1
|
Li Y, Yang X, Cai H, Wang F. Homologous recombination deficiency among patients with germline or somatic non-BRCA1/2 homologous recombination repair gene variations. NPJ Precis Oncol 2025; 9:192. [PMID: 40527918 DOI: 10.1038/s41698-025-00999-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 06/05/2025] [Indexed: 06/20/2025] Open
Abstract
This study examined the relationship between homologous recombination deficiency (HRD) and variations in non-BRCA1/2 homologous recombination repair (HRR) genes. 27.3% (132/483) of the patients with ovarian, breast, endometrial, prostate, and pancreatic cancers carrying non-BRCA1/2 HRR variations were HRD + . Germline mutations were associated with significantly higher HRD+ rates than somatic mutations, while biallelic alterations did not show stronger associations with HRD compared to monoallelic alterations. High HRD+ rates (66.7-100.0%) were associated with variations in PALB2, RAD51C/D, and RAD54L, while low HRD+ rates (0-37.5%) corresponded with variations in PTEN, ATM, BRIP1, CDK12, and NBN, which may be influenced by variation grade and tissue origin. HRD positivity was mutually exclusive with HER2+ status in breast cancer and with TMB-H/MSI-H in endometrial cancer. Overall, these findings highlight the different strengths of the correlation between non-BRCA1/2 HRR gene variations and HRD and guide HRD testing in cases of "BRCA1/2-wildtype" results.
Collapse
Affiliation(s)
- Yue Li
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Xinhua Yang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Haoyang Cai
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
| |
Collapse
|
2
|
Jaidee R, Pocasap P, Jusakul A, Senggunprai L, Prawan A, Hong JH, Heng HL, Kukongviriyapan V, Teh BT, Kongpetch S. Synergistic suppression of cholangiocarcinoma cells via DNA damage response and cell cycle arrest by dual targeting PARP and ATM in DNA damage repair pathway. Biomed Pharmacother 2025; 189:118273. [PMID: 40527032 DOI: 10.1016/j.biopha.2025.118273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 05/28/2025] [Accepted: 06/13/2025] [Indexed: 06/19/2025] Open
Abstract
This study investigated alterations in DNA damage repair (DDR) genes in cholangiocarcinoma (CCA) influencing cellular vulnerability to DDR inhibitors, notably poly (ADP-ribose) polymerase inhibitors (PARPi) and ataxia-telangiectasia mutated inhibitors (ATMi). Genomic DDR alterations in CCA tumors were identified via cBioPortal. The cytotoxic effect of the inhibitors was determined in 8 CCA cell lines. Cell cycle arrest and apoptosis were determined using flow cytometry. The levels of γ-H2A histone family member X (γ-H2AX) were evaluated using an immunofluorescence assay. Proteins associated with the cell cycle and DDR were evaluated using Western blotting. Among these CCA cells, BRCA-mutated KKU-023 cells exhibited the highest PARPi sensitivity. Co-treatment with ATMi induced a synthetic homologous recombination deficiency (HRD) state in BRCA-proficient CCA cells, thereby sensitizing them to PARPi. This drug combination suppressed CCA cell growth via G2/M phase arrest, downregulated cyclin A and B1 expression, and enhanced apoptosis. Mechanistically, this synergistic cytotoxicity was attributed to increased DNA double-strand breaks, evidenced by elevated γ-H2AX and reduced RAD51 expression, signifying compromised DNA repair. These preclinical findings support the therapeutic potential of DDR inhibitors, individually or in combination, for CCA subsets characterized by DDR gene alterations.
Collapse
Affiliation(s)
- Rattanaporn Jaidee
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Piman Pocasap
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand; Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Jing Han Hong
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Hong Lee Heng
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre of Singapore, Singapore
| | - Veerapol Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Bin Tean Teh
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre of Singapore, Singapore
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
3
|
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
|
4
|
Sisman Y, Schejbel L, Schnack TH, Høgdall C, Høgdall E. Clinical Characteristics and Survival of Ovarian Cancer Patients According to Homologous Recombination Deficiency Status. Cancers (Basel) 2025; 17:1628. [PMID: 40427127 PMCID: PMC12109656 DOI: 10.3390/cancers17101628] [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: 04/10/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND HRD is a key biomarker in ovarian cancer, predicting response to PARP inhibitors. However, it remains unclear whether HRD-positive patients differ from HRD-negative patients in terms of clinical characteristics in PARP inhibitor-naïve populations. This study aims to evaluate platinum-sensitive PARP-inhibitor naïve ovarian cancer patients' clinical characteristics and survival outcomes based on HRD status. Secondly, to investigate whether platinum-resistant patients with homologous recombination repair (HRR) gene mutations are HRD-positive. METHODS Two distinct HRD algorithms-an in-house genomic instability score (GIS) and the normalized large-scale state transitions score (nLST)-were used to stratify patients as HRD-positive or HRD-negative. Clinical data and survival in PARP inhibitor-naïve, platinum-sensitive HGSC patients were analyzed. RESULTS A total of 71 platinum-sensitive PARP-inhibitor naïve patients were analyzed. By in-house GIS, 37 patients (52%) were classified as HRD-positive and 34 (48%) as HRD-negative. Using nLST, 43 (61%) were HRD-positive and 28 (39%) were HRD-negative. Our analysis revealed no significant differences in clinical parameters or survival between HRD-positive and HRD-negative platinum-sensitive patients. The only observed difference was that somatic BRCA1/2-mutated patients were younger. In the subgroup of six platinum-resistant patients harboring HRR gene mutations, four patients (67%) were classified as HRD positive. CONCLUSIONS Our findings suggest that HRD status does not significantly influence clinical characteristics or survival outcomes in platinum-sensitive, PARP inhibitor-naïve HGSC patients. As some platinum-resistant patients with HRR gene mutations were HRD positive; this subgroup may benefit from further investigation into the potential effect of PARP inhibitors.
Collapse
Affiliation(s)
- Yagmur Sisman
- Department of Gynecology, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark; (Y.S.); (C.H.)
- Department of Pathology, Copenhagen University Hospital, Herlev University Hospital, 2730 Herlev, Denmark;
| | - Lone Schejbel
- Department of Pathology, Copenhagen University Hospital, Herlev University Hospital, 2730 Herlev, Denmark;
| | | | - Claus Høgdall
- Department of Gynecology, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark; (Y.S.); (C.H.)
| | - Estrid Høgdall
- Department of Pathology, Copenhagen University Hospital, Herlev University Hospital, 2730 Herlev, Denmark;
| |
Collapse
|
5
|
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
|
6
|
Lin YY, Noghabi HS, Volik S, Bell R, Sar F, Haegert A, Chung HC, Fazli L, Oo HZ, Daugaard M, Kuo MH, Hsu SC, Imeda EL, Zanettini C, Queiroz L, Schlotmann B, Gheybi K, Cooper C, Kote-Jarai Z, Eeles R, Kung HJ, Marchionni L, Weischenfeldt J, Miller KD, Rabinowitz A, Wang Y, Zhang HF, Sorensen PH, Carey MS, Gleave M, Hayes VM, Gibson WT, Collins CC. Identifying Rare Germline Variants Associated with Metastatic Prostate Cancer Through an Extreme Phenotype Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.28.25326584. [PMID: 40343042 PMCID: PMC12060958 DOI: 10.1101/2025.04.28.25326584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Background Studies of germline variants in prostate cancer (PCa) have largely focused on their connections to cancer predisposition. However, an understanding of how heritable factors contribute to cancer progression and metastasis remain limited. Objective To identify low frequency to rare germline nonsynonymous variants associated with increased risk for metastatic PCa (mPCa), while providing functional validation. Design We assembled an extreme phenotype cohort (EPC) of 52 patients diagnosed with predominantly high-grade (Gleason Score (GS) ≥ 8) PCa and > 7 years of follow-up for which localized treatment naïve tumor tissues were available. In half of the cases, the tumor had metastasized to bone, providing an even distribution of bone mPCa and non mPCa cases. Tumor and matched distant benign DNA samples were exome sequenced and analyzed for germline variants with population-wide minor allelic frequencies ≤ 2%. Findings were validated using two independent PCa germline cohorts, including a closely matched Australian study biased to aggressive disease (n = 53) and Pan Prostate Cancer Group (PPCG, n = 976). Two mPCa-promoting candidate variants in KDM6B and BRCA2 were engineered into cell lines and functionalized. Results Germline nonsynonymous rare variants (gnsRVs) identified in 25 DNA Damage Repair (DDR) genes were significantly enriched in the mPCa patients (p=4.57e-06). Conversely, the prevalence of synonymous variants at minor allele frequencies of ≤ 2% were similar between the mPCa and non mPCa patients. The predictive power of variants in 53 non-DDR genes was validated in the Australian cohort (p=0.028) and correlated with high-risk PCa in PPCG (p=0.03). KDM6B K973Q showed functional significance despite being annotated as benign in ClinVar, while BRCA2 I1962T showed sensitivity to Olaparib. In total, six EPC variants related to DNA repair or epigenetics were found to alter enzymatic activity. Conclusions EPCs coupled with low frequency/rare variant analyses may advance understanding of interactions between the germline and tumor in PCa. We identified a series of germline variants that were enriched among mPCa patients. Moreover, we showed that one of these variants confers a metastatic phenotype. Our findings suggest that germline testing at diagnosis may improve treatment stratification in PCa. Patient summary The presence of specific genetic variants among men with PCa may elevate the risk of mPCa once PCa develops. Knowledge of the variant burden at time of diagnosis may enable accurate stratification of some patients for aggressive therapeutic interventions.
Collapse
Affiliation(s)
- Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- These authors are joint first authors
| | - Hamideh Sharifi Noghabi
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- These authors are joint first authors
| | - Stanislav Volik
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Robert Bell
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Funda Sar
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Hee Chul Chung
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Ming-Han Kuo
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Chieh Hsu
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Eddie L Imeda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Claudio Zanettini
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Lucio Queiroz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Balthasar Schlotmann
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Kazzem Gheybi
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Colin Cooper
- The Institute of Cancer Research, London, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Rosalind Eeles
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Hsing-Jien Kung
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA
| | - Joachim Weischenfeldt
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Alan Rabinowitz
- Rural Coordination Center of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, British Columbia, Canada
| | - Hai-Feng Zhang
- Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark S Carey
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Vanessa M Hayes
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - William T Gibson
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- These authors are joint last authors
| | - Colin C Collins
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- These authors are joint last authors
| |
Collapse
|
7
|
Zhao F, Cai C, Gao H, Moon J, Christyani G, Qin S, Hao Y, Liu T, Lou Z, Kim W. Mono-ubiquitination of TopBP1 by PHRF1 enhances ATR activation and genomic stability. Nucleic Acids Res 2025; 53:gkaf073. [PMID: 40052822 PMCID: PMC11886831 DOI: 10.1093/nar/gkaf073] [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: 03/12/2024] [Revised: 12/28/2024] [Accepted: 01/28/2025] [Indexed: 03/10/2025] Open
Abstract
The TopBP1-ATR axis is critical for maintaining genomic stability during DNA replication stress, yet the precise regulation of TopBP1 in replication stress responses remains poorly understood. In this study, we identified PHD and Ring Finger Domains 1 (PHRF1) as an important ATR activator through its interaction with TopBP1. Our analysis revealed a correlation between PHRF1 and genomic stability in cancer patients. Mechanistically, PHRF1 is recruited to DNA lesions in a manner dependent on its PHD domain and histone methylation. Subsequently, PHRF1 mono-ubiquitinates TopBP1 at lysine 73, which enhances the TopBP1-ATR interaction and activates ATR. Depletion of PHRF1 disrupts ATR activation and sensitizes cells to replication stress-inducing agents. Furthermore, conditional knockout of Phrf1 in mice leads to early lethality and impaired ATR-Chk1 axis signaling. Collectively, our findings establish PHRF1 as a novel E3 ligase for TopBP1, coordinating the replication stress response by enhancing TopBP1-ATR signaling.
Collapse
Affiliation(s)
- Fei Zhao
- College of Biology, Hunan University, Changsha 410082, China
| | - Chenghui Cai
- College of Biology, Hunan University, Changsha 410082, China
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, United States
| | - Jaeyoung Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151 Chungcheongnam-do, Republic of Korea
| | - Grania Christyani
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151 Chungcheongnam-do, Republic of Korea
| | - Sisi Qin
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151 Chungcheongnam-do, Republic of Korea
| | - Yalan Hao
- Analytical Instrumentation Center, Hunan University, Changsha 410082, China
| | - Tongzheng Liu
- College of Pharmacy/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Zhenkun Lou
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, United States
| | - Wootae Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151 Chungcheongnam-do, Republic of Korea
| |
Collapse
|
8
|
Sánchez-Beltrán J, Soler Díaz J, Herraiz C, Olivares C, Cerdido S, Cerezuela-Fuentes P, García-Borrón JC, Jiménez-Cervantes C. An MGRN1-Based Biomarker Combination Accurately Predicts Melanoma Patient Survival. Int J Mol Sci 2025; 26:1739. [PMID: 40004203 PMCID: PMC11855888 DOI: 10.3390/ijms26041739] [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: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
With ever-increasing incidence and high metastatic potential, cutaneous melanoma is the deadliest skin cancer. Risk prediction based on the Tumor-Node-Metastasis (TNM) staging system has medium accuracy with intermediate IIB-IIIB stages, as roughly 25% of patients with low-medium-grade TNM, and hence a favorable prognostic, undergo an aggressive disease with short survival and around 15% of deaths arise from metastases of thin, low-risk lesions. Therefore, reliable prognostic biomarkers are required. We used genomic and clinical information of melanoma patients from the TCGA-SKCM cohort and two GEO studies for discovery and validation of potential biomarkers, respectively. Neither mutation nor overexpression of major melanoma driver genes provided significant prognostic information. Conversely, expression of MGRN1 and the melanocyte-specific genes MLANA, PMEL, and TYRP1 provided a simple 4-gene signature identifying with high-sensitivity (>80%), low-medium TNM patients with adverse outcomes. Transcriptomic analysis of tumors with this signature, or from low-medium-grade TNM patients with poor outcomes, revealed comparable dysregulation of an inflammatory response, cell cycle progression, and DNA damage/repair programs. A functional analysis of MGRN1-knockout cells confirmed these molecular features. Therefore, the simple MGRN1-MLANA-PMEL-TYRP1 combination of biomarkers complemented TNM staging prognostic accuracy and pointed to the dysregulation of immunological responses and genomic stability as determinants of a melanoma outcome.
Collapse
Affiliation(s)
- José Sánchez-Beltrán
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Javier Soler Díaz
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
| | - Cecilia Herraiz
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Conchi Olivares
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Sonia Cerdido
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Pablo Cerezuela-Fuentes
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
- Medical Oncology Department, Hospital Clínico Universitario Virgen de La Arrixaca, 30120 Murcia, Spain
| | - José Carlos García-Borrón
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Celia Jiménez-Cervantes
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| |
Collapse
|
9
|
Arce-Gallego S, Cresta Morgado P, Delgado-Serrano L, Simonetti S, Gonzalez M, Romero-Lozano P, Marmolejo D, Morales-Barrera R, Arnau GM, Semidey ME, Aguilar D, Cordoba-Terreros S, Mast R, de Albert M, Planas J, Cuadras M, Maldonado X, Suarez C, Casanova-Salas I, Figols M, Cros S, Mas A, Nonell L, Dienstmann R, Nuciforo P, Vivancos A, Llop-Guevara A, Carles J, Serra V, Mateo J. Homologous recombination repair status in metastatic prostate cancer by next-generation sequencing and functional immunofluorescence. Cell Rep Med 2025; 6:101937. [PMID: 39914385 PMCID: PMC11866514 DOI: 10.1016/j.xcrm.2025.101937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/19/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025]
Abstract
Metastatic prostate cancer (mPC) is enriched for homologous recombination repair (HRR) gene alterations, which have prognostic and predictive value. Routine clinical implementation of next-generation sequencing (NGS) is still limited. We investigated the association between genomic and functional loss of HRR, using NGS and RAD51 immunofluorescence (RAD51-IF) in 219 primary or metastatic biopsies from 187 patients with stage IV prostate cancer. NGS showed frequent genomic alterations in TP53 (40%), AR (15%), PTEN (14%), FOXA1 (12%), MYC (10%), BRCA2 (9%), ATM (8%), and BRCA1 (2%). We pursued RAD51-IF in 206 samples; of those, 139/206 (67%) were evaluable. 21% of samples had RAD51-low score compatible with HRR deficiency (HRD). RAD51-IF showed high sensitivity (71%) and specificity (86%) for BRCA1/2 alterations. Patients with RAD51-low scores experienced longer progression-free survival (PFS) on poly(ADP-ribose) polymerase inhibitors (PARPi) or platinum chemotherapy. RAD51-IF is feasible in routine clinical samples from patients with mPC and is associated with clinically relevant HRR gene alterations.
Collapse
Affiliation(s)
- Sara Arce-Gallego
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pablo Cresta Morgado
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Oncology Data Science (OdysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Luisa Delgado-Serrano
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Sara Simonetti
- Molecular Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Macarena Gonzalez
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paula Romero-Lozano
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - David Marmolejo
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rafael Morales-Barrera
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Gisela Mir Arnau
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Daniel Aguilar
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Richard Mast
- Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Matias de Albert
- Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jacques Planas
- Department of Urology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Mercè Cuadras
- Department of Urology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Xavier Maldonado
- Department of Radiation Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Cristina Suarez
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Irene Casanova-Salas
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Mariona Figols
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Manresa, Spain
| | - Sara Cros
- Department of Medical Oncology, Granollers Hospital, Granollers, Spain
| | - Alba Mas
- Bioinformatics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lara Nonell
- Bioinformatics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (OdysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; University of Vic - Central University of Catalonia, Vic, Spain
| | - Paolo Nuciforo
- Molecular Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Alba Llop-Guevara
- Experimental Therapeutics, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Joan Carles
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Violeta Serra
- Experimental Therapeutics, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Joaquin Mateo
- Prostate Cancer Research, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
10
|
Xu Z, Nie C, Liao J, Ma Y, Zhou XA, Li X, Li S, Lin H, Luo Y, Cheng K, Mao Z, Zhang L, Pan Y, Chen Y, Wang W, Wang J. DDX39A resolves replication fork-associated RNA-DNA hybrids to balance fork protection and cleavage for genomic stability maintenance. Mol Cell 2025; 85:490-505.e11. [PMID: 39706185 DOI: 10.1016/j.molcel.2024.11.029] [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: 05/28/2024] [Revised: 10/21/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024]
Abstract
Safeguarding replication fork stability in transcriptionally active regions is crucial for precise DNA replication and mutation prevention. Here, we discover the pervasive existence of replication fork-associated RNA-DNA hybrids (RF-RDs) in transcriptionally active regions of human cells. These hybrids function as protective barriers, preventing DNA2-mediated nascent DNA degradation and replication fork collapse under replication stress. We also identify DDX39A as a RAD51-associated protein that binds to stalled forks and resolves RF-RDs, facilitating proper DNA2-mediated DNA resection and replication fork restart. Excessive dissolution of RF-RDs causes replication fork collapse and genomic instability, while insufficient dissolution of RF-RDs under replication stress increases fork stability, resulting in chemoresistance that can be reversed by eliminating RF-RDs. In summary, we elucidated the prevalence of RF-RDs at replication forks within transcriptionally active regions, revealed their pivotal role in safeguarding replication fork stability, and proposed that targeting RF-RDs holds promise for augmenting chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Zhanzhan Xu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Chen Nie
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Junwei Liao
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yujie Ma
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xiao Albert Zhou
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Shiwei Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Haodong Lin
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yefei Luo
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Kaiqi Cheng
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Zuchao Mao
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Lei Zhang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yichen Pan
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yuke Chen
- Department of Urology, Peking University First Hospital, Beijing 100034, China
| | - Weibin Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China.
| | - Jiadong Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University International Cancer Institute, Institute of Advanced Clinical Medicine, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China; Department of Gastrointestinal Translational Research, Peking University Cancer Hospital, Beijing 100142, China.
| |
Collapse
|
11
|
Fougner V, Urup T, Poulsen HS, Grunnet K, Westmose CY, Melchior LC, Larsen KB, Højgaard M, Spanggaard I, Belcaid L, Rohrberg KS, Lassen U, Hasselbalch B, Nørøxe DS. Actionable alterations in glioblastoma: Insights from the implementation of genomic profiling as the standard of care from 2016 to 2023. Neurooncol Pract 2025; 12:34-44. [PMID: 39917766 PMCID: PMC11798607 DOI: 10.1093/nop/npae082] [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] [Indexed: 02/09/2025] Open
Abstract
Background In 2016, genomic profiling was implemented for patients with grade 4 primary brain tumors at Rigshospitalet, Denmark. The aim of this study was to discover actionable alterations and to match these with targeted therapies. Methods Between January 2016 and December 2023, 483 brain tumor patients were profiled. We retrieved clinical data and molecular data. Whole exome, whole genome, or panel sequencing, along with SNP array analyses, and RNA-seq were performed on resected primary tumor tissue. Alterations were classified according to the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT) following the European Association of Neuro-Oncology (EANO) guideline on rational molecular testing. Results A total of 200 (41.4%) patients' tumors harbored an alteration of interest according to the EANO guideline. Twenty (4.1%) patients had an ESCAT high-tier alteration (tier I or II), while 155 patients (32.1%) had an alteration corresponding to ESCAT IIIA. Thirty-five patients (7.2%) had an actionable alteration, and 15 (3.1%) received targeted therapy. The treated targets were BRAFV600E mutations, FGFR alterations, NTRK fusions, PDGFRA fusions, PTPRZ1-MET fusions, and TMB-high. The overall response rate was 20%, with a median duration of response of 12 months, and 47% achieved stable disease as the best response. Conclusions Genomic profiling uncovers alterations of interest in a substantial number of patients, but only a minority are considered by the Danish National Molecular Tumor Board to have actionable alterations, and even fewer receive targeted therapy. Nevertheless, factors, such as promising targets and the increasing availability of trials, may contribute to a future increase in the number of patients benefiting from targeted therapies based on genomic profiling.
Collapse
Affiliation(s)
- Vincent Fougner
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| | - Thomas Urup
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| | - Kirsten Grunnet
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| | - Christina Yde Westmose
- Center for Genomic Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Linea Cecilie Melchior
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Karen Bonde Larsen
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Martin Højgaard
- Phase 1 Unit, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Iben Spanggaard
- Phase 1 Unit, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Laila Belcaid
- Phase 1 Unit, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kristoffer Staal Rohrberg
- Phase 1 Unit, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Lassen
- Phase 1 Unit, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| | - Benedikte Hasselbalch
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| | - Dorte Schou Nørøxe
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Danish Comprehensive Cancer Center - Brain Tumor Center (DCCC-BTC), Copenhagen, Denmark
| |
Collapse
|
12
|
Subramanian DN, Zethoven M, Pishas KI, Marinović ER, McInerny S, Rowley SM, Allan PE, Devereux L, Cheasley D, James PA, Campbell IG. Assessment of candidate high-grade serous ovarian carcinoma predisposition genes through integrated germline and tumour sequencing. NPJ Genom Med 2025; 10:1. [PMID: 39794353 PMCID: PMC11724014 DOI: 10.1038/s41525-024-00447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/07/2024] [Indexed: 01/13/2025] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) has a significant hereditary component, only half of which is explained. Previously, we performed germline exome sequencing on BRCA1 and BRCA2-negative HGSOC patients, revealing three proposed and 43 novel candidate genes enriched with rare loss-of-function variants. For validation, we undertook case-control analyses using genomic data from disease-free controls. This confirms enrichment for nearly all previously identified genes. Additionally, one-hundred-and-eleven HGSOC tumours from variant carriers were sequenced alongside other complementary studies, seeking evidence of biallelic inactivation as supportive evidence. PALB2 and ATM validate as HGSOC predisposition genes, with 6/8 germline carrier tumours exhibiting biallelic inactivation accompanied by characteristic mutational signatures. Among candidate genes, only LLGL2 consistently shows biallelic inactivation and protein expression loss, supporting it as a novel HGSOC susceptibility gene. The remaining candidate genes fail to validate. Integrating case-control analyses with tumour sequencing is thus crucial for accurate gene discovery in familial cancer studies.
Collapse
Affiliation(s)
- Deepak N Subramanian
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Maia Zethoven
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Kathleen I Pishas
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Evanny R Marinović
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Simone McInerny
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Simone M Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Prue E Allan
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lisa Devereux
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Lifepool Cohort, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul A James
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Ian G Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
13
|
Schejbel L, Poulsen TS, Vestergaard LK, Christensen IJ, Høgdall E. Evaluation of the Oncomine Comprehensive Assay Plus NGS Panel and the OncoScan CNV Assay for Homologous Recombination Deficiency Detection. Mol Diagn Ther 2025; 29:117-127. [PMID: 39312094 PMCID: PMC11742463 DOI: 10.1007/s40291-024-00745-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Testing for homologous recombination deficiency (HRD) as a biomarker in relation to poly (ADP-ribose) polymerase inhibitor (PARPi) treatment in ovarian cancer is done by sequencing of the BRCA1/2 genes and/or by assessing a genomic instability signature. Here we present data obtained with two different methods for genomic instability testing: the Oncomine™ Comprehensive Assay Plus (OCA Plus) NGS panel and the OncoScan CNV assay. METHODS The retrospective analytical study included 80 ovarian cancer samples of patients previously referred to clinical Myriad testing (reference cohort), and 50 ovarian cancer samples from patients collected as part of the Pelvic Mass study. OCA Plus NGS libraries were sequenced with the Ion S5™XL Sequencer and analyzed with the Ion Reporter™ Software v5.20 for calculation of the genomic instability metric (GIM). In addition, all samples were tested with the OncoScan CNV FFPE Assay and analyzed with a previously published R-algorithm for generation of an in-house genomic instability score (in-house GIS). RESULTS The OCA Plus assay had a concordance to the reference of 89% on samples with a tumor fraction ≥ 30% (auto-calculated or via molecular estimation). A total of 15 samples in the reference cohort had a calculated tumor fraction < 30% in the OCA Plus assay. In these, the concordance to reference was only 60%. For the OncoScan CNV in-house GIS a local cutoff point of ≥ 50 was calculated. This gave a concordance to the reference of 85%, with 91% of the samples in the reference cohort passing quality control (QC) on tumor fraction. Both assays had a high sensitivity for the detection of genomic instability in samples with pathogenic or likely pathogenic BRCA1/2 mutations, with 12/13 being GIM positive (OCA Plus assay) and 13/13 being in-house GIS positive (OncoScan CNV assay). CONCLUSIONS The OCA Plus assay and the OncoScan CNV assay show a high but not complete concordance to reference standard homologous recombination deficiency (HRD) detection. The main reason for QC failure or non-concordance in our study was a low tumor fraction estimated in the assay, despite the selection of material by a pathologist with an inclusion criterion of > 30% tumor. QC steps should include careful tumor content evaluation, and results on samples with < 30% tumor should not be reported.
Collapse
Affiliation(s)
- Lone Schejbel
- Department of Pathology, Copenhagen University Hospital, Herlev, Denmark.
| | | | | | | | - Estrid Høgdall
- Department of Pathology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
14
|
Tsilingiri K, Chalari A, Christopoulou G, Voutsina A, Constantoulakis P, Potaris Κ, Vamvakaris I, Hatzidaki D, Zachou G, Vatsellas G, Georgoulias V, Kotsakis A, Klinakis A. Genomic scarring score predicts the response to PARP inhibitors in non-small cell lung cancer. NPJ Precis Oncol 2024; 8:291. [PMID: 39725687 DOI: 10.1038/s41698-024-00777-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
PARP inhibitors (PARPi) have shown efficacy in tumours harbouring mutations in homologous recombination repair (HRR) genes. Somatic HRR mutations have been described in patients with Non-Small Cell Lung Cancer (NSCLC), but PARP inhibitors (PARPi) are not yet a therapeutic option. Here we assessed the homologous recombination status of early-stage NSCLC and explored the therapeutic benefit of PARPi in preclinical models. The Genomic Scarring Score GSS (GSS) and HRR mutation profile of 136 patients were assessed. High GSS (h-GSS) was observed in 39 (28.7%) patients half of which carried pathogenic/likely pathogenic somatic HRR mutations. TP53 mutations were significantly enriched in h-GSS tumours (p < 0.001). Olaparib significantly delayed tumour growth in h-GSS but not l-GSS Patient-derived Xenografts (PDXs), while patients with h-GSS/TP53mut tumours respond favourably to adjuvant platinum-based chemotherapy. Our functional data clearly support the idea that the use of GSS rather than the mutational status of HRR genes could select patients for administration of PARPi.
Collapse
Affiliation(s)
| | - Anna Chalari
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Georgia Christopoulou
- Genotypos MSA, Private Molecular Biology and Cytogenetics Diagnostic Center, Athens, Greece
| | - Alexandra Voutsina
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | | | | | | | | | | | - Giannis Vatsellas
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larisa, Larisa, Greece
| | - Apostolos Klinakis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
15
|
Wang L, Ge Y, Yin L, Zong D, Li Y, Wu J, He X. Dynamic changes in serum adenosine and the adenosine metabolism-based signature for prognosis in HER2-positive metastatic breast cancer patients. Heliyon 2024; 10:e39545. [PMID: 39669152 PMCID: PMC11636132 DOI: 10.1016/j.heliyon.2024.e39545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 12/14/2024] Open
Abstract
Aims Adenosine metabolism in the breast cancer microenvironment is critical for tumor immunity. However, the prognostic significance of adenosine in breast cancer remains unclear. We aimed to dynamically monitor serum adenosine levels in patients with HER2-positive metastatic breast cancer (MBC) patients and to explore its predictive significance in trastuzumab therapy. Methods The sequencing and clinical data were downloaded from TCGA and GSE176078. Adenosine-related differentially expressed genes was analyzed by "DESeq2" package. Multivariate Cox and lasso-penalized Cox regressions were used to construct prognostic risk signatures. The risk scores were calculated from the identified expression of the hub genes. Bioinformatic analyses were performed using R with related packages. We also enrolled the metastatic breast cancer patients with HER2-positive from in our center and classified them into different groups according to the clinical outcomes assessed by enhanced CT. The adenosine levels were dynamically detected, and the difference in immune microenvironment between the subgroups was assessed by the immune cells that were recorded in our center. Results A total of 109 breast cancer patients with HER2-positive MBC were enrolled, and the expressions of 22 adenosine-related genes were filtered and matched from the TCGA database. The survival model based on the 15 differentially expressed genes was established, and the risk scores of each patient were the prognostic risk factors. Single-cell transcriptome sequencing data identified transcriptomic differences in patients with HER2-positive breast cancer. We also confirmed the predictive value of serum adenosine in the clinical progression of HER2-positive MBC patients. The different immune microenvironment between the subgroups supported the reliability of the predictive ability of adenosine in HER2-positive MBC patients. Conclusions The dynamic change of adenosine is a predictive biomarker for monitoring disease progression. The adenosine metabolism-based signature has the potential application in the prognosis of HER2-positive MBC patients.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Li Yin
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Dan Zong
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Yang Li
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Jianfeng Wu
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
16
|
Dodson AE, Shenker S, Sullivan P, Nayak SU, Middleton C, McGuire M, Chipumuro E, Mishina Y, Tobin ER, Cadzow L, Wylie AA, Sangurdekar D. Pan-Cancer Analysis of Homologous Recombination Deficiency in Cell Lines. CANCER RESEARCH COMMUNICATIONS 2024; 4:3084-3098. [PMID: 39485057 PMCID: PMC11621922 DOI: 10.1158/2767-9764.crc-24-0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/10/2024] [Accepted: 10/30/2024] [Indexed: 11/03/2024]
Abstract
SIGNIFICANCE HRD is common in cancer and can be exploited therapeutically, as it sensitizes cells to DNA-damaging agents. Here, we scored more than 1,300 cancer cell lines for HRD using two different bioinformatic approaches, thereby enabling large-scale analyses that provide insights into the etiology and features of HRD.
Collapse
|
17
|
He W, Zhu H, Zhang S, Shu G, Lei H, Wang M, Yin G, Ni X, Wu Q. Epigenetic editing of BRCA1 promoter increases cisplatin and olaparib sensitivity of ovarian cancer cells. Epigenetics 2024; 19:2357518. [PMID: 38796857 PMCID: PMC11135871 DOI: 10.1080/15592294.2024.2357518] [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: 01/24/2024] [Revised: 04/23/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Drug resistance is the primary contributor to the high mortality rate of ovarian cancer (OC). The loss of BRCA1/2 function is linked to drug sensitivity in OC cells. The aim of this study is to enhance the drug sensitivity of OC cells by inducing BRCA1 dysfunction through promoter epigenetic editing. Epigenetic regulatory regions within the BRCA1 promoter, affecting gene expression, were initially discerned through analysis of clinical samples. Subsequently, we designed and rigorously validated epigenetic editing tools. Ultimately, we evaluated the cisplatin and olaparib sensitivity of the OC cells after editing. The BRCA1 promoter contains two CpG-rich regions, with methylation of the region covering the transcription start site (TSS) strongly correlating with transcription and influencing OC development, prognosis, and homologous recombination (HR) defects. Targeting this region in OC cells using our designed epigenetic editing tools led to substantial and persistent DNA methylation changes, accompanied by significant reductions in H3K27ac histone modifications. This resulted in a notable suppression of BRCA1 expression and a decrease in HR repair capacity. Consequently, edited OC cells exhibited heightened sensitivity to cisplatin and olaparib, leading to increased apoptosis rates. Epigenetic inactivation of the BRCA1 promoter can enhance cisplatin and olaparib sensitivity of OC cells through a reduction in HR repair capacity, indicating the potential utility of epigenetic editing technology in sensitization therapy for OC.
Collapse
Affiliation(s)
- Wanhong He
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Haijun Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Sufen Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Guang Shu
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Han Lei
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xiaohua Ni
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| |
Collapse
|
18
|
Planas-Paz L, Pauli C. Leveraging homologous recombination deficiency for sarcoma : Unravelling homologous recombination repair deficiency and therapeutic opportunities in soft tissue and bone sarcoma. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:14-19. [PMID: 39535612 PMCID: PMC11695455 DOI: 10.1007/s00292-024-01381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Homologous recombination deficiency (HRD) in tumors correlates with poor prognosis and metastases development. Determining HRD is of major clinical relevance as it can be treated with PARP inhibitors (PARPi). HRD remains poorly investigated in sarcoma, a rare and heterogeneous cancer of mesenchymal origin. OBJECTIVE We aimed (i) to investigate predictive biomarkers of HRD in several independent sarcoma cohorts using a cross-functional strategy by combining genomic, transcriptomic and phenotypic approaches and (ii) to evaluate the therapeutic potential of PARPi and DNA damage response (DDR)-based therapies ex vivo. MATERIALS AND METHODS We performed a comprehensive genomic and transcriptomic characterization of sarcoma using datasets from The Cancer Genome Atlas (TCGA) and Therapeutically Applicable Research to Generate Effective Treatments (TARGET), and our own bone and soft tissue sarcoma cohorts. We evaluated PARP1/2 and WEE1 inhibition ex vivo in patient-derived sarcoma cell models as monotherapy and in combination with chemotherapeutic agents to identify synergistic effects. RESULTS Firstly, we identified genomic traits of HRD in a subset of sarcomas associated with molecular alterations in homologous recombination repair (HRR) pathway genes and high chromosomal instability. Secondly, we identified and validated distinct SARC-HRD transcriptional signatures that predicted sensitivity to PARPi. Finally, we showed functional defects in HRR in sarcoma cells that were associated with functional dependency towards PARPi and WEE1i and support the clinical use of RAD51 as a predictive biomarker for PARPi sensitivity. CONCLUSION We provide a personalized oncological approach to potentially improve the treatment of sarcoma patients. We encourage the evaluation of gene expression signatures to enhance the identification of patients who might benefit from DDR-based therapies.
Collapse
Affiliation(s)
- Lara Planas-Paz
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland.
- Medical Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Ichikawa H, Aizawa M, Kano Y, Hanyu T, Muneoka Y, Hiroi S, Ueki H, Moro K, Hirose Y, Miura K, Shimada Y, Sakata J, Yabusaki H, Nakagawa S, Kawasaki T, Okuda S, Wakai T. Landscape of homologous recombination deficiency in gastric cancer and clinical implications for first-line chemotherapy. Gastric Cancer 2024; 27:1273-1286. [PMID: 39110344 DOI: 10.1007/s10120-024-01542-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/25/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Homologous recombination deficiency (HRD) is one of the crucial hallmarks of cancer. It is associated with a favorable response to platinum-based chemotherapy. We explored the distinctive clinicopathological features of gastric cancer (GC) with HRD and the clinical significance of HRD in platinum-based first-line chemotherapy for unresectable metastatic GC. METHODS We enrolled 160 patients with GC in this study. Their tumor samples were subjected to genomic profiling utilizing targeted tumor sequencing. HRD was defined as the presence of alterations in any of 16 HR genes (BARD1, BLM, BRCA1, BRCA2, BRIP1, MRE11A, NBN, PALB2, PARP1, POLD1, RAD50, RAD51, RAD51C, RAD51D, WRN, and XRCC2). The clinicopathological features and treatment outcomes of first-line chemotherapy for unresectable metastatic GC were compared between HRD and non-HRD groups. RESULTS Forty-seven patients (29.4%) were classified into the HRD group. This group had a significantly lower proportion of macroscopic type 3 or 4 tumors and higher TMB than the non-HRD group. Among patients who underwent platinum-based first-line chemotherapy, the HRD group had a greater response rate and longer progression-free survival after treatment (median 8.0 months vs. 3.0 months, P = 0.010), with an adjusted hazard ratio of 0.337 (95% confidence interval 0.151-0.753). HRD status was not associated with treatment outcomes in patients who did not undergo platinum-based chemotherapy. CONCLUSIONS Low proportion of macroscopic type 3 or 4 tumors and a high TMB are distinctive features of GC with HRD. HRD status is a potential predictive marker in platinum-based first-line chemotherapy for unresectable metastatic GC.
Collapse
Affiliation(s)
- Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | - Masaki Aizawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Yosuke Kano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Takaaki Hanyu
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yusuke Muneoka
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Sou Hiroi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hiroto Ueki
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Kazuki Moro
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yuki Hirose
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Kohei Miura
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Jun Sakata
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hiroshi Yabusaki
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Satoru Nakagawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Takashi Kawasaki
- Department of Pathology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 2-5274, Gakkocho-dori, Chuo-ku, Niigata City, Niigata, 951-8514, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| |
Collapse
|
20
|
Bertucci F, Guille A, Lerebours F, Ceccarelli M, Syed N, Adélaïde J, Finetti P, Ueno NT, Van Laere S, Viens P, De Nonneville A, Goncalves A, Birnbaum D, Callens C, Bedognetti D, Mamessier E. Whole-exome profiles of inflammatory breast cancer and pathological response to neoadjuvant chemotherapy. J Transl Med 2024; 22:969. [PMID: 39465437 PMCID: PMC11514970 DOI: 10.1186/s12967-024-05790-8] [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: 08/14/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) became a standard treatment strategy for patients with inflammatory breast cancer (IBC) because of high disease aggressiveness. However, given the heterogeneity of IBC, no molecular feature reliably predicts the response to chemotherapy. Whole-exome sequencing (WES) of clinical tumor samples provides an opportunity to identify genomic alterations associated with chemosensitivity. METHODS We retrospectively applied WES to 44 untreated IBC primary tumor samples and matched normal DNA. The pathological response to NACT, assessed on operative specimen, distinguished the patients with versus without pathological complete response (pCR versus no-pCR respectively). We compared the mutational profiles, spectra and signatures, pathway mutations, copy number alterations (CNAs), HRD, and heterogeneity scores between pCR versus no-pCR patients. RESULTS The TMB, HRD, and mutational spectra were not different between the complete (N = 13) versus non-complete (N = 31) responders. The two most frequently mutated genes were TP53 and PIK3CA. They were more frequently mutated in the complete responders, but the difference was not significant. Only two genes, NLRP3 and SLC9B1, were significantly more frequently mutated in the complete responders (23% vs. 0%). By contrast, several biological pathways involved in protein translation, PI3K pathway, and signal transduction showed significantly higher mutation frequency in the patients with pCR. We observed a higher abundance of COSMIC signature 7 (due to ultraviolet light exposure) in tumors from complete responders. The comparison of CNAs of the 3808 genes included in the GISTIC regions between both patients' groups identified 234 genes as differentially altered. The CIN signatures were not differentially represented between the complete versus non-complete responders. Based on the H-index, the patients with heterogeneous tumors displayed a lower pCR rate (11%) than those with less heterogeneous tumors (35%). CONCLUSIONS This is the first study aiming at identifying correlations between the WES data of IBC samples and the achievement of pCR to NACT. Our results, obtained in this 44-sample series, suggest a few subtle genomic alterations associated with pathological response. Additional investigations are required in larger series.
Collapse
Affiliation(s)
- François Bertucci
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France.
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France.
| | - Arnaud Guille
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Florence Lerebours
- Department of Medical Oncology, Institut Curie Saint-Cloud, Paris, France
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
- Department of Public Health Sciences, University of Miami, Miami, USA
| | - Najeeb Syed
- University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - José Adélaïde
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Naoto T Ueno
- University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Patrice Viens
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Alexandre De Nonneville
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Anthony Goncalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Céline Callens
- Department of Medical Oncology, Institut Curie Saint-Cloud, Paris, France
| | - Davide Bedognetti
- Tumor Biology and Immunology Laboratory, Research Branch, Sidra Medicine, Doha, Qatar
| | - Emilie Mamessier
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| |
Collapse
|
21
|
Zhou Y, Xu J. Impact of PARP inhibitors on progression-free survival in platinum-sensitive recurrent epithelial ovarian cancer: a retrospective analysis. World J Surg Oncol 2024; 22:276. [PMID: 39434111 PMCID: PMC11492665 DOI: 10.1186/s12957-024-03562-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Poly (ADP-ribose) polymerase (PARP) inhibitors such as olaparib and niraparib have shown promise in extending progression-free survival (PFS) in patients with platinum-sensitive recurrent (PSR) epithelial ovarian cancer. In this retrospective study, we aimed to present our own data on the effect of PARP inhibitors on PFS in recurrent epithelial ovarian cancer. METHODS 82 patients diagnosed with PSR epithelial ovarian, tubal, or primary peritoneal cancer between May 2017 and September 2023 were initially enrolled from our hospital. However, 16 patients had prior exposure to PARP inhibitors during primary treatment, and 11 were lost to follow-up. Consequently, the study focused on 55 eligible patients. PFS was compared between patients receiving PARP inhibitor maintenance therapy and those who did not. RESULTS Among the 55 patients with PSR epithelial ovarian cancer, 18 received olaparib as maintenance therapy, 19 received niraparib, and 18 opted for observation. PARP inhibitor therapy significantly extended PFS (mean 24.0 months) compared to observation (mean 9.0 months, p = 0.0005), regardless of BRCA mutation status (HR = 0.20, 95% CI: 0.08-0.50). Subgroup analysis showed no statistical difference between olaparib and niraparib. Additionally, there was no PFS difference based on BRCA mutation status within both PARP inhibitor groups. CONCLUSION Our retrospective study demonstrates that PARP inhibitor maintenance therapy, including olaparib and niraparib, significantly prolongs PFS in patients with PSR epithelial ovarian, tubal, or primary peritoneal cancer, These findings support the broad utilization of PARP inhibitors as a standard maintenance therapy for PSR epithelial ovarian cancer irrespective of BRCA mutation status.
Collapse
Affiliation(s)
- Yumei Zhou
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
22
|
Bergstrom EN, Abbasi A, Díaz-Gay M, Galland L, Ladoire S, Lippman SM, Alexandrov LB. Deep Learning Artificial Intelligence Predicts Homologous Recombination Deficiency and Platinum Response From Histologic Slides. J Clin Oncol 2024; 42:3550-3560. [PMID: 39083703 PMCID: PMC11469627 DOI: 10.1200/jco.23.02641] [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: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/28/2024] [Indexed: 08/02/2024] Open
Abstract
PURPOSE Cancers with homologous recombination deficiency (HRD) can benefit from platinum salts and poly(ADP-ribose) polymerase inhibitors. Standard diagnostic tests for detecting HRD require molecular profiling, which is not universally available. METHODS We trained DeepHRD, a deep learning platform for predicting HRD from hematoxylin and eosin (H&E)-stained histopathological slides, using primary breast (n = 1,008) and ovarian (n = 459) cancers from The Cancer Genome Atlas (TCGA). DeepHRD was compared with four standard HRD molecular tests using breast (n = 349) and ovarian (n = 141) cancers from multiple independent data sets, including platinum-treated clinical cohorts with RECIST progression-free survival (PFS), complete response (CR), and overall survival (OS) endpoints. RESULTS DeepHRD predicted HRD from held-out H&E-stained breast cancer slides in TCGA with an AUC of 0.81 (95% CI, 0.77 to 0.85). This performance was confirmed in two independent primary breast cancer cohorts (AUC, 0.76 [95% CI, 0.71 to 0.82]). In an external platinum-treated metastatic breast cancer cohort, samples predicted as HRD had higher complete CR (AUC, 0.76 [95% CI, 0.54 to 0.93]) with 3.7-fold increase in median PFS (14.4 v 3.9 months; P = .0019) and hazard ratio (HR) of 0.45 (P = .0047). There were no significant differences in nonplatinum treatment outcome by predicted HRD status in three breast cancer cohorts, including CR (AUC, 0.39) and PFS (HR, 0.98, P = .95) in taxane-treated metastatic breast cancer. Through transfer learning to high-grade serous ovarian cancer, DeepHRD-predicted HRD samples had better OS after first-line (HR, 0.46; P = .030) and neoadjuvant (HR, 0.49; P = .015) platinum therapy in two cohorts. CONCLUSION DeepHRD can predict HRD in breast and ovarian cancers directly from routine H&E slides across multiple external cohorts, slide scanners, and tissue fixation variables. When compared with molecular testing, DeepHRD classified 1.8- to 3.1-fold more patients with HRD, which exhibited better OS in high-grade serous ovarian cancer and platinum-specific PFS in metastatic breast cancer.
Collapse
Affiliation(s)
- Erik N. Bergstrom
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Ammal Abbasi
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Marcos Díaz-Gay
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Loïck Galland
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
- Platform of Transfer in Biological Oncology, Centre Georges-François Leclerc, Dijon, France
- University of Burgundy-Franche Comté, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
- Platform of Transfer in Biological Oncology, Centre Georges-François Leclerc, Dijon, France
- University of Burgundy-Franche Comté, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | | | - Ludmil B. Alexandrov
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA
| |
Collapse
|
23
|
Belli C, Boscolo Bielo L, Repetto M, Crimini E, Scalia R, Diana A, Orefice J, Ascione L, Pellizzari G, Fusco N, Barberis M, Daniele B, Guerini-Rocco E, Curigliano G. Deleterious alterations in homologous recombination repair genes and efficacy of platinum-based chemotherapy in biliary tract cancers. Oncologist 2024; 29:707-715. [PMID: 38823036 PMCID: PMC11299956 DOI: 10.1093/oncolo/oyae125] [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: 01/09/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Platinum-based chemotherapy represents the standard first-line treatment for biliary tract cancers (BTC). Deficits in genes involved in the homologous recombination (HR) and DNA damage response (DDR) may confer higher sensitivity to platinum agents. METHODS We retrospectively included patients affected by BTC from 2 Italian institutions. Inclusion criteria consist of the receipt of platinum-based chemotherapy in the metastatic setting and the availability of comprehensive genomic profiling using next-generation sequencing (NGS). Patients were included in the HRD-like group if demonstrated oncogenic or likely oncogenic alterations in HR-/DDR-genes. Clinical endpoints were compared between the HRD-like group and the non-HRD-like group. RESULTS Seventy-four patients were included, of whom 25 (33%) in the HRD-like group and 49 (66%) in the non-HRD group. With a median follow-up of 26.04 months (interquartile-range [IQR] 9.41-29.27) in the HRD-like group and of 22.48 months (IQR 16.86-40.53) in the non-HRD group, no PFS difference emerged, with a mPFS of 5.18 months in the HRD-like group compared to 6.04 months in the non-HRD group (hazard ratio [HR], 1.017, 95% CI 0.58-1.78; P = .95). No differences were observed in DCR (64% [95 CI 45%-83%] vs 73% [95 CI 61%-86%]; P = .4), and CBR (45% [95% CI 28%-73%] vs 50% [95% CI, 37%-68%]; P = .9) between the HRD-like group and non-HRD groups, respectively. Median OS did not statistically differ between the HRD-like group and non-HRD group (26.7 vs 18.0 months, respectively; HR, 0.670, 0.33 to 1.37, P = .27). CONCLUSION HR-/DDR-genes, when assessed with regular tumor-only NGS panels, provide limited clinical validity to identify patients with BTC more likely to benefit from platinum-based chemotherapy.
Collapse
Affiliation(s)
- Carmen Belli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
| | - Matteo Repetto
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
- Early Drug Development Service, Memorial Sloan-Kettering Cancer Center, New York 10065, United States
| | - Edoardo Crimini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
| | - Raimondo Scalia
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
| | - Anna Diana
- Medical Oncology Unit, Ospedale del Mare, Naples 80147, Italy
| | - Jessica Orefice
- Medical Oncology Unit, Ospedale del Mare, Naples 80147, Italy
| | - Liliana Ascione
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
| | - Gloria Pellizzari
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Bruno Daniele
- Medical Oncology Unit, Ospedale del Mare, Naples 80147, Italy
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan 20122, Italy
| |
Collapse
|
24
|
Zhang H, Huang W, Chen M, Liu Y, Yan B, Mou S, Jiang W, Mei H. Research on molecular characteristics of ADME-related genes in kidney renal clear cell carcinoma. Sci Rep 2024; 14:16834. [PMID: 39039118 PMCID: PMC11263354 DOI: 10.1038/s41598-024-67516-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024] Open
Abstract
Genes involved in drug absorption, distribution, metabolism, and excretion (ADME) are named ADME genes. However, the comprehensive role of ADME genes in kidney renal clear cell carcinoma (KIRC) remains unclear. Using the clinical and gene expression data of KIRC patients downloaded from The Cancer Genome Atlas (TCGA), ArrayExpress, and the Gene Expression Omnibus (GEO) databases, we cluster patients into two patterns, and the population with a relatively poor prognosis demonstrated higher level of immunosuppressive cell infiltration and higher proportion of glycolytic subtypes. Then, 17 ADME genes combination identified through the least absolute shrinkage and selection operator algorithm (LASSO, 1000 times) was utilized to calculate the ADME score. The ADME score was found to be an independent predictor of prognosis in KIRC and to be tightly associated with the infiltration level of immune cells, metabolic properties, tumor-related signaling pathways, genetic variation, and responses to chemotherapeutics. Our work revealed the characteristics of ADME in KIRC. Assessing the ADME profiles of individual patients can deepen our comprehension of tumor microenvironment (TME) features in KIRC and can aid in developing more personalized and effective therapeutic strategies.
Collapse
Affiliation(s)
- Haiyu Zhang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weisheng Huang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mutong Chen
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuhan Liu
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bing Yan
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shuanzhu Mou
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wendong Jiang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
- Department of Urology, Shantou University Medical College, Shantou, China.
- Shenzhen Second People's Hospital, Clinical Medicine College of Anhui Medical University, Shenzhen, China.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
25
|
Pan JN, Li PC, Wang M, Li MW, Ding XW, Zhou T, Wang HN, Wang YK, Chen LB, Wang R, Ye WW, Wu WZ, Lou F, Wang XJ, Cao WM. AcornHRD: an HRD algorithm highly associated with anthracycline-based neoadjuvant chemotherapy in breast cancer in China. Eur J Med Res 2024; 29:366. [PMID: 39014466 PMCID: PMC11253386 DOI: 10.1186/s40001-024-01936-y] [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: 12/16/2023] [Accepted: 06/11/2024] [Indexed: 07/18/2024] Open
Abstract
PURPOSE Our study aimed to develop and validate a homologous recombination deficiency (HRD) scoring algorithm in the Chinese breast cancer population. METHODS AND MATERIALS Ninety-six in-house breast cancer (BC) samples and 6 HRD-positive standard cells were analyzed by whole-genome sequencing (WGS). Besides, 122 BCs from the TCGA database were down-sampled to ~ 1X WGS. We constructed an algorithm named AcornHRD for HRD score calculated based on WGS at low coverage as input data to estimate large-scale copy number alteration (LCNA) events on the genome. A clinical cohort of 50 BCs (15 cases carrying BRCA mutation) was used to assess the association between HRD status and anthracyclines-based neoadjuvant treatment outcomes. RESULTS A 100-kb window was defined as the optimal size using 41 in-house cases and the TCGA dataset. HRD score high threshold was determined as HRD score ≥ 10 using 55 in-house BCs with BRCA mutation to achieve a 95% BRCA-positive agreement rate. Furthermore, the HRD status agreement rate of AcornHRD is 100%, while the ShallowHRD is 60% in standard cells. BRCA mutation was significantly associated with a high HRD score evaluated by AcornHRD and ShallowHRD (p = 0.008 and p = 0.003, respectively) in the TCGA dataset. However, AcornHRD showed a higher positive agreement rate than did the ShallowHRD algorithm (70% vs 60%). In addition, the BRCA-positive agreement rate of AcornHRD was superior to that of ShallowHRD (87% vs 13%) in the clinical cohort. Importantly, the high HRD score assessed by AcornHRD was significantly correlated with a residual cancer burden score of 0 or 1 (RCB0/1). Besides, the HRD-positive group was more likely to respond to anthracycline-based chemotherapy than the HRD-negative group (pCR [OR = 9.5, 95% CI 1.11-81.5, p = 0.040] and RCB0/1 [OR = 10.29, 95% CI 2.02-52.36, p = 0.005]). CONCLUSION Using the AcornHRD algorithm evaluation, our analysis demonstrated the high performance of the LCNA genomic signature for HRD detection in breast cancers.
Collapse
Affiliation(s)
- Jia-Ni Pan
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310018, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, 999078, SAR, China
| | - Pu-Chun Li
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Meng Wang
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China
| | - Ming-Wei Li
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China
| | - Xiao-Wen Ding
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Tao Zhou
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China
| | - Hui-Na Wang
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China
| | - Yun-Kai Wang
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China
| | - Li-Bin Chen
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China
| | - Rong Wang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Wei-Wu Ye
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Wei-Zhu Wu
- Lihuili Hospital of Ningbo Medical Center, Ningbo, 315040, China
| | - Feng Lou
- AcornMed Biotechnology Co., Ltd., Floor 18, Block 5, Yard 18, Kechuang 13 RD, Beijing, 100176, China.
| | - Xiao-Jia Wang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
26
|
Chen C, Dettman EJ, Zhou W, Gozman A, Jin F, Lee LC, Ren Y, Zhou H, Cristescu R, Shao C. Prevalence of homologous recombination biomarkers in multiple tumor types: an observational study. Future Oncol 2024; 20:2357-2370. [PMID: 39011875 PMCID: PMC11520556 DOI: 10.1080/14796694.2024.2367957] [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: 12/15/2023] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
Aim: To determine the prevalence of deleterious mutations in BRCA1 and BRCA2 and in 13 genes involved in homologous recombination repair (HRR), the prevalence of genomic loss of heterozygosity and the allelic and hereditary status of BRCA1, BRCA2 and other HRR gene mutations in multiple solid tumor types.Patients & methods: This was a retrospective observational study of patients with an advanced/metastatic diagnosis in one of 15 solid tumor types, who were identified in a real-world clinico-genomic database.Results: Tumor tissue samples from 9457 patients were analyzed, among which 4.7% had known or suspected deleterious BRCA1/2 mutations. The prevalence (range) of mutations in HRR genes was 13.6% (2.4%-26.0%) and genomic loss of heterozygosity ≥16% was 20.6% (2.6-34.4%) across all tumor types.Conclusion: The prevalence of mutations varied significantly depending on the type of tumor.
Collapse
Affiliation(s)
- Cai Chen
- Merck & Co., Inc., Rahway, NJ07065, USA
| | | | - Wei Zhou
- Merck & Co., Inc., Rahway, NJ07065, USA
| | | | - Fan Jin
- Merck & Co., Inc., Rahway, NJ07065, USA
| | | | - Yixin Ren
- Merck & Co., Inc., Rahway, NJ07065, USA
| | - Heng Zhou
- Merck & Co., Inc., Rahway, NJ07065, USA
| | | | | |
Collapse
|
27
|
Abbasi A, Steele CD, Bergstrom EN, Khandekar A, Farswan A, Mckay RR, Pillay N, Alexandrov LB. Detecting HRD in whole-genome and whole-exome sequenced breast and ovarian cancers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.14.24310383. [PMID: 39040162 PMCID: PMC11261949 DOI: 10.1101/2024.07.14.24310383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Breast and ovarian cancers harboring homologous recombination deficiency (HRD) are sensitive to PARP inhibitors and platinum chemotherapy. Conventionally, detecting HRD involves screening for defects in BRCA1, BRCA2, and other relevant genes. Recent analyses have shown that HRD cancers exhibit characteristic mutational patterns due to the activities of HRD-associated mutational signatures. At least three machine learning tools exist for detecting HRD based on mutational patterns. Here, using sequencing data from 1,043 breast and 182 ovarian cancers, we trained Homologous Recombination Proficiency Profiler (HRProfiler), a machine learning method for detecting HRD using six mutational features. HRProfiler's performance is assessed against prior approaches using additional independent datasets of 417 breast and 115 ovarian cancers, including retrospective data from a clinical trial involving patients treated with PARP inhibitors. Our results demonstrate that HRProfiler is the only tool that robustly and consistently predicts clinical response from whole-exome sequenced breast and ovarian cancers.
Collapse
Affiliation(s)
- Ammal Abbasi
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Bioinformatics and Systems Biology Graduate Program, UC San Diego, La Jolla, CA, 92093, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92037
| | - Christopher D. Steele
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
| | - Erik N. Bergstrom
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
| | - Azhar Khandekar
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Bioinformatics and Systems Biology Graduate Program, UC San Diego, La Jolla, CA, 92093, USA
| | - Akanksha Farswan
- Research Department of Pathology, Cancer Institute, University College London, London, WC1E 6BT, UK
- Department of Cellular and Molecular Pathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, HA7 4LP, UK
| | - Rana R. Mckay
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
| | - Nischalan Pillay
- Research Department of Pathology, Cancer Institute, University College London, London, WC1E 6BT, UK
- Department of Cellular and Molecular Pathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, HA7 4LP, UK
| | - Ludmil B. Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Bioinformatics and Systems Biology Graduate Program, UC San Diego, La Jolla, CA, 92093, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
28
|
Wang Z, Li A, Lu Y, Han M, Ruan M, Wang C, Zhang X, Zhu C, Shen K, Dong L, Chen X. Association of tumor immune infiltration and prognosis with homologous recombination repair genes mutations in early triple-negative breast cancer. Front Immunol 2024; 15:1407837. [PMID: 39026672 PMCID: PMC11254810 DOI: 10.3389/fimmu.2024.1407837] [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: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
The aim of this study was to evaluate the mutation spectrum of homologous recombination repair (HRR) genes and its association with tumor immune infiltration and prognosis in triple-negative breast cancer (TNBC). TNBC patients (434 patients from Ruijin cohort) were evaluated with targeted next-generating sequencing for mutations in HRR genes. The frequencies of mutations were compared with public reference cohorts (320 TNBC patients from METABRIC, 105 from TCGA, and 225 from MSKCC 2018). Associations between mutation status and tumor immune infiltration and prognosis were analyzed. HRR genes mutations were seen in 21.89% patients, with BRCA1/2 mutations significantly enriched in tumors with breast/ovarian cancer family history (P = 0.025) and high Ki-67 levels (P = 0.018). HRR genes mutations were not related with recurrence-free survival (RFS) (adjusted P = 0.070) and overall survival (OS) (adjusted P = 0.318) for TNBC patients, regardless of carboplatin treatment (P > 0.05). Moreover, tumor immune infiltration and PD-L1 expression was positively associated with HRR or BRCA1/2 mutation (all P < 0.001). Patients with both HRR mutation and high CD8+ T cell counts had the best RFS and OS, whereas patients with no HRR mutation and low CD8+ T cell counts had the worst outcomes (RFS P < 0.001, OS P = 0.019). High frequency of HRR gene mutations was found in early TNBC, with no prognostic significance. Immune infiltration and PD-L1 expression was positively associated with HRR mutation, and both HRR mutation and high CD8+ T cell infiltration levels were associated with superior disease outcome.
Collapse
Affiliation(s)
- Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anqi Li
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujie Lu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyuan Han
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Ruan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaotian Zhang
- Department of Translational Oncology, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Changbin Zhu
- Department of Translational Oncology, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Dong
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Abel J, Jain S, Rajan D, Padigela H, Leidal K, Prakash A, Conway J, Nercessian M, Kirkup C, Javed SA, Biju R, Harguindeguy N, Shenker D, Indorf N, Sanghavi D, Egger R, Trotter B, Gerardin Y, Brosnan-Cashman JA, Dhoot A, Montalto MC, Parmar C, Wapinski I, Khosla A, Drage MG, Yu L, Taylor-Weiner A. AI powered quantification of nuclear morphology in cancers enables prediction of genome instability and prognosis. NPJ Precis Oncol 2024; 8:134. [PMID: 38898127 PMCID: PMC11187064 DOI: 10.1038/s41698-024-00623-9] [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: 08/03/2023] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
While alterations in nucleus size, shape, and color are ubiquitous in cancer, comprehensive quantification of nuclear morphology across a whole-slide histologic image remains a challenge. Here, we describe the development of a pan-tissue, deep learning-based digital pathology pipeline for exhaustive nucleus detection, segmentation, and classification and the utility of this pipeline for nuclear morphologic biomarker discovery. Manually-collected nucleus annotations were used to train an object detection and segmentation model for identifying nuclei, which was deployed to segment nuclei in H&E-stained slides from the BRCA, LUAD, and PRAD TCGA cohorts. Interpretable features describing the shape, size, color, and texture of each nucleus were extracted from segmented nuclei and compared to measurements of genomic instability, gene expression, and prognosis. The nuclear segmentation and classification model trained herein performed comparably to previously reported models. Features extracted from the model revealed differences sufficient to distinguish between BRCA, LUAD, and PRAD. Furthermore, cancer cell nuclear area was associated with increased aneuploidy score and homologous recombination deficiency. In BRCA, increased fibroblast nuclear area was indicative of poor progression-free and overall survival and was associated with gene expression signatures related to extracellular matrix remodeling and anti-tumor immunity. Thus, we developed a powerful pan-tissue approach for nucleus segmentation and featurization, enabling the construction of predictive models and the identification of features linking nuclear morphology with clinically-relevant prognostic biomarkers across multiple cancer types.
Collapse
|
30
|
Stiegeler N, Garsed DW, Au-Yeung G, Bowtell DDL, Heinzelmann-Schwarz V, Zwimpfer TA. Homologous recombination proficient subtypes of high-grade serous ovarian cancer: treatment options for a poor prognosis group. Front Oncol 2024; 14:1387281. [PMID: 38894867 PMCID: PMC11183307 DOI: 10.3389/fonc.2024.1387281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Approximately 50% of tubo-ovarian high-grade serous carcinomas (HGSCs) have functional homologous recombination-mediated (HR) DNA repair, so-called HR-proficient tumors, which are often associated with primary platinum resistance (relapse within six months after completion of first-line therapy), minimal benefit from poly(ADP-ribose) polymerase (PARP) inhibitors, and shorter survival. HR-proficient tumors comprise multiple molecular subtypes including cases with CCNE1 amplification, AKT2 amplification or CDK12 alteration, and are often characterized as "cold" tumors with fewer infiltrating lymphocytes and decreased expression of PD-1/PD-L1. Several new treatment approaches aim to manipulate these negative prognostic features and render HR-proficient tumors more susceptible to treatment. Alterations in multiple different molecules and pathways in the DNA damage response are driving new drug development to target HR-proficient cancer cells, such as inhibitors of the CDK or P13K/AKT pathways, as well as ATR inhibitors. Treatment combinations with chemotherapy or PARP inhibitors and agents targeting DNA replication stress have shown promising preclinical and clinical results. New approaches in immunotherapy are also being explored, including vaccines or antibody drug conjugates. Many approaches are still in the early stages of development and further clinical trials will determine their clinical relevance. There is a need to include HR-proficient tumors in ovarian cancer trials and to analyze them in a more targeted manner to provide further evidence for their specific therapy, as this will be crucial in improving the overall prognosis of HGSC and ovarian cancer in general.
Collapse
Affiliation(s)
| | - Dale W. Garsed
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - George Au-Yeung
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - David D. L. Bowtell
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Tibor A. Zwimpfer
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Gynecological Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
31
|
Ball M, Ourailidis I, Kluck K, Menzel M, Kirchner M, Allgäuer M, Tay TKY, Schnecko F, Volckmar AL, Goldschmid H, Neuman O, Fröhling S, Schirmacher P, Budczies J, Stenzinger A, Kazdal D. Leveraging Off-Target Reads in Panel Sequencing for Homologous Recombination Repair Deficiency Screening in Tumor. J Mol Diagn 2024; 26:479-486. [PMID: 38522840 PMCID: PMC12178388 DOI: 10.1016/j.jmoldx.2024.02.008] [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: 09/22/2023] [Revised: 01/24/2024] [Accepted: 02/14/2024] [Indexed: 03/26/2024] Open
Abstract
Targeted tumor only sequencing has become a standard practice in cancer diagnostics. This study aims to develop an approach for robust copy number variant calling in tumor samples using only off-target region (OTR) reads. We also established a clinical use case for homologous recombination deficiency (HRD) score estimation (HRDest) using the sum of telomeric-allelic imbalance and large-scale state transition scores without the need for loss of heterozygosity information. A strong correlation was found between HRD score and the sum of telomeric-allelic imbalance + large-scale state transition in The Cancer Genome Atlas cohort (ρ = 0.99, P < 2.2 × 10-16) and in a clinical in-house cohort of 34 tumors (ρ = 0.9, P = 5.1 × 10-13) comparing whole-exome sequencing and targeted sequencing data. HRDest scores from 1086 clinical cases were compared with The Cancer Genome Atlas data set. There were no significant differences in HRD score distribution within the analyzed tumor types. As a control, commercially available HRD standards were also sequenced, and the HRDest scores obtained from the OTR reads were well within the HRD reference range provided by the manufacturer. In conclusion, OTR reads of tumor-only panel sequencing can be used to determine genome-wide copy number variant profiles and to approximate HRD scores.
Collapse
Affiliation(s)
- Markus Ball
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Klaus Kluck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Menzel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martina Kirchner
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Allgäuer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Timothy Kwang Yong Tay
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Department of Anatomical Pathology, Singapore General Hospital, Singapore; Department of Molecular Pathology, Singapore General Hospital, Singapore
| | - Fabian Schnecko
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hannah Goldschmid
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Olaf Neuman
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine Heidelberg, Heidelberg, Germany
| | - Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium, Heidelberg, Germany; Center for Personalized Medicine Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium, Heidelberg, Germany; Center for Personalized Medicine Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany.
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium, Heidelberg, Germany; Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany.
| |
Collapse
|
32
|
Zhang X, Joseph S, Wu D, Bowser JL, Vaziri C. The DNA Damage Response (DDR) landscape of endometrial cancer defines discrete disease subtypes and reveals therapeutic opportunities. NAR Cancer 2024; 6:zcae015. [PMID: 38596432 PMCID: PMC11000323 DOI: 10.1093/narcan/zcae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/12/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
Genome maintenance is an enabling characteristic that allows neoplastic cells to tolerate the inherent stresses of tumorigenesis and evade therapy-induced genotoxicity. Neoplastic cells also deploy many mis-expressed germ cell proteins termed Cancer Testes Antigens (CTAs) to promote genome maintenance and survival. Here, we present the first comprehensive characterization of the DNA Damage Response (DDR) and CTA transcriptional landscapes of endometrial cancer in relation to conventional histological and molecular subtypes. We show endometrial serous carcinoma (ESC), an aggressive endometrial cancer subtype, is defined by gene expression signatures comprising members of the Replication Fork Protection Complex (RFPC) and Fanconi Anemia (FA) pathway and CTAs with mitotic functions. DDR and CTA-based profiling also defines a subset of highly aggressive endometrioid endometrial carcinomas (EEC) with poor clinical outcomes that share similar profiles to ESC yet have distinct characteristics based on conventional histological and genomic features. Using an unbiased CRISPR-based genetic screen and a candidate gene approach, we confirm that DDR and CTA genes that constitute the ESC and related EEC gene signatures are required for proliferation and therapy-resistance of cultured endometrial cancer cells. Our study validates the use of DDR and CTA-based tumor classifiers and reveals new vulnerabilities of aggressive endometrial cancer where none currently exist.
Collapse
Affiliation(s)
- Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
| | - Sayali Joseph
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
| | - Di Wu
- Department of Biostatistics, University of North Carolina at Chapel Hill, School of Dentistry, Chapel Hill, NC - 27599, USA
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC - 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC - 27599, USA
| |
Collapse
|
33
|
Wu Y, Chen R, Ni S, Hu K. Biomimetic "nano-spears" for CAFs-targeting: splintered three "shields" with enhanced cisplatin anti-TNBC efficiency. J Control Release 2024; 370:556-569. [PMID: 38697316 DOI: 10.1016/j.jconrel.2024.04.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
The treatment dilemma of triple-negative breast cancer (TNBC) revolves around drug resistance and metastasis. Cancer-associated fibroblasts (CAFs) contribute to cisplatin (Cis) resistance and further metastasis in TNBC, making TNBC a difficult-to-treat disease. The dense stromal barrier which restricts drug delivery, invasive phenotype of tumor cells, and immunosuppressive tumor microenvironment (TME) induced by CAFs serve as three "shields" for TNBC against Cis therapy. Here, we designed a silybin-loaded biomimetic nanoparticle coated with anisamide-modified red blood cell membrane (ARm@SNP) as a "nanospear" for CAFs-targeting, which could shatter the "shields" and significantly exhibit inhibitory effect on 4T1 cells in combination with Cis both in vitro and in vivo. The ARm@SNP/Cis elicited 4T1 tumor growth arrest and destroyed three "shields" as follows: disintegrating the stromal barrier by inhibiting blood vessels growth and the expression of fibronectin; decreasing 4T1 cell invasion and metastasis by affecting the TGF-β/Twist/EMT pathway which impeded EMT activation; reversing the immunosuppressive microenvironment by increasing the activity and infiltration of immunocompetent cells. Based on CAFs-targeting, ARm@SNP reversed the resistance of Cis, remodeled the TME and inhibited invasion and metastasis while significantly improving the therapeutic effect of Cis on 4T1 tumor-bearing mice, providing a promising approach for treating intractable TNBC.
Collapse
Affiliation(s)
- Yufan Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rujing Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
34
|
Lee M, Yoo TK, Chae BJ, Lee A, Cha YJ, Lee J, Ahn SG, Kang J. Luminal androgen receptor subtype and tumor-infiltrating lymphocytes groups based on triple-negative breast cancer molecular subclassification. Sci Rep 2024; 14:11278. [PMID: 38760384 PMCID: PMC11101432 DOI: 10.1038/s41598-024-61640-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 05/08/2024] [Indexed: 05/19/2024] Open
Abstract
In our previous study, we developed a triple-negative breast cancer (TNBC) subtype classification that correlated with the TNBC molecular subclassification. In this study, we aimed to evaluate the predictor variables of this subtype classification on the whole slide and to validate the model's performance by using an external test set. We explored the characteristics of this subtype classification and investigated genomic alterations, including genomic scar signature scores. First, TNBC was classified into the luminal androgen receptor (LAR) and non-luminal androgen receptor (non-LAR) subtypes based on the AR Allred score (≥ 6 and < 6, respectively). Then, the non-LAR subtype was further classified into the lymphocyte-predominant (LP), lymphocyte-intermediate (LI), and lymphocyte-depleted (LD) groups based on stromal tumor-infiltrating lymphocytes (TILs) (< 20%, > 20% but < 60%, and ≥ 60%, respectively). This classification showed fair agreement with the molecular classification in the test set. The LAR subtype was characterized by a high rate of PIK3CA mutation, CD274 (encodes PD-L1) and PDCD1LG2 (encodes PD-L2) deletion, and a low homologous recombination deficiency (HRD) score. The non-LAR LD TIL group was characterized by a high frequency of NOTCH2 and MYC amplification and a high HRD score.
Collapse
Affiliation(s)
- Miseon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tae-Kyung Yoo
- Division of Breast Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byung Joo Chae
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jieun Lee
- Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Gwe Ahn
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
35
|
D'Agostino E, Mastrodomenico L, Ponzoni O, Baldessari C, Piombino C, Pipitone S, Giuseppa Vitale M, Sabbatini R, Dominici M, Toss A. Molecular characterization as new driver in prognostic signatures and therapeutic strategies for endometrial cancer. Cancer Treat Rev 2024; 126:102723. [PMID: 38555857 DOI: 10.1016/j.ctrv.2024.102723] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/28/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Endometrial cancer (EC) incidence and mortality rates have been increasing, particularly among young females. Although more than 90% of ECs are sporadic, 5-10% are hereditary, a majority of which occurs within Hereditary Non-Polyposis Colorectal Cancer syndrome (HNPCC) or Lynch syndrome. The traditional histopathological classification differentiates EC between two main groups: type I (or endometrioid) and type II (including all other histopathological subtypes). However, this classification lacks reproducibility and does not account for the emerging molecular heterogeneity. In 2013, The Cancer Genome Atlas (TCGA) project proposed EC molecular classification defining four groups with different prognostic and predictive values and the current international guidelines are progressively establishing EC risk stratification and treatment based on both histopathological and molecular criteria. Our manuscript aims to summarize the current state of EC molecular characterizations, including germline alterations at the basis of hereditary EC predisposition, to discuss their clinical utility as prognostic and predictive markers.
Collapse
Affiliation(s)
- Elisa D'Agostino
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| | - Luciana Mastrodomenico
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| | - Ornella Ponzoni
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| | - Cinzia Baldessari
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| | - Claudia Piombino
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| | - Stefania Pipitone
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Giuseppa Vitale
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| | - Roberto Sabbatini
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy.
| | - Massimo Dominici
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Angela Toss
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
36
|
Bertucci F, Lerebours F, Ceccarelli M, Guille A, Syed N, Finetti P, Adélaïde J, Van Laere S, Goncalves A, Viens P, Birnbaum D, Mamessier E, Callens C, Bedognetti D. Mutational landscape of inflammatory breast cancer. J Transl Med 2024; 22:374. [PMID: 38637846 PMCID: PMC11025259 DOI: 10.1186/s12967-024-05198-4] [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: 03/08/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Inflammatory breast cancer (IBC) is the most pro-metastatic form of BC. Better understanding of its enigmatic pathophysiology is crucial. We report here the largest whole-exome sequencing (WES) study of clinical IBC samples. METHODS We retrospectively applied WES to 54 untreated IBC primary tumor samples and matched normal DNA. The comparator samples were 102 stage-matched non-IBC samples from TCGA. We compared the somatic mutational profiles, spectra and signatures, copy number alterations (CNAs), HRD and heterogeneity scores, and frequencies of actionable genomic alterations (AGAs) between IBCs and non-IBCs. The comparisons were adjusted for the molecular subtypes. RESULTS The number of somatic mutations, TMB, and mutational spectra were not different between IBCs and non-IBCs, and no gene was differentially mutated or showed differential frequency of CNAs. Among the COSMIC signatures, only the age-related signature was more frequent in non-IBCs than in IBCs. We also identified in IBCs two new mutational signatures not associated with any environmental exposure, one of them having been previously related to HIF pathway activation. Overall, the HRD score was not different between both groups, but was higher in TN IBCs than TN non-IBCs. IBCs were less frequently classified as heterogeneous according to heterogeneity H-index than non-IBCs (21% vs 33%), and clonal mutations were more frequent and subclonal mutations less frequent in IBCs. More than 50% of patients with IBC harbored at least one high-level of evidence (LOE) AGA (OncoKB LOE 1-2, ESCAT LOE I-II), similarly to patients with non-IBC. CONCLUSIONS We provide the largest mutational landscape of IBC. Only a few subtle differences were identified with non-IBCs. The most clinically relevant one was the higher HRD score in TN IBCs than in TN non-IBCs, whereas the most intriguing one was the smaller intratumor heterogeneity of IBCs.
Collapse
Affiliation(s)
- François Bertucci
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France.
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France.
| | - Florence Lerebours
- Department of Medical Oncology, Institut Curie Saint-Cloud, Paris, France
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
- Department of Public Health Sciences, University of Miami, Miami, USA
| | - Arnaud Guille
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Najeeb Syed
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Pascal Finetti
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - José Adélaïde
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Anthony Goncalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Daniel Birnbaum
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Emilie Mamessier
- Département d'Oncologie Médicale, Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, 232, Boulevard de Sainte-Marguerite, 13009, Marseille, France
| | - Céline Callens
- Department of Medical Oncology, Institut Curie Saint-Cloud, Paris, France
| | - Davide Bedognetti
- Tumor Biology and Immunology Laboratory, Research Branch, Sidra Medicine, Doha, Qatar
| |
Collapse
|
37
|
Hijazo‐Pechero S, Alay A, Cordero D, Marín R, Vilariño N, Palmero R, Brenes J, Montalban‐Casafont A, Nadal E, Solé X. Transcriptional analysis of landmark molecular pathways in lung adenocarcinoma results in a clinically relevant classification with potential therapeutic implications. Mol Oncol 2024; 18:453-470. [PMID: 37943164 PMCID: PMC10850798 DOI: 10.1002/1878-0261.13550] [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: 05/09/2023] [Revised: 09/11/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a molecularly heterogeneous disease. In addition to genomic alterations, cancer transcriptional profiling can be helpful to tailor cancer treatment and to estimate each patient's outcome. Transcriptional activity levels of 50 molecular pathways were inferred in 4573 LUAD patients using Gene Set Variation Analysis (GSVA) method. Seven LUAD subtypes were defined and independently validated based on the combined behavior of the studied pathways: AD (adenocarcinoma subtype) 1-7. AD1, AD4, and AD5 subtypes were associated with better overall survival. AD1 and AD4 subtypes were enriched in epidermal growth factor receptor (EGFR) mutations, whereas AD2 and AD6 showed higher tumor protein p53 (TP53) alteration frequencies. AD2 and AD6 subtypes correlated with higher genome instability, proliferation-related pathway expression, and specific sensitivity to chemotherapy, based on data from LUAD cell lines. LUAD subtypes were able to predict immunotherapy response in addition to CD274 (PD-L1) gene expression and tumor mutational burden (TMB). AD2 and AD4 subtypes were associated with potential resistance and response to immunotherapy, respectively. Thus, analysis of transcriptomic data could improve patient stratification beyond genomics and single biomarkers (i.e., PD-L1 and TMB) and may lay the foundation for more personalized treatment avenues, especially in driver-negative LUAD.
Collapse
Affiliation(s)
- Sara Hijazo‐Pechero
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
- Translational Genomics and Targeted Therapies in Solid TumorsAugust Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Ania Alay
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
| | - David Cordero
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
| | - Raúl Marín
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
| | - Noelia Vilariño
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
- Thoracic Oncology Unit, Department of Medical Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
- Neuro‐Oncology Unit, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
| | - Ramón Palmero
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
- Thoracic Oncology Unit, Department of Medical Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
| | - Jesús Brenes
- Thoracic Oncology Unit, Department of Medical Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
| | - Aina Montalban‐Casafont
- Molecular Biology CORE, Center for Biomedical Diagnostics (CDB)Hospital Clínic de BarcelonaSpain
| | - Ernest Nadal
- Preclinical and Experimental Research in Thoracic Tumors (PrETT), Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de LlobregatBarcelonaSpain
- Thoracic Oncology Unit, Department of Medical Oncology, Catalan Institute of Oncology (ICO)L'Hospitalet de LlobregatBarcelonaSpain
| | - Xavier Solé
- Translational Genomics and Targeted Therapies in Solid TumorsAugust Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
- Molecular Biology CORE, Center for Biomedical Diagnostics (CDB)Hospital Clínic de BarcelonaSpain
| |
Collapse
|
38
|
Yang Y, Wu SF, Bao W. Molecular subtypes of endometrial cancer: Implications for adjuvant treatment strategies. Int J Gynaecol Obstet 2024; 164:436-459. [PMID: 37525501 DOI: 10.1002/ijgo.14969] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND When determining adjuvant treatment for endometrial cancer, the decision typically relies on factors such as cancer stage, histologic grade, subtype, and a few histopathologic markers. The Cancer Genome Atlas revealed molecular subtyping of endometrial cancer, which can provide more accurate prognostic information and guide personalized treatment plans. OBJECTIVE To summarize the expression and molecular basis of the main biomarkers of endometrial cancer. SEARCH STRATEGY PubMed was searched from January 2000 to March 2023. SELECTION CRITERIA Studies evaluating molecular subtypes of endometrial cancer and implications for adjuvant treatment strategies. DATA COLLECTION AND ANALYSIS Three authors independently performed a comprehensive literature search, collected and extracted data, and assessed the methodological quality of the included studies. MAIN RESULTS We summarized the molecular subtyping of endometrial cancer, including mismatch repair deficient, high microsatellite instability, polymerase epsilon (POLE) exonuclease domain mutated, TP53 gene mutation, and non-specific molecular spectrum. We also summarized planned and ongoing clinical trials and common therapy methods in endometrial cancer. POLE mutated endometrial cancer consistently exhibits favorable patient outcomes, regardless of adjuvant therapy. Genomic similarities between p53 abnormality endometrial cancer and high-grade serous ovarian cancer suggested possible overlapping treatment strategies. High levels of immune checkpoint molecules, such as programmed cell death 1 and programmed cell death 1 ligand 1 can counterbalance mismatch repair deficient endometrial cancer immune phenotype. Hormonal treatment is an appealing option for high-risk non-specific molecular spectrum endometrial cancers, which are typically endometrioid and hormone receptor positive. Combining clinical and pathologic characteristics to guide treatment decisions for patients, including concurrent radiochemotherapy, chemotherapy, inhibitor therapy, endocrine therapy, and immunotherapy, might improve the management of endometrial cancer and provide more effective treatment options for patients. CONCLUSIONS We have characterized the molecular subtypes of endometrial cancer and discuss their value in terms of a patient-tailored therapy in order to prevent significant under- or overtreatment.
Collapse
Affiliation(s)
- Ye Yang
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Su Fang Wu
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Wei Bao
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
39
|
Bruchim I, Capasso I, Polonsky A, Meisel S, Salutari V, Werner H, Lorusso D, Scambia G, Fanfani F. New therapeutic targets for endometrial cancer: a glimpse into the preclinical sphere. Expert Opin Ther Targets 2024; 28:29-43. [PMID: 38327111 DOI: 10.1080/14728222.2024.2316739] [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: 08/30/2023] [Accepted: 02/06/2024] [Indexed: 02/09/2024]
Abstract
INTRODUCTION Endometrial cancer (EC) is the only gynecologic malignancy showing increasing trends in incidence and mortality. While standard treatment has been effective primarily for early-stage EC, precision medicine with tailored therapy has revolutionized the management of this disease. Genome sequencing analyses have identified four sub-types of EC. Treatments for primary and metastatic disease can now be tailored more accurately to achieve better oncologic results. AREAS COVERED This review provides an overview of the most relevant and updated evidence in the literature regarding EC molecular analysis and its role in risk classification, prognostication, and guidance for tailored and target therapies in early and advanced/metastatic stages. In addition, it provides updated information on optimal surgical management based on molecular classification and highlights key advances and future strategies. EXPERT OPINION EC molecular analysis yields the potential of tailoring adjuvant treatment by escalating or deescalating therapy, as shown for POLE-mutated and p53-mutated tumors. Moreover, the expression of specific molecular signatures offers the possibility to employ novel target therapies, such as immune-checkpoint inhibitors that have demonstrated a significant benefit on prognosis. New treatment guidelines are still being established, and ongoing studies are exploring the potential prognostic role of further sub-stratifications of the four molecular classes and treatment options.
Collapse
Affiliation(s)
- Ilan Bruchim
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Technion, Institute of Technology, Haifa, Israel
| | - Ilaria Capasso
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ariel Polonsky
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Shilhav Meisel
- Gynecology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Technion, Institute of Technology, Haifa, Israel
| | - Vanda Salutari
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Haim Werner
- The Technion, Institute of Technology, Haifa, Israel
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Domenica Lorusso
- Università Cattolica del Sacro Cuore, Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Fanfani
- Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
40
|
van Wijk LM, Vermeulen S, Ter Haar NT, Kramer CJH, Terlouw D, Vrieling H, Cohen D, Vreeswijk MPG. Performance of a RAD51-based functional HRD test on paraffin-embedded breast cancer tissue. Breast Cancer Res Treat 2023; 202:607-616. [PMID: 37725154 PMCID: PMC10564840 DOI: 10.1007/s10549-023-07102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE BRCA-deficient breast cancers (BC) are highly sensitive to platinum-based chemotherapy and PARP inhibitors due to their deficiency in the homologous recombination (HR) pathway. However, HR deficiency (HRD) extends beyond BRCA-associated BC, highlighting the need for a sensitive method to enrich for HRD tumors in an alternative way. A promising approach is the use of functional HRD tests which evaluate the HR capability of tumor cells by measuring RAD51 protein accumulation at DNA damage sites. This study aims to evaluate the performance of a functional RAD51-based HRD test for the identification of HRD BC. METHODS The functional HR status of 63 diagnostic formalin-fixed paraffin-embedded (FFPE) BC samples was determined by applying the RAD51-FFPE test. Samples were screened for the presence of (epi)genetic defects in HR and matching tumor samples were analyzed with the RECAP test, which requires ex vivo irradiated fresh tumor tissue on the premise that the HRD status as determined by the RECAP test faithfully represented the functional HR status. RESULTS The RAD51-FFPE test identified 23 (37%) of the tumors as HRD, including three tumors with pathogenic variants in BRCA1/2. The RAD51-FFPE test showed a sensitivity of 88% and a specificity of 76% in determining the HR-class as defined by the RECAP test. CONCLUSION Given its high sensitivity and compatibility with FFPE samples, the RAD51-FFPE test holds great potential to enrich for HRD tumors, including those associated with BRCA-deficiency. This potential extends to situations where DNA-based testing may be challenging or not easily accessible in routine clinical practice. This is particularly important considering the potential implications for treatment decisions and patient stratification.
Collapse
Affiliation(s)
- Lise M van Wijk
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Sylvia Vermeulen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Natalja T Ter Haar
- Department of Pathology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Claire J H Kramer
- Department of Pathology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Diantha Terlouw
- Department of Pathology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Harry Vrieling
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Danielle Cohen
- Department of Pathology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Maaike P G Vreeswijk
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
41
|
Prosz A, Duan H, Tisza V, Sahgal P, Topka S, Klus GT, Börcsök J, Sztupinszki Z, Hanlon T, Diossy M, Vizkeleti L, Stormoen DR, Csabai I, Pappot H, Vijai J, Offit K, Ried T, Sethi N, Mouw KW, Spisak S, Pathania S, Szallasi Z. Nucleotide excision repair deficiency is a targetable therapeutic vulnerability in clear cell renal cell carcinoma. Sci Rep 2023; 13:20567. [PMID: 37996508 PMCID: PMC10667362 DOI: 10.1038/s41598-023-47946-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Due to a demonstrated lack of DNA repair deficiencies, clear cell renal cell carcinoma (ccRCC) has not benefitted from targeted synthetic lethality-based therapies. We investigated whether nucleotide excision repair (NER) deficiency is present in an identifiable subset of ccRCC cases that would render those tumors sensitive to therapy targeting this specific DNA repair pathway aberration. We used functional assays that detect UV-induced 6-4 pyrimidine-pyrimidone photoproducts to quantify NER deficiency in ccRCC cell lines. We also measured sensitivity to irofulven, an experimental cancer therapeutic agent that specifically targets cells with inactivated transcription-coupled nucleotide excision repair (TC-NER). In order to detect NER deficiency in clinical biopsies, we assessed whole exome sequencing data for the presence of an NER deficiency associated mutational signature previously identified in ERCC2 mutant bladder cancer. Functional assays showed NER deficiency in ccRCC cells. Some cell lines showed irofulven sensitivity at a concentration that is well tolerated by patients. Prostaglandin reductase 1 (PTGR1), which activates irofulven, was also associated with this sensitivity. Next generation sequencing data of the cell lines showed NER deficiency-associated mutational signatures. A significant subset of ccRCC patients had the same signature and high PTGR1 expression. ccRCC cell line-based analysis showed that NER deficiency is likely present in this cancer type. Approximately 10% of ccRCC patients in the TCGA cohort showed mutational signatures consistent with ERCC2 inactivation associated NER deficiency and also substantial levels of PTGR1 expression. These patients may be responsive to irofulven, a previously abandoned anticancer agent that has minimal activity in NER-proficient cells.
Collapse
Affiliation(s)
- Aurel Prosz
- Danish Cancer Institute, Copenhagen, Denmark
| | - Haohui Duan
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA, USA
- Department of Biology, University of Massachusetts, Boston, MA, USA
| | - Viktoria Tisza
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Pranshu Sahgal
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT), Harvard University, Cambridge, MA, USA
| | - Sabine Topka
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Niehaus Center for Inherited Cancer Genomics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gregory T Klus
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Judit Börcsök
- Danish Cancer Institute, Copenhagen, Denmark
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Zsofia Sztupinszki
- Danish Cancer Institute, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Timothy Hanlon
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Miklos Diossy
- Danish Cancer Institute, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Laura Vizkeleti
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
| | - Dag Rune Stormoen
- Department of Oncology, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Istvan Csabai
- Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | - Helle Pappot
- Department of Oncology, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Joseph Vijai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Niehaus Center for Inherited Cancer Genomics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering, New York, NY, USA
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Niehaus Center for Inherited Cancer Genomics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering, New York, NY, USA
| | - Thomas Ried
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Nilay Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT), Harvard University, Cambridge, MA, USA
| | - Kent W Mouw
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Department of Radiation Oncology, Brigham & Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sandor Spisak
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Shailja Pathania
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA, USA.
- Department of Biology, University of Massachusetts, Boston, MA, USA.
| | - Zoltan Szallasi
- Danish Cancer Institute, Copenhagen, Denmark.
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA.
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
42
|
McGrail DJ, Li Y, Smith RS, Feng B, Dai H, Hu L, Dennehey B, Awasthi S, Mendillo ML, Sood AK, Mills GB, Lin SY, Yi SS, Sahni N. Widespread BRCA1/2-independent homologous recombination defects are caused by alterations in RNA-binding proteins. Cell Rep Med 2023; 4:101255. [PMID: 37909041 PMCID: PMC10694618 DOI: 10.1016/j.xcrm.2023.101255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/02/2022] [Accepted: 09/29/2023] [Indexed: 11/02/2023]
Abstract
Defects in homologous recombination DNA repair (HRD) both predispose to cancer development and produce therapeutic vulnerabilities, making it critical to define the spectrum of genetic events that cause HRD. However, we found that mutations in BRCA1/2 and other canonical HR genes only identified 10%-20% of tumors that display genomic evidence of HRD. Using a networks-based approach, we discovered that over half of putative genes causing HRD originated outside of canonical DNA damage response genes, with a particular enrichment for RNA-binding protein (RBP)-encoding genes. These putative drivers of HRD were experimentally validated, cross-validated in an independent cohort, and enriched in cancer-associated genome-wide association study loci. Mechanistic studies indicate that some RBPs are recruited to sites of DNA damage to facilitate repair, whereas others control the expression of canonical HR genes. Overall, this study greatly expands the repertoire of known drivers of HRD, with implications for basic biology, genetic screening, and therapy stratification.
Collapse
Affiliation(s)
- Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH 44106, USA; Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Yang Li
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roger S Smith
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Bin Feng
- GSK Oncology Experimental Medicine Unit, Waltham, MA 02451, USA
| | - Hui Dai
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Limei Hu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Briana Dennehey
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sharad Awasthi
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marc L Mendillo
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Simpson Querrey Center for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gordon B Mills
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR 97201, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - S Stephen Yi
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Program in Quantitative and Computational Biosciences (QCB), Baylor College of Medicine, Houston, TX 77030, USA; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
43
|
Zhang X, Joseph S, Wu D, Bowser JL, Vaziri C. The DNA Damage Response (DDR) landscape of endometrial cancer defines discrete disease subtypes and reveals therapeutic opportunities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567919. [PMID: 38045328 PMCID: PMC10690150 DOI: 10.1101/2023.11.20.567919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Genome maintenance is an enabling characteristic that allows neoplastic cells to tolerate the inherent stresses of tumorigenesis and evade therapy-induced genotoxicity. Neoplastic cells also deploy mis-expressed germ cell proteins termed Cancer Testes Antigens (CTAs) to promote genome maintenance and survival. Here, we present the first comprehensive characterization of the DNA Damage Response (DDR) and CTA transcriptional landscapes of endometrial cancer in relation to conventional histological and molecular subtypes. We show endometrial serous carcinoma (ESC), an aggressive endometrial cancer subtype, is defined by gene expression signatures comprising members of the Replication Fork Protection Complex (RFPC) and Fanconi Anemia (FA) pathway and CTAs with mitotic functions. DDR and CTA- based profiling also defines a subset of highly aggressive endometrioid endometrial carcinomas (EEC) with poor clinical outcomes that share similar profiles to ESC yet have distinct characteristics based on conventional histological and genomic features. Using an unbiased CRISPR-based genetic screen and a candidate gene approach, we confirm that DDR and CTA genes that constitute the ESC and related EEC gene signatures are required for proliferation and therapy-resistance of cultured endometrial cancer cells. Our study validates the use of DDR and CTA-based tumor classifiers and reveals new vulnerabilities of aggressive endometrial cancer where none currently exist.
Collapse
|
44
|
Dong Q, Yu T, Chen B, Liu M, Sun X, Cao H, Liu K, Xu H, Wang Y, Zhuang S, Jin Z, Liang H, Hui Y, Gu Y. Mutant RB1 enhances therapeutic efficacy of PARPis in lung adenocarcinoma by triggering the cGAS/STING pathway. JCI Insight 2023; 8:e165268. [PMID: 37937640 PMCID: PMC10721263 DOI: 10.1172/jci.insight.165268] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/25/2023] [Indexed: 11/09/2023] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPis) are approved for cancer therapy according to their synthetic lethal interactions, and clinical trials have been applied in non-small cell lung cancer. However, the therapeutic efficacy of PARPis in lung adenocarcinoma (LUAD) is still unknown. We explored the effect of a mutated retinoblastoma gene (RB1) on PARPi sensitivity in LUAD. Bioinformatic screening was performed to identify PARPi-sensitive biomarkers. Here, we showed that viability of LUAD cell lines with mutated RB1 was significantly decreased by PARPis (niraparib, rucaparib, and olaparib). RB1 deficiency induced genomic instability, prompted cytosolic double-stranded DNA (dsDNA) formation, activated the cGAS/STING pathway, and upregulated downstream chemokines CCL5 and CXCL10, triggering immune cell infiltration. Xenograft experiments indicated that PARPi treatment reduced tumorigenesis in RB1-KO mice. Additionally, single-cell RNA sequencing analysis showed that malignant cells with downregulated expression of RB1 had more communications with other cell types, exhibiting activation of specific signaling such as GAS, IFN response, and antigen-presenting and cytokine activities. Our findings suggest that RB1 mutation mediates the sensitivity to PARPis through a synthetic lethal effect by triggering the cGAS/STING pathway and upregulation of immune infiltration in LUAD, which may be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Qi Dong
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Tong Yu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
| | - Bo Chen
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
| | - Mingyue Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
| | - Xiang Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Huiying Cao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Kaidong Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
| | - Huanhuan Xu
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
| | - Yuquan Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shuping Zhuang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zixin Jin
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, and
| |
Collapse
|
45
|
Jacobson DH, Pan S, Fisher J, Secrier M. Multi-scale characterisation of homologous recombination deficiency in breast cancer. Genome Med 2023; 15:90. [PMID: 37919776 PMCID: PMC10621207 DOI: 10.1186/s13073-023-01239-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Homologous recombination is a robust, broadly error-free mechanism of double-strand break repair, and deficiencies lead to PARP inhibitor sensitivity. Patients displaying homologous recombination deficiency can be identified using 'mutational signatures'. However, these patterns are difficult to reliably infer from exome sequencing. Additionally, as mutational signatures are a historical record of mutagenic processes, this limits their utility in describing the current status of a tumour. METHODS We apply two methods for characterising homologous recombination deficiency in breast cancer to explore the features and heterogeneity associated with this phenotype. We develop a likelihood-based method which leverages small insertions and deletions for high-confidence classification of homologous recombination deficiency for exome-sequenced breast cancers. We then use multinomial elastic net regression modelling to develop a transcriptional signature of heterogeneous homologous recombination deficiency. This signature is then applied to single-cell RNA-sequenced breast cancer cohorts enabling analysis of homologous recombination deficiency heterogeneity and differential patterns of tumour microenvironment interactivity. RESULTS We demonstrate that the inclusion of indel events, even at low levels, improves homologous recombination deficiency classification. Whilst BRCA-positive homologous recombination deficient samples display strong similarities to those harbouring BRCA1/2 defects, they appear to deviate in microenvironmental features such as hypoxic signalling. We then present a 228-gene transcriptional signature which simultaneously characterises homologous recombination deficiency and BRCA1/2-defect status, and is associated with PARP inhibitor response. Finally, we show that this signature is applicable to single-cell transcriptomics data and predict that these cells present a distinct milieu of interactions with their microenvironment compared to their homologous recombination proficient counterparts, typified by a decreased cancer cell response to TNFα signalling. CONCLUSIONS We apply multi-scale approaches to characterise homologous recombination deficiency in breast cancer through the development of mutational and transcriptional signatures. We demonstrate how indels can improve homologous recombination deficiency classification in exome-sequenced breast cancers. Additionally, we demonstrate the heterogeneity of homologous recombination deficiency, especially in relation to BRCA1/2-defect status, and show that indications of this feature can be captured at a single-cell level, enabling further investigations into interactions between DNA repair deficient cells and their tumour microenvironment.
Collapse
Affiliation(s)
- Daniel H Jacobson
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Shi Pan
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jasmin Fisher
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
46
|
Cussenot O, Cancel-Tassin G, Rao SR, Woodcock DJ, Lamb AD, Mills IG, Hamdy FC. Aligning germline and somatic mutations in prostate cancer. Are genetics changing practice? BJU Int 2023; 132:472-484. [PMID: 37410655 DOI: 10.1111/bju.16120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
OBJECTIVE To review the current status of germline and somatic (tumour) genetic testing for prostate cancer (PCa), and its relevance for clinical practice. METHODS A narrative synthesis of various molecular profiles related to their clinical context was carried out. Current guidelines for genetic testing and its feasibility in clinical practice were analysed. We report the main identified genetic sequencing results or functional genomic scores for PCa published in the literature or obtained from the French PROGENE study. RESULTS The molecular alterations observed in PCa are mostly linked to disruption of the androgen receptor (AR) pathway or DNA repair deficiency. The main known germline mutations affect the BReast CAncer gene 2 (BRCA2) and homeobox B13 (HOXB13) genes, whereas AR and tumour protein p53 (TP53) are the genes with most frequent somatic alterations in tumours from men with metastatic PCa. Molecular tests are now available for detecting some of these germline or somatic alterations and sometimes recommended by guidelines, but their utilisation must combine rationality and feasibility. They can guide specific therapies, notably for the management of metastatic disease. Indeed, following androgen deprivation, targeted therapies for PCa currently include poly-(ADP-ribose)-polymerase (PARP) inhibitors, immune checkpoint inhibitors, and prostate-specific membrane antigen (PSMA)-guided radiotherapy. The genetic tests currently approved for targeted therapies remain limited to the detection of BRCA1 and BRCA2 mutation and DNA mismatch repair deficiency, while large panels are recommended for germline analyses, not only for inherited cancer predisposing syndrome, but also for metastatic PCa. CONCLUSIONS Further consensus aligning germline with somatic molecular analysis in metastatic PCa is required, including genomics scars, emergent immunohistochemistry, or functional pre-screen imaging. With rapid advances in knowledge and technology in the field, continuous updating of guidelines to help the clinical management of these individuals, and well-conducted studies to evaluate the benefits of genetic testing are needed.
Collapse
Affiliation(s)
- Olivier Cussenot
- Centre de Recherche sur les Pathologies Prostatiques et Urologiques (CeRePP), Paris, France
- GRC 5 Predictive Onco-Urology, Sorbonne University, Paris, France
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Geraldine Cancel-Tassin
- Centre de Recherche sur les Pathologies Prostatiques et Urologiques (CeRePP), Paris, France
- GRC 5 Predictive Onco-Urology, Sorbonne University, Paris, France
| | - Srinivasa R Rao
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Dan J Woodcock
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
47
|
Feng J, Lan Y, Liu F, Yuan Y, Ge J, Wei S, Luo H, Li J, Luo T, Bian X. Combination of genomic instability score and TP53 status for prognosis prediction in lung adenocarcinoma. NPJ Precis Oncol 2023; 7:110. [PMID: 37907595 PMCID: PMC10618567 DOI: 10.1038/s41698-023-00465-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023] Open
Abstract
The genomic instability (GI) /homologous recombination deficiency (HRD) score, calculated as the sum of the events of loss of heterozygosity (LOH), large-scale state transition (LST) and telomere allele imbalance (TAI), is used to guide the choice of treatment in several cancers, but its relationship with genomic features, clinicopathological characteristics and prognosis in lung cancer is poorly understood, which could lead to population bias in prospective studies. We retrospectively analyzed 1011 lung cancer patients whose tumor samples were successfully profiled by high-throughput sequencing panel including GI/HRD score. Alterations of many cancer suppressor genes were associated with higher GI/HRD scores, biallelic inactivation of TP53 was correlated with a high GI/HRD score. A combination of two gene alterations exhibited a higher GI/HRD scores than single gene alterations. The GI/HRD score was associated with advanced stages in lung adenocarcinoma but not in lung squamous cell carcinoma. Furthermore, patients with higher GI/HRD scores had significantly shorter overall survival and progression-free survival than patients with lower GI/HRD scores. Finally, patients with a combination of a higher GI/HRD scores and TP53 alteration exhibited an extremely poor prognosis compared with patients with a lower GI/HRD scores and wild-type TP53 (overall survival, training cohort, hazard ratio (HR) = 8.56, P < 0.001; validation cohort, HR = 6.47, P < 0.001; progression-free survival, HR = 4.76, P < 0.001). Our study revealed the prognostic value of the GI/HRD score in lung adenocarcinoma, but not for all lung cancer. Moreover, the combination of the GI/HRD score and TP53 status could be a promising strategy to predict the prognosis of patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Juan Feng
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China
| | - Yang Lan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China
| | - Feng Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China
| | - Ye Yuan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China
| | - Jia Ge
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China
| | - Sen Wei
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China
| | - Hu Luo
- Department of Respiratory and Critical Care Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Jianjun Li
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Tao Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China.
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, 400038, Chongqing, China.
| |
Collapse
|
48
|
Guarischi-Sousa R, Kroll JE, Bonaldi A, Pierry PM, Villela D, Souza CA, Silva JS, Bürger MC, Oliveira FA, de Paula MG, Meliso FM, de Almeida LG, Monfredini PM, de Oliveira AG, Milanezi F, Scapulatempo-Neto C, Yamamoto GL. A Benchmark of In-House Homologous Recombination Repair Deficiency Testing Solutions for High-Grade Serous Ovarian Cancer Diagnosis. Diagnostics (Basel) 2023; 13:3293. [PMID: 37958189 PMCID: PMC10648202 DOI: 10.3390/diagnostics13213293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 11/15/2023] Open
Abstract
Homologous recombination deficiency (HRD) has become an important prognostic and predictive biomarker for patients with high-grade serous ovarian cancer who may benefit from poly-ADP ribose polymerase inhibitors (PARPi) and platinum-based therapies. HRD testing provides relevant information to personalize patients' treatment options and has been progressively incorporated into diagnostic laboratories. Here, we assessed the performance of an in-house HRD testing system deployable in a diagnostic clinical setting, comparing results from two commercially available next-generation sequencing (NGS)-based tumor tests (SOPHiA DDMTM HRD Solution and AmoyDx® (HRD Focus Panel)) with the reference assay from Myriad MyChoice® (CDx). A total of 85 ovarian cancer samples were subject to HRD testing. An overall strong correlation was observed across the three assays evaluated, regardless of the different underlying methods employed to assess genomic instability, with the highest pairwise correlation between Myriad and SOPHiA (R = 0.87, p-value = 3.39 × 10-19). The comparison of the assigned HRD status to the reference Myriad's test revealed a positive predictive value (PPV) and negative predictive value (NPV) of 90.9% and 96.3% for SOPHiA's test, while AmoyDx's test achieved 75% PPV and 100% NPV. This is the largest HRD testing evaluation using different methodologies and provides a clear picture of the robustness of NGS-based tests currently offered in the market. Our data shows that the implementation of in-house HRD testing in diagnostic laboratories is technically feasible and can be reliably performed with commercial assays. Also, the turnaround time is compatible with clinical needs, making it an ideal alternative to offer to a broader number of patients while maintaining high-quality standards at more accessible price tiers.
Collapse
|
49
|
Li W, Gao L, Yi X, Shi S, Huang J, Shi L, Zhou X, Wu L, Ying J. Patient Assessment and Therapy Planning Based on Homologous Recombination Repair Deficiency. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:962-975. [PMID: 36791952 PMCID: PMC10928375 DOI: 10.1016/j.gpb.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 12/23/2022] [Accepted: 02/05/2023] [Indexed: 02/16/2023]
Abstract
Defects in genes involved in the DNA damage response cause homologous recombination repair deficiency (HRD). HRD is found in a subgroup of cancer patients for several tumor types, and it has a clinical relevance to cancer prevention and therapies. Accumulating evidence has identified HRD as a biomarker for assessing the therapeutic response of tumor cells to poly(ADP-ribose) polymerase inhibitors and platinum-based chemotherapies. Nevertheless, the biology of HRD is complex, and its applications and the benefits of different HRD biomarker assays are controversial. This is primarily due to inconsistencies in HRD assessments and definitions (gene-level tests, genomic scars, mutational signatures, or a combination of these methods) and difficulties in assessing the contribution of each genomic event. Therefore, we aim to review the biological rationale and clinical evidence of HRD as a biomarker. This review provides a blueprint for the standardization and harmonization of HRD assessments.
Collapse
Affiliation(s)
- Wenbin Li
- Department of Pathology, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Gao
- Geneplus-Shenzhen, Shenzhen 518000, China; Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Yi
- Geneplus-Beijing, Beijing 102206, China
| | | | - Jie Huang
- National Institutes for Food and Drug Control, Beijing 100050, China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lingying Wu
- Department of Gynecologic Oncology, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
50
|
Horeweg N, Nout RA, Jürgenliemk-Schulz IM, Lutgens LC, Jobsen JJ, Haverkort MA, Mens JWM, Slot A, Wortman BG, de Boer SM, Stelloo E, Verhoeven-Adema KW, Putter H, Smit VT, Bosse T, Creutzberg CL. Molecular Classification Predicts Response to Radiotherapy in the Randomized PORTEC-1 and PORTEC-2 Trials for Early-Stage Endometrioid Endometrial Cancer. J Clin Oncol 2023; 41:4369-4380. [PMID: 37487144 PMCID: PMC10522107 DOI: 10.1200/jco.23.00062] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/20/2023] [Accepted: 06/09/2023] [Indexed: 07/26/2023] Open
Abstract
PURPOSE The molecular classification of endometrial cancer (EC) has proven to have prognostic value and is predictive of response to adjuvant chemotherapy. Here, we investigate its predictive value for response to external beam radiotherapy (EBRT) and vaginal brachytherapy (VBT) in early-stage endometrioid EC (EEC). METHODS Data of the randomized PORTEC-1 trial (n = 714) comparing pelvic EBRT with no adjuvant therapy in early-stage intermediate-risk EC and the PORTEC-2 trial (n = 427) comparing VBT with EBRT in early-stage high-intermediate-risk EC were used. Locoregional (including vaginal and pelvic) recurrence-free survival was compared between treatment groups across the four molecular classes using Kaplan-Meier's methodology and log-rank tests. RESULTS A total of 880 molecularly classified ECs, 484 from PORTEC-1 and 396 from PORTEC-2, were included. The majority were FIGO-2009 stage I EEC (97.2%). The median follow-up was 11.3 years. No locoregional recurrences were observed in EC with a pathogenic mutation of DNA polymerase-ε (POLEmut EC). In mismatch repair-deficient (MMRd) EC, locoregional recurrence-free survival was similar after EBRT (94.2%), VBT (94.2%), and no adjuvant therapy (90.3%; P = .74). In EC with a p53 abnormality (p53abn EC), EBRT (96.9%) had a substantial benefit over VBT (64.3%) and no adjuvant therapy (72.2%; P = .048). In EC with no specific molecular profile (NSMP EC), both EBRT (98.3%) and VBT (96.2%) yielded better locoregional control than no adjuvant therapy (87.7%; P < .0001). CONCLUSION The molecular classification of EC predicts response to radiotherapy in stage I EEC and may guide adjuvant treatment decisions. Omitting radiotherapy seems to be safe in POLEmut EC. The benefit of radiotherapy seems to be limited in MMRd EC. EBRT yields a significantly better locoregional recurrence-free survival than VBT or no adjuvant therapy in p53abn EC. VBT is the treatment of choice for NSMP EC as it is as effective as EBRT and significantly better than no adjuvant therapy for locoregional tumor control.
Collapse
Affiliation(s)
- Nanda Horeweg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Remi A. Nout
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | | | - Jan J. Jobsen
- Department of Radiotherapy, Medisch Spectrum Twente, Enschede, the Netherlands
| | | | - Jan Willem M. Mens
- Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Annerie Slot
- Radiotherapeutic Institute Friesland, Leeuwarden, the Netherlands
| | - Bastiaan G. Wortman
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Radiotherapy, Catharina Ziekenhuis, Eindhoven, the Netherlands
| | - Stephanie M. de Boer
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ellen Stelloo
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Hein Putter
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carien L. Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|