients face a dire prognosis and alternative therapeutic options are desperately needed. Inhibition of checkpoint kinase 1 (CHK1) represents a potential therapeutic strategy for GBM through regulation of the DNA damage response (DDR) pathway, but no suitable brain-penetrant CHK1 inhibitors have been reported to date. In this study, we disclose the discovery and optimization of clinical candidate 38 (BEN-28010) as a freely brain-penetrant, potent, and selective CHK1 inhibitor, derived from virtual screening hit 1. In vivo pharmacological studies demonstrated efficacy of orally administered 38 in several GBM CDX and PDX models as a monotherapy and in combination with ionizing radiation, including improved overall survival in an intracranially implanted GBM PDX mouse model. Additionally, 38 utilizes an underrepresented aminoimidazole kinase hinge-binding motif that may have broader utility within kinase inhibitor drug discovery.
Collapse
Affiliation(s)
- Michael J Rawling
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | - Michael Bestwick
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | - Sandeep K Chahal
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, U.K
| | | | | | | | - Jennie Gold
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Scott H Henderson
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | - Andrew Malcolm
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Fernando Ortega
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Anne Phelan
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Edward D Savory
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Alejandro C Schwartz
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Neil G Stevenson
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Emma L Turner
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Márton Vass
- BenevolentAI, 4-8 Maple Street, London W1T 5HD, U.K
| | - Jamie A Wright
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Christine Watson
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| |
Collapse
|
2
|
Ferguson ML, Ciufolini MA. Diversity Oriented Routes to Thiopeptide Antibiotics: A Solution to the "Thiazole Problem". J Org Chem 2024; 89:18729-18732. [PMID: 39604077 DOI: 10.1021/acs.joc.4c02362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The Goossen decarboxylative coupling reaction enables the union of thiazole-2-carboxylic acids with a 2-pyridyl triflate, leading to the formation of pyridine-thiazole clusters of the kind found in thiopeptide antibiotics. The method avoids problematic or technically challenging reaction sequences involving 2-thiazolyl organometallics, facilitating the investigation of the structure-activity relationship of the thiopeptides.
Collapse
Affiliation(s)
- Mai Lam Ferguson
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Marco A Ciufolini
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| |
Collapse
|
3
|
Luo J, Li Y, Zhang Y, Wu D, Ren Y, Liu J, Wang C, Zhang J. An update on small molecule compounds targeting synthetic lethality for cancer therapy. Eur J Med Chem 2024; 278:116804. [PMID: 39241482 DOI: 10.1016/j.ejmech.2024.116804] [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] [Received: 05/07/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Targeting cancer-specific vulnerabilities through synthetic lethality (SL) is an emerging paradigm in precision oncology. A SL strategy based on PARP inhibitors has demonstrated clinical efficacy. Advances in DNA damage response (DDR) uncover novel SL gene pairs. Beyond BRCA-PARP, emerging SL targets like ATR, ATM, DNA-PK, CHK1, WEE1, CDK12, RAD51, and RAD52 show clinical promise. Selective and bioavailable small molecule inhibitors have been developed to induce SL, but optimization for potency, specificity, and drug-like properties remains challenging. This article illuminated recent progress in the field of medicinal chemistry centered on the rational design of agents capable of eliciting SL specifically in neoplastic cells. It is envisioned that innovative strategies harnessing SL for small molecule design may unlock novel prospects for targeted cancer therapeutics going forward.
Collapse
Affiliation(s)
- Jiaxiang Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yang Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yiwen Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Defa Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jie Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Chengdi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
4
|
Hu S, Jiang C, Jin Q. Discovery of pyrido[3,2-d]pyrimidin-6(5H)-one derivatives as checkpoint kinase 1 (CHK1) inhibitors with potent antitumor efficacy. Eur J Med Chem 2024; 269:116351. [PMID: 38547734 DOI: 10.1016/j.ejmech.2024.116351] [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] [Received: 01/21/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024]
Abstract
Checkpoint kinase 1 (CHK1) plays a crucial role in the DNA damage response pathway, making it an attractive target for cancer therapy. Herein, we present the synthesis, optimization, and evaluation of selective CHK1 inhibitors with a pyrido[3,2-d]pyrimidin-6(5H)-one scaffold. Among them, compound 11 showed single-digit nanomolar potency against CHK1 (IC50: 0.55 nM) with good kinase selectivity. Notably, 11 showed anti-proliferative effect in MV-4-11 cells singly (IC50 = 202 nM) and a synergistic effect in combination with gemcitabine in HT-29 cells (IC50 = 63.53 nM). Furthermore, the combination of 11 and gemcitabine exhibited synergistic effect in the HT-29 xenograft mouse model. Overall, this work provides a strong foundation for the development of selective CHK1 inhibitors and the therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Shihe Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China; SkyRun Pharma Co., Ltd., No. 9 Weidi Road, Nanjing, 210046, China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Qiaomei Jin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| |
Collapse
|
5
|
Qin L, Su X, Wang J, Wu L, Zou M, Gu J, Xu Y, Nie Y. Regulating the stereoselectivity of medium-chain dehydrogenase/reductase by creating an additional active pocket accommodating prochiral heterocyclic ketones. Chem Commun (Camb) 2023; 59:13042-13045. [PMID: 37846488 DOI: 10.1039/d3cc04361d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Medium chain dehydrogenase/reductase (MDR) is a robust catalyst for the asymmetric synthesis of chiral heterocyclic alcohols, essential building blocks in pharmaceuticals. However, the regulatory mechanism of stereoselective complementary reduction of heterocyclic ketones by carbonyl reductase (CR) is unclear. Structure-guided creation of an additional substrate-binding active pocket inversed the stereoselectivity of SpCR from Spathaspora passalidarum. The mutant m48 showed improved catalytic activity towards the 12 tested heterocyclic ketones (conversion rate >99%, ee value > 99%). Hence, we regulated the stereoselectivity of MDR by creating an active pocket suitable for substrate localisation. This strategy has a guiding significance in addition to the conventional method for stereoselectivity modification of MDR.
Collapse
Affiliation(s)
- Lei Qin
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Xin Su
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Jun Wang
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Lunjie Wu
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Man Zou
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Jie Gu
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Yan Xu
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yao Nie
- Laboratory of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
6
|
Xu Z, Zhuang Y, Chen Q. Current scenario of pyrazole hybrids with in vivo therapeutic potential against cancers. Eur J Med Chem 2023; 257:115495. [PMID: 37209450 DOI: 10.1016/j.ejmech.2023.115495] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Chemotherapeutics occupy a pivotal role in the medication of different types of cancers, but the prevalence and mortality rates of cancer remain high. The drug resistance and low specificity of current available chemotherapeutics are the main barriers for the effective cancer chemotherapy, evoking an immediate need for the development of novel anticancer agents. Pyrazole is a highly versatile five-membered heterocycle with two adjacent nitrogen atoms and possesses remarkable therapeutic effects and robust pharmacological potency. The pyrazole derivatives especially pyrazole hybrids have demonstrated potent in vitro and in vivo efficacies against cancers through multiple mechanisms, inclusive of apoptosis induction, autophagy regulation, and cell cycle disruption. Moreover, several pyrazole hybrids such as crizotanib (pyrazole-pyridine hybrid), erdafitinib (pyrazole-quinoxaline hybrid) and ruxolitinib (pyrazole-pyrrolo [2,3-d]pyrimidine hybrid) have already been approved for the cancer therapy, revealing that pyrazole hybrids are useful scaffolds to develop novel anticancer agents. The purpose of this review is to summarize the current scenario of pyrazole hybrids with potential in vivo anticancer efficacy along with mechanisms of action, toxicity, and pharmacokinetics, covering papers published in recent 5 years (2018-present), to facilitate further rational exploitation of more effective candidates.
Collapse
Affiliation(s)
- Zhi Xu
- Industry Innovation & Research and Development Institute of Zhumadian, Huanghuai University, Zhumadian, 463000, China.
| | - Yafei Zhuang
- Industry Innovation & Research and Development Institute of Zhumadian, Huanghuai University, Zhumadian, 463000, China
| | - Qingtai Chen
- College of Chemistry Pharmaceutical Engineering, Huanghuai University, Zhumadian, 463000, China
| |
Collapse
|
7
|
Dual inhibition of CHK1/FLT3 enhances cytotoxicity and overcomes adaptive and acquired resistance in FLT3-ITD acute myeloid leukemia. Leukemia 2023; 37:539-549. [PMID: 36526736 DOI: 10.1038/s41375-022-01795-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
FLT3 inhibitors (FLT3i) are widely used for the treatment of acute myeloid leukemia (AML), but adaptive and acquired resistance remains a primary challenge. Inhibitors simultaneously blocking adaptive and acquired resistance are highly demanded. Here, we observed the potential of CHK1 inhibitors to synergistically improve the therapeutic effect of FLT3i in FLT3-mutated AML cells. Notably, the combination overcame adaptive resistance. The simultaneous targeting of FLT3 and CHK1 kinases may overcome acquired and adaptive resistance. A dual FLT3/CHK1 inhibitor 30 with a good oral PK profile was identified. Mechanistic studies indicated that 30 inhibited FLT3 and CHK1, downregulated the c-Myc pathway and further activated the p53 pathway. Functional studies showed that 30 was more selective against cells with various FLT3 mutants, overcame adaptive resistance in vitro, and effectively inhibited resistant FLT3-ITD AML in vivo. Moreover, 30 showed favorable druggability without significant blood toxicity or myelosuppression and exhibited a good oral PK profile with a T1/2 over 12 h in beagles. These findings support the targeting of FLT3 and CHK1 as a novel strategy for overcoming adaptive and acquired resistance to FLT3i therapy in AML and suggest 30 as a potential clinical candidate.
Collapse
|
8
|
Deng M, Wang P, Long X, Xu G, Wang C, Li J, Zhou Y, Liu T. Design, Synthesis, and Biological Evaluation of 2-Aminothiazole Derivatives as Novel Checkpoint Kinase 1 (CHK1) Inhibitors. ChemMedChem 2023; 18:e202200664. [PMID: 36732891 DOI: 10.1002/cmdc.202200664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/19/2023] [Indexed: 02/04/2023]
Abstract
A series of 2-aminothiazole derivatives were designed, synthesized on the basis of bioisosterism strategy and evaluated for their CHK1 inhibitory activity. Most of them exhibited potent CHK1 inhibition, and excellent antiproliferative activity against MV-4-11 and Z-138 cell lines. Systematic structure-activity relationship (SAR) efforts led to the discovery of a promising compound 8 n, which showed potent CHK1 inhibitory activity with IC50 value of 4.25±0.10 nM, excellent antiproliferative activity against MV-4-11 and Z-138 cells with IC50 value of 42.10±5.77 nM and 24.16±6.67 nM, respectively, as well as moderate oral exposure (AUC(0-t) =1076.25 h ⋅ ng/mL) in mice. Additionally, treatment of MV-4-11 cells with compound 8 n for 2 h led to robust inhibition of CHK1 autophosphorylation on serine 296. Furthermore, kinase selectivity assay revealed that 8 n displayed acceptable selectivity toward 15 kinases. These results demonstrated that compound 8 n may be a promising potential anticancer agent for further development.
Collapse
Affiliation(s)
- Minjie Deng
- College of Pharmaceutical Sciences, ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang University, 310058, Hangzhou, P. R. China
| | - Peipei Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 210023, Nanjing, P. R. China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P. R. China
| | - Xiubing Long
- Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, 200131, Shanghai, P. R. China
| | - Gaoya Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 210023, Nanjing, P. R. China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P. R. China
| | - Chang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P. R. China
| | - Jia Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 210023, Nanjing, P. R. China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 528400, Zhongshan, P. R. China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, 264117, Yantai, P. R. China
| | - Yubo Zhou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 210023, Nanjing, P. R. China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 528400, Zhongshan, P. R. China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P. R. China
| | - Tao Liu
- College of Pharmaceutical Sciences, ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang University, 310058, Hangzhou, P. R. China.,Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, Zhejiang University, 310058, Hangzhou, P. R. China
| |
Collapse
|
9
|
Shen Z, Zhuang W, Li K, Guo Y, Qu B, Chen S, Gao J, Liu J, Xu L, Dong X, Che J, Li Q. Identification of Novel Covalent XPO1 Inhibitors Based on a Hybrid Virtual Screening Strategy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082543. [PMID: 35458742 PMCID: PMC9024667 DOI: 10.3390/molecules27082543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022]
Abstract
Nuclear export protein 1 (XPO1), a member of the nuclear export protein-p (Karyopherin-P) superfamily, regulates the transport of “cargo” proteins. To facilitate this important process, which is essential for cellular homeostasis, XPO1 must first recognize and bind the cargo proteins. To inhibit this process, small molecule inhibitors have been designed that inhibit XPO1 activity through covalent binding. However, the scaffolds for these inhibitors are very limited. While virtual screening may be used to expand the diversity of the XPO1 inhibitor skeleton, enormous computational resources would be required to accomplish this using traditional screening methods. In the present study, we report the development of a hybrid virtual screening workflow and its application in XPO1 covalent inhibitor screening. After screening, several promising XPO1 covalent molecules were obtained. Of these, compound 8 performed well in both tumor cell proliferation assays and a nuclear export inhibition assay. In addition, molecular dynamics simulations were performed to provide information on the mode of interaction of compound 8 with XPO1. This research has identified a promising new scaffold for XPO1 inhibitors, and it demonstrates an effective and resource-saving workflow for identifying new covalent inhibitors.
Collapse
Affiliation(s)
- Zheyuan Shen
- Department of Urology, Rui’an People’s Hospital, the Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China;
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China; (S.C.); (J.G.); (X.D.)
| | - Weihao Zhuang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Kang Li
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai 222000, China;
| | - Yu Guo
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Bingxue Qu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Sikang Chen
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China; (S.C.); (J.G.); (X.D.)
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Jian Gao
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China; (S.C.); (J.G.); (X.D.)
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Jing Liu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China;
| | - Xiaowu Dong
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China; (S.C.); (J.G.); (X.D.)
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
| | - Jinxin Che
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China; (S.C.); (J.G.); (X.D.)
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (Y.G.); (B.Q.); (J.L.)
- Correspondence: (J.C.); (Q.L.)
| | - Qimeng Li
- Department of Urology, Rui’an People’s Hospital, the Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China;
- Correspondence: (J.C.); (Q.L.)
| |
Collapse
|
10
|
Downregulation of c-Myc expression confers sensitivity to CHK1 inhibitors in hematologic malignancies. Acta Pharmacol Sin 2022; 43:220-228. [PMID: 33782542 PMCID: PMC8724279 DOI: 10.1038/s41401-021-00652-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/12/2021] [Indexed: 01/03/2023]
Abstract
Checkpoint kinase 1 inhibitors (CHK1i) have shown impressive single-agent efficacy in treatment of certain tumors, as monotherapy or potentiators of chemotherapy in clinical trials, but the sensitive tumor types and downstream effectors to dictate the therapeutic responses to CHK1i remains unclear. In this study we first analyzed GDSC (Genomics of Drug Sensitivity in Cancer) and DepMap database and disclosed that hematologic malignancies (HMs) were relatively sensitive to CHK1i or CHK1 knockdown. This notion was confirmed by examining PY34, a new and potent in-house selective CHK1i, which exhibited potent anti-HM effect in vitro and in vivo, as single agent. We demonstrated that the downregulation of c-Myc and its signaling pathway was the common transcriptomic profiling response of sensitive HM cell lines to PY34, whereas overexpressing c-Myc could partially rescue the anticancer effect of PY34. Strikingly, we revealed the significant correlations between downregulation of c-Myc and cell sensitivity to PY34 in 17 HM cell lines and 39 patient-derived cell (PDC) samples. Thus, our results demonstrate that HMs are more sensitive to CHK1i than solid tumors, and c-Myc downregulation could represent the CHK1i efficacy in HMs.
Collapse
|
11
|
Highly efficient synthesis of pharmaceutically relevant chiral 3-N-substituted-azacyclic alcohols using two enantiocomplementary short chain dehydrogenases. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2021.108300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
12
|
Jin T, Xu L, Wang P, Hu X, Zhang R, Wu Z, Du W, Kan W, Li K, Wang C, Zhou Y, Li J, Liu T. Discovery and Development of a Potent, Selective, and Orally Bioavailable CHK1 Inhibitor Candidate: 5-((4-((3-Amino-3-methylbutyl)amino)-5-(trifluoromethyl)pyrimidin-2-yl)amino)picolinonitrile. J Med Chem 2021; 64:15069-15090. [PMID: 34665631 DOI: 10.1021/acs.jmedchem.1c00994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Checkpoint kinase 1 (CHK1) plays an important role in the DNA damage response pathway, being a potential anti-cancer drug target. In this study, we used a strategy for trifluoromethyl substitution to obtain orally bioavailable CHK1 inhibitors to overcome the limitations of lead compound 1, which can only be administered intravenously. After detailed investigation, we identified compound 6c as an oral CHK1 inhibitor, which demonstrated a considerably higher plasma exposure in mice. Compound 6c also showed good kinase selectivity. Moreover, it exhibited a significant antiproliferative effect in MV-4-11 cells singly and a synergistic effect in combination with gemcitabine in HT-29, A549, and RPMI-8226 cells. Additionally, compound 6c could inhibit tumor growth in the MV-4-11 xenograft mouse model. The combination of 6c and gemcitabine exhibited synergistic effect in the HT-29 xenograft mouse model. Thus, compound 6c was found to be a selective and oral potential anticancer CHK1 inhibitor.
Collapse
Affiliation(s)
- Tingting Jin
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Lei Xu
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Peipei Wang
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaobei Hu
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Runyuan Zhang
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Zhiqi Wu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Wenxin Du
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Weijuan Kan
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Li
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Chang Wang
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yubo Zhou
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Jia Li
- State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangdong 528400, China
| | - Tao Liu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| |
Collapse
|
13
|
Arshad M, Khan MS, Nami SAA, Ahmad SI, Kashif M, Anjum A. Synthesis, characterization, biological, and molecular docking assessment of bioactive 1,3-thiazolidin-4-ones fused with 1-(pyrimidin-2-yl)-1H-imidazol-4-yl) moieties. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2021. [DOI: 10.1007/s13738-020-02144-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Jin T, Wang P, Long X, Jiang K, Song P, Wu W, Xu G, Zhou Y, Li J, Liu T. Design, Synthesis, and Biological Evaluation of Orally Bioavailable CHK1 Inhibitors Active against Acute Myeloid Leukemia. ChemMedChem 2021; 16:1477-1487. [PMID: 33591599 DOI: 10.1002/cmdc.202000882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/02/2021] [Indexed: 11/06/2022]
Abstract
Checkpoint kinase 1 (CHK1) is a central component in DNA damage response and has emerged as a target for antitumor therapeutics. Herein, we describe the design, synthesis, and biological evaluation of a novel series of potent diaminopyrimidine CHK1 inhibitors. The compounds exhibited moderate to potent CHK1 inhibition and could suppress the proliferation of malignant hematological cell lines. The optimized compound 13 had a CHK1 IC50 value of 7.73±0.74 nM, and MV-4-11 cells were sensitive to it (IC50 =0.035±0.007 μM). Furthermore, compound 13 was metabolically stable in mouse liver microsomes in vitro and displayed moderate oral bioavailability in vivo. Moreover, treatment of MV-4-11 cells with compound 13 for 2 h led to robust inhibition of CHK1 autophosphorylation on serine 296. Based on these biochemical results, we consider compound 13 to be a promising CHK1 inhibitor and potential anticancer therapeutic agent.
Collapse
Affiliation(s)
- Tingting Jin
- College of Pharmaceutical Sciences, ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Peipei Wang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
| | - Xiubing Long
- College of Pharmaceutical Sciences, ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Kailong Jiang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China.,University of Chinese Academy of Sciences, No. 19 A Yuquan Road, Beijing, 100049, P. R. China
| | - Pinrao Song
- Shanghai Jemincare Pharmaceuticals Co. Ltd, Jemincare Group Research Institute, 1118 Halei Road, Shanghai, 201203, P. R. China
| | - Wenbiao Wu
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China.,University of Chinese Academy of Sciences, No. 19 A Yuquan Road, Beijing, 100049, P. R. China
| | - Gaoya Xu
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
| | - Yubo Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China.,University of Chinese Academy of Sciences, No. 19 A Yuquan Road, Beijing, 100049, P. R. China.,Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, P. R. China
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China.,University of Chinese Academy of Sciences, No. 19 A Yuquan Road, Beijing, 100049, P. R. China.,Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, P. R. China
| | - Tao Liu
- College of Pharmaceutical Sciences, ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
15
|
Chen X, Wu X, Gao J, Ying H, Dong X, Che J, Shen Z. Identification of new IDH2 R140Q inhibitors by discriminatory analysis-based molecular docking and biological evaluation. Arch Pharm (Weinheim) 2020; 354:e2000063. [PMID: 33184958 DOI: 10.1002/ardp.202000063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/29/2020] [Accepted: 10/24/2020] [Indexed: 11/07/2022]
Abstract
Isocitrate dehydrogenase 2 (IDH2) is a key enzyme in the regulation of cell metabolism. Its mutated type can lead to the accumulation of 2-hydroxyglutarate, which is often related to malignancies such as acute myeloid leukemia. Therefore, it is necessary to find new inhibitors targeting mutant IDH2. Discriminatory analysis-based molecular docking was employed to screen the ChemDiv compound library, which resulted in the identification of three new IDH2R140Q inhibitors with moderate-to-good IC50 values. Among them, compounds 1 and 3 displayed good selectivity against other mutant or wild-type IDH proteins. The most potent compound 1, bearing the [1,2,4]triazolo[1,5-a]pyrimidin scaffold, was subjected to dynamic simulations to provide more information on the binding mode with IDH2R140Q , providing structural clues to further optimize compound 1 as a new mutant IDH2 inhibitor.
Collapse
Affiliation(s)
- Xiaoyun Chen
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianmin Wu
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Gao
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Huazhou Ying
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhijian Shen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Che J, Feng R, Gao J, Yu H, Weng Q, He Q, Dong X, Wu J, Yang B. Evaluation of Artificial Intelligence in Participating Structure-Based Virtual Screening for Identifying Novel Interleukin-1 Receptor Associated Kinase-1 Inhibitors. Front Oncol 2020; 10:1769. [PMID: 33014870 PMCID: PMC7494739 DOI: 10.3389/fonc.2020.01769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/07/2020] [Indexed: 01/06/2023] Open
Abstract
Interleukin-1 receptor associated kinase-1 (IRAK1) exhibits important roles in inflammation, infection, and autoimmune diseases; however, only a few inhibitors have been discovered. In this study, at first, a discriminatory structure-based virtual screening (SBVS) was employed, but only one active compound (compound 1, IC50 = 2.25 μM) was identified. The low hit rate (2.63%) which derives from the weak discriminatory power of docking among high-scored molecules was observed in our virtual screening (VS) process for IRAK1 inhibitor. Furthermore, an artificial intelligence (AI) method, which employed a support vector machine (SVM) model, integrated information of molecular docking, pharmacophore scoring and molecular descriptors was constructed to enhance the traditional IRAK1-VS protocol. Using AI, it was found that VS of IRAK1 inhibitors excluded by over 50% of the inactive compounds, which could significantly improve the prediction accuracy of the SBVS model. Moreover, four active molecules (two of which exhibited comparative IC50 with compound 1) were accurately identified from a set of highly similar candidates. Amongst, compounds with better activity exhibited good selectivity against IRAK4. The AI assisted workflow could serve as an effective tool for enhancement of SBVS.
Collapse
Affiliation(s)
- Jinxin Che
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ruiwei Feng
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China.,Real Doctor AI Research Centre, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Gao
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hongyun Yu
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China.,Real Doctor AI Research Centre, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qinjie Weng
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China.,Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China.,Cancer Center of Zhejiang University, Hangzhou, China
| | - Jian Wu
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China.,Real Doctor AI Research Centre, School of Medicine, Zhejiang University, Hangzhou, China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Chen Z, Wang C, Lei C, Feng X, Li C, Jung SY, Qin J, Chen J. Phosphoproteomics Analysis Reveals a Potential Role of CHK1 in Regulation of Innate Immunity through IRF3. J Proteome Res 2020; 19:2264-2277. [DOI: 10.1021/acs.jproteome.9b00829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Zhen Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Chao Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Caoqi Lei
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Xu Feng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Chen Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|