1
|
Zhang Y, Liu X, Li Z, Wang X, Tang C. Development of 9H-purine scaffold as novel CDK2 inhibitors: Design, synthesis, and biological evaluation. Bioorg Med Chem Lett 2025; 122:130166. [PMID: 40057135 DOI: 10.1016/j.bmcl.2025.130166] [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: 12/10/2023] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Cyclin-dependent kinase 2 (CDK2), a crucial regulator in multiple oncogenic signaling pathways, has emerged as a promising target for the development of innovative anticancer therapies and overcoming resistance to CDK4/6 inhibitors. In this study, three series of compounds were designed and synthesized, using the CDK2 inhibitor fadraciclib (CYC065) as the lead compound, with 9H-purine as the core structure. The design incorporated reported structure-activity relationship data and utilized computer-aided drug design techniques. Compounds in series 1 explored the binding mode between the ATP ribose binding site in CDK2 and C2 substituents, while compounds in series 2 and 3 validated the feasibility of modifying the specific binding region with different substituents and investigated the effects of filling the CDK2 hydrophobic pocket at the N9 position with alkyl substituents. Three compounds, 1f, 2e, and 3a, demonstrated remarkable activity against CDK2-cyclin E2. Notably, 3a exhibited the most potent effect, with a CDK2-cyclin E2 IC50 value of 6.0 ± 0.1 nM, an MV4-11 IC50 value of 489.2 ± 0.2 nM, and excellent selectivity for CDK2. This study evaluated the impact of substitutions at the 2, 6, and 9 positions of the purine ring on the activity of CDK2 small molecule inhibitors. The findings offer a theoretical foundation for future research, broadening the structural diversity and scope of CDK2 inhibitor studies.
Collapse
Affiliation(s)
- Yan Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Xiya Liu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Ziming Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Xia Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Chunlei Tang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
2
|
Kang Y, Han B, Kong Y, Liu S, Zhang N. Efficacy and safety of first-line CDK4/6 inhibitors plus AI therapy for patients with HR +/HER2- advanced breast cancer: a network meta-analysis. BMC Cancer 2025; 25:843. [PMID: 40340824 PMCID: PMC12063220 DOI: 10.1186/s12885-025-14194-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND CDK4/6 inhibitors plus aromatase inhibitors (AI) significantly improve the therapeutic effect of initial treatment for HR + /HER2- advanced breast cancer. However, there is a lack of head-to-head randomized controlled trials involving the four CDK4/6 inhibitors in current clinical treatments. This article aims to compare the efficacy and safety of the four CDK4/6 inhibitors in previously untreated HR + /HER2- advanced breast cancer for a better clinical medication selection. METHODS We performed a systematic search on databases published up to May 19, 2024 in the PubMed, Embase, and Cochrane library, and we focused on the data of phase II/III trials that met inclusion criteria. Pooled data on progression-free survival (PFS), objective response rate (ORR), clinical benefit rate (CBR), all adverse events (AE), and 3/4 adverse events were analyzed using the fixed-effect consistency models. RESULTS Dalpiciclib plus AI showed the best survival benefit in PFS (SUCRA value 77.9%) for all patients. In terms of ORR and CBR, Abemaciclib plus AI were ranked the best benefit (89.3% and 68.9%, respectively). Furthermore, Abemaciclib plus AI was ranked at the top for prolonging PFS in majority of the subgroups. In terms of AEs and grade 3/4 AEs, dalpiciclib plus AI had the greatest probability (91.3% and 99.8%). Ribociclib plus AI had lowest adverse events (29.3%), and grade 3/4 adverse events of abemaciclib plus AI were the least (26.2%). CONCLUSION There was no statistically significant difference in PFS among the four CDK4/6 inhibitors. Dalpiciclib has the best therapeutic effect in PFS. Meanwhile, dalpiciclib has the highest risk of adverse events and the 3/4 adverse events incidence compared with the others.
Collapse
Affiliation(s)
- Yanrong Kang
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, 264001, China
| | - Bing Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, 250013, China
| | - Yongmei Kong
- Department of Body Radiotherapy, Shandong Second Province General Hospital, Jinan, Shandong, 250021, China
| | - Shuai Liu
- Department of Breast Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China.
| | - Nan Zhang
- Department of Breast Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China.
| |
Collapse
|
3
|
Alrouji M, Alshammari MS, Anwar S, Venkatesan K, Shamsi A. Mechanistic Roles of Transcriptional Cyclin-Dependent Kinases in Oncogenesis: Implications for Cancer Therapy. Cancers (Basel) 2025; 17:1554. [PMID: 40361480 PMCID: PMC12071579 DOI: 10.3390/cancers17091554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Cyclin-dependent kinases (CDKs) are pivotal in regulating cell cycle progression and transcription, making them crucial targets in cancer research. The two types of CDKs that regulate different biological activities are transcription-associated CDKs (e.g., CDK7, 8, 9, 12, and 13) and cell cycle-associated CDKs (e.g., CDK1, 2, 4, and 6). One characteristic of cancer is the dysregulation of CDK activity, which results in unchecked cell division and tumor expansion. Targeting transcriptional CDKs, which control RNA polymerase II activity and gene expression essential for cancer cell survival, has shown promise as a therapeutic approach in recent research. While research into selective inhibitors for transcriptional CDKs is ongoing, inhibitors that target CDK4/6, such as palbociclib and ribociclib, have demonstrated encouraging outcomes in treating breast cancer. CDK7, CDK8, and CDK9 are desirable targets for therapy since they have shown oncogenic roles in a variety of cancer types, such as colorectal, ovarian, and breast malignancies. Even with significant advancements, creating selective inhibitors with negligible off-target effects is still difficult. This review highlights the need for more research to optimize therapeutic strategies and improve patient outcomes by giving a thorough overview of the non-transcriptional roles of CDKs in cancer biology, their therapeutic potential, and the difficulties in targeting these kinases for cancer treatment.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Mohammed S. Alshammari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Saleha Anwar
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, Saudi Arabia
| |
Collapse
|
4
|
Azuma N, Yamaguchi Y, Tanaka T, Matsuzaka E, Saida Y, Yokoi T, Handa H, Hirayama J, Nishina H. INTS15, A Subunit of the Integrator Complex, Plays a Key Regulatory Role in Cell Cycle and Differentiation. Genes Cells 2025; 30:e70015. [PMID: 40194948 DOI: 10.1111/gtc.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025]
Abstract
We previously reported that Integrator complex subunit 15 (INTS15) is a causative gene for an autosomal-dominant eye disease named variable panocular malformations (VPMs) and that INTS15 stably interacts with the Integrator complex to support snRNA 3' end processing, thereby controlling mRNA splicing. Here we report another critical function of INTS15 in cell cycle control. HeLa cells and human iPS cells were engineered to overexpress INTS15 expression in a cumate-responsive manner and used to study its role in the regulation of cell cycle and differentiation. INTS15 activates the expression of p53 and p21 to induce G1 arrest when overexpressed. In in vitro differentiation of iPS cells, INTS15 promotes the formation of the three germ layers as well as differentiation into late retinal tissues. Meanwhile, INTS15 knockdown results in defects in G2/M progression and apoptosis. Moreover, INTS15 expression levels vary substantially by cell type and flactuate during the cell cycle. Thus, this study reveals a novel biological aspect of the Integrator complex and demonstrates its potential practical applications.
Collapse
Affiliation(s)
- Noriyuki Azuma
- Department of Ophthalmology and Laboratory for Visual Science, National Centre for Child Health and Development, Tokyo, Japan
- Department of Developmental and Regenerative Biology, Medical Research Institute, Institute of Science Tokyo, Tokyo, Japan
| | - Yuki Yamaguchi
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Taku Tanaka
- Department of Ophthalmology and Laboratory for Visual Science, National Centre for Child Health and Development, Tokyo, Japan
- Department of Developmental and Regenerative Biology, Medical Research Institute, Institute of Science Tokyo, Tokyo, Japan
| | - Emiko Matsuzaka
- Department of Ophthalmology and Laboratory for Visual Science, National Centre for Child Health and Development, Tokyo, Japan
| | - Yuki Saida
- Department of Ophthalmology and Laboratory for Visual Science, National Centre for Child Health and Development, Tokyo, Japan
| | - Tadashi Yokoi
- Department of Ophthalmology and Laboratory for Visual Science, National Centre for Child Health and Development, Tokyo, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Jun Hirayama
- Department of Developmental and Regenerative Biology, Medical Research Institute, Institute of Science Tokyo, Tokyo, Japan
- Department of Clinical Engineering, Faculty of Health Science, Komatsu University, Ishikawa, Japan
| | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Zhang X, Wang B, Qi X, Qian Z, Gao X, Cheng Y, Wang X. A Glutathione-Responsive System with Prodrug and Sensitization Strategies for Targeted Therapy of Glioma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2501620. [PMID: 40119786 DOI: 10.1002/smll.202501620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/08/2025] [Indexed: 03/24/2025]
Abstract
Glioblastoma represents a highly aggressive form of malignant tumor within the central nervous system. Although chemotherapy remains the primary therapeutic strategy, its efficacy is often limited. To overcome the limitations associated with chemotherapeutic agents, such as high toxicity and non-specific adverse effects, a novel nanoparticle system comprising cRGD-modified and glutathione (GSH)-responsive polymers, and PEG-ss-Dox and apatinib (AP) (PDOX-AP/cRGD-NPs) is developed. PDOX-AP/cRGD-NPs show effective penetration of the blood-brain barrier (BBB), facilitate targeted delivery to brain tumors, and exhibit controlled drug release. PDOX-AP/cRGD-NPs show more effect in reducing the viability of GL-261, U87-MG, and LN-229 cells, inhibiting clonogenicity, and suppressing anti-apoptotic protein expression than PDOX/cRGD-NPs or AP/cRGD-NPs. Additionally, PDOX-AP/cRGD-NPs substantially increase drug uptake, BBB penetration, apoptosis rates, and the proportion of cells in the G2 phase. In vivo experiments further reveal that cRGD-directed nanoparticles exhibit superior accumulation in glioma regions compared to their non-cRGD-modified counterparts. In the interim, PDOX-AP/cRGD-NPs demonstrate significant efficacy in suppressing both ectopic and orthotopic growth of GL-261 gliomas, as well as orthotopic LN-229 gliomas, thereby markedly extending the median survival duration. This study introduces a promising targeted co-delivery system for combination chemotherapy.
Collapse
Affiliation(s)
- Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xin Qi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
6
|
Hung C, Nguyen TTT, Poulikakos PI, Polsky D. Recent Developments in Targeting the Cell Cycle in Melanoma. Cancers (Basel) 2025; 17:1291. [PMID: 40282469 PMCID: PMC12025829 DOI: 10.3390/cancers17081291] [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: 01/22/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Melanoma is an aggressive cancer with rising incidence, particularly among older individuals. Despite advancements in targeted therapies for BRAF and MEK proteins and immunotherapies, many patients either fail to respond or develop resistance. For those progressing on immunotherapy, limited treatment options remain. The Cyclin D-CDK4/6-RB pathway is commonly dysregulated in melanoma, with up to 90% of cases showing alterations that activate it. Although targeting Cyclin-CDK complexes has shown promise in preclinical models, clinical responses have been suboptimal. This review explores the molecular mechanisms behind Cyclin-CDK dysregulation in melanoma and the challenges of targeting this pathway. It also discusses strategies to improve the efficacy of CDK4/6 inhibitors, including combination therapies to overcome resistance and enhance patient outcomes. Understanding these mechanisms can guide the development of more effective treatments for melanoma.
Collapse
Affiliation(s)
- Christie Hung
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA; (C.H.); (T.T.T.N.)
| | - Trang T. T. Nguyen
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA; (C.H.); (T.T.T.N.)
| | - Poulikos I. Poulikakos
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - David Polsky
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA; (C.H.); (T.T.T.N.)
| |
Collapse
|
7
|
Sun F, Ji C, Zhou X, Zhang Y, Cheng H, Ye Z. Targeting RACGAP1 suppresses growth hormone pituitary adenoma growth. Endocrine 2025; 88:234-248. [PMID: 39607642 DOI: 10.1007/s12020-024-04116-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
PURPOSE Growth hormone pituitary adenoma (GHPA) is a major subtype of pituitary adenoma (PA), with tumor enlargement and abnormal secretion of growth hormone (GH) often causing complications. Rac GTPase-activating protein 1 (RACGAP1), a member of the guanine triphosphatase-activating protein family, is highly overexpressed in multiple tumors and promotes tumor growth. However, the role of RACGAP1 in GHPA remains unelucidated. Besides, specific inhibitors targeting RACGAP1 have not yet been developed. In this study, we aimed to determine the expression and function of RACGAP1 in GHPA and identify effective inhibitors against RACGAP1. METHODS Immunohistochemistry was used to detect the expression of RACGAP1 in GHPA and normal pituitary tissues. The effect of RACGAP1 on cell proliferation, apoptosis, and cell cycle was evaluated by knockdown of RACGAP1 in GH3 cells in vitro and xenograft models of GHPA in vivo. The downstream mechanism of RACGAP1 was explored by RNA sequencing, bioinformatic analysis, and Western blot. Inhibitors targeting RACGAP1 were screened and verified through a structure-based virtual docking method, cell viability assays, and surface plasmon resonance (SPR) experiments. RESULTS RACGAP1 expression was increased in GHPA compared with normal pituitary tissues. Knocking down RACGAP1 suppressed cell growth in vitro and in vivo. Preliminary mechanism studies indicated that inhibition of RACGAP1 led to the upregulation of p21 and the downregulation of several genes involved in the cell cycle signaling pathway, such as Cyclin A, CDK1, and CDK2. Moreover, DB07268 was identified for the first time as an effective RACGAP1 inhibitor that could prominently restrain the proliferation of GH3 cells. CONCLUSION This study demonstrates that RACGAP1 plays a critical role in GHPA, highlighting the novel inhibitor DB07268 as a promising therapeutic approach.
Collapse
Affiliation(s)
- Feifan Sun
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
| | - Chenxing Ji
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
| | - Xiang Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Haixia Cheng
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, Shanghai, 200040, China.
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China.
- Shanghai Key laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China.
| |
Collapse
|
8
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
9
|
Sun J, Liang S, Liu X, Zhang S, Li M, Zhang Q, Chen J. Insights into the selectivity of a brain-penetrant CDK4/6 vs CDK1/2 inhibitor for glioblastoma used in multiple replica molecular dynamics simulations. J Biomol Struct Dyn 2025; 43:2223-2242. [PMID: 38112295 DOI: 10.1080/07391102.2023.2294175] [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: 07/27/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Cyclin dependent kinases (CDKs) play an important role in cell cycle regulation and their dysfunction is associated with many cancers. That is why CDKs have been attractive targets for the treatment of cancer. Glioblastoma is a cancer caused by the aberrant expression of CDK4/6, so exploring the mechanism of the selection of CDK4/6 toward inhibitors relative to the other family members CDK1/2 is essential. In this work, multiple replica molecular dynamics (MRMD) simulations, principal component analysis (PCA), free energy landscapes (FELs), molecular mechanics Poisson-Boltzmann/Generalized Born surface area (MM-PB/GBSA) and other methods were integrated to decipher the selectively binding mechanism of the inhibitor N1J to CDK4/6 and CDK1/2. Molecular electrostatic potential (MESP) analysis provides an explanation for the N1J selectivity. Residue-based free energy decomposition reveals that most of the hot residues are located at the same location of CDKs proteins, but the different types of residues in different proteins cause changes in binding energy, which is considered as a potential developmental direction to improve the selectivity of inhibitors to CDK4/6. These results provide insights into the source of inhibitor and CDK4/6 selectivity for the future development of more selective inhibitors.
Collapse
Affiliation(s)
- Jiahao Sun
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Shanshan Liang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Xinguo Liu
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Shaolong Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Meng Li
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Qinggang Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan, China
| |
Collapse
|
10
|
Glaviano A, Singh SK, Lee EHC, Okina E, Lam HY, Carbone D, Reddy EP, O'Connor MJ, Koff A, Singh G, Stebbing J, Sethi G, Crasta KC, Diana P, Keyomarsi K, Yaffe MB, Wander SA, Bardia A, Kumar AP. Cell cycle dysregulation in cancer. Pharmacol Rev 2025; 77:100030. [PMID: 40148026 DOI: 10.1016/j.pharmr.2024.100030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer is a systemic manifestation of aberrant cell cycle activity and dysregulated cell growth. Genetic mutations can determine tumor onset by either augmenting cell division rates or restraining normal controls such as cell cycle arrest or apoptosis. As a result, tumor cells not only undergo uncontrolled cell division but also become compromised in their ability to exit the cell cycle accurately. Regulation of cell cycle progression is enabled by specific surveillance mechanisms known as cell cycle checkpoints, and aberrations in these signaling pathways often culminate in cancer. For instance, DNA damage checkpoints, which preclude the generation and augmentation of DNA damage in the G1, S, and G2 cell cycle phases, are often defective in cancer cells, allowing cell division in spite of the accumulation of genetic errors. Notably, tumors have evolved to become dependent on checkpoints for their survival. For example, checkpoint pathways such as the DNA replication stress checkpoint and the mitotic checkpoint rarely undergo mutations and remain intact because any aberrant activity could result in irreparable damage or catastrophic chromosomal missegregation leading to cell death. In this review, we initially focus on cell cycle control pathways and specific functions of checkpoint signaling involved in normal and cancer cells and then proceed to examine how cell cycle control and checkpoint mechanisms can provide new therapeutic windows that can be exploited for cancer therapy. SIGNIFICANCE STATEMENT: DNA damage checkpoints are often defective in cancer cells, allowing cell division in spite of the accumulation of genetic errors. Conversely, DNA replication stress and mitotic checkpoints rarely undergo mutations because any aberrant activity could result in irreparable damage or catastrophic chromosomal missegregation, leading to cancer cell death. This review focuses on the checkpoint signaling mechanisms involved in cancer cells and how an emerging understanding of these pathways can provide new therapeutic opportunities for cancer therapy.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Samarendra K Singh
- School of Biotechnology, Institute of Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark J O'Connor
- Discovery Centre, AstraZeneca, Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Andrew Koff
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York
| | - Garima Singh
- School of Biotechnology, Institute of Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Justin Stebbing
- School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Karen Carmelina Crasta
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore, Singapore
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael B Yaffe
- MIT Center for Precision Cancer Medicine, Koch Institute for Integrative Cancer Research, Broad Institute, Massachusetts Institute of Technology, Cambridge, Boston, Massachusetts
| | - Seth A Wander
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
11
|
Yu Y, Arrigo A, Chandra A, Zhuang C, Najjar MK, Khan MS, Zhu D, Dono A, Strowd RE, Tandon N, Zhu JJ, Hsu SH, Esquenazi Y, Chan M, Lo HW. Targeting tGLI1, a novel mediator of tumor therapeutic resistance, using Ketoconazole sensitizes glioblastoma to CDK4/6 therapy and chemoradiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639359. [PMID: 40060625 PMCID: PMC11888219 DOI: 10.1101/2025.02.20.639359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Glioblastoma (GBM) remains the most aggressive primary brain tumor in adults, with no effective treatments. While cyclin-dependent kinase 4/6 inhibitors (CDK4/6is) show clinical promise in some cancers, they have not significantly improved survival in GBM patients. This lack of response is attributed to the treatment-resistant glioma stem cell (GSC) population. We previously identified truncated glioma-associated oncogene homolog 1 (tGLI1) as a novel transcription factor promoting GSCs; however, its role in CDK4/6i resistance has never been investigated in any cancer type. Here, we found positive correlations between tGLI1 and CDK4/6 therapeutic resistance in patient datasets and in vitro studies. Pharmacological inhibition of tGLI1 using FDA-approved ketoconazole (KCZ), a tGLI1-specific inhibitor, sensitized GBM and GSCs to CDK4/6is. KCZ+CDK4/6i combination therapy demonstrated synergistic anti-proliferative effects, significantly inhibiting GBM stemness and cell cycle progression while increasing apoptosis. The combination was more efficacious than monotherapies in two orthotopic GBM mouse models. tGLI1 promoted GBM resistance to radiation therapy and temozolomide, while KCZ potentiated effects of these treatments. Collectively, we report for the first time that tGLI1 is a novel mediator of GBM resistance to CDK4/6is, and KCZ sensitizes GBM to CDK4/6is, thereby supporting future clinical utility of novel KCZ+CDK4/6i combinatorial therapy for GBM patients.
Collapse
Affiliation(s)
- Yang Yu
- Department of Cancer Biology, Wake Forest School University of Medicine, Winston-Salem, NC
| | - Austin Arrigo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ankush Chandra
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chuling Zhuang
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mariana K Najjar
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Munazza S Khan
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dongqin Zhu
- Department of Cancer Biology, Wake Forest School University of Medicine, Winston-Salem, NC
| | - Antonio Dono
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Roy E Strowd
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Nitin Tandon
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sigmund H Hsu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael Chan
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest School University of Medicine, Winston-Salem, NC
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
12
|
Schirripa A, Schöppe H, Nebenfuehr S, Zojer M, Klampfl T, Kugler V, Maw BS, Ceylan H, Uras IZ, Scheiblecker L, Gamper E, Stelzl U, Stefan E, Kaserer T, Sexl V, Kollmann K. Cdk6's functions are critically regulated by its unique C-terminus. iScience 2025; 28:111697. [PMID: 39898030 PMCID: PMC11787673 DOI: 10.1016/j.isci.2024.111697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/09/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
The vital cell cycle machinery is tightly regulated and alterations of its central signaling hubs are a hallmark of cancer. The activity of CDK6 is controlled by interaction with several partners including cyclins and INK4 proteins, which have been shown to mainly bind to the amino-terminal lobe. We analyzed the impact of CDK6's C-terminus on its functions in a leukemia model, revealing a central role in promoting proliferation. C-terminally truncated Cdk6 (Cdk6 ΔC) shows reduced nuclear translocation and therefore chromatin interaction and fails to enhance proliferation and disease progression. The combination of proteomic analysis and protein modeling highlights that Cdk6's C-terminus is essential for protein flexibility and for its binding potential to cyclin D, p27Kip1 and INK4 proteins but not cyclin B. We demonstrate that the C-terminus is a unique and essential part of the CDK6 protein, regulating interaction partner binding and therefore CDK6's functionality.
Collapse
Affiliation(s)
- Alessia Schirripa
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Helge Schöppe
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Thorsten Klampfl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Valentina Kugler
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Belinda S. Maw
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Huriye Ceylan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Iris Z. Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Lisa Scheiblecker
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Elisabeth Gamper
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Ulrich Stelzl
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Eduard Stefan
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|
13
|
Kumarasamy V, Wang J, Roti M, Wan Y, Dommer AP, Rosenheck H, Putta S, Trub A, Bisi J, Strum J, Roberts P, Rubin SM, Frangou C, McLean K, Witkiewicz AK, Knudsen ES. Discrete vulnerability to pharmacological CDK2 inhibition is governed by heterogeneity of the cancer cell cycle. Nat Commun 2025; 16:1476. [PMID: 39924553 PMCID: PMC11808123 DOI: 10.1038/s41467-025-56674-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Cyclin dependent kinase 2 (CDK2) regulates cell cycle and is an emerging target for cancer therapy. There are relatively small numbers of tumor models that exhibit strong dependence on CDK2 and undergo G1 cell cycle arrest following CDK2 inhibition. The expression of P16INK4A and cyclin E1 determines this sensitivity to CDK2 inhibition. The co-expression of these genes occurs in breast cancer patients highlighting their clinical significance as predictive biomarkers for CDK2-targeted therapies. In cancer models that are genetically independent of CDK2, pharmacological inhibitors suppress cell proliferation by inducing 4N cell cycle arrest and increasing the expressions of phospho-CDK1 (Y15) and cyclin B1. CRISPR screens identify CDK2 loss as a mediator of resistance to a CDK2 inhibitor, INX-315. Furthermore, CDK2 deletion reverses the G2/M block induced by CDK2 inhibitors and restores cell proliferation. Complementary drug screens define multiple means to cooperate with CDK2 inhibition beyond G1/S. These include the depletion of mitotic regulators as well as CDK4/6 inhibitors cooperate with CDK2 inhibition in multiple phases of the cell cycle. Overall, this study underscores two fundamentally distinct features of response to CDK2 inhibitors that are conditioned by tumor context and could serve as the basis for differential therapeutic strategies in a wide range of cancers.
Collapse
Affiliation(s)
- Vishnu Kumarasamy
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Michelle Roti
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yin Wan
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adam P Dommer
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Hanna Rosenheck
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sivasankar Putta
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | | | | | | | | | - Seth M Rubin
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Costakis Frangou
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Karen McLean
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
14
|
Liu X, Qian K, Su L, Tian X, Zhao X, Wang T, Han L, Li Z, Zhang P, Wang R, Liu B, Li Y, Tan X, Zhang Y. Comprehensive genomic and transcriptomic profiling of pulmonary nodules in synchronous multiple primary lung cancer. Eur J Med Res 2025; 30:74. [PMID: 39905556 PMCID: PMC11792379 DOI: 10.1186/s40001-025-02327-7] [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: 11/26/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Synchronous multiple primary lung cancer (sMPLC) exhibits distinct histopathological characteristics among pulmonary nodules. However, a comprehensive understanding of the somatic mutation landscape and transcriptome heterogeneity is lacking. Therefore, our study aims to meticulously investigate genomic distinctions among multiple pulmonary nodules within individual patients. METHODS We performed targeted DNA sequencing on tumor specimens and conducted bulk RNA transcriptome analysis on 53 multiple nodules originating from 26 lung cancer patients. The multiple nodules from the same patient was determined as major nodule and minor nodule. Additionally, the tumor tissues underwent histopathological evaluation through H&E staining, complemented by a comprehensive series of immunohistochemistry (IHC) analyses to detect protein expression. The detected protein markers encompassed PD-L1, Ki67, and others. RESULTS For the 53 nodule samples from 26 MPLCs patients, EGFR was the mostly mutated genes, and the TP53 mutation frequency was notably different between major and minor nodules. Furthermore, pathway enrichment analysis based on the differentially expressed genes (DEGs) between major and minor nodules revealed the significantly active cell cycle and p53 pathways in the major nodules. Additionally, both major and minor nodules demonstrated mostly similar immune microenvironment and PD-L1 protein expression, and a significantly higher expression of Ki67. A noteworthy suppression was observed in the immune microenvironment in nodules, revealed by the expression of macrophage, neutrophils, and NK cells. Furthermore, minor nodules exhibited a modestly elevated expression of macrophages compared to major nodules. Additionally, among the significantly up-regulated cell cycle-related genes in the major nodules when compared with minor nodules, CCNE1 mRNA expression demonstrated significant correlation with poor prognosis in the lung cancer. Furthermore, the MYC inhibitor demonstrated more sensitivity for the major nodules than minor nodules. CONCLUSIONS This study validated molecular distinctions between samples from major and minor nodules in patients with sMPLC at both genomic and transcriptomic levels. The major nodules exhibited heightened activity in tumor cell proliferation pathways and demonstrated malignancy-related biological characteristics, which correlated with pathological assessment results.
Collapse
Affiliation(s)
- Xingsheng Liu
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Kun Qian
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Lei Su
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Xiaoru Tian
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Xin Zhao
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Tengteng Wang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Li Han
- Kanghui Biotech Co., Ltd., Liaoning, 110170, China
| | - Zhenzhen Li
- Kanghui Biotech Co., Ltd., Liaoning, 110170, China
| | - Peilong Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Ruotian Wang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Baodong Liu
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Yuanbo Li
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Xiaogang Tan
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University Lung Cancer Center, Beijing, 100053, China.
| |
Collapse
|
15
|
Cuan X, Wang J, Zhao Y, Yan J, Sheng J, Huang Y. Network Pharmacology Approach and Experimental Verification to Explore the Anti-NSCLC Mechanism of Grifolic Acid. Int J Mol Sci 2025; 26:629. [PMID: 39859343 PMCID: PMC11765843 DOI: 10.3390/ijms26020629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related death. Non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancers and over 60% express wild-type EGFR (WT-EGFR); however, EGFR tyrosine kinase inhibitors (TKIs) have limited effect in most patients with WT-EGFR tumors. In this study, we applied network pharmacology screening and MTT screening of bioactive compounds to obtain one novel grifolic acid that may inhibit NSCLC through the EGFR-ERK1/2 pathway. Through the PPI network and machine learning, we identified two hub genes, EGFR and AKT1, as potential therapeutic targets. Molecular docking confirmed that the grifolic acid could effectively bind to the key target, EGFR. Using the NSCLC cell line NCI-H1781 as an in vitro model, we evaluated the effect of the drugs' combination on viability, apoptosis, and clonogenicity capacity. In vitro studies showed that combined treatment decreased cell viability, increased activation PARP, and caused cell cycle redistribution and significantly greater inhibition of pEGFR and pAKT. This study not only provides new insights into the mechanism of grifolic acid against NSCLC but also important information and new research ideas for the discovery of anti-NSCLC compounds from natural products.
Collapse
Affiliation(s)
- Xiangdan Cuan
- Key Laboratory of Pu-Er Tea Science, Ministry of Education, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China; (X.C.); (J.W.); (Y.Z.); (J.Y.)
| | - Jinxian Wang
- Key Laboratory of Pu-Er Tea Science, Ministry of Education, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China; (X.C.); (J.W.); (Y.Z.); (J.Y.)
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China
| | - Yue Zhao
- Key Laboratory of Pu-Er Tea Science, Ministry of Education, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China; (X.C.); (J.W.); (Y.Z.); (J.Y.)
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China
| | - Jingyun Yan
- Key Laboratory of Pu-Er Tea Science, Ministry of Education, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China; (X.C.); (J.W.); (Y.Z.); (J.Y.)
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China
| | - Jun Sheng
- Key Laboratory of Pu-Er Tea Science, Ministry of Education, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China; (X.C.); (J.W.); (Y.Z.); (J.Y.)
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China
| | - Yanping Huang
- Key Laboratory of Pu-Er Tea Science, Ministry of Education, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China; (X.C.); (J.W.); (Y.Z.); (J.Y.)
- College of Science, Yunnan Agricultural University, Heilongtan, North of Kunming, Kunming 650201, China
| |
Collapse
|
16
|
Pellarin I, Dall'Acqua A, Favero A, Segatto I, Rossi V, Crestan N, Karimbayli J, Belletti B, Baldassarre G. Cyclin-dependent protein kinases and cell cycle regulation in biology and disease. Signal Transduct Target Ther 2025; 10:11. [PMID: 39800748 PMCID: PMC11734941 DOI: 10.1038/s41392-024-02080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin Dependent Kinases (CDKs) are closely connected to the regulation of cell cycle progression, having been first identified as the kinases able to drive cell division. In reality, the human genome contains 20 different CDKs, which can be divided in at least three different sub-family with different functions, mechanisms of regulation, expression patterns and subcellular localization. Most of these kinases play fundamental roles the normal physiology of eucaryotic cells; therefore, their deregulation is associated with the onset and/or progression of multiple human disease including but not limited to neoplastic and neurodegenerative conditions. Here, we describe the functions of CDKs, categorized into the three main functional groups in which they are classified, highlighting the most relevant pathways that drive their expression and functions. We then discuss the potential roles and deregulation of CDKs in human pathologies, with a particular focus on cancer, the human disease in which CDKs have been most extensively studied and explored as therapeutic targets. Finally, we discuss how CDKs inhibitors have become standard therapies in selected human cancers and propose novel ways of investigation to export their targeting from cancer to other relevant chronic diseases. We hope that the effort we made in collecting all available information on both the prominent and lesser-known CDK family members will help in identify and develop novel areas of research to improve the lives of patients affected by debilitating chronic diseases.
Collapse
Affiliation(s)
- Ilenia Pellarin
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessandra Dall'Acqua
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Andrea Favero
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Ilenia Segatto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Valentina Rossi
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Nicole Crestan
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Javad Karimbayli
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Barbara Belletti
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
17
|
Takeuchi R, Nomura T, Yaguchi M, Kuwahara N, Amino Y, Taguchi C, Suzuki I, Suzuki H, Nagashima T, Arikawa K, Okada Y, Nomoto T, Hiratsuka K. Cyclosporine A causes gingival overgrowth via reduced G1 cell cycle arrest in gingival fibroblasts. PLoS One 2024; 19:e0309189. [PMID: 39705288 DOI: 10.1371/journal.pone.0309189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 12/22/2024] Open
Abstract
Gingival overgrowth caused by cyclosporine A is due to increased fibroblast proliferation in gingival tissues. Cell cycle system balances proliferation and anti-proliferation of gingival fibroblasts and plays a role in the maintenance of its population in gingival tissues. When cells detect and respond to abnormalities (e.g. DNA damage), cell cycle progression is arrested in the G1 phase until the completion of damage restoration. In this study, we investigated the effects of cyclosporine A on G1 cell cycle arrest and on its regulators in gingival fibroblasts to clarify the mechanism of cyclosporine A-induced gingival overgrowth. Human gingival fibroblasts from healthy donors were cultured to semi-confluence and were then treated with or without 200 ng/mL (166 nM) cyclosporine A in D-MEM with 2% fetal bovine serum. Cell proliferation was assessed by counting total cell numbers. The distribution of cell cycle phases was assessed using flow cytometric analysis. The levels of mRNA and protein expression for cell cycle regulators were quantified using reverse transcription-quantitative PCR and western blot analysis, respectively. Treatment with cyclosporine A markedly increased cell proliferation, inhibited G1 cell cycle arrest, significantly increased CDC25A and CYCLIN E1 mRNA expression levels, significantly decreased P21, SMAD3 and SMAD4 mRNA expression levels, significantly upregulated the protein expression levels of CDC25A, CYCLIN E1, pCDK2 and pRB1 and significantly downregulated the protein expression levels of P21, SMAD3 and SMAD4. Treatment with cyclosporine A also increased MYC and ATM mRNA expression levels and decreased CDK2, ATR, P27, P53 and RB1 mRNA expression levels but not significantly. These results demonstrate that cyclosporine A causes gingival overgrowth due to the following mechanism in gingival fibroblasts: cyclosporine A increases levels of phospho-CDK2 and CYCLIN E1 by upregulating CDC25A and downregulating P21 with the downregulation of SMAD3 and SMAD4, which results in the inhibition of G1 cell cycle arrest.
Collapse
Affiliation(s)
- Reiri Takeuchi
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Takatoshi Nomura
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Manabu Yaguchi
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Noriko Kuwahara
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Yuta Amino
- Department of Oral Implantology, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Chieko Taguchi
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Itaru Suzuki
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Haruka Suzuki
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Teruaki Nagashima
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
- Department of Community Oral Health, Nihon University Graduate School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Kazumune Arikawa
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Yuichiro Okada
- Department of Histology, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Takato Nomoto
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| | - Koichi Hiratsuka
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan
| |
Collapse
|
18
|
Takeuchi R, Kuwahara N, Amino Y, Hayashi S, Taguchi C, Suzuki I, Suzuki H, Nagashima T, Arikawa K, Okada Y, Nomoto T, Hiratsuka K. Cyclosporine A Causes Gingival Overgrowth by Promoting Entry into the S Phase at the G1/S Cell Cycle Checkpoint in Gingival Fibroblasts Exposed to Lipopolysaccharide. Diseases 2024; 12:322. [PMID: 39727652 PMCID: PMC11727098 DOI: 10.3390/diseases12120322] [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: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES Cyclosporine A promotes gingival fibrosis by enhancing the proliferation of gingival fibroblasts, leading to gingival overgrowth. The population of gingival fibroblasts is regulated by cell cycle machinery, which balances cell growth and inhibition. Cells that detect DNA damage pause at the G1/S checkpoint to repair the damage instead of progressing to the S phase. Previous studies have linked drug-induced gingival overgrowth to the response of fibroblasts to lipopolysaccharide (LPS) and cyclosporine A. This research investigates the effects of cyclosporine A on the G1/S checkpoint and its mediators in LPS-treated gingival fibroblasts to clarify the mechanisms behind cyclosporine-A-induced gingival overgrowth. METHODS Semi-confluent human gingival fibroblasts were treated with LPS or cyclosporine A in DMEM. Cell proliferation was evaluated by counting the total number of cells. The distribution of the cell cycle phases was analyzed using flow cytometry. Additionally, the expression levels of mRNAs and proteins related to cell cycle regulators were quantified by reverse-transcription quantitative PCR and Western blotting, respectively. RESULTS Cyclosporine A treatment significantly enhanced cell proliferation and the G1-S cell cycle transition. It increased the mRNA levels of CDC25A and CYCLIN D while decreasing those of RB1, SMAD3, and SMAD4. Additionally, it upregulated the protein levels of CDC25A, CYCLIN D, CDK4, CDK6, and pRB and downregulated the protein levels of SMAD3 and SMAD4. CONCLUSIONS Gingival overgrowth induced by cyclosporine A could be attributed to these alterations.
Collapse
Affiliation(s)
- Reiri Takeuchi
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (N.K.); (K.H.)
| | - Noriko Kuwahara
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (N.K.); (K.H.)
| | - Yuta Amino
- Department of Oral Implantology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan;
| | - Sachiyo Hayashi
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (S.H.); (T.N.)
| | - Chieko Taguchi
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Itaru Suzuki
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Haruka Suzuki
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Teruaki Nagashima
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
- Department of Community Oral Health, Nihon University Graduate School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan
| | - Kazumune Arikawa
- Department of Community Oral Health, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (C.T.); (I.S.); (H.S.); (K.A.)
| | - Yuichiro Okada
- Department of Histology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan
| | - Takato Nomoto
- Department of Special Needs Dentistry, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (S.H.); (T.N.)
| | - Koichi Hiratsuka
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan; (N.K.); (K.H.)
| |
Collapse
|
19
|
Oriakhi K, Erharuyi O, Orumwensodia KO, Essien EE, Falodun A, Uadia PO, Bernhard F, Engel N. Pro-apoptotic and anti-proliferative activities of cassane diterpenoids on squamous carcinoma cells: An in vitro and in silico study. Toxicol Rep 2024; 13:101833. [PMID: 39717850 PMCID: PMC11665704 DOI: 10.1016/j.toxrep.2024.101833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Squamous carcinoma of the head and neck is characterized by aberrant apoptosis that prolongs the proliferative capacity of the cells and by uncontrolled cell growth. This study aimed to examine the pro-apoptotic and antiproliferative effects of Caesalpinia pulcherrima cassane diterpenoids on squamous carcinoma cells in vitro. The cytotoxicity of four (4) cassane diterpenoids {Six-cinnamoyl- 7-hydroxyvouacapen-5-ol(1), pulcherrimin A(2), C(3), and E(4)} isolated from C. pulcherrima was determined in squamous carcinoma cell lines (CAL33, FaDu, and Detroit 562) and in non tumorigenic fibroblast cells. The results showed that compounds 1 and 4 had the highest cytotoxic potential, significantly reducing cell viability in all squamous cell lines in a concentration dependent manner. Compounds 1 and 4 may inhibit the proliferation of CAL33 cells by reducing their ability to divide, decreasing PCNA expression, and suppressing migration. Additionally, treatment with compounds 1 and 4 led to an activation of Caspase 3 expression in FaDu cells. Molecular docking analysis revealed strong binding affinities of compounds 1 and 4 to the Caspase 3 receptor, with values of -8.5 and -8.8 kcal/mol, respectively. These results suggest that Pulcherrimin E and 6-cinnamoyl-7-hydroxylvouacapen-5-ol have potential antitumor effects based on their selective cytotoxic effect on squamous carcinoma cells in vitro.
Collapse
Affiliation(s)
- Kelly Oriakhi
- Department of Medical Biochemistry, School of Basic Medical Sciences, College of Medical Sciences, University of Benin, Nigeria
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, USA
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University, Medical Center, Schillingallee 35, Rostock 18057, Germany
| | - Osayemwenre Erharuyi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Benin, Nigeria
| | | | | | - Abiodun Falodun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Benin, Nigeria
| | - Patrick O. Uadia
- Department of Biochemistry, Faculty of Life Sciences, University of Benin, Nigeria
| | - Frerich Bernhard
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University, Medical Center, Schillingallee 35, Rostock 18057, Germany
| | - Nadja Engel
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University, Medical Center, Schillingallee 35, Rostock 18057, Germany
| |
Collapse
|
20
|
Roy R, Gampa SC, Garimella SV. Role of specific CDKs in regulating DNA damage repair responses and replication stress. Curr Opin Pharmacol 2024; 79:102485. [PMID: 39265226 DOI: 10.1016/j.coph.2024.102485] [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: 09/01/2022] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/14/2024]
Abstract
Cyclins along with their catalytic units, Cyclin-dependent kinases (CDKs) regulate the cell cycle transition and transcription; and are essentially known as 'master regulators' in modulating DNA damage response (DDR) and replication stress. In addition to influencing DNA repair and damage signaling, CDKs also play a pivotal role in cell division fidelity and the maintenance of genomic integrity after DNA damage. In this review, we focus on the intricate ways by which specific CDKs mainly CDK7, CDK9, and CDK12/13, regulate the cell cycle progression and transcription and how their modulation can lead to lethal effects on the integrity of the genome. With a better knowledge of how these CDKs control the DDR and replication stress, it is now possible to combine CDK inhibitors with chemotherapeutic drugs that damage DNA in ways that can be applied in clinical settings as successful therapeutic strategies.
Collapse
Affiliation(s)
- Rahul Roy
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi, 110016, India
| | - Siri Chandana Gampa
- Department of Biotechnology, School of Science, GITAM (deemed to be University), Visakhapatnam, 530045, India
| | - Sireesha V Garimella
- Department of Biotechnology, School of Science, GITAM (deemed to be University), Visakhapatnam, 530045, India.
| |
Collapse
|
21
|
Łupicka-Słowik A, Cossu F, Sieńczyk M. Palbociclib as an Antitumor Drug: A License to Kill. Molecules 2024; 29:5334. [PMID: 39598723 PMCID: PMC11596203 DOI: 10.3390/molecules29225334] [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: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Neoplastic cells are characterized by uncontrolled cell divisions caused by cell cycle dysregulation. Key regulatory proteins governing the transition from the G1 to the S phase are the CDK4 and CDK6 kinases, which are controlled by D-type cyclins. The CDK4/6 kinases enable the use of these proteins as targets for anticancer therapy because they prevent the growth and the development of malignant cells by inhibiting their activity. This paper surveys the clinical trial results concerning palbociclib, the first in-class FDA-approved anticancer drug for hormone-dependent breast cancer. It discusses the therapeutic applications in breast cancer as well as in solid tumors and hematopoietic malignancies. Additionally, the paper presents an analysis of palbociclib resistance acquired during therapy and explores new approaches, such as modifications to palbociclib that enhance its desired activity or open up new therapeutic possibilities (PROTACs).
Collapse
Affiliation(s)
- Agnieszka Łupicka-Słowik
- Division of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| | - Federica Cossu
- National Research Council, Institute of Biophysics (IBF-CNR), Milan Unit, Via Corti, 12, 20133 Milan, Italy;
| | - Marcin Sieńczyk
- Division of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| |
Collapse
|
22
|
Araki K, Torii T, Takeuchi K, Kinoshita N, Urano R, Nakajima R, Zhou Y, Kobayashi T, Hanyu T, Ohtani K, Ambe K, Kawauchi K. Non-canonical olfactory pathway activation induces cell fusion of cervical cancer cells. Neoplasia 2024; 57:101044. [PMID: 39222591 PMCID: PMC11402306 DOI: 10.1016/j.neo.2024.101044] [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: 07/25/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Multinucleation occurs in various types of advanced cancers and contributes to their malignant characteristics, including anticancer drug resistance. Therefore, inhibiting multinucleation can improve cancer prognosis; however, the molecular mechanisms underlying multinucleation remain elusive. Here, we introduced a genetic mutation in cervical cancer cells to induce cell fusion-mediated multinucleation. The olfactory receptor OR1N2 was heterozygously mutated in these fused cells; the same OR1N2 mutation was detected in multinucleated cells from clinical cervical cancer specimens. The mutation-induced structural change in the OR1N2 protein activated protein kinase A (PKA), which, in turn, mediated the non-canonical olfactory pathway. PKA phosphorylated and activated furin protease, resulting in the cleavage of the fusogenic protein syncytin-1. Because this cleaved form of syncytin-1, processed by furin, participates in cell fusion, furin inhibitors could suppress multinucleation and reduce surviving cell numbers after anticancer drug treatment. The improved anticancer drug efficacy indicates a promising therapeutic approach for advanced cervical cancers.
Collapse
Affiliation(s)
- Keigo Araki
- Department of Morphological Biology, School of Dentistry, Ohu University, Koriyama, Fukushima 963-8611, Japan.
| | - Takeru Torii
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo 650-0047, Japan
| | - Kohei Takeuchi
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo 650-0047, Japan
| | - Natsuki Kinoshita
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo 650-0047, Japan
| | - Ryoto Urano
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo 650-0047, Japan
| | - Rinka Nakajima
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Hyogo 669-1330, Japan
| | - Yaxuan Zhou
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Hyogo 669-1330, Japan
| | - Tokuo Kobayashi
- Department of Morphological Biology, School of Dentistry, Ohu University, Koriyama, Fukushima 963-8611, Japan
| | - Tadayoshi Hanyu
- Department of Gynecology, Tsuboi Cancer Center Hospital, Koriyama, Fukushima 963-0197, Japan
| | - Kiyoshi Ohtani
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Hyogo 669-1330, Japan
| | - Kimiharu Ambe
- Department of Morphological Biology, School of Dentistry, Ohu University, Koriyama, Fukushima 963-8611, Japan
| | - Keiko Kawauchi
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
23
|
Advani D, Kumar P. Uncovering Cell Cycle Dysregulations and Associated Mechanisms in Cancer and Neurodegenerative Disorders: A Glimpse of Hope for Repurposed Drugs. Mol Neurobiol 2024; 61:8600-8630. [PMID: 38532240 DOI: 10.1007/s12035-024-04130-7] [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: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
The cell cycle is the sequence of events orchestrated by a complex network of cell cycle proteins. Unlike normal cells, mature neurons subsist in a quiescent state of the cell cycle, and aberrant cell cycle activation triggers neuronal death accompanied by neurodegeneration. The periodicity of cell cycle events is choreographed by various mechanisms, including DNA damage repair, oxidative stress, neurotrophin activity, and ubiquitin-mediated degradation. Given the relevance of cell cycle processes in cancer and neurodegeneration, this review delineates the overlapping cell cycle events, signaling pathways, and mechanisms associated with cell cycle aberrations in cancer and the major neurodegenerative disorders. We suggest that dysregulation of some common fundamental signaling processes triggers anomalous cell cycle activation in cancer cells and neurons. We discussed the possible use of cell cycle inhibitors for neurodegenerative disorders and described the associated challenges. We propose that a greater understanding of the common mechanisms driving cell cycle aberrations in cancer and neurodegenerative disorders will open a new avenue for the development of repurposed drugs.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India.
| |
Collapse
|
24
|
Saito A, Omura I, Imaizumi K. CREB3L1/OASIS: cell cycle regulator and tumor suppressor. FEBS J 2024; 291:4853-4866. [PMID: 38215153 DOI: 10.1111/febs.17052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/09/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Cell cycle checkpoints detect DNA errors, eventually arresting the cell cycle to promote DNA repair. Failure of such cell cycle arrest causes aberrant cell proliferation, promoting the pathogenesis of multiple diseases, including cancer. Endoplasmic reticulum (ER) stress transducers activate the unfolded protein response, which not only deals with unfolded proteins in ER lumen but also orchestrates diverse physiological phenomena such as cell differentiation and lipid metabolism. Among ER stress transducers, cyclic AMP-responsive element-binding protein 3-like protein 1 (CREB3L1) [also known as old astrocyte specifically induced substance (OASIS)] is an ER-resident transmembrane transcription factor. This molecule is cleaved by regulated intramembrane proteolysis, followed by activation as a transcription factor. OASIS is preferentially expressed in specific cells, including astrocytes and osteoblasts, to regulate their differentiation. In accordance with its name, OASIS was originally identified as being upregulated in long-term-cultured astrocytes undergoing cell cycle arrest because of replicative stress. In the context of cell cycle regulation, previously unknown physiological roles of OASIS have been discovered. OASIS is activated as a transcription factor in response to DNA damage to induce p21-mediated cell cycle arrest. Although p21 is directly induced by the master regulator of the cell cycle, p53, no crosstalk occurs between p21 induction by OASIS or p53. Here, we summarize previously unknown cell cycle regulation by ER-resident transcription factor OASIS, particularly focusing on commonalities and differences in cell cycle arrest between OASIS and p53. This review also mentions tumorigenesis caused by OASIS dysfunctions, and OASIS's potential as a tumor suppressor and therapeutic target.
Collapse
Affiliation(s)
- Atsushi Saito
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Issei Omura
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| |
Collapse
|
25
|
Gao Y, Yu Y, Zhang M, Yu W, Kang L. Mechanisms of endocrine resistance in hormone receptor-positive breast cancer. Front Oncol 2024; 14:1448687. [PMID: 39544302 PMCID: PMC11560879 DOI: 10.3389/fonc.2024.1448687] [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: 06/14/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Hormone receptor-positive breast cancer may recur or metastasize years or decades after its diagnosis. Furthermore, hormone receptor expression may persist in relapsed or metastatic cancer cells. Endocrine therapy is one of the most efficacious treatments for hormone receptor-positive breast cancers. Nevertheless, a considerable proportion of patients develop resistance to endocrine therapy. Previous studies have identified numerous mechanisms underlying drug resistance, such as epigenetic abnormalities in the estrogen receptor (ER) genome, activation of ER-independent ligands, and alterations in signaling pathways including PI3K/AKT/mTOR, Notch, NF-κB, FGFR, and IRE1-XBP1. This article reviews the mechanisms of endocrine resistance in hormone receptor-positive advanced breast cancer, drawing from previous studies, and discusses the latest research advancements and prospects.
Collapse
Affiliation(s)
| | | | | | | | - Lihua Kang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
26
|
Li XZ, Song W, Zhao ZH, Lu YH, Xu GL, Yang LJ, Yin S, Sun QY, Chen LN. Flavopiridol induces cell cycle arrest and apoptosis by interfering with CDK1 signaling pathway in human ovarian granulosa cells. Sci Rep 2024; 14:26239. [PMID: 39482384 PMCID: PMC11528022 DOI: 10.1038/s41598-024-77032-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
Several clinical trials have been conducted to evaluate the use of flavopiridol (FP) to treat a variety of cancers, and almost all cancer drugs were found to be associated with toxicity and side effects. It is not clear whether the use of FP will affect the female reproductive system. Granulosa cells, as the important cells that constitute the follicle, play a crucial role in determining the reproductive ability of females. In this study, we investigated whether different concentrations of FP have a toxic effect on the growth of immortalized human ovarian granulosa cells. The results showed that FP had an inhibitory effect on cell proliferation at a level of nanomole concentration. FP reduced cell proliferation and induced apoptosis by inducing mitochondrial dysfunction and oxidative stress, as well as increasing BAX/BCL2 and pCDK1 levels. These results suggest that toxicity to the reproductive system should be considered when FP is used in clinical applications.
Collapse
Affiliation(s)
- Xiao-Zhen Li
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Song
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zheng-Hui Zhao
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - You-Hui Lu
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gen-Lu Xu
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Li-Jia Yang
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shen Yin
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Lei-Ning Chen
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
27
|
Liu Y, Deng Y, Yang C, Naranmandura H. Double-Faced Immunological Effects of CDK4/6 Inhibitors on Cancer Treatment: Challenges and Perspectives. Bioengineering (Basel) 2024; 11:1084. [PMID: 39593745 PMCID: PMC11591775 DOI: 10.3390/bioengineering11111084] [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: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Cyclin-dependent kinases (CDKs) are generally involved in the progression of cell cycle and cell division in normal cells, while abnormal activations of CDKs are deemed to be a driving force for accelerating cell proliferation and tumorigenesis. Therefore, CDKs have become ideal therapeutic targets for cancer treatment. The U.S FDA has approved three CDK4/6 inhibitors (CDK4/6is) for the treatment of patients with hormone receptor-positive (HR+) or human epidermal growth factor receptor 2-negative (HER2-) advanced or metastatic breast cancer, and these drugs showed impressive results in clinics. Besides cell-cycle arrest, there is growing evidence that CDK4/6is exert paradoxical roles on cancer treatment by altering the immune system. Indeed, clinical data showed that CDK4/6is could change the immune system to exert antitumor effects, while these changes also caused tumor resistance to CDK4/6i. However, the molecular mechanism for the regulation of the immune system by CDK4/6is is unclear. In this review, we comprehensively discuss the paradoxical immunological effects of CDK4/6is in cancer treatment, elucidating their anticancer mechanisms through immunomodulatory activity and induction of acquired drug resistance by dysregulating the immune microenvironment. More importantly, we suggest a few strategies including combining CDK4/6is with immunotherapy to overcome drug resistance.
Collapse
Affiliation(s)
- Yongqin Liu
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiying Deng
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chang Yang
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hua Naranmandura
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
28
|
Yang M, Xiang H, Luo G. Targeting Protein Kinase, Membrane-Associated Tyrosine/Threonine 1 (PKMYT1) for Precision Cancer Therapy: From Discovery to Clinical Trial. J Med Chem 2024; 67:17997-18016. [PMID: 39383322 DOI: 10.1021/acs.jmedchem.4c01619] [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: 10/11/2024]
Abstract
\Protein kinase membrane-associated tyrosine/threonine 1 (PKMYT1), an overlooked member of the WEE family responsible for regulating cell cycle transition, has recently emerged as a compelling therapeutic target for precision cancer therapy due to its established synthetic lethal relationship with CCNE1 (cyclin E1) amplification. Since the first-in-class selective PKMYT1 inhibitor, RP-6306, entered clinical trials in 2021, the field has experienced renewed interest underscored by the growing number of inhibitor patents and the exploration of additional gene alterations, such as KRAS/p53 mutations, FBXW7 mutation, and PPP2R1A mutation, as novel synthetic lethal partners. This perspective summarizes, for the first time, the PKMYT1 structure, function, and inhibitors in both the literature and patent applications reported to date. Compounds are described focusing on their design and optimization process, structural features, and biological activity with the aim to promoting further drug discovery efforts targeting PKMYT1 as a potential precision therapy.
Collapse
Affiliation(s)
- Ming Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Hua Xiang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Guoshun Luo
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
29
|
Wu M, Zhao Y, Zhang C, Pu K. Advancing Proteolysis Targeting Chimera (PROTAC) Nanotechnology in Protein Homeostasis Reprograming for Disease Treatment. ACS NANO 2024; 18:28502-28530. [PMID: 39377250 DOI: 10.1021/acsnano.4c09800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) represent a transformative class of therapeutic agents that leverage the intrinsic protein degradation machinery to modulate the hemostasis of key disease-associated proteins selectively. Although several PROTACs have been approved for clinical application, suboptimal therapeutic efficacy and potential adverse side effects remain challenging. Benefiting from the enhanced targeted delivery, reduced systemic toxicity, and improved bioavailability, nanomedicines can be tailored with precision to integrate with PROTACs which hold significant potential to facilitate PROTAC nanomedicines (nano-PROTACs) for clinical translation with enhanced efficacy and reduced side effects. In this review, we provide an overview of the recent progress in the convergence of nanotechnology with PROTAC design, leveraging the inherent properties of nanomaterials, such as lipids, polymers, inorganic nanoparticles, nanohydrogels, proteins, and nucleic acids, for precise PROTAC delivery. Additionally, we discuss the various categories of PROTAC targets and provide insights into their clinical translational potential, alongside the challenges that need to be addressed.
Collapse
Affiliation(s)
- Mengyao Wu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yilan Zhao
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chi Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, 636921, Singapore
| |
Collapse
|
30
|
Susanti NMP, Kurniawan F, Damayanti S, Kartasasmita RE, Tjahjono DH. The novel selective inhibitors of cyclin-dependent kinase 4/6: in vitro and in silico study. Sci Rep 2024; 14:23505. [PMID: 39379427 PMCID: PMC11461483 DOI: 10.1038/s41598-024-71865-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 10/10/2024] Open
Abstract
One of the main regulators in the cell cycle is cyclin-dependent kinase 4 and 6 (CDK4/6). FDA has approved CDK4/6 inhibitors for the treatment of patients with metastatic breast cancer. However, the development of selective agents remains problematic due to the conservation of their ATP binding sites. In the previous in silico study, ZINC585292724, ZINC585292587, ZINC585291674, and ZINC585291474 have been identified as potential inhibitors. Therefore, the present study aimed to analyze the selectivity and inhibitory activity of the four compounds against CDK4/6 in vitro as well as determine the potential for their further development in silico. The in vitro results showed that the four compounds had good selectivity towards both kinases, due to their similar structure. In agreement with the in silico results, ZINC585291674 produced the best inhibitory activity against CDK4 and CDK6, with IC50 of 184.14 nM and 111.78 nM, respectively. Their ADMET profile were also similar to reference compound of Palbociclib. Based on this, ZINC585291674 can be used as a lead compound for further inhibitor development.
Collapse
Affiliation(s)
- Ni Made Pitri Susanti
- School of Pharmacy, Bandung Institute of Technology, Bandung, 40132, Indonesia
- Study Program of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Udayana, Badung, 80361, Indonesia
| | - Fransiska Kurniawan
- School of Pharmacy, Bandung Institute of Technology, Bandung, 40132, Indonesia
| | - Sophi Damayanti
- School of Pharmacy, Bandung Institute of Technology, Bandung, 40132, Indonesia
| | | | | |
Collapse
|
31
|
Brito Nunes C, Borges MC, Freathy RM, Lawlor DA, Qvigstad E, Evans DM, Moen GH. Understanding the Genetic Landscape of Gestational Diabetes: Insights into the Causes and Consequences of Elevated Glucose Levels in Pregnancy. Metabolites 2024; 14:508. [PMID: 39330515 PMCID: PMC11434570 DOI: 10.3390/metabo14090508] [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: 08/26/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Background/Objectives: During pregnancy, physiological changes in maternal circulating glucose levels and its metabolism are essential to meet maternal and fetal energy demands. Major changes in glucose metabolism occur throughout pregnancy and consist of higher insulin resistance and a compensatory increase in insulin secretion to maintain glucose homeostasis. For some women, this change is insufficient to maintain normoglycemia, leading to gestational diabetes mellitus (GDM), a condition characterized by maternal glucose intolerance and hyperglycaemia first diagnosed during the second or third trimester of pregnancy. GDM is diagnosed in approximately 14.0% of pregnancies globally, and it is often associated with short- and long-term adverse health outcomes in both mothers and offspring. Although recent studies have highlighted the role of genetic determinants in the development of GDM, research in this area is still lacking, hindering the development of prevention and treatment strategies. Methods: In this paper, we review recent advances in the understanding of genetic determinants of GDM and glycaemic traits during pregnancy. Results/Conclusions: Our review highlights the need for further collaborative efforts as well as larger and more diverse genotyped pregnancy cohorts to deepen our understanding of the genetic aetiology of GDM, address research gaps, and further improve diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Caroline Brito Nunes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Rachel M. Freathy
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 4PY, UK;
| | - Deborah A. Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Elisabeth Qvigstad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - David M. Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1QU, UK
- Frazer Institute, University of Queensland, Brisbane 4102, Australia
| | - Gunn-Helen Moen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4067, Australia
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Frazer Institute, University of Queensland, Brisbane 4102, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| |
Collapse
|
32
|
Gaballah A, Elsherbiny A, Sharaky M, Hamed N, Raslan N, Almilaibary A, Fayyad R, Ousman M, Hamdan A, Fahim S. Dexamethasone-tamoxifen combination exerts synergistic therapeutic effects in tamoxifen-resistance breast cancer cells. Biosci Rep 2024; 44:BSR20240367. [PMID: 38864530 PMCID: PMC11230869 DOI: 10.1042/bsr20240367] [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: 03/20/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/13/2024] Open
Abstract
Tamoxifen (TAM) is a key player in estrogen receptor-positive (ER+) breast cancer (BC); however, ∼30% of patients experience relapse and a lower survival rate due to TAM resistance. TAM resistance was related to the over expression of SOX-2 gene, which is regulated by the E2F3 transcription factor in the Wnt signaling pathway. It was suggested that SOX-2 overexpression was suppressed by dexamethasone (DEX), a glucocorticoid commonly prescribed to BC patients. The aim of the present study is to explore the effect of combining DEX and TAM on the inhibition of TAM-resistant LCC-2 cells (TAMR-1) through modulating the E2F3/SOX-2-mediated Wnt signaling pathway. The effect of the combination therapy on MCF-7 and TAMR-1 cell viability was assessed. Drug interactions were analyzed using CompuSyn and SynergyFinder softwares. Cell cycle distribution, apoptotic protein expression, gene expression levels of SOX-2 and E2F3, and cell migration were also assessed. Combining DEX with TAM led to synergistic inhibition of TAMR-1 cell proliferation and migration, induced apoptosis, reduced SOX-2 and E2F3 expression and was also associated with S and G2-M phase arrest. Therefore, combining DEX with TAM may present an effective therapeutic option to overcome TAM resistance, by targeting the E2F3/SOX-2/Wnt signaling pathway, in addition to its anti-inflammatory effect.
Collapse
Affiliation(s)
- Aliaa I. Gaballah
- School of Pharmacy, Newgiza University (NGU), Newgiza, km 22 Cairo-Alexandria Desert Road, Giza, P.O. Box 12577, Egypt
| | - Aliaa A. Elsherbiny
- Department of Biochemistry, School of Pharmacy, Newgiza University (NGU), Newgiza, km 22 Cairo-Alexandria Desert Road, Giza, P.O. Box 12577, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Najat O. Hamed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Nahed A. Raslan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
- Clinical Pharmacy Program, College of Health Sciences and Nursing, Al-Rayan Colleges, Medina 42541, Saudi Arabia
| | - Abdullah Almilaibary
- Department of Family and Community Medicine, Faculty of Medicine, Al-Baha University, AlBaha, Saudi Arabia
| | - Reda Mohamed Abdrabbou Fayyad
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
- Department of Pharmacology, General Medicine Practice Program, Batterjee Medical College, Aseer 61961, Saudi Arabia
| | - Mona S. Ousman
- Emergency Medical Services, College of Applied Sciences, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Ahmed M.E. Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Sally A. Fahim
- Department of Biochemistry, School of Pharmacy, Newgiza University (NGU), Newgiza, km 22 Cairo-Alexandria Desert Road, Giza, P.O. Box 12577, Egypt
| |
Collapse
|
33
|
Ni K, Li ZL, Hu ZY, Hong L. Antitumor Effect of Apcin on Endometrial Carcinoma via p21-Mediated Cell Cycle Arrest and Apoptosis. Curr Med Sci 2024; 44:623-632. [PMID: 38853192 DOI: 10.1007/s11596-024-2877-z] [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: 10/06/2023] [Accepted: 03/27/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVE Endometrial carcinoma (EC) is a prevalent gynecological malignancy characterized by increasing incidence and mortality rates. This underscores the critical need for novel therapeutic targets. One such potential target is cell division cycle 20 (CDC20), which has been implicated in oncogenesis. This study investigated the effect of the CDC20 inhibitor Apcin on EC and elucidated the underlying mechanism involved. METHODS The effects of Apcin on EC cell proliferation, apoptosis, and the cell cycle were evaluated using CCK8 assays and flow cytometry. RNA sequencing (RNA-seq) was subsequently conducted to explore the underlying molecular mechanism, and Western blotting and coimmunoprecipitation were subsequently performed to validate the results. Animal studies were performed to evaluate the antitumor effects in vivo. Bioinformatics analysis was also conducted to identify CDC20 as a potential therapeutic target in EC. RESULTS Treatment with Apcin inhibited proliferation and induced apoptosis in EC cells, resulting in cell cycle arrest. Pathways associated with apoptosis and the cell cycle were activated following treatment with Apcin. Notably, Apcin treatment led to the upregulation of the cell cycle regulator p21, which was verified to interact with CDC20 and consequently decrease the expression of downstream cyclins in EC cells. In vivo experiments confirmed that Apcin treatment significantly impeded tumor growth. Higher CDC20 expression was observed in EC tissue than in nonmalignant tissue, and increased CDC20 expression in EC patients was associated with shorter overall survival and progress free interval. CONCLUSION CDC20 is a novel molecular target in EC, and Apcin could be developed as a candidate antitumor drug for EC treatment.
Collapse
Affiliation(s)
- Ke Ni
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Li Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi-Yong Hu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
34
|
Magge T, Rajendran S, Brufsky AM, Foldi J. CDK4/6 inhibitors: The Devil is in the Detail. Curr Oncol Rep 2024; 26:665-678. [PMID: 38713311 DOI: 10.1007/s11912-024-01540-7] [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] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE OF REVIEW Update on the most recent clinical evidence on CDK4/6 inhibitors (CDK4/6i) in the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor (HER)2-negative breast cancer. RECENT FINDINGS Over the past decade, CDK4/6i have become part of the standard of care treatment of patients with both metastatic and high-risk early HR + /HER2- breast cancers. The three available CDK4/6i (palbociclib, ribociclib and abemaciclib) have been extensively studied in combination with endocrine therapy (ET) in metastatic breast cancer (mBC) with consistent prolongation of progression free survival; however, ribociclib has emerged as the preferred first line agent in mBC given overall survival benefit over endocrine monotherapy. In early BC, abemaciclib is the only currently approved agent while ribociclib has early positive clinical trial data. Toxicities and financial burden limit the use of CDK4/6i in all patients and resource-poor settings, and optimal timing of their use in mBC remains unclear. There is considerable evidence for the use of CDK4/6i in metastatic and early HR + /HER2- breast cancer, but knowledge gaps remain, and further research is necessary to better define their optimal use.
Collapse
Affiliation(s)
- Tara Magge
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Sneha Rajendran
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Adam M Brufsky
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Julia Foldi
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Breast Medical Oncology, Magee Women's Hospital, University of Pittsburgh Medical Center, 300 Halket Street, Suite 3524, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
35
|
Lashen A, Alqahtani S, Shoqafi A, Algethami M, Jeyapalan JN, Mongan NP, Rakha EA, Madhusudan S. Clinicopathological Significance of Cyclin-Dependent Kinase 2 (CDK2) in Ductal Carcinoma In Situ and Early-Stage Invasive Breast Cancers. Int J Mol Sci 2024; 25:5053. [PMID: 38732271 PMCID: PMC11084890 DOI: 10.3390/ijms25095053] [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: 02/26/2024] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
Cyclin-dependent kinase 2 (CDK2) is a key cell cycle regulator, with essential roles during G1/S transition. The clinicopathological significance of CDK2 in ductal carcinomas in situ (DCIS) and early-stage invasive breast cancers (BCs) remains largely unknown. Here, we evaluated CDK2's protein expression in 479 BC samples and 216 DCIS specimens. Analysis of CDK2 transcripts was completed in the METABRIC cohort (n = 1980) and TCGA cohort (n = 1090), respectively. A high nuclear CDK2 protein expression was significantly associated with aggressive phenotypes, including a high tumour grade, lymph vascular invasion, a poor Nottingham prognostic index (all p-values < 0.0001), and shorter survival (p = 0.006), especially in luminal BC (p = 0.009). In p53-mutant BC, high nuclear CDK2 remained linked with worse survival (p = 0.01). In DCIS, high nuclear/low cytoplasmic co-expression showed significant association with a high tumour grade (p = 0.043), triple-negative and HER2-enriched molecular subtypes (p = 0.01), Comedo necrosis (p = 0.024), negative ER status (p = 0.004), negative PR status (p < 0.0001), and a high proliferation index (p < 0.0001). Tumours with high CDK2 transcripts were more likely to have higher expressions of genes involved in the cell cycle, homologous recombination, and p53 signaling. We provide compelling evidence that high CDK2 is a feature of aggressive breast cancers. The clinical evaluation of CDK2 inhibitors in early-stage BC patients will have a clinical impact.
Collapse
MESH Headings
- Humans
- Female
- Cyclin-Dependent Kinase 2/metabolism
- Cyclin-Dependent Kinase 2/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Prognosis
- Middle Aged
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Neoplasm Staging
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/mortality
- Aged
- Gene Expression Regulation, Neoplastic
- Neoplasm Invasiveness
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Ayat Lashen
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
- Department of Pathology, Nottingham University Hospital, City Campus, Nottingham NG5 1PB, UK
| | - Shatha Alqahtani
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Ahmed Shoqafi
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Mashael Algethami
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Jennie N. Jeyapalan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
- Faculty of Medicine and Health Sciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
| | - Nigel P. Mongan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
- Faculty of Medicine and Health Sciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emad A. Rakha
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
- Department of Pathology, Nottingham University Hospital, City Campus, Nottingham NG5 1PB, UK
| | - Srinivasan Madhusudan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (S.A.); (A.S.); (M.A.); (J.N.J.); (N.P.M.); (E.A.R.)
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| |
Collapse
|
36
|
Guo Z, Tang Y, Wang S, Huang Y, Chi Q, Xu K, Xue L. Natural product fargesin interferes with H3 histone lactylation via targeting PKM2 to inhibit non-small cell lung cancer tumorigenesis. Biofactors 2024; 50:592-607. [PMID: 38149461 DOI: 10.1002/biof.2031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common malignant tumors. There is an urgent need to find more effective drugs that inhibit NSCLC. Fargesin (FGS) has demonstrated anti-tumor effects; however, its efficacy and the molecular mechanism of inhibiting NSCLC are unclear. Herein, we investigated FGS' inhibitory effects on NSCLC by CCK8 and EdU assays and cell cycle analysis of A549 cells in vitro and in a nude mouse tumor transplantation model in vivo. FGS (10-50 μM) significantly inhibited cell proliferation and down-regulated expression levels of CDK1 and CCND1. Transcriptomic analysis showed that FGS regulated the cell metabolic process pathway. Differential metabolites with FGS treatment were enriched in glycolysis and pyruvate pathways. Cell metabolism assay were used to evaluate the oxygen consumption rate (OCR), Extracellular acidification rate (ECAR) in A549 cells. FGS also inhibited the production of cellular lactate and the expression of LDHA, LDHB, PKM2, and SLC2A1. These genes were identified as important oncogenes in lung cancer, and their binding to FGS was confirmed by molecular docking simulation. Notably, the over-expression and gene silencing experiments signified PKM2 as the molecular target of FGS for anti-tumorigenesis. Moreover, the H3 histone lactylation, were correlated with tumorigenesis, were inhibited with FGS treatment. Conclusively, FGS inhibited the aerobic glycolytic and H3 histone lactylation signaling pathways in A549 NSCLC cells by targeting PKM2. These findings provide evidence of the therapeutic potential of FGS in NSCLC.
Collapse
Affiliation(s)
- Zizhang Guo
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yeqing Tang
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shunshun Wang
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuming Huang
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingjia Chi
- Department of Mechanics and Engineering Structure, Hubei Key Laboratory of Theory and Application of Advanced Materials Mechanics, Wuhan University of Technology, China
| | - Kang Xu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Lei Xue
- Department of Thoracic Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
37
|
Niu P, Tao Y, Lin G, Xu H, Meng Q, Yang K, Huang W, Song M, Ding K, Ma D, Fan M. Design and Synthesis of Novel Macrocyclic Derivatives as Potent and Selective Cyclin-Dependent Kinase 7 Inhibitors. J Med Chem 2024; 67:6099-6118. [PMID: 38586950 DOI: 10.1021/acs.jmedchem.3c01832] [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: 04/09/2024]
Abstract
The duality of function (cell cycle regulation and gene transcription) of cyclin-dependent kinase 7 (CDK7) makes it an attractive oncology target and the discovery of CDK7 inhibitors has been a long-term pursuit by academia and pharmaceutical companies. However, achieving selective leading compounds is still difficult owing to the similarities among the ATP binding pocket. Herein, we detail the design and synthesis of a series of macrocyclic derivatives with pyrazolo[1,5-a]-1,3,5-triazine core structure as potent and selective CDK7 inhibitors. The diverse manners of macrocyclization led to distinguished selectivity profiles of the CDK family. Molecular dynamics (MD) simulation explained the binding difference between 15- and 16-membered macrocyclic compounds. Further optimization generated compound 37 exhibiting good CDK7 inhibitory activity and high selectivity over other CDKs. This work clearly demonstrated macrocyclization is a versatile method to finely tune the selectivity profile of small molecules and MD simulation can be a valuable tool in prioritizing designs of the macrocycle.
Collapse
Affiliation(s)
- Pengpeng Niu
- Academy of Medical Engineering and Translational Medicine (AMT), Tianjin University, Tianjin 300072, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Yanxin Tao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Guohao Lin
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Huiqi Xu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qingyuan Meng
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
- Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Kang Yang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Weixue Huang
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 20032, China
| | - Meiru Song
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Ke Ding
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 20032, China
| | - Dawei Ma
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 20032, China
| | - Mengyang Fan
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
38
|
Niu P, Tao Y, Meng Q, Huang Y, Li S, Ding K, Ma D, Ye Z, Fan M. Discovery of novel macrocyclic derivatives as potent and selective cyclin-dependent kinase 2 inhibitors. Bioorg Med Chem 2024; 104:117711. [PMID: 38583237 DOI: 10.1016/j.bmc.2024.117711] [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: 03/23/2024] [Accepted: 03/31/2024] [Indexed: 04/09/2024]
Abstract
Cyclin-dependent kinase 2 (CDK2) is a member of CDK family of kinases (CDKs) that regulate the cell cycle. Its inopportune or over-activation leads to uncontrolled cell cycle progression and drives numerous types of cancers, especially ovarian, uterine, gastric cancer, as well as those associated with amplified CCNE1 gene. However, developing selective lead compound as CDK2 inhibitors remains challenging owing to similarities in the ATP pockets among different CDKs. Herein, we described the optimization of compound 1, a novel macrocyclic inhibitor targeting CDK2/5/7/9, aiming to discover more selective and metabolically stable lead compound as CDK2 inhibitor. Molecular dynamic (MD) simulations were performed for compound 1 and 9 to gain insights into the improved selectivity against CDK5. Further optimization efforts led to compound 22, exhibiting excellent CDK2 inhibitory activity, good selectivity over other CDKs and potent cellular effects. Based on these characterizations, we propose that compound 22 holds great promise as a potential lead candidate for drug development.
Collapse
Affiliation(s)
- Pengpeng Niu
- Academy of Medical Engineering and Translational Medicine (AMT), Tianjin University, Tianjin 300072, China; Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Yanxin Tao
- School of Life Sciences, Tianjin University, Tianjin 300072, China; Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Qingyuan Meng
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China; Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yixing Huang
- Department of Otorhinolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310022, China
| | - Shan Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Ke Ding
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 20032, China
| | - Dawei Ma
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 20032, China
| | - Zu Ye
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China.
| | - Mengyang Fan
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
39
|
Wang BS, Zhang CL, Cui X, Li Q, Yang L, He ZY, Yang Z, Zeng MM, Cao N. Curcumin inhibits the growth and invasion of gastric cancer by regulating long noncoding RNA AC022424.2. World J Gastrointest Oncol 2024; 16:1437-1452. [PMID: 38660661 PMCID: PMC11037052 DOI: 10.4251/wjgo.v16.i4.1437] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Gastric cancer, characterized by a multifactorial etiology and high heterogeneity, continues to confound researchers in terms of its pathogenesis. Curcumin, a natural anticancer agent, exhibits therapeutic promise in gastric cancer. Its effects include promoting cell apoptosis, curtailing tumor angiogenesis, and enhancing sensitivity to radiation and chemotherapy. Long noncoding RNAs (lncRNAs) have garnered significant attention as biomarkers for early screening, diagnosis, treatment, and drug response because of their remarkable specificity and sensitivity. Recent investigations have revealed an association between aberrant lncRNA expression and early diagnosis, clinical staging, metastasis, drug sensitivity, and prognosis in gastric cancer. A profound understanding of the intricate mechanisms through which lncRNAs influence gastric cancer development can provide novel insights for precision treatment and tailored management of patients with gastric cancer. This study aimed to unravel the potential of curcumin in suppressing the malignant behavior of gastric cancer cells by upregulating specific lncRNAs and modulating gastric cancer onset and progression. AIM To identify lncRNAs associated with curcumin treatment and investigate the role of lncRNA AC022424.2 in the effects of curcumin on gastric cancer cell apoptosis, proliferation, and invasion. Furthermore, these findings were validated in clinical samples. METHODS The study employed CCK-8 assays to assess the impact of curcumin on gastric cancer cell proliferation, flow cytometry to investigate its effects on apoptosis, and scratch and Transwell assays to evaluate its influence on the migration and invasion of BGC-823 and MGC-803 cells. Western blotting was used to gauge changes in the protein expression levels of CDK6, CDK4, Bax, Bcl-2, caspase-3, P65, and the PI3K/Akt/mTOR pathway in gastric cancer cell lines after curcumin treatment. Differential expression of lncRNAs before and after curcumin treatment was assessed using lncRNA sequencing and validated using quantitative reverse transcription polymerase chain reaction (qRT-PCR) in BGC-823 and MGC-803 cells. AC022424.2-1 knockdown BGC-823 and MGC-803 cells were generated to scrutinize the impact of lncRNA AC022424.2 on apoptosis, proliferation, migration, and invasion of gastric cancer cells. Western blotting was performed to ascertain changes in the expression of proteins implicated in the PI3K/Akt/mTOR and NF-κB signaling pathways. RT-PCR was employed to measure lncRNA AC022424.2 expression in clinical gastric cancer tissues and to correlate its expression with clinical pathological characteristics. RESULTS Curcumin induced apoptosis and hindered proliferation, migration, and invasion of gastric cancer cells in a dose- and time-dependent manner. LncRNA AC022424.2 was upregulated after curcumin treatment, and its knockdown enhanced cancer cell aggressiveness. LncRNA AC022424.2 may have affected cancer cells via the PI3K/Akt/mTOR and NF-κB signaling pathways. LncRNA AC022424.2 downregulation was correlated with lymph node metastasis, making it a potential diagnostic and prognostic marker. CONCLUSION Curcumin has potential anticancer effects on gastric cancer cells by regulating lncRNA AC022424.2. This lncRNA plays a significant role in cancer cell behavior and may have clinical implications in diagnosis and prognosis evaluation. The results of this study enhance our understanding of gastric cancer development and precision treatment.
Collapse
Affiliation(s)
- Bin-Sheng Wang
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Chen-Li Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Xiang Cui
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Qiang Li
- Third Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Lei Yang
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Zhi-Yun He
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ze Yang
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Miao-Miao Zeng
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Nong Cao
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
40
|
Lashen A, Algethami M, Alqahtani S, Shoqafi A, Sheha A, Jeyapalan JN, Mongan NP, Rakha EA, Madhusudan S. The Clinicopathological Significance of the Cyclin D1/E1-Cyclin-Dependent Kinase (CDK2/4/6)-Retinoblastoma (RB1/pRB1) Pathway in Epithelial Ovarian Cancers. Int J Mol Sci 2024; 25:4060. [PMID: 38612869 PMCID: PMC11012085 DOI: 10.3390/ijms25074060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Cyclin-dependent kinases (CDK2, CDK4, CDK6), cyclin D1, cyclin E1 and phosphorylated retinoblastoma (pRB1) are key regulators of the G1/S cell cycle checkpoint and may influence platinum response in ovarian cancers. CDK2/4/6 inhibitors are emerging targets in ovarian cancer therapeutics. In the current study, we evaluated the prognostic and predictive significance of the CDK2/4/6-cyclin D1/E1-pRB1 axis in clinical ovarian cancers (OC). The CDK2/4/6, cyclin D1/E1 and RB1/pRB1 protein expression were investigated in 300 ovarian cancers and correlated with clinicopathological parameters and patient outcomes. CDK2/4/6, cyclin D1/E1 and RB1 mRNA expression were evaluated in the publicly available ovarian TCGA dataset. We observed nuclear and cytoplasmic staining for CDK2/4/6, cyclins D1/E1 and RB1/pRB1 in OCs with varying percentages. Increased nuclear CDK2 and nuclear cyclin E1 expression was linked with poor progression-free survival (PFS) and a shorter overall survival (OS). Nuclear CDK6 was associated with poor OS. The cytoplasmic expression of CDK4, cyclin D1 and cyclin E1 also has predictive and/or prognostic significance in OCs. In the multivariate analysis, nuclear cyclin E1 was an independent predictor of poor PFS. Tumours with high nuclear cyclin E1/high nuclear CDK2 have a worse PFS and OS. Detailed bioinformatics in the TCGA cohort showed a positive correlation between cyclin E1 and CDK2. We also showed that cyclin-E1-overexpressing tumours are enriched for genes involved in insulin signalling and release. Our data not only identified the prognostic/predictive significance of these key cell cycle regulators but also demonstrate the importance of sub-cellular localisation. CDK2 targeting in cyclin-E1-amplified OCs could be a rational approach.
Collapse
Affiliation(s)
- Ayat Lashen
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
- Department of Pathology, Nottingham University Hospital, City Campus, Nottingham NG5 1PB, UK
| | - Mashael Algethami
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Shatha Alqahtani
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Ahmed Shoqafi
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Amera Sheha
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Jennie N. Jeyapalan
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
- Faculty of Medicine and Health Sciences, Centre for Cancer Sciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
| | - Nigel P. Mongan
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
- Faculty of Medicine and Health Sciences, Centre for Cancer Sciences, University of Nottingham, Sutton Bonington Campus, Sutton Bonington LE12 5RD, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emad A. Rakha
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
| | - Srinivasan Madhusudan
- Naaz Coker Ovarian Cancer Research Centre, Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (A.L.); (M.A.); (S.A.); (A.S.); (A.S.); (J.N.J.); (N.P.M.); (E.A.R.)
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| |
Collapse
|
41
|
Zhi D, An Z, Li L, Zheng C, Yuan X, Lan Y, Zhang J, Xu Y, Ma H, Li N, Wang J. 3-Amide-β-carbolines block the cell cycle by targeting CDK2 and DNA in tumor cells potentially as anti-mitotic agents. Bioorg Chem 2024; 145:107216. [PMID: 38387396 DOI: 10.1016/j.bioorg.2024.107216] [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: 12/12/2023] [Revised: 02/04/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
β-Carboline alkaloids are natural and synthetic products with outstanding antitumor activity. C3 substituted and dimerized β-carbolines exert excellent antitumor activity. In the present research, 37 β-carboline derivatives were synthesized and characterized. Their cytotoxicity, cell cycle, apoptosis, and CDK2- and DNA-binding affinity were evaluated. β-Carboline monomer M3 and dimer D4 showed selective activity and higher cytotoxicity in tumor cells than in normal cells. Structure-activity relationships (SAR) indicated that the amide group at C3 enhanced the antitumor activity. M3 blocked the A549 (IC50 = 1.44 ± 1.10 μM) cell cycle in the S phase and inhibited A549 cell migration, while D4 blocked the HepG2 (IC50 = 2.84 ± 0.73 μM) cell cycle in the G0/G1 phase, both of which ultimately induced apoptosis. Furthermore, associations of M3 and D4 with CDK2 and DNA were proven by network pharmacology analysis, molecular docking, and western blotting. The expression level of CDK2 was downregulated in M3-treated A549 cells and D4-treated HepG2 cells. Moreover, M3 and D4 interact with DNA and CDK2 at sub-micromolar concentrations in endothermic interactions caused by entropy-driven adsorption processes, which means that the favorable entropy change (ΔS > 0) overcomes the unfavorable enthalpy change (ΔH > 0) and drives the spontaneous reaction (ΔG < 0). Overall, these results clarified the antitumor mechanisms of M3 and D4 through disrupting the cell cycle by binding DNA and CDK2, which demonstrated the potential of M3 and D4 as novel antiproliferative drugs targeting mitosis.
Collapse
Affiliation(s)
- Dongming Zhi
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Zhiyuan An
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Lishan Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Chaojia Zheng
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Xiaorong Yuan
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Yu Lan
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Jinghan Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Yujie Xu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Huiya Ma
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Na Li
- College of Chemistry and Life Science, Chifeng University, Inner Mongolia Autonomous Region, China.
| | - Junru Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, China.
| |
Collapse
|
42
|
Purohit L, Jones C, Gonzalez T, Castrellon A, Hussein A. The Role of CD4/6 Inhibitors in Breast Cancer Treatment. Int J Mol Sci 2024; 25:1242. [PMID: 38279242 PMCID: PMC10816395 DOI: 10.3390/ijms25021242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
Over the last decade, treatment paradigms for breast cancer have undergone a renaissance, particularly in hormone-receptor-positive/HER2-negative breast cancer. These revolutionary therapies are based on the selective targeting of aberrancies within the cell cycle. This shift towards targeted therapies has also changed the landscape of disease monitoring. In this article, we will review the fundamentals of cell cycle progression in the context of the new cyclin-dependent kinase inhibitors. In addition to discussing the currently approved cyclin-dependent kinase inhibitors for breast cancer, we will explore the ongoing development and search for predictive biomarkers and modalities to monitor treatment.
Collapse
Affiliation(s)
| | | | | | - Aurelio Castrellon
- Memorial Health System, Pembroke Pines, FL 33024, USA; (L.P.); (C.J.); (T.G.); (A.H.)
| | | |
Collapse
|
43
|
Youness RA, Habashy DA, Khater N, Elsayed K, Dawoud A, Hakim S, Nafea H, Bourquin C, Abdel-Kader RM, Gad MZ. Role of Hydrogen Sulfide in Oncological and Non-Oncological Disorders and Its Regulation by Non-Coding RNAs: A Comprehensive Review. Noncoding RNA 2024; 10:7. [PMID: 38250807 PMCID: PMC10801522 DOI: 10.3390/ncrna10010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Recently, myriad studies have defined the versatile abilities of gasotransmitters and their synthesizing enzymes to play a "Maestro" role in orchestrating several oncological and non-oncological circuits and, thus, nominated them as possible therapeutic targets. Although a significant amount of work has been conducted on the role of nitric oxide (NO) and carbon monoxide (CO) and their inter-relationship in the field of oncology, research about hydrogen sulfide (H2S) remains in its infancy. Recently, non-coding RNAs (ncRNAs) have been reported to play a dominating role in the regulation of the endogenous machinery system of H2S in several pathological contexts. A growing list of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are leading the way as upstream regulators for H2S biosynthesis in different mammalian cells during the development and progression of human diseases; therefore, their targeting can be of great therapeutic benefit. In the current review, the authors shed the light onto the biosynthetic pathways of H2S and their regulation by miRNAs and lncRNAs in various oncological and non-oncological disorders.
Collapse
Affiliation(s)
- Rana A. Youness
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
- Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, Cairo 11835, Egypt
| | - Danira Ashraf Habashy
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
- Clinical Pharmacy Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Nour Khater
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Kareem Elsayed
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Sousanna Hakim
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Heba Nafea
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Carole Bourquin
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, 1211 Geneva, Switzerland;
| | - Reham M. Abdel-Kader
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| | - Mohamed Z. Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11835, Egypt
| |
Collapse
|
44
|
Maleki EH, Bahrami AR, Matin MM. Cancer cell cycle heterogeneity as a critical determinant of therapeutic resistance. Genes Dis 2024; 11:189-204. [PMID: 37588236 PMCID: PMC10425754 DOI: 10.1016/j.gendis.2022.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/20/2022] [Accepted: 11/16/2022] [Indexed: 01/15/2023] Open
Abstract
Intra-tumor heterogeneity is now arguably one of the most-studied topics in tumor biology, as it represents a major obstacle to effective cancer treatment. Since tumor cells are highly diverse at genetic, epigenetic, and phenotypic levels, intra-tumor heterogeneity can be assumed as an important contributing factor to the nullification of chemotherapeutic effects, and recurrence of the tumor. Based on the role of heterogeneous subpopulations of cancer cells with varying cell-cycle dynamics and behavior during cancer progression and treatment; herein, we aim to establish a comprehensive definition for adaptation of neoplastic cells against therapy. We discuss two parallel and yet distinct subpopulations of tumor cells that play pivotal roles in reducing the effects of chemotherapy: "resistant" and "tolerant" populations. Furthermore, this review also highlights the impact of the quiescent phase of the cell cycle as a survival mechanism for cancer cells. Beyond understanding the mechanisms underlying the quiescence, it provides an insightful perspective on cancer stem cells (CSCs) and their dual and intertwined functions based on their cell cycle state in response to treatment. Moreover, CSCs, epithelial-mesenchymal transformed cells, circulating tumor cells (CTCs), and disseminated tumor cells (DTCs), which are mostly in a quiescent state of the cell cycle are proved to have multiple biological links and can be implicated in our viewpoint of cell cycle heterogeneity in tumors. Overall, increasing our knowledge of cell cycle heterogeneity is a key to identifying new therapeutic solutions, and this emerging concept may provide us with new opportunities to prevent the dreadful cancer recurrence.
Collapse
Affiliation(s)
- Ebrahim H. Maleki
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 31-007 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, 917751376 Mashhad, Iran
| |
Collapse
|
45
|
Knight H, Abis G, Kaur M, Green HL, Krasemann S, Hartmann K, Lynham S, Clark J, Zhao L, Ruppert C, Weiss A, Schermuly RT, Eaton P, Rudyk O. Cyclin D-CDK4 Disulfide Bond Attenuates Pulmonary Vascular Cell Proliferation. Circ Res 2023; 133:966-988. [PMID: 37955182 PMCID: PMC10699508 DOI: 10.1161/circresaha.122.321836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a chronic vascular disease characterized, among other abnormalities, by hyperproliferative smooth muscle cells and a perturbed cellular redox and metabolic balance. Oxidants induce cell cycle arrest to halt proliferation; however, little is known about the redox-regulated effector proteins that mediate these processes. Here, we report a novel kinase-inhibitory disulfide bond in cyclin D-CDK4 (cyclin-dependent kinase 4) and investigate its role in cell proliferation and PH. METHODS Oxidative modifications of cyclin D-CDK4 were detected in human pulmonary arterial smooth muscle cells and human pulmonary arterial endothelial cells. Site-directed mutagenesis, tandem mass-spectrometry, cell-based experiments, in vitro kinase activity assays, in silico structural modeling, and a novel redox-dead constitutive knock-in mouse were utilized to investigate the nature and definitively establish the importance of CDK4 cysteine modification in pulmonary vascular cell proliferation. Furthermore, the cyclin D-CDK4 oxidation was assessed in vivo in the pulmonary arteries and isolated human pulmonary arterial smooth muscle cells of patients with pulmonary arterial hypertension and in 3 preclinical models of PH. RESULTS Cyclin D-CDK4 forms a reversible oxidant-induced heterodimeric disulfide dimer between C7/8 and C135, respectively, in cells in vitro and in pulmonary arteries in vivo to inhibit cyclin D-CDK4 kinase activity, decrease Rb (retinoblastoma) protein phosphorylation, and induce cell cycle arrest. Mutation of CDK4 C135 causes a kinase-impaired phenotype, which decreases cell proliferation rate and alleviates disease phenotype in an experimental mouse PH model, suggesting this cysteine is indispensable for cyclin D-CDK4 kinase activity. Pulmonary arteries and human pulmonary arterial smooth muscle cells from patients with pulmonary arterial hypertension display a decreased level of CDK4 disulfide, consistent with CDK4 being hyperactive in human pulmonary arterial hypertension. Furthermore, auranofin treatment, which induces the cyclin D-CDK4 disulfide, attenuates disease severity in experimental PH models by mitigating pulmonary vascular remodeling. CONCLUSIONS A novel disulfide bond in cyclin D-CDK4 acts as a rapid switch to inhibit kinase activity and halt cell proliferation. This oxidative modification forms at a critical cysteine residue, which is unique to CDK4, offering the potential for the design of a selective covalent inhibitor predicted to be beneficial in PH.
Collapse
Affiliation(s)
- Hannah Knight
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Giancarlo Abis
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, United Kingdom (G.A.)
| | - Manpreet Kaur
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Hannah L.H. Green
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Centre Hamburg-Eppendorf, Germany (S.K., K.H.)
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Centre Hamburg-Eppendorf, Germany (S.K., K.H.)
| | - Steven Lynham
- Proteomics Core Facility, Centre of Excellence for Mass Spectrometry (S.L.), King’s College London, United Kingdom
| | - James Clark
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom (L.Z.)
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center Giessen Biobank, Justus-Liebig-University Giessen, Germany (C.R.)
| | - Astrid Weiss
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Member of the German Center for Lung Research (DZL), Germany (A.W., R.T.S.)
| | - Ralph T. Schermuly
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Member of the German Center for Lung Research (DZL), Germany (A.W., R.T.S.)
| | - Philip Eaton
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (P.E.)
| | - Olena Rudyk
- School of Cardiovascular and Metabolic Medicine and Sciences, British Heart Foundation Centre of Research Excellence (H.K., M.K., H.L.H.G., J.C., O.R.), King’s College London, United Kingdom
| |
Collapse
|
46
|
Hu W, Wang L, Luo J, Zhang J, Li N. The Potent Novel CDK4/6 Inhibitor TQB3616 in Hormone Receptor Positive Breast Cancer: Preclinical Characterization with in vitro and Human Tumor Xenograft Models. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:899-912. [PMID: 38090281 PMCID: PMC10715022 DOI: 10.2147/bctt.s434973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024]
Abstract
PURPOSE Inhibition of the cyclin-dependent kinase (CDK) 4/6-retinoblastoma (RB) pathway exerts a considerable inhibitory effect, preventing the spread and metastasis of breast cancer cells and promoting tumor regression. In this study, we examined the antitumor activity of TQB3616, a novel inhibitor of CDK4/6 activity, which showed a greater efficacy improvement in antitumor effects. METHODS TQB3616 group, abemaciclib group and endocrine or HER-2 targeted combination therapy group were set up respectively. The effects of drugs on cell proliferation activity, cell cycle, apoptosis, downstream protein expression and gene expression of HR positive (T47D, MCF-7) and HER-2 positive (BT474, MDA-MB-361) breast cancer cell lines were studied. The antiproliferative effect of TQB3616 was also measured in vivo. RESULTS TQB3616 showed a remarkable inhibitory effect on the proliferation of hormone receptor-positive breast cancer cells in vitro. In addition, TQB3616 combined with endocrine therapy or Human Epidermal Growth Factor Receptor 2 (HER2) targeted therapy showed significant synergistic antitumor activity in estrogen receptor (ER)-positive/HER2-negative or HER2-positive breast cancer. In contrast to abemaciclib, which targets the CDK4/6 pathway with proven efficacy, the oral agent TQB3616 not only induced G1 stalling, leading to a profound reduction in the level of RB protein phosphorylated at Ser807/811, but also showed enhanced tumor killing effects by promoting cell apoptosis. Oral administration of TQB3616 showed more potent antitumor activity than abemaciclib in an in vitro breast cancer xenograft model, causing significant tumor regression associated with sustained target inhibition in tumor tissue and manageable in vivo toxicity. CONCLUSION The results of this study indicate that TQB3616 is a novel CDK4/6 inhibitor, and its highly effective antitumor activity against breast cancer is expected to yield promising therapeutic effects in clinical studies.
Collapse
Affiliation(s)
- Wenyu Hu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Lei Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - JiaLing Luo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jian Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Nanlin Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
47
|
Yang B, Quan Y, Zhao W, Ji Y, Yang X, Li J, Li Y, Liu X, Wang Y, Li Y. Design, synthesis and biological evaluation of 2-((4-sulfamoylphenyl)amino)-pyrrolo[2,3-d]pyrimidine derivatives as CDK inhibitors. J Enzyme Inhib Med Chem 2023; 38:2169282. [PMID: 36656085 PMCID: PMC9858427 DOI: 10.1080/14756366.2023.2169282] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
To explore the potential use of CDK inhibitors in pancreatic ductal adenocarcinoma (PDAC) therapy, a series of novel 2-((4-sulfamoylphenyl)amino)-pyrrolo[2,3-d]pyrimidine derivatives was designed, synthesised, and investigated for inhibition on both CDK kinase activity and cellular proliferation of pancreatic cancer. Most of new sulphonamide-containing derivatives demonstrated strong inhibitory activity on CDK9 and obvious anti-proliferative activity in cell culture. Moreover, two new compounds suppressed cell proliferation of multiple human pancreatic cancer cell lines. The most potent compound 2g inhibited cancer cell proliferation by blocking Rb phosphorylation and induced apoptosis via downregulation of CDK9 downstream proteins Mcl-1 and c-Myc in MIA PaCa-2 cells. CDK9 knockdown experiment suggests its anti-proliferative activity is mainly mediated by CDK9. Additionally, 2g displayed moderate tumour inhibition effect in AsPC-1 derived xenograft mice model. Altogether, this study provided a new start for further optimisation to develop potential CDK inhibitor candidates for PDAC treatment by alone or combination use.
Collapse
Affiliation(s)
- Bo Yang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yanni Quan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yingjie Ji
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaotang Yang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jianrui Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ying Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,Ying Wang Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100050, China
| | - Yanping Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,CONTACT Yanping Li
| |
Collapse
|
48
|
Čižmáriková M, Michalková R, Mirossay L, Mojžišová G, Zigová M, Bardelčíková A, Mojžiš J. Ellagic Acid and Cancer Hallmarks: Insights from Experimental Evidence. Biomolecules 2023; 13:1653. [PMID: 38002335 PMCID: PMC10669545 DOI: 10.3390/biom13111653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a complex and multifaceted disease with a high global incidence and mortality rate. Although cancer therapy has evolved significantly over the years, numerous challenges persist on the path to effectively combating this multifaceted disease. Natural compounds derived from plants, fungi, or marine organisms have garnered considerable attention as potential therapeutic agents in the field of cancer research. Ellagic acid (EA), a natural polyphenolic compound found in various fruits and nuts, has emerged as a potential cancer prevention and treatment agent. This review summarizes the experimental evidence supporting the role of EA in targeting key hallmarks of cancer, including proliferation, angiogenesis, apoptosis evasion, immune evasion, inflammation, genomic instability, and more. We discuss the molecular mechanisms by which EA modulates signaling pathways and molecular targets involved in these cancer hallmarks, based on in vitro and in vivo studies. The multifaceted actions of EA make it a promising candidate for cancer prevention and therapy. Understanding its impact on cancer biology can pave the way for developing novel strategies to combat this complex disease.
Collapse
Affiliation(s)
- Martina Čižmáriková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Gabriela Mojžišová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| | - Martina Zigová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Annamária Bardelčíková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| |
Collapse
|
49
|
Fernezlian S, Baldavira C, de Souza M, Farhat C, de Vilhena A, Pereira J, de Campos J, Takagaki T, Balancin M, Ab'Saber A, Capelozzi V. A semi-automated microscopic image analysis method for scoring Ki-67 nuclear immunostaining. Braz J Med Biol Res 2023; 56:e12922. [PMID: 37970922 PMCID: PMC10644968 DOI: 10.1590/1414-431x2023e12922] [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: 07/12/2023] [Accepted: 09/11/2023] [Indexed: 11/19/2023] Open
Abstract
Nuclear proliferation marker MIB-1 (Ki-67) immunohistochemistry (IHC) is used to examine tumor cell proliferation. However, the diagnostic or prognostic value of the Ki-67 nuclear staining intensity and location, defined as nuclear gradient (NG), has not been assessed. This study examined the potential association between Ki-67 NG and cell cycle phases and its effect on the prognosis of pulmonary typical carcinoid (PTC) tumors. We propose a method for classifying the NG of Ki-67 during the cell cycle and compare the results between PTC, pulmonary adenocarcinoma (PAD), and breast ductal carcinoma (BDC). A literature review and objective analysis of IHC-stained paraffin sections were used to determine the Ki-67 labeling index and composed a stratification of the NG into NG1, NG2, and NG3/4 categories. A semi-automated image analysis protocol was established to determine the Ki-67 NG in PTC, PAD, and BDC. High intraobserver consistency and moderate interobserver agreement were achieved in the determination of Ki-67 NG in tumor specimens. NG1 and NG2 were lower in PTC than in PAD and BDC. Cox multivariate analysis of PTC after adjusting for age and number of metastatic lymph nodes showed that Ki-67 NG1 and NG2 significantly predicted clinical outcomes. The semi-automated method for quantification of Ki-67 nuclear immunostaining proposed in this study could become a valuable diagnostic and prognostic tool in PTC.
Collapse
Affiliation(s)
- S.M. Fernezlian
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - C.M. Baldavira
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M.L.F. de Souza
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - C. Farhat
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A.F. de Vilhena
- Departamento de Cirurgia Torácica, Instituto do Coração, São Paulo, SP, Brasil
| | - J.C.N. Pereira
- Department of General Thoracic Surgery, Georges Pompidou European Hospital, Paris, France
- International Perioperative Europrogram, Paris, France
| | - J.R.M. de Campos
- Departamento de Cirurgia Torácica, Instituto do Coração, São Paulo, SP, Brasil
- Departamento de Cirurgia Torácica, Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | - T. Takagaki
- Divisão de Pneumologia, Instituto do Coração, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M.L. Balancin
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A.M. Ab'Saber
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V.L. Capelozzi
- Laboratório de Genômica e Histomorfometria, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
50
|
Zhao Y, Culman J, Cascorbi I, Nithack N, Marx M, Zuhayra M, Lützen U. PSMA-617 inhibits proliferation and potentiates the 177Lu-PSMA-617-induced death of human prostate cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3315-3326. [PMID: 37284895 PMCID: PMC10567812 DOI: 10.1007/s00210-023-02539-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/17/2023] [Indexed: 06/08/2023]
Abstract
The human prostate-specific membrane antigen (PSMA) is substantially up-regulated in metastatic prostate cancer (PCa) cells. PSMA can be targeted by 177Lu conjugated to PSMA-617, a high-affinity ligand for the PSMA. The binding of the radioligand, 177Lu-PSMA-617, results in its internalisation and delivery of β-radiation into the cancer cells. However, PSMA-617, a component of the final product in the synthesis of the radioligand, may also play a role in the pathophysiology of PCa cells. The present study aimed to clarify the effects of PSMA-617 (10, 50 and 100 nM) on the expression of PSMA in PSMA-positive LNCaP cells, their proliferation, 177Lu-PSMA-617-induced cell death by WST-1 and lactate dehydrogenase assays, immunohistochemistry, western blotting, immunofluorescence staining and uptake of 177Lu-PSMA-617. PSMA-617 at 100 nM concentration induced cell-growth arrest, down-regulated cyclin D1 and cyclin E1 (by 43 and 36%, respectively) and up-regulated the cyclin-dependent kinase inhibitor p21Waf1/Cip1 (by 48%). Immunofluorescence staining demonstrated reduced content of DNA, pointing to a lower rate of cell division. PSMA-617 (up to 100 nM) did not alter the uptake of 177Lu-PSMA-617 into the LNCaP cells. Interestingly, simultaneous treatment with 177Lu-PSMA-617 and PSMA-617 for 24 and 48 h substantially potentiated the cell-death promoting effects of the radioligand. In conclusion, the combination of impeding tumour cell proliferation by PSMA-617 and its potentiation of the radiation-induced cell death brought about by 177Lu-PSMA-617 in PCa cells may considerably improve the outcome of the radiation therapy with 177Lu-PSMA-617, especially in patients with decreased radiosensitivity of PCa cells to the radioligand.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - Juraj Culman
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Niklas Nithack
- Central Rhine Community Hospital-Clinic for Urology and Pediatric Urology, Koblenz, Germany
| | - Marlies Marx
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Maaz Zuhayra
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ulf Lützen
- Department of Nuclear Medicine, Molecular Imaging, Diagnostics and Therapy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|