1
|
Malik U, Pal D. Isoxazole compounds: Unveiling the synthetic strategy, in-silico SAR & toxicity studies and future perspective as PARP inhibitor in cancer therapy. Eur J Med Chem 2024; 279:116898. [PMID: 39353240 DOI: 10.1016/j.ejmech.2024.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Latest developments in cancer treatment have shed a light on the crucial role of PARP inhibitors that enhance the treatment effectiveness by modifying abnormal repair pathways. PARP inhibitors, such as Olaparib, Rucaparib, Niraparib, and Talazoparib have been approved in a number of cancers including BRCA 1/BRCA2 associated malignancies although there are many difficulties as therapeutical resistance. Besides the conventional synthetic drugs, natural compounds such as flavones and flavonoids have been found to be PARP inhibitors but only in preclinical studies. Isoxazole is very important class of potential candidates for medicinal chemistry with anti-cancer and other pharmacological activities. At present, there are no approved PARP inhibitors of isoxazole origin but their ability to hit many pathways inside the cancer cells points out on its importance for future treatments design. In drug development, isoxazoles are helpful because of the molecular design flexibility that may be enhanced using various synthetic approaches. This review highlights the molecular mechanisms of PARP inhibition, importance of isoxazole compounds and present advances in their synthetic strategies that demonstrate promise for these agents as new anticancer drugs. It emphasizes that isoxazole-based PARP inhibitors compounds could be novel anti-cancer drugs. Through this review, we hope to grow a curiosity in additional explorations of isoxazole-based PARP inhibitors and their applications in the trends of novel insights towards precision cancer therapy.
Collapse
Affiliation(s)
- Udita Malik
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495009, India
| | - Dilipkumar Pal
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495009, India.
| |
Collapse
|
2
|
Luo J, Li Y, Zhang Y, Wu D, Ren Y, Liu J, Wang C, Zhang J. An update on small molecule compounds targeting synthetic lethality for cancer therapy. Eur J Med Chem 2024; 278:116804. [PMID: 39241482 DOI: 10.1016/j.ejmech.2024.116804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Targeting cancer-specific vulnerabilities through synthetic lethality (SL) is an emerging paradigm in precision oncology. A SL strategy based on PARP inhibitors has demonstrated clinical efficacy. Advances in DNA damage response (DDR) uncover novel SL gene pairs. Beyond BRCA-PARP, emerging SL targets like ATR, ATM, DNA-PK, CHK1, WEE1, CDK12, RAD51, and RAD52 show clinical promise. Selective and bioavailable small molecule inhibitors have been developed to induce SL, but optimization for potency, specificity, and drug-like properties remains challenging. This article illuminated recent progress in the field of medicinal chemistry centered on the rational design of agents capable of eliciting SL specifically in neoplastic cells. It is envisioned that innovative strategies harnessing SL for small molecule design may unlock novel prospects for targeted cancer therapeutics going forward.
Collapse
Affiliation(s)
- Jiaxiang Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yang Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yiwen Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Defa Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Jie Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Chengdi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Health and Frontiers Science Center for Disease-related Molecular Network and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
3
|
Lu G, Zou Z, Xin M, Meng Y, Cheng Z, Du Z, Gu J, Zhang X, Zou Y. Carbamoylation at C-8 position of natural 3-arylcoumarin scaffold for the discovery of novel PARP-1 inhibitors with potent anticancer activity. Eur J Med Chem 2024; 277:116726. [PMID: 39116535 DOI: 10.1016/j.ejmech.2024.116726] [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: 06/14/2024] [Revised: 07/21/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Structural modification based on natural privileged scaffolds has proven to be an attractive approach to generate potential antitumor candidates with high potency and specific targeting. As a continuation of our efforts to identify potent PARP-1 inhibitors, natural 3-arylcoumarin scaffold was served as the starting point for the construction of novel structural unit for PARP-1 inhibition. Herein, a series of novel 8-carbamyl-3-arylcoumarin derivatives were designed and synthesized. The antiproliferative activities of target compounds against four BRCA-mutated cancer cells (SUM149PT, HCC1937, MDA-MB-436 and Capan-1) were evaluated. Among them, compound 9b exhibited excellent antiproliferative effects against SUM149PT, HCC1937 and Capan-1 cells with IC50 values of 0.62, 1.91 and 4.26 μM, respectively. Moreover, 9b could significantly inhibit the intracellular PARP-1/2 activity in SUM149PT cells with IC50 values of 2.53 nM and 6.45 nM, respectively. Further mechanism studies revealed that 9b could aggravate DNA double-strand breaks, increase ROS production, decrease mitochondrial membrane potential, arrest cell cycle at G2/M phase and ultimately induce apoptosis in SUM149PT cells. In addition, molecular docking study demonstrated that the binding mode of 9b with PARP-1 was similar to that of niraparib, forming multiple hydrogen bond interactions with the active site of PARP-1. Taken together, these findings suggest that 8-carbamyl-3-arylcoumarin scaffold could serve as an effective structural unit for PARP-1 inhibition and offer a valuable paradigm for the structural modification of natural products.
Collapse
Affiliation(s)
- Guoqing Lu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Zhiru Zou
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Meixiu Xin
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yingfen Meng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Zhuo Cheng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Zhibo Du
- Zhongshan Wanhan Pharmaceuticals Co., Ltd., Zhongshan, 528451, PR China
| | - Jiayi Gu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xuejing Zhang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yong Zou
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
Almohdar D, Kamble P, Basavannacharya C, Gulkis M, Calbay O, Huang S, Narayan S, Çağlayan M. Impact of DNA ligase inhibition on the nick sealing of polβ nucleotide insertion products at the downstream steps of base excision repair pathway. Mutagenesis 2024; 39:263-279. [PMID: 38736258 DOI: 10.1093/mutage/geae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024] Open
Abstract
DNA ligase (LIG) I and IIIα finalize base excision repair (BER) by sealing a nick product after nucleotide insertion by DNA polymerase (pol) β at the downstream steps. We previously demonstrated that a functional interplay between polβ and BER ligases is critical for efficient repair, and polβ mismatch or oxidized nucleotide insertions confound the final ligation step. Yet, how targeting downstream enzymes with small molecule inhibitors could affect this coordination remains unknown. Here, we report that DNA ligase inhibitors, L67 and L82-G17, slightly enhance hypersensitivity to oxidative stress-inducing agent, KBrO3, in polβ+/+ cells more than polβ-/- null cells. We showed less efficient ligation after polβ nucleotide insertions in the presence of the DNA ligase inhibitors. Furthermore, the mutations at the ligase inhibitor binding sites (G448, R451, A455) of LIG1 significantly affect nick DNA binding affinity and nick sealing efficiency. Finally, our results demonstrated that the BER ligases seal a gap repair intermediate by the effect of polβ inhibitor that diminishes gap filling activity. Overall, our results contribute to understand how the BER inhibitors against downstream enzymes, polβ, LIG1, and LIGIIIα, could impact the efficiency of gap filling and subsequent nick sealing at the final steps leading to the formation of deleterious repair intermediates.
Collapse
Affiliation(s)
- Danah Almohdar
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| | - Pradnya Kamble
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| | - Chandrakala Basavannacharya
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| | - Mitchell Gulkis
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| | - Ozlem Calbay
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, United States
| | - Shuang Huang
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, United States
| | - Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, United States
| | - Melike Çağlayan
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| |
Collapse
|
5
|
Fu XL, Guo SM, Ma JQ, Ma FY, Wang X, Tang YX, Li Y, Zhang WY, Ye LH. HBXIP induces PARP1 via WTAP-mediated m 6A modification and CEBPA-activated transcription in cisplatin resistance to hepatoma. Acta Pharmacol Sin 2024; 45:2405-2419. [PMID: 38871923 PMCID: PMC11489769 DOI: 10.1038/s41401-024-01309-5] [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: 01/26/2024] [Accepted: 05/08/2024] [Indexed: 06/15/2024] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is a DNA-binding protein that is involved in various biological functions, including DNA damage repair and transcription regulation. It plays a crucial role in cisplatin resistance. Nevertheless, the exact regulatory pathways governing PARP1 have not yet been fully elucidated. In this study, we present evidence suggesting that the hepatitis B X-interacting protein (HBXIP) may exert regulatory control over PARP1. HBXIP functions as a transcriptional coactivator and is positively associated with PARP1 expression in tissues obtained from hepatoma patients in clinical settings, and its high expression promotes cisplatin resistance in hepatoma. We discovered that the oncogene HBXIP increases the level of PARP1 m6A modification by upregulating the RNA methyltransferase WTAP, leading to the accumulation of the PARP1 protein. In this process, on the one hand, HBXIP jointly activates the transcription factor ETV5, promoting the activation of the WTAP promoter and further facilitating the promotion of the m6A modification of PARP1 by WTAP methyltransferase, enhancing the RNA stability of PARP1. On the other hand, HBXIP can also jointly activate the transcription factor CEBPA, enhance the activity of the PARP1 promoter, and promote the upregulation of PARP1 expression, ultimately leading to enhanced DNA damage repair capability and promoting cisplatin resistance in hepatoma. Notably, aspirin inhibits HBXIP, thereby reducing the expression of PARP1. Overall, our research revealed a novel mechanism for increasing PARP1 abundance, and aspirin therapy could overcome cisplatin resistance in hepatoma.
Collapse
Affiliation(s)
- Xue-Li Fu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Shi-Man Guo
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jia-Qi Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Fang-Yuan Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xue Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan-Xin Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ye Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Wei-Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Li-Hong Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
6
|
Yang Y, Mou Y, Wan LX, Zhu S, Wang G, Gao H, Liu B. Rethinking therapeutic strategies of dual-target drugs: An update on pharmacological small-molecule compounds in cancer. Med Res Rev 2024; 44:2600-2623. [PMID: 38769656 DOI: 10.1002/med.22057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/06/2023] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Oncogenes and tumor suppressors are well-known to orchestrate several signaling cascades, regulate extracellular and intracellular stimuli, and ultimately control the fate of cancer cells. Accumulating evidence has recently revealed that perturbation of these key modulators by mutations or abnormal protein expressions are closely associated with drug resistance in cancer therapy; however, the inherent drug resistance or compensatory mechanism remains to be clarified for targeted drug discovery. Thus, dual-target drug development has been widely reported to be a promising therapeutic strategy for improving drug efficiency or overcoming resistance mechanisms. In this review, we provide an overview of the therapeutic strategies of dual-target drugs, especially focusing on pharmacological small-molecule compounds in cancer, including small molecules targeting mutation resistance, compensatory mechanisms, synthetic lethality, synergistic effects, and other new emerging strategies. Together, these therapeutic strategies of dual-target drugs would shed light on discovering more novel candidate small-molecule drugs for the future cancer treatment.
Collapse
Affiliation(s)
- Yiren Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yi Mou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Lin-Xi Wan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shiou Zhu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Huiyuan Gao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Du J, Zhang J, Liu D, Gao L, Liao H, Chu L, Lin J, Li W, Meng X, Zou F, Cai S, Zou M, Dong H. 1G6-D7 Inhibits Homologous Recombination Repair by Targeting Extracellular HSP90α to Promote Apoptosis in Non-Small Cell Lung Cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:4884-4898. [PMID: 38899512 DOI: 10.1002/tox.24356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
Despite recent advances in treatment, non-small cell lung cancer (NSCLC) continues to have a high mortality rate. Currently, NSCLC pathogenesis requires further investigation, and therapeutic drugs are still under development. Homologous recombination repair (HRR) repairs severe DNA double-strand breaks. Homologous recombination repair deficiency (HRD) occurs when HRR is impaired and causes irreparable double-strand DNA damage, leading to genomic instability and increasing the risk of cancer development. Poly(ADP-ribose) polymerase (PARP) inhibitors can effectively treat HRD-positive tumors. Extracellular heat shock protein 90α (eHSP90α) is highly expressed in hypoxic environments and inhibits apoptosis, thereby increasing cellular tolerance. Here, we investigated the relationship between eHSP90α and HRR in NSCLC. DNA damage models were established in NSCLC cell lines (A549 and H1299). The activation of DNA damage and HRR markers, apoptosis, proliferation, and migration were investigated. In vivo tumor models were established using BALB/c nude mice and A549 cells. We found that human recombinant HSP90α stimulation further activated HRR and reduced DNA damage extent; however, eHSP90α monoclonal antibody, 1G6-D7, effectively inhibited HRR. HRR inhibition and increased apoptosis were observed after LRP1 knockdown; this effect could not be reversed with hrHSP90α addition. The combined use of 1G6-D7 and olaparib caused significant apoptosis and HRR inhibition in vitro and demonstrated promising anti-tumor effects in vivo. Extracellular HSP90α may be involved in HRR in NSCLC through LRP1. The combined use of 1G6-D7 and PARP inhibitors may exert anti-tumor effects by inhibiting DNA repair and further inducing apoptosis of NSCLC cells.
Collapse
Affiliation(s)
- Jiangzhou Du
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinming Zhang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Gao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Liao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lanhe Chu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Lin
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Li
- Department of Dermatology, The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, California, Los Angeles, USA
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengchen Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Valenza C, Marsicano RM, Trapani D, Curigliano G. PARP inhibitor resistant BRCA-mutated advanced breast cancer: current landscape and emerging treatments. Curr Opin Oncol 2024; 36:474-479. [PMID: 39246166 DOI: 10.1097/cco.0000000000001092] [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: 09/10/2024]
Abstract
PURPOSE OF REVIEW Patients with advanced breast cancer (aBC) treated with PARP inhibitors (PARPi) can eventually experience disease progression for emerging treatment resistance. This review aims to depict the treatment the molecular landscape, and the innovative therapies for patients with PARPi-resistant BRCA-mutated aBC. RECENT FINDINGS No specific therapy is specifically available in the setting post-PARPi-failure, with antibody-drug conjugates or nonplatinum-based chemotherapy (PBC) representing the best treatment options in this setting. Mechanisms of on-target PARPi resistance can be classified in reversions (60%) and nonreversion (40%); reverse mutations restore PARP functions. According to the first evidence of clinical validity, these alterations are associated with lower efficacy of PARPi and PBC. However, their clinical utility needs to be assessed. SUMMARY PARPi-resistant aBC represents a clinical unmet need due to the lack of specific targeted therapies and validated prognostic and predictive biomarkers. Constant efforts are required to better define the mechanisms of PARPi resistance and, consequently, develop biomarker-based treatment approach to prevent or overcame resistance.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Renato Maria Marsicano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Fukumoto W, Okamura S, Tamai M, Arima J, Kawahara I, Fukuda I, Mitsuke A, Sakaguchi T, Sugita S, Matsushita R, Tatarano S, Yamada Y, Nakagawa M, Enokida H, Yoshino H. Development of a novel treatment based on PKMYT1 inhibition for cisplatin-resistant bladder cancer with miR-424-5p-dependent cyclin E1 amplification. BMC Cancer 2024; 24:1333. [PMID: 39472827 PMCID: PMC11523841 DOI: 10.1186/s12885-024-13109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Chemotherapy including cisplatin is recommended for the treatment of advanced bladder cancer, but its effectiveness is limited due to the acquisition of drug resistance. Although several mechanisms of cisplatin resistance have been reported, there are still many unknowns, and treatment of cisplatin-resistant bladder cancer remains difficult. Accordingly, in this study, we aimed to identify and characterize microRNAs involved in cisplatin resistance. METHODS Small RNA sequencing analysis was performed to search for microRNAs related to cisplatin resistance. The identified microRNAs were then characterized using gain-of-function studies, sensitivity analysis, target gene analysis, and cellular assays. RESULTS We identified miR-424-5p as a candidate microRNA that was downregulated in cisplatin-resistant strains compared with parental strains. Notably, in gain-of-function studies, miR-424-5p suppressed the proliferative ability of cisplatin-resistant bladder cancer (CDDP-R BC). Furthermore, miR-424-5p restored sensitivity to cisplatin. RNA sequence analysis revealed seven candidate genes targeted by this microRNA. Among them, cyclin E1 (CCNE1) was chosen for subsequent analyses because its expression was upregulated in cisplatin-resistant cells compared with parental cells and because recent studies have shown that CCNE1 amplification is synthetic lethal with PKMYT1 kinase inhibition. Therefore, we performed functional analysis using the PKMYT1 inhibitor RP-6306 and demonstrated that RP-6306 inhibited cell growth through suppression of mitotic entry and restored cisplatin sensitivity in CDDP-R BC. CONCLUSIONS Overall, our findings provided insights into the development of novel therapeutic strategies for CDDP-R BC.
Collapse
Affiliation(s)
- Wataru Fukumoto
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shunsuke Okamura
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Motoki Tamai
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Junya Arima
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Ichiro Kawahara
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Ikumi Fukuda
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Akihiko Mitsuke
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Takashi Sakaguchi
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Satoshi Sugita
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Ryosuke Matsushita
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Shuichi Tatarano
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yasutoshi Yamada
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Masayuki Nakagawa
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Hideki Enokida
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Hirofumi Yoshino
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan.
| |
Collapse
|
10
|
He M, Jiang H, Li S, Xue M, Wang H, Zheng C, Tong J. The crosstalk between DNA-damage responses and innate immunity. Int Immunopharmacol 2024; 140:112768. [PMID: 39088918 DOI: 10.1016/j.intimp.2024.112768] [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: 05/20/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
DNA damage is typically caused during cell growth by DNA replication stress or exposure to endogenous or external toxins. The accumulation of damaged DNA causes genomic instability, which is the root cause of many serious disorders. Multiple cellular organisms utilize sophisticated signaling pathways against DNA damage, collectively known as DNA damage response (DDR) networks. Innate immune responses are activated following cellular abnormalities, including DNA damage. Interestingly, recent studies have indicated that there is an intimate relationship between the DDR network and innate immune responses. Diverse kinds of cytosolic DNA sensors, such as cGAS and STING, recognize damaged DNA and induce signals related to innate immune responses, which link defective DDR to innate immunity. Moreover, DDR components operate in immune signaling pathways to induce IFNs and/or a cascade of inflammatory cytokines via direct interactions with innate immune modulators. Consistently, defective DDR factors exacerbate the innate immune imbalance, resulting in severe diseases, including autoimmune disorders and tumorigenesis. Here, the latest progress in understanding crosstalk between the DDR network and innate immune responses is reviewed. Notably, the dual function of innate immune modulators in the DDR network may provide novel insights into understanding and developing targeted immunotherapies for DNA damage-related diseases, even carcinomas.
Collapse
Affiliation(s)
- Mei He
- College of Life Sciences, Hebei University, Baoding 071002, China; Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Hua Jiang
- Department of Hematology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Shun Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610041, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Jie Tong
- College of Life Sciences, Hebei University, Baoding 071002, China.
| |
Collapse
|
11
|
Ma X, Tian F, Xiao Y, Huang M, Song D, Chen X, Xu H. Synergistic effects of bloom helicase (BLM) inhibitor AO/854 with cisplatin in prostate cancer. Sci Rep 2024; 14:24962. [PMID: 39438537 PMCID: PMC11496540 DOI: 10.1038/s41598-024-75938-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
To determine the synergistic effect and mechanism of AO/854, a new Bloom syndrome protein (BLM) helicase inhibitor, and cisplatin (CDDP), a DNA-crosslinking agent, cell viability assays, neutral comet assays, and Western blotting (WB) were performed on prostate cancer (PCa) cells. According to our findings, combining AO/854 and CDDP enhanced the antiproliferative capabilities of PC3 cell lines. As evidenced by the upregulation of γH2AX, cleaved caspase-3/caspase-3, and BAX/Bcl-2, AO/854 dramatically increased PC3 apoptosis and DNA damage induced by CDDP. Furthermore, combining AO/854 and CDDP synergistically inhibited PC3 cell migration and invasion. In addition, AO/854 inhibited CDDP-induced S-phase cell-cycle arrest in PC3 cells while enhancing G2/M-phase cell-cycle arrest. In vivo, the antitumor efficacy of the combination therapy group was greater than that of the groups treated with AO/854 or CDDP alone. Our findings indicate that synergistic chemotherapy with AO/854 and CDDP may be a novel anticancer strategy for PCa.
Collapse
Affiliation(s)
- Xiaoyan Ma
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang, 550003, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Life Sciences, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Fu Tian
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang, 550003, China
| | - Yuanpin Xiao
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang, 550003, China
| | - Mengqiu Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Life Sciences, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Dandan Song
- Department of Brewing Engineering, Moutai Institute, Renhuai, 564500, China
| | - Xinlin Chen
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang, 550003, China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Life Sciences, Guizhou University, Guiyang, 550025, China.
- College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
12
|
Salazar Saez R, Zorrilla M, Sánchez R, Cebollero A, Manrique I, Martín A, de Ávila L, Lacalle-Emborujo A, Martin-Rodriguez S, Bernardo-González I, Alonso M. Molecular analysis of BRCA1 and BRCA2 genes in La Rioja (Spain): five new variants. Hered Cancer Clin Pract 2024; 22:22. [PMID: 39438962 PMCID: PMC11495111 DOI: 10.1186/s13053-024-00296-2] [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: 07/05/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND To study BRCA1/2 gene variants in La Rioja in the northcentral area of Spain. METHODS We performed a molecular analysis of BRCA1 and BRCA2 in 642 individuals from 427 different families from June 2008 to December 2019. RESULTS We identified 71 families with pathogenic variants in these genes, 32 families with BRCA1 variants and 39 families with BRCA2 variants. The pathogenic variants c.959delG in BRCA1 and c.1363_1369delTCAGAGA, c.1397dupA, c.4234_4236delACTinsC and c.8387delC in BRCA2 have not been previously described. The c.81-2 A > T variant in BRCA1, detected in two unrelated families, has not been reported previously in the Spanish population. Two large genomic deletions were found in the BRCA1 gene in exons (Ex) 23-24 and Ex1A-1B-2, and one deletion was found in the BRCA2 gene in Ex2. The pathogenic variant c.5123 C > A in BRCA1 was detected in 8 unrelated families and was the most frequent pathogenic variant in our population. The c.6024dupG mutation in BRCA2 was detected in 6 unrelated families; the c.2808_2011delACAA mutation in BRCA2 was found in 5 different families; the c.211 A > G mutation in BRCA1 was found in three different families; and the c.68_69delAG, c81-2 A > T, c.4038_4039delAA, and c.5266dupC variants in BRCA1 and the c.2457delA, c.2701delC, c.5116_5119delAATA, c.6275delTT, c.7558 C > T and c.7617 + 1G > A variants in BRCA2 were found in two different families. CONCLUSIONS The spectrum of pathogenic variants in the BRCA1/2 genes in La Rioja is similar to that in other Spanish regions, with some unique characteristics. The pathogenic c.6024dupG variant in the BRCA2 gene was detected in a large number of families and could have a founding effect in the Ebro riverside areas in the regions of La Rioja and Navarra. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Raquel Salazar Saez
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain.
- Servicio de Oncología Médica, Hospital Universitario San Pedro, 3ª planta, Calle Piqueras nº 98, Logroño, La Rioja, 26006, Spain.
| | - Miriam Zorrilla
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| | - Rosa Sánchez
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| | - Ana Cebollero
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| | - Isabel Manrique
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| | - Alfonso Martín
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| | - Leticia de Ávila
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| | | | | | | | - Martina Alonso
- Clinical Oncology Unit, Hospital Universitario San Pedro de Logroño, La Rioja, Spain
| |
Collapse
|
13
|
Conte M, Cabeza Fernández V, Oliver FJ, Alarcón T, Soler J. Emergence of cyclic hypoxia and the impact of PARP inhibitors on tumor progression. NPJ Syst Biol Appl 2024; 10:122. [PMID: 39433780 PMCID: PMC11494044 DOI: 10.1038/s41540-024-00453-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Tumor hypoxia is a dynamic phenomenon marked by fluctuations in oxygen levels across both rapid (seconds to minutes) and slow (hours to days) time scales. While short hypoxia cycles are relatively well understood, the mechanisms behind longer cycles remain largely unclear. In this paper, we present a novel mechanistic mathematical model that explains slow hypoxia cycles through feedback loops involving vascular expansion and regression, oxygen-regulated tumor growth, and toxic cytokine production. Our study reveals that, for the emergence of slow hypoxia cycles, endothelial cells must adapt by decreasing receptor activation as ligand concentration increases. Additionally, the interaction between tumor cells and toxic cytokines influences frequency and intensity of these cycles. By examining the effects of pharmacological interventions, specifically poly (ADP-ribose) polymerase inhibitors, we also demonstrate how targeting cell proliferation can help regulate oxygen levels. Our findings enhance the understanding of hypoxia regulation and suggest PARP proteins as promising therapeutic targets.
Collapse
Affiliation(s)
- Martina Conte
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parco Area delle Scienze 53/A, 43124, Parma, Italy.
- Division of Mathematical Oncology and Computational Systems Biology Department of Computational and Quantitative Medicine, Beckman Research Institute City of Hope National Medical Center, 1500 E Duarte Rd., Duarte, 91010, CA, USA.
| | - Vanesa Cabeza Fernández
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento 17, Armilla, 18016, Spain
| | - F Javier Oliver
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, Parque Tecnológico de Ciencias de la Salud, Avenida del Conocimiento 17, Armilla, 18016, Spain
| | - Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys 23, Barcelona, 08010, Spain
- Centre de Recerca Matemàtica, Campus Universitat Autònoma de Barcelona, Carrer de l'Albareda, Bellaterra, 08193, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Carrer de l'Albareda, Bellaterra, 08193, Spain
| | - Juan Soler
- Departamento de Matemática Aplicada and Research Unit "Modeling Nature" (MNat), Universidad de Granada, Avenida de la Fuente Nueva S/N, Granada, 18071, Spain.
| |
Collapse
|
14
|
Feng Y, Long Y, Wang H, Ouyang Y, Li Q, Wu M, Zheng J. Benchmarking machine learning methods for synthetic lethality prediction in cancer. Nat Commun 2024; 15:9058. [PMID: 39428397 PMCID: PMC11491473 DOI: 10.1038/s41467-024-52900-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Synthetic lethality (SL) is a gold mine of anticancer drug targets, exposing cancer-specific dependencies of cellular survival. To complement resource-intensive experimental screening, many machine learning methods for SL prediction have emerged recently. However, a comprehensive benchmarking is lacking. This study systematically benchmarks 12 recent machine learning methods for SL prediction, assessing their performance across diverse data splitting scenarios, negative sample ratios, and negative sampling techniques, on both classification and ranking tasks. We observe that all the methods can perform significantly better by improving data quality, e.g., excluding computationally derived SLs from training and sampling negative labels based on gene expression. Among the methods, SLMGAE performs the best. Furthermore, the methods have limitations in realistic scenarios such as cold-start independent tests and context-specific SLs. These results, together with source code and datasets made freely available, provide guidance for selecting suitable methods and developing more powerful techniques for SL virtual screening.
Collapse
Affiliation(s)
- Yimiao Feng
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Yahui Long
- Bioformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - He Wang
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yang Ouyang
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Quan Li
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Min Wu
- Institute for Infocomm Research, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Jie Zheng
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Engineering Research Center of Intelligent Vision and Imaging, Shanghai, China.
| |
Collapse
|
15
|
Huang YZ, Sang MY, Xi PW, Xu RX, Cai MY, Wang ZW, Zhao JY, Li YH, Wei JF, Ding Q. FANCI Inhibition Induces PARP1 Redistribution to Enhance the Efficacy of PARP Inhibitors in Breast Cancer. Cancer Res 2024; 84:3447-3463. [PMID: 39037758 DOI: 10.1158/0008-5472.can-23-2738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/27/2024] [Accepted: 07/19/2024] [Indexed: 07/23/2024]
Abstract
Breast cancer is a global public health concern with high mortality rates, necessitating the development of innovative treatment strategies. PARP inhibitors have shown efficacy in certain patient populations, but their application is largely limited to cancers with homologous recombination deficiency. Here, we identified the suppression of FANCI as a therapeutic strategy to enhance the efficacy of PARP inhibitors in breast cancer. Elevated FANCI expression in breast cancer was associated with poor prognosis and increased cell proliferation and migration. FANCI interacted with PARP1, and suppressing FANCI limited the nuclear localization and functionality of PARP1. Importantly, FANCI inhibition sensitized breast cancer cells to the PARP inhibitor talazoparib in the absence of BRCA mutations. Additionally, the CDK4/6 inhibitor palbociclib enhanced the sensitivity of breast cancer cells to talazoparib through FANCI inhibition. These findings highlight the potential of targeting FANCI to enhance the efficacy of PARP inhibitors in treating breast cancer. Significance: Targeting FANCI is a promising therapeutic strategy for enhancing PARP inhibitor sensitivity in breast cancer that holds potential for broader therapeutic applications beyond cancers harboring BRCA mutations.
Collapse
Affiliation(s)
- Yu-Zhou Huang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Ming-Yi Sang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Pei-Wen Xi
- Health Management Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Ruo-Xi Xu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Meng-Yuan Cai
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Zi-Wen Wang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Jian-Yi Zhao
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Yi-Han Li
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Ji-Fu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, PR China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
16
|
da Costa AA, Somuncu O, Ravindranathan R, Mukkavalli S, Martignetti DB, Nguyen H, Jiao Y, Lamarre BP, Sadatrezaei G, Moreau L, Liu J, Iyer DR, Lazaro JB, Shapiro GI, Parmar K, D’Andrea AD. Single-Stranded DNA Gap Accumulation Is a Functional Biomarker for USP1 Inhibitor Sensitivity. Cancer Res 2024; 84:3435-3446. [PMID: 38885312 PMCID: PMC11474172 DOI: 10.1158/0008-5472.can-23-4007] [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: 01/25/2024] [Revised: 04/15/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Recent studies suggest that PARP and POLQ inhibitors confer synthetic lethality in BRCA1-deficient tumors by accumulation of single-stranded DNA (ssDNA) gaps at replication forks. Loss of USP1, a deubiquitinating enzyme, is also synthetically lethal with BRCA1 deficiency, and USP1 inhibitors are now undergoing clinical development for these cancers. Herein, we show that USP1 inhibitors also promote the accumulation of ssDNA gaps during replication in BRCA1-deficient cells, and this phenotype correlates with drug sensitivity. USP1 inhibition increased monoubiquitinated proliferating cell nuclear antigen at replication forks, mediated by the ubiquitin ligase RAD18, and knockdown of RAD18 caused USP1 inhibitor resistance and suppression of ssDNA gaps. USP1 inhibition overcame PARP inhibitor resistance in a BRCA1-mutated xenograft model and induced ssDNA gaps. Furthermore, USP1 inhibition was synergistic with PARP and POLQ inhibition in BRCA1-mutant cells, with enhanced ssDNA gap accumulation. Finally, in patient-derived ovarian tumor organoids, sensitivity to USP1 inhibition alone or in combination correlated with the accumulation of ssDNA gaps. Assessment of ssDNA gaps in ovarian tumor organoids represents a rapid approach for predicting response to USP1 inhibition in ongoing clinical trials. Significance: USP1 inhibitors kill BRCA1-deficient cells and cause ssDNA gap accumulation, supporting the potential of using ssDNA gap detection as a functional biomarker for clinical trials on USP1 inhibitors.
Collapse
Affiliation(s)
- Alexandre A. da Costa
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Ozge Somuncu
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Ramya Ravindranathan
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Sirisha Mukkavalli
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - David B. Martignetti
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Huy Nguyen
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Yuqing Jiao
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Benjamin P. Lamarre
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Golbahar Sadatrezaei
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Lisa Moreau
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Joyce Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Divya R. Iyer
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Jean-Bernard Lazaro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Geoffrey I. Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Kalindi Parmar
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Alan D. D’Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
17
|
Cadzow L, Brenneman J, Tobin E, Sullivan P, Nayak S, Ali JA, Shenker S, Griffith J, McGuire M, Grasberger P, Mishina Y, Murray M, Dodson AE, Gannon H, Krall E, Hixon J, Chipumuro E, Sinkevicius K, Gokhale PC, Ganapathy S, Matulonis UA, Liu JF, Olaharski A, Sangurdekar D, Liu H, Wilt J, Schlabach M, Stegmeier F, Wylie AA. The USP1 Inhibitor KSQ-4279 Overcomes PARP Inhibitor Resistance in Homologous Recombination-Deficient Tumors. Cancer Res 2024; 84:3419-3434. [PMID: 39402989 PMCID: PMC11474170 DOI: 10.1158/0008-5472.can-24-0293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/24/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024]
Abstract
Defects in DNA repair pathways play a pivotal role in tumor evolution and resistance to therapy. At the same time, they create vulnerabilities that render tumors dependent on the remaining DNA repair processes. This phenomenon is exemplified by the clinical activity of PARP inhibitors in tumors with homologous recombination (HR) repair defects, such as tumors with inactivating mutations in BRCA1 or BRCA2. However, the development of resistance to PARP inhibitors in BRCA-mutant tumors represents a high unmet clinical need. In this study, we identified deubiquitinase ubiquitin-specific peptidase-1 (USP1) as a critical dependency in tumors with BRCA mutations or other forms of HR deficiency and developed KSQ-4279, the first potent and selective USP1 inhibitor to enter clinical testing. The combination of KSQ-4279 with a PARP inhibitor was well tolerated and induced durable tumor regression across several patient-derived PARP-resistant models. These findings indicate that USP1 inhibitors represent a promising therapeutic strategy for overcoming PARP inhibitor resistance in patients with BRCA-mutant/HR-deficient tumors and support continued testing in clinical trials. Significance: KSQ-4279 is a potent and selective inhibitor of USP1 that induces regression of PARP inhibitor-resistant tumors when dosed in combination with PARP inhibitors, addressing an unmet clinical need for BRCA-mutant tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Elsa Krall
- KSQ Therapeutics, Lexington, Massachusetts.
| | - Jeff Hixon
- KSQ Therapeutics, Lexington, Massachusetts.
| | | | | | - Prafulla C. Gokhale
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Suthakar Ganapathy
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | | | - Joyce F. Liu
- Dana Farber Cancer Institute, Boston, Massachusetts.
| | | | | | - Hanlan Liu
- KSQ Therapeutics, Lexington, Massachusetts.
| | | | | | | | | |
Collapse
|
18
|
Akamandisa MP, Boddicker NJ, Yadav S, Hu C, Hart SN, Ambrosone C, Anton-Culver H, Auer PL, Bodelon C, Burnside ES, Chen F, Eliassen HA, Goldgar DE, Haiman C, Hodge JM, Huang H, John EM, Karam R, Lacey JV, Lindstroem S, Martinez E, Na J, Neuhausen SL, O'Brien KM, Olson JE, Pal T, Palmer JR, Patel AV, Pesaran T, Polley EC, Richardson ME, Ruddy K, Sandler DP, Teras LR, Trentham-Dietz A, Vachon CM, Weinberg C, Winham SJ, Yao S, Zirpoli G, Kraft P, Weitzel JN, Domchek SM, Couch FJ, Nathanson KL. Association of Gene Variant Type and Location with Breast Cancer Risk in the General Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315237. [PMID: 39417132 PMCID: PMC11482981 DOI: 10.1101/2024.10.11.24315237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Importance Pathogenic variants (PVs) in ATM, BRCA1, BRCA2, CHEK2 , and PALB2 are associated with increased breast cancer risk. However, it is unknown whether breast cancer risk differs by PV type or location in carriers ascertained from the general population. Objective To evaluate breast cancer risks associated with PV type and location in ATM, BRCA1, BRCA2, CHEK2 , and PALB2 . Design Age adjusted case-control association analysis for all participants, subsets of PV carriers, and women with no breast cancer family history in population-based and clinical testing cohorts. Setting Twelve US population-based studies within the Cancer Risk Estimates Related to Susceptibility (CARRIERS) Consortium, and breast cancer cases from the UK-Biobank and an Ambry Genetics clinical testing cohort. Participants 32,247 women with and 32,544 age-matched women without a breast cancer diagnosis from CARRIERS; 237 and 1351 women with BRCA2 PVs and breast cancer from the UKBB and Ambry Genetics, respectively. Exposures PVs in ATM, BRCA1, BRCA2, CHEK2, and PALB2. Main Outcomes and Measures PVs were grouped by type and location within genes and assessed for risks of breast cancer (odds ratios (OR), 95% confidence intervals (CI), and p-values) using logistic regression. Mean ages at diagnosis were compared using linear regression. Results Compared to women carrying BRCA2 exon 11 protein truncating variants (PTVs) in the CARRIERS population-based study, women with BRCA2 ex13-27 PTVs (OR=2.7, 95%CI 1.1-7.9) and ex1-10 PTVs (OR=1.6, 95%CI 0.8-3.5) had higher breast cancer risks, lower rates of ER-negative breast cancer (ex13-27 OR=0.5, 95%CI 0.2-0.9; ex1-10 OR=0.5, 95%CI 0.1-1.0), and earlier age of breast cancer diagnosis (ex13-27 5.5 years, p<0.001; ex1-10 2.4 years, p=0.17). These associations with ER-negative breast cancer and age replicated in a high-risk clinical cohort and the population-based UK Biobank cohort. No differences in risk or age at diagnosis by gene region were observed for PTVs in other predisposition genes. Conclusions and Relevance Population-based and clinical high-risk cohorts establish that PTVs in exon 11 of BRCA2 are associated with reduced risk of breast cancer, later age at diagnosis, and greater risk of ER-negative disease. These differential risks may improve individualized risk prediction and clinical management for women carrying BRCA2 PTVs. Key Points Question: Does ATM , BRCA1 , BRCA2 , CHEK2 and PALB2 pathogenic variant type and location influence breast cancer risk in population-based studies? Findings: Breast cancer risk and estrogen receptor status differ based on the type and location of pathogenic variants in BRCA2 . Women carrying protein truncating variants in exon 11 have a lower breast cancer risk in the population-based cohorts, older age at diagnosis and higher rates of estrogen receptor negative breast cancer than women with exon 1-10 or exon 13-27 truncation variants in population-based and clinical testing cohorts. Meaning: Incorporating pathogenic variant type and location in cancer risk models may improve individualized risk prediction.
Collapse
|
19
|
Jiang Y, Wang J, Zhang Y, Cao Z, Zhang Q, Su J, He S, Bo X. Graph based recurrent network for context specific synthetic lethality prediction. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-023-2618-y. [PMID: 39422810 DOI: 10.1007/s11427-023-2618-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/10/2024] [Indexed: 10/19/2024]
Abstract
The concept of synthetic lethality (SL) has been successfully used for targeted therapies. To further explore SL for cancer therapy, identifying more SL interactions with therapeutic potential are essential. Recently, graph neural network-based deep learning methods have been proposed for SL prediction, which reduce the SL search space of wet-lab based methods. However, these methods ignore that most SL interactions depend strongly on genetic context, which limits the application of the predicted results. In this study, we proposed a graph recurrent network-based model for specific context-dependent SL prediction (SLGRN). In particular, we introduced a Graph Recurrent Network-based encoder to acquire a context-specific, low-dimensional feature representation for each node, facilitating the prediction of novel SL. SLGRN leveraged gate recurrent unit (GRU) and it incorporated a context-dependent-level state to effectively integrate information from all nodes. As a result, SLGRN outperforms the state-of-the-arts models for SL prediction. We subsequently validate novel SL interactions under different contexts based on combination therapy or patient survival analysis. Through in vitro experiments and retrospective clinical analysis, we emphasize the potential clinical significance of this context-specific SL prediction model.
Collapse
Affiliation(s)
- Yuyang Jiang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Jing Wang
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yixin Zhang
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - ZhiWei Cao
- School of Informatics, Xiamen University, Xiamen, 361005, China
| | - Qinglong Zhang
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Jinsong Su
- School of Informatics, Xiamen University, Xiamen, 361005, China.
| | - Song He
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| | - Xiaochen Bo
- Department of Bioinformatics, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| |
Collapse
|
20
|
Miao R, Blue K, Sommerer K, Shah A, Bottiglieri S, del Cueto A, Berry DK, Ho TT, Hicks JK, Kim DW. PARP Inhibitors in Pancreatic Cancer with Homologous Recombination Repair Gene Mutations: A Single-Institution Experience. Cancers (Basel) 2024; 16:3447. [PMID: 39456541 PMCID: PMC11505755 DOI: 10.3390/cancers16203447] [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: 09/01/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Limited data are available regarding the anticancer activity of PARP inhibitors (PARPis) in pancreatic cancer with mutations in HRR genes other than BRCA and PALB2. METHODS We retrospectively reviewed the clinical characteristics and outcomes of 48 patients with advanced pancreatic cancer harboring pathogenic germline and/or somatic HRR mutations who were treated with PARPis. RESULTS Thirty patients had germline (g)HRR mutations only, twelve had somatic (s)HRR mutations only, and six had concomitant gHRR and sHRR mutations. The objective response rate (ORR) was 22%. The median progression-free survival (mPFS) and overall survival (mOS) were 6.9 and 11.5 months, respectively. Five patients received olaparib in the front-line setting due to borderline performance status. Their ORR was 20%, and their mPFS and mOS were both 11.3 months. The ORR was higher in patients with BRCA or PALB2 mutations (germline or somatic) than in those with non-BRCA/PALB2 mutations. Patients with somatic non-BRCA/PALB2 variants had a shorter mPFS. Patients with concomitant gHRR/sHRR mutations or gHRR mutations alone had a significantly longer mPFS than those with sHRR mutations only. CONCLUSIONS PARP inhibitors may be considered for patients with advanced pancreatic cancer harboring pathogenic alterations of BRCA who cannot tolerate standard chemotherapy. Maintenance PARPis can be considered in selected patients with non-BRCA/non-PALB2 HRR mutations.
Collapse
Affiliation(s)
- Ruoyu Miao
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Kirsten Blue
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Katelyn Sommerer
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Anand Shah
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Sal Bottiglieri
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Alex del Cueto
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Darcy K. Berry
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Teresa T. Ho
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - James Kevin Hicks
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Dae Won Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| |
Collapse
|
21
|
Li W, Chen G, Wang Y, Jiang Y, Wu N, Hu M, Wu T, Yue W. Functional Analysis of BARD1 Missense Variants on Homology-Directed Repair in Ovarian and Breast Cancers. Mol Carcinog 2024. [PMID: 39387837 DOI: 10.1002/mc.23829] [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: 04/22/2024] [Revised: 09/07/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
Women with germline BRCA1 mutations face an increased risk of developing breast and ovarian cancers. BARD1 (BRCA1 associated RING domain 1) is an essential heterodimeric partner of BRCA1, and mutations in BARD1 are also associated with these cancers. While BARD1 mutations are recognized for their cancer susceptibility, the exact roles of numerous BARD1 missense mutations remain unclear. In this study, we conducted functional assays to assess the homology-directed DNA repair (HDR) activity of all BARD1 missense substitutions identified in 55 breast and ovarian cancer samples, using the real-world data from the COSMIC and cBioPortal databases. Seven BARD1 variants (V85M, P187A, G491R, R565C, P669L, T719R, and Q730L) were confirmed to impair DNA damage repair. Furthermore, cells harboring these BARD1 variants exhibited increased sensitivity to the chemotherapeutic drugs, cisplatin, and olaparib, compared to cells expressing wild-type BARD1. These findings collectively suggest that these seven missense BARD1 variants are likely pathogenic and may respond well to cisplatin-olaparib combination therapy. This study not only enhances our understanding of BARD1's role in DNA damage repair but also offers valuable insights into predicting therapy responses in patients with specific BARD1 missense mutations.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Gynecology and Obstetrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Guansheng Chen
- Department of Gynecology and Obstetrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- Department of Gynecology and Obstetrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yuening Jiang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
| | - Nanlin Wu
- Department of Pathology, Chuzhou First People's Hospital, Anhui, China
| | - Mingjie Hu
- School of Life Science, Bengbu Medical University, Anhui, China
| | - Taju Wu
- School of Life Science, Bengbu Medical University, Anhui, China
| | - Wei Yue
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Interdisciplinary Eye Research Institute (EYE-X Institute), Bengbu Medical University, Anhui, China
| |
Collapse
|
22
|
Wang J, Wang ZQ, Zong W. ADP-ribose hydrolases: biological functions and potential therapeutic targets. Expert Rev Mol Med 2024; 26:e21. [PMID: 39375922 PMCID: PMC11488344 DOI: 10.1017/erm.2024.17] [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: 11/30/2023] [Revised: 03/19/2024] [Accepted: 05/15/2024] [Indexed: 10/09/2024]
Abstract
ADP-ribosylation (ADPRylation), which encompasses poly(ADP-ribosyl)ation and mono(ADP-ribosyl)ation, is an important post-translational modification catalysed by the poly(ADP-ribose) polymerase (PARP) enzyme superfamily. The process involves writers (PARPs) and erasers (ADP-ribose hydrolases), which work together to precisely regulate diverse cellular and molecular responses. Although the PARP-mediated synthesis of ADP-ribose (ADPr) has been well studied, ADPr degradation by degrading enzymes deserves further investigation. Nonetheless, recent studies have provided important new insights into the biology and functions of ADPr hydrolases. Notably, research has illuminated the significance of the poly(ADP-ribose) degradation pathway and its activation by the coordinated actions of poly(ADP-ribose) glycohydrolase and other ADPr hydrolases, which have been identified as key components of ADPRylation signalling networks. The degradation pathway has been proposed to play crucial roles in key cellular processes, such as DNA damage repair, chromatin dynamics, transcriptional regulation and cell death. A deep understanding of these ADPr erasing enzymes provides insights into the biological roles of ADPRylation in human health and disease aetiology and paves the road for the development of novel therapeutic strategies. This review article provides a summary of current knowledge about the biochemical and molecular functions of ADPr erasers and their physiological implications in human pathology.
Collapse
Affiliation(s)
- Jingpeng Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Zhao-Qi Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Jena 07743, Germany
| | - Wen Zong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| |
Collapse
|
23
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
24
|
Gong Z, Zhou D, Shen H, Ma C, Wu D, Hou L, Wang H, Xu T. Development of a prognostic model related to homologous recombination deficiency in glioma based on multiple machine learning. Front Immunol 2024; 15:1452097. [PMID: 39434883 PMCID: PMC11491349 DOI: 10.3389/fimmu.2024.1452097] [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: 06/20/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Background Despite advances in neuro-oncology, treatments of glioma and tools for predicting the outcome of patients remain limited. The objective of this research is to construct a prognostic model for glioma using the Homologous Recombination Deficiency (HRD) score and validate its predictive capability for glioma. Methods We consolidated glioma datasets from TCGA, various cancer types for pan-cancer HRD analysis, and two additional glioma RNAseq datasets from GEO and CGGA databases. HRD scores, mutation data, and other genomic indices were calculated. Using machine learning algorithms, we identified signature genes and constructed an HRD-related prognostic risk model. The model's performance was validated across multiple cohorts. We also assessed immune infiltration and conducted molecular docking to identify potential therapeutic agents. Results Our analysis established a correlation between higher HRD scores and genomic instability in gliomas. The model, based on machine learning algorithms, identified seven key genes, significantly predicting patient prognosis. Moreover, the HRD score prognostic model surpassed other models in terms of prediction efficacy across different cancers. Differential immune cell infiltration patterns were observed between HRD risk groups, with potential implications for immunotherapy. Molecular docking highlighted several compounds, notably Panobinostat, as promising for high-risk patients. Conclusions The prognostic model based on the HRD score threshold and associated genes in glioma offers new insights into the genomic and immunological landscapes, potentially guiding therapeutic strategies. The differential immune profiles associated with HRD-risk groups could inform immunotherapeutic interventions, with our findings paving the way for personalized medicine in glioma treatment.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Haotian Shen
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chao Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hongxiang Wang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
25
|
Xia Y, Huang P, Qian YY, Wang Z, Jin N, Li X, Pan W, Wang SY, Jin P, Drokow EK, Li X, Zhang Q, Zhang Z, Li P, Fang Y, Yang XP, Han Z, Gao QL. PARP inhibitors enhance antitumor immune responses by triggering pyroptosis via TNF-caspase 8-GSDMD/E axis in ovarian cancer. J Immunother Cancer 2024; 12:e009032. [PMID: 39366751 PMCID: PMC11459312 DOI: 10.1136/jitc-2024-009032] [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] [Accepted: 09/22/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND In addition to their established action of synthetic lethality in tumor cells, poly(ADP-ribose) polymerase inhibitors (PARPis) also orchestrate tumor immune microenvironment (TIME) that contributes to suppressing tumor growth. However, it remains not fully understood whether and how PARPis trigger tumor-targeting immune responses. METHODS To decode the immune responses reshaped by PARPis, we conducted T-cell receptor (TCR) sequencing and immunohistochemical (IHC) analyses of paired clinical specimens before and after niraparib monotherapy obtained from a prospective study, as well as ID8 mouse ovarian tumors. To validate the induction of immunogenic cell death (ICD) by PARPis, we performed immunofluorescence/IHC staining with homologous recombination deficiency tumor cells and patient-derived xenograft tumor tissues, respectively. To substantiate that PARPis elicited tumor cell pyroptosis, we undertook comprehensive assessments of the cellular morphological features, cleavage of gasdermin (GSDM) proteins, and activation of TNF-caspase signaling pathways through genetic downregulation/depletion and selective inhibition. We also evaluated the critical role of pyroptosis in tumor suppression and immune activation following niraparib treatment using a syngeneic mouse model with implanting CRISPR/Cas9 edited Gsdme-/ - ID8 tumor cells into C57BL/6 mice. RESULTS Our findings revealed that PARPis augmented the proportion of neoantigen-recognized TCR clones and TCR clonal expansion, and induced an inflamed TIME characterized by increased infiltration of both innate and adaptive immune cells. This PARPis-strengthened immune response was associated with the induction of ICD, specifically identified as pyroptosis, which possessed distinctive morphological features and GSDMD/E cleavage. It was validated that the cleavage of GSDMD/E was due to elevated caspase 8 activity downstream of the TNFR1, rather than FAS and TRAIL-R. On PARP inhibition, the NF-κB signaling pathway was activated, leading to increased secretion of TNF-α and subsequent initiation of the TNFR1-caspase 8 cascade. Impeding pyroptosis through the depletion of Gsdme significantly compromised the tumor-suppressing effects of PARP inhibition and undermined the anti-immune response in the syngeneic ID8 mouse model. CONCLUSIONS PARPis induce a specific type of ICD called pyroptosis via TNF-caspase 8-GSDMD/E axis, resulting in an inflamed TIME and augmentation of tumor-targeting immune responses. These findings deepen our understanding of PARPis activities and point toward a promising avenue for synergizing PARPis with immunotherapeutic interventions. TRIAL REGISTRATION NUMBER NCT04507841.
Collapse
Affiliation(s)
- Yu Xia
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pu Huang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yi-yu Qian
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zanhong Wang
- Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Ning Jin
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Pan
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si-Yuan Wang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Jin
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Emmanuel Kwateng Drokow
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Central South University, Changsha, Hunan, China
| | - Xiong Li
- Department of Gynecology & Obstetrics, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Zhengmao Zhang
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pingfei Li
- Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yong Fang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang-Ping Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiqiang Han
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Qing-lei Gao
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education, Hubei Provincial Key Laboratory of Tumor Invasion and Metastasis), Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrics and Gynecology, Department of Gynecological Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
26
|
Peng Y, Liu D, Huang D, Inuzuka H, Liu J. PROTAC as a novel anti-cancer strategy by targeting aging-related signaling. Semin Cancer Biol 2024; 106-107:143-155. [PMID: 39368654 DOI: 10.1016/j.semcancer.2024.09.004] [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: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024]
Abstract
Aging and cancer share common cellular hallmarks, including cellular senescence, genomic instability, and abnormal cell death and proliferation, highlighting potential areas for therapeutic interventions. Recent advancements in targeted protein degradation technologies, notably Proteolysis-Targeting Chimeras (PROTACs), offer a promising approach to address these shared pathways. PROTACs leverage the ubiquitin-proteasome system to specifically degrade pathogenic proteins involved in cancer and aging, thus offering potential solutions to key oncogenic drivers and aging-related cellular dysfunction. This abstract summarizes the recent progress of PROTACs in targeting critical proteins implicated in both cancer progression and aging, and explores future perspectives in integrating these technologies for more effective cancer treatments.
Collapse
Affiliation(s)
- Yunhua Peng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Donghua Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an 710061, China
| | - Daoyuan Huang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Jing Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an 710061, China.
| |
Collapse
|
27
|
Xie X, Chen C, Wang C, Guo Y, Sun B, Tian J, Yan J, Li D, Chen G. Targeting GPX4-mediated ferroptosis protection sensitizes BRCA1-deficient cancer cells to PARP inhibitors. Redox Biol 2024; 76:103350. [PMID: 39265497 PMCID: PMC11415882 DOI: 10.1016/j.redox.2024.103350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024] Open
Abstract
BRCA1 is one of the most frequently-mutated tumor suppressor genes in ovarian and breast cancers. Loss of BRCA1 triggers homologous recombination (HR) repair deficiency, consequently leading to genomic instability and PARP inhibitors (PARPi)-associated synthetic lethality. Although, the roles of BRCA1 in DNA repair and replication have been extensively investigated, its tumor suppressive functions beyond genome safeguard remain poorly understood. Here, we report that BRCA1 promotes ferroptosis susceptibility through catalyzing K6-linked polyubiquitination of GPX4 and subsequently accelerating GPX4 degradation. Depletion of BRCA1 induces ferroptosis resistance in ovarian cancer cells due to elevated GPX4 protein, and silence of GPX4 significantly suppresses the growth of BRCA1-deficient ovarian cancer xenografts. Importantly, we found that PARPi triggers ferroptosis in ovarian cancer cells, inhibition of GPX4 markedly increase PARPi-induced ferroptosis in BRCA1-deficient ovarian cancer cells. Combined treatment of GPX4 inhibitor and PARPi produces synergistic anti-tumor efficacy in BRCA1-deficient ovarian cancer cells, patient derived organoid (PDO) and xenografts. Thus, our study uncovers a novel mechanism via which BRCA1 exerts tumor suppressive function through regulating ferroptosis, and demonstrates the potential of GPX4 as a therapeutic target for BRCA1-mutant cancers.
Collapse
Affiliation(s)
- Xuexia Xie
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China; Department of Anesthesiology and General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China
| | - Congcong Chen
- Department of Anesthesiology and General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China
| | - Cong Wang
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yongjian Guo
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Binghe Sun
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaxin Tian
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jin Yan
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Dake Li
- Department of Gynecology, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Guo Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
28
|
Di Marco T, Mazzoni M, Greco A, Cassinelli G. Non-oncogene dependencies: Novel opportunities for cancer therapy. Biochem Pharmacol 2024; 228:116254. [PMID: 38704100 DOI: 10.1016/j.bcp.2024.116254] [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: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Targeting oncogene addictions have changed the history of subsets of malignancies and continues to represent an excellent therapeutic opportunity. Nonetheless, alternative strategies are required to treat malignancies driven by undruggable oncogenes or loss of tumor suppressor genes and to overcome drug resistance also occurring in cancers addicted to actionable drivers. The discovery of non-oncogene addiction (NOA) uncovered novel therapeutically exploitable "Achilles' heels". NOA refers to genes/pathways not oncogenic per sé but essential for the tumor cell growth/survival while dispensable for normal cells. The clinical success of several classes of conventional and molecular targeted agents can be ascribed to their impact on both tumor cell-associated intrinsic as well as microenvironment-related extrinsic NOA. The integration of genetic, computational and pharmacological high-throughput approaches led to the identification of an expanded repertoire of synthetic lethality interactions implicating NOA targets. Only a few of them have been translated into the clinics as most NOA vulnerabilities are not easily druggable or appealing targets. Nonetheless, their identification has provided in-depth knowledge of tumor pathobiology and suggested novel therapeutic opportunities. Here, we summarize conceptual framework of intrinsic and extrinsic NOA providing exploitable vulnerabilities. Conventional and emerging methodological approaches used to disclose NOA dependencies are reported together with their limits. We illustrate NOA paradigmatic and peculiar examples and outline the functional/mechanistic aspects, potential druggability and translational interest. Finally, we comment on difficulties in exploiting the NOA-generated knowledge to develop novel therapeutic approaches to be translated into the clinics and to fully harness the potential of clinically available drugs.
Collapse
Affiliation(s)
- Tiziana Di Marco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Mara Mazzoni
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Angela Greco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
29
|
Collier KA, Stover DG. Confirmation of large BRCA2 reversion deletions leading to reconstituted in-frame transcripts detected via circulating tumor DNA. Ann Oncol 2024; 35:917-919. [PMID: 39048081 DOI: 10.1016/j.annonc.2024.07.720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/22/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024] Open
Affiliation(s)
- K A Collier
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus; Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer, Columbus
| | - D G Stover
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus; Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer, Columbus; Department of Biomedical Informatics, Ohio State University, Columbus, USA.
| |
Collapse
|
30
|
Ikeuchi H, Matsuno Y, Kusumoto-Matsuo R, Kojima S, Ueno T, Ikegami M, Kitada R, Sumiyoshi-Okuma H, Kojima Y, Yonemori K, Yatabe Y, Takamochi K, Suzuki K, Yoshioka KI, Mano H, Kohsaka S. GLI1 confers resistance to PARP inhibitors by activating the DNA damage repair pathway. Oncogene 2024; 43:3037-3048. [PMID: 39095584 DOI: 10.1038/s41388-024-03105-1] [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: 01/14/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024]
Abstract
Identifying the mechanisms of action of anticancer drugs is an important step in the development of new drugs. In this study, we established a comprehensive screening platform consisting of 68 oncogenes (MANO panel), encompassing 243 genetic variants, to identify predictive markers for drug efficacy. Validation was performed using drugs that targeted EGFR, BRAF, and MAP2K1, which confirmed the utility of this functional screening panel. Screening of a BRCA2-knockout DLD1 cell line (DLD1-KO) revealed that cells expressing SMO and GLI1 were resistant to olaparib. Gene set enrichment analysis identified genes associated with DNA damage repair that were enriched in cells overexpressing SMO and GLI1. The expression of genes associated with homologous recombination repair (HR), such as the FANC family and BRCA1/2, was significantly upregulated by GLI1 expression, which is indicative of PARP inhibitor resistance. Although not all representative genes of the nucleotide excision repair (NER) pathway were upregulated, NER activity was enhanced by GLI1. The GLI1 inhibitor was effective against DLD1-KO cells overexpressing GLI1 both in vitro and in vivo. Furthermore, the combination therapy of olaparib and GLI1 inhibitor exhibited a synergistic effect on DLD1-KO, suggesting the possible clinical application of GLI1 inhibitor targeting cancer with defective DNA damage repair. This platform enables the identification of biomarkers associated with drug sensitivity, and is a useful tool for drug development.
Collapse
Affiliation(s)
- Hiroshi Ikeuchi
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yusuke Matsuno
- Laboratory of Genome Stability Maintenance, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Rika Kusumoto-Matsuo
- Laboratory of Genome Stability Maintenance, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Shinya Kojima
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Masachika Ikegami
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Rina Kitada
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | - Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken-Ichi Yoshioka
- Laboratory of Genome Stability Maintenance, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan.
| |
Collapse
|
31
|
Fang M, Lin Y, Xue C, Sheng K, Guo Z, Han Y, Lin H, Wu Y, Sang Y, Chen X, Howell SB, Lin X, Lin X. The AKT inhibitor AZD5363 elicits synthetic lethality in ARID1A-deficient gastric cancer cells via induction of pyroptosis. Br J Cancer 2024; 131:1080-1091. [PMID: 39003371 PMCID: PMC11405682 DOI: 10.1038/s41416-024-02778-5] [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: 07/03/2023] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a deadly disease with poor overall survival and limited therapeutic options. Genetic alterations such as mutations and/or deletions in chromatin remodeling gene AT-rich interactive domain 1 A (ARID1A) occur frequently in GC. Although ARID1A mutations/deletions are not a druggable target for conventional treatments, novel therapeutic strategies based on a synthetic lethal approach may be effective for the treatment of ARID1A-deficient cancers. METHODS A kinase inhibitor library containing 551 compounds was screened in ARID1A isogenic GC cells for the ability to induce synthetic lethality effect. Selected hits' activity was validated, and the mechanism of the most potent candidate drug, AKT inhibitor AD5363 (capivasertib), on induction of the synthetic lethality with ARID1A deficiency was investigated. RESULTS After robust vulnerability screening of 551 diverse protein kinase inhibitors, we identified the AKT inhibitor AZD5363 as being the most potent lead compound in inhibiting viability of ARID1A-/- cells. A synthetic lethality between loss of ARID1A expression and AKT inhibition by AZD5363 was validated in both GC cell model system and xenograft model. Mechanistically, AZD5363 treatment induced pyroptotic cell death in ARID1A-deficient GC cells through activation of the Caspase-3/GSDME pathway. Furthermore, ARID1A occupied the AKT gene promoter and regulated its transcription negatively, thus the GC cells deficient in ARID1A showed increased expression and phosphorylation of AKT. CONCLUSIONS Our study demonstrates a novel synthetic lethality interaction and unique mechanism between ARID1A loss and AKT inhibition, which may provide a therapeutic and mechanistic rationale for targeted therapy on patients with ARID1A-defective GC who are most likely to be beneficial to AZD5363 treatment.
Collapse
Affiliation(s)
- Menghan Fang
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Youfen Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chaorong Xue
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Kaiqin Sheng
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Zegeng Guo
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuting Han
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Hanbin Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuecheng Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Yuchao Sang
- Scientific Research Center, Anxi County Hospital, Quanzhou, China
| | - Xintan Chen
- Scientific Research Center, Anxi County Hospital, Quanzhou, China
| | - Stephen B Howell
- Department of Medicine and the Moores Cancer Center, University of California, San Diego, CA, USA
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| | - Xinjian Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Scientific Research Center, Anxi County Hospital, Quanzhou, China.
| |
Collapse
|
32
|
Ashworth A. Thirty years since the race to the BRCA1 gene. Nature 2024; 634:1062-1063. [PMID: 39478204 DOI: 10.1038/d41586-024-03358-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
|
33
|
Yin Z, Tao J, Liu Y, Chen H, Hu K, Wang Y, Xiong M. In Silico Analysis Uncovers FOXA1 as a Potential Biomarker for Predicting Neoadjuvant Chemotherapy Response in Fine-Needle Aspiration Biopsies. J Cancer 2024; 15:6052-6072. [PMID: 39440050 PMCID: PMC11493000 DOI: 10.7150/jca.101901] [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: 08/04/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
Background: The preoperative identification of neoadjuvant chemotherapy (NAC) treatment responsiveness in breast cancer (BC) patients is advantageous for tailoring treatment regimens. There is a relative scarcity in the current research exploring NAC treatment responsive biomarkers using bulk sequencing data obtained from fine-needle aspiration (FNA). Materials and Methods: Limma was employed for the selection of differentially expressed genes. Additionally, WGCNA, machine learning, and Genetic Perturbation Similarity Analysis (GPSA) were utilized to identify key genes associated with NAC treatment response. ConsensusClusterPlus was employed for unsupervised clustering. Rt-qPCR and WB were conducted to assess gene expression and protein levels in clinical tissues and cell lines. The Seahorse XF96 Extracellular Flux Analyzer was utilized to evaluate Extracellular Acidification Rate (ECAR) and Oxygen Consumption Rate (OCR). The "pRRophetic" package was used for drug sensitivity prediction, while CB-Dock2 was applied for molecular docking and optimal pose presentation. Spatial transcriptomic analysis was based on the CROST database. Results: Eleven biomarkers were identified associated with NAC treatment response in BC patients, with FOXA1 identified as a pivotal hub gene among them. The expression levels of FOXA1 showed a significant positive correlation with genomic stability and a marked negative correlation with the homologous recombination deficiency (HRD) score. Downregulation of the FOXA1 gene resulted in reduced glycolysis in MCF-7 cells.Additionally, FOXA1 were found to serve as a biomarker for both NAC and PARP inhibitor treatment sensitivity in BC patients. Spatial transcriptomic analysis indicates significantly elevated infiltration of T follicular helper (T-FH) cells and mast cells surrounding tumors exhibiting high FOXA1 expression. Conclusion: In summary, our study involved the analysis of diverse sequencing datasets derived from various FNA samples to identify biomarkers sensitive to NAC, thereby offering novel insights into resources for future personalized clinical treatment strategies.
Collapse
Affiliation(s)
- Zhenglang Yin
- Department of General surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of General surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
| | - Jianfei Tao
- Department of General surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of General surgery, The People's Hospital of Feidong County, Hefei, 231699, China
- Department of Thoracic Surgery, The People's Hospital of Feidong County, Hefei, 231699, China
| | - Yanyan Liu
- Department of General surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
| | - Haohao Chen
- Department of General surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
| | - Kongwang Hu
- Department of General surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yao Wang
- Digestive Endoscopy Department, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Maoming Xiong
- Department of General surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
34
|
Ma J, Qin X, Le W, Chen X, Wang X, Xu C. Identification of BBC3 as a novel indicator for predicting prostate cancer development and olaparib resistance. Discov Oncol 2024; 15:496. [PMID: 39331229 PMCID: PMC11436583 DOI: 10.1007/s12672-024-01373-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Prostate cancer (PCa) is a commonly occurring malignancy in elderly men. Olaparib, a poly ADP-ribose polymerase inhibitor, is utilized in PCa treatment. However, patients often develop resistance to olaparib after a period of treatment. Genetic alterations may play a significant role in this resistance, but the specific genes involved remain unclear. This study collected RNA-sequence data from the Gene Expression Omnibus database on both olaparib-sensitive and -resistant PCa cells to identify genes crucial for resistance. Subsequently, the enriched pathways of these genes were analyzed, and a protein-protein interaction (PPI) network was constructed to identify hub genes. The effect of these hub genes on PCa occurrence, progression, and prognosis was assessed using data from The Cancer Genome Atlas and Chinese Prostate Cancer Genome and Epigenome Atlas databases. Finally, this study validated our findings in clinical PCa samples and cells. From the GSE189186 dataset, 50 upregulated genes and 2 downregulated genes were identified in olaparib-resistant C4-2B and LNCaP cells. Utilizing the PPI network, eight upregulated genes (BBC3, TP53I3, FDXR, DDB2, CDKN1A, GADD45A, ZMAT3, and SESN1) were identified as hub genes for olaparib-resistant PCa cells. Furthermore, some of these genes were central to PCa occurrence, with BBC3 also influencing progression and prognosis. Importantly, BBC3 expression was upregulated in clinical PCa samples and affected PCa cells sensitive to olaparib, suggesting its potential as a predictive marker for PCa development and olaparib resistance.
Collapse
Affiliation(s)
- Junjie Ma
- Department of Urology, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China
| | - Xin Qin
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China
| | - Wei Le
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China
| | - Xi Chen
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China
| | - Xiao Wang
- Department of Urology, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China.
| | - Chengdang Xu
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China.
| |
Collapse
|
35
|
Li J, Zhou M, Xie J, Chen J, Yang M, Ye C, Cheng S, Liu M, Li R, Tan R. Organoid modeling meets cancers of female reproductive tract. Cell Death Discov 2024; 10:410. [PMID: 39333482 PMCID: PMC11437045 DOI: 10.1038/s41420-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Diseases of the female reproductive system, especially malignant tumors, pose a serious threat to women's health worldwide. One of the key factors limiting research progress in this area is the lack of representative models. Organoid technology, especially tumor organoids, has been increasingly applied in the study of female reproductive system tumors due to their high heterogeneity, close resemblance to the physiological state, easy acquisition and cultivation advantages. They play a significant role in understanding the origin and causes of tumors, drug screening, and personalized treatment and more. This article reviews the organoid models for the female reproductive system, focusing on the cancer research advancements. It discusses the methods for constructing tumor organoids of the female reproductive tract and summarizes the limitations of current research. The aim is to offer a reference for future development and application of these organoid models, contributing to the advancement of anti-tumor drugs and treatment strategies for female reproductive tract cancer patients.
Collapse
Affiliation(s)
- Jiao Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengting Zhou
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Jiani Chen
- Chongqing Medical University, Chongqing, China
| | - Mengni Yang
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjun Ye
- Rehabilitation Department, Changgeng Yining Hospital, Wenzhou, China
| | - Shihu Cheng
- Geriatric Department, Changgeng Yining Hospital, Wenzhou, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
36
|
Li J, Jia Z, Dong L, Cao H, Huang Y, Xu H, Xie Z, Jiang Y, Wang X, Liu J. DNA damage response in breast cancer and its significant role in guiding novel precise therapies. Biomark Res 2024; 12:111. [PMID: 39334297 PMCID: PMC11437670 DOI: 10.1186/s40364-024-00653-2] [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: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
DNA damage response (DDR) deficiency has been one of the emerging targets in treating breast cancer in recent years. On the one hand, DDR coordinates cell cycle and signal transduction, whose dysfunction may lead to cell apoptosis, genomic instability, and tumor development. Conversely, DDR deficiency is an intrinsic feature of tumors that underlies their response to treatments that inflict DNA damage. In this review, we systematically explore various mechanisms of DDR, the rationale and research advances in DDR-targeted drugs in breast cancer, and discuss the challenges in its clinical applications. Notably, poly (ADP-ribose) polymerase (PARP) inhibitors have demonstrated favorable efficacy and safety in breast cancer with high homogenous recombination deficiency (HRD) status in a series of clinical trials. Moreover, several studies on novel DDR-related molecules are actively exploring to target tumors that become resistant to PARP inhibition. Before further clinical application of new regimens or drugs, novel and standardized biomarkers are needed to develop for accurately characterizing the benefit population and predicting efficacy. Despite the promising efficacy of DDR-related treatments, challenges of off-target toxicity and drug resistance need to be addressed. Strategies to overcome drug resistance await further exploration on DDR mechanisms, and combined targeted drugs or immunotherapy will hopefully provide more precise or combined strategies and expand potential responsive populations.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Heng Cao
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yansong Huang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhixuan Xie
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Yiwen Jiang
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Xiang Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
37
|
Nemetova U, Si̇yah P, Boran T, Bi̇lgi̇ Ç, Özyürek M, Şahi̇nler Ayla S. Synthesis and In Silico Evaluation of Piperazine-Substituted 2,3-Dichloro-5,8-dihydroxy-1,4-naphthoquinone Derivatives as Potential PARP-1 Inhibitors. ACS OMEGA 2024; 9:39733-39742. [PMID: 39346823 PMCID: PMC11425603 DOI: 10.1021/acsomega.4c04915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
PARP-1 (poly(ADP-ribose)-polymerase 1) inhibitors are vital in synthetic lethality, primarily due to their specificity for PARP-1 over PARP-2 (PARP-1 > PARP-2). This specificity is crucial as it allows precise inhibition of PARP-1 in tumor cells with Breast Cancer 1 protein (BRCA1) or BRCA2 deficiencies. The development of highly specific PARP-1 inhibitors not only meets the therapeutic needs of tumor treatment but also has the potential to minimize the adverse effects associated with nonselective PARP-2 inhibition. In this study, a series of novel 2,3-dichloro-5,8-dihydroxy-1,4-naphthoquinone (DDNO) derivatives were synthesized, characterized, and evaluated regarding their PARP-1 inhibitory and cytotoxic activity. Compound 3 exhibited the highest cytotoxic potential against all cell lines, except for MDA-MB-231 cells. The inhibitory potential of these molecules against PARP-1 was evaluated through in silico molecular docking and molecular dynamics studies. Notably, compounds 5, 9, and 13 exhibited significant inhibitory activity in silico results, interacting with critical amino acids known to be important for PARP-1 inhibition during simulations. These compounds exhibited target-specific and strong binding profiles, with docking scores of -7.17, -7.41, and -7.37 kcal/mol, respectively, and MM/GBSA scores of -52.51, -43.77, and -62.87 kcal/mol, respectively. These novel compounds (DDNO derivatives) hold promise as potential PARP-1 inhibitors for the development of targeted therapeutics against cancer.
Collapse
Affiliation(s)
- Ulviyye Nemetova
- Engineering
Faculty, Department of Chemistry, Organic Chemistry, Istanbul University-Cerrahpaşa, 34320 Istanbul, Turkey
| | - Pınar Si̇yah
- Department
of Biochemistry, Faculty of Pharmacy, Bahcesehir
University, 34353 Istanbul, Turkey
| | - Tuğçe Boran
- Faculty
of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul University-Cerrahpaşa, 34500 Istanbul, Turkey
| | - Çiğdem Bi̇lgi̇
- Faculty
of Pharmacy, Department of Pharmacognosy, Istanbul University-Cerrahpaşa, 34500 Istanbul, Turkey
| | - Mustafa Özyürek
- Engineering
Faculty, Department of Chemistry, Analytical Chemistry, Istanbul University-Cerrahpaşa, 34320 Istanbul, Turkey
| | - Sibel Şahi̇nler Ayla
- Engineering
Faculty, Department of Chemistry, Organic Chemistry, Istanbul University-Cerrahpaşa, 34320 Istanbul, Turkey
| |
Collapse
|
38
|
Bhandari SK, Wiest N, Sallmyr A, Du R, Tomkinson A. Redundant but essential functions of PARP1 and PARP2 in DNA ligase I-independent DNA replication. Nucleic Acids Res 2024; 52:10341-10354. [PMID: 39106163 PMCID: PMC11417376 DOI: 10.1093/nar/gkae672] [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: 02/09/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/09/2024] Open
Abstract
While DNA ligase I (LigI) joins most Okazaki fragments, a backup pathway involving poly(ADP-ribose) synthesis, XRCC1 and DNA ligase IIIα (LigIIIα) functions along with the LigI-dependent pathway and is also capable of supporting DNA replication in the absence of LigI. Here we have addressed for the first time the roles of PARP1 and PARP2 in this pathway using isogenic null derivatives of mouse CH12F3 cells. While single and double null mutants of the parental cell line and single mutants of LIG1 null cells were viable, loss of both PARP1 and PARP2 was synthetically lethal with LigI deficiency. Thus, PARP1 and PARP2 have a redundant essential role in LigI-deficient cells. Interestingly, higher levels of PARP2 but not PARP1 associated with newly synthesized DNA in the LIG1 null cells and there was a much higher increase in PARP2 chromatin retention in LIG1 null cells incubated with the PARP inhibitor olaparib with this effect occurring independently of PARP1. Together our results suggest that PARP2 plays a major role in specific cell types that are more dependent upon the backup pathway to complete DNA replication and that PARP2 retention at unligated Okazaki fragments likely contributes to the side effects of current clinical PARP inhibitors.
Collapse
Affiliation(s)
- Seema Khattri Bhandari
- Cancer Research Facility, Departments of Internal Medicine and Molecular Genetics & Microbiology, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, 915 Camino de Salud, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Nathaniel Wiest
- Cancer Research Facility, Departments of Internal Medicine and Molecular Genetics & Microbiology, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, 915 Camino de Salud, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Annahita Sallmyr
- Cancer Research Facility, Departments of Internal Medicine and Molecular Genetics & Microbiology, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, 915 Camino de Salud, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Ruofei Du
- Cancer Research Facility, Departments of Internal Medicine and Molecular Genetics & Microbiology, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, 915 Camino de Salud, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Alan E Tomkinson
- Cancer Research Facility, Departments of Internal Medicine and Molecular Genetics & Microbiology, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, 915 Camino de Salud, 1 University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
39
|
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: 0] [Impact Index Per Article: 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
|
40
|
Chen Q, Sun Y, Li H, Liu S, Zhang H, Cheng Z, Wang Y. Pixantrone as a novel MCM2 inhibitor for ovarian cancer treatment. Eur J Pharmacol 2024; 979:176835. [PMID: 39032764 DOI: 10.1016/j.ejphar.2024.176835] [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: 05/08/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Mini-chromosome maintenance protein 2 (MCM2) is a potential target for the development of cancer therapeutics. However, small molecule inhibitors targeting MCM2 need further investigation. METHODS Molecular dynamics simulation was performed to identify active pockets in the MCM2 protein structure (6EYC). The active pocket was used as a docking model to discover MCM2 inhibitors by using structure-based virtual screening and surface plasmon resonance (SPR) assay. Furthermore, the efficacy of pixantrone targeting MCM2 in ovarian cancer was evaluated in vitro and in vivo. RESULTS Pixantrone was identified as a novel inhibitor of MCM2 by virtual screening. SPR binding affinity analysis confirmed the direct binding of pixantrone to MCM2 protein. Pixantrone significantly reduced the viability of ovarian cancer cells A2780 and SKOV3 in a dose- and time-dependent manner. In addition, pixantrone inhibited DNA replication, and induced cell cycle arrest and apoptosis in ovarian cancer cells via targeting MCM2. Knockdown of MCM2 could attenuate the inhibitory activity of pixantrone in ovarian cancer cells. Furthermore, pixantrone significantly suppressed ovarian cancer growth in the A2780 cell xenograft mouse model and showed favorable safety. CONCLUSION These findings suggest that pixantrone may be a promising drug for ovarian cancer patients by targeting MCM2 in the clinic.
Collapse
Affiliation(s)
- Qingshan Chen
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China
| | - Yaoqi Sun
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tong Ji University, Shanghai, China
| | - Hao Li
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China
| | - Shupeng Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tong Ji University, Shanghai, China
| | - Hai Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China.
| | - Zhongping Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tong Ji University, Shanghai, China.
| | - Yu Wang
- Department of Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China.
| |
Collapse
|
41
|
Roggia M, Natale B, Amendola G, Grasso N, Di Maro S, Taliani S, Castellano S, Reina SCR, Salvati E, Amato J, Cosconati S. Discovering Dually Active Anti-cancer Compounds with a Hybrid AI-structure-based Approach. J Chem Inf Model 2024. [PMID: 39276072 DOI: 10.1021/acs.jcim.4c01132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
Cancer's persistent growth often relies on its ability to maintain telomere length and tolerate the accumulation of DNA damage. This study explores a computational approach to identify compounds that can simultaneously target both G-quadruplex (G4) structures and poly(ADP-ribose) polymerase (PARP)1 enzyme, offering a potential multipronged attack on cancer cells. We employed a hybrid virtual screening (VS) protocol, combining the power of machine learning with traditional structure-based methods. PyRMD, our AI-powered tool, was first used to analyze vast chemical libraries and to identify potential PARP1 inhibitors based on known bioactivity data. Subsequently, a structure-based VS approach selected compounds from these identified inhibitors for their G4 stabilization potential. This two-step process yielded 50 promising candidates, which were then experimentally validated for their ability to inhibit PARP1 and stabilize G4 structures. Ultimately, four lead compounds emerged as promising candidates with the desired dual activity and demonstrated antiproliferative effects against specific cancer cell lines. This study highlights the potential of combining Artificial Intelligence and structure-based methods for the discovery of multitarget anticancer compounds, offering a valuable approach for future drug development efforts.
Collapse
Affiliation(s)
- Michele Roggia
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Benito Natale
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Giorgio Amendola
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Nicola Grasso
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, Naples 80131, Italy
| | - Salvatore Di Maro
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Sabrina Castellano
- Dipartimento di Farmacia, Università di Salerno, Via Giovanni Paolo II 132, 84084 Fisciano Salerno, Italy
| | | | - Erica Salvati
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, Naples 80131, Italy
| | - Sandro Cosconati
- DiSTABiF, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| |
Collapse
|
42
|
Zhang J, Chen Y, Gong X, Yang Y, Gu Y, Huang L, Fu J, Zhao M, Huang Y, Li L, Liu W, Wan Y, He X, Ma Z, Zhao W, Zhang M, Tang T, Wang Y, Thiery JP, Zheng X, Chen L. GATA factor TRPS1, a new DNA repair protein, cooperates with reversible PARylation to promote chemoresistance in patients with breast cancer. J Biol Chem 2024; 300:107780. [PMID: 39276941 PMCID: PMC11490888 DOI: 10.1016/j.jbc.2024.107780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Resistance to DNA-damaging agents is a major unsolved challenge for breast cancer patients undergoing chemotherapy. Here, we show that elevated expression of transcriptional repressor GATA binding 1 (TRPS1) is associated with lower drug sensitivity, reduced response rate, and poor prognosis in chemotherapy-treated breast cancer patients. Mechanistically, elevated TRPS1 expression promotes hyperactivity of DNA damage repair (DDR) in breast cancer cells. We provide evidence that TRPS1 dynamically localizes to DNA breaks in a Ku70-and Ku80-dependent manner and that TRPS1 is a new member of the DDR protein family. We also discover that the dynamics of TRPS1 assembly at DNA breaks is regulated by its reversible PARylation in the DDR, and that mutations of the PARylation sites on TRPS1 lead to increased sensitivity to chemotherapeutic drugs. Taken together, our findings provide new mechanistic insights into the DDR and chemoresistance in breast cancer patients and identify TRPS1 as a critical DDR protein. TRPS1 may also be considered as a target to improve chemo-sensitization strategies and, consequently, clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yatao Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xue Gong
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yongfeng Yang
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Yun Gu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Ling Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jianfeng Fu
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Menglu Zhao
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yehong Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lulu Li
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenzhuo Liu
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yajie Wan
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xilin He
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhifang Ma
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Weiyong Zhao
- Department of Radiation Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meng Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tao Tang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yuzhi Wang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | | | - Xiaofeng Zheng
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China.
| | - Liming Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
43
|
Keggenhoff FL, Castven D, Becker D, Stojkovic S, Castven J, Zimpel C, Straub BK, Gerber T, Langer H, Hähnel P, Kindler T, Fahrer J, O'Rourke CJ, Ehmer U, Saborowski A, Ma L, Wang XW, Gaiser T, Matter MS, Sina C, Derer S, Lee JS, Roessler S, Kaina B, Andersen JB, Galle PR, Marquardt JU. PARP-1 selectively impairs KRAS-driven phenotypic and molecular features in intrahepatic cholangiocarcinoma. Gut 2024; 73:1712-1724. [PMID: 38857989 PMCID: PMC11420749 DOI: 10.1136/gutjnl-2023-331237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/24/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVE Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer with limited therapeutic options. KRAS mutations are among the most abundant genetic alterations in iCCA associated with poor clinical outcome and treatment response. Recent findings indicate that Poly(ADP-ribose)polymerase1 (PARP-1) is implicated in KRAS-driven cancers, but its exact role in cholangiocarcinogenesis remains undefined. DESIGN PARP-1 inhibition was performed in patient-derived and established iCCA cells using RNAi, CRISPR/Cas9 and pharmacological inhibition in KRAS-mutant, non-mutant cells. In addition, Parp-1 knockout mice were combined with iCCA induction by hydrodynamic tail vein injection to evaluate an impact on phenotypic and molecular features of Kras-driven and Kras-wildtype iCCA. Clinical implications were confirmed in authentic human iCCA. RESULTS PARP-1 was significantly enhanced in KRAS-mutant human iCCA. PARP-1-based interventions preferentially impaired cell viability and tumourigenicity in human KRAS-mutant cell lines. Consistently, loss of Parp-1 provoked distinct phenotype in Kras/Tp53-induced versus Akt/Nicd-induced iCCA and abolished Kras-dependent cholangiocarcinogenesis. Transcriptome analyses confirmed preferential impairment of DNA damage response pathways and replicative stress response mediated by CHK1. Consistently, inhibition of CHK1 effectively reversed PARP-1 mediated effects. Finally, Parp-1 depletion induced molecular switch of KRAS-mutant iCCA recapitulating good prognostic human iCCA patients. CONCLUSION Our findings identify the novel prognostic and therapeutic role of PARP-1 in iCCA patients with activation of oncogenic KRAS signalling.
Collapse
Affiliation(s)
- Friederike L Keggenhoff
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Diana Becker
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stojan Stojkovic
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Jovana Castven
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Carolin Zimpel
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Beate K Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Tissue Biobank of the University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Tiemo Gerber
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Harald Langer
- Cardiology Angiology, University Medical Centre, Mannheim, Germany
| | - Patricia Hähnel
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Kindler
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jörg Fahrer
- Department of Chemistry, RPTU Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Colm J O'Rourke
- Department of Health and Medical Sciences, University of Copenhagen Biotech Research & Innovation Centre, Kobenhavn, Denmark
| | - Ursula Ehmer
- Department of Internal Medicine II, Klinikum rechts der Isar der Technischen Universitat, München, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Timo Gaiser
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Matthias S Matter
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Christian Sina
- Institute of Nutritional Medicine, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Stefanie Derer
- Institute of Nutritional Medicine, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Bernd Kaina
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jesper B Andersen
- Department of Health and Medical Sciences, University of Copenhagen Biotech Research & Innovation Centre, Kobenhavn, Denmark
| | - Peter R Galle
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jens U Marquardt
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| |
Collapse
|
44
|
Li P, Zhu X, Qu H, Han Z, Yao X, Wei Y, Li B, Chen H. Synergistic Effect of Ubiquitin-Specific Protease 14 and Poly(ADP-Ribose) Glycohydrolase Co-Inhibition in BRCA1-Mutant, Poly(ADP-Ribose) Polymerase Inhibitor-Resistant Triple-Negative Breast Cancer Cells. Onco Targets Ther 2024; 17:741-753. [PMID: 39258222 PMCID: PMC11385694 DOI: 10.2147/ott.s463217] [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: 02/06/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
Purpose The clinical benefits of poly(ADP-ribose) polymerase (PARP) inhibitors are limited to triple-negative breast cancer (TNBC) with BRCA deficiency due to primary and acquired resistance. Thus, there is a pressing need to develop alternative treatment regimens to target BRCA-mutated TNBC tumors that are resistant to PARP inhibition. Similar to PARP, poly(ADP-ribose) glycohydrolase (PARG) plays a role in DNA replication and repair. However, there are conflicting reports on the vulnerability of BRCA1-deficient tumor cells to PARG inhibition. This study aims to investigate the synergistically lethal effect of the PARG inhibitor COH34 and the ubiquitin-specific protease (USP) 14 inhibitor IU1-248 and the underlying mechanisms in BRCA1-mutant, PARP inhibitor-resistant TNBC cells. Methods The cytotoxicity of PARG inhibition alone or in combination with USP14 inhibition in the BRCA-mutant, PARP inhibitor-resistant TNBC cell lines, HCC1937 and SUM149PT, was analyzed using cell viability and proliferation assays and flow cytometry. The molecular mechanisms underlying the synergistic effects of IU1-248 and COH34 were evaluated by immunofluorescence staining, DNA repair reporter assays and Western blot analysis. Results It was found that HCC1937 and SUM149PT cells exhibited moderate responsiveness to PARG inhibition alone. To the best of our knowledge, this research is the first to demonstrate that the combination of IU1-248 and COH34 produces synergistic effects against TNBC cells in the same setting. Mechanistically, the blockade of USP14 by IU1-248 was shown to increase DNA damage and promote error-prone non-homologous end joining (NHEJ), as evidenced by the accumulation of γH2AX and 53BP1 in the nucleus and the activation of a reporter assay. Additionally, it was demonstrated that the inhibition of NHEJ repair activity attenuates the synergistic effects of concomitant PARG and USP14 inhibition. IU1-248 promotes NHEJ repair through the downregulation of the expression of c-Myc. Conclusion USP14 inhibition may be a plausible strategy for expanding the utility of PARG inhibitors in TNBC in BRCA-mutant, PARP inhibitor-resistant settings.
Collapse
Affiliation(s)
- Pisong Li
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Xiaoyu Zhu
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Hui Qu
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Zhongbin Han
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Xingyu Yao
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Yuan Wei
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Baijun Li
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Hongshen Chen
- Department of Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| |
Collapse
|
45
|
Bertlin JAC, Pauzaite T, Liang Q, Wit N, Williamson JC, Sia JJ, Matheson NJ, Ortmann BM, Mitchell TJ, Speak AO, Zhang Q, Nathan JA. VHL synthetic lethality screens uncover CBF-β as a negative regulator of STING. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610968. [PMID: 39282259 PMCID: PMC11398426 DOI: 10.1101/2024.09.03.610968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) represents the most common form of kidney cancer and is typified by biallelic inactivation of the von Hippel-Lindau (VHL) tumour suppressor gene. Here, we undertake genome-wide CRISPR/Cas9 screening to reveal synthetic lethal interactors of VHL, and uncover that loss of Core Binding Factor β (CBF-β) causes cell death in VHL-null ccRCC cell lines and impairs tumour establishment and growth in vivo. This synthetic relationship is independent of the elevated activity of hypoxia inducible factors (HIFs) in VHL-null cells, but does involve the RUNX transcription factors that are known binding partners of CBF-β. Mechanistically, CBF-β loss leads to upregulation of type I interferon signalling, and we uncover a direct inhibitory role for CBF-β at the STING locus controlling Interferon Stimulated Gene expression. Targeting CBF-β in kidney cancer both selectively induces tumour cell lethality and promotes activation of type I interferon signalling.
Collapse
Affiliation(s)
- James A C Bertlin
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Tekle Pauzaite
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Qian Liang
- Simmons Comprehensive Cancer Center, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Niek Wit
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - James C Williamson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Jia Jhing Sia
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Brian M Ortmann
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Thomas J Mitchell
- Early Cancer Institute and Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Anneliese O Speak
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Qing Zhang
- Simmons Comprehensive Cancer Center, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| |
Collapse
|
46
|
Liu Y, Chen X, Lu H, Wu X, Liu X, Xu F, Ye D, Ding B, Lu X, Qiu L, Zhu J, Wang Y, Huang X, Shen Z, Zhu T, Shen Y, Zhou Y. Is the Homologous Recombination Repair Mutation Defined by a 15-Gene Panel Associated with the Prognosis of Epithelial Ovarian Cancer? Mol Diagn Ther 2024; 28:621-632. [PMID: 38967864 DOI: 10.1007/s40291-024-00726-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND There is no consensus regarding the specific genes included in the homologous recombination repair (HRR) gene panel for identifying the HRR deficiency (HRD) status and predicting the prognosis of epithelial ovarian cancer (EOC) patients. OBJECTIVE We aimed to explore a 15-gene panel involving the HRR pathway as a predictive prognostic indicator in Chinese patients newly diagnosed with EOC. PATIENTS AND METHODS We reviewed the previously published reports about different HRR gene panels and prespecified the 15-gene panel. The genetic testing results in a 15-gene panel from 308 EOC patients diagnosed between 2014 and 2022 from six centers were collected. The association of clinicopathologic characteristics, the use of poly (adenosine diphosphate-ribose) polymerase inhibitors (PARPis) and progression-free survival (PFS) with 15-gene panel HRR mutations (HRRm) status was assessed. RESULTS 43.2% (133/308) of patients were determined to carry 144 deleterious HRRm, among which 68.1% (98/144) were germline mutations and 32.8% (101/308) were BRCA1/2 gene lethal mutations. The hazard ratio (HR) (95% confidence interval, CI) for PFS (HRRm v HRR wild type, HRRwt) using the 15-gene panel HRRm was 0.42 (0.28-0.64) at all stages and 0.42 (0.27-0.65) at stages IIIC-IV. However, a prognostic difference was observed only between the BRCA mutation group and the HRRwt group, not between the non-BRCA HRRm group and the HRRwt group. For the subgroups of patients not using PARPis, the HR (95% CI) was 0.41 (0.24-0.68) at stages IIIC-IV. CONCLUSIONS This study provides evidence that 15-gene panel HRRm can predict the prognosis of EOC, of these only the BRCA1/2 mutations, not non-BRCA HRRm, contribute to prognosis prediction. Among patients without PARPis, the HRRm group presented a better PFS. This is the first study of this kind in the Chinese population.
Collapse
Affiliation(s)
- Yi Liu
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xiaojun Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Huaiwu Lu
- Department of Gynecological Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Xin Wu
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200082, China
| | - Xuehan Liu
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Fei Xu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Dongdong Ye
- Department of Gynecological Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Bo Ding
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoyan Lu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Ling Qiu
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200082, China
| | - Jing Zhu
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yingying Wang
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xinya Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Zhen Shen
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Tao Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
47
|
Aubuchon LN, Verma P. Endogenous base damage as a driver of genomic instability in homologous recombination-deficient cancers. DNA Repair (Amst) 2024; 141:103736. [PMID: 39096699 DOI: 10.1016/j.dnarep.2024.103736] [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: 03/31/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024]
Abstract
Homologous recombination (HR) is a high-fidelity DNA double-strand break (DSB) repair pathway. Both familial and somatic loss of function mutation(s) in various HR genes predispose to a variety of cancer types, underscoring the importance of error-free repair of DSBs in human physiology. While environmental sources of DSBs have been known, more recent studies have begun to uncover the role of endogenous base damage in leading to these breaks. Base damage repair intermediates often consist of single-strand breaks, which if left unrepaired, can lead to DSBs as the replication fork encounters these lesions. This review summarizes various sources of endogenous base damage and how these lesions are repaired. We highlight how conversion of base repair intermediates, particularly those with 5'or 3' blocked ends, to DSBs can be a predominant source of genomic instability in HR-deficient cancers. We also discuss how endogenous base damage and ensuing DSBs can be exploited to enhance the efficacy of Poly (ADP-ribose) polymerase inhibitors (PARPi), that are widely used in the clinics for the regimen of HR-deficient cancers.
Collapse
Affiliation(s)
- Lindsey N Aubuchon
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Priyanka Verma
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Cancer Biology Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
48
|
DE Morais GP, Machado CB, Dias Nogueira BM, DE Pinho Pessoa FMC, DE Sousa Oliveira D, Ribeiro RM, DA Silva JBS, Seabra AD, Mello Júnior FAR, Burbano RR, Khayat AS, DE Moraes Filho MO, DE Moraes MEA, Moreira-Nunes CA. Association of PARP1 Expression Levels and Clinical Parameters in Different Leukemic Subtypes With BCR::ABL1 p190+ Translocation. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:592-598. [PMID: 39238631 PMCID: PMC11372699 DOI: 10.21873/cdp.10368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/21/2024] [Accepted: 07/12/2024] [Indexed: 09/07/2024]
Abstract
Background/Aim Although the reciprocal translocation t(9;22)(q34;q11) is a hallmark of chronic myeloid leukemia (CML), it is also present in acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). Depending on the gene's breakpoint, it is possible to obtain three isoforms, among which p190 stands out for the poor prognosis it induces whenever it appears. Due to the genomic instability induced by BCR::ABL1, it is proposed to expand the applicability of poly-ADP-ribose polymerase-1 (PARP1) and its inhibitors in hematological neoplasms. Materials and Methods We measured the expression levels of PARP1 by quantitative real-time PCR (qPCR) using TaqMan®, correlating its expression with BCR::ABL1 p190+, to evaluate its influence in the clinic of adult patients. Results We found that PARP1 is expressed differently in ALL, AML and CML and that p190 transcripts do not follow a linear pattern in these populations. We also found that PARP1 expression is not correlated with age, white blood cell and the amount of p190 transcripts. Conclusion Despite the lack of statistical correlation between the variables analyzed, the role of PARP1 in BCR::ABL1 leukemia cannot be ruled out, given the instability profile promoted by this translocation. Finally, further studies involving a larger sample of patients are needed, as well as investigations into other molecular pathways that may impact on the pathogenesis of different BCR::ABL1 leukemic subtypes.
Collapse
Affiliation(s)
| | - Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Beatriz Maria Dias Nogueira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Flávia Melo Cunha DE Pinho Pessoa
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | | | | | | - Rommel Rodriguez Burbano
- Molecular Biology Laboratory, Ophir Loyola Hospital, Belem, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belem, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belem, Brazil
| | - Manoel Odorico DE Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Caroline Aquino Moreira-Nunes
- Unichristus University Center, Faculty of Biomedicine, Fortaleza, Brazil
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belem, Brazil
- Clementino Fraga Group, Central Unity, Molecular Biology Laboratory, Fortaleza, Brazil
| |
Collapse
|
49
|
Wu H, Han BW, Liu T, Zhang M, Wu Y, Nie J. Epstein-Barr virus deubiquitinating enzyme BPLF1 is involved in EBV carcinogenesis by affecting cellular genomic stability. Neoplasia 2024; 55:101012. [PMID: 38875930 PMCID: PMC11225014 DOI: 10.1016/j.neo.2024.101012] [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: 12/22/2023] [Revised: 05/20/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024]
Abstract
Increased mutational burden and EBV load have been revealed from normal tissues to Epstein-Barr virus (EBV)-associated gastric carcinomas (EBVaGCs). BPLF1, encoded by EBV, is a lytic cycle protein with deubiquitinating activity has been found to participate in disrupting repair of DNA damage. We first confirmed that BPLF1 gene in gastric cancer (GC) significantly increased the DNA double strand breaks (DSBs). Ubiquitination mass spectrometry identified histones as BPLF1 interactors and potential substrates, and co-immunoprecipitation and in vitro experiments verified that BPLF1 regulates H2Bub by targeting Rad6. Over-expressing Rad6 restored H2Bub but partially reduced γ-H2AX, suggesting that other downstream DNA repair processes were affected. mRNA expression of BRCA2 were significantly down-regulated by next-generation sequencing after over-expression of BPLF1, and over-expression of p65 facilitated the repair of DSBs. We demonstrated BPLF1 may lead to the accumulation of DSBs by two pathways, reducing H2B ubiquitination (H2Bub) and blocking homologous recombination which may provide new ideas for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Hantao Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China; Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Bo-Wei Han
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Min Zhang
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yingsong Wu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Jing Nie
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
50
|
Dibitetto D, Widmer CA, Rottenberg S. PARPi, BRCA, and gaps: controversies and future research. Trends Cancer 2024; 10:857-869. [PMID: 39004561 DOI: 10.1016/j.trecan.2024.06.008] [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: 04/30/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
In recent years, various poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have been approved for the treatment of several cancers to target the vulnerability of homologous recombination (HR) deficiency (e.g., due to BRCA1/2 dysfunction). In this review we analyze the ongoing debates and recent breakthroughs in the use of PARPis for BRCA1/2-deficient cancers, juxtaposing the 'double-strand break (DSB)' and 'single-stranded DNA (ssDNA) gap' models of synthetic lethality induced by PARPis. We spotlight the complexity of this interaction, highlighting emerging research on the role of DNA polymerase theta (POLθ) and ssDNA gaps in shaping therapy responses. We scrutinize the clinical ramifications of these findings, especially concerning PARPi efficacy and resistance mechanisms, underscoring the heterogeneity of BRCA-mutated tumors and the urgent need for advanced research to bridge the gap between laboratory models and patient outcomes.
Collapse
Affiliation(s)
- Diego Dibitetto
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Bern Center for Precision Medicine and Cancer Therapy Research Cluster, Department for Biomedical Research, University of Bern, 3012 Bern, Switzerland; Molecular Oncology and DNA Damage Response Laboratory, Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy.
| | - Carmen A Widmer
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Bern Center for Precision Medicine and Cancer Therapy Research Cluster, Department for Biomedical Research, University of Bern, 3012 Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Bern Center for Precision Medicine and Cancer Therapy Research Cluster, Department for Biomedical Research, University of Bern, 3012 Bern, Switzerland; Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands.
| |
Collapse
|