1
|
Wen M, Yu A, Park Y, Calarese D, Gerber HP, Yin G. Homogeneous antibody-drug conjugates with dual payloads: potential, methods and considerations. MAbs 2025; 17:2498162. [PMID: 40322862 PMCID: PMC12054377 DOI: 10.1080/19420862.2025.2498162] [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: 02/07/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
The development of site-specific dual-payload antibody-drug conjugates (ADCs) represents a potential advancement in targeted cancer therapy, enabling the simultaneous delivery of two distinct drugs into the same cancer cells to overcome payload resistance and enhance therapeutic efficacy. Here, we examine various methodologies for achieving site-specific dual-payload conjugation, including the use of multi-functional linkers, canonical amino acids, non-canonical amino acids, and enzyme-mediated methods, all of which facilitate precise control over payload attachment while ensuring homogeneity. We explore the implications of different conjugation techniques on drug-to-antibody ratios and the ratios of the two payloads, as well as their impact on process complexity and manufacturability. Additionally, we address the potential advantages of dual-payload ADCs compared to ADCs combined with traditional chemotherapy or single-payload ADC/ADC combinations. By evaluating these innovative methods, we aim to provide a comprehensive understanding of the current landscape in dual-payload ADC development and outline emerging directions necessary for further advancement of this promising therapeutic strategy.
Collapse
Affiliation(s)
- Miao Wen
- Sutro Biopharma Inc, South San Francisco, CA, USA
| | - Abigail Yu
- Sutro Biopharma Inc, South San Francisco, CA, USA
| | - Young Park
- Sutro Biopharma Inc, South San Francisco, CA, USA
| | | | | | - Gang Yin
- Sutro Biopharma Inc, South San Francisco, CA, USA
| |
Collapse
|
2
|
Yao R, Zeng Y, Zhang Y, Cao X, Mao J, Li W, Xu K, Liu L. Identification of a new micropeptide altKLF4 derived from KLF4 that influences myeloma chemotherapeutic sensitivity. Cell Signal 2025; 131:111767. [PMID: 40147548 DOI: 10.1016/j.cellsig.2025.111767] [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: 11/04/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Multiple myeloma (MM) is a common yet incurable hematological malignancy characterized by bone marrow infiltration. A major clinical challenge is the resistance to chemotherapy, highlighting the urgent need to better understand the molecular mechanisms underlying chemotherapeutic resistance to available drugs. Recent studies have emphasized the role of micropeptides in solid tumors and leukemia, but their functions in MM remain unclear. In this study, we identified a novel micropeptide, altKLF4, derived from the transcription factor KLF4, which is highly expressed in newly diagnosed myeloma patient samples. We found that ectopic expression of altKLF4 interfered with chemotherapy sensitivity induced by proteasome inhibitors in myeloma cells. Additionally, confocal microscopy and transcriptome sequencing revealed that altKLF4 co-localizes with the mitochondrial inner marker TOMM20 and participates in mitochondria-related biological processes, suggesting that altKLF4 partially localizes to the mitochondria. Mitochondria may also play a role in regulating ferroptosis. Our results further demonstrated that altKLF4 inhibited drug sensitivity and ferroptosis induced by the GPX4 inhibitor RSL3 in multiple myeloma cells through a direct interaction with GPX4. In vivo experiments showed that RSL3 significantly suppressed primary myeloma growth, which could be rescued by the micropeptide altKLF4. Taken together, our study identifies altKLF4 as a novel micropeptide that serves as a potential biomarker for chemotherapeutic resistance in multiple myeloma, offering insights for diagnosis and management of drug-resistant MM.
Collapse
Affiliation(s)
- Ruosi Yao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yindi Zeng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yaxin Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Cao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiwei Mao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenjing Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Linlin Liu
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
3
|
Wang X, Zhou J, Xu B. Engaging an engineered PARP-2 catalytic domain mutant to solve the complex structures harboring approved drugs for structure analyses. Bioorg Chem 2025; 160:108471. [PMID: 40228437 DOI: 10.1016/j.bioorg.2025.108471] [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: 02/20/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
The PARP-1/2 inhibitors have been approved for the treatment of cancers by modulating the enzymatic activity and/or the trapping ability for damaged DNA of PARP-1 and/or PARP-2, and the selective PARP-1 inhibitors are now attracting considerable attention with an aim to search for drug candidates with an improved safety. Exploring the structural basis of the selectivity and trapping capability of known PARP-1/2 inhibitors would be beneficial for the discovery of the improved inhibitors. Herein, a mutated PARP-2 catalytic domain, designated as catPARP-2SE, was engineered. It could be expressed in an elevated level and had capability to crystalize at 25 °C, which greatly facilitated obtaining PARP-2 crystals. Consequently, the complex structures of Fluzoparib, Pamiparib, Rucaparib, and Niraparib within PARP-2 were achieved. Taking advantage of these complexed structures, the detailed and quantitative analyses of protein-ligand and intra-protein interactions (αB-αF, αJ-αB, αJ-αF, ASL-αD and ASL-αF interfaces) were conducted with quantum chemistry methods (GFN2-xTB and IGMH). It suggested that the residues adjacent to Asp766 in the HD and ASL domains and the αJ-αF and ASL-αD interfaces were closely related to the selectivity and trapping mechanism. These results would provide some insights for the design and development of novel PARP-1/2 inhibitors with improved pharmacodynamic properties.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Zhou
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Bailing Xu
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
4
|
Zhao J, Tang B, Shen P, Zeng H, Wei Q. Empowering PARP inhibition through rational combination: Mechanisms of PARP inhibitors and combinations with a focus on the treatment of metastatic castration-resistant prostate cancer. Crit Rev Oncol Hematol 2025; 210:104698. [PMID: 40089046 DOI: 10.1016/j.critrevonc.2025.104698] [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: 12/21/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have revolutionized the treatment of many cancers. Metastatic castration-resistant prostate cancer (mCRPC) is an area where PARP inhibitors are intensively studied; the efficacy with PARP inhibitor monotherapy in patients with homologous recombination repair mutations following novel hormonal therapy have prompted the investigation of combination therapy, with adding an androgen receptor pathway inhibitor (ARPI) being one focus of research. Data on PARP inhibitor monotherapy and combination therapy for mCRPC are accumulating, and it is important to navigate through the complex data to inform treatment decision. Here we review the mechanisms of action of PARP inhibitors, their pharmacological properties, the synergistic activity of PARP inhibitors plus other drug classes, and the clinical evidence on monotherapy and combination therapy in patients with mCRPC. We propose key considerations in the selection of agents and treatment sequence for mCRPC, such as efficacy, toxicity profiles, biomarkers, and interactions with concomitant medications.
Collapse
Affiliation(s)
- Jinge Zhao
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Tang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Dennler O, Ryan CJ. Evaluating sequence and structural similarity metrics for predicting shared paralog functions. NAR Genom Bioinform 2025; 7:lqaf051. [PMID: 40290317 PMCID: PMC12034104 DOI: 10.1093/nargab/lqaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/07/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Gene duplication is the primary source of new genes, resulting in most genes having identifiable paralogs. Over time, paralog pairs may diverge in some respects but many retain the ability to perform the same functional role. Protein sequence identity is often used as a proxy for functional similarity and can predict shared functions between paralogs as revealed by synthetic lethal experiments. However, the advent of alternative protein representations, including embeddings from protein language models (PLMs) and predicted structures from AlphaFold, raises the possibility that alternative similarity metrics could better capture functional similarity between paralogs. Here, using two species (budding yeast and human) and two different definitions of shared functionality (shared protein-protein interactions and synthetic lethality), we evaluated a variety of alternative similarity metrics. For some tasks, predicted structural similarity or PLM similarity outperform sequence identity, but more importantly these similarity metrics are not redundant with sequence identity, i.e. combining them with sequence identity leads to improved predictions of shared functionality. By adding contextual features, representing similarity to homologous proteins within and across species, we can significantly enhance our predictions of shared paralog functionality. Overall, our results suggest that alternative similarity metrics capture complementary aspects of functional similarity beyond sequence identity alone.
Collapse
Affiliation(s)
- Olivier Dennler
- School of Medicine, University College Dublin, Dublin 4, D04 V1W8, Ireland
- School of Computer Science, University College Dublin, Dublin 4, D04 V1W8, Ireland
- Conway Institute, University College Dublin, Dublin 4, D04 V1W8, Ireland
| | - Colm J Ryan
- School of Medicine, University College Dublin, Dublin 4, D04 V1W8, Ireland
- School of Computer Science, University College Dublin, Dublin 4, D04 V1W8, Ireland
- Conway Institute, University College Dublin, Dublin 4, D04 V1W8, Ireland
| |
Collapse
|
6
|
Abida W, Beltran H, Raychaudhuri R. State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2025; 45:e473636. [PMID: 40112242 DOI: 10.1200/edbk-25-473636] [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: 03/22/2025]
Abstract
Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (177Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruben Raychaudhuri
- University of Washington and the Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
7
|
Cohen Z, Petrenko E, Barisaac A, Abu-Zhayia E, Yanovich-Ben-Uriel C, Ayoub N, Aran D. SLAYER: a computational framework for identifying synthetic lethal interactions through integrated analysis of cancer dependencies. NAR Genom Bioinform 2025; 7:lqaf052. [PMID: 40276038 PMCID: PMC12019633 DOI: 10.1093/nargab/lqaf052] [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: 01/09/2025] [Revised: 03/17/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Synthetic lethality represents a promising therapeutic approach in precision oncology, yet systematic identification of clinically relevant synthetic lethal interactions remains challenging. Here, we present SLAYER (Synthetic Lethality AnalYsis for Enhanced taRgeted therapy), a computational framework that integrates cancer genomic data and genome-wide CRISPR knockout screens to identify potential synthetic lethal interactions. SLAYER employs parallel analytical approaches examining both direct mutation-dependency associations and pathway-mediated relationships across 1080 cancer cell lines. Our integrative method identified 682 putative interactions, which were refined to 148 high-confidence candidates through stringent filtering for effect size, druggability, and clinical prevalence. Systematic validation against protein interaction databases revealed an ∼14-fold enrichment of known associations among SLAYER predictions compared with random gene pairs. Through pathway-level analysis, we identified inhibition of the aryl hydrocarbon receptor (AhR) as potentially synthetically lethal with RB1 mutations in bladder cancer. Experimental studies demonstrated selective sensitivity to AhR inhibition in RB1-mutant versus wild-type bladder cancer cells, which probably operates through indirect pathway-mediated mechanisms rather than direct genetic interaction. In summary, by integrating mutation profiles, gene dependencies, and pathway relationships, our approach provides a resource for investigating genetic vulnerabilities across cancer types.
Collapse
Affiliation(s)
- Ziv Cohen
- The Taub Faculty of Computer Science, Technion—Israel Institute of Technology, Haifa 3200003, Israel
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| | - Ekaterina Petrenko
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| | - Alma Sophia Barisaac
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| | - Enas R Abu-Zhayia
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| | | | - Nabieh Ayoub
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| | - Dvir Aran
- The Taub Faculty of Computer Science, Technion—Israel Institute of Technology, Haifa 3200003, Israel
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
8
|
Xiong J, Deng C, Fu Y, Tang J, Xie J, Chen Y. Prognostic and Potential Therapeutic Roles of PRKDC Expression in Lung Cancer. Mol Biotechnol 2025; 67:2455-2466. [PMID: 39044064 DOI: 10.1007/s12033-024-01209-3] [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: 05/06/2024] [Indexed: 07/25/2024]
Abstract
PRKDC is a key factor involved in the ligation step of the non-homologous end joining pathway. Its dysfunction has proven to be a biomarker for radiosensitivity of cancer cells. However, the prognostic value of PRKDC and its underlying mechanisms have not been clarified yet. In this study, we found that PRKDC overexpressed in lung adenocarcinoma (LUAD) and is significantly related to unfavorable survival, while downregulation of PRKDC is link to inflamed tumor immune signature. Our further in vitro results also showed a potent antitumor efficacy of PRKDC inhibitors alone or combined with cisplatin in human lung cancer cells. This study demonstrated that PRKDC is a potential prognostic biomarker, immunotherapy target, and promising combination candidate for chemotherapy for lung cancer, and highlighted the potential of PRKDC-targeted inhibitors for the treatment of lung cancer.
Collapse
Affiliation(s)
- Jiani Xiong
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Cuimin Deng
- Department of Pharmacy, QuanZhou Women's and Children's Hospital, Quanzhou, Fujian Province, People's Republic of China
| | - YunRong Fu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingji Tang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Jieming Xie
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
| |
Collapse
|
9
|
Li J, Zhao L, Zheng F, Zhu H, Li E, Zhou W, Yao G, Liu J, Zheng J, Pan S, Hu J, Shao F, Wu X. PARP Inhibitors Rechallenge in Patients With Recurrent Ovarian Cancer: A Multicentre Real-World Study in China. BJOG 2025; 132 Suppl 4:45-51. [PMID: 40275473 DOI: 10.1111/1471-0528.18181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE To evaluate the treatment pattern, outcomes, safety and identify patient populations benefiting from PARP inhibitor (PARPi) rechallenge for recurrent ovarian cancer. DESIGN A multicentre, retrospective, real-world study. SETTING Twelve hospitals in China. POPULATION Seventy patients with recurrent ovarian cancer underwent PARPi rechallenge between 1 June 2019 and 10 March 2023. METHODS Data, including demographic, clinical characteristics and treatment-related information, were retrospectively collected from electronic health records. MAIN OUTCOME MEASURES The primary outcome was progression-free survival (PFS) of PARPi rechallenge (PARPi2) as maintenance therapy. We also conducted exploratory analysis to identify factors influencing PFS and characteristics associated with favourable outcomes. RESULTS Of the 70 patients, 37.1% had BRCA1/2 mutations. PARPi2 was used as a maintenance therapy in 81.4% of patients, with a median PFS of 8.6 months (95% confidence interval [CI]: 6.0-13.5). PFS did not significantly differ by BRCA status (hazard ratio = 1.25 [95% CI: 0.60-2.60], p = 0.55). Achieving complete response (CR) to the last chemotherapy was a significant predictor for receiving PARPi2 for ≥ 6 months (vs. partial response, odds ratio = 4.25 [95% CI: 1.21-14.9], p = 0.02). Patients receiving combination therapies (33.3%) had longer median PFS than those receiving monotherapy (11.0 [95% CI: 5.2-15.3] vs. 7.7 [95% CI: 5.0-13.5] months). Overall, 2.9% of patients discontinued PARPi2 due to adverse events. CONCLUSIONS PARPi rechallenge as maintenance therapy may be feasible and tolerable. Achieving CR after the last chemotherapy is associated with longer PFS and combined therapies may improve outcomes, indicating potential to overcome PARPi resistance.
Collapse
Affiliation(s)
- Jin Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lingjun Zhao
- Department of Gynecology, Ningbo Women and Children's Hospital, Ningbo, China
| | - Fei Zheng
- Department of Gynecology, Ningbo No. 2 Hospital, Ningbo, China
| | - Hua Zhu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Enchun Li
- Department of Gynecology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Wei Zhou
- Department of Gynecology, Taizhou Hospital of Zhejiang Province, Taizhou, China
| | - Guorong Yao
- Department of Integrated Chinese and Western Medicine, Huzhou Central Hospital, Huzhou, China
| | - Jie Liu
- Department of Gynecology, Jinhua People's Hospital, Jinhua, China
| | - Jianxiao Zheng
- Department of Integrated Chinese and Western Medicine, Yueqing People's Hospital, Wenzhou, China
| | - Shan Pan
- Department of Gynecology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, China
| | - Jinghui Hu
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Shao
- Department of Gynecologic Surgical, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
10
|
Wang Y, Hess JD, Wang C, Ma L, Luo M, Jossart J, Perry JJ, Kwon D, Wang Z, Pei X, Shen C, Wang Y, Zhou M, Yin H, Horne D, Nussenzweig A, Zheng L, Shen B. Discovery and Characterization of Small Molecule Inhibitors Targeting Exonuclease 1 for Homologous Recombination-Deficient Cancer Therapy. ACS Chem Biol 2025. [PMID: 40378357 DOI: 10.1021/acschembio.5c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Human exonuclease 1 (EXO1), a member of the structure-specific nuclease family, plays a critical role in maintaining genome stability by processing DNA double-strand breaks (DSBs), nicks, and replication intermediates during DNA replication and repair. As its exonuclease activity is essential for homologous recombination (HR) and replication fork processing, EXO1 has emerged as a compelling therapeutic target, especially in cancers marked by heightened DNA damage and replication stress. Through high-throughput screening of 45,000 compounds, we identified seven distinct chemical scaffolds that demonstrated effective and selective inhibition of EXO1. Representative compounds from two of the most potent scaffolds, C200 and F684, underwent a comprehensive docking analysis and subsequent site-directed mutagenesis studies to evaluate their binding mechanisms. Biochemical assays further validated their potent and selective inhibition of the EXO1 nuclease activity. Tumor cell profiling experiments revealed that these inhibitors exploit synthetic lethality in BRCA1-deficient cells, emphasizing their specificity and therapeutic potential for targeting genetically HR-deficient (HRD) cancers driven by deleterious mutations in HR genes like BRCA1/2. Mechanistically, EXO1 inhibition suppressed DNA end resection, stimulated the accumulation of DNA double-strand breaks, and triggered S-phase PARylation, effectively disrupting DNA repair pathways that are essential for cancer cell survival. These findings establish EXO1 inhibitors as promising candidates for the treatment of HRD cancers and lay the groundwork for the further optimization and development of these compounds as targeted therapeutics.
Collapse
Affiliation(s)
- Yixing Wang
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Jessica D Hess
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Chen Wang
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Lingzi Ma
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Megan Luo
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Jennifer Jossart
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - John J Perry
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - David Kwon
- Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Zhe Wang
- Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Xinyu Pei
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Changxian Shen
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Yingying Wang
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Mian Zhou
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Holly Yin
- Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - André Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute NIH, Bethesda, Maryland 20892, United States
| | - Li Zheng
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| | - Binghui Shen
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, Duarte, California 91010, United States
| |
Collapse
|
11
|
Hattori M, Mikami Y, Sato M, Nagai H, Fujii K, Horiguchi Y, Shuzui M, Fukuda K, Miyamoto Y, Mori M, Hinata M, Kawakami M, Mitani A, Tanaka G, Kage H. Olaparib-induced interstitial lung disease: A case series analysis. Respir Investig 2025; 63:629-632. [PMID: 40381525 DOI: 10.1016/j.resinv.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Olaparib-induced interstitial lung disease (OILD) is a rare but potentially serious adverse event, and its imaging characteristics and clinical course remain unclear. Ishimoto et al. previously reported three cases of OILD, but further characterisation is needed. We present three additional cases, all presenting with fever and bilateral ground-glass opacities (GGO) and fine reticulonodular opacities on computed tomography (CT). Bronchoalveolar lavage fluid (BALF) analysis showed marked lymphocytosis (>75 %). Prednisolone (0.5-1 mg/kg/day) was effective without fibrosis. BALF lymphocytosis may aid in the diagnosis of OILD. OILD should be considered in febrile patients receiving olaparib.
Collapse
Affiliation(s)
- Mototaka Hattori
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yu Mikami
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Midori Sato
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroyuki Nagai
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Koki Fujii
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yuki Horiguchi
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masahiro Shuzui
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kensuke Fukuda
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yuichiro Miyamoto
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mayuyo Mori
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Munetoshi Hinata
- Department of Pathology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masanori Kawakami
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Akihisa Mitani
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Goh Tanaka
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
12
|
Lu X, Sauter B, Keller A, Zhanybekova S, Gillingham D. Exploring the Potential of Homologous Recombination Protein PALB2 in Synthetic Lethal Combinations. ACS Chem Biol 2025; 20:1099-1106. [PMID: 40300769 DOI: 10.1021/acschembio.5c00111] [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: 05/01/2025]
Abstract
Cells with defective homologous recombination (HR) are highly sensitive to poly(ADP-ribose) polymerase (PARP) inhibition. Current therapeutic approaches leverage this vulnerability by using PARP inhibitors in cells with genetically compromised HR. However, if HR factors in cancer cells could be inhibited or degraded pharmacologically, it might reveal other opportunities for synergistic combinations. In this study, we developed a model system that recapitulates PARP/HR synthetic lethality by integrating a small-molecule responsive zinc-finger degron into the HR factor Partner and Localizer of BRCA2 (PALB2). We further tested a series of peptide ligands for PALB2 based on its natural binding partners, which led to the discovery of a high affinity peptide that will support future work on PALB2 and HR. Together, our findings validate PALB2 as a promising drug target and provide the tools and starting points for developing molecules with therapeutic applications.
Collapse
Affiliation(s)
- Xinyan Lu
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Basilius Sauter
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Aramis Keller
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Saule Zhanybekova
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Dennis Gillingham
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
13
|
Gohari Z, Stojanovic L, Rassool FV. Combining STING Agonists with PARP Inhibitors Mounts an NK-Dependent Defense against Therapy-Resistant Breast Cancer. Cancer Res 2025; 85:1747-1749. [PMID: 40370063 DOI: 10.1158/0008-5472.can-25-0831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 05/16/2025]
Abstract
Breast cancers with BRCA1 or BRCA2 mutations are defective in repair of DNA double-strand breaks by homologous recombination, resulting in compensatory error-prone repair that causes genomic instability. Poly(ADP-ribose) polymerase inhibitors (PARPi) are FDA-approved to treat homologous recombination-defective cancers, inducing therapy responses by synthetic lethality. PARPis increase micronuclei formation and cytosolic double-stranded DNA accumulation, activating stimulator of interferon genes (STING). Activation of STING can mediate anticancer innate immune responses by increasing T-cell infiltration into the tumor microenvironment. However, PARPi responses are not durable, and therapy resistance ensues with limited therapeutic options available for these patients. Using PARPi-sensitive and -resistant patient-derived xenografts and mouse-derived allografts, Pedretti and colleagues show in this issue of Cancer Research that the PARPi olaparib in combination with the next-generation STING agonist diABZI can overcome PARPi resistance in a manner dependent on NK cell function in the tumor microenvironment. Their study highlights a novel component of the STING-dependent innate immune response repertoire required for fighting PARPi-resistant cancer. Potent and specific next-generation STING agonists are being tested in the clinic in solid and liquid tumors, indicating a resurgence of these drugs after a long period of modest clinical activity, with a special focus on combination therapy strategies to fight therapy-resistant cancer. See related article by Pedretti et al., p. 1888.
Collapse
Affiliation(s)
- Zahra Gohari
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lora Stojanovic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Feyruz V Rassool
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| |
Collapse
|
14
|
Wang Z, Zhang Z, Liu R, Ren X, Liu S, Huang Y. Synthesis of Chiral 3-Piperidin-2-ones and 3-Piperidines via Ni-Catalyzed Reductive Coupling. Org Lett 2025. [PMID: 40358021 DOI: 10.1021/acs.orglett.5c01184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Herein, an approach to a wide range of chiral 3-substituted δ-lactams from reductive coupling of Csp2-hybridized organohalides and 3-chloro-δ-lactams was described, and the products are versatile precursors for accessing enantioenriched 3-substituted piperidines. The utility of the reaction was highlighted by the economic synthesis of the chiral precursors of Preclamol and Niraparib. Notably, the modified chiral Bilm ligands were found to be the key factor for the reactivity and enantioselectivity.
Collapse
Affiliation(s)
- Zhaoqing Wang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhuo Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Rui Liu
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaolin Ren
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Song Liu
- Department of Chongqing Key Laboratory of Environmental Materials and Remediation Technologies, College of Chemistry and Environmental Engineering, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Yuan Huang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
15
|
Abraham JE, O'Connor LO, Grybowicz L, Alba KP, Dayimu A, Demiris N, Harvey C, Drewett LM, Lucey R, Fulton A, Roberts AN, Worley JR, Chhabra MA, Qian W, Brown J, Hardy R, Vallier AL, Chan S, Cidon MEU, Sherwin E, Chakrabarti A, Sadler C, Barnes J, Persic M, Smith S, Raj S, Borley A, Braybrooke JP, Staples E, Scott LC, Palmer CA, Moody M, Churn MJ, Pilger D, Zagnoli-Vieira G, Wijnhoven PWG, Mukesh MB, Roylance RR, Schouten PC, Levitt NC, McAdam K, Armstrong AC, Copson ER, McMurtry E, Galbraith S, Tischkowitz M, Provenzano E, O'Connor MJ, Earl HM. Neoadjuvant PARP inhibitor scheduling in BRCA1 and BRCA2 related breast cancer: PARTNER, a randomized phase II/III trial. Nat Commun 2025; 16:4269. [PMID: 40360463 PMCID: PMC12075821 DOI: 10.1038/s41467-025-59151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) exploit DNA repair deficiency in germline BRCA1 and BRCA2 pathogenic variant (gBRCAm) cancers. Haematological toxicity limits chemotherapy-PARPi treatment combinations. In preclinical models we identified a schedule combining olaparib and carboplatin that avoids enhanced toxicity but maintains anti-tumour activity. We investigated this schedule in a neoadjuvant, phase II-III, randomised controlled trial for gBRCAm breast cancers (ClinicalTrials.gov ID:NCT03150576; PARTNER). The research arm included carboplatin (Area Under the Curve 5, 3-weekly); paclitaxel (80 mg/m2, weekly) day 1, plus olaparib (150 mg twice daily) day 3-14 (4 cycles), followed by anthracycline-containing chemotherapy (3 cycles); control arm gave chemotherapy alone. The primary endpoint, pathological complete response rate, showed no statistical difference between research 64.1% (25/39); control 69.8% (30/43) (p = 0.59). However, estimated survival outcomes at 36-months demonstrated improved event-free survival: research 96.4%, control 80.1% (p = 0.04); overall survival: research 100%, control 88.2% (p = 0.04) and breast cancer specific survival: research 100%, control 88.2% (p = 0.04). There were no statistical differences in relapse-free survival and distant disease-free survival, both were: research 96.4%, control 87.9% (p = 0.20). Similarly, local recurrence-free survival and time to second cancer were both: research 96.4%, control 87.8% (p = 0.20). The PARTNER trial identified a safe, tolerable schedule combining neoadjuvant chemotherapy with olaparib. This combination demonstrated schedule-dependent overall survival benefit in early-stage gBRCAm breast cancer. This result needs confirmation in larger trials.
Collapse
Affiliation(s)
- Jean E Abraham
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom.
| | | | - Louise Grybowicz
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Karen Pinilla Alba
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Alimu Dayimu
- Cambridge Clinical Trials Centre, Cancer Theme, University of Cambridge, Cambridge, United Kingdom
| | - Nikolaos Demiris
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - Caron Harvey
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Lynsey M Drewett
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Rebecca Lucey
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Alexander Fulton
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Anne N Roberts
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Joanna R Worley
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ms Anita Chhabra
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Wendi Qian
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Richard Hardy
- Cambridge Clinical Trials Centre, Cancer Theme, University of Cambridge, Cambridge, United Kingdom
| | - Anne-Laure Vallier
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Steve Chan
- The City Hospital, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Maria Esther Una Cidon
- Royal Bournemouth General Hospital, University Hospitals Dorset NHS Foundation Trust, Bournemouth, United Kingdom
| | - Elizabeth Sherwin
- Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Ipswich, United Kingdom
| | | | - Claire Sadler
- Apconix Ltd, Alderley Edge, Cheshire, United Kingdom
| | | | - Mojca Persic
- University Hospital of Derby and Burton, Derby, United Kingdom
| | - Sarah Smith
- Bedford Hospital, Bedfordshire Hospitals NHS Foundation Trust, Bedford, United Kingdom
| | - Sanjay Raj
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Hampshire Hospitals NHS Foundation Trust, Hampshire, United Kingdom
| | | | - Jeremy P Braybrooke
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Emma Staples
- Queens Hospital, Barking, Havering and Redbridge University Hospitals NHS Trust, Romford, United Kingdom
| | - Lucy C Scott
- Beatson West Of Scotland Cancer Centre, Glasgow, Scotland, United Kingdom
| | - Cheryl A Palmer
- Hinchingbrooke Hospital, North West Anglia NHS Foundation Trust, Huntingdon, United Kingdom
| | - Margaret Moody
- Macmillan Unit, West Suffolk Hospital NHS Foundation Trust, Bury St Edmunds, United Kingdom
| | - Mark J Churn
- Worcestershire Acute Hospitals NHS Trust, Worcester, United Kingdom
| | | | | | | | - Mukesh B Mukesh
- Colchester General Hospital, East Suffolk & North Essex NHS Trust, Colchester, United Kingdom
| | - Rebecca R Roylance
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Philip C Schouten
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Nicola C Levitt
- Oxford University Hospital NHS Foundation Trust, Oxford, United Kingdom
| | - Karen McAdam
- Peterborough City Hospital, North West Anglia NHS Foundation Trust, Peterborough, United Kingdom
| | | | - Ellen R Copson
- Cancer Sciences Academic Unit, University of Southampton, Southampton, United Kingdom
| | | | | | - Marc Tischkowitz
- Department of Genomic Medicine, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Elena Provenzano
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Helena M Earl
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Saito S, Kato S, Arai U, En A, Tsunezumi J, Mizushima T, Tateishi K, Adachi N. HR eye & MMR eye: one-day assessment of DNA repair-defective tumors eligible for targeted therapy. Nat Commun 2025; 16:4239. [PMID: 40355434 PMCID: PMC12069580 DOI: 10.1038/s41467-025-59462-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Homologous recombination (HR) and mismatch repair (MMR) act as guardians of the human genome, and defects in HR or MMR are causative in at least a quarter of all malignant tumors. Although these DNA repair-deficient tumors are eligible for effective targeted therapies, fully reliable diagnostic strategies based on functional assay have yet to be established, potentially limiting safe and proper application of the molecular targeted drugs. Here we show that transient transfection of artificial DNA substrates enables ultrarapid detection of HR and MMR. This finding led us to develop a diagnostic strategy that can determine the cellular HR/MMR status within one day without the need for control cells or tissues. Notably, the accuracy of this method allowed the discovery of a pathogenic RAD51D mutation, which was missed by existing companion diagnostic tests. Our methods, termed HR eye and MMR eye, are applicable to frozen tumor tissues and roughly predict the response to therapy. Overall, the findings presented here could pave the way for accurately assessing malignant tumors with functional defects in HR or MMR, a step forward in accelerating precision medicine.
Collapse
Affiliation(s)
- Shinta Saito
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Shingo Kato
- Department of Clinical Cancer Genomics, Yokohama City University Hospital, Yokohama, 236-0004, Japan
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Usaki Arai
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Atsuki En
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Jun Tsunezumi
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Taichi Mizushima
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Kensuke Tateishi
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Noritaka Adachi
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan.
| |
Collapse
|
17
|
Bourgeois NM, Wei L, Kaushansky A, Aitchison JD. Exploiting Host Kinases to Combat Dengue Virus Infection and Disease. Antiviral Res 2025:106172. [PMID: 40348023 DOI: 10.1016/j.antiviral.2025.106172] [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: 12/31/2024] [Revised: 04/03/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025]
Abstract
The burden of dengue on human health has dramatically increased in recent years, underscoring the urgent need for effective therapeutic interventions. Despite decades of research since the discovery of the dengue virus, no specific antiviral treatments are available and strategies to reliably prevent severe disease remain limited. Direct-acting antivirals against dengue are under active investigation but have shown limited efficacy to date. An underappreciated Achille's heal of the virus is its dependence on host factors for infection and pathogenesis, each of which presents a potential avenue for therapeutic intervention. We and others have demonstrated that dengue virus relies on multiple host kinases, some of which are already targeted by clinically approved inhibitors. These offer drug repurposing opportunities for host-directed dengue treatment. Here, we summarize findings on the role of kinases in dengue infection and disease and highlight potential kinase targets for the development of innovative host-directed therapeutics.
Collapse
Affiliation(s)
- Natasha M Bourgeois
- Department of Global Health, University of Washington, Seattle WA 98195, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA
| | - Ling Wei
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA
| | - Alexis Kaushansky
- Department of Global Health, University of Washington, Seattle WA 98195, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA.
| | - John D Aitchison
- Department of Global Health, University of Washington, Seattle WA 98195, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA.
| |
Collapse
|
18
|
Doruk Y, Diolaiti ME, Ashworth A, Talele TT. Discovery of a Novel [6-6-5-5-6] Pentacyclic Tetrahydrocyclopentaphthalazinone as a Promising PARP Inhibitor Scaffold. ACS Med Chem Lett 2025; 16:776-783. [PMID: 40365384 PMCID: PMC12067119 DOI: 10.1021/acsmedchemlett.4c00603] [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: 12/16/2024] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Inhibitors of poly(ADP-ribose) polymerases (PARPs) have revolutionized the treatment of cancers with DNA repair deficiencies. Here we describe the structure-based discovery and synthesis of 6-6-5-5-6-fused pentacyclic scaffolds 5 and cis-(±)-6 as a novel class of PARP1 inhibitors. Chiral supercritical fluid chromatographic separation of cis-(±)-6 afforded inactive ent-6_P1 and active ent-6_P2. Compound 5 (P-gp ER = 0.9) and ent-6_P2 (P-gp ER = 1.1) demonstrated good Caco-2 permeability and are not actively effluxed by ABC transporters. In vitro analysis in HEK293T cells found that 5, cis-(±)-6, and ent-6_P2 showed near complete inhibition of PARP1 activity at 10 μM. Furthermore, compounds 5, cis-(±)-6, and ent-6_P2 displayed selective cytotoxic activity in BRCA mutant cancer cells but not isogenic BRCA-proficient cells. Taken together, 5 and ent-6_P2 define a novel class of lead PARP inhibitors for further development.
Collapse
Affiliation(s)
- Yagmur
U. Doruk
- UCSF
Helen Diller Family Comprehensive Cancer Center, University of California, San
Francisco, California 94158, United States
| | - Morgan E. Diolaiti
- UCSF
Helen Diller Family Comprehensive Cancer Center, University of California, San
Francisco, California 94158, United States
| | - Alan Ashworth
- UCSF
Helen Diller Family Comprehensive Cancer Center, University of California, San
Francisco, California 94158, United States
- Department
of Medicine, University of California, San Francisco, California 94158, United States
| | - Tanaji T. Talele
- Department
of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York 11439, United States
| |
Collapse
|
19
|
Liu J, Jiao X, Mu W, Li H, Xia Y, Wu Y, Zhu L, Zhong Q, Pan W, Liu X, Xiang M, Cheng J, Lin H, Zhao X, Ding Z, Hu G, Mills GB, Ma D, Gao Q, Fang Y. Mitigating T cell DNA damage during PARP inhibitor treatment enhances antitumor efficacy. Sci Transl Med 2025; 17:eadr5861. [PMID: 40333991 DOI: 10.1126/scitranslmed.adr5861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/12/2025] [Indexed: 05/09/2025]
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARPis) are a class of agents targeting DNA damage repair that have become standard therapy for epithelial ovarian cancer (EOC) and multiple other solid tumors. In addition to targeting DNA damage repair, PARPis actively modulate antitumor immune responses, with efficacy being partially dependent on T cell activity. Here, we found that patient T cells sustain DNA damage during PARPi treatment, which reduces treatment efficacy. Leveraging paired pre- and posttreatment tumor samples from a clinical trial of patients with EOC treated with neoadjuvant niraparib as monotherapy, we showed that the PARPi caused DNA damage, slowed proliferation, and increased apoptosis in T cells, which we validated both in vitro and in mouse models. A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) knockout screen in primary human T cells identified PARP1 as the principal mediator of PARPi-induced T cell death. T cell-specific deletion of PARP1 or mutating Parp1 at its binding sites in transgenic mice led to reduced T cell DNA damage during PARPi treatment, resulting in improved efficacy of PARPis, alone or in combination with immune checkpoint inhibition. We then engineered PARPi-tolerant CAR T cells using cytosine base editing, which decreased PARPi-induced PARP1 trapping and led to reduced PARPi-induced DNA damage, resulting in superior antitumor efficacy in xenograft models compared with parental CAR T cells. This study highlights the relevance of PARPi-induced DNA damage to T cells and suggests opportunities to improve the efficacy of PARPis as monotherapy or in combination with immunotherapy.
Collapse
Affiliation(s)
- Jiahao Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaofei Jiao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huayi Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xia
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yijie Wu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qing Zhong
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen Pan
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xingzhe Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minghua Xiang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiali Cheng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haolong Lin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuejiao Zhao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Division of Oncological Sciences, Oregon Heath and Sciences University, Portland, OR 97201, USA
- Knight Cancer Institute, Portland, OR 97201, USA
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, 250101, China
| | - Guang Hu
- Nanjing IASO Biotherapeutics Ltd., Nanjing, 210043, China
| | - Gordon B Mills
- Division of Oncological Sciences, Oregon Heath and Sciences University, Portland, OR 97201, USA
- Knight Cancer Institute, Portland, OR 97201, USA
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
20
|
Biju T, Venkatesh C, Honnasiddappa DB, Sajjan M, Mahadeva NK, Dinesh BGH, Kumar BS, Ganjipete S, Ramar M, Kunjiappan S, Theivendren P, Madasamy S, Chidambaram K, Ammunje DN, Pavadai P. ATAD2 bromodomain in cancer therapy: current status and future perspectives. Int J Biol Macromol 2025; 311:143948. [PMID: 40334884 DOI: 10.1016/j.ijbiomac.2025.143948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
ATPase family AAA domain-containing protein 2, or ATAD2, is a novel carcinogen, essential for cancer development, chromatin remodeling, and transcriptional control. It contains a bromodomain, which binds to acetylated histones to control gene expression. It also impacts pathways that regulate the cell cycle, DNA replication, and hormone signalling. ATAD2 is overexpressed in several malignancies, including colorectal, lung, ovarian, and breast cancers, and cancer metastasis. Investigations into the function of ATAD2 in oncogenesis and its interactions may offer fresh approaches to creating cancer treatment plans. Although preclinical research is very encouraging, many unresolved aspects regarding therapeutic development remain, including toxicity being explored concurrently. Investigations into the function of ATAD2 in oncogenesis may offer fresh approaches to developing chemotherapy strategies. Most of ATAD2's molecular mechanisms behind carcinogenesis and functions are discussed here. Additionally, we included progress, including potential monoclonal antibodies, RNA-based therapies, and small chemical inhibitors, in the review. Therefore, we guarantee this study will provide researchers with new opportunities and directions for cancer therapeutics.
Collapse
Affiliation(s)
- Tincy Biju
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Chidananda Venkatesh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Darshana Ballagere Honnasiddappa
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Mallikarjun Sajjan
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Nayan Kumar Mahadeva
- Department of Pharmacognosy, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Basavana Gowda Hosur Dinesh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Bandral Sunil Kumar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Srinivas Ganjipete
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Mohankumar Ramar
- Department of Pharmaceutical Sciences, UConn School of Pharmacy, Storrs CT-06269, USA
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry & Analysis, School of Pharmaceutical Sciences, Vels Institute of Science, Technology & Advanced Studies, Pallavaram, Chennai, Tamil Nadu 600117, India
| | - Sundar Madasamy
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Damodar Nayak Ammunje
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India.
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India.
| |
Collapse
|
21
|
Simoneau A, Pratt CB, Wu HJ, Rajeswaran SS, Comer CG, Sudsakorn S, Zhang W, Liu S, Meier SR, Choi AH, Khendu T, Stowe H, Shen B, Whittington DA, Chen Y, Yu Y, Mallender WD, Feng T, Andersen JN, Maxwell JP, Throner S. Characterization of TNG348: A Selective, Allosteric USP1 Inhibitor That Synergizes with PARP Inhibitors in Tumors with Homologous Recombination Deficiency. Mol Cancer Ther 2025; 24:678-691. [PMID: 39886906 PMCID: PMC12046316 DOI: 10.1158/1535-7163.mct-24-0515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/01/2025]
Abstract
Inhibition of the deubiquitinating enzyme USP1 can induce synthetic lethality in tumors characterized by homologous recombination deficiency (HRD) and represents a novel therapeutic strategy for the treatment of BRCA1/2-mutant cancers, potentially including patients whose tumors have primary or acquired resistance to PARP inhibitors (PARPi). In this study, we present a comprehensive characterization of TNG348, an allosteric, selective, and reversible inhibitor of USP1. TNG348 induces dose-dependent accumulation of ubiquitinated protein substrates both in vitro and in vivo. CRISPR screens show that TNG348 exerts its antitumor effect by disrupting the translesion synthesis pathway of DNA damage tolerance through RAD18-dependent ubiquitinated PCNA. Although TNG348 and PARPi share the ability to selectively kill HRD tumor cells, CRISPR screens reveal that TNG348 and PARPi do so through discrete mechanisms. Particularly, knocking out PARP1 causes resistance to PARPi but sensitizes cells to TNG348 treatment. Consistent with these findings, combination of TNG348 with PARPi leads to synergistic antitumor effects in HRD tumors, resulting in tumor growth inhibition and regression in multiple mouse xenograft tumor models. Importantly, our data on human cancer models further show that the addition of TNG348 to PARPi treatment can overcome acquired PARPi resistance in vivo. Although the clinical development of TNG348 has been discontinued because of unexpected liver toxicity in patients (NCT06065059), the present data provide preclinical and mechanistic support for the continued exploration of USP1 as a drug target for the treatment of patients with BRCA1/2-mutant or HRD cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yi Yu
- Tango Therapeutics, Boston, Massachusetts
| | | | | | | | | | | |
Collapse
|
22
|
Khoury R, Longobardi G, Barnatan TT, Venkert D, García Alvarado A, Yona A, Green Buzhor M, Shahar S, Wang Q, Acúrcio RC, Guedes RC, Florindo HF, Zhao JJ, Satchi-Fainaro R. Radiation-guided nanoparticles enhance the efficacy of PARP inhibitors in primary and metastatic BRCA1-deficient tumors via immunotherapy. J Control Release 2025; 383:113812. [PMID: 40319918 DOI: 10.1016/j.jconrel.2025.113812] [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: 12/17/2024] [Revised: 04/06/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) have revolutionized the treatment landscape for patients suffering from BRCA1-mutated breast and ovarian cancers. However, responses are not durable. We demonstrate that treatment with PARPi, niraparib, increases programmed death-ligand 1 (PD-L1) expression in BRCA1-deficient cancer cells, contributing to immune evasion. To circumvent this, we developed P-selectin-targeted poly (lactic-co-glycolic) acid (PLGA)-poly (ethylene glycol) (PEG)-based nanoparticles (NPs) encapsulating PARP and PD-L1 inhibitors at a synergistic ratio. To further enhance tumor targeting, we leveraged radiation-induced P-selectin upregulation in BRCA1-deficient cancer cells and their associated angiogenic endothelial cells, improving NP accumulation in the primary tumors and hard-to-target metastatic sites, including brain metastasis. Using a combination of traditional 2-dimensional (2D) cell cultures, advanced 3-dimensional (3D) spheroids, tumor-on-a-chip platforms, and in vivo models, we demonstrate the enhanced accumulation and efficacy of the radiation-guided P-selectin-targeted NPs in primary and brain-metastatic BRCA1-deficient tumors.
Collapse
Affiliation(s)
- Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Giuseppe Longobardi
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tania T Barnatan
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Venkert
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - América García Alvarado
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adi Yona
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Marina Green Buzhor
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shir Shahar
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Qiwei Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Rita C Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Tel Aviv University, Center for Nanoscience and Nanotechnology, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
23
|
Kamii M, Kamata R, Saito H, Yamamoto G, Mashima C, Yamauchi T, Nakao T, Sakae Y, Yamamori-Morita T, Nakai K, Hakozaki Y, Takenaka M, Okamoto A, Ohashi A. PARP inhibitors elicit a cellular senescence mediated inflammatory response in homologous recombination proficient cancer cells. Sci Rep 2025; 15:15458. [PMID: 40316566 PMCID: PMC12048520 DOI: 10.1038/s41598-025-00336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have improved the prognosis of homologous recombination deficient (HRD) ovarian cancer (OC), while effective therapeutic strategies for HR-proficient (HRP) OC still need to be established. This study investigates senescence-mediated inflammation as a novel mechanism of action for PARP inhibitors in HRP cancers. Transcriptome analyses were performed in olaparib-treated HeLa cells as a HRP model. Interferon regulatory factor-Lucia luciferase (IRF-Luc) reporter activity was assessed. The effects of PARP inhibitors on senescence-like phenotypes were assessed in seven HRP cancer cell lines, based on morphological changes, senescence-associated β-galactosidase (SA-β-GAL) activity, cellular granularity, and senescence-associated secretory phenotype (SASP)-related gene expression. Peripheral blood mononuclear cell (PBMC) migration assays were also performed with the conditioned medium in treatment with the PARP inhibitor. Transcriptome analyses revealed numbers of inflammatory cytokine- and chemokine-related pathways were significantly upregulated in olaparib-treated HeLa cells, which were confirmed by IRF-Luc reporter assays. The PARP inhibitors induced senescent phenotypes in HRP cancer cell lines: flattened and enlarged morphology, increased SA-β-GAL activity, elevated cellular granularity, and upregulated expressions of SASP-related genes (e.g., IL1B, IL6, and CXCL10). Furthermore, in vitro migration assays revealed that PARP inhibitor-treated HRP cancer cells attracted PBMCs more abundantly, suggesting the potential for recruiting immune cells to HRP cancer cells through senescence-mediated immunological activation. Our findings suggest that PARP inhibitors recruit immune cells to HRP cancer cells, potentially activating immune responses in the tumor microenvironment, providing new insights into the clinical benefits of PARP inhibitors in immunotherapy for patients with HRP OC.
Collapse
Affiliation(s)
- Misato Kamii
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryo Kamata
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Hitoshi Saito
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Gaku Yamamoto
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Chiaki Mashima
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Toyohiro Yamauchi
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
- Department of Integrated Bioscience, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan
| | - Takehiro Nakao
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yuta Sakae
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Tomoko Yamamori-Morita
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Kazuki Nakai
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yumi Hakozaki
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Masataka Takenaka
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Akihiro Ohashi
- Division of Collaborative Research and Developments, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
- Department of Integrated Bioscience, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan.
| |
Collapse
|
24
|
Zhang T, Zhang Y, Wang X, Hu H, Lin CG, Xu Y, Zheng H. Genome-wide CRISPR activation screen identifies ARL11 as a sensitivity determinant of PARP inhibitor therapy. Cancer Gene Ther 2025; 32:521-537. [PMID: 40123001 DOI: 10.1038/s41417-025-00893-w] [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: 07/28/2024] [Revised: 02/16/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Resistance to poly-(ADP)-ribose polymerase inhibitors (PARPi) remains a significant challenge in clinical practice, leading to treatment failure in many patients. It is crucial to better understand the molecular mechanisms that underlie PARPi resistance. In this study, utilizing a genome-wide CRISPR activation screen with olaparib, we identified ARL11 as a potential modulator of PARPi treatment response in BRCA-wild-type MDA-MB-231 cells. Mechanistically, ARL11 interacts with STING to enhance innate immunity and forms positive feedback with type I interferon (IFN) induction, which induces ARL11 up-regulation and contributes to resistance to PARPi therapy. Additionally, we observed that ARL11 interacts with the RUVBL1 and RUVBL2 (RUVBL1/2) complex, the key DNA double-strand repair proteins, facilitating DNA homologous recombination (HR) repair and significantly reducing PARPi-induced DNA double-strand damages. Clinical sample analysis reveals that the expression levels of ARL11 and RUVBL1/2 are significantly elevated in breast cancer patients compared to healthy controls. Collectively, our findings suggested that ARL11 and RUVBL1/2 may be promising therapeutic targets to sensitize breast cancer cells to PARPi therapy.
Collapse
Affiliation(s)
- Tengjiang Zhang
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yuan Zhang
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xuxiang Wang
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Haitian Hu
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Christopher G Lin
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yaru Xu
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi Province, China.
| |
Collapse
|
25
|
Hao S, Liu Z, Lenz HJ, Yu J, Zhang L. Werner helicase as a therapeutic target in mismatch repair deficient colorectal cancer. DNA Repair (Amst) 2025; 149:103831. [PMID: 40203476 DOI: 10.1016/j.dnarep.2025.103831] [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: 12/06/2024] [Revised: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths in the United States. A key driver of CRC development is microsatellite instability (MSI), which is caused by DNA mismatch repair deficiency and characterized by hypermutability of short-tandem repeat sequences. A significant portion of MSI CRCs do not respond to checkpoint immunotherapy treatments, highlighting an unmet need for improved therapies. Recent studies have revealed that MSI cancer cells require Werner (WRN), a RecQ family DNA helicase, for survival. Inhibiting WRN has emerged as a promising approach for targeting MSI CRCs that are insensitive to standard therapies. Several highly potent small-molecule WRN inhibitors have been developed and exhibited striking in vitro and in vivo activities against MSI cancers. Two of these WRN inhibitors, HRO761 and VVD-133214, have recently entered clinical trials. In this review, we summarize recent studies on WRN as a synthetic lethal target in MSI CRC and the development of WRN inhibitors as a new class of anticancer agents.
Collapse
Affiliation(s)
- Suisui Hao
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Zhaojin Liu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Heinz-Josef Lenz
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Jian Yu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA.
| |
Collapse
|
26
|
Jordan MR, Mendoza-Munoz PL, Pawelczak KS, Turchi JJ. Targeting DNA damage sensors for cancer therapy. DNA Repair (Amst) 2025; 149:103841. [PMID: 40339280 DOI: 10.1016/j.dnarep.2025.103841] [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: 01/30/2025] [Revised: 04/18/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
DNA damage occurs from both endogenous and exogenous sources and DNA damaging agents are a mainstay in cancer therapeutics. DNA damage sensors (DDS) are proteins that recognize and bind to unique DNA structures that arise from direct DNA damage or replication stress and are the first step in the DNA damage response (DDR). DNA damage sensors are responsible for recruiting transducer proteins that signal downstream DNA repair pathways. As the initiating proteins, DDS are excellent candidates for anti-cancer drug targeting to limit DDR activation. Here, we review four major DDS: PARP1, RPA, Ku, and the MRN complex. We briefly describe the cellular DDS functions before analyzing the structural mechanisms of DNA damage sensing. Lastly, we examine the current state of the field towards inhibiting each DDS for anti-cancer therapeutics and broadly discuss the therapeutic potential for DDS targeting.
Collapse
Affiliation(s)
- Matthew R Jordan
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Pamela L Mendoza-Munoz
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - John J Turchi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States; NERx BioSciences, Indianapolis, IN, United States.
| |
Collapse
|
27
|
Rodríguez-Moreno JF, de Velasco G, Álvarez-Fernández C, Collado R, Fernández R, Vázquez S, Virizuela JA, Gajate P, Font A, Lainez N, Sevillano-Fernández E, Graña-Castro O, Beltrán L, Madurga R, Rodríguez-Antona C, Berraondo P, Ruiz-Llorente S, García-Donas J. Treatment Efficacy and Molecular Dynamics of Neoadjuvant Durvalumab and Olaparib in Resectable Urothelial Bladder Cancer: The NEODURVARIB Trial. Clin Cancer Res 2025; 31:1644-1656. [PMID: 40298406 PMCID: PMC12010967 DOI: 10.1158/1078-0432.ccr-24-2890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/20/2024] [Accepted: 02/21/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Neoadjuvant treatment of bladder cancer is evolving, with immunotherapy demonstrating promising activity. PARP inhibition combined with immune activation has been proposed as a synergistic strategy. We conducted a comprehensive molecular characterization of tumors treated with this combination in the neoadjuvant setting to provide crucial results for rational development. PATIENTS AND METHODS A phase II clinical trial was designed to evaluate the combination of anti-PDL1 inhibitor durvalumab and PARP inhibitor olaparib, focusing on biomarker dynamics in both pre- and post-treatment settings. A total of 29 patients were enrolled. Genomic and transcriptomic profiling, as well as analyses of immune cell populations, was conducted at baseline and at the time of cystectomy. RESULTS Of the 29 patients treated, a pathologic complete response was observed in 13 cases (44.8%). No major safety concerns were associated with the treatment, and 26 patients (90%) underwent cystectomy. Mutational patterns, tumor mutation burden, and homologous recombination deficiency remained stable throughout treatment and were not predictive of outcomes. However, a shift toward stromal phenotypes and increased expression of epithelial-mesenchymal transition signatures were observed following therapy, particularly in resistant tumors. Moreover, an increase in circulating CD4+ CD27- CD28- T cells was noted among responders. CONCLUSIONS The combination of neoadjuvant durvalumab and olaparib shows therapeutic activity in bladder cancer. Resistance mechanisms seem to be driven by transcriptional adaptations rather than the emergence of new mutations.
Collapse
Affiliation(s)
- Juan F. Rodríguez-Moreno
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA), Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| | | | | | | | - Ricardo Fernández
- Medical Oncology Department, Hospital Universitario de Cruces, Barakaldo, Spain
| | | | | | - Pablo Gajate
- Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Albert Font
- Instituto Catalán de Oncología (ICO), Badalona, Spain
| | - Nuria Lainez
- Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Elena Sevillano-Fernández
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA), Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| | - Osvaldo Graña-Castro
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA), Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| | - Luis Beltrán
- Department of Cellular Pathology, Barts Health NHS Trust, London, United Kingdom
| | - Rodrigo Madurga
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | | | - Pedro Berraondo
- Program of Immunology and Immunotherapy, CIMA Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sergio Ruiz-Llorente
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA), Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
- Laboratory of Innovation in Oncology, HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
- Departamento de Biomedicina y Biotecnología, Área de Genética, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Jesús García-Donas
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA), Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Urbanización Montepríncipe, Madrid, Spain
| |
Collapse
|
28
|
Habaka M, Daly GR, Shinyanbola D, Alabdulrahman M, McGrath J, Dowling GP, Hehir C, Huang HYR, Hill ADK, Varešlija D, Young LS. PARP Inhibitors in the Neoadjuvant Setting; A Comprehensive Overview of the Rationale for their Use, Past and Ongoing Clinical Trials. Curr Oncol Rep 2025; 27:533-551. [PMID: 40192976 DOI: 10.1007/s11912-025-01669-z] [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] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
PURPOSEOF REVIEW Poly (ADP-ribose) polymerases (PARPs) are enzymes essential for detecting and repairing DNA damage through poly-ADP-ribosylation. In cancer, cells with deficiencies in homologous recombination repair mechanisms often become more dependent on PARP-mediated repair mechanisms to effectively repair dsDNA breaks. As such, PARP inhibitors (PARPis) were introduced into clinical practice, serving as a key targeted therapy option through synthetic lethality in the treatment of cancers with homologous recombination repair deficiency (HRD). Though PARPis are currently approved in the adjuvant setting for several cancer types such as ovarian, breast, prostate and pancreatic cancer, their potential role in the neoadjuvant setting remains under investigation. This review outlines the rationale for using PARPi in the neoadjuvant setting and evaluates findings from early and ongoing clinical trials. RECENT FINDINGS Our analysis indicates that numerous studies have explored PARPi as a neoadjuvant treatment for HRD-related cancers. The majority of neoadjuvant PARPi trials have been performed in breast and ovarian cancer, while phase II/III evidence supporting efficacy in prostate and pancreatic cancers remains limited. Studies are investigating PARPi in the neoadjuvant setting of HRD-related cancers. Future research should prioritize combination strategies with immune checkpoint inhibitors and expand outcome measures to include patient satisfaction and quality-of-life metrics.
Collapse
Affiliation(s)
- Minatoullah Habaka
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Gordon R Daly
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Deborah Shinyanbola
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Jason McGrath
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gavin P Dowling
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Cian Hehir
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Helen Ye Rim Huang
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Arnold D K Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| | - Damir Varešlija
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| | - Leonie S Young
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
29
|
Pai Bellare G, Kundu K, Dey P, Philip KT, Chauhan N, Sharma M, Rajput SK, Patro BS. Targeting Replication Fork Processing Synergizes with PARP Inhibition to Potentiate Lethality in Homologous Recombination Proficient Ovarian Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410718. [PMID: 40089867 PMCID: PMC12079468 DOI: 10.1002/advs.202410718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/20/2025] [Indexed: 03/17/2025]
Abstract
Synthetic lethality in homologous recombination (HR)-deficient cancers caused by Poly (ADP-ribose) polymerase inhibitors (PARPi) has been classically attributed to its role in DNA repair. The mode of action of PARPi and resistance thereof are now believed to be predominantly replication associated. Therefore, effective combinatorial approaches of targeting replication fork processing along with HR-downregulation to target HR-proficient and possibly PARPi-resistant tumors are warranted. Stilbenes are a privileged class of molecules, which include resveratrol, pterostilbene, piceatannol, etc, that modulate both replication processes and RAD51-expression. In this investigation, by screening a small library of stilbenes, including in-house synthesized molecules, trans-4,4'-dihydroxystilbene (DHS) was discovered as a potent natural agent, which downregulates RAD51 expression and HR repair (GFP-reporter assay). DHS induces extensive synergistic cell death in ovarian cancers when combined with talazoparib (PARPi). Mechanistically, DHS elicits replication-stress through severely impeding replication fork progress, speed, and inducing fork-asymmetry. This leads to robust induction of single stranded DNA (ssDNA) gaps and poly-ADP-ribosylation (PARylation) in S-phase cells, signifying issues related to lagging (Okazaki) strand synthesis. PARPi, which abrogates PARylation, potentiates DHS induced ssDNA gaps, and their conversion into lethal double strand breaks through MRE11 action. Furthermore, the combination is highly effective in mitigating ovarian tumor xenograft growth in SCID mice and exhibited a good therapeutic-index with no/minimal tissue-toxicity.
Collapse
Affiliation(s)
- Ganesh Pai Bellare
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
- Homi Bhabha National InstituteAnushaktinagarMumbai400094India
| | - Kshama Kundu
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
| | - Papiya Dey
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
| | - Krupa Thankam Philip
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
- Homi Bhabha National InstituteAnushaktinagarMumbai400094India
| | - Nitish Chauhan
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
- Homi Bhabha National InstituteAnushaktinagarMumbai400094India
| | - Muskan Sharma
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
| | | | - Birija Sankar Patro
- Bio‐Organic DivisionBhabha Atomic Research CentreMumbai400085India
- Homi Bhabha National InstituteAnushaktinagarMumbai400094India
| |
Collapse
|
30
|
Call N, Tomkinson AE. Joining of DNA breaks- interplay between DNA ligases and poly (ADP-ribose) polymerases. DNA Repair (Amst) 2025; 149:103843. [PMID: 40347914 DOI: 10.1016/j.dnarep.2025.103843] [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: 12/31/2024] [Revised: 03/28/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
The joining of DNA single- and double-strand breaks (SSB and DSB) is essential for maintaining genome stability and integrity. While this is ultimately accomplished in human cells by the DNA ligases encoded by the LIG1, LIG3 and LIG4 genes, these enzymes are recruited to DNA breaks through specific interactions with proteins involved in break sensing and recognition and/or break processing. In this review, we focus on the interplay between the DNA break-activated poly (ADP-ribose) polymerases, PARP1 and PARP2, poly (ADP-ribose) (PAR) and the DNA ligases in DNA replication and repair. The most extensively studied example of this interplay is the recruitment of DNA ligase IIIα (LigIIIα) and other repair proteins to SSBs through an interaction between XRCC1, a scaffold protein and partner protein of nuclear LigIIIα, and PAR synthesized by PARP1 and to a lesser extent PARP2. Recently, these proteins have been implicated in a back-up pathway for joining Okazaki fragments that appears to have a critical function even in cells with no defect in the major LigI-dependent pathway. Finally, we discuss the effects of FDA-approved PARP1/2 inhibitors on DNA replication and repair in cancer and non-malignant cells and the potential utility of DNA ligase inhibitors as cancer therapeutics.
Collapse
Affiliation(s)
- Nicolas Call
- University of New Mexico Comprehensive Cancer Center and the Departments of Internal Medicine, and Molecular Genetics & Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Alan E Tomkinson
- University of New Mexico Comprehensive Cancer Center and the Departments of Internal Medicine, and Molecular Genetics & Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.
| |
Collapse
|
31
|
Sweatman E, Bayley R, Selemane R, Higgs MR. SETD1A-dependent EME1 transcription drives PARPi sensitivity in HR deficient tumour cells. Br J Cancer 2025; 132:690-702. [PMID: 39994444 PMCID: PMC11997087 DOI: 10.1038/s41416-025-02963-0] [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: 07/10/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cells deficient in DNA repair factors breast cancer susceptibility 1/2 (BRCA1/2) or ataxia-telangiectasia mutated (ATM) are sensitive to poly-ADP ribose polymerase (PARP) inhibitors. Building on our previous findings, we asked how the lysine methyltransferase SETD1A contributed to PARP inhibitor-mediated cell death in these contexts and determined the mechanisms responsible. METHODS We used cervical, breast, lung and ovarian cancer cells bearing mutations in BRCA1 or ATM and depleted SETD1A using siRNA or CRISPR/Cas9. We assessed the effects of the PARPi Olaparib on cell viability, homologous recombination, and DNA repair. We assessed underlying transcriptional perturbations using RNAseq. We used The Cancer Genomics Atlas (TCGA) and DepMap to investigate patient survival and cancer cell characteristics. RESULTS Loss of SETD1A from both BRCA1-deficient and ATM-deficient cancer cells was associated with resistance to Olaparib, explained by partial restoration of homologous recombination. Mechanistically, SETD1A-dependent transcription of the crossover junction endonuclease EME1 correlated with sensitivity to Olaparib in these cells. Accordingly, when SETD1A or EME1 was lost, BRCA1 or ATM-mutated cells became resistant to Olaparib, and homologous recombination was partially restored. CONCLUSIONS Loss of SETD1A or EME1 drives cellular resistance to Olaparib in certain genetic contexts and may help explain why patients develop resistance to PARP inhibitors in the clinic.
Collapse
Affiliation(s)
- Ellie Sweatman
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Rachel Bayley
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Richad Selemane
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Martin R Higgs
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
32
|
Gui F, Jiang B, Jiang J, He Z, Tsujino T, Takai T, Arai S, Pana C, Köllermann J, Bradshaw GA, Eisert R, Kalocsay M, Fassl A, Balk SP, Kibel AS, Jia L. Acute BRCAness induction and AR pathway blockage through CDK12/7/9 degradation enhances PARP inhibitor sensitivity in prostate cancer. SCIENCE ADVANCES 2025; 11:eadu0847. [PMID: 40267193 PMCID: PMC12017310 DOI: 10.1126/sciadv.adu0847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/17/2025] [Indexed: 04/25/2025]
Abstract
Current treatments for advanced prostate cancer (PCa) primarily target the androgen receptor (AR) pathway. However, the emergence of castration-resistant prostate cancer (CRPC) and resistance to AR pathway inhibitors (APPIs) remains ongoing challenges. Here, we present BSJ-5-63, a proteolysis-targeting chimera (PROTAC) targeting cyclin-dependent kinases (CDKs) CDK12, CDK7, and CDK9, offering a multipronged approach to CRPC therapy. BSJ-5-63 degrades CDK12, diminishing BRCA1 and BRCA2 expression and inducing a sustained "BRCAness" state. This sensitizes cancer cells to PARP inhibitors (PARPis) regardless of their homologous recombination repair (HRR) status. Furthermore, CDK7 and CDK9 degradation attenuates AR signaling, enhancing its therapeutic efficacy. Preclinical studies, including both in vitro and in vivo CRPC models, demonstrate that BSJ-5-63 exerts potent antitumor activity in both AR-positive and AR-negative setting. This study introduces BSJ-5-63 as a promising therapeutic agent that addresses both DNA repair and AR signaling mechanisms, with potential benefits for a board patient population.
Collapse
Affiliation(s)
- Fu Gui
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jie Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Takuya Tsujino
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Tomoaki Takai
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Seiji Arai
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Department of Urology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Celine Pana
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Jens Köllermann
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Pathology, Frankfurt am Main, Germany
| | | | - Robyn Eisert
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Marian Kalocsay
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne Fassl
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Steven P. Balk
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Adam S. Kibel
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Jia
- Department of Urology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Wu L, Liang F, Chen C, Zhang Y, Huang H, Pan Y. Identification of prognostic and therapeutic biomarkers associated with macrophage and lipid metabolism in pancreatic cancer. Sci Rep 2025; 15:14584. [PMID: 40281115 PMCID: PMC12032141 DOI: 10.1038/s41598-025-99144-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Although macrophages and lipid metabolism significantly influence the progression of various cancers, their precise roles in pancreatic cancer (PC) remain unclear. This study focuses on identifying and validating biomarkers associated with macrophage-related genes (MRGs) and lipid metabolism-related genes (LMRGs), providing new targets and strategies for therapeutic intervention. This research utilized datasets from TCGA-PAAD, GSE62452, and GSE57495. Candidate genes were identified by overlapping differentially expressed genes with MRGs from WGCNA and LMRGs. Regression analyses were performed to pinpoint potential biomarkers and construct a risk model, which underwent evaluation. A nomogram was subsequently developed and validated. Additional analyses, including functional enrichment, somatic mutation profiling, immune landscape assessment, and RT-qPCR, were performed to investigate the underlying biological mechanisms in PC. The study identified ADH1A, ACACB, CD36, CERS4, PDE3B, ALOX5, and CRAT as biomarkers for PC. RT-qPCR results revealed reduced expression of ADH1A, ACACB, CD36, CERS4, PDE3B, and CRAT in tumor samples compared to adjacent tissues, whereas ALOX5 expression was significantly elevated in tumor samples. A risk model utilizing these biomarkers classified PC patients into high- and low-risk cohorts, with high-risk patients showing lower survival probabilities. Subsequently, risk score and N stage were identified as independent prognostic factors, leading to the development of a nomogram. Notably, both risk cohorts showed significant enrichment in the "cell cycle" pathway. Furthermore, TP53 mutations were prevalent in both high-risk (76%) and low-risk (50%) cohorts. Correlation analysis indicated that PVRL2 (an immunosuppressive factor), CD276 (an immunoactivator), and CCL20 (a chemotactic factor) had the highest positive correlation with the risk score. In this study, ADH1A, ACACB, CD36, CERS4, PDE3B, ALOX5, and CRAT were identified as biomarkers for PC, with their expression levels validated in clinical samples. These findings offered a potential theoretical foundation for developing targeted treatments for PC.
Collapse
Affiliation(s)
- Lili Wu
- Department of Surgical Nursing, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Feihong Liang
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Changgan Chen
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Yaxin Zhang
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China.
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.
| |
Collapse
|
34
|
Akamandisa MP, Boddicker NJ, Yadav S, Hu C, Hart SN, Ambrosone C, Anton-Culver H, Auer PL, Bodelon C, Burnside ES, Chen F, Eliassen AH, 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. Ann Oncol 2025:S0923-7534(25)00170-X. [PMID: 40288678 DOI: 10.1016/j.annonc.2025.04.010] [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: 12/03/2024] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Pathogenic variants (PVs) in ATM, BRCA1, BRCA2, CHEK2, and PALB2 are associated with increased breast cancer risk. However, it is unknown whether this risk differs by PV type or location in carriers ascertained from the general population. PATIENTS AND METHODS To evaluate breast cancer risks associated with PV type and location in ATM, BRCA1, BRCA2, CHEK2, and PALB2, we performed age adjusted case-control association analysis in 32,247 women with and 32,544 age-matched women without breast cancer from the CARRIERS Consortium. 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. RESULTS Compared to women carrying BRCA2 exon 11 protein truncating variants (PTVs) in the CARRIERS population-based study, women with BRCA2 ex1-10 PTVs (OR=13.5, 95%CI 6.0-38.7, P<0.001) and ex13-27 PTVs (OR=9.0, 95%CI 4.9-18.5, P<0.001) had higher breast cancer risks, lower rates of ER-negative breast cancer (ex13-27 OR=0.5, 95%CI 0.2-0.9, P=0.035; ex1-10 OR=0.5, 95%CI 0.1-1.0, P=0.065), and earlier age at breast cancer diagnosis (ex13-27 5.5 years, P<0.001; ex1-10 2.4 years, P=0.169). These associations with ER-negative breast cancer and age replicated in a high-risk clinical cohort from Ambry Genetics and the population-based UK Biobank cohort. No differences in risk by gene region were observed for PTVs in other predisposition genes. CONCLUSION Population-based and clinical high-risk cohorts establish that PTVs in exon 11 of BRCA2 are associated with reduced breast cancer risk, 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.
Collapse
Affiliation(s)
- M P Akamandisa
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - N J Boddicker
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - S Yadav
- Department of Oncology, Mayo Clinic, Rochester, MN
| | - C Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - S N Hart
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - C Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Center, Buffalo, NY
| | | | - P L Auer
- Division of Biostatistics, Institute for Health & Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - C Bodelon
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - E S Burnside
- Department of Radiology, University of Wisconsin, Madison, WI
| | - F Chen
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - A H Eliassen
- Harvard TH Chan School of Public Health, Harvard University, Cambridge, MA
| | | | - C Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - J M Hodge
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - H Huang
- Harvard TH Chan School of Public Health, Harvard University, Cambridge, MA
| | - E M John
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Palo Alto, CA
| | - R Karam
- Ambry Genetics, Aliso Viejo, CA
| | - J V Lacey
- Beckman Research Institute, City of Hope Cancer Center, Duarte, CA
| | - S Lindstroem
- Department of Epidemiology, University of Washington, Seattle, WA
| | - E Martinez
- Department of Family Medicine and Public Health, University of California San Diego, San Diego, CA
| | - J Na
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - S L Neuhausen
- Beckman Research Institute, City of Hope Cancer Center, Duarte, CA
| | - K M O'Brien
- National Institute of Environmental Health Sciences, Durham, NC
| | - J E Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - T Pal
- Division of Genetic Medicine in the Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - J R Palmer
- Slone Epidemiology Center, Boston University, Boston, MA
| | - A V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA
| | | | - E C Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL
| | | | - K Ruddy
- Department of Oncology, Mayo Clinic, Rochester, MN
| | - D P Sandler
- National Institute of Environmental Health Sciences, Durham, NC
| | - L R Teras
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - A Trentham-Dietz
- University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - C M Vachon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - C Weinberg
- National Institute of Environmental Health Sciences, Durham, NC
| | - S J Winham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - S Yao
- Department of Cancer Prevention and Control, Roswell Park Cancer Center, Buffalo, NY
| | - G Zirpoli
- Slone Epidemiology Center, Boston University, Boston, MA
| | - P Kraft
- Trans-Divisional Research Program, National Cancer Institute, Rockville, MD
| | - J N Weitzel
- The University of Kansas Cancer Center, Kansas City, KS
| | - S M Domchek
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - F J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - K L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
35
|
Zhu R, Eason K, Chin SF, Edwards PAW, Manzano Garcia R, Moulange R, Pan JW, Teo SH, Mukherjee S, Callari M, Caldas C, Sammut SJ, Rueda OM. Detecting homologous recombination deficiency for breast cancer through integrative analysis of genomic data. Mol Oncol 2025. [PMID: 40260608 DOI: 10.1002/1878-0261.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/25/2025] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Homologous recombination deficiency (HRD) leads to genomic instability, and patients with HRD can benefit from HRD-targeting therapies. Previous studies have primarily focused on identifying HRD biomarkers using data from a single technology. Here we integrated features from different genomic data types, including total copy number (CN), allele-specific copy number (ASCN) and single nucleotide variants (SNV). Using a semi-supervised method, we developed HRD classifiers from 1404 breast tumours across two datasets based on their BRCA1/2 status, demonstrating improved HRD identification when aggregating different data types. Notably, HRD-positive tumours in ER-negative disease showed improved survival post-adjuvant chemotherapy, while HRD status strongly correlated with neoadjuvant treatment response. Furthermore, our analysis of cell lines highlighted a sensitivity to PARP inhibitors, particularly rucaparib, among predicted HRD-positive lines. Exploring somatic mutations outside BRCA1/2, we confirmed variants in several genes associated with HRD. Our method for HRD classification can adapt to different data types or resolutions and can be used in various scenarios to help refine patient selection for HRD-targeting therapies that might lead to better clinical outcomes.
Collapse
Affiliation(s)
- Rong Zhu
- School of Mathematics and Statistics, Beijing Institute of Technology, Beijing, China
- MRC Biostatistics Unit, University of Cambridge, UK
| | - Katherine Eason
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | | | | | | | | | | | - Sach Mukherjee
- MRC Biostatistics Unit, University of Cambridge, UK
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- University of Bonn, Bonn, Germany
| | | | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, UK
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | | |
Collapse
|
36
|
Jiang KC, Zhu YH, Jiang ZL, Liu Y, Hussain W, Luo HY, Sun WH, Ji XY, Li DX. Regulation of PEST-containing nuclear proteins in cancer cells: implications for cancer biology and therapy. Front Oncol 2025; 15:1548886. [PMID: 40330830 PMCID: PMC12052563 DOI: 10.3389/fonc.2025.1548886] [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: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
The PEST-containing nuclear protein (PCNP) is a nuclear protein involved in the regulation of cell cycle progression, protein degradation, and tumorigenesis. PCNP contains a PEST sequence, a polypeptide structural motif rich in proline (P), glutamic acid (E), serine (S), and threonine (T), which serves as a proteolytic recognition signal. The degradation of specific proteins via the PEST sequence plays a crucial role in modulating signaling pathways that control cell growth, differentiation, apoptosis, and stress responses. PCNP is primarily degraded through the ubiquitin-proteasome system (UPS) and the calpain pathway, with phosphorylation of threonine and serine residues further accelerating its degradation. The ubiquitination of PCNP by the ring finger protein NIRF in an E3 ligase-dependent manner is well documented, along with its involvement in the MAPK and PI3K/AKT/mTOR signaling pathways. Additionally, PCNP is implicated in p53-mediated cell cycle arrest and apoptosis, which are essential for inhibiting tumor growth. To explore the role of PCNP in cancer, this review examines its effects on cell growth, differentiation, proliferation, and apoptosis in lung adenocarcinoma, thyroid cancer, ovarian cancer, and other malignancies derived from glandular epithelial cells. By focusing on PCNP and its regulatory mechanisms, this study provides a scientific basis for further research on the biological functions of the PEST sequence in tumor development and cancer progression.
Collapse
Affiliation(s)
- Kai-Chun Jiang
- Department of Traditional Chinese Medicine, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Yong-Hao Zhu
- School of Stomatology, Henan University, Kaifeng, Henan, China
| | - Zhi-Liang Jiang
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Yi Liu
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wahab Hussain
- School of Stomatology, Henan University, Kaifeng, Henan, China
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Huang-Yin Luo
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Wei-Hang Sun
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Xin-Ying Ji
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Oncology, Huaxian County Hospital, Anyang, Henan, China
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Ding-Xi Li
- The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
37
|
Peng S, Long M, Chen Q, Yin Z, Zeng C, Zhang W, Wen Q, Zhang X, Ke W, Wu Y. Perspectives on cancer therapy-synthetic lethal precision medicine strategies, molecular mechanisms, therapeutic targets and current technical challenges. Cell Death Discov 2025; 11:179. [PMID: 40240755 PMCID: PMC12003663 DOI: 10.1038/s41420-025-02418-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/27/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, synthetic lethality has become an important theme in the field of targeted cancer therapy. Synthetic lethality refers to simultaneous defects in two or more genes leading to cell death, whereas defects in any single gene do not lead to cell death. Taking advantage of the genetic vulnerability that exists within cancer cells, it theoretically has no negative impact on healthy cells and has fewer side effects than non-specific chemotherapy. Currently, targeted cancer therapies focus on inhibiting key pathways in cancer. However, it has been found that over-activation of oncogenic-related signaling pathways can also induce cancer cell death, which is a major breakthrough in the new field of targeted therapies. In this review, we summarize the conventional gene targets in synthetic lethality (PARP, ATR, ATM, WEE1, PRMT) and provide an in-depth analysis of their latest potential mechanisms. We explore the impact of over-activation of pathways such as PI3K/AKT, MAPK, and WNT on cancer cell survival, and present the technical challenges of current research. Important theoretical foundations and insights are provided for the application of synthetic lethal strategies in cancer therapy, as well as future research directions.
Collapse
Affiliation(s)
- Shixuan Peng
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Mengle Long
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Qisheng Chen
- Department of Anesthesiology, The First People's Hospital of Chenzhou, The Chenzhou Affiliated Hospital, Hengyang Medical School, University of South China, Chenzhou, Hunan, 423000, China
| | - Zhijian Yin
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Chang Zeng
- Department of Pathology, Yueyang Central Hospital, Yueyang, China
| | - Wanyong Zhang
- Department of Pathology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, 437100, Hubei, China
| | - Qingyang Wen
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Xinwen Zhang
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Weiqi Ke
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China.
| | - Yongjun Wu
- Department of Pathology, Xiangtan Center Hospital, Xiangtan City, Hunan province, 411100, China.
- Department of Pathology, The Affiliated Hospital of Hunan University, Xiangtan City, Hunan Province, China.
| |
Collapse
|
38
|
Zhou D, Liu W, Zhang Y, Li C. Ivosidenib Confers BRCAness Phenotype and Synthetic Lethality to Poly (ADP-Ribose) Polymerase Inhibition in BRCA1/2-Proficient Cancer Cells. Biomedicines 2025; 13:958. [PMID: 40299557 PMCID: PMC12025137 DOI: 10.3390/biomedicines13040958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/30/2025] [Accepted: 04/05/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objectives: PARP inhibitors (PARPi) are pivotal to treating homologous recombination repair-deficient (HRD) cancers, particularly BRCA1/2-mutated ovarian and breast cancers. However, most ovarian and breast cancers harbor wild-type (WT) BRCA1/2, limiting PARPi eligibility. This study aims to identify an approved drug that could induce a BRCAness phenotype, thereby sensitizing WT BRCA cancers to PARPi. Methods: Ovarian and breast cancer cell lines with WT BRCA1/2 were treated with ivosidenib. HR repair efficiency was assessed via RAD51 foci formation and reporter assays. Synthetic lethality with PARPi was evaluated using viability and colony formation assays. Mechanistic studies included RNA-binding protein pulldown, co-immunoprecipitation, and functional analyses of DNA repair pathways. YTHDC2's role in HR was investigated through siRNA knockdown and rescue experiments. Results: Ivosidenib significantly reduced HR repair efficiency and sensitized cells to PARPi, inducing synthetic lethality. Mechanistically, ivosidenib directly bound YTHDC2, an m6A reader critical for HR. This interaction disrupted YTHDC2's ability to promote DNA double-strand break repair via HR, evidenced by impaired recruitment of repair proteins (e.g., BRCA1, RAD51) and accumulation of DNA damage (γH2AX foci). YTHDC2 knockdown phenocopied ivosidenib effects, while overexpression rescued HR defects. Conclusions: Ivosidenib induces BRCAness in WT BRCA ovarian and breast cancers by targeting YTHDC2, thereby suppressing HR repair and enhancing PARPi sensitivity. This uncovers a novel, metabolism-independent mechanism of ivosidenib, repositioning it as a therapeutic agent for HRD tumors. These findings propose a strategy to expand PARPi eligibility to WT BRCA cancers, addressing a critical unmet need in oncology.
Collapse
Affiliation(s)
- Danyang Zhou
- Department of Oncology, The Affiliated Dazu’s Hospital of Chongqing Medical University, Chongqing 402360, China; (D.Z.); (W.L.); (Y.Z.)
- Department of Respiratory, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210012, China
| | - Wei Liu
- Department of Oncology, The Affiliated Dazu’s Hospital of Chongqing Medical University, Chongqing 402360, China; (D.Z.); (W.L.); (Y.Z.)
| | - Yanyan Zhang
- Department of Oncology, The Affiliated Dazu’s Hospital of Chongqing Medical University, Chongqing 402360, China; (D.Z.); (W.L.); (Y.Z.)
| | - Chong Li
- Department of Oncology, The Affiliated Dazu’s Hospital of Chongqing Medical University, Chongqing 402360, China; (D.Z.); (W.L.); (Y.Z.)
| |
Collapse
|
39
|
Yan L, He Q, Verma SP, Zhang X, Giel AS, Maj C, Graz K, Naderi E, Chen J, Ali MW, Gharahkhani P, Shu X, Offit K, Shah PM, Gerdes H, Molena D, Srivastava A, MacGregor S, Palles C, Thieme R, Vieth M, Gockel I, Vaughan TL, Schumacher J, Buas MF. Biologically targeted discovery-replication scan identifies G×G interaction in relation to risk of Barrett's esophagus and esophageal adenocarcinoma. HGG ADVANCES 2025; 6:100399. [PMID: 39755942 PMCID: PMC11815673 DOI: 10.1016/j.xhgg.2025.100399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025] Open
Abstract
Inherited genetics represents an important contributor to risk of esophageal adenocarcinoma (EAC), and its precursor Barrett's esophagus (BE). Genome-wide association studies have identified ∼30 susceptibility variants for BE/EAC, yet genetic interactions remain unexamined. To address challenges in large-scale G×G scans, we combined knowledge-guided filtering and machine learning approaches, focusing on genes with (1) known/plausible links to BE/EAC pathogenesis (n = 493) or (2) prior evidence of biological interactions (n = 4,196). Approximately 75 × 106 SNP×SNP interactions were screened via hierarchical group lasso (glinternet) using BEACON GWAS data. The top ∼2,000 interactions retained in each scan were prioritized using p values from single logistic models. Identical scans were repeated among males only (78%), with two independent GWAS datasets used for replication. In overall and male-specific primary replications, 11 of 187 and 20 of 191 interactions satisfied p < 0.05, respectively. The strongest evidence for secondary replication was for rs17744726×rs3217992 among males, with consistent directionality across all cohorts (Pmeta = 2.19 × 10-8); rs3217992 "T" was associated with reduced risk only in individuals homozygous for rs17744726 "G." Rs3217992 maps to the CDKN2B 3' UTR and reportedly disrupts microRNA-mediated repression. Rs17744726 maps to an intronic enhancer region in BLK. Through in silico prioritization and experimental validation, we identified a nearby proxy variant (rs4841556) as a functional modulator of enhancer activity. Enhancer-gene mapping and eQTLs implicated BLK and FAM167A as targets. The first systematic G×G investigation in BE/EAC, this study uncovers differential risk associations for CDKN2B variation by BLK genotype, suggesting novel biological dependency between two risk loci encoding key mediators of tumor suppression and inflammation.
Collapse
Affiliation(s)
- Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Qianchuan He
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shiv P Verma
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xu Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ann-Sophie Giel
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - Carlo Maj
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - Kathryn Graz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elnaz Naderi
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jianhong Chen
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mourad Wagdy Ali
- Department of Genome Sciences, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Xiang Shu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Offit
- Clinical Genetics, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pari M Shah
- Gastroenterology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans Gerdes
- Gastroenterology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amitabh Srivastava
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Claire Palles
- Department of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - René Thieme
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander-Universiät Erlangen-Nürnberg, Klinikum Bayreuth, Bayreuth, Germany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Thomas L Vaughan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Epidemiology, University of Washington, School of Public Health, Seattle, WA, USA
| | | | - Matthew F Buas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
40
|
Gonzalez-Leal C, Cai J, de Groot BFJ, Wegerer A, Preisser J, Luijsterburg M, Ladurner A. Poly-(ADP-ribose) serves as a scaffold for the methyltransferase METTL3/14 complex in the DNA damage response. Nucleic Acids Res 2025; 53:gkaf244. [PMID: 40219966 PMCID: PMC11992677 DOI: 10.1093/nar/gkaf244] [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: 02/24/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
PARP1, a crucial DNA break sensor, synthesizes poly-(ADP-ribose) (PAR), a nucleic acid that promotes the recruitment of DNA repair proteins. Emerging evidence highlights a role of RNA and RNA-binding proteins in DNA repair. Notably, the RNA-m6A methyltransferase complex METTL3/14 is implicated in repairing ultraviolet-induced DNA lesions. Here, we dissected the interplay between the two nucleic acids PAR and RNA and how METTL3/14 recruitment and m6A accumulation at laser-induced DNA lesions responds to PAR dynamics. In vitro, METTL3/14 recognized both PAR and RNA, yet PAR presence did not inhibit the methyltransferase complex's catalytic activity. Acute knock-out of METTL3 rendered cells sensitive to transcription-blocking DNA damage and resulted in defects in transcription recovery and transcription-coupled DNA repair. Furthermore, combining METTL3 and PARP inhibitors led to an enhanced antiproliferative effect on cancer cells. Future therapeutic avenues may thus leverage the interplay between the nucleic acids PAR and RNA.
Collapse
Affiliation(s)
- Claudia Gonzalez-Leal
- Department of Physiological Chemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg – Martinsried, Germany
- International Max Planck Research School (IMPRS) for Molecules of Life, 82152 Planegg-Martinsried, Germany
| | - Jin Cai
- Department of Physiological Chemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg – Martinsried, Germany
- International Max Planck Research School (IMPRS) for Molecules of Life, 82152 Planegg-Martinsried, Germany
| | - Bram A F J de Groot
- Department of Human Genetics, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Andreas Wegerer
- Department of Physiological Chemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg – Martinsried, Germany
| | - Julia Preisser
- Department of Physiological Chemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg – Martinsried, Germany
| | - Martijn S Luijsterburg
- Department of Human Genetics, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Andreas G Ladurner
- Department of Physiological Chemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg – Martinsried, Germany
- International Max Planck Research School (IMPRS) for Molecules of Life, 82152 Planegg-Martinsried, Germany
- Eisbach Bio GmbH, Am Klopferspitz 19, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
41
|
Chauhan R, Damerla RR, Dhyani VS. Synthetic lethality in cancer: a protocol for scoping review of gene interactions from synthetic lethal screens and functional studies. Syst Rev 2025; 14:81. [PMID: 40200332 PMCID: PMC11978169 DOI: 10.1186/s13643-025-02814-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Two genes are synthetically lethal if loss of function of either one of the two genes does not result in cell death, whereas loss of function of both genes together results in being detrimental to cell survival. This concept has been the basis for developing personalized, precision treatments, which can selectively damage tumor cells and minimize toxicity to normal tissues. Tumor cells often harbor mutations in genes involved in DNA repair pathways, forcing them to switch to alternative repair pathways, leading to chemotherapeutic resistance. These interactions, if targeted, could be synthetically lethal. We aimed to summarize synthetically lethal gene pairs that could be utilized to selectively target cancer cells and minimize side effects on normal tissues. The objective of this review is to study druggable synthetically lethal gene pairs for targeted cancer therapy that have been identified through various genetic screens and functional studies. METHODS A systematic literature search will be conducted to extract synthetically lethal gene pairs that can be specifically targeted to cancer cells. Owing to the relatively recent research pertaining to this field, the literature search will incorporate data from 1956. The search will be conducted on PubMed, Web of Science, Embase, and Scopus. The narrative approach will guide the analysis and synthesis of the results. DISCUSSION This review highlights scientific articles that report druggable synthetically lethal gene pairs by testing the efficacy of targeted inhibitors in clonogenic assays. These include research studies that identify synthetically lethal gene pairs detected through CRISPR screens by knocking out one or two genes within the same cell and testing the potency of inhibitors to specifically kill malignant cells. SYSTEMATIC REVIEW REGISTRATION https://doi.org/10.17605/OSF.IO/5BCW6 .
Collapse
Affiliation(s)
- Raashi Chauhan
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rama Rao Damerla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Vijay Shree Dhyani
- Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
42
|
Bai D, Nowak M, Lu D, Wang Q, Fitzgerald M, Zhang H, MacDonald R, Xu Z, Luo L. The outcast of medicine: metals in medicine--from traditional mineral medicine to metallodrugs. Front Pharmacol 2025; 16:1542560. [PMID: 40260378 PMCID: PMC12010122 DOI: 10.3389/fphar.2025.1542560] [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: 12/10/2024] [Accepted: 03/07/2025] [Indexed: 04/23/2025] Open
Abstract
Metals have long held a significant role in the human body and have been utilized as mineral medicines for thousands of years. The modern advancement of metals in pharmacology, particularly as metallodrugs, has become crucial in disease treatment. As the machanism of metallodurgsare increasingly uncovered, some metallodrugs are already approved by FDA and widely used in treating antitumor, antidiabetes, and antibacterial. Therefore, a thorough understanding of metallodrug development is essential for advancing future study. This review offers an in-depth examination of the evolution of mineral medicines and the applications of metallodrugs within contemporary medicine. We specifically aim to summarize the historical trajectory of metals and mineral medicines in Traditional Chinese Mineral Medicine by analyzing key historical texts and representative mineral medicines. Additionally, we discuss recent advancements in understanding metallodrugs' mechanisms, such as protein interactions, enzyme inhibition, DNA interactions, reactive oxygen species (ROS) generation, and cellular structure targeting. Furthermore, we address the challenges in metallodrug development and propose potential solutions. Lastly, we outline future directions for metallodrugs to enhance their efficacy and effectiveness. The progression of metallodrugs has broadened their applications and contributed significantly to patient health, creating good healthcare solutions for the global population.
Collapse
Affiliation(s)
- Donghan Bai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Michal Nowak
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Dajun Lu
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Qiaochu Wang
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, United States
| | | | - Hui Zhang
- Institute of Traditional Chinese Medicine, European University of Chinese Medicine, Horsens, Denmark
| | - Remy MacDonald
- Department of Statistics, George Mason University, Virginia, VA, United States
| | - Ziwen Xu
- Department of Nursing, The University of Melbourne, Parkville, VIC, Australia
| | - Lu Luo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
43
|
Meléndez-Flórez MP, Ortega-Recalde O, Rangel N, Rondón-Lagos M. Chromosomal Instability and Clonal Heterogeneity in Breast Cancer: From Mechanisms to Clinical Applications. Cancers (Basel) 2025; 17:1222. [PMID: 40227811 PMCID: PMC11988187 DOI: 10.3390/cancers17071222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Chromosomal instability (CIN) and clonal heterogeneity (CH) are fundamental hallmarks of breast cancer that drive tumor evolution, disease progression, and therapeutic resistance. Understanding the mechanisms underlying these phenomena is essential for improving cancer diagnosis, prognosis, and treatment strategies. METHODS In this review, we provide a comprehensive overview of the biological processes contributing to CIN and CH, highlighting their molecular determinants and clinical relevance. RESULTS We discuss the latest advances in detection methods, including single-cell sequencing and other high-resolution techniques, which have enhanced our ability to characterize intratumoral heterogeneity. Additionally, we explore how CIN and CH influence treatment responses, their potential as therapeutic targets, and their role in shaping the tumor immune microenvironment, which has implications for immunotherapy effectiveness. CONCLUSIONS By integrating recent findings, this review underscores the impact of CIN and CH on breast cancer progression and their translational implications for precision medicine.
Collapse
Affiliation(s)
- María Paula Meléndez-Flórez
- Departamento de Morfología, Facultad de Medicina e Instituto de Genética, Universidad Nacional de Colombia, Bogotá 110231, Colombia; (M.P.M.-F.); (O.O.-R.)
| | - Oscar Ortega-Recalde
- Departamento de Morfología, Facultad de Medicina e Instituto de Genética, Universidad Nacional de Colombia, Bogotá 110231, Colombia; (M.P.M.-F.); (O.O.-R.)
- Department of Pathology, Instituto Nacional de Cancerología, Bogotá 110231, Colombia
| | - Nelson Rangel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Milena Rondón-Lagos
- Escuela de Ciencias Biológicas, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| |
Collapse
|
44
|
Martires LCM, Ahronian LG, Pratt CB, Das NM, Zhang X, Whittington DA, Zhang H, Shen B, Come J, McCarren P, Liu MS, Min C, Feng T, Jahic H, Ali JA, Aird DR, Li F, Andersen JN, Huang A, Mallender WD, Nicholson HE. LIG1 Is a Synthetic Lethal Target in BRCA1 Mutant Cancers. Mol Cancer Ther 2025; 24:618-627. [PMID: 39868490 PMCID: PMC11962389 DOI: 10.1158/1535-7163.mct-24-0598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/08/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Synthetic lethality approaches in BRCA1/2-mutated cancers have focused on PARP inhibitors, which are subject to high rates of innate or acquired resistance in patients. In this study, we used CRISPR/Cas9-based screening to identify DNA ligase I (LIG1) as a novel target for synthetic lethality in BRCA1-mutated cancers. Publicly available data supported LIG1 hyperdependence of BRCA1 mutant cells across a variety of breast and ovarian cancer cell lines. We used CRISPRn, CRISPRi, RNAi, and protein degradation to confirm the lethal effect of LIG1 inactivation at the DNA, RNA, and protein level in BRCA1 mutant cells in vitro. LIG1 inactivation resulted in viability loss across multiple BRCA1-mutated cell lines, whereas no effect was observed in BRCA1/2 wild-type cell lines, demonstrating target selectivity for the BRCA1 mutant context. On-target nature of the phenotype was demonstrated through rescue of viability with exogenous wild-type LIG1 cDNA. Next, we demonstrated a concentration-dependent relationship of LIG1 protein expression and BRCA1 mutant cell viability using a titratable, degradable LIG1 fusion protein. BRCA1 mutant viability required LIG1 catalytic activity, as catalytically dead mutant LIG1K568A failed to rescue viability loss caused by endogenous LIG1 depletion. LIG1 perturbation produced proportional increases in PAR staining in BRCA1 mutant cells, indicating a mechanism consistent with the function of LIG1 in sealing ssDNA nicks. Finally, we confirmed LIG1 hyperdependence in vivo using a xenograft model in which LIG1 loss resulted in tumor stasis in all mice. Our cumulative findings demonstrate that LIG1 is a promising synthetic lethal target for development in patients with BRCA1-mutant cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jon Come
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | - Mu-Sen Liu
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | | | - Haris Jahic
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | | | - Fang Li
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | - Alan Huang
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | | |
Collapse
|
45
|
Conceição CJF, Moe E, Ribeiro PA, Raposo M. PARP1: A comprehensive review of its mechanisms, therapeutic implications and emerging cancer treatments. Biochim Biophys Acta Rev Cancer 2025; 1880:189282. [PMID: 39947443 DOI: 10.1016/j.bbcan.2025.189282] [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: 05/22/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
The Poly (ADP-ribose) polymerase-1 (PARP1) enzyme is involved in several signalling pathways related to homologous repair (HR), base excision repair (BER), and non-homologous end joining (NHEJ). Studies demonstrated that the deregulation of PARP1 function and control mechanisms can lead to cancer emergence. On the other side, PARP1 can be a therapeutic target to maximize cancer treatment. This is done by molecules that can modulate radiation effects, such as DNA repair inhibitors (PARPi). With this approach, tumour cell viability can be undermined by targeting DNA repair mechanisms. Thus, treatment using PARPi represents a new era for cancer therapy, and even new horizons can be attained by coupling these molecules with a nano-delivery system. For this, drug delivery systems such as liposomes encompass all the required features due to its excellent biocompatibility, biodegradability, and low toxicity. This review presents a comprehensive overview of PARP1 biological features and mechanisms, its role in cancer development, therapeutic implications, and emerging cancer treatments by PARPi-mediated therapies. Although there are a vast number of studies regarding PARP1 biological function, some PARP1 mechanisms are not clear yet, and full-length PARP1 structure is missing. Nevertheless, literature reports demonstrate already the high usefulness and vast possibilities offered by combined PARPi cancer therapy.
Collapse
Affiliation(s)
- Carlota J F Conceição
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal.
| | - Elin Moe
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal; Department of Chemistry, UiT-The Arctic University of Norway, N-9037 Tromsø, Norway.
| | - Paulo A Ribeiro
- Laboratory of Instrumentation, Biomedical Engineering and Radiation Physics (LIBPhys-UNL), Department of Physics, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal.
| | - Maria Raposo
- Laboratory of Instrumentation, Biomedical Engineering and Radiation Physics (LIBPhys-UNL), Department of Physics, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal.
| |
Collapse
|
46
|
Lahiri S, Hamilton G, Moore G, Goehring L, Huang TT, Jensen RB, Rothenberg E. BRCA2 prevents PARPi-mediated PARP1 retention to protect RAD51 filaments. Nature 2025; 640:1103-1111. [PMID: 40140565 DOI: 10.1038/s41586-025-08749-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/06/2025] [Indexed: 03/28/2025]
Abstract
The tumour-suppressor protein BRCA2 has a central role in homology-directed DNA repair by enhancing the formation of RAD51 filaments on resected single-stranded DNA generated at double-stranded DNA breaks and stimulating RAD51 activity1,2. Individuals with BRCA2 mutations are predisposed to cancer; however, BRCA2-deficient tumours are often responsive to targeted therapy with PARP inhibitors (PARPi)3-6. The mechanism by which BRCA2 deficiency renders cells sensitive to PARPi but with minimal toxicity in cells heterozygous for BRCA2 mutations remains unclear. Here we identify a previously unknown role of BRCA2 that is directly linked to the effect of PARP1 inhibition. Using biochemical and single-molecule approaches, we demonstrate that PARPi-mediated PARP1 retention on a resected DNA substrate interferes with RAD51 filament stability and impairs RAD51-mediated DNA strand exchange. Full-length BRCA2 protects RAD51 filaments and counteracts the instability conferred by PARPi-mediated retention by preventing the binding of PARP1 to DNA. Extending these findings to a cellular context, we use quantitative single-molecule localization microscopy to show that BRCA2 prevents PARPi-induced PARP1 retention at homologous-recombination repair sites. By contrast, BRCA2-deficient cells exhibit increased PARP1 retention at these lesions in response to PARPi. These results provide mechanistic insights into the role of BRCA2 in maintaining RAD51 stability and protecting homologous-recombination repair sites by mitigating PARPi-mediated PARP1 retention.
Collapse
Affiliation(s)
- Sudipta Lahiri
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - George Hamilton
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Gemma Moore
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Liana Goehring
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Tony T Huang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ryan B Jensen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
47
|
Li P, Wu D, Yu X. Targeting dePARylation in cancer therapy. DNA Repair (Amst) 2025; 148:103824. [PMID: 40056493 DOI: 10.1016/j.dnarep.2025.103824] [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: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025]
Abstract
Poly(ADP-ribosyl)ation (PARylation), a reversible post-translational modification mediated by poly(ADP-ribose) polymerases (PARPs), plays crucial roles in DNA replication and DNA damage repair. Since interfering PARylation induces selective cytotoxicity in tumor cells with homologous recombination defects, PARP inhibitors (PARPi) have significant clinical impacts in treating BRCA-mutant cancer patients. Likewise, dePARylation is also essential for optimal DNA damage response and genomic stability. This process is mediated by a group of dePARylation enzymes, such as poly(ADP-ribose) glycohydrolase (PARG). Currently, several novel PARG inhibitors have been developed and examined in preclinical and clinical studies, demonstrating promising anti-cancer activity distinct from PARP inhibitors. This review discusses the role of dePARylation in genome stability and the potential of PARG inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Peng Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Duo Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaochun Yu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Yang J, Wan SY, Song QY, Xie YH, Wan J, Zhou YH, Zhang ZT, Xiao YS, Li X, Chen H, Liu XR, Xu L, You HJ, Hu DS, Petersen RB, Zhang YH, Zheng L, Zhang Y, Huang K. Angiopoietin-like protein 8 directs DNA damage responses towards apoptosis by stabilizing PARP1-DNA condensates. Cell Death Differ 2025; 32:672-688. [PMID: 39592710 PMCID: PMC11982567 DOI: 10.1038/s41418-024-01422-2] [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: 05/23/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024] Open
Abstract
Upon genotoxic stresses, cells employ various DNA damage responses (DDRs), including DNA damage repair or apoptosis, to safeguard genome integrity. However, the determinants among different DDRs choices are largely unknown. Here, we report angiopoietin-like protein 8 (ANGPTL8), a secreted regulator of lipid metabolism, localizes to the nucleus and acts as a dynamic switch that directs DDRs towards apoptosis rather than DNA repair after genotoxin exposure. ANGPTL8 deficiency alleviates DNA damage and apoptosis in cells exposed to genotoxins, as well as in the liver or kidney of mice injured by hepatic ischemia/reperfusion or cisplatin treatment. Mechanistically, ANGPTL8 physically interacts with Poly (ADP-ribose) polymerase 1 (PARP1), in a PARylation-independent manner, and reduces the fluidity of PARP1-DNA condensates, thereby enhancing the pro-apoptotic accumulation of PARP1 and PAR chains on DNA lesions. However, the transcription of ANGPTL8 is gradually decreased following genotoxin treatment, partly due to downregulation of CCAAT enhancer binding protein alpha (CEBPA), presumably to avoid further cytotoxicity. Together, we provide new insights by which genotoxic stress induced DDRs are channeled to suicidal apoptosis to safeguard genome integrity.
Collapse
Affiliation(s)
- Jing Yang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shi-Yuan Wan
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiu-Yi Song
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hao Xie
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Jun Wan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yi-Hao Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Zi-Tong Zhang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Shuo Xiao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xi Li
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Chen
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin-Ran Liu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Xu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Juan You
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - De-Sheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI, 48858, USA
| | - Yong-Hui Zhang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yu Zhang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Tongji-Rong Cheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
49
|
Frederick MI, Fyle E, Clouvel A, Abdesselam D, Hassan S. Targeting FEN1/EXO1 to enhance efficacy of PARP inhibition in triple-negative breast cancer. Transl Oncol 2025; 54:102337. [PMID: 40054125 PMCID: PMC11928819 DOI: 10.1016/j.tranon.2025.102337] [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: 10/05/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. The only targeted therapeutic approach that has emerged for early TNBC patients with BRCA-mutations (BRCAMUT) are PARP inhibitors (PARPi). In combination, PARPi may benefit a larger cohort of TNBC patients. We used our previously identified 63-gene signature that was associated with PARPi response to identify candidate genes that could be therapeutic targets. We selected FEN1 for further investigation since its knockdown was associated with an increase in G2/M arrest, DNA damage, and apoptosis. We first tested LNT1, a FEN1/EXO1 inhibitor, in a panel of 10 TNBC cell lines. LNT1 sensitivity was identified predominantly in BRCA1-mutant/deficient cell lines. However, the combination of PARPi and LNT1 demonstrated a synergistic or additive effect in 7/10 cell lines, mainly in BRCA1/2 wild-type (BRCAWT) and BRCA2-mutant cell lines, with intrinsic and acquired resistance to PARPi. The greatest synergy was observed in a BRCA2-mutant cell line with acquired resistance to olaparib (HCC1395-OlaR), with a combination index value of 0.20. In the synergistic cell lines, BT549 (BRCAWT) and HCC1395-OlaR, the combination was associated with a rapid progression in DNA replication fork speed, an early and sustained increase in DNA damage in comparison to each of the single-agents. However, in the additive BRCA1/2 wild-type cell lines, MDAMB231 and HCC1806, the combination demonstrated a high DNA damage response that was largely driven by either talazoparib or LNT1. Therefore, targeting FEN1/EXO1 with PARPi is a promising targeted combination approach, particularly in the context of PARPi-resistant and BRCAWT TNBC.
Collapse
Affiliation(s)
- Mallory I Frederick
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Elicia Fyle
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Anna Clouvel
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Djihane Abdesselam
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Saima Hassan
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada; Division of Surgical Oncology, Department of Surgery, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, QC H2X0C1, Canada.
| |
Collapse
|
50
|
Pires MJ, Alam S, Lovric A, Fabbrizi E, Rotili D, Altun M, Valerie NCK. Duplexed CeTEAM drug biosensors reveal determinants of PARP inhibitor selectivity in cells. J Biol Chem 2025; 301:108361. [PMID: 40021124 PMCID: PMC11986510 DOI: 10.1016/j.jbc.2025.108361] [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: 09/18/2024] [Revised: 01/31/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) targeting PARP1 and PARP2 have revolutionized cancer therapy by selectively killing cancer cells with defective DNA repair. However, achieving PARP1 or PARP2-selective inhibitors is difficult due to structural homology. Selectivity profiling is typically done with purified proteins, but these lack the complexity of intracellular environments and could therefore be inaccurate. Here, we duplex PARP1 L713F-GFP and PARP2 L269A-mCherry cellular target engagement by accumulation of mutant (CeTEAM) drug biosensors to systematically characterize binding and cell cycle alterations of 27 PARPi. Our results reveal that most PARPi are equipotent for both PARPs, including the next-generation drug, senaparib. However, benzimidazole carboxamide (niraparib) derivatives demonstrated PARP1-selective tendencies, while phthalazinones (olaparib) favored PARP2. AZD5305, a reported PARP1-selective inhibitor with characteristics of both series, was the exception and appears ∼1600-fold more potent toward PARP1. In agreement with current understanding, we see that trapping-associated S/G2-phase transitions positively correlate with PARP1/2 binding potency, while some potent binders, such as veliparib, did not - likely reflecting their allosteric influence on DNA retention. We also assessed the effect of the PARP1/2 active site component, histone PARylation factor 1, on intracellular PARPi binding and see that its depletion elicits slight deviations in apparent binding potency, while contributing additively to trapping-like phenotypes. The PARP1/2 CeTEAM platform thus provides a structural roadmap for the development of selective PARPi and should facilitate the discovery of targeted therapies. Furthermore, our results highlight that multiplexing CeTEAM biosensors and layered genetic perturbations can systematically profile determinants of intracellular drug selectivity.
Collapse
Affiliation(s)
- Maria J Pires
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Sweden
| | - Seher Alam
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Sweden
| | - Alen Lovric
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Sweden
| | - Emanuele Fabbrizi
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome Roma RM, Italy
| | - Dante Rotili
- Department of Science, "Roma Tre" University, Rome, Italy; INBB - Biostructures and Biosystems National Institute, Rome, Italy
| | - Mikael Altun
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Sweden.
| | - Nicholas C K Valerie
- Science for Life Laboratory, Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Sweden.
| |
Collapse
|