1
|
Lu J, Du L, Zhang P, Ma N, Zhang Q, Guo X, Li X, Lei X, Qu B. CKS1B regulates the radiosensitivity of lung cancer via activating the PI3K/AKT signaling pathway. Cell Signal 2025; 132:111828. [PMID: 40262716 DOI: 10.1016/j.cellsig.2025.111828] [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: 12/29/2024] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
Radiotherapy is the mainstay and first-line treatment for non-small-cell lung cancer (NSCLC). However, there are no effective strategies for regulating tumor radiosensitivity. This study aimed to examine whether CDC28 protein kinase regulatory subunit 1B (CKS1B) knockdown can radiosensitize NSCLC cells. The results indicated that CKS1B overexpression promoted the proliferation, migration, and invasion of NSCLC cells following exposure to ionizing radiation (IR). In addition, A549 cell xenografts with CKS1B knockdown exhibited significantly enhanced radiosensitivity compared to wild-type xenografts. Mechanistically, it was observed that CKS1B silencing stimulated apoptosis, inhibited cell cycle progression, and weakened DNA damage repair, thereby increasing the sensitivity of NSCLC cells to IR treatment. Moreover, the CKS1B-induced radioresistance was mediated by the PI3K/AKT signaling pathway. These findings demonstrate that CKS1B influences the NSCLC treatment, suggesting that it is a potential prognostic marker for predicting the radiosensitivity of NSCLC cells.
Collapse
Affiliation(s)
- Jiangyue Lu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China; Medical School of Chinese PLA, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Lehui Du
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Pei Zhang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Na Ma
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Qian Zhang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xingdong Guo
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xianwen Li
- School of Nursing, Nanjing Medical University, No.101 Long Mian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China.
| | - Xiao Lei
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China.
| | - Baolin Qu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing 100853, China.
| |
Collapse
|
2
|
Li B, Liu T, Zhao M, Guo D, Peng W, Tao Z, Hu X, Jin J. MYC amplification sensitizes TNBC to CHK1 inhibitors. Cell Signal 2025; 131:111709. [PMID: 40037423 DOI: 10.1016/j.cellsig.2025.111709] [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: 12/09/2024] [Revised: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, for which effective therapy is urgently needed. We demonstrated that MYC overexpression was associated with TNBC subtype and promoted the cell proliferation, invasion and migration in TNBC cells. Moreover, MYC overexpression induced replication stress and DNA damage in TNBC cells. Our subsequent results revealed that the novel second-generation CHK1 inhibitor, prexasertib, exhibited a more pronounced inhibitory effect in MYC-overexpressed TNBC cells compared to other DNA damage repair inhibitors, including ATR, WEE1, and PARP inhibitors. Prexasertib induced synergistic lethality with MYC-overexpressed TNBC cells by generating excessive DNA damage. Intriguingly, RNA-seq analysis identified an increase in MYC levels and activation of MYC-related pathways following prexasertib treatment, while western blot results showed that prexasertib led to MYC protein degradation independent of proteasome pathway. In addition, MYC overexpression was associated with an immunosuppressive microenvironment and high PD-L1 expression. Prexasertib activated cGAS-STING pathway by inducing DNA damage. Therefore, combination of prexasertib and immune checkpoint inhibitors will be a potential therapeutic strategy for MYC-overexpressed TNBC. In conclusion, our findings demonstrated that MYC overexpression characterizes an aggressive TNBC subtype, enabling synergistic lethality with CHK1 inhibitors. CHK1 inhibitors will be a potential therapeutic strategy in TNBC patients with MYC overexpression.
Collapse
Affiliation(s)
- Bin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tiantian Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingchuan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Duancheng Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenxia Peng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Juan Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Wang X, Li X, Li D, Zhang Y, Bai B, Chai B, Wen Z. A novel lncRNA YIL163C enhances genomic stability and antifungal resistance via the DNA damage response in Saccharomyces cerevisiae. Front Microbiol 2025; 16:1571797. [PMID: 40376465 PMCID: PMC12078222 DOI: 10.3389/fmicb.2025.1571797] [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: 02/06/2025] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Long non-coding RNAs (lncRNAs) are increasingly recognized as key regulators in cellular processes, including the DNA damage response (DDR). In Saccharomyces cerevisiae, DDR is critical for maintaining genomic integrity under stress, mediated by proteins like Mec1 and Rad53. However, the involvement of lncRNAs in DDR pathways, remains largely unexplored. This study investigates the function of a novel lncRNA, YIL163C, in promoting cell survival and genomic stability under DNA damage conditions. Methods Genetic suppressor screening was employed to assess the role of YIL163C in rescuing lethality in mec1Δ sml1Δ and rad53Δ sml1Δ exposed to DNA damage. Proteomic and phosphoproteomic analyses were conducted to evaluate changes in protein abundance and phosphorylation states. The impact of YIL163C on DDR and antifungal drug tolerance, specifically to 5-fluorocytosine, was also examined. Results Overexpression of YIL163C was found to rescue lethality in mec1Δ sml1Δ and rad53Δ sml1Δ under DNA damage conditions. Proteomic analyses revealed that YIL163C modulates pathways related to DNA replication, ER stress response, and ribosome biogenesis, enhancing cellular resilience to HU-induced stress. Additionally, YIL163C reduced sensitivity to 5-fluorocytosine, indicating a role in antifungal drug tolerance. Phosphoproteomic data suggested YIL163C influences phosphorylation states, potentially acting downstream of the Mec1-Rad53 signaling pathway. Conclusion This study provides new insights into the regulatory mechanisms of lncRNAs in DDR, with broader implications for antifungal therapy and genomic stability research, emphasizing the role of lncRNAs in stress responses beyond traditional protein-centric mechanisms.
Collapse
Affiliation(s)
- Xueting Wang
- Department of Dermatology, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for New Technology Research of Vegetables, Vegetable Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Xuemei Li
- Department of Dermatology, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Duoyun Li
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Yiying Zhang
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Bing Bai
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Bao Chai
- Department of Dermatology, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| | - Zewen Wen
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, Shenzhen Nanshan People’s Hospital, Affiliated Nanshan Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Halim CE, Deng S, Crasta KC, Yap CT. Interplay Between the Cytoskeleton and DNA Damage Response in Cancer Progression. Cancers (Basel) 2025; 17:1378. [PMID: 40282554 PMCID: PMC12025774 DOI: 10.3390/cancers17081378] [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: 03/05/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
DNA damage has emerged as a critical factor in fuelling the development and progression of cancer. DNA damage response (DDR) pathways lie at the crux of cell fate decisions following DNA damage induction, which can either trigger the repair of detrimental DNA lesions to protect cancer cells or induce the cell death machinery to eliminate damaged cells. Cytoskeletal dynamics have a critical role to play and influence the proper function of DDR pathways. Microfilaments, intermediate filaments, microtubules, and their associated proteins are well involved in the DDR. For instance, they are not only implicated in the recruitment of specific DDR molecules to the sites of DNA damage but also in the regulation of the mobility of the damaged DNA to repair sites in the periphery of the nucleus. The exquisite roles that these cytoskeletal proteins play in different DDR pathways, such as non-homologous end joining (NHEJ), homologous recombination (HR), base excision repair (BER), and nucleotide excision repair (NER), in cancer cells are extensively discussed in this review. Many cancer treatments are reliant upon inducing DNA damage in cancer cells to eliminate them; thus, it is important to shed light on factors that could affect their efficacy. Although the cytoskeleton is intricately involved in the DDR process, this has often been overlooked in cancer research and has not been exploited in developing DDR-targeting cancer therapy. Understanding the interplay between the cytoskeleton and the DDR in cancer will then provide insights into improving the development of cancer therapies that can leverage the synergistic action of DDR inhibitors and cytoskeleton-targeting agents.
Collapse
Affiliation(s)
- Clarissa Esmeralda Halim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Karen Carmelina Crasta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (C.E.H.); (S.D.); (K.C.C.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
5
|
Peng S, Long M, Chen Q, Yin Z, Zeng C, Zhang W, Wen Q, Zhang X, Ke W, Wu Y. Perspectives on cancer therapy-synthetic lethal precision medicine strategies, molecular mechanisms, therapeutic targets and current technical challenges. Cell Death Discov 2025; 11:179. [PMID: 40240755 PMCID: PMC12003663 DOI: 10.1038/s41420-025-02418-8] [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: 11/17/2024] [Revised: 02/27/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, synthetic lethality has become an important theme in the field of targeted cancer therapy. Synthetic lethality refers to simultaneous defects in two or more genes leading to cell death, whereas defects in any single gene do not lead to cell death. Taking advantage of the genetic vulnerability that exists within cancer cells, it theoretically has no negative impact on healthy cells and has fewer side effects than non-specific chemotherapy. Currently, targeted cancer therapies focus on inhibiting key pathways in cancer. However, it has been found that over-activation of oncogenic-related signaling pathways can also induce cancer cell death, which is a major breakthrough in the new field of targeted therapies. In this review, we summarize the conventional gene targets in synthetic lethality (PARP, ATR, ATM, WEE1, PRMT) and provide an in-depth analysis of their latest potential mechanisms. We explore the impact of over-activation of pathways such as PI3K/AKT, MAPK, and WNT on cancer cell survival, and present the technical challenges of current research. Important theoretical foundations and insights are provided for the application of synthetic lethal strategies in cancer therapy, as well as future research directions.
Collapse
Affiliation(s)
- Shixuan Peng
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Mengle Long
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Qisheng Chen
- Department of Anesthesiology, The First People's Hospital of Chenzhou, The Chenzhou Affiliated Hospital, Hengyang Medical School, University of South China, Chenzhou, Hunan, 423000, China
| | - Zhijian Yin
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Chang Zeng
- Department of Pathology, Yueyang Central Hospital, Yueyang, China
| | - Wanyong Zhang
- Department of Pathology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, 437100, Hubei, China
| | - Qingyang Wen
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Xinwen Zhang
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Weiqi Ke
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China.
| | - Yongjun Wu
- Department of Pathology, Xiangtan Center Hospital, Xiangtan City, Hunan province, 411100, China.
- Department of Pathology, The Affiliated Hospital of Hunan University, Xiangtan City, Hunan Province, China.
| |
Collapse
|
6
|
Cheng B, Ding Z, Hong Y, Wang Y, Zhou Y, Chen J, Peng X, Zeng C. Research progress in DNA damage response (DDR)-targeting modulators: From hits to clinical candidates. Eur J Med Chem 2025; 287:117347. [PMID: 39908794 DOI: 10.1016/j.ejmech.2025.117347] [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: 12/25/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
In recent years, synthetic lethality has been regarded as a sound example of cancer treatment. Identifying a growing number of synthetic lethality targets has led to a substantial broadening of the application of synthetic lethality, well beyond the PAPR inhibitors employed for treating tumors with BRCA1/2 deficiencies. Especially, molecular targets within the DDR have furnished inhibitor sources and have rapidly advanced to clinical trials. In this review, we summarize the DDR-associated synthetic lethality targets such as WRN, USP1, PARP, ATR, DNA-PK, PRMT5, POLQ, and WEE1. These targets allow for the development of targeted modulators like inhibitors and degraders. Additionally, we emphasize the rational design, advantages, and potential limitations. Furthermore, we outline the promising future of DDR-targeted drug development.
Collapse
Affiliation(s)
- Binbin Cheng
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China; Department of Cardiology, Central Laboratory of Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Zongbao Ding
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Yimeng Hong
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China
| | - Yaping Wang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China
| | - Yingxing Zhou
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China; Huangshi Key Laboratory of Molecular Diagnosis and Individualized Treatment, Huangshi Love&health Hospital Affiliated of Hubei Polytechnic University, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaopeng Peng
- College of Pharmacy, Gannan Medical University, Ganzhou, 314000, China.
| | - Chunlai Zeng
- Department of Cardiology, Central Laboratory of Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China.
| |
Collapse
|
7
|
Wang C, Han X, Kong S, Zhang S, Ning H, Wu F. Deciphering the mechanisms of PARP inhibitor resistance in prostate cancer: Implications for precision medicine. Biomed Pharmacother 2025; 185:117955. [PMID: 40086424 DOI: 10.1016/j.biopha.2025.117955] [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: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Prostate cancer is a leading malignancy among men. While early-stage prostate cancer can be effectively managed, metastatic prostate cancer remains incurable, with a median survival of 3-5 years. The primary treatment for advanced prostate cancer is androgen deprivation therapy (ADT), but resistance to ADT often leads to castrationresistant prostate cancer (CRPC), presenting a significant therapeutic challenge. The advent of precision medicine has introduced promising new treatments, including PARP inhibitors (PARPi), which target defects in DNA repair mechanisms in cancer cells. PARPi have shown efficacy in treating advanced prostate cancer, especially in patients with metastatic CRPC (mCRPC) harboring homologous recombination (HR)-associated gene mutations. Despite these advancements, resistance to PARPi remains a critical issue. Here, we explored the primary mechanisms of PARPi resistance in prostate cancer. Key resistance mechanisms include homologous recombination recovery through reverse mutations in BRCA genes, BRCA promoter demethylation, and non-degradation of mutated BRCA proteins. The tumor microenvironment and overactivation of the base excision repair pathway also play significant roles in bypassing PARPi-induced synthetic lethality. In addition, we explored the clinical implications and therapeutic strategies to overcome resistance,emphasizing the need for precision medicine approaches. Our findings highlight the need for comprehensive strategies to improve PARPi sensitivity and effectiveness,ultimately aiming to extend patient survival and improve the quality of life for those with advanced prostate cancer. As our understanding of PARPi resistance evolves, more diverse and effective individualized treatment regimens will emerge.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China
| | - Xiaoran Han
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shaoqiu Kong
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shanhua Zhang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Hao Ning
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| | - Fei Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| |
Collapse
|
8
|
Choe H, Wang Z, Huang J, Yang Y, Zhao Z, Jo H, Pak H, Ali T, Ding K, Ma J, Li L, Shang D, Zhang L. Broussoflavonol B induces S-phase arrest and apoptosis in pancreatic cancer cells by modulating the cell cycle checkpoint through inhibition of the AURKA/PLK1 pathway. Cancer Cell Int 2025; 25:100. [PMID: 40098146 PMCID: PMC11916852 DOI: 10.1186/s12935-025-03717-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Broussoflavonol B (Bf-B), a flavonoid compound identified in the roots of Daphne giraldii Nitsche, has been extensively investigated for its potential anti-inflammatory, antioxidant, and anticancer properties. However, the precise mechanism underlying the regulation of AURKA/PLK1 pathway-mediated cell cycle arrest by Bf-B in pancreatic cancer remains poorly understood. PURPOSE The objective of this study was to investigate the inhibitory effect of Bf-B on pancreatic ductal adenocarcinoma (PDAC) and its underlying mechanism. METHODS A CCK8 assay was conducted to identify the flavonoids with the highest inhibitory activity against PANC-1, the pancreatic cancer cell line among the 25 flavonoids. Through bioinformatics analysis and molecular docking, the pathogenic targets of pancreatic cancer and flavonoid-related targets were explored, and the key targets and signaling pathways of drug intervention in pancreatic cancer were analyzed. The viability and migration ability of pancreatic cancer cells were assessed following treatment with Bf-B via the CCK8, colony formation, and wound healing assays. The cell cycle distribution and cell apoptosis were analyzed through flow cytometry and Hoechst staining. Western blotting and qPCR were employed to investigate the expression of relevant proteins and genes. For in vivo experiments, we employed a xenograft mouse model to evaluate the anticancer efficacy of Bf-B. Immunohistochemistry and immunofluorescence assays were employed to investigate the expression of relevant proteins. RESULTS In this study, the structure‒activity relationships of 25 flavonoids were evaluated. The results demonstrated that Bf-B with diisopentenyl has potent cytotoxic effects on PANC-1 cancer cells. AURKA, PLK1, and MET might serve as key targets for Bf-B inhibition of disease progression in PDAC patients. The results demonstrated that Bf-B inhibits the proliferation and migration of PANC-1 and BXPC-3 cells and induces cell cycle S-phase arrest, apoptosis, and DNA damage. Moreover, the results of western blot and qPCR experiments indicated that Bf-B exerts anticancer effects by downregulating the expression of the genes encoding AURKA/PLK1, the cell cycle checkpoint kinase ATR/CHK1/CDC25C, and Cyclin B1/CDK1 signaling pathway-related proteins and upregulating the expression of PP53, P21, and histone H2A. XS139ph expression. In xenograft-bearing mice, AURKA/PLK1 expression was reduced in a dose-dependent manner, accompanied by an increase in histone H2A. XS139ph expression. CONCLUSION Bf-B might be a potent therapeutic agent for pancreatic cancer because of its ability to suppress the expression of AURKA/PLK1.
Collapse
Affiliation(s)
- HyokChol Choe
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
- Department of Clinical Medicine, Sinuiju Medical University, Sinuiju, Korea
| | - Zhen Wang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianhua Huang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Yutong Yang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Zhihao Zhao
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - HyonSu Jo
- Pyongyang University of Medical Sciences, Pyongyang, Korea
| | - HyonU Pak
- Pyongyang University of Medical Sciences, Pyongyang, Korea
| | - Tanveer Ali
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Kaiyue Ding
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Junnan Ma
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China
| | - Lingzhi Li
- The College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, China
| | - Dong Shang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China.
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Lin Zhang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 0411, China.
| |
Collapse
|
9
|
Luo L, Jiang C, Xie S. The mechanism of high mobility group box-1 in the proliferation and macrophage polarization in esophageal squamous cell carcinoma cells. Eur J Med Res 2025; 30:144. [PMID: 40022250 PMCID: PMC11869724 DOI: 10.1186/s40001-025-02390-0] [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: 11/07/2024] [Accepted: 02/17/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Previous studies showed that high mobility group box-1 (HMGB1) facilitates the initiation and progression of esophageal squamous cell carcinoma (ESCC), and the current research investigated the detailed mechanisms implicated. METHODS The impact of HMGB1 and IGFBP3 levels on the survival of ESCC was examined by plotting Kaplan-Meier (KM) curves based on the data collected from The Cancer Genome Atlas (TCGA). Quantitative real-time PCR (qRT-PCR) was performed to detect the expressions of HMGB1 in both human esophageal epithelial cells (HEEC) and ESCC cells. After cell transfection, the proliferation of ESCC cells was measured, and the cell metastasis was determined based on the levels of cadherins (CDHs) and Vimentin (VIM). Macrophage polarization was determined by calculating the mean fluorescence intensity (MFI) of CD206 and CD86. In addition, co-immunoprecipitation and immunoblotting were applied to evaluate the interaction between insulin-like growth factor binding protein 3 (IGFBP3)/DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and HMGB1. RESULTS A high level of HMGB1 was predictive of an unfavorable prognosis of ESCC (p < 0.05). HMGB1 showed a higher expression in ESCC cells (p < 0.05), while knockdown of HMGB1 inhibited ESCC cell proliferation, downregulated the levels of CDH2 and VIM and upregulated the level of CDH1 (p < 0.05). In contrast, overexpressed HMGB1 showed the opposite effects (p < 0.05), suggesting the role of HMGB1 in the epithelial-mesenchymal transition (EMT) of ESCC. After the knockout of HMGB1, the MFI of CD86 was increased but that of CD206 was reduced, indicating the polarization towards M1 macrophages (p < 0.05). However, the results were reversed when HMGB1 was overexpressed (p < 0.05). Meanwhile, HMGB1 could interact with the IGFBP3/DNA-PKcs complex (p < 0.05). Low-expressed IGFBP3 was predictive of an unfavorable prognosis of ESCC, and IGFBP3 silencing promoted the proliferation of ESCC cells (p < 0.05). Besides, HMGB1 and IGFBP3 could act antagonistically in influencing the proliferation of ESCC cells and macrophage polarization. CONCLUSIONS Through in vitro experiments, this study found that HMGB1 was linked to the proliferation and polarization of macrophages in ESCC, providing novel evidence for the role of HMGB1 in ESCC development.
Collapse
Affiliation(s)
- Liling Luo
- Department of Radiation Oncology, Guangdong Provincial People'S Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, China
| | - Chao Jiang
- Department of Radiation Oncology, The People's Hospital of Shenzhen Baoan District, the Second Affiliated Hospital of Shenzhen University, Shenzhen, 518100, China
| | - Songxi Xie
- Department of Radiation Oncology, Guangdong Provincial People'S Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
10
|
Nadin SB, Cuello-Carrión FD, Cayado-Gutiérrez N, Fanelli MA. Overview of Wnt/β-Catenin Pathway and DNA Damage/Repair in Cancer. BIOLOGY 2025; 14:185. [PMID: 40001953 PMCID: PMC11851563 DOI: 10.3390/biology14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
The Wnt/β-catenin pathway takes part in important cellular processes in tumor cells, such as gene expression, adhesion, and survival. The canonical pathway is activated in several tumors, and β-catenin is its major effector. The union of Wnt to the co-receptor complex causes the inhibition of GSK3β activity, thus preventing the phosphorylation and degradation of β-catenin, which accumulates in the cytoplasm, to subsequently be transported to the nucleus to associate with transcription factors. The relationship between Wnt/β-catenin and DNA damage/repair mechanisms has been a focus for the last few years. Studying the Wnt/β-catenin network interactions with DNA damage/repair proteins has become a successful research field. This review provides an overview of the participation of Wnt/β-catenin in DNA damage/repair mechanisms and their future implications as targets for cancer therapy.
Collapse
Affiliation(s)
- Silvina B. Nadin
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro Científico Tecnológico (CCT), Mendoza 5500, Argentina
| | - F. Darío Cuello-Carrión
- Laboratorio de Oncología, IMBECU, CONICET, CCT, Mendoza 5500, Argentina; (F.D.C.-C.); (N.C.-G.); (M.A.F.)
| | - Niubys Cayado-Gutiérrez
- Laboratorio de Oncología, IMBECU, CONICET, CCT, Mendoza 5500, Argentina; (F.D.C.-C.); (N.C.-G.); (M.A.F.)
- Cátedra de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina
| | - Mariel A. Fanelli
- Laboratorio de Oncología, IMBECU, CONICET, CCT, Mendoza 5500, Argentina; (F.D.C.-C.); (N.C.-G.); (M.A.F.)
| |
Collapse
|
11
|
Ivanenkov YA, Malyshev AS, Terentiev VA, Korzhenevskaya AA, Evteev SA, Vatsadze SZ, Medved'ko AV, Shegai PV, Kaprin AD. Small molecule ATM inhibitors as potential cancer therapy: a patent review (2003-present). Expert Opin Ther Pat 2025; 35:111-136. [PMID: 39727182 DOI: 10.1080/13543776.2024.2446228] [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: 09/05/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION The ataxia telangiectasia mutated kinase (ATM) is key in coordinating the DDR signaling network essential for responding to double-strand breaks (DSBs). Several ATM inhibitors are being investigated for potential anticancer treatment in clinical trials. AREAS COVERED This review aims to provide a comprehensive overview of patents and patent applications since 2003, with a particular focus on the structural properties, activity and efficacy of the claimed ATM kinase small-molecule inhibitors. The search was conducted using SciFinder, Cortellis Drug Discovery Intelligence Database, and Espacenet. After filtering, 44 records were identified for further analysis. This paper also discusses the recent progress in the clinical trials and development history. EXPERT OPINION ATM kinase is a promising target for cancer therapy. Small-molecule ATM kinase inhibitors hold significant potential in cancer treatment by enhancing the efficacy of existing DNA-damaging therapies. Patent analysis revealed that the majority of these compounds contain imidazo[4,5-c]quinolinone scaffold or its bioisosteric variations which are optimal in terms of good ATM inhibitory activity and selectivity over closely related enzymes. Clinical trials explore combinations with RT or DNA-targeted compounds like PARP inhibitors, which induce DSBs. The medicinal chemistry field anticipates that these therapeutic options will soon be available on the pharmaceutical market.
Collapse
Affiliation(s)
- Yan A Ivanenkov
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russian Federation
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, Russian Federation
| | - Alexander S Malyshev
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russian Federation
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, Russian Federation
| | - Victor A Terentiev
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russian Federation
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, Russian Federation
| | | | - Sergei A Evteev
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russian Federation
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, Russian Federation
| | - Sergey Z Vatsadze
- N.D. Zelinsky Institute of Organic Chemistry, Moscow, Russian Federation
| | | | - Petr V Shegai
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russian Federation
| | - Andrey D Kaprin
- P. Hertsen Moscow Oncology Research Institute, Moscow, Russian Federation
- Peoples' Friendship University of Russia (RUDN), Moscow, Russian Federation
| |
Collapse
|
12
|
Yang T, Fan Y, Bai G, Huang Y. RFC4 confers radioresistance of esophagus squamous cell carcinoma through regulating DNA damage response. Am J Physiol Cell Physiol 2025; 328:C367-C380. [PMID: 39672175 DOI: 10.1152/ajpcell.00533.2024] [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: 07/31/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/15/2024]
Abstract
Radioresistance in esophageal squamous cell carcinoma (ESCC) is a critical factor leading to treatment failure and recurrence, yet its underlying molecular mechanisms remain unclear. This study aimed to investigate the role of replication factor C4 (RFC4) in ESCC radioresistance and to explore the underlying mechanisms. We utilized online bioinformatics tools to analyze the properties, functions, and prognostic significance of RFC4 in ESCC. We established cell lines with varying RFC4 expression levels and subjected them to radiation exposure. RFC4 expression was assessed using quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry, and immunoblotting. Cell proliferation was evaluated with MTT, 5-ethynyl-2'-deoxyuridine (EdU), and colony formation assays. Apoptosis and cell cycle distribution were analyzed by flow cytometry. Western blotting and immunofluorescence were used to study the impact of RFC4 on the DNA damage response in ESCC cells. A xenograft mouse model was employed to assess tumor growth in vivo. RFC4 expression was significantly upregulated in ESCC tissues and cells, particularly in radioresistant cases. Functional experiments revealed that RFC4 promotes cell proliferation, inhibits apoptosis, induces cell cycle arrest, and mitigates radiation-induced DNA damage responses. Mechanistically, RFC4-mediated radioresistance in ESCC may involve the inactivation of the p53 signaling pathway. In animal studies, RFC4 knockdown, either alone or in combination with radiation therapy, effectively suppressed the growth of xenograft tumors. These findings highlight the potential of targeting RFC4 to overcome radioresistance by modulating the DNA damage response in ESCC, offering promising therapeutic avenues for patients with ESCC.NEW & NOTEWORTHY Our research indicates that replication factor C4 (RFC4) plays a role in conferring radioresistance to esophageal squamous cell carcinoma (ESCC) by bolstering DNA damage repair, primarily through the inhibition of the p53 signaling pathway. This finding positions RFC4 as a promising therapeutic target for combating radioresistance in ESCC, although further research is required to fully comprehend its intricate role in the disease.
Collapse
Affiliation(s)
- Tao Yang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yue Fan
- Discipline Construction Office, Baoan Central Hospital of Shenzhen, Shenzhen, China
| | - Guang Bai
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yinpeng Huang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
13
|
Mi L, Zhang H. Myriad factors and pathways influencing tumor radiotherapy resistance. Open Life Sci 2024; 19:20220992. [PMID: 39655194 PMCID: PMC11627069 DOI: 10.1515/biol-2022-0992] [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: 05/07/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 12/12/2024] Open
Abstract
Radiotherapy is a cornerstone in the treatment of various tumors, yet radioresistance often leads to treatment failure and tumor recurrence. Several factors contribute to this resistance, including hypoxia, DNA repair mechanisms, and cancer stem cells. This review explores the diverse elements that drive tumor radiotherapy resistance. Historically, resistance has been attributed to cellular repair and tumor repopulation, but recent research has expanded this understanding. The tumor microenvironment - characterized by hypoxia, immune evasion, and stromal interactions - further complicates treatment. Additionally, molecular mechanisms such as aberrant signaling pathways, epigenetic modifications, and non-B-DNA structures play significant roles in mediating resistance. This review synthesizes current knowledge, highlighting the interplay of these factors and their clinical implications. Understanding these mechanisms is crucial for developing strategies to overcome resistance and improve therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Lanjuan Mi
- School of Life and Health Sciences, Huzhou College, Hu Zhou, China
| | - Hongquan Zhang
- The First Affiliated Hospital of Huzhou University, Hu Zhou, China
| |
Collapse
|
14
|
Hu D, Li M, Chen X. DLX5 Promotes Radioresistance in Renal Cell Carcinoma by Upregulating c-Myc Expression. FRONT BIOSCI-LANDMRK 2024; 29:400. [PMID: 39614438 DOI: 10.31083/j.fbl2911400] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a prevalent and aggressive kidney cancer with notable metastatic potential. While radiotherapy is effective for treating metastatic RCC, the emergence of radioresistance presents a major challenge. This study explores the role of DLX5, previously identified as an oncogene in various cancers, in the development of radioresistance in RCC. METHODS Distal-less homeobox 5 (DLX5) expression was measured using western blot analysis. To study the effects of DLX5, its expression was knocked down in 786-O and Caki-1 RCC cell lines through si-DLX5 transfection, and the impact of DLX5 on RCC cell proliferation and radioresistance was assessed using cell counting kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU) incorporation assay, flow cytometry, colony formation, immunofluorescence, and western blot assays. The underlying mechanisms were explored through western blot, colony formation, and CCK-8 assays. In vivo effects were examined using a xenograft mouse model. RESULTS In silico results showed increased DLX5 levels in RCC tissues. Similarly, DLX5 expression was elevated in RCC cell lines. Silencing DLX5 reduced RCC cell proliferation and induced apoptosis in vitro. Additionally, DLX5 knockdown decreased radioresistance and increased DNA damage in RCC cells. Mechanistic studies revealed that DLX5 promotes radioresistance through the upregulation of c-Myc. In vivo, DLX5 silencing impeded tumor growth and reduced radioresistance. CONCLUSION DLX5 contributes to RCC cell growth and radioresistance by upregulating c-Myc expression, highlighting its potential as a target for overcoming radioresistance in RCC.
Collapse
Affiliation(s)
- Danfei Hu
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, 315000,Ningbo, Zhejiang, China
| | - Mingyao Li
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, 315000,Ningbo, Zhejiang, China
| | - Xiaodong Chen
- Department of Urology, Ningbo Urology and Nephrology Hospital, 315100 Ningbo, Zhejiang, China
| |
Collapse
|
15
|
Satish S, Athavale M, Kharkar PS. Targeted therapies for Glioblastoma multiforme (GBM): State-of-the-art and future prospects. Drug Dev Res 2024; 85:e22261. [PMID: 39485272 DOI: 10.1002/ddr.22261] [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: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024]
Abstract
Glioblastoma multiforme (GBM) remains one of the most aggressive and lethal forms of brain cancer, characterized by rapid growth and resistance to conventional therapies. The present review explores the latest advancements in targeted therapies for GBM, emphasizing the critical role of the blood-brain barrier (BBB), blood-brain-tumor barrier, tumor microenvironment, and genetic mutations in influencing treatment outcomes. The impact of the key hallmarks of GBM, for example, chemoresistance, hypoxia, and the presence of glioma stem cells on the disease progression and multidrug resistance are discussed in detail. The major focus is on the innovative strategies aimed at overcoming these challenges, such as the use of monoclonal antibodies, small-molecule inhibitors, and novel drug delivery systems designed to enhance drug penetration across the BBB. Additionally, the potential of immunotherapy, specifically immune checkpoint inhibitors and vaccine-based approaches, to improve patient prognosis was explored. Recent clinical trials and preclinical studies are reviewed to provide a comprehensive overview of the current landscape and future prospects in GBM treatment. The integration of advanced computational models and personalized medicine approaches is also considered, aiming to tailor therapies to individual patient profiles for better efficacy. Overall, while significant progress has been made in understanding and targeting the complex biology of GBM, continued research and clinical innovation are imperative to develop more effective and sustainable therapeutic options for patients battling this formidable disease.
Collapse
Affiliation(s)
- Smera Satish
- Sathgen Therapeutics, Godavari Biorefineries Limited, Somaiya Group Company, Mumbai, India
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Maithili Athavale
- Sathgen Therapeutics, Godavari Biorefineries Limited, Somaiya Group Company, Mumbai, India
| | - Prashant S Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
16
|
Moosavi F, Hassani B, Nazari S, Saso L, Firuzi O. Targeting DNA damage response in pancreatic ductal adenocarcinoma: A review of preclinical and clinical evidence. Biochim Biophys Acta Rev Cancer 2024; 1879:189185. [PMID: 39326802 DOI: 10.1016/j.bbcan.2024.189185] [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: 03/06/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with one of the most unfavorable prognoses across all malignancies. In this review, we investigate the role of inhibitors targeting crucial regulators of DNA damage response (DDR) pathways, either as single treatments or in combination with chemotherapeutic agents and targeted therapies in PDAC. The most prominent clinical benefit of PARP inhibitors' monotherapy is related to the principle of synthetic lethality in individuals harboring BRCA1/2 and other DDR gene mutations as predictive biomarkers. Moreover, induction of BRCAness with inhibitors of RTKs, including VEGFR and c-MET and their downstream signaling pathways, RAS/RAF/MEK/ERK and PI3K/AKT/mTOR in order to expand the application of PARP inhibitors in patients without DDR mutations, has also been addressed. Other DDR-targeting agents beyond PARP inhibitors, including inhibitors of ATM, ATR, CHEK1/2, and WEE1 have also demonstrated their potential in preclinical models of PDAC and may hold promise in future studies.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Hassani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
17
|
Sun T, Liu C, Kong L, Zha J, Ni G. Cold plasma irradiation inhibits skin cancer via ferroptosis. Biomed Phys Eng Express 2024; 10:065036. [PMID: 39390682 DOI: 10.1088/2057-1976/ad8200] [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: 06/05/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Cold atmospheric plasma (CAP) has been extensively utilized in medical treatment, particularly in cancer therapy. However, the underlying mechanism of CAP in skin cancer treatment remains elusive. In this study, we established a skin cancer model using CAP treatmentin vitro. Also, we established the Xenograft experiment modelin vivo. The results demonstrated that treatment with CAP induced ferroptosis, resulting in a significant reduction in the viability, migration, and invasive capacities of A431 squamous cell carcinoma, a type of skin cancer. Mechanistically, the significant production of reactive oxygen species (ROS) by CAP induces DNA damage, which then activates Ataxia-telangiectasia mutated (ATM) and p53 through acetylation, while simultaneously suppressing the expression of Solute Carrier Family 7 Member 11 (SLC7A11). Consequently, this cascade led to the down-regulation of intracellular Glutathione peroxidase 4 (GPX4), ultimately resulting in ferroptosis. CAP exhibits a favorable impact on skin cancer treatment, suggesting its potential medical application in skin cancer therapy.
Collapse
Affiliation(s)
- Tao Sun
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, People's Republic of China
- University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Changqing Liu
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, People's Republic of China
- University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Ling Kong
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, People's Republic of China
- University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Jingjing Zha
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, People's Republic of China
- Anhui Province Key Laboratory of Medical Physics and Technology, Hefei 230031, People's Republic of China
| | - Guohua Ni
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, People's Republic of China
- Anhui Province Key Laboratory of Medical Physics and Technology, Hefei 230031, People's Republic of China
| |
Collapse
|
18
|
Li L, Wang X, Jiang M, Li L, Wang D, Li Y. Advancements in a novel model of autophagy and immune network regulation in radioresistance of cancer stem cells. Biomed Pharmacother 2024; 179:117420. [PMID: 39255736 DOI: 10.1016/j.biopha.2024.117420] [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: 06/25/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Radiotherapy, a precise modality for treating malignant tumors, has undergone rapid advancements in primary and clinical research. The mechanisms underlying tumor radioresistance have become significant research. With the introduction and in-depth study of cancer stem cells (CSCs) theory, CSCs have been identified as the primary factor contributing to the development of tumor radioresistance. The "stemness" of CSCs is a biological characteristic of a small subset of cells within tumor tissues, characterized by self-renewal solid ability. This characteristic leads to resistance to radiotherapy, chemotherapy, and targeted therapies, driving tumor recurrence and metastasis. Another study revealed that cellular autophagy plays a pivotal role in maintaining the "stemness" of CSCs. Autophagy is a cellular mechanism that degrades proteins and organelles to generate nutrients and energy in response to stress. This process maintains cellular homeostasis and contributes to CSCs radioresistance. Furthermore, ionizing radiation (IR) facilitates epithelial-to-mesenchymal transition (EMT), vascular regeneration, and other tumor processes by influencing the infiltration of M2-type tumor-associated macrophages (TAMs). IR promotes the activation of the classical immunosuppressive "switch," PD-1/PD-L1, which diminishes T-cell secretion, leading to immune evasion and promoting radioresistance. Interestingly, recent studies have found that the immune pathway PD-1/PD-L1 is closely related to cellular autophagy. However, the interrelationships between immunity, autophagy, and radioresistance of CSCs and the regulatory mechanisms involved remain unclear. Consequently, this paper reviews recent research to summarize these potential connections, aiming to establish a theoretical foundation for future studies and propose a new model for the network regulation of immunity, autophagy, and radioresistance of tumor cells.
Collapse
Affiliation(s)
- Leyao Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Xin Wang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Mei Jiang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Lei Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Di Wang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yajun Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China.
| |
Collapse
|
19
|
Huang C, Zhang K, Ren Y, Liu X, Li Y, Yang B, Chen P, Zhang M, Lu X, Zhuo Y, Qi C, Cai K. A manganese-doped layered double hydroxide loaded with lactate oxidase and DNA repair inhibitors for synergistically enhanced tumor immunotherapy. Acta Biomater 2024; 187:340-351. [PMID: 39218280 DOI: 10.1016/j.actbio.2024.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Tumor immunotherapy has gained more and more attention in tumor treatment. However, the accumulation of lactic acid in tumor tissue inhibits the response of immune cells to form an immunosuppressive microenvironment (ISME). To reverse the ISME, an acid-responsive nanoplatform (termed as MLLN@HA) is reported for synergistically enhanced tumor immunotherapy. MLLN@HA is constructed by the co-loading of lactate oxidase (LOX) and DNA repair inhibitor (NU7441) in a manganese-doped layered double hydroxide (Mn-LDH), and then modified with hyaluronic acid (HA) for tumor-targeted delivery. After endocytosis by tumor cells, MLLN@HA decomposes and releases LOX, NU7441 and Mn2+ ions in the acidic tumor microenvironment. The released LOX catalyzes the conversion of lactic acid into hydrogen peroxide (H2O2), which not only alleviates the ISME, but also provides reactants for the Mn2+-mediated Fenton-like reaction to enhance chemodynamic therapy (CDT). Released NU7441 prevents CDT-induced DNA damage from being repaired, thereby increasing double-stranded DNA (dsDNA) fragments within tumor cells. Importantly, the released Mn2+ ions enhance the sensitivity of cyclic GMP-AMP synthase (cGAS) to dsDNA fragments, and activate the stimulator of interferon genes (STING) to induce an anti-tumor immune response. Such an orchestrated immune-boosting strategy ultimately achieves effective tumor growth inhibition and prevents tumor lung metastasis. STATEMENT OF SIGNIFICANCE: To improve the efficacy of tumor immunotherapy, an innovative acid-responsive MLLN@HA nanoplatform was developed for synergistically enhanced tumor immunotherapy. The MLLN@HA actively targets to tumor cells through the interaction of HA with CD44, and then degrades to release LOX, NU7441 and Mn2+ ions in the acidic tumor microenvironment. The released LOX generates H2O2 for the Mn2+-mediated Fenton reaction and reverses the ISME by consuming lactate. NU7441 prevents DNA damage repair, leading to an increased concentration of free DNA fragments, while Mn2+ ions activate the cGAS-STING pathway, enhancing the systemic anti-tumor immune response. The orchestrated immune-boosting nanoplatform effectively inhibits tumor growth and lung metastasis, presenting a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Chengyao Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ke Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yu Ren
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xihong Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yan Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Bangliu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Peiran Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mingyue Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xiaotong Lu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yuhong Zhuo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Chao Qi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
20
|
Qian J, Liao G, Chen M, Peng RW, Yan X, Du J, Huang R, Pan M, Lin Y, Gong X, Xu G, Zheng B, Chen C, Yang Z. Advancing cancer therapy: new frontiers in targeting DNA damage response. Front Pharmacol 2024; 15:1474337. [PMID: 39372203 PMCID: PMC11449873 DOI: 10.3389/fphar.2024.1474337] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Genomic instability is a core characteristic of cancer, often stemming from defects in DNA damage response (DDR) or increased replication stress. DDR defects can lead to significant genetic alterations, including changes in gene copy numbers, gene rearrangements, and mutations, which accumulate over time and drive the clonal evolution of cancer cells. However, these vulnerabilities also present opportunities for targeted therapies that exploit DDR deficiencies, potentially improving treatment efficacy and patient outcomes. The development of PARP inhibitors like Olaparib has significantly improved the treatment of cancers with DDR defects (e.g., BRCA1 or BRCA2 mutations) based on synthetic lethality. This achievement has spurred further research into identifying additional therapeutic targets within the DDR pathway. Recent progress includes the development of inhibitors targeting other key DDR components such as DNA-PK, ATM, ATR, Chk1, Chk2, and Wee1 kinases. Current research is focused on optimizing these therapies by developing predictive biomarkers for treatment response, analyzing mechanisms of resistance (both intrinsic and acquired), and exploring the potential for combining DDR-targeted therapies with chemotherapy, radiotherapy, and immunotherapy. This article provides an overview of the latest advancements in targeted anti-tumor therapies based on DDR and their implications for future cancer treatment strategies.
Collapse
Affiliation(s)
- Jiekun Qian
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Cardiothoracic Surgery, Fujian Medical University, Fuzhou, China
| | - Guoliang Liao
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Maohui Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Ren-Wang Peng
- Division of General Thoracic Surgery, Department of BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xin Yan
- Department of Cardiac Medical Center Nursing, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianting Du
- Fujian Key Laboratory of Cardiothoracic Surgery, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Renjie Huang
- Fujian Key Laboratory of Cardiothoracic Surgery, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Maojie Pan
- Fujian Key Laboratory of Cardiothoracic Surgery, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Yuxing Lin
- Fujian Key Laboratory of Cardiothoracic Surgery, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Xian Gong
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Guobing Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Bin Zheng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Cardiothoracic Surgery, Fujian Medical University, Fuzhou, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| | - Zhang Yang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China
| |
Collapse
|
21
|
Yu Z, Teng Y, Yang H, Wang Y, Li X, Feng L, Xu W, Hao Y, Li Y. Inhibiting H2AX Can Ameliorate Myocardial Ischemia/Reperfusion Injury by Regulating P53/JNK Signaling Pathway. Cardiol Res Pract 2024; 2024:1905996. [PMID: 39257436 PMCID: PMC11387088 DOI: 10.1155/2024/1905996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/13/2024] [Indexed: 09/12/2024] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a significant area of focus in cardiovascular disease research. I/R injury can increase intracellular oxidative stress, leading to DNA damage. H2AX plays a crucial role in DNA repair. This study utilized mouse and cell models of myocardial I/R to investigate the impact of H2AX on cardiomyocytes during I/R. This study initially assessed the expression of H2AX in MI/R mice compared to a sham surgery group. Subsequently, cardiac function, infarct area, and mitochondrial damage were evaluated after inhibiting H2AX in MI/R mice and a negative control group. Furthermore, the study delved into the molecular mechanisms by analyzing the expression of H2AX, P53, p-JNK, SHP2, p-SHP2, p-RAS, parkin, Drp1, Cyt-C, Caspase-3, and Caspase-8 in cardiomyocytes following the addition of JNK or P53 agonists. The results from western blotting in vivo indicated significantly higher H2AX expression in the MI/R group compared to the sham group. Inhibiting H2AX improved cardiac function, reduced myocardial infarct area, and mitigated mitochondrial damage in the MI/R group. In vitro experiments demonstrated that inhibiting H2AX could attenuate mitochondrial damage and apoptosis in myocardial cells by modulating the P53 and JNK signaling pathways. These findings suggested that inhibiting H2AX may alleviate myocardial I/R injury through the regulation of the P53/JNK pathway, highlighting H2AX as a potential target for the treatment of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ziyang Yu
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yirong Teng
- Department of General Practice The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Hongbo Yang
- Department of Cardiology Fuwai Yunnan Hospital Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Yudi Wang
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Xichen Li
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Lei Feng
- Department of Laboratory Yan'an Hospital of Kunming City, Kunming, Yunnan, China
| | - Wenbo Xu
- Department of Laboratory The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yinglu Hao
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yanping Li
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| |
Collapse
|
22
|
Hui Z, Deng H, Zhang X, Garrido C, Lirussi F, Ye XY, Xie T, Liu ZQ. Development and therapeutic potential of DNA-dependent protein kinase inhibitors. Bioorg Chem 2024; 150:107608. [PMID: 38981210 DOI: 10.1016/j.bioorg.2024.107608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
The deployment of DNA damage response (DDR) combats various forms of DNA damage, ensuring genomic stability. Cancer cells' propensity for genomic instability offers therapeutic opportunities to selectively kill cancer cells by suppressing the DDR pathway. DNA-dependent protein kinase (DNA-PK), a nuclear serine/threonine kinase, is crucial for the non-homologous end joining (NHEJ) pathway in the repair of DNA double-strand breaks (DSBs). Therefore, targeting DNA-PK is a promising cancer treatment strategy. This review elaborates on the structures of DNA-PK and its related large protein, as well as the development process of DNA-PK inhibitors, and recent advancements in their clinical application. We emphasize our analysis of the development process and structure-activity relationships (SARs) of DNA-PK inhibitors based on different scaffolds. We hope this review will provide practical information for researchers seeking to develop novel DNA-PK inhibitors in the future.
Collapse
Affiliation(s)
- Zi Hui
- Xiangya School of Pharmaceutical Sciences, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410013, P. R. China; School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, P.R. China
| | - Haowen Deng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xuelei Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Carmen Garrido
- INSERM U1231, Label LipSTIC and Ligue Nationale contre le Cancer, Dijon, France; Faculté de médecine, Université de Bourgogne, Dijon, Centre de lutte contre le cancer Georges François Leclerc, 21000, Dijon, France
| | - Frédéric Lirussi
- INSERM U1231, Label LipSTIC and Ligue Nationale contre le Cancer, Dijon, France; Université de Franche Comté, France, University Hospital of Besançon (CHU), France
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, P.R. China.
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, P.R. China.
| | - Zhao-Qian Liu
- Xiangya School of Pharmaceutical Sciences, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410013, P. R. China.
| |
Collapse
|
23
|
Waldrip ZJ, Acharya B, Armstrong D, Hanafi M, Rainwater RR, Amole S, Fulmer M, Azevedo-Pouly AC, Burns A, Burdine L, Frett B, Burdine MS. Discovery of the DNA-PKcs inhibitor DA-143 which exhibits enhanced solubility relative to NU7441. Sci Rep 2024; 14:19999. [PMID: 39198533 PMCID: PMC11358143 DOI: 10.1038/s41598-024-70858-w] [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: 03/29/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
DNA-dependent protein kinase catalytic subunit (DNA-PKcs) plays a vital role in DNA damage repair and lymphocyte function, presenting a significant target in cancer and immune diseases. Current DNA-PKcs inhibitors are undergoing Phase I/II trials as adjuncts to radiotherapy and chemotherapy in cancer. Nevertheless, clinical utility is limited by suboptimal bioavailability. This study introduces DNA-PKcs inhibitors designed to enhance bioavailability. We demonstrate that a novel DNA-PKcs inhibitor, DA-143, surpasses NU7441 in aqueous solubility as well as other available inhibitors. In addition, DA-143 displayed an improvement in DNA-PKcs inhibition relative to NU7441 achieving an IC50 of 2.5 nM. Consistent with current inhibitors, inhibition of DNA-PKcs by DA-143 resulted in increased tumor cell sensitivity to DNA-damage from chemotherapy and inhibition of human T cell function. The improved solubility of DA-143 is critical for enhanced efficacy at reduced doses and facilitates more effective evaluation of DNA-PKcs inhibition in both preclinical and clinical development.
Collapse
Affiliation(s)
- Zachary J Waldrip
- Division of Surgical Research, Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Baku Acharya
- Department of Pharmaceutical Science, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Daniel Armstrong
- Department of Pharmaceutical Science, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Maha Hanafi
- Department of Pharmaceutical Science, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11526, Egypt
| | - Randall R Rainwater
- Division of Surgical Research, Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Sharon Amole
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Madeline Fulmer
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Ana Clara Azevedo-Pouly
- Division of Surgical Research, Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Alaina Burns
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Lyle Burdine
- Division of Surgical Research, Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Department of Transplant Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Brendan Frett
- Department of Pharmaceutical Science, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Marie Schluterman Burdine
- Division of Surgical Research, Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
- Arkansas Children's Research Institute, Little Rock, AR, 72202, USA.
| |
Collapse
|
24
|
Gao Z, Luan X, Wang X, Han T, Li X, Li Z, Li P, Zhou Z. DNA damage response-related ncRNAs as regulators of therapy resistance in cancer. Front Pharmacol 2024; 15:1390300. [PMID: 39253383 PMCID: PMC11381396 DOI: 10.3389/fphar.2024.1390300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
The DNA damage repair (DDR) pathway is a complex signaling cascade that can sense DNA damage and trigger cellular responses to DNA damage to maintain genome stability and integrity. A typical hallmark of cancer is genomic instability or nonintegrity, which is closely related to the accumulation of DNA damage within cancer cells. The treatment principles of radiotherapy and chemotherapy for cancer are based on their cytotoxic effects on DNA damage, which are accompanied by severe and unnecessary side effects on normal tissues, including dysregulation of the DDR and induced therapeutic tolerance. As a driving factor for oncogenes or tumor suppressor genes, noncoding RNA (ncRNA) have been shown to play an important role in cancer cell resistance to radiotherapy and chemotherapy. Recently, it has been found that ncRNA can regulate tumor treatment tolerance by altering the DDR induced by radiotherapy or chemotherapy in cancer cells, indicating that ncRNA are potential regulatory factors targeting the DDR to reverse tumor treatment tolerance. This review provides an overview of the basic information and functions of the DDR and ncRNAs in the tolerance or sensitivity of tumors to chemotherapy and radiation therapy. We focused on the impact of ncRNA (mainly microRNA [miRNA], long noncoding RNA [lncRNA], and circular RNA [circRNA]) on cancer treatment by regulating the DDR and the underlying molecular mechanisms of their effects. These findings provide a theoretical basis and new insights for tumor-targeted therapy and the development of novel drugs targeting the DDR or ncRNAs.
Collapse
Affiliation(s)
- Ziru Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xinchi Luan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xuezhe Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Tianyue Han
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaoyuan Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zeyang Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhixia Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
25
|
Ye BJ, Li DF, Li XY, Hao JL, Liu DJ, Yu H, Zhang CD. Methylation synthetic lethality: Exploiting selective drug targets for cancer therapy. Cancer Lett 2024; 597:217010. [PMID: 38849016 DOI: 10.1016/j.canlet.2024.217010] [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: 04/28/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
In cancer, synthetic lethality refers to the drug-induced inactivation of one gene and the inhibition of another in cancer cells by a drug, resulting in the death of only cancer cells; however, this effect is not present in normal cells, leading to targeted killing of cancer cells. Recent intensive epigenetic research has revealed that aberrant epigenetic changes are more frequently observed than gene mutations in certain cancers. Recently, numerous studies have reported various methylation synthetic lethal combinations involving DNA damage repair genes, metabolic pathway genes, and paralogs with significant results in cellular models, some of which have already entered clinical trials with promising results. This review systematically introduces the advantages of methylation synthetic lethality and describes the lethal mechanisms of methylation synthetic lethal combinations that have recently demonstrated success in cellular models. Furthermore, we discuss the future opportunities and challenges of methylation synthetic lethality in targeted anticancer therapies.
Collapse
Affiliation(s)
- Bing-Jie Ye
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Di-Fei Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Jia-Lin Hao
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Di-Jie Liu
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Hang Yu
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Chun-Dong Zhang
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
26
|
Uboveja A, Aird KM. Interplay between altered metabolism and DNA damage and repair in ovarian cancer. Bioessays 2024; 46:e2300166. [PMID: 38873912 PMCID: PMC11955923 DOI: 10.1002/bies.202300166] [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: 08/31/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
Ovarian cancer is the most lethal gynecological malignancy and is often associated with both DNA repair deficiency and extensive metabolic reprogramming. While still emerging, the interplay between these pathways can affect ovarian cancer phenotypes, including therapeutic resistance to the DNA damaging agents that are standard-of-care for this tumor type. In this review, we will discuss what is currently known about cellular metabolic rewiring in ovarian cancer that may impact DNA damage and repair in addition to highlighting how specific DNA repair proteins also promote metabolic changes. We will also discuss relevant data from other cancers that could be used to inform ovarian cancer therapeutic strategies. Changes in the choice of DNA repair mechanism adopted by ovarian cancer are a major factor in promoting therapeutic resistance. Therefore, the impact of metabolic reprogramming on DNA repair mechanisms in ovarian cancer has major clinical implications for targeted combination therapies for the treatment of this devastating disease.
Collapse
Affiliation(s)
- Apoorva Uboveja
- Department of Pharmacology & Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Katherine M Aird
- Department of Pharmacology & Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
27
|
Liu Z, Jiang K, Liu Y, Li J, Huang S, Li P, Xu L, Xu X, Hu X, Zeng X, Huang Z, Zhou Y, Li J, Long K, Wang M. Discovery of Preclinical Candidate AD1058 as a Highly Potent, Selective, and Brain-Penetrant ATR Inhibitor for the Treatment of Advanced Malignancies. J Med Chem 2024. [PMID: 39053006 DOI: 10.1021/acs.jmedchem.4c00734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
The ataxia telangiectasia-mutated and Rad3-related protein (ATR) plays a crucial role in regulating the cellular DNA-damage response (DDR), making it a promising target for antitumor drug development through synthetic lethality. In this study, we present the discovery and detailed characterization of AD1058, a highly potent and selective ATR inhibitor, with good preclinical pharmacokinetic profiles. AD1058 exhibits superior efficacy in inhibiting cell proliferation, disrupting the cell cycle, and inducing apoptosis compared to AZD6738. AD1058 displays potent antitumor effects as a single agent or in combination with clinically approved tumor therapies such as PARP inhibitors, ionizing radiotherapy, or chemotherapy in vivo. Considering its enhanced ability to permeate the blood-brain barrier, AD1058 is a promising clinical candidate for the treatment of brain metastases and leptomeningeal metastases in solid tumors. Additionally, among reported ATR inhibitors, AD1058 features the shortest synthesis route and the highest efficiency to date.
Collapse
Affiliation(s)
- Zhi Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kailong Jiang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yan Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junfei Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Siqi Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ping Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Lei Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xiaomin Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xiaobei Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xia Zeng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Zehui Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jia Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kai Long
- Shanghai Annova Biotechnology Co., Ltd., Shanghai 201203, China
| | - Mingliang Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
28
|
Hu YM, Liu XC, Hu L, Dong ZW, Yao HY, Wang YJ, Zhao WJ, Xiang YK, Liu Y, Wang HB, Yin QK. Inhibition of the ATR-DNAPKcs-RB axis drives G1/S-phase transition and sensitizes triple-negative breast cancer (TNBC) to DNA holliday junctions. Biochem Pharmacol 2024; 225:116310. [PMID: 38788960 DOI: 10.1016/j.bcp.2024.116310] [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: 02/05/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Targeting the DNA damage response (DDR) is a promising strategy in oncotherapy, as most tumor cells are sensitive to excess damage due to their repair defects. Ataxia telangiectasia mutated and RAD3-related protein (ATR) is a damage response signal transduction sensor, and its therapeutic potential in tumor cells needs to be precisely investigated. Herein, we identified a new axis that could be targeted by ATR inhibitors to decrease the DNA-dependent protein kinase catalytic subunit (DNAPKcs), downregulate the expression of the retinoblastoma (RB), and drive G1/S-phase transition. Four-way DNA Holliday junctions (FJs) assembled in this process could trigger S-phase arrest and induce lethal chromosome damage in RB-positive triple-negative breast cancer (TNBC) cells. Furthermore, these unrepaired junctions also exerted toxic effects to RB-deficient TNBC cells when the homologous recombination repair (HRR) was inhibited. This study proposes a precise strategy for treating TNBC by targeting the DDR and extends our understanding of ATR and HJ in tumor treatment.
Collapse
Affiliation(s)
- Yue-Miao Hu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Xue-Cun Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Lei Hu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Zhi-Wen Dong
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China; Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| | - Hong-Ying Yao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Ying-Jie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Wen-Jing Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Yu-Ke Xiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yi Liu
- School of Chemistry and Chemical Engineering, Yantai University, Yantai 264005, China
| | - Hong-Bo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China.
| | - Qi-Kun Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China.
| |
Collapse
|
29
|
Gu L, Liu M, Zhang Y, Zhou H, Wang Y, Xu ZX. Telomere-related DNA damage response pathways in cancer therapy: prospective targets. Front Pharmacol 2024; 15:1379166. [PMID: 38910895 PMCID: PMC11190371 DOI: 10.3389/fphar.2024.1379166] [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: 01/30/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Maintaining the structural integrity of genomic chromosomal DNA is an essential role of cellular life and requires two important biological mechanisms: the DNA damage response (DDR) mechanism and telomere protection mechanism at chromosome ends. Because abnormalities in telomeres and cellular DDR regulation are strongly associated with human aging and cancer, there is a reciprocal regulation of telomeres and cellular DDR. Moreover, several drug treatments for DDR are currently available. This paper reviews the progress in research on the interaction between telomeres and cellular DNA damage repair pathways. The research on the crosstalk between telomere damage and DDR is important for improving the efficacy of tumor treatment. However, further studies are required to confirm this hypothesis.
Collapse
Affiliation(s)
- Liting Gu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Yuning Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| |
Collapse
|
30
|
Ng YB, Akincilar SC. Shaping DNA damage responses: Therapeutic potential of targeting telomeric proteins and DNA repair factors in cancer. Curr Opin Pharmacol 2024; 76:102460. [PMID: 38776747 DOI: 10.1016/j.coph.2024.102460] [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: 12/10/2022] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 05/25/2024]
Abstract
Shelterin proteins regulate genomic stability by preventing inappropriate DNA damage responses (DDRs) at telomeres. Unprotected telomeres lead to persistent DDR causing cell cycle inhibition, growth arrest, and apoptosis. Cancer cells rely on DDR to protect themselves from DNA lesions and exogenous DNA-damaging agents such as chemotherapy and radiotherapy. Therefore, targeting DDR machinery is a promising strategy to increase the sensitivity of cancer cells to existing cancer therapies. However, the success of these DDR inhibitors depends on other mutations, and over time, patients develop resistance to these therapies. This suggests the need for alternative approaches. One promising strategy is co-inhibiting shelterin proteins with DDR molecules, which would offset cellular fitness in DNA repair in a mutation-independent manner. This review highlights the associations and dependencies of the shelterin complex with the DDR proteins and discusses potential co-inhibition strategies that might improve the therapeutic potential of current inhibitors.
Collapse
Affiliation(s)
- Yu Bin Ng
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Semih Can Akincilar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore.
| |
Collapse
|
31
|
Mirzayans R. Changing the Landscape of Solid Tumor Therapy from Apoptosis-Promoting to Apoptosis-Inhibiting Strategies. Curr Issues Mol Biol 2024; 46:5379-5396. [PMID: 38920994 PMCID: PMC11202608 DOI: 10.3390/cimb46060322] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
The many limitations of implementing anticancer strategies under the term "precision oncology" have been extensively discussed. While some authors propose promising future directions, others are less optimistic and use phrases such as illusion, hype, and false hypotheses. The reality is revealed by practicing clinicians and cancer patients in various online publications, one of which has stated that "in the quest for the next cancer cure, few researchers bother to look back at the graveyard of failed medicines to figure out what went wrong". The message is clear: Novel therapeutic strategies with catchy names (e.g., synthetic "lethality") have not fulfilled their promises despite decades of extensive research and clinical trials. The main purpose of this review is to discuss key challenges in solid tumor therapy that surprisingly continue to be overlooked by the Nomenclature Committee on Cell Death (NCCD) and numerous other authors. These challenges include: The impact of chemotherapy-induced genome chaos (e.g., multinucleation) on resistance and relapse, oncogenic function of caspase 3, cancer cell anastasis (recovery from late stages of apoptosis), and pitfalls of ubiquitously used preclinical chemosensitivity assays (e.g., cell "viability" and tumor growth delay studies in live animals) that score such pro-survival responses as "lethal" events. The studies outlined herein underscore the need for new directions in the management of solid tumors.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
32
|
Zhou J, Zhang MY, Gao AA, Zhu C, He T, Herman JG, Guo MZ. Epigenetic silencing schlafen-11 sensitizes esophageal cancer to ATM inhibitor. World J Gastrointest Oncol 2024; 16:2060-2073. [PMID: 38764821 PMCID: PMC11099458 DOI: 10.4251/wjgo.v16.i5.2060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/26/2024] [Accepted: 04/01/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Targeting DNA damage response (DDR) pathway is a cutting-edge strategy. It has been reported that Schlafen-11 (SLFN11) contributes to increase chemosensitivity by participating in DDR. However, the detailed mechanism is unclear. AIM To investigate the role of SLFN11 in DDR and the application of synthetic lethal in esophageal cancer with SLFN11 defects. METHODS To reach the purpose, eight esophageal squamous carcinoma cell lines, 142 esophageal dysplasia (ED) and 1007 primary esophageal squamous cell carcinoma (ESCC) samples and various techniques were utilized, including methylation-specific polymerase chain reaction, CRISPR/Cas9 technique, Western blot, colony formation assay, and xenograft mouse model. RESULTS Methylation of SLFN11 was exhibited in 9.15% of (13/142) ED and 25.62% of primary (258/1007) ESCC cases, and its expression was regulated by promoter region methylation. SLFN11 methylation was significantly associated with tumor differentiation and tumor size (both P < 0.05). However, no significant associations were observed between promoter region methylation and age, gender, smoking, alcohol consumption, TNM stage, or lymph node metastasis. Utilizing DNA damaged model induced by low dose cisplatin, SLFN11 was found to activate non-homologous end-joining and ATR/CHK1 signaling pathways, while inhibiting the ATM/CHK2 signaling pathway. Epigenetic silencing of SLFN11 was found to sensitize the ESCC cells to ATM inhibitor (AZD0156), both in vitro and in vivo. CONCLUSION SLFN11 is frequently methylated in human ESCC. Methylation of SLFN11 is sensitive marker of ATM inhibitor in ESCC.
Collapse
Affiliation(s)
- Jing Zhou
- School of Medicine, NanKai University, Tianjin 300071, China
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Mei-Ying Zhang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ai-Ai Gao
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Cheng Zhu
- School of Medicine, NanKai University, Tianjin 300071, China
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Tao He
- Departments of Pathology, Characteristic Medical Center of The Chinese People’s Armed Police Force, Tianjin 300162, China
| | - James G Herman
- The Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, United States
| | - Ming-Zhou Guo
- School of Medicine, NanKai University, Tianjin 300071, China
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
33
|
Chen XY, Wu ZX, Wang JQ, Teng QX, Tang H, Liu Q, Chen ZS, Chen W. Multidrug resistance transporters P-gp and BCRP limit the efficacy of ATR inhibitor ceralasertib in cancer cells. Front Pharmacol 2024; 15:1400699. [PMID: 38756373 PMCID: PMC11096521 DOI: 10.3389/fphar.2024.1400699] [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: 03/14/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
The therapeutic effect of chemotherapy and targeted therapy are known to be limited by drug resistance. Substantial evidence has shown that ATP-binding cassette (ABC) transporters P-gp and BCRP are significant contributors to multidrug resistance (MDR) in cancer cells. In this study, we demonstrated that a clinical-staged ATR inhibitor ceralasertib is susceptible to P-gp and BCRP-mediated MDR. The drug resistant cancer cells were less sensitive to ceralasertib compared to the parental cells. Moreover, ceralasertib resistance can be reversed by inhibiting the drug efflux activity of P-gp and BCRP. Interestingly, ceralasertib was able to downregulate the level of P-gp but not BCRP, suggesting a potential regulation between ATR signaling and P-gp expression. Furthermore, computational docking analysis predicted high affinities between ceralasertib and the drug-binding sites of P-gp and BCRP. In summary, overexpression of P-gp and BCRP are sufficient to confer cancer cells resistance to ceralasertib, underscoring their role as biomarkers for therapeutic efficacy.
Collapse
Affiliation(s)
- Xuan-Yu Chen
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Zhuo-Xun Wu
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
| | - Jing-Quan Wang
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
| | - Qiu-Xu Teng
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qianwen Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Wenkuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
34
|
Zhanataev AK, Kulakova AV, Luzina OA, Khomenko TM, Volcho KP, Salakhutdinov NF, Durnev AD. Enhancement of the Clastogenic Effects of Topotecan In Vivo by Tyrosyl-DNA Phosphodiesterase 1 Inhibitors. Bull Exp Biol Med 2024; 177:30-34. [PMID: 38954304 DOI: 10.1007/s10517-024-06125-9] [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: 09/07/2023] [Indexed: 07/04/2024]
Abstract
Topotecan administered intraperitoneally at single doses of 0.25, 0.5, and 1 mg/kg induced chromosomal aberrations in bone marrow cells of F1(CBA×C57BL/6) hybrid mice in a dose-dependent manner. A tyrosyl-DNA phosphodiesterase 1 (TDP1) inhibitor, an usnic acid derivative OL9-116 was inactive in a dose range of 20-240 mg/kg, but enhanced the cytogenetic effect of topotecan (0.25 mg/kg) at a dose of 40 mg/kg (per os). The TDP1 inhibitor, a coumarin derivative TX-2552 (at doses of 20, 40, 80, and 160 mg/kg per os), increased the level of aberrant metaphases induced by topotecan (0.25 mg/kg) by 2.1-2.6 times, but was inactive at a dose of 10 mg/kg. The results indicate that TDP1 inhibitors enhance the clastogenic activity of topotecan in mouse bone marrow cells in vivo and are characterized by different dose profiles of the co-mutagenic effects.
Collapse
Affiliation(s)
- A K Zhanataev
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia.
| | - A V Kulakova
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - O A Luzina
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - T M Khomenko
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - K P Volcho
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N F Salakhutdinov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A D Durnev
- V. V. Zakusov Research Institute of Pharmacology, Moscow, Russia
| |
Collapse
|
35
|
Pusch FF, Dorado García H, Xu R, Gürgen D, Bei Y, Brückner L, Röefzaad C, von Stebut J, Bardinet V, Chamorro Gonzalez R, Eggert A, Schulte JH, Hundsdörfer P, Seifert G, Haase K, Schäfer BW, Wachtel M, Kühl AA, Ortiz MV, Wengner AM, Scheer M, Henssen AG. Elimusertib has Antitumor Activity in Preclinical Patient-Derived Pediatric Solid Tumor Models. Mol Cancer Ther 2024; 23:507-519. [PMID: 38159110 PMCID: PMC10985474 DOI: 10.1158/1535-7163.mct-23-0094] [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: 02/13/2023] [Revised: 09/12/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
The small-molecule inhibitor of ataxia telangiectasia and Rad3-related protein (ATR), elimusertib, is currently being tested clinically in various cancer entities in adults and children. Its preclinical antitumor activity in pediatric malignancies, however, is largely unknown. We here assessed the preclinical activity of elimusertib in 38 cell lines and 32 patient-derived xenograft (PDX) models derived from common pediatric solid tumor entities. Detailed in vitro and in vivo molecular characterization of the treated models enabled the evaluation of response biomarkers. Pronounced objective response rates were observed for elimusertib monotherapy in PDX, when treated with a regimen currently used in clinical trials. Strikingly, elimusertib showed stronger antitumor effects than some standard-of-care chemotherapies, particularly in alveolar rhabdomysarcoma PDX. Thus, elimusertib has strong preclinical antitumor activity in pediatric solid tumor models, which may translate to clinically meaningful responses in patients.
Collapse
Affiliation(s)
- Fabian F. Pusch
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Heathcliff Dorado García
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robin Xu
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dennis Gürgen
- Experimental Pharmacology and Oncology (EPO), Berlin, Germany
| | - Yi Bei
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lotte Brückner
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (BIMSB/BIH), Berlin, Germany
| | - Claudia Röefzaad
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jennifer von Stebut
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor Bardinet
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| | - Rocío Chamorro Gonzalez
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes H. Schulte
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, University Hospital Tübingen, Tübingen, Germany
| | - Patrick Hundsdörfer
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Georg Seifert
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kerstin Haase
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| | | | | | - Anja A. Kühl
- iPATH.Berlin—Core Unit Immunopathology for Experimental Models, Charité Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York City, New York
| | | | - Monika Scheer
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anton G. Henssen
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (BIMSB/BIH), Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Odhiambo DA, Pittman AN, Rickard AG, Castillo RJ, Bassil AM, Chen J, Ravotti ML, Xu ES, Himes JE, Daniel AR, Watts TL, Williams NT, Luo L, Kirsch DG, Mowery YM. Preclinical Evaluation of the ATR Inhibitor BAY 1895344 as a Radiosensitizer for Head and Neck Squamous Cell Carcinoma. Int J Radiat Oncol Biol Phys 2024; 118:1315-1327. [PMID: 38104870 PMCID: PMC11294978 DOI: 10.1016/j.ijrobp.2023.12.012] [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: 08/16/2023] [Revised: 11/17/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Despite aggressive multimodal treatment that typically includes definitive or adjuvant radiation therapy (RT), locoregional recurrence rates approach 50% for patients with locally advanced human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC). Thus, more effective therapeutics are needed to improve patient outcomes. We evaluated the radiosensitizing effects of ataxia telangiectasia and RAD3-related (ATR) inhibitor (ATRi) BAY 1895344 in preclinical models of HNSCC. METHODS AND MATERIALS Murine and human HPV-negative HNSCC cells (MOC2, MOC1, JHU-012) were treated with vehicle or ATRi with or without 4 Gy. Checkpoint kinase 1 phosphorylation and DNA damage (γH2AX) were evaluated by Western blot, and ATRi half-maximal inhibitory concentration was determined by MTT assay for HNSCC cells and immortalized murine oral keratinocytes. In vitro radiosensitization was tested by clonogenic assay. Cell cycle distribution and mitotic catastrophe were evaluated by flow cytometry. Mitotic aberrations were quantified by fluorescent microscopy. Tumor growth delay and survival were assessed in mice bearing MOC2 or JHU-012 transplant tumors treated with vehicle, ATRi, RT (10 Gy × 1 or 8 Gy × 3), or combined ATRi + RT. RESULTS ATRi caused dose-dependent reduction in checkpoint kinase 1 phosphorylation at 1 hour post-RT (4 Gy) and dose-dependent increase in γH2AX at 18 hours post-RT. Addition of RT to ATRi led to decreased BAY 1895344 half-maximal inhibitory concentration in HNSCC cell lines but not in normal tissue surrogate immortalized murine oral keratinocytes. Clonogenic assays demonstrated radiosensitization in the HNSCC cell lines. ATRi abrogated the RT-induced G2/M checkpoint, leading to mitosis with unrepaired DNA damage and increased mitotic aberrations (multinucleated cells, micronuclei, nuclear buds, nucleoplasmic bridges). ATRi and RT significantly delayed tumor growth in MOC2 and JHU-012 in vivo models, with improved overall survival in the MOC2 model. CONCLUSIONS These findings demonstrated that BAY 1895344 increased in vitro and in vivo radiosensitivity in HPV-negative HNSCC preclinical models, suggesting therapeutic potential warranting evaluation in clinical trials for patients with locally advanced or recurrent HPV-negative HNSCC.
Collapse
Affiliation(s)
| | | | - Ashlyn G Rickard
- Dept. of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh
| | | | | | - Joshua Chen
- College of Arts and Sciences, Duke University
| | - Madison L Ravotti
- Dept. of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh
| | - Eric S Xu
- Dept. of Radiation Oncology, Duke University
| | | | | | - Tammara L Watts
- Dept. of Head and Neck Surgery & Communication Sciences, Duke University
| | | | - Lixia Luo
- Dept. of Radiation Oncology, Duke University
| | - David G Kirsch
- Dept. of Radiation Oncology, Duke University
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network
- Dept. of Radiation Oncology and Dept. of Medical Biophysics, University of Toronto
| | - Yvonne M Mowery
- Dept. of Radiation Oncology, Duke University
- Dept. of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh
- Dept. of Head and Neck Surgery & Communication Sciences, Duke University
| |
Collapse
|
37
|
Chiappa M, Guffanti F, Grasselli C, Panini N, Corbelli A, Fiordaliso F, Damia G. Different Patterns of Platinum Resistance in Ovarian Cancer Cells with Homologous Recombination Proficient and Deficient Background. Int J Mol Sci 2024; 25:3049. [PMID: 38474294 DOI: 10.3390/ijms25053049] [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: 01/19/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Platinum compounds are very active in first-line treatments of ovarian carcinoma. In fact, high rates of complete remission are achieved, but most patients eventually relapse with resistant disease. Many mechanisms underlying the platinum-resistant phenotype have been reported. However, there are no data in the same isogenic cell system proficient and deficient in homologous recombination (HR) on platinum-acquired resistance that might unequivocally clarify the most important mechanism associated with resistance. We generated and characterized cisplatin (DDP)-resistant murine ovarian ID8 cell lines in a HR-deficient and -proficient background. Specific upregulation of the NER pathway in the HR-proficient and -resistant cells and partial restoration of HR in Brca1-/--resistant cells were found. Combinations of different inhibitors of the DNA damage response pathways with cisplatin were strongly active in both resistant and parental cells. The data from the ID8 isogenic system are in line with current experimental and clinical evidence and strongly suggest that platinum resistance develops in different ways depending on the cell DNA repair status (i.e., HR-proficient or HR-deficient), and the upregulation and/or restoration of repair pathways are major determinants of DDP resistance.
Collapse
Affiliation(s)
- Michela Chiappa
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Federica Guffanti
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Chiara Grasselli
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Nicolò Panini
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Alessandro Corbelli
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Fabio Fiordaliso
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Giovanna Damia
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| |
Collapse
|
38
|
Mavroeidi D, Georganta A, Panagiotou E, Syrigos K, Souliotis VL. Targeting ATR Pathway in Solid Tumors: Evidence of Improving Therapeutic Outcomes. Int J Mol Sci 2024; 25:2767. [PMID: 38474014 DOI: 10.3390/ijms25052767] [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: 01/23/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The DNA damage response (DDR) system is a complicated network of signaling pathways that detects and repairs DNA damage or induces apoptosis. Critical regulators of the DDR network include the DNA damage kinases ataxia telangiectasia mutated Rad3-related kinase (ATR) and ataxia-telangiectasia mutated (ATM). The ATR pathway coordinates processes such as replication stress response, stabilization of replication forks, cell cycle arrest, and DNA repair. ATR inhibition disrupts these functions, causing a reduction of DNA repair, accumulation of DNA damage, replication fork collapse, inappropriate mitotic entry, and mitotic catastrophe. Recent data have shown that the inhibition of ATR can lead to synthetic lethality in ATM-deficient malignancies. In addition, ATR inhibition plays a significant role in the activation of the immune system by increasing the tumor mutational burden and neoantigen load as well as by triggering the accumulation of cytosolic DNA and subsequently inducing the cGAS-STING pathway and the type I IFN response. Taken together, we review stimulating data showing that ATR kinase inhibition can alter the DDR network, the immune system, and their interplay and, therefore, potentially provide a novel strategy to improve the efficacy of antitumor therapy, using ATR inhibitors as monotherapy or in combination with genotoxic drugs and/or immunomodulators.
Collapse
Affiliation(s)
- Dimitra Mavroeidi
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Anastasia Georganta
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Emmanouil Panagiotou
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Konstantinos Syrigos
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Vassilis L Souliotis
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece
| |
Collapse
|
39
|
Black WC, Abdoli A, An X, Auger A, Beaulieu P, Bernatchez M, Caron C, Chefson A, Crane S, Diallo M, Dorich S, Fader LD, Ferraro GB, Fournier S, Gao Q, Ginzburg Y, Hamel M, Han Y, Jones P, Lanoix S, Lacbay CM, Leclaire ME, Levy M, Mamane Y, Mulani A, Papp R, Pellerin C, Picard A, Skeldon A, Skorey K, Stocco R, St-Onge M, Truchon JF, Truong VL, Zimmermann M, Zinda M, Roulston A. Discovery of the Potent and Selective ATR Inhibitor Camonsertib (RP-3500). J Med Chem 2024; 67:2349-2368. [PMID: 38299539 DOI: 10.1021/acs.jmedchem.3c01917] [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: 02/02/2024]
Abstract
ATR is a key kinase in the DNA-damage response (DDR) that is synthetic lethal with several other DDR proteins, making it an attractive target for the treatment of genetically selected solid tumors. Herein we describe the discovery of a novel ATR inhibitor guided by a pharmacophore model to position a key hydrogen bond. Optimization was driven by potency and selectivity over the related kinase mTOR, resulting in the identification of camonsertib (RP-3500) with high potency and excellent ADME properties. Preclinical evaluation focused on the impact of camonsertib on myelosuppression, and an exploration of intermittent dosing schedules to allow recovery of the erythroid compartment and mitigate anemia. Camonsertib is currently undergoing clinical evaluation both as a single agent and in combination with talazoparib, olaparib, niraparib, lunresertib, or gemcitabine (NCT04497116, NCT04972110, NCT04855656). A preliminary recommended phase 2 dose for monotherapy was identified as 160 mg QD given 3 days/week.
Collapse
Affiliation(s)
- W Cameron Black
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Abbas Abdoli
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Xiuli An
- New York Blood Center Enterprises, New York, New York 10065, United States
| | - Anick Auger
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | | | | | - Cathy Caron
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Amandine Chefson
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Sheldon Crane
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Mohamed Diallo
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Stéphane Dorich
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Lee D Fader
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Gino B Ferraro
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Sara Fournier
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Qi Gao
- J-Star Research, Inc., 3001 Hadley Road, Suites 1-5A, South Plainfield, New Jersey 07080, United States
| | - Yelena Ginzburg
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Martine Hamel
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Yongshuai Han
- New York Blood Center Enterprises, New York, New York 10065, United States
| | - Paul Jones
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Stéphanie Lanoix
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Cyrus M Lacbay
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Marie-Eve Leclaire
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Maayan Levy
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yael Mamane
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Amina Mulani
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Robert Papp
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Charles Pellerin
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Audrey Picard
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Alexander Skeldon
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Kathryn Skorey
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Rino Stocco
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Miguel St-Onge
- Ventus Therapeutics, Inc., 7150 Frederick-Banting, Saint-Laurent, Quebec H4S 2A1, Canada
| | - Jean-François Truchon
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Vouy Linh Truong
- Nuchem Sciences, Inc., 2350 Rue Cohen, Suite 201, Saint-Laurent, Quebec H4R 2N6, Canada
| | - Michal Zimmermann
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Michael Zinda
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| | - Anne Roulston
- Repare Therapeutics, Inc., 7171 Frederick-Banting, Building 2, Saint-Laurent, Quebec H4S 1Z9, Canada
| |
Collapse
|
40
|
Huang F, Zhang C, Yang W, Zhou Y, Yang Y, Yang X, Guo W, Wang B. Identification of a DNA damage repair-related LncRNA signature for predicting the prognosis and immunotherapy response of hepatocellular carcinoma. BMC Genomics 2024; 25:155. [PMID: 38326754 PMCID: PMC10851502 DOI: 10.1186/s12864-024-10055-1] [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: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND DNA damage repair (DDR) may affect tumorigenesis and therapeutic response in hepatocellular carcinoma (HCC). Long noncoding RNAs (LncRNAs) can regulate DDR and play a vital role in maintaining genomic stability in cancers. Here, we identified a DDR-related prognostic signature in HCC and explored its potential clinical value. METHODS Data of HCC samples were obtained from the Cancer Genome Atlas (TCGA), and a list of DDR-related genes was extracted from the Molecular Signatures database (MSigDB). A DDR-related lncRNAs signature associated to overall survival (OS) was constructed using the least absolute shrinkage and selection operator-cox regression, and was further validated by the Kaplan-Meier curve and receiver operating characteristic curve. A nomogram integrating other clinical risk factors was established. Moreover, the relationships between the signature with somatic mutation, immune landscape and drug sensitivity were explored. RESULTS The prognostic model of 5 DDR-related lncRNAs was constructed and classified patients into two risk groups at median cut-off. The low-risk group had a better OS, and the signature was an independent prognostic indicator in HCC. A nomogram of the signature combined with TNM stage was constructed. TP53 gene was more frequently mutated in the high-risk group. Marked differences in immune cells were observed, such as CD4 + T cells, NK cells and macrophages, between the two groups. Moreover, an increase in the expression of immune checkpoint molecules was found in the high-risk group. The low-risk group presented with a significantly higher response to sorafenib or cisplatin. Finally, potential value of this signature was validated in real-world HCC patients. CONCLUSION Our findings provided a promising insight into DDR-related lncRNAs in HCC and a personalized prediction tool for prognosis and therapeutic response.
Collapse
Affiliation(s)
- Fei Huang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Centre, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Zhou
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yihui Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinrong Yang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Laboratory Medicine, Shanghai Geriatric Medical Centre, Shanghai, China.
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Di Bona M, Bakhoum SF. Micronuclei and Cancer. Cancer Discov 2024; 14:214-226. [PMID: 38197599 PMCID: PMC11265298 DOI: 10.1158/2159-8290.cd-23-1073] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 01/11/2024]
Abstract
Chromosome-containing micronuclei are a feature of human cancer. Micronuclei arise from chromosome mis-segregation and characterize tumors with elevated rates of chromosomal instability. Although their association with cancer has been long recognized, only recently have we broadened our understanding of the mechanisms that govern micronuclei formation and their role in tumor progression. In this review, we provide a brief historical account of micronuclei, depict the mechanisms underpinning their creation, and illuminate their capacity to propel tumor evolution through genetic, epigenetic, and transcriptional transformations. We also posit the prospect of leveraging micronuclei as biomarkers and therapeutic targets in chromosomally unstable cancers. SIGNIFICANCE Micronuclei in chromosomally unstable cancer cells serve as pivotal catalysts for cancer progression, instigating transformative genomic, epigenetic, and transcriptional alterations. This comprehensive review not only synthesizes our present comprehension but also outlines a framework for translating this knowledge into pioneering biomarkers and therapeutics, thereby illuminating novel paths for personalized cancer management.
Collapse
Affiliation(s)
- Melody Di Bona
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samuel F. Bakhoum
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
42
|
Chen W, Kang Y, Sheng W, Huang Q, Cheng J, Pei S, Meng Y. A new 4-gene-based prognostic model accurately predicts breast cancer prognosis and immunotherapy response by integrating WGCNA and bioinformatics analysis. Front Immunol 2024; 15:1331841. [PMID: 38370403 PMCID: PMC10869553 DOI: 10.3389/fimmu.2024.1331841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Background Breast cancer (BRCA) is a common malignancy in women, and its resistance to immunotherapy is a major challenge. Abnormal expression of genes is important in the occurrence and development of BRCA and may also affect the prognosis of patients. Although many BRCA prognosis model scores have been developed, they are only applicable to a limited number of disease subtypes. Our goal is to develop a new prognostic score that is more accurate and applicable to a wider range of BRCA patients. Methods BRCA patient data from The Cancer Genome Atlas database was used to identify breast cancer-related genes (BRGs). Differential expression analysis of BRGs was performed using the 'limma' package in R. Prognostic BRGs were identified using co-expression and univariate Cox analysis. A predictive model of four BRGs was established using Cox regression and the LASSO algorithm. Model performance was evaluated using K-M survival and receiver operating characteristic curve analysis. The predictive ability of the signature in immune microenvironment and immunotherapy was investigated. In vitro experiments validated POLQ function. Results Our study identified a four-BRG prognostic signature that outperformed conventional clinicopathological characteristics in predicting survival outcomes in BRCA patients. The signature effectively stratified BRCA patients into high- and low-risk groups and showed potential in predicting the response to immunotherapy. Notably, significant differences were observed in immune cell abundance between the two groups. In vitro experiments demonstrated that POLQ knockdown significantly reduced the viability, proliferation, and invasion capacity of MDA-MB-231 or HCC1806 cells. Conclusion Our 4-BRG signature has the potential as an independent biomarker for predicting prognosis and treatment response in BRCA patients, complementing existing clinicopathological characteristics.
Collapse
Affiliation(s)
- Wenlong Chen
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yakun Kang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenyi Sheng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Qiyan Huang
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jiale Cheng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - You Meng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
43
|
Chotiprasidhi P, Sato-Espinoza AK, Wangensteen KJ. Germline Genetic Associations for Hepatobiliary Cancers. Cell Mol Gastroenterol Hepatol 2023; 17:623-638. [PMID: 38163482 PMCID: PMC10899027 DOI: 10.1016/j.jcmgh.2023.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Hepatobiliary cancers (HBCs) include hepatocellular carcinoma, cholangiocarcinoma, and gallbladder carcinoma, which originate from the liver, bile ducts, and gallbladder, respectively. They are responsible for a substantial burden of cancer-related deaths worldwide. Despite knowledge of risk factors and advancements in therapeutics and surgical interventions, the prognosis for most patients with HBC remains bleak. There is evidence from familial aggregation and case-control studies to suggest a familial risk component in HBC susceptibility. Recent progress in genomics research has led to the identification of germline variants including single nucleotide polymorphisms (SNPs) and pathogenic or likely pathogenic (P/LP) variants in cancer-associated genes associated with HBC risk. These findings emerged from genome-wide association studies and next-generation sequencing techniques such as whole-exome sequencing. Patients with other cancer types, including breast, colon, ovarian, prostate, and pancreatic cancer, are recommended by guidelines to undergo germline genetic testing, but similar recommendations are lagging in HBC. This prompts the question of whether multi-gene panel testing should be integrated into clinical guidelines for HBC management. Here, we review the hereditary genetics of HBC, explore studies investigating SNPs and P/LP variants in HBC patients, discuss the clinical implications and potential for personalized treatments and impact on patient's family members, and conclude that additional studies are needed to examine how genetic testing can be applied clinically.
Collapse
Affiliation(s)
- Perapa Chotiprasidhi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Kirk J Wangensteen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
44
|
Du J, Dong Y, Zuo W, Deng Y, Zhu H, Yu Q, Li M. Mec1-Rad53 Signaling Regulates DNA Damage-Induced Autophagy and Pathogenicity in Candida albicans. J Fungi (Basel) 2023; 9:1181. [PMID: 38132782 PMCID: PMC10744610 DOI: 10.3390/jof9121181] [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: 11/09/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
DNA damage activates the DNA damage response and autophagy in C. albicans; however, the relationship between the DNA damage response and DNA damage-induced autophagy in C. albicans remains unclear. Mec1-Rad53 signaling is a critical pathway in the DNA damage response, but its role in DNA damage-induced autophagy and pathogenicity in C. albicans remains to be further explored. In this study, we compared the function of autophagy-related (Atg) proteins in DNA damage-induced autophagy and traditional macroautophagy and explored the role of Mec1-Rad53 signaling in regulating DNA damage-induced autophagy and pathogenicity. We found that core Atg proteins are required for these two types of autophagy, while the function of Atg17 is slightly different. Our results showed that Mec1-Rad53 signaling specifically regulates DNA damage-induced autophagy but has no effect on macroautophagy. The recruitment of Atg1 and Atg13 to phagophore assembly sites (PAS) was significantly inhibited in the mec1Δ/Δ and rad53Δ/Δ strains. The formation of autophagic bodies was obviously affected in the mec1Δ/Δ and rad53Δ/Δ strains. We found that DNA damage does not induce mitophagy and ER autophagy. We also identified two regulators of DNA damage-induced autophagy, Psp2 and Dcp2, which regulate DNA damage-induced autophagy by affecting the protein levels of Atg1, Atg13, Mec1, and Rad53. The deletion of Mec1 or Rad53 significantly reduces the ability of C. albicans to systematically infect mice and colonize the kidneys, and it makes C. albicans more susceptible to being killed by macrophages.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China; (J.D.); (Y.D.); (W.Z.); (Y.D.); (H.Z.); (Q.Y.)
| |
Collapse
|
45
|
Vokes NI, Galan Cobo A, Fernandez-Chas M, Molkentine D, Treviño S, Druker V, Qian Y, Patel S, Schmidt S, Hong L, Lewis J, Rinsurongkawong W, Rinsurongkawong V, Lee JJ, Negrao MV, Gibbons DL, Vaporciyan A, Le X, Wu J, Zhang J, Rigney U, Iyer S, Dean E, Heymach JV. ATM Mutations Associate with Distinct Co-Mutational Patterns and Therapeutic Vulnerabilities in NSCLC. Clin Cancer Res 2023; 29:4958-4972. [PMID: 37733794 PMCID: PMC10690143 DOI: 10.1158/1078-0432.ccr-23-1122] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/16/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Ataxia-telangiectasia mutated (ATM) is the most frequently mutated DNA damage repair gene in non-small cell lung cancer (NSCLC). However, the molecular correlates of ATM mutations and their clinical implications have not been fully elucidated. EXPERIMENTAL DESIGN Clinicopathologic and genomic data from 26,587 patients with NSCLC from MD Anderson, public databases, and a de-identified nationwide (US-based) NSCLC clinicogenomic database (CGDB) were used to assess the co-mutation landscape, protein expression, and mutational processes in ATM-mutant tumors. We used the CGDB to evaluate ATM-associated outcomes in patients treated with immune checkpoint inhibitors (ICI) with or without chemotherapy, and assessed the effect of ATM loss on STING signaling and chemotherapy sensitivity in preclinical models. RESULTS Nonsynonymous mutations in ATM were observed in 11.2% of samples (2,980/26,587) and were significantly associated with mutations in KRAS, but mutually exclusive with EGFR (q < 0.1). KRAS mutational status constrained the ATM co-mutation landscape, with strong mutual exclusivity with TP53 and KEAP1 within KRAS-mutated samples. Those ATM mutations that co-occurred with TP53 were more likely to be missense mutations and associate with high mutational burden, suggestive of non-functional passenger mutations. In the CGDB cohort, dysfunctional ATM mutations associated with improved OS only in patients treated with ICI-chemotherapy, and not ICI alone. In vitro analyses demonstrated enhanced upregulation of STING signaling in ATM knockout cells with the addition of chemotherapy. CONCLUSIONS ATM mutations define a distinct subset of NSCLC associated with KRAS mutations, increased TMB, decreased TP53 and EGFR co-occurrence, and potential increased sensitivity to ICIs in the context of DNA-damaging chemotherapy.
Collapse
Affiliation(s)
- Natalie I. Vokes
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana Galan Cobo
- Department of Molecular Diagnostics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - David Molkentine
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Santiago Treviño
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vitaly Druker
- Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Yu Qian
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sonia Patel
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie Schmidt
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lingzhi Hong
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeff Lewis
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Waree Rinsurongkawong
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - J. Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marcelo V. Negrao
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Don L. Gibbons
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ara Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiuning Le
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Una Rigney
- Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sonia Iyer
- Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emma Dean
- Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - John V. Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
46
|
Wei W, Shi F, Xu Y, Jiao Y, Zhang Y, Ou Q, Wu X, Yang L, Lai J. The enrichment of Fanconi anemia/homologous recombination pathway aberrations in ATM/ATR-mutated NSCLC was accompanied by unique molecular features and poor prognosis. J Transl Med 2023; 21:874. [PMID: 38041093 PMCID: PMC10690992 DOI: 10.1186/s12967-023-04634-1] [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: 06/19/2023] [Accepted: 10/14/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND ATM and ATR are two critical factors to regulate DNA damage response (DDR), and their mutations were frequently observed in different types of cancer, including non-small cell lung cancer (NSCLC). Given that the majority of identified ATM/ATR mutations were variants of uncertain significance, the clinical/molecular features of pathogenic ATM/ATR aberrations have not been comprehensively investigated in NSCLC. METHODS Next-generation sequencing (NGS) analyses were conducted to investigate the molecular features in 191 NSCLC patients who harbored pathogenic/likely pathogenic ATM/ATR mutations and 308 NSCLC patients who did not have any types of ATM/ATR variants. The results were validated using an external cohort of 2727 NSCLC patients (including 48 with ATM/ATR pathogenic mutations). RESULTS Most pathogenic ATM/ATR genetic alterations were frameshift and nonsense mutations that disrupt critical domains of the two proteins. ATM/ATR-mutated patients had significantly higher tumor mutational burdens (TMB; P < 0.001) and microsatellite instabilities (MSI; P = 0.023), but not chromosomal instabilities, than those without any ATM/ATR variations. In particular, KRAS mutations were significantly enriched in ATM-mutated patients (P = 0.014), whereas BRCA2 mutations (P = 0.014), TP53 mutations (P = 0.014), and ZNF703 amplification (P = 0.008) were enriched in ATR-mutated patients. Notably, patients with ATM/ATR pathogenic genetic alterations were likely to be accompanied by mutations in Fanconi anemia (FA) and homologous recombination (HR) pathways, which were confirmed using both the study (P < 0.001) and validation (P < 0.001) cohorts. Furthermore, the co-occurrence of FA/HR aberrations could contribute to increased TMB and MSI, and patients with both ATM/ATR and FA/HR mutations tended to have worse overall survival. CONCLUSIONS Our results demonstrated the unique clinical and molecular features of pathogenic ATM/ATR mutations in NSCLC, which helps better understand the cancerous involvement of these DDR regulators, as well as directing targeted therapies and/or immunotherapies to treat ATM/ATR-mutated NSCLC, especially those with co-existing FA/HR aberrations.
Collapse
Affiliation(s)
- Wei Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yang Xu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, 210032, Jiangsu, China
| | - Yang Jiao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Ying Zhang
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, Jiangsu, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, 210032, Jiangsu, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, 210032, Jiangsu, China
| | - Lingyi Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
| | - Jinhuo Lai
- Department of Medical Oncology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350025, Fujian, China.
| |
Collapse
|
47
|
Huang X, You L, Nepovimova E, Psotka M, Malinak D, Valko M, Sivak L, Korabecny J, Heger Z, Adam V, Wu Q, Kuca K. Inhibitors of phosphoinositide 3-kinase (PI3K) and phosphoinositide 3-kinase-related protein kinase family (PIKK). J Enzyme Inhib Med Chem 2023; 38:2237209. [PMID: 37489050 PMCID: PMC10392309 DOI: 10.1080/14756366.2023.2237209] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/11/2023] [Indexed: 02/02/2024] Open
Abstract
Phosphoinositide 3-kinases (PI3K) and phosphoinositide 3-kinase-related protein kinases (PIKK) are two structurally related families of kinases that play vital roles in cell growth and DNA damage repair. Dysfunction of PIKK members and aberrant stimulation of the PI3K/AKT/mTOR signalling pathway are linked to a plethora of diseases including cancer. In recent decades, numerous inhibitors related to the PI3K/AKT/mTOR signalling have made great strides in cancer treatment, like copanlisib and sirolimus. Notably, most of the PIKK inhibitors (such as VX-970 and M3814) related to DNA damage response have also shown good efficacy in clinical trials. However, these drugs still require a suitable combination therapy to overcome drug resistance or improve antitumor activity. Based on the aforementioned facts, we summarised the efficacy of PIKK, PI3K, and AKT inhibitors in the therapy of human malignancies and the resistance mechanisms of targeted therapy, in order to provide deeper insights into cancer treatment.
Collapse
Affiliation(s)
- Xueqin Huang
- College of Life Science, Yangtze University, Jingzhou, China
| | - Li You
- College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
| | - Miroslav Psotka
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - David Malinak
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Bratislava, Slovakia
| | - Ladislav Sivak
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
48
|
Zhang X, Zhao Q, Wang T, Long Q, Sun Y, Jiao L, Gullerova M. DNA damage response, a double-edged sword for vascular aging. Ageing Res Rev 2023; 92:102137. [PMID: 38007046 DOI: 10.1016/j.arr.2023.102137] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/03/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Vascular aging is a major risk factor for age-related cardiovascular diseases, which have high rates of morbidity and mortality. It is characterized by changes in the blood vessels, such as macroscopically increased vascular diameter and intima-medial thickness, chronic inflammation, vascular calcification, arterial stiffening, and atherosclerosis. DNA damage and the subsequent various DNA damage response (DDR) pathways are important causative factors of vascular aging. Deficient DDR, which may result in the accumulation of unrepaired damaged DNA or mutations, can lead to vascular aging. On the other hand, over-activation of some DDR proteins, such as poly (ADP ribose) polymerase (PARP) and ataxia telangiectasia mutated (ATM), also can enhance the process of vascular aging, suggesting that DDR can have both positive and negative effects on vascular aging. Despite the evidence reviewed in this paper, the role of DDR in vascular aging and potential therapeutic targets remain poorly understood and require further investigation.
Collapse
Affiliation(s)
- Xiao Zhang
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; China International Neuroscience Institute (China-INI), Beijing 100053, China
| | - Qing Zhao
- M.D. Program, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; China International Neuroscience Institute (China-INI), Beijing 100053, China
| | - Qilin Long
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Yixin Sun
- First Hospital, Peking University, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; China International Neuroscience Institute (China-INI), Beijing 100053, China; Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Monika Gullerova
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom.
| |
Collapse
|
49
|
Chen JW, Shan TK, Wei TW, Jiang QQ, Du C, Gu LF, Yang TT, Zhou LH, Wang SB, Bao YL, Wang H, Ji Y, Xie LP, Gu AH, Sun CQ, Wang QM, Wang LS. SIRT3-dependent mitochondrial redox homeostasis mitigates CHK1 inhibition combined with gemcitabine treatment induced cardiotoxicity in hiPSC-CMs and mice. Arch Toxicol 2023; 97:3209-3226. [PMID: 37798514 DOI: 10.1007/s00204-023-03611-3] [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: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 10/07/2023]
Abstract
Administration of CHK1-targeted anticancer therapies is associated with an increased cumulative risk of cardiac complications, which is further amplified when combined with gemcitabine. However, the underlying mechanisms remain elusive. In this study, we generated hiPSC-CMs and murine models to elucidate the mechanisms underlying CHK1 inhibition combined with gemcitabine-induced cardiotoxicity and identify potential targets for cardioprotection. Mice were intraperitoneally injected with 25 mg/kg CHK1 inhibitor AZD7762 and 20 mg/kg gemcitabine for 3 weeks. hiPSC-CMs and NMCMs were incubated with 0.5 uM AZD7762 and 0.1 uM gemcitabine for 24 h. Both pharmacological inhibition or genetic deletion of CHK1 and administration of gemcitabine induced mtROS overproduction and pyroptosis in cardiomyocytes by disrupting mitochondrial respiration, ultimately causing heart atrophy and cardiac dysfunction in mice. These toxic effects were further exacerbated with combination administration. Using mitochondria-targeting sequence-directed vectors to overexpress CHK1 in cardiomyocyte (CM) mitochondria, we identified the localization of CHK1 in CM mitochondria and its crucial role in maintaining mitochondrial redox homeostasis for the first time. Mitochondrial CHK1 function loss mediated the cardiotoxicity induced by AZD7762 and CHK1-knockout. Mechanistically, mitochondrial CHK1 directly phosphorylates SIRT3 and promotes its expression within mitochondria. On the contrary, both AZD7762 or CHK1-knockout and gemcitabine decreased mitochondrial SIRT3 abundance, thus resulting in respiration dysfunction. Further hiPSC-CMs and mice experiments demonstrated that SIRT3 overexpression maintained mitochondrial function while alleviating CM pyroptosis, and thereby improving mice cardiac function. In summary, our results suggest that targeting SIRT3 could represent a novel therapeutic approach for clinical prevention and treatment of cardiotoxicity induced by CHK1 inhibition and gemcitabine.
Collapse
Affiliation(s)
- Jia-Wen Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Tian-Kai Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Tian-Wen Wei
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Qi-Qi Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Chong Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Ling-Feng Gu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Tong-Tong Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Liu-Hua Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Si-Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Yu-Lin Bao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Li-Ping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ai-Hua Gu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Chong-Qi Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.
| | - Qi-Ming Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China.
| | - Lian-Sheng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
50
|
Lapa BS, Costa MI, Figueiredo D, Jorge J, Alves R, Monteiro AR, Serambeque B, Laranjo M, Botelho MF, Carreira IM, Sarmento-Ribeiro AB, Gonçalves AC. AZD-7648, a DNA-PK Inhibitor, Induces DNA Damage, Apoptosis, and Cell Cycle Arrest in Chronic and Acute Myeloid Leukemia Cells. Int J Mol Sci 2023; 24:15331. [PMID: 37895013 PMCID: PMC10607085 DOI: 10.3390/ijms242015331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The non-homologous end joining pathway is vital for repairing DNA double-strand breaks (DSB), with DNA-dependent protein kinase (DNA-PK) playing a critical role. Altered DNA damage response (DDR) in chronic (CML) and acute myeloid leukemia (AML) offers potential therapeutic opportunities. We studied the therapeutic potential of AZD-7648 (DNA-PK inhibitor) in CML and AML cell lines. This study used two CML (K-562 and LAMA-84) and five AML (HEL, HL-60, KG-1, NB-4, and THP-1) cell lines. DDR gene mutations were obtained from the COSMIC database. The copy number and methylation profile were evaluated using MS-MLPA and DDR genes, and telomere length using qPCR. p53 protein expression was assessed using Western Blot, chromosomal damage through cytokinesis-block micronucleus assay, and γH2AX levels and DSB repair kinetics using flow cytometry. Cell density and viability were analyzed using trypan blue assay after treatment with AZD-7648 in concentrations ranging from 10 to 200 µM. Cell death, cell cycle distribution, and cell proliferation rate were assessed using flow cytometry. The cells displayed different DNA baseline damage, DDR gene expressions, mutations, genetic/epigenetic changes, and p53 expression. Only HEL cells displayed inefficient DSB repair. The LAMA-84, HEL, and KG-1 cells were the most sensitive to AZD-7648, whereas HL-60 and K-562 showed a lower effect on density and viability. Besides the reduction in cell proliferation, AZD-7648 induced apoptosis, cell cycle arrest, and DNA damage. In conclusion, these results suggest that AZD-7648 holds promise as a potential therapy for myeloid leukemias, however, with variations in drug sensitivity among tested cell lines, thus supporting further investigation to identify the specific factors influencing sensitivity to this DNA-PK inhibitor.
Collapse
Affiliation(s)
- Beatriz Santos Lapa
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Maria Inês Costa
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Diana Figueiredo
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| | - Ana Raquel Monteiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
| | - Beatriz Serambeque
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Institute of Biophysics, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mafalda Laranjo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Institute of Biophysics, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Institute of Biophysics, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Isabel Marques Carreira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Cytogenetics and Genomics Laboratory, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-061 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.L.); (M.I.C.); (J.J.); (R.A.); (A.R.M.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (B.S.); (I.M.C.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
| |
Collapse
|