1
|
Shin YZ, Yum YA, Bae ES, Jarhad DB, Aswar VR, Tripathi SK, Kwon EJ, Kim YJ, Kim M, Lee SK, Jeong LS, Cha HJ. Targeting HASPIN in gemcitabine-resistant pancreatic cancer cells by lead optimization of thioadenosine analogue. Biomed Pharmacother 2025; 188:118135. [PMID: 40378773 DOI: 10.1016/j.biopha.2025.118135] [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: 03/17/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Despite multiple kinase inhibitors having been developed for cancer therapy, mitotic kinases remain difficult to target with small molecules due to severe adverse effects on proliferating normal cells. Recently, HASPIN, a mitotic kinase responsible for histone H3 phosphorylation, has emerged as a promising cancer-specific target. In this study, we synthesized a novel thioadenosine analogue, LJ5157, based on the structure of the previously developed HASPIN inhibitor LJ4827. In silico transcriptome analysis of pancreatic cancer patient data from The Cancer Genome Atlas identified HASPIN as not only a cancer-specific target but also a potential key player in overcoming gemcitabine resistance. To evaluate the therapeutic potential of LJ5157, we tested its efficacy in pancreatic cancer cells, particularly gemcitabine-resistant Panc-1 (GR) cells. The inhibitor exhibited potent anti-cancer activity, effectively suppressing the growth of GR cells, which showed more dysregulated cell cycle progression and greater proportion of polyploid cells compared to wild-type Panc-1 cells. Furthermore, it demonstrated superior efficacy in reducing the mitotic population of polyploid GR cells, which correlated with significant tumor growth inhibition in a GR-cell-derived xenograft model. Further optimization of LJ4827 led to development of LJ5242, an analogue with enhanced selectivity for HASPIN and improved cell cycle inhibitory potency. These findings highlight HASPIN inhibition as a promising strategy for targeting chemoresistant pancreatic cancer and further identify thioadenosine as a valuable pharmacophore for developing clinically viable HASPIN inhibitors.
Collapse
Affiliation(s)
- Yoon-Ze Shin
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yun A Yum
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Dnyandev B Jarhad
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Vikas R Aswar
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | | | - Eun-Ji Kwon
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yun-Jeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Minjae Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Lak-Shin Jeong
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Future Medicine Co., Ltd, Seongnam, Gyeonggi-do, Republic of Korea.
| | - Hyuk-Jin Cha
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Abd El Hadi SR, Eldinary MA, Ghith A, Haffez H, Salman A, Sayed GA. Unravelling the potency of the 4-oxo-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carbonitrile scaffold with S-arylamide hybrids as PIM-1 kinase inhibitors: synthesis, biological activity and in silico studies. RSC Med Chem 2025:d5md00021a. [PMID: 40162200 PMCID: PMC11951167 DOI: 10.1039/d5md00021a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
PIM-1 is a type of serine/threonine kinase that plays a crucial role in controlling several vital processes, including proliferation and apoptosis. New synthetic S-amide tetrahydropyrimidinone derivatives were designed and synthesized as PIM-1 inhibitors with potential anticancer activity. Several biochemical assays were performed for anticancer assessment, including PIM-1 inhibitory assays, MTT, apoptosis and cell cycle, gene expression analysis, c-MYC analysis, and ATPase inhibitory assays. Compounds (8c, 8d, 8g, 8h, 8k, and 8l) exhibited strong in vitro broad antiproliferative activity against MCF-7, DU-145, and PC-3, with a relatively higher SI index suggesting minimal cytotoxicity to normal cells. Furthermore, these compounds induced mixed late apoptosis and necrosis with cell cycle arrest at the G2/M phase. Moreover, compounds 8b, 8f, 8g, 8k, and 8l showed potent inhibitory action against PIM-1 kinase, with corresponding IC50 values of 660, 909, 373, 518, and 501 nM. In silico prediction studies of physiochemical properties, molecular dynamics, and induced fit docking studies were performed for these compounds to explain their potent biological activity. In conclusion, new pyrimidinone compounds (8c, 8d, 8g, 8h, 8k, and 8l) exhibit potential PIM-1 inhibitory activity and can be used as promising scaffolds for further optimization of new leads with selective PIM-inhibitors and anticancer activity.
Collapse
Affiliation(s)
- Soha R Abd El Hadi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City Cairo 11829 Egypt
| | - Manar A Eldinary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City Cairo 11829 Egypt
| | - Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital Woodville South SA 5011 Australia
- Robinson Research Institute, University of Adelaide Adelaide SA 5006 Australia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt Cairo 11835 Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University P.O. Box 11795 Cairo Egypt
- Center of Scientific Excellence "Helwan Structural Biology Research (HSBR)", Helwan University Cairo 11795 Egypt
| | - Aya Salman
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829 Cairo Egypt
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829 Cairo Egypt
| |
Collapse
|
3
|
Teli G, Maji L, Pal R, Maheshwari N, Purawarga Matada GS, Chawla PA, Chawla V. Recent advancements in mechanistic research, therapeutic potential, and structure-activity relationships of aurora kinase inhibitors in cancer therapies. Bioorg Chem 2025; 154:107976. [PMID: 39603069 DOI: 10.1016/j.bioorg.2024.107976] [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: 09/20/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Aurora kinases (AURKs)-a family of serine/threonine protein kinases consisting of AURK-A, AURK-B, and AURK-C, are critical regulators of chromosomal segregation, centrosome maturation, and cytokinesis during the cell cycle. Each kinase is activated via phosphorylation at unique threonine residues: Thr288 (AURK-A), Thr232 (AURK-B), and Thr195 (AURK-C). Activation of AURK-A and AURK-B through phosphorylation triggers a series of downstream signaling pathways, including RalA, NF-κB, p53, PLK1, BRCA1/BRCA2, H2AX, and Kif2C, as well as multiple transmembrane kinase receptors. Dysregulation of these pathways has been implicated in cancer development and progression, positioning AURKs as pivotal targets for anticancer drug research. Inhibition of AURKs has demonstrated significant efficacy in tumor growth suppression and induction of cancer cell death, thereby focusing recent research on the development of potent AURK inhibitors. This review provides an in-depth exploration of AURK inhibitors, discussing their biological activities, structure-activity relationships, selectivity profiles, and mechanisms of action. Notably, compounds 6, 27, and 16 exhibit potent AURK-A inhibition with IC50 values of 1.7 nM, 11.83 nM, and 15 nM, respectively. Similarly, compounds 28, 16, and 7 demonstrate strong AURK-B inhibitory activity, with IC50 values of 10.5 nM, 12 nM, and 14.09 nM, respectively. This comprehensive overview aims to support medicinal chemists in developing more potent, selective, and safe AURK inhibitors as potential anticancer therapeutics.
Collapse
Affiliation(s)
- Ghanshyam Teli
- School of Pharmacy, Sangam University, NH-79, Atoon, Bhilwara, 311001, Rajasthan, India; University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab 151203 India
| | - Lalmohan Maji
- Department of Pharmaceutical Chemistry, Tarifa Memorial Institute of Pharmacy, Murshidabad, 742166, West Bengal, India
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, Karnataka, India
| | - Neelesh Maheshwari
- School of Pharmacy, Sangam University, NH-79, Atoon, Bhilwara, 311001, Rajasthan, India
| | | | - Pooja A Chawla
- University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab 151203 India.
| | - Viney Chawla
- University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab 151203 India.
| |
Collapse
|
4
|
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
|
5
|
Zhang Y, Xie Z, Xiao F, Yu J, Fan Z, Sun S, Shi J, Fu Z, Li X, Wang D, Zheng M, Luo X. Prediction of Multi-Pharmacokinetics Property in Multi-Species: Bayesian Neural Network Stacking Model with Uncertainty. Mol Pharm 2024. [PMID: 39508275 DOI: 10.1021/acs.molpharmaceut.4c00406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Pharmacokinetic (PK) properties of a drug are vital attributes influencing its therapeutic effectiveness, playing an important role in the drug development process. Focusing on the difficult task of predicting PK parameters, we compiled an extensive data set comprising parameters across multiple species. Building upon this groundwork, we introduced the PKStack ensemble model to predict PK parameters across diverse species. PKStack integrates a variety of base models and includes uncertainty in its predictions. We also manually collected PK data from animals as an external test set. We predicted a total of 45 tasks for nine PK parameters in five species, and in general, the prediction accuracy was better for intravenous injections, including parameters such as human Vd (R2 = 0.72, RMSE = 0.31), human CL (R2 = 0.52, RMSE = 0.32), and others. In addition to predictive accuracy, we also considered the interpretability of the results and the definition of the model's application domain. Based on the findings, our model has great potential for practical applications in drug discovery.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyin Xie
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fu Xiao
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Yu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Information Science and Technology, Shanghai Tech University, Shanghai 201210, China
- Lingang Laboratory, Shanghai 200031, China
| | - Zhehuan Fan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shihui Sun
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiangshan Shi
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zunyun Fu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaomin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
6
|
Zhang J, Zeng X, Guo Q, Sheng Z, Chen Y, Wan S, Zhang L, Zhang P. Small cell lung cancer: emerging subtypes, signaling pathways, and therapeutic vulnerabilities. Exp Hematol Oncol 2024; 13:78. [PMID: 39103941 DOI: 10.1186/s40164-024-00548-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024] Open
Abstract
Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by early metastasis, rapid tumor growth and poor prognosis. In recent decades, the epidemiology, initiation and mutation characteristics of SCLC, as well as abnormal signaling pathways contributing to its progression, have been widely studied. Despite extensive investigation, fewer drugs have been approved for SCLC. Recent advancements in multi-omics studies have revealed diverse classifications of SCLC that are featured by distinct characteristics and therapeutic vulnerabilities. With the accumulation of SCLC samples, different subtypes of SCLC and specific treatments for these subtypes were further explored. The identification of different molecular subtypes has opened up novel avenues for the treatment of SCLC; however, the inconsistent and uncertain classification of SCLC has hindered the translation from basic research to clinical applications. Therefore, a comprehensives review is essential to conclude these emerging subtypes and related drugs targeting specific therapeutic vulnerabilities within abnormal signaling pathways. In this current review, we summarized the epidemiology, risk factors, mutation characteristics of and classification, related molecular pathways and treatments for SCLC. We hope that this review will facilitate the translation of molecular subtyping of SCLC from theory to clinical application.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Xiaoping Zeng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qiji Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Zhenxin Sheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yan Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Shiyue Wan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lele Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
7
|
Chang CP, Yeh TK, Chen CT, Wang WP, Chen YT, Tsai CH, Chen YF, Ke YY, Wang JY, Chen CP, Hsieh TC, Wu MH, Huang CL, Chen YP, Zhuang H, Chi YH. Discovery of a Long Half-Life AURKA Inhibitor to Treat MYC-Amplified Solid Tumors as a Monotherapy and in Combination with Everolimus. Mol Cancer Ther 2024; 23:766-779. [PMID: 38592383 DOI: 10.1158/1535-7163.mct-23-0602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/20/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024]
Abstract
Aurora kinase inhibitors, such as alisertib, can destabilize MYC-family oncoproteins and have demonstrated compelling antitumor efficacy. In this study, we report 6K465, a novel pyrimidine-based Aurora A inhibitor, that reduces levels of c-MYC and N-MYC oncoproteins more potently than alisertib. In an analysis of the antiproliferative effect of 6K465, the sensitivities of small cell lung cancer (SCLC) and breast cancer cell lines to 6K465 were strongly associated with the protein levels of c-MYC and/or N-MYC. We also report DBPR728, an acyl-based prodrug of 6K465 bearing fewer hydrogen-bond donors, that exhibited 10-fold improved oral bioavailability. DBPR728 induced durable tumor regression of c-MYC- and/or N-MYC-overexpressing xenografts including SCLC, triple-negative breast cancer, hepatocellular carcinoma, and medulloblastoma using a 5-on-2-off or once-a-week dosing regimen on a 21-day cycle. A single oral dose of DBPR728 at 300 mg/kg induced c-MYC reduction and cell apoptosis in the tumor xenografts for more than 7 days. The inhibitory effect of DBPR728 at a reduced dosing frequency was attributed to its uniquely high tumor/plasma ratio (3.6-fold within 7 days) and the long tumor half-life of active moiety 6K465. Furthermore, DBPR728 was found to synergize with the mTOR inhibitor everolimus to suppress c-MYC- or N-MYC-driven SCLC. Collectively, these results suggest DBPR728 has the potential to treat cancers overexpressing c-MYC and/or N-MYC.
Collapse
Affiliation(s)
- Chun-Ping Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Wan-Ping Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Yen-Ting Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Chia-Hua Tsai
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Yan-Fu Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Yi-Yu Ke
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Tsung-Chih Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Mine-Hsine Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Ya-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Hong Zhuang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
8
|
Lee H, Kim E, Hwang N, Yoo J, Nam Y, Hwang I, Park JG, Park SE, Chung KS, Won Chung H, Song C, Ji MJ, Park HM, Lee IK, Lee KT, Joo Roh E, Hur W. Discovery of N-benzylbenzamide-based allosteric inhibitors of Aurora kinase A. Bioorg Med Chem 2024; 102:117658. [PMID: 38460487 DOI: 10.1016/j.bmc.2024.117658] [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: 12/13/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/11/2024]
Abstract
Aurora kinases (AurkA/B/C) regulate the assembly of bipolar mitotic spindles and the fidelity of chromosome segregation during mitosis, and are attractive therapeutic targets for cancers. Numerous ATP-competitive AurkA inhibitors have been developed as potential anti-cancer agents. Recently, a few allosteric inhibitors have been reported that bind to the allosteric Y-pocket within AurkA kinase domain and disrupt the interaction between AurkA and its activator TPX2. Herein we report a novel allosteric AurkA inhibitor (6h) of N-benzylbenzamide backbone. Compound 6h suppressed the both catalytic activity and non-catalytic functions of AurkA. The inhibitory activity of 6h against AurkA (IC50 = 6.50 μM) was comparable to that of the most potent allosteric AurkA inhibitor AurkinA. Docking analysis against the Y-pocket revealed important pharmacophores and interactions that were coherent with structure-activity relationship. In addition, 6h suppressed DNA replication in G1-S phase, which is a feature of allosteric inhibition of AurA. Our current study may provide a useful insight in designing potent allosteric AurkA inhibitors.
Collapse
Affiliation(s)
- Hyomin Lee
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Biomedical Science and Technology, UST KIST School, Seoul 02792, Republic of Korea
| | - Euijung Kim
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Narae Hwang
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jesik Yoo
- Division of Biomedical Science and Technology, UST KIST School, Seoul 02792, Republic of Korea; Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Yunju Nam
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Injeoung Hwang
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; HY-KIST Bioconvergence, Hanyang University, Seoul 04763, Republic of Korea
| | - Jin-Gyeong Park
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Eun Park
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hwan Won Chung
- Computational Science Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Chiman Song
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Mi-Jung Ji
- Advanced Analysis Data Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hyun-Mee Park
- Advanced Analysis Data Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - In-Kyun Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eun Joo Roh
- Division of Biomedical Science and Technology, UST KIST School, Seoul 02792, Republic of Korea; Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Wooyoung Hur
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; HY-KIST Bioconvergence, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
9
|
Kwon Y, Choi Y, Kim M, Jo H, Jeong MS, Jung HS, Jeoung D. HDAC6-MYCN-CXCL3 axis mediates allergic inflammation and is necessary for allergic inflammation-promoted cellular interactions. Mol Immunol 2024; 166:1-15. [PMID: 38176167 DOI: 10.1016/j.molimm.2023.12.007] [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: 09/01/2023] [Revised: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
Histone deacetylase 6 (HDAC6) has been shown to play an important role in allergic inflammation. This study hypothesized that novel downstream targets of HDAC6 would mediate allergic inflammation. Experiments employing HDAC6 knock out C57BL/6 mice showed that HDAC6 mediated passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). Antigen stimulation increased expression of N-myc (MYCN) and CXCL3 in an HDAC6-dependent manner in the bone marrow-derived mast cells. MYCN and CXCL3 were necessary for both PCA and PSA. The role of early growth response 3 (EGR3) in the regulation of HDAC6 expression has been reported. ChIP assays showed EGR3 as a direct regulator of MYCN. miR-34a-5p was predicted to be a negative regulator of MYCN. Luciferase activity assays showed miR-34a-5p as a direct regulator of MYCN. miR-34a-5p mimic negatively regulated PCA and PSA. MYCN decreased miR-34a-5p expression in antigen-stimulated rat basophilic leukemia cells (RBL2H3). MYCN was shown to bind to the promoter sequence of CXCL3. In an IgE-independent manner, recombinant CXCL3 protein increased expression of HDAC6, MYCN, and β-hexosaminidase activity in RBL2H3 cells. Mouse recombinant CXCL3 protein enhanced the angiogenic potential of the culture medium of RBL2H3. CXCL3 was necessary for the enhanced angiogenic potential of the culture medium of antigen-stimulated RBL2H3. The culture medium of RBL2H3 was able to induce M2 macrophage polarization in a CXCL3-dependent manner. Recombinant CXCL3 protein also increased the expression of markers of M2 macrophage. Thus, the identification of the novel role of HDAC6-MYCN-CXCL3 axis can help better understand the pathogenesis of anaphylaxis.
Collapse
Affiliation(s)
- Yoojung Kwon
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Yunji Choi
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Misun Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, South Korea
| | - Hyein Jo
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Myeong Seon Jeong
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
10
|
Guo H, Li W, Guo Y, Chen N, Cui J. Molecular classification of small cell lung cancer subtypes: Characteristics, prognostic factors, and clinical translation. Chin Med J (Engl) 2024; 137:130-139. [PMID: 37660289 PMCID: PMC10798698 DOI: 10.1097/cm9.0000000000002693] [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: 02/15/2023] [Indexed: 09/04/2023] Open
Abstract
ABSTRACT Small cell lung cancer (SCLC) is a highly malignant tumor with a very poor prognosis; therefore, more effective treatments are urgently needed for patients afflicted with the disease. In recent years, emerging molecular classifications based on key transcription factors of SCLC have provided more information on the tumor pathophysiology, metastasis, immune microenvironment, and acquired therapeutic resistance and reflected the intertumoral heterogeneity of the various SCLC phenotypes. Additionally, advances in genomics and single-cell sequencing analysis have further revealed the high intratumoral heterogeneity and plasticity of the disease. Herein, we review and summarize these recent lines of evidence and discuss the possible pathogenesis of SCLC.
Collapse
Affiliation(s)
| | | | | | | | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
11
|
Jaber AM, Al-Mahadeen MM, Al-Qawasmeh RA, Taha MO. Synthesis, anticancer evaluation and docking studies of novel adamantanyl-1,3,4-oxadiazol hybrid compounds as Aurora-A kinase inhibitors. Med Chem Res 2023; 32:2394-2404. [DOI: 10.1007/s00044-023-03145-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/19/2023] [Indexed: 07/10/2024]
|
12
|
Jadhav M, Sankhe K, Bhandare RR, Edis Z, Bloukh SH, Khan TA. Synthetic Strategies of Pyrimidine-Based Scaffolds as Aurora Kinase and Polo-like Kinase Inhibitors. Molecules 2021; 26:5170. [PMID: 34500603 PMCID: PMC8434097 DOI: 10.3390/molecules26175170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/19/2022] Open
Abstract
The past few decades have witnessed significant progress in anticancer drug discovery. Small molecules containing heterocyclic moieties have attracted considerable interest for designing new antitumor agents. Of these, the pyrimidine ring system is found in multitude of drug structures, and being the building unit of DNA and RNA makes it an attractive scaffold for the design and development of anticancer drugs. Currently, 22 pyrimidine-containing entities are approved for clinical use as anticancer drugs by the FDA. An exhaustive literature search indicates several publications and more than 59 patents from the year 2009 onwards on pyrimidine derivatives exhibiting potent antiproliferative activity. These pyrimidine derivatives exert their activity via diverse mechanisms, one of them being inhibition of protein kinases. Aurora kinase (AURK) and polo-like kinase (PLK) are protein kinases involved in the regulation of the cell cycle. Within the numerous pyrimidine-based small molecules developed as anticancer agents, this review focuses on the pyrimidine fused heterocyclic compounds modulating the AURK and PLK proteins in different phases of clinical trials as anticancer agents. This article aims to provide a comprehensive overview of synthetic strategies for the preparation of pyrimidine derivatives and their associated biological activity on AURK/PLK. It will also present an overview of the synthesis of the heterocyclic-2-aminopyrimidine, 4-aminopyrimidine and 2,4-diaminopyrimidine scaffolds, and one of the pharmacophores in AURK/PLK inhibitors is described systematically.
Collapse
Affiliation(s)
- Mrunal Jadhav
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India; (M.J.); (K.S.)
| | - Kaksha Sankhe
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India; (M.J.); (K.S.)
| | - Richie R. Bhandare
- Department of Pharmaceutical Chemistry, College of Pharmacy & Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
| | - Zehra Edis
- Department of Pharmaceutical Chemistry, College of Pharmacy & Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
| | - Samir Haj Bloukh
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
- Department of Clinical Sciences, College of Pharmacy & Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Tabassum Asif Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India; (M.J.); (K.S.)
| |
Collapse
|