1
|
Huang D, Zhang J, Yang X, Tan X, Chai T, Ma L, Zhao B, Chen Y, Yang J, Zhang Y. Structure-guided Development of Novel Benzothiophene Derivatives as PLK1-PBD Inhibitors. LETT DRUG DES DISCOV 2024; 21:2163-2174. [DOI: 10.2174/1570180820666230602153458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 05/29/2025]
Abstract
Background:
Polo-like kinase 1 (PLK1), a validated target for tumor therapy, plays a key role
in mitosis and is over-expressed in many tumors. In addition to its N-terminal kinase domain, PLk1 also
harbors a C-terminal polo-box domain (PBD).
Objective:
A candidate based on PLK1-PBD was developed as a promising compound for future development.
Methods:
Seventeen small molecule PLK1-PBD inhibitors were designed, synthesized and evaluated for
PLK1-PBD inhibitory activities by fluorescence polarization (FP) assay. The compounds with better inhibitory
activities were further assessed for their anti-proliferative activities using a CCK-8 method.
Results:
The inhibitory rates of compounds 7a, 7d, 14a, 14d, 14e and 14f exceeded 98%. The IC50 values
of compounds 7d, 14d, 14e, and 14f were 0.73 μM, 0.67 μM, 0.89 μM and 0.26 μM, proving better than
MCC1019. Compound 14f showed the best inhibitory activity (IC50: 0.26 μM) and antiproliferative activities
against three cancer cell lines (HeLa, HepG2 and MG63). Especially, compound 14f also exhibited
acceptable safety profiles in the human ether-a-go-go related gene (hERG) and normal cell tests. The
results of docking and prediction studies indicated that compound 14f had a high binding affinity to the
target, with good drug-like absorption, distribution, metabolism, elimination, and toxicity (ADMET)
properties.
Conclusion:
Compound 14f can be a promising compound for future development.
Collapse
Affiliation(s)
- Daowei Huang
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| | - Jing Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing, 100050, China
| | - Xiaocong Yang
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| | - Xiangduan Tan
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Tingting Chai
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| | - Lindeng Ma
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| | - Bingyang Zhao
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| | - Ying Chen
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| | - Jixia Yang
- School of
Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yue Zhang
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang,
050018, China
| |
Collapse
|
2
|
Stafford JM, Wyatt MD, McInnes C. Inhibitors of the PLK1 polo-box domain: drug design strategies and therapeutic opportunities in cancer. Expert Opin Drug Discov 2023; 18:65-81. [PMID: 36524399 DOI: 10.1080/17460441.2023.2159942] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Polo Like Kinase 1 (PLK1) is a key regulator of mitosis and its overexpression is frequently observed in a wide variety of human cancers, while often being associated with poor survival rates. Therefore, it is considered a potential and attractive target for cancer therapeutic development. The Polo like kinase family is characterized by the presence of a unique C terminal polobox domain (PBD) involved in regulating kinase activity and subcellular localization. Among the two functionally essential, druggable sites with distinct properties that PLK1 offers, targeting the PBD presents an alternative approach for therapeutic development. AREAS COVERED Significant progress has been made in progressing from the peptidic PBD inhibitors first identified, to peptidomimetic and recently drug-like small molecules. In this review, the rationale for targeting the PBD over the ATP binding site is discussed, along with recent progress, challenges, and outlook. EXPERT OPINION The PBD has emerged as a viable alternative target for the inhibition of PLK1, and progress has been made in using compounds to elucidate mechanistic aspects of activity regulation and in determining roles of the PBD. Studies have resulted in proof of concept of in vivo efficacy suggesting promise for PBD binders in clinical development.
Collapse
Affiliation(s)
- Jessy M Stafford
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Michael D Wyatt
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Campbell McInnes
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
3
|
Yan H, Liu Z, Yan G, Liu X, Liu X, Wang Y, Chen Y. A robust high-throughput fluorescence polarization assay for rapid screening of SARS-CoV-2 papain-like protease inhibitors. Virology 2022; 574:18-24. [PMID: 35870326 PMCID: PMC9287753 DOI: 10.1016/j.virol.2022.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 12/01/2022]
Abstract
The global scourge of COVID-19 is a serious threat to public health, but effective therapies remain very limited for this disease. Therefore, the discovery of novel antiviral agents is urgently needed to fight against COVID-19. In the lifecycle of SARS-CoV-2, the causing pathogen of COVID-19, papain-like protease (PLpro) is responsible for the cleavage of polyprotein into functional units as well as immune evasion of vaccines. Hence, PLpro has been regarded as an attractive target to develop antiviral agents. Herein, we first developed a robust and simple sandwich-like fluorescence polarization (FP) screening assay for the discovery of PLpro inhibitors, and identified anacardic acid as a novel competitive inhibitor against PLpro in vitro with an IC50 value of 24.26 ± 0.4 μM. This reliable FP screening assay could provide a prospective avenue for rapid discovery of antiviral agents targeting PLpro in a large-scale screening.
Collapse
Affiliation(s)
- Haohao Yan
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, China
| | - Zhicheng Liu
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, China
| | - Gangan Yan
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, China
| | - Xiaoli Liu
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, China
| | - Xiaoping Liu
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, China
| | - Yanchang Wang
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, United States.
| | - Yunyu Chen
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, China.
| |
Collapse
|
4
|
Gunasekaran P, Yim MS, Ahn M, Soung NK, Park JE, Kim J, Bang G, Shin SC, Choi J, Kim M, Kim HN, Lee YH, Chung YH, Lee K, EunKyeong Kim E, Jeon YH, Kim MJ, Lee KR, Kim BY, Lee KS, Ryu EK, Bang JK. Development of a Polo-like Kinase-1 Polo-Box Domain Inhibitor as a Tumor Growth Suppressor in Mice Models. J Med Chem 2020; 63:14905-14920. [PMID: 33142063 PMCID: PMC8919061 DOI: 10.1021/acs.jmedchem.0c01451] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Polo-like kinase-1 (Plk1) plays a key role in mitosis and has been identified as an attractive anticancer drug target. Plk1 consists of two drug-targeting sites, namely, N-terminal kinase domain (KD) and C-terminal polo-box domain (PBD). As KD-targeting inhibitors are associated with severe side effects, here we report on the pyrazole-based Plk1 PBD inhibitor, KBJK557, which showed a remarkable in vitro anticancer effect by inducing Plk1 delocalization, mitotic arrest, and apoptosis in HeLa cells. Further, in vivo optical imaging analysis and antitumorigenic activities in mouse xenograft models demonstrate that KBJK557 preferentially accumulates in cancer cells and selectively inhibits cancer cell proliferation. Pharmacokinetic profiles and partition coefficients suggest that KBJK557 was exposed in the blood and circulated through the organs with an intermediate level of clearance (t1/2, 7.73 h). The present investigation offers a strategy for specifically targeting cancer using a newly identified small-molecule inhibitor that targets the Plk1 PBD.
Collapse
Affiliation(s)
- Pethaiah Gunasekaran
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
| | - Min Su Yim
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
- Department of Bio-analytical Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Mija Ahn
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Jung-Eun Park
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Jaehi Kim
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
| | - Geul Bang
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Chung-Buk 363-883, Republic of Korea
| | - Sang Chul Shin
- Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Joonhyeok Choi
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
| | - Minkyoung Kim
- College of Pharmacy, Dongguk University, 52 Dongguk-ro, Ilsandong-gu, Goyang 10320, Republic of Korea
| | - Hak Nam Kim
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
| | - Young-Ho Lee
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
- Department of Bio-analytical Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Young-Ho Chung
- Drug & Disease Target Research Team, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University, 52 Dongguk-ro, Ilsandong-gu, Goyang 10320, Republic of Korea
| | - Eunice EunKyeong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Young-Ho Jeon
- Laboratory of Biochemistry and Structural Biology, College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Min Ju Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Bo-Yeon Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Kyung S Lee
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Eun Kyoung Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
- Department of Bio-analytical Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Jeong Kyu Bang
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Chung Buk 28119, Republic of Korea
- Department of Bio-analytical Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
5
|
Alverez CN, Park JE, Toti KS, Xia Y, Krausz KW, Rai G, Bang JK, Gonzalez FJ, Jacobson KA, Lee KS. Identification of a New Heterocyclic Scaffold for Inhibitors of the Polo-Box Domain of Polo-like Kinase 1. J Med Chem 2020; 63:14087-14117. [PMID: 33175530 PMCID: PMC7769008 DOI: 10.1021/acs.jmedchem.0c01669] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As a mitotic-specific target widely deregulated in various human cancers, polo-like kinase 1 (Plk1) has been extensively explored for anticancer activity and drug discovery. Although multiple catalytic domain inhibitors were tested in preclinical and clinical studies, their efficacies are limited by dose-limiting cytotoxicity, mainly from off-target cross reactivity. The C-terminal noncatalytic polo-box domain (PBD) of Plk1 has emerged as an attractive target for generating new protein-protein interaction inhibitors. Here, we identified a 1-thioxo-2,4-dihydro-[1,2,4]triazolo[4,3-a]quinazolin-5(1H)-one scaffold that efficiently inhibits Plk1 PBD but not its related Plk2 and Plk3 PBDs. Structure-activity relationship studies led to multiple inhibitors having ≥10-fold higher inhibitory activity than the previously characterized Plk1 PBD-specific phosphopeptide, PLHSpT (Kd ∼ 450 nM). In addition, S-methyl prodrugs effectively inhibited mitotic progression and cell proliferation and their metabolic stability was determined. These data describe a novel class of small-molecule inhibitors that offer a promising avenue for future drug discovery against Plk1-addicted cancers.
Collapse
Affiliation(s)
- Celeste N Alverez
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Jung-Eun Park
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kiran S Toti
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yangliu Xia
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kristopher W Krausz
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ganesha Rai
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Jeong K Bang
- Division of Magnetic Resonance, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Frank J Gonzalez
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kyung S Lee
- Chemistry Section, Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
6
|
Chen Y, Fu Z, Li D, Yue Y, Liu X. Optimizations of a novel fluorescence polarization-based high-throughput screening assay for β-catenin/LEF1 interaction inhibitors. Anal Biochem 2020; 612:113966. [PMID: 32956692 DOI: 10.1016/j.ab.2020.113966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/21/2020] [Accepted: 09/15/2020] [Indexed: 01/20/2023]
Abstract
Aberrant activation of the Wnt/β-catenin signaling pathway is prominent in the development and metastasis of non-small cell lung cancer (NSCLC). Highly effective inhibition of this pathway highlights a therapeutic avenue against NSCLC. Moreover, β-catenin/LEF1 interaction regulates β-catenin nuclear transport as well as the transcriptions of the key oncogenes in Wnt/β-catenin signaling pathway. Therefore, interruption of this interaction would be a promising therapeutic strategy for NSCLC metastasis. To date, no economical and rapid high-throughput screening (HTS) assay has been reported for the discovery of β-catenin/LEF1 interaction inhibitors. In this study, we developed a novel fluorescence polarization (FP)-based HTS assay to identify β-catenin/LEF1 interaction inhibitors. The FITC-LEF1 sequence, incubation time, temperature, and DMSO resistance were optimized, and then a high Z' factor of 0.77 was achieved. A pilot screening of a natural product library via this established FP screening assay identified sanguinarine analogues as potential β-catenin/LEF1 interaction inhibitors. GST pull-down and surface plasmon resonance (SPR) assay demonstrated that β-catenin/LEF1 interaction is a potential anticancer target of sanguinarine in vitro. This newly developed FP screening assay will be vital for the rapid discovery of novel Wnt inhibitors targeting β-catenin/LEF1 interaction.
Collapse
Affiliation(s)
- Yunyu Chen
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, 241002, China
| | - Zhenghao Fu
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, 241002, China
| | - Dongsheng Li
- Department of Medicinal Chemistry, Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, 201203, China
| | - Yuhuan Yue
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, 130122, China.
| | - Xiaoping Liu
- Institute for Drug Screening and Evaluation, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
7
|
Liang H, Liu H, Kuang Y, Chen L, Ye M, Lai L. Discovery of Targeted Covalent Natural Products against PLK1 by Herb-Based Screening. J Chem Inf Model 2020; 60:4350-4358. [PMID: 32407091 DOI: 10.1021/acs.jcim.0c00074] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Natural products (NPs) are a rich source of drug discovery, and some of them act by covalently binding to the targets. Recently, targeted covalent natural product (TCNP) design has gained considerable attention since this approach offers significant benefits in improving biological efficacy and decreasing the off-target side effects. However, most of the known TCNPs were discovered by chance. Rational approaches for a systematic screen of TCNPs are much needed. Here, we developed a combined computational and experimental approach to carry out herb-based screening to identify TCNPs against proper cysteine residues in the target proteins. The herb-based TCNP screening approach (HB-TCNP) starts from a druggable pocket and cysteine residue prediction, followed by virtual screening of a covalent NP database and herb-based mapping to identify candidate herbs for experimental validation. Herbs with time-dependent activity are selected, and their NPs are experimentally tested to further screen covalent NPs. We have successfully applied HB-TCNP to screen anti-PLK1 herbs and NPs with high efficacy. Cys67 and Cys133 in the ATP binding pocket of PLK1 were used in the search. Five herbs were tested and exhibited PLK1 inhibition activity to some extent, among which Scutellaria baicalensis showed the most potent activity with time dependency. Further experimental studies showed that the main active compounds in Scutellaria baicalensis, baicalein and baicalin, covalently bind PLK1 through Cys133. Our study provided an efficient way to rationally design TCNPs and to make better use of herb medicines. The Cys133 residue in PLK1 serves as a novel covalent site for further drug discovery against PLK1.
Collapse
Affiliation(s)
- Hao Liang
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Hongbo Liu
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yi Kuang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Limin Chen
- Peking-Tsinghua Center for Life Sciences at Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Luhua Lai
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences at Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
8
|
Abdelfatah S, Fleischer E, Klinger A, Wong VKW, Efferth T. Identification of inhibitors of the polo-box domain of polo-like kinase 1 from natural and semisynthetic compounds. Invest New Drugs 2020; 38:1-9. [PMID: 30877426 DOI: 10.1007/s10637-019-00752-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/25/2019] [Indexed: 12/30/2022]
Abstract
PLK1 has an important role in the regulation of cell cycle and represents an important target for cancer treatment. This enzyme belongs to the Polo-like kinases family, which is characterized by a regulatory domain named Polo-box domain (PBD). Rather than regular kinase inhibitors, this domain provides high selectivity to PLK1. Here, we report on four novel PLK1 PBD inhibitors identified by cytotoxicity screening and fluorescence polarization assay of a chemical library of natural and semisynthetic compounds. These compounds revealed two- to three-fold higher selectivity to the PDB of PLK1 than to those of the related family members, PLK2 and PLK3. These four substances inhibited tumor cell growth of sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 leukemia cells. The tested compounds increased the apoptotic cell fraction, which indicates apoptosis as a major mechanism of cell death. Cell cycle analysis showed compound (5) arrested the cell cycle of CCRF-CEM cells in the G2/M phase, while the other three molecules ((compound (3), compound (4), and compound (6)) exerted pronounced cytotoxicity with an increase of cells in the sub-G1 population. Molecular docking was performed for the understanding of ligand-protein interaction, the tested candidates showed strong binding affinity to PLK1 PBD. In conclusion, we identified four new chemical scaffolds that may serve as lead compounds for the development of selective PLK1 inhibitors in the future.
Collapse
Affiliation(s)
- Sara Abdelfatah
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, 55128, Mainz, Germany
| | | | | | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, 55128, Mainz, Germany.
| |
Collapse
|
9
|
Abdeldayem A, Raouf YS, Constantinescu SN, Moriggl R, Gunning PT. Advances in covalent kinase inhibitors. Chem Soc Rev 2020; 49:2617-2687. [DOI: 10.1039/c9cs00720b] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This comprehensive review details recent advances, challenges and innovations in covalent kinase inhibition within a 10 year period (2007–2018).
Collapse
Affiliation(s)
- Ayah Abdeldayem
- Department of Chemical & Physical Sciences
- University of Toronto
- Mississauga
- Canada
- Department of Chemistry
| | - Yasir S. Raouf
- Department of Chemical & Physical Sciences
- University of Toronto
- Mississauga
- Canada
- Department of Chemistry
| | | | - Richard Moriggl
- Institute of Animal Breeding and Genetics
- University of Veterinary Medicine
- 1210 Vienna
- Austria
| | - Patrick T. Gunning
- Department of Chemical & Physical Sciences
- University of Toronto
- Mississauga
- Canada
- Department of Chemistry
| |
Collapse
|
10
|
Malde AK, Hill TA, Iyer A, Fairlie DP. Crystal Structures of Protein-Bound Cyclic Peptides. Chem Rev 2019; 119:9861-9914. [DOI: 10.1021/acs.chemrev.8b00807] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alpeshkumar K. Malde
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Timothy A. Hill
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Abishek Iyer
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David P. Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
11
|
Rubner S, Schubert S, Berg T. Poloxin-2HT+: changing the hydrophobic tag of Poloxin-2HT increases Plk1 degradation and apoptosis induction in tumor cells. Org Biomol Chem 2019; 17:3113-3117. [PMID: 30848278 DOI: 10.1039/c9ob00080a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We report the hydrophobically-tagged Plk1 PBD inhibitor Poloxin-2HT+, which selectively degrades the tumor target Plk1 and induces apoptosis in human tumor cells with higher potency than the hydrophobically-tagged inhibitor Poloxin-2HT. Our data provide further evidence that hydrophobically tagged inhibitors of protein-protein interactions can target and destroy disease-relevant proteins.
Collapse
Affiliation(s)
- Stefan Rubner
- Institute of Organic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany.
| | | | | |
Collapse
|
12
|
High expression of CDK1 and BUB1 predicts poor prognosis of pancreatic ductal adenocarcinoma. Gene 2019; 701:15-22. [PMID: 30898709 DOI: 10.1016/j.gene.2019.02.081] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most common causes of cancer-related death. Increasing evidence suggests that cell cycle dysregulation is one of the hallmarks of cancer. In this study, by using the GEO database, we predicted the cell cycle-related protein CDK1 and BUB1 to be significantly overexpressed in PDAC tissues. Thus, this study aimed to investigate the clinical pathological significance of CDK1 and BUB1 in PDAC. METHODS To explore the role of CDK1 and BUB1 in PDAC progression and evaluate their prognostic value, we investigated the expression patterns of CDK1 and BUB1 by using immunohistochemical staining in 99 PDAC and 71 normal pancreatic tissues with complete pathological parameters and survival data. RESULTS CDK1 and BUB1 were significantly overexpressed in PDAC tissues. The expression of CDK1 was correlated with tumor size and histological grade, and the expression of BUB1 was correlated with the tumor size of PDAC. With regard to survival, a high expression of either CDK1 or BUB1 was correlated with a short survival of PDAC patients. Additionally, PDAC patients with a concurrent high expression of CDK1 and BUB1 showed the shortest survival. CONCLUSIONS Our study demonstrated that CDK1 and BUB1 may play a role in PDAC progression and could be prognostic biomarkers for PDAC patients.
Collapse
|
13
|
Rubner S, Scharow A, Schubert S, Berg T. Selective Degradation of Polo-like Kinase 1 by a Hydrophobically Tagged Inhibitor of the Polo-Box Domain. Angew Chem Int Ed Engl 2018; 57:17043-17047. [PMID: 30351497 DOI: 10.1002/anie.201809640] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/09/2018] [Indexed: 12/18/2022]
Abstract
Hydrophobic tagging (HT) of bioactive compounds can induce target degradation via the proteasomal pathway. The first application of hydrophobic tagging to an existing inhibitor of protein-protein interactions is now presented. We developed Poloxin-2HT by fusing an adamantyl tag to Poloxin-2, an inhibitor of the polo-box domain of the protein kinase Plk1, which is a target for tumor therapy. Poloxin-2HT selectively reduced the protein levels of Plk1 in HeLa cells and had a significantly stronger effect on cell viability and the induction of apoptosis than the untagged PBD inhibitor Poloxin-2. The change in cellular phenotype associated with the addition of the hydrophobic tag to Poloxin-2 demonstrated that Poloxin-2HT targets Plk1 in living cells. Our data validate hydrophobic tagging of selective inhibitors of protein-protein interactions as a novel strategy to target and destroy disease-relevant proteins.
Collapse
Affiliation(s)
- Stefan Rubner
- Leipzig University, Institute of Organic Chemistry, Johannisallee 29, 04103, Leipzig, Germany
| | - Andrej Scharow
- Leipzig University, Institute of Organic Chemistry, Johannisallee 29, 04103, Leipzig, Germany
| | - Sabine Schubert
- Leipzig University, Institute of Organic Chemistry, Johannisallee 29, 04103, Leipzig, Germany
| | - Thorsten Berg
- Leipzig University, Institute of Organic Chemistry, Johannisallee 29, 04103, Leipzig, Germany
| |
Collapse
|
14
|
IMB-T130 targets 3-dehydroquinate synthase and inhibits Mycobacterium tuberculosis. Sci Rep 2018; 8:17439. [PMID: 30487577 PMCID: PMC6262012 DOI: 10.1038/s41598-018-35701-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 11/02/2018] [Indexed: 11/08/2022] Open
Abstract
The anti-tuberculosis (TB) agent IMB-T130 was speculated to be a multi-target compound. In this research, we found that IMB-T130 inhibits the catalytic activity of Mycobacterium tuberculosis 3-dehydroquinate synthase (MtDHQS), the enzyme in the second step of the shikimate pathway. IMB-T130 was identified as a selective inhibitor of MtDHQS with an IC50 value of 0.87 μg/mL. The interaction between the compound and protein was analysed by surface plasmon resonance and circular dichroism. Based on the in silico molecular docking results, the essential amino acids in the binding pocket were then confirmed by site-directed mutagenesis. Overexpression of DHQS reduced the antibacterial activity of IMB-T130 in cells, verifying that DHQS is the target of IMB-T130. IMB-T130 inhibited standard and drug-resistant M. tuberculosis strains by targeting DHQS. Our findings improve our understanding of MtDHQS and make it to be a potential target for new anti-TB drug discovery.
Collapse
|
15
|
Rubner S, Scharow A, Schubert S, Berg T. Selective Degradation of Polo‐like Kinase 1 by a Hydrophobically Tagged Inhibitor of the Polo‐Box Domain. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201809640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Stefan Rubner
- Leipzig University Institute of Organic Chemistry Johannisallee 29 04103 Leipzig Germany
| | - Andrej Scharow
- Leipzig University Institute of Organic Chemistry Johannisallee 29 04103 Leipzig Germany
| | - Sabine Schubert
- Leipzig University Institute of Organic Chemistry Johannisallee 29 04103 Leipzig Germany
| | - Thorsten Berg
- Leipzig University Institute of Organic Chemistry Johannisallee 29 04103 Leipzig Germany
| |
Collapse
|
16
|
Abstract
Mitosis is controlled by reversible protein phosphorylation involving specific kinases and phosphatases. A handful of major mitotic protein kinases, such as the cyclin B-CDK1 complex, the Aurora kinases, and Polo-like kinase 1 (PLK1), cooperatively regulate distinct mitotic processes. Research has identified proteins and mechanisms that integrate these kinases into signaling cascades that guide essential mitotic events. These findings have important implications for our understanding of the mechanisms of mitotic regulation and may advance the development of novel antimitotic drugs. We review collected evidence that in vertebrates, the Aurora kinases serve as catalytic subunits of distinct complexes formed with the four scaffold proteins Bora, CEP192, INCENP, and TPX2, which we deem "core" Aurora cofactors. These complexes and the Aurora-PLK1 cascades organized by Bora, CEP192, and INCENP control crucial aspects of mitosis and all pathways of spindle assembly. We compare the mechanisms of Aurora activation in relation to the different spindle assembly pathways and draw a functional analogy between the CEP192 complex and the chromosomal passenger complex that may reflect the coevolution of centrosomes, kinetochores, and the actomyosin cleavage apparatus. We also analyze the roles and mechanisms of Aurora-PLK1 signaling in the cell and centrosome cycles and in the DNA damage response.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, Saint-Petersburg 197758, Russian Federation.
| | | |
Collapse
|
17
|
Sun J, Liu HY, Xu RF, Zhu HL. Identification of nitroimidazole-oxime derivatives targeting the polo-box domain of polo-like kinase 1. Bioorg Med Chem 2017; 25:6581-6588. [PMID: 29100732 DOI: 10.1016/j.bmc.2017.10.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/15/2017] [Accepted: 10/25/2017] [Indexed: 11/23/2022]
Abstract
Recent progress in the development of small molecular skeleton-derived polo-like kinase (PLK1) catalytic domain (KD) inhibitors has led to the synthesis of multiple ligands with high binding affinity. However, few systematic analyses have been conducted to identify key PLK1-PBD domain and characterize their interactions with potent PLK1 inhibitors. Therefore, we designed a series of PLK1-PBD inhibitors with an in silico scaffold modification strategy. A docking simulation combined with a primary screen in vitro were performed to filter for the lead compound, which was then substituted, synthesized and evaluated by a variety of bioassays. The biological profile of 4v suggests that this compound may be developed as a potential anticancer agent.
Collapse
Affiliation(s)
- Juan Sun
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China; Elion Nature Biological Technology Co., Ltd, Nanjing 210046, PR China.
| | - Han-Yu Liu
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China
| | - Ruo-Fei Xu
- Shandong Experimental High School, Jinan 250001, PR China
| | - Hai-Liang Zhu
- Elion Nature Biological Technology Co., Ltd, Nanjing 210046, PR China
| |
Collapse
|
18
|
Stahl D, Braun M, Gentles AJ, Lingohr P, Walter A, Kristiansen G, Gütgemann I. Low BUB1 expression is an adverse prognostic marker in gastric adenocarcinoma. Oncotarget 2017; 8:76329-76339. [PMID: 29100315 PMCID: PMC5652709 DOI: 10.18632/oncotarget.19357] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/19/2017] [Indexed: 01/12/2023] Open
Abstract
Gastric adenocarcinomas are associated with a poor prognosis due to the fact that the tumor has often metastasized by the time of diagnosis and prognostic markers are urgently needed to tailor treatment. We examined the expression of the mitotic spindle checkpoint protein BUB1 (budding uninhibited by benzimidazoles 1) and Ki-67 protein expression by immunohistochemistry in 218 patients with primary gastric adenocarcinomas. Tumors with low frequency of BUB1 expression were associated with larger tumor size (pT) (p < 0.001), higher incidence of lymph node metastases (pN) (p = 0.027), distant metastases (pM) (p = 0.006) and higher UICC stage (p < 0.001). Furthermore, BUB1 expression was inversely correlated with residual tumor stage (p = 0.038). Abundant BUB1 protein expression correlated with frequent Ki-67 protein expression (p < 0.001) and low BUB1 expression was associated with shorter survival (p < 0.001). Univariate and multivariate analyses confirmed BUB1 to be an independent prognostic marker in gastric cancer (p = 0.021).
Collapse
Affiliation(s)
- David Stahl
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Martin Braun
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Andrew J Gentles
- Center for Cancer Systems Biology (CCSB), Stanford University, Stanford, California, USA
| | - Philipp Lingohr
- Department of General, Visceral, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Adeline Walter
- Department of Gynecology and Obstetrics, University Hospital Bonn, Bonn, Germany
| | | | - Ines Gütgemann
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
19
|
Park JE, Hymel D, Burke TR, Lee KS. Current progress and future perspectives in the development of anti-polo-like kinase 1 therapeutic agents. F1000Res 2017; 6:1024. [PMID: 28721210 PMCID: PMC5497816 DOI: 10.12688/f1000research.11398.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2017] [Indexed: 12/21/2022] Open
Abstract
Although significant levels of side effects are often associated with their use, microtubule-directed agents that primarily target fast-growing mitotic cells have been considered to be some of the most effective anti-cancer therapeutics. With the hope of developing new-generation anti-mitotic agents with reduced side effects and enhanced tumor specificity, researchers have targeted various proteins whose functions are critically required for mitotic progression. As one of the highly attractive mitotic targets, polo-like kinase 1 (Plk1) has been the subject of an extensive effort for anti-cancer drug discovery. To date, a variety of anti-Plk1 agents have been developed, and several of them are presently in clinical trials. Here, we will discuss the current status of generating anti-Plk1 agents as well as future strategies for designing and developing more efficacious anti-Plk1 therapeutics.
Collapse
Affiliation(s)
- Jung-Eun Park
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Hymel
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Kyung S Lee
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
20
|
Archambault V, Normandin K. Several inhibitors of the Plk1 Polo-Box Domain turn out to be non-specific protein alkylators. Cell Cycle 2017; 16:1220-1224. [PMID: 28521657 PMCID: PMC5499904 DOI: 10.1080/15384101.2017.1325043] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 12/16/2022] Open
Abstract
For almost a decade, there has been much interest in the development of chemical inhibitors of Polo-like kinase 1 (Plk1) protein interactions. Plk1 is a master regulator of the cell division cycle that controls numerous substrates. It is a promising target for cancer drug development. Inhibitors of the kinase domain of Plk1 had some success in clinical trials. However, they are not perfectly selective. In principle, Plk1 can also be inhibited by interfering with its protein interaction domain, the Polo-Box Domain (PBD). Selective chemical inhibitors of the PBD would constitute tools to probe for PBD-dependent functions of Plk1 and could be advantageous in cancer therapy. The discovery of Poloxin and thymoquinone as PBD inhibitors indicated that small, cell-permeable chemical inhibitors could be identified. Other efforts followed, including ours, reporting additional molecules capable of blocking the PBD. It is now clear that, unfortunately, most of these compounds are non-specific protein alkylators (defined here as groups covalently added via a carbon) that have little or no potential for the development of real Plk1 PBD-specific drugs. This situation should be minded by biologists potentially interested in using these compounds to study Plk1. Further efforts are needed to develop selective, cell-permeable PBD inhibitors.
Collapse
Affiliation(s)
- Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montreal, Canada
| | - Karine Normandin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| |
Collapse
|