1
|
Mao J, Wang K, Tong J, Zhang W, Shen G, Wang D, Liao Z, Zhang Z, Miao Q, Jiang S, Zhang K. Discovery of dual PARP/NAMPT inhibitors for the treatment of BRCA wild-type triple-negative breast cancer. Bioorg Med Chem Lett 2025; 120:130117. [PMID: 39889972 DOI: 10.1016/j.bmcl.2025.130117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Simultaneous inhibition of poly(ADP-ribose) polymerase (PARP) and nicotinamide phosphoribosyltransferase (NAMPT) has been shown to be synergistically effective against breast cancer susceptibility (BRCA) wild-type triple-negative breast cancer (TNBC) through synthetic lethality, which may be explored to broaden the clinical utility of PARP inhibitors. Herein, we report the discovery of dual PARP/NAMPT inhibitors through a pharmacophore linking approach. The lead compound 13j with potent inhibitory activity against both PARP1 and NAMPT (IC50 = 0.8 and 18 nM, respectively) effectively inhibited the proliferation of TNBC MDA-MB-231 cells with wild-type BRCA at submicromolar level. Mechanically, 13j disrupted the homologous recombination repair (HRR) pathway, caused the accumulation of DNA double-strand breaks (DSBs) and ultimately induced apoptotic cell death. In addition, this compound exhibited potent inhibitory potency on the migration of MDA-MB-231 cells. This study demonstrates that compound 13j is a promising lead compound for the development of better PARP/NAMPT inhibitors to treat TNBC with wild-type BRCA.
Collapse
Affiliation(s)
- Jie Mao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Kaizhen Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Jun Tong
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Wanheng Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Guoqing Shen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Dexiang Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Zepeng Liao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Zhiyi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Qi Miao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China.
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China.
| |
Collapse
|
2
|
Qin CH, Zhang SM, Huo XO, Song RP, Ling J. Effects of SB939 are mediated by STAT3 to inhibit breast cancer cell metastasis-related genes. Oncol Lett 2025; 29:236. [PMID: 40151421 PMCID: PMC11948958 DOI: 10.3892/ol.2025.14982] [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: 09/25/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
The histone deacetylase inhibitor pracinostat (SB939) may inhibit metastasis of triple-negative breast cancer by downregulating fibronectin (FN1) expression through the STAT3 signaling pathway. SB939 exhibits low cytotoxicity and is a potential targeted agent against breast cancer. The present study investigated the value of STAT3 and FN1 as breast cancer treatment targets and integrated cancer databases and bioinformatics tools to evaluate the effect of SB939 on breast cancer metastasis. Gene Set Enrichment Analysis, Gene Expression Profiling Interactive Analysis, Gene Expression Database of Normal and Tumor Tissues 2, The University of Alabama at Birmingham Cancer data analysis portal, GeneMANIA, Search Tool for the Retrieval of Interacting Genes/Proteins, LinkedOmics and Tumor Immune Estimation Resource databases were used in the present study. SB939 inhibited enrichment of the STAT3 pathway and decreased the expression of FN1. FN1 and STAT3 expression was markedly higher in breast cancer tissues compared with normal tissues. Kaplan-Meier curves demonstrated that increased expression of STAT3 and FN1 was associated with low survival in patients with breast cancer with overall, recurrence-free and disease-specific survival and FN1 having the strongest association with MMP2, which facilitating extracellular matrix degradation and metastatic niche formation. Furthermore, MMP2 exhibits crosstalk STAT3 to induce metastasis of breast cancer cells. To conclude, SB939 may be used as a small molecule compound for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Chen-Hui Qin
- Department of Oncology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Shu-Min Zhang
- Department of Oncology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Xiao-Ou Huo
- Department of Oncology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Ruo-Piao Song
- Department of Oncology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Jun Ling
- Clinical Laboratory Department, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
3
|
Sood R, Niemierko A, Ryan L, Spring L, Moy B, Bardia A, Vidula N. Impact of Race/Ethnicity on Clinical and Genomic Characteristics, Trial Participation, and Genotype-Matched Therapy among Patients with Metastatic Breast Cancer. Clin Cancer Res 2025; 31:1315-1322. [PMID: 39869305 DOI: 10.1158/1078-0432.ccr-24-2825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/03/2024] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
PURPOSE Race/ethnicity may affect outcomes in metastatic breast cancer (MBC) due to biological and social determinants. We evaluated the impact of race/ethnicity on clinical, socioeconomic, and genomic characteristics, clinical trial participation, and receipt of genotype-matched therapy among patients with MBC. EXPERIMENTAL DESIGN A retrospective study of patients with MBC who underwent cell-free DNA testing (cfDNA, Guardant360, 74 gene panel) between 11/2016 and 11/2020 was conducted. Receipt of genotype-matched therapy targeted at a cfDNA actionable mutation was determined. Pearson χ2 and Wilcoxon rank-sum tests were used to compare categorical and continuous variables between groups. Multivariable logistic regression was used to assess the association of race and receiving matched therapy. RESULTS A total of 425 patients with MBC and cfDNA results were identified (White: 369, Black: 27, Hispanic: 15, and Asian: 14). White patients traveled further for cancer care than other groups (P < 0.001). White patients had the highest rates of commercial insurance, Black patients had the highest rates of state-supported insurance, and Asian patients had the highest uninsured rates (P < 0.001). Clinical trial enrollment did not differ by race/ethnicity (P = 0.34). The proportion of patients with ≥1 actionable mutation in cfDNA did not vary by race/ethnicity (P = 0.18). The highest rates of matched therapy were observed in White patients (P < 0.001). After multivariable logistic regression adjusting for subtype, commercial versus other insurance, Charlson Comorbidity Index, and distance to center, White patients remained more likely to receive matched therapy (P = 0.024). CONCLUSIONS Racial/ethnic minority patients were less likely to receive matched therapy. Further research is needed to identify barriers to precision medicine.
Collapse
Affiliation(s)
- Rupali Sood
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Andrzej Niemierko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Lianne Ryan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Laura Spring
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Aditya Bardia
- University of California Los Angeles, Los Angeles, California
| | - Neelima Vidula
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4
|
Torrisi R, Gerosa R, Miggiano C, Saltalamacchia G, Benvenuti C, Santoro A. Beyond failure of endocrine-based therapies in HR+/HER2 negative advanced breast cancer: What before chemotherapy? A glimpse into the future. Crit Rev Oncol Hematol 2025; 208:104634. [PMID: 39900320 DOI: 10.1016/j.critrevonc.2025.104634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite the impressive improvements achieved by endocrine therapy and CDK4/6 inhibitors (CDK4/6i) and the forthcoming availability of alternative endocrine manipulations and targeted therapies, hormone-receptor positive/HER2 negative (HR+/HER2-) advanced breast cancer (ABC) is almost inevitably destined to become endocrine- refractory. At this time chemotherapy has been recently challenged and partly replaced by new targeted options as antibody-drug conjugated (ADCs). Trastuzumab-deruxtecan has been proven meaningfully superior to chemotherapy either in 1st and later lines after progression to CDK4/6i in HER2-low ABC and results with other ADCs as Sacituzumab Govitecan and Datopotamab-deruxtecan are promising, but the definition of cross-resistance between these drugs sharing either antibody or payload is crucial before implementing them in a useful sequence. While PARP inhibitors are the standard 2nd line in patients with gBRCA mutation, it is not still known whether patients with mutations of PALB2 or of other homologous recombinant defect (HRD)-related genes will benefit of the same treatment. On the other hand, the results obtained with immune checkpoint inhibitors (ICIs) in HR+ /HER2-ABC contrarily to the early setting are disappointing up to now, but investigations of ICIs in combination with other targeted drugs which may increase immune response and the search for better markers of activity are under way. Moreover the anticipation in upfront treatment of ADCs or PARPi in patients with features of putative endocrine resistance and/or of less sensitiviy to CDK4/6i and the choice of therapy in patients recurring during or soon after adjuvant CDK4/6i and olaparib represent further challenges for the future.
Collapse
Affiliation(s)
- Rosalba Torrisi
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy.
| | - Riccardo Gerosa
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Chiara Miggiano
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Giuseppe Saltalamacchia
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy
| | - Chiara Benvenuti
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Armando Santoro
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| |
Collapse
|
5
|
Jahan N, Taraba J, Boddicker NJ, Giridhar KV, Leon-Ferre RA, Tevaarwerk AJ, Cathcart-Rake E, O'Sullivan CC, Peethambaram PP, Hobday TJ, Mina LA, Batalini F, Advani P, Sideras K, Haddad TC, Ruddy KJ, Goetz MP, Couch FJ, Yadav S. Real-World Evidence on Prescribing Patterns and Clinical Outcomes of Metastatic Breast Cancer Patients Treated with PARP Inhibitors: The Mayo Clinic Experience. Clin Breast Cancer 2025; 25:e211-e219.e2. [PMID: 39516069 PMCID: PMC11936386 DOI: 10.1016/j.clbc.2024.10.006] [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: 06/22/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE This study evaluates real-world outcomes, toxicities, and prescribing patterns of PARP inhibitors (PARPis) for the treatment of metastatic breast cancer (MBC). PATIENTS AND METHODS Electronic health records of 62 MBC patients treated with olaparib (n = 48) or talazoparib (n = 14) at Mayo Clinic System between 2017 and 2022 were analyzed. Time-to-treatment-failure (TTF) was assessed utilizing the Kaplan-Meier method. Predictors of TTF were identified in a multivariate Cox-proportional hazard regression model adjusting for relevant tumor and demographic characteristics. RESULTS Among 62 patients who received PARPis for MBC, 55 had germline (g) pathogenic variants (PVs) (gBRCA1 = 24, gBRCA2 = 26, and gPALB2 = 4) and 8 patients had somatic (s) PVs (sBRCA1 = 4, sBRCA2 = 2, sATM = 1, sCDKN2A = 1). Median TTF in the gBRCA1, gBRCA2, and gPALB2 PV carriers were 7, 8, and 9 months, respectively (P = .37). Complete or partial responses were observed among 51.8% of patients with gBRCA or gPALB2 PVs. In multivariate analysis, HER2 positivity (hazard ratio, HR: 4.9, P = .007) and somatic PVs in homologous recombination repair (HRR) genes other than BRCA (sATM or sCDKN2A) (HR: 11.7, P = .01) were associated with a shorter TTF. No significant difference in TTF was observed by the type of PARPi, estrogen and progesterone receptor status, age, or number of prior therapies. Eight (16.7%) patients receiving olaparib and seven (50%) receiving talazoparib required dose reductions due to toxicities. CONCLUSIONS In real-world practice, PARPis are well-tolerated with promising TTF in gBRCA1/2 and gPALB2 carriers. Further studies will delineate the clinical efficacy of PARPis in other MBC subsets, such as sBRCA mutations, HER2-positive disease, and CNS metastasis.
Collapse
Affiliation(s)
- Nusrat Jahan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL; Department of Oncology, Mayo Clinic, Rochester, MN.
| | - Jodi Taraba
- Department of Oncology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | - Lida A Mina
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ
| | - Felipe Batalini
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ
| | - Pooja Advani
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | | | | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
6
|
Gitto SB, Pantel AR, Maxwell KN, Pryma DA, Farwell MD, Liu F, Cao Q, O'Brien SR, Clark AS, Shah PD, McDonald ES. [ 18F]FluorThanatrace PET imaging as a biomarker of response to PARP inhibitors in breast cancer. COMMUNICATIONS MEDICINE 2025; 5:90. [PMID: 40133542 PMCID: PMC11937411 DOI: 10.1038/s43856-025-00791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Poly (ADP-ribose) polymerase inhibitors (PARPi) are approved for Breast Cancer gene (BRCA)-mutant HER2- breast cancer, and there is clinical interest in expanding indications to include homologous recombination deficient (HRD) breast cancers. Yet, response in these populations remains variable, suggesting clinical utility in developing a better biomarker to select patients for PARPi and predict response. Here, we evaluate a radiolabeled PARPi, [18F]FluorThanatrace ([18F]FTT), as a functional biomarker of PARPi response in breast cancer. METHODS A single-arm prospective observational trial was conducted at the University of Pennsylvania. [18F]FTT-PET uptake was measured in 24 women with untreated primary breast cancer and correlated with tumor HRD score. In a separate cohort of ten subjects with metastatic HER- breast cancer, [18F]FTT-PET uptake was measured at baseline and after a short interval on a PARPi (a measure of drug-target engagement) and correlated to progression free survival (PFS). RESULTS Here we show that baseline [18F]FTT-PET uptake does not correlate to HRD tissue score, supporting that [18F]FTT provides distinct information from genetic features. Baseline [18F]FTT-PET uptake and the change in uptake from baseline to after PARPi initiation significantly correlates to PFS in woman with breast cancer who received a PARPi (ρ = 0.74, P = 0.023 and ρ = -0.86, P = 0.012, respectively). CONCLUSIONS These early results suggest the potential of [18F]FTT-PET to select patients for PARPi treatment and monitor in vivo pharmacodynamics after therapy start. Absence of association with HRD scores supports [18F]FTT uptake as a novel measure that may be leveraged as a biomarker. Further studies are warranted.
Collapse
Affiliation(s)
- Sarah B Gitto
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Austin R Pantel
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Department of Medicine, Division of Hematology and Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fang Liu
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Quy Cao
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia R O'Brien
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy S Clark
- Department of Medicine, Division of Hematology and Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Payal D Shah
- Department of Medicine, Division of Hematology and Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth S McDonald
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Kazmi F, Shrestha N, Liu TFD, Foord T, Heesen P, Booth S, Dodwell D, Lord S, Yeoh KW, Blagden SP. Next-generation sequencing for guiding matched targeted therapies in people with relapsed or metastatic cancer. Cochrane Database Syst Rev 2025; 3:CD014872. [PMID: 40122129 PMCID: PMC11930395 DOI: 10.1002/14651858.cd014872.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
BACKGROUND Matched targeted therapies (MTT) given alone or in combination with systemic anti-cancer therapies have delivered proven survival benefit for many people with newly diagnosed cancer. However, there is little evidence of their effectiveness in the recurrent or late-stage setting. With this uncertainty, alongside the perception that late-stage cancers are too genetically heterogenous or too mutationally diverse to benefit from matched targeted therapies, next-generation sequencing (NGS) of tumours in people with refractory cancer remains a low priority. As a result, next-generation sequencing testing of recurrent or late-stage disease is discouraged. We lack evidence to support the utility of next generation sequencing in guiding matched targeted therapies in this setting. OBJECTIVES To evaluate the benefits and harms of matched targeted therapies in people with advanced cancers in randomised controlled trials. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, ClinicalTrials.gov, and the World Health Organisation International Clinical Trials Registry Platform (WHO-ICTRP) search portal up to 30th October 2024. We also screened reference lists of included studies and also the publications that cited these studies. SELECTION CRITERIA We included randomised controlled trials (RCTs) that had enroled participants with advanced/refractory solid or haematological cancers who had progressed through at least one line of standard anti-cancer systemic therapy. To be eligible, all participants should have received matched targeted therapy based on next-generation sequencing carried out on their tumour (tumour tissue, blood or bone marrow). DATA COLLECTION AND ANALYSIS We systematically searched medical databases (e.g. MEDLINE, Embase) and trial registers for randomised controlled trials (RCTs). Outcomes of interest were progression-free survival (PFS), overall survival (OS), overall response rates (ORR), serious (grade 3 or 4) adverse events (AEs) and quality of life (QOL). We used a random-effects model to pool outcomes across studies and compared predefined subgroups using interaction tests. Grading of Recommendations Assessment, Development and Evaluation (GRADE) assessment of certainty was used to evaluate the quality of evidence. MAIN RESULTS We identified a total of 37 studies, out of which 35 studies (including 9819 participants) were included in the meta-analysis. All included studies compared a matched targeted therapy intervention to standard-of-care treatment, non-matched targeted therapies or no treatment (best supportive care): Matched targeted therapy versus standard-of-care treatment Matched targeted therapy (MTT) compared with standard systematic therapy probably reduces the risk of disease progression by 34% (hazard ratio (HR) = 0.66, 95% confidence interval (CI) 0.59 to 0.74; 14 studies, 3848 participants; moderate-certainty evidence). However, MTT might have little to no difference in risk of death (HR = 0.85, 95% CI 0.75 to 0.97; 14 studies, 3848 participants; low-certainty evidence) and may increase overall response rates (low-certainty evidence). There was no clear evidence of a difference in severe (grade 3/4) adverse events between matched targeted therapy and standard-of-care treatment (low-certainty evidence). There was limited evidence of a difference in quality of life between groups (very low-certainty of evidence). Matched targeted therapy in combination with standard-of-care treatment versus standard-of-care treatment alone Matched targeted therapy in combination with standard-of-care treatment compared with standard-of-care treatment alone probably reduces the risk of disease progression by 39% (HR = 0.61, 95% CI 0.53-0.70, 14 studies, 2,637 participants; moderate-certainty evidence) and risk of death by 21% (HR = 0.79, 95% CI 0.70 to 0.89; 11 studies, 2575 participants, moderate-certainty evidence). The combination of MTT and standard-of-care treatment may also increase overall response rates (low-certainty evidence). There was limited evidence of a difference in the incidence of severe adverse events (very low-certainty evidence) and quality of life between the groups (very low-certainty of evidence). Matched targeted therapy versus non-matched targeted therapy Matched targeted therapy compared with non-matched targeted therapy probably reduces the risk of disease progression by 24% (HR = 0.76, 95% CI 0.64 to 0.89; 3 studies, 1568 participants; moderate-certainty evidence) and may reduce the risk of death by 25% (HR = 0.75, 95% CI 0.65 to 0.86, 1307 participants; low-certainty evidence). There was little to no effect on overall response rates between MTT and non-MTT. There was no clear evidence of a difference in overall response rates (low-certainty evidence) and severe adverse events between MTT and non-MTT (low-certainty evidence). None of the studies comparing MTT and non-MTT reported quality of life. Matched targeted therapy versus best supportive care Matched targeted therapy compared with the best supportive care (BSC) i.e. no active treatment probably reduces the risk of disease progression by 63% (HR 0.37, 95% CI 0.28 to 0.50; 4 studies, 858 participants; moderate-certainty evidence). There was no clear evidence of a difference in overall survival between groups (HR = 0.88, 95% CI 0.73 to 1.06, 3 studies, 783 participants; low-certainty evidence). There was no clear evidence of a difference in overall response rates (very low-certainty of evidence) and incidence of severe adverse events (very low-certainty of evidence) between the groups. Quality of life was reported in a single study but did not provide composite scores. Risk of bias The overall risk of bias was judged low for eight studies, unclear for two studies, and the remaining 27 studies were high risk. AUTHORS' CONCLUSIONS Matched targeted therapies guided by next-generation sequencing in people with advanced cancer prolongs the time before cancer progresses compared to standard therapies. However, there is limited evidence to suggest that it prolongs overall survival, improves the quality of life or increases adverse events. Importantly, this review supports equitable access to next-generation sequencing technology for all people with advanced cancer and offers them the opportunity to access genotype-matched targeted therapies.
Collapse
Affiliation(s)
- Farasat Kazmi
- Department of Oncology, University of Oxford, Oxford, UK
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Nipun Shrestha
- Health Evidence Synthesis, Recommendations and Impact (HESRI), School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Tik Fung Dave Liu
- Department of Oncology, Norfolk and Norwich University Hospital, Norwich, UK
| | | | | | - Stephen Booth
- Department of Haematology, Royal Berkshire Hospital, Reading, UK
| | - David Dodwell
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Oxford, UK
| | - Kheng-Wei Yeoh
- Radiation Oncology, National Cancer Centre, Singapore, Singapore
| | | |
Collapse
|
8
|
Sekine Y, Oka D, Ohtsu A, Nakayama H, Miyao T, Miyazawa Y, Arai S, Koike H, Matsui H, Shibata Y, Suzuki K. The combination of poly(ADP-ribose) polymerase inhibitor and statin inhibits the proliferation of human castration-resistant and taxane-resistant prostate cancer cells in vitro and in vivo. BMC Cancer 2025; 25:521. [PMID: 40119293 PMCID: PMC11929194 DOI: 10.1186/s12885-025-13895-6] [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: 12/07/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Olaparib exhibits antitumor effects in castration-resistant prostate cancer patients with germline mutations in DNA repair genes. We previously reported that simvastatin reduced the expression of DNA repair genes in PC-3 cells. The efficacy of combination therapy using olaparib and simvastatin as "BRCAness" in castration-resistant and taxane-resistant prostate cancers was evaluated in this study. METHODS PC-3, LNCaP, and 22Rv1 human prostate cancer cell lines were used to develop androgen-independent LNCaP cells (LNCaP-LA). mRNA and protein expression levels were evaluated by quantitative real-time polymerase chain reaction and western blot analysis, respectively. Cell viability was determined using the MTS assay and cell counts. All evaluations were performed on cells treated with simvastatin with or without olaparib. RESULTS The mRNA levels of BRCA1, BRCA2, RAD51, FANCD2, FANCG, FANCA, BARD1, RFC3, RFC4, and RFC5, which are known DNA repair genes, were downregulated by simvastatin in androgen-independent prostate cancer cells, such as PC-3, LNCaP-LA, and 22Rv1 cells. In contrast, the expression of all these genes remained unchanged in androgen-dependent LNCaP cells following treatment with simvastatin. Furthermore, simvastatin increased the expression of above stated genes in normal prostate stromal cells (PrSC). The reduction in BRCA1 and BRCA2 expression following siRNA transfection increased the cytocidal effects of Olaparib in PC-3 and LNCaP-LA cells. The combination of olaparib and simvastatin further inhibited cell proliferation compared to monotherapy with either drug in PC-3, 22Rv1, and LNCaP-LA cells but not in PrSC cells. In a 22Rv1-derived mouse xenograft model, the combination of olaparib and simvastatin enhanced the inhibition of cell proliferation. Moreover, we established a 22Rv1 cell line with acquired resistance to Cabazitaxel (22Rv1-CR). In 22Rv1-CR cells, simvastatin also decreased the expression of BRCA1, BRCA2, and FANCA, and the combination of olaparib and simvastatin further enhanced the inhibition of cell proliferation compared with treatment with either of the drugs alone. CONCLUSIONS Simvastatin altered the expression of several genes associated with DNA repair in castration-resistant and taxane-resistant prostate cancer cells. The combination of poly (ADP-ribose) polymerase inhibitors and drugs that decrease DNA repair gene expression can potentially affect castration-resistant and taxane-resistant prostate cancer growth.
Collapse
Affiliation(s)
- Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Daisuke Oka
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Akira Ohtsu
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroshi Nakayama
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takeshi Miyao
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yoshiyuki Miyazawa
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Seiji Arai
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hidekazu Koike
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroshi Matsui
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yasuhiro Shibata
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Kazuhiro Suzuki
- Department of Urology, Gunma University Graduate School of Medicine, 3-9-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
9
|
Zhu Z, Shi Y. Poly (ADP-ribose) polymerase inhibitors in cancer therapy. Chin Med J (Engl) 2025; 138:634-650. [PMID: 39932206 PMCID: PMC11925422 DOI: 10.1097/cm9.0000000000003471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Indexed: 03/17/2025] Open
Abstract
ABSTRACT Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have emerged as critical agents for cancer therapy. By inhibiting the catalytic activity of PARP enzymes and trapping them in the DNA, PARPis disrupt DNA repair, ultimately leading to cell death, particularly in cancer cells with homologous recombination repair deficiencies, such as those harboring BRCA mutations. This review delves into the mechanisms of action of PARPis in anticancer treatments, including the inhibition of DNA repair, synthetic lethality, and replication stress. Furthermore, the clinical applications of PARPis in various cancers and their adverse effects as well as their combinations with other therapies and the mechanisms underlying resistance are summarized. This review provides comprehensive insights into the role and mechanisms of PARP and PARPis in DNA repair, with a particular focus on the potential of PARPi-based therapies in precision medicine for cancer treatment.
Collapse
Affiliation(s)
- Ziqi Zhu
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yujun Shi
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
10
|
Renaudin X, Campalans A. Modulation of OGG1 enzymatic activities by small molecules, promising tools and current challenges. DNA Repair (Amst) 2025; 149:103827. [PMID: 40120404 DOI: 10.1016/j.dnarep.2025.103827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
Oxidative DNA damage, resulting from endogenous cellular processes and external sources plays a significant role in mutagenesis, cancer progression, and the pathogenesis of neurological disorders. Base Excision Repair (BER) is involved in the repair of base modifications such as oxidations or alkylations as well as single strand breaks. The DNA glycosylase OGG1, initiates the BER pathway by the recognition and excision of 8oxoG, the most common oxidative DNA lesion, in both nuclear and mitochondrial DNA. Beyond DNA repair, OGG1 modulates transcription, particularly pro-inflammatory genes, linking oxidative DNA damage to broader biological processes like inflammation and aging. In cancer therapy, BER inhibition has emerged as a promising strategy to enhance treatment efficacy. Targeting OGG1 sensitizes cells to chemotherapies, radiotherapies, and PARP inhibitors, presenting opportunities to overcome therapy resistance. Additionally, OGG1 activators hold potential in mitigating oxidative damage associated with aging and neurological disorders. This review presents the development of several inhibitors and activators of OGG1 and how they have contributed to advance our knowledge in the fundamental functions of OGG1. We also discuss the new opportunities they provide for clinical applications in treating cancer, inflammation and neurological disorders. Finally, we also highlight the challenges in targeting OGG1, particularly regarding the off-target effects recently reported for some inhibitors and how we can overcome these limitations.
Collapse
Affiliation(s)
- Xavier Renaudin
- Université Paris-Saclay, iRCM/IBFJ, CEA, Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses F-92260, France; Université Paris Cité, iRCM/IBFJ, CEA, Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses F-92260, France
| | - Anna Campalans
- Université Paris-Saclay, iRCM/IBFJ, CEA, Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses F-92260, France; Université Paris Cité, iRCM/IBFJ, CEA, Genetic Stability, Stem Cells and Radiation, Fontenay-aux-Roses F-92260, France.
| |
Collapse
|
11
|
Bouzid RS, Bouzid R, Labed H, Serhani I, Hellal D, Oumeddour L, Boudhiaf I, Ibrir M, Khadraoui H, Belaaloui G. Molecular subtyping and target identification in triple negative breast cancer through immunohistochemistry biomarkers. BMC Cancer 2025; 25:454. [PMID: 40082760 PMCID: PMC11905517 DOI: 10.1186/s12885-025-13832-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The Triple-Negative Breast Cancer (TNBC) molecular subtyping and target identification based on Immunohistochemistry (IHC) is of considerable worth for routine use. Yet, literature on this topic is limited worldwide and needs to be enriched with data from different populations. METHODS We assessed the IHC expression of subtyping biomarkers (Cytokeratins 5, 14 and 17, Epidermal Growth Factor Receptor, Claudins 3 and 7, E-cadherin, Vimentin and Androgen receptor) and predictive biomarkers (Tumor-infiltrating lymphocytes (TILs) density, Breast Cancer Antigen 1 (BRCA1) and P53) in a cohort of TNBC patients. Clinicopathologic parameters and overall survival (OS) were investigated as well. RESULTS The patients were aged 50.11 ± 12.13y (more than 40y in 76.56% of patients), and 23.44% had a BC family history. They were in a non-advanced stage: 51.6% T2 stage, 56.2% negative lymph node involvement, 76.6% without metastasis and 64.1% grade II Scarff-Bloom-Richardson classification (SBR). The IHC subtypes were: 53.1% Basal-like1 (BL1), 6.3% Basal-like2 (BL2), 17.2% Mesenchymal (MES), 9.4% Luminal Androgen Receptor (LAR), 4.7% Mixed subtype and 9.4% "Unclassified" type. The LAR subtype involved the youngest patients (40.17 ± 8.68y, p = 0.02). The "Unclassified" subtype expressed the p53 mutated-type pattern more frequently (100%, p = 0.07). The BRCA1 mutated pattern and TILs infiltration were present in (23.44% and 37.5% of patients, respectively). The OS of the subtypes differed significantly (p = 0.007, log-rank test). The subtypes median OS were, respectively, 15.47 mo. (Unclassified), 18.94 mo. (BL2), 27.23 mo. (MES), 27.28 mo. (Mixed), 30.88 mo. (BL1), and 45.07 mo. (LAR). There was no difference in the OS following age, BRCA1 expression, p53 pattern and TILs density. Though, the OS following the TNM stage was different (p = 0.001). A multivariable Cox proportional hazards regression analysis showed that TNM staging and TNBC subtypes, independently influence the OS (p < 0.001 and p = 0.017, respectively). Hence, IHC is useful in TNBC subtyping for prognostic purposes and in the identification of therapeutic biomarkers. Further investigation is required to confirm our results and to implement IHC as a routine tool to improve patient's care.
Collapse
Affiliation(s)
- Rima Saad Bouzid
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Radhia Bouzid
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Housna Labed
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Iman Serhani
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Dounia Hellal
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Leilia Oumeddour
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Ines Boudhiaf
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Massouda Ibrir
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, University Hospital, Batna, Algeria
| | - Hachani Khadraoui
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Ghania Belaaloui
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria.
- Faculty of Medicine, University Batna 2, Batna, Algeria.
| |
Collapse
|
12
|
Ryspayeva D, Seyhan AA, MacDonald WJ, Purcell C, Roady TJ, Ghandali M, Verovkina N, El-Deiry WS, Taylor MS, Graff SL. Signaling pathway dysregulation in breast cancer. Oncotarget 2025; 16:168-201. [PMID: 40080721 PMCID: PMC11906143 DOI: 10.18632/oncotarget.28701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Collapse
Affiliation(s)
- Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Tyler J. Roady
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| | - Martin S. Taylor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Brown Center on the Biology of Aging, Brown University, RI 02903, USA
| | - Stephanie L. Graff
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| |
Collapse
|
13
|
Cruellas M, Papakonstantinou A, López-Fernández A, Castillo E, Matito J, Gómez M, Rezqallah A, Vega S, Navarro V, Torres M, Moles-Fernández A, Saura C, Vivancos A, Balmaña J, Oliveira M. Identifying germline pathogenic variants in breast cancer using tumor sequencing. Breast 2025; 81:104439. [PMID: 40090122 PMCID: PMC11952849 DOI: 10.1016/j.breast.2025.104439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
PURPOSE To investigate the performance of an in-house tumor sequencing panel to identify patients with breast cancer and a germline pathogenic variant (gPV). PATIENTS AND METHODS Retrospective and blinded tumor sequencing analysis in 90 patients with breast cancer and prior germline genetic testing (45 non-carriers and 45 carriers of a gPV) using an in-house panel (VHIO-300). Sensitivity (S), specificity (Sp), positive predictive value (PPV), and negative predictive value (NPV) of tumor sequencing were calculated. A Cohen's kappa coefficient ≥0.80 was predefined as minimum to be reliably acceptable for clinical implementation. RESULTS The cohort included 84 women and 6 men with a median age of 48 years (29-84). Tumors of germline carriers were mainly stage II (47 % vs 31 %, P = 0.047), luminal B-like (56 % vs 31 %, p = 0.037) or triple negative (22 % vs 16 %, = 0.037). The in-house tumor panel identified 91 % (40/44) of the gPV. The analysis did not detect any of the 2 patients with germline large rearrangement alterations nor 2 of the 7 patients with intronic variants included. The tumor sequencing panel yielded 7 % of false positive results (ie, genetic alterations suggestive of germline origin). Hence, S was 91 %, Sp 93 % and Cohen's kappa coefficient between tumor and germline testing was 0.84 (95 % CI 0.73-0.95). CONCLUSION Tumor tissue sequencing with our in-house panel demonstrated an acceptable performance to identify patients with breast cancer carriers of a gPV.
Collapse
Affiliation(s)
- Mara Cruellas
- Medical Oncology Service, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Spain; Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Spain
| | - Andri Papakonstantinou
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; Department Breast Cancer, Endocrine Tumors and Sarcoma, Theme Cancer, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Adrià López-Fernández
- Medical Oncology Service, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Spain; Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Spain
| | - Ester Castillo
- Genomics Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Judit Matito
- Genomics Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Marina Gómez
- Genomics Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Alejandra Rezqallah
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Spain
| | - Sharela Vega
- Medical Oncology Service, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Spain; Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Spain
| | - Víctor Navarro
- Statistics Unit, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Maite Torres
- Clinical Genetics Service, Vall d'Hebron Barcelona Hospital Campus, Spain
| | | | - Cristina Saura
- Medical Oncology Service, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Spain; Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Spain
| | - Ana Vivancos
- Genomics Cancer Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Judith Balmaña
- Medical Oncology Service, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Spain; Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Spain.
| | - Mafalda Oliveira
- Medical Oncology Service, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Spain; Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Spain
| |
Collapse
|
14
|
Wang YW, Allen I, Funingana G, Tischkowitz M, Joko-Fru YW. Predictive biomarkers for the efficacy of PARP inhibitors in ovarian cancer: an updated systematic review. BJC REPORTS 2025; 3:14. [PMID: 40069561 PMCID: PMC11897386 DOI: 10.1038/s44276-025-00122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/06/2024] [Accepted: 01/09/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND PARP inhibitors are effective in treating ovarian cancer, especially for BRCA1/2 pathogenic variant carriers and those with HRD (homologous recombination deficiency). Concerns over toxicity and costs have led to the search for predictive biomarkers. We present an updated systematic review, expanding on a previous ESMO review on PARP inhibitor biomarkers. METHODS Following ESMO's 2020 review protocol, we extended our search to March 31, 2023, including PubMed and clinical trial data. We also reviewed the reference lists of review articles. We conducted a meta-analysis using a random-effects model to evaluate hazard ratios and assess the predictive potential of biomarkers and the effectiveness of PARP inhibitors in survival. RESULTS We found 375 articles, 103 of which were included after screening (62 primary research, 41 reviews). HRD remained the primary biomarker (95%), particularly BRCA1/2 variants (77%). In the non-HRD category, six articles (10%) introduced innovative biomarkers, including ADP-ribosylation, HOXA9 promoter methylation, patient-derived organoids, KELIM, and SLFN11. DISCUSSION Prospective assessment of real-time homologous recombination repair via nuclear RAD51 levels shows promise but needs validation. Emerging biomarkers like ADP-ribosylation, HOXA9 promoter methylation, patient-derived organoids, KELIM, and SLFN11 offer potential but require large-scale validation.
Collapse
Affiliation(s)
- Ying-Wen Wang
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynaecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Isaac Allen
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Yvonne Walburga Joko-Fru
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Galuia M, Fedorova J, McHayleh W, Mamounas E, Ahmad S, Pavri S. Perioperative Drug Management of Systemic Therapies in Breast Cancer: A Literature Review and Treatment Recommendations. Curr Oncol 2025; 32:154. [PMID: 40136358 PMCID: PMC11941735 DOI: 10.3390/curroncol32030154] [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: 01/18/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer accounts for about 30% of all new female cancers each year, and its incidence is increasing 0.6% per year. An enhanced understanding of the molecular mechanisms of carcinogenesis has led to the development of constantly evolving strategies for local and systemic therapies. Perioperative chemotherapy, immunotherapy, and endocrine therapy play pivotal roles in the overall treatment plan. Guidelines on the appropriate use of these drugs in patients undergoing extirpative breast surgery and/or breast reconstruction are lacking. Clear indications for the management of systemic therapies relative to the timing of surgery is crucial to ensure consistent treatment outcomes and to minimize complications. Our purpose is to propose evidence-based recommendations to optimize the perioperative management of systemic therapies in patients undergoing breast cancer surgery and breast reconstructive surgery. In this review, we outline the basic tenets of breast cancer therapies, provide an overview on wound-healing principles, delineate relevant pharmacodynamic concepts, summarize literature and pharmacologic data from various preclinical studies and clinical trials, and propose treatment recommendations. Synopsis: This review proposes evidence-based recommendations regarding systemic therapies management for outcome optimization in the perioperative period in breast cancer patients.
Collapse
Affiliation(s)
- Mariem Galuia
- Department of Internal Medicine, AdventHealth Hospital, Orlando, FL 32804, USA;
| | - Julia Fedorova
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| | - Wassim McHayleh
- Department of Medical Oncology, AdventHealth Hospital, Altamonte Springs, FL 32701, USA
| | - Eleftherios Mamounas
- Division of Breast Surgery, Department of Surgery, AdventHealth Hospital, Orlando, FL 32804, USA;
| | - Sarfraz Ahmad
- Gynecologic Oncology Program, AdventHealth Cancer Institute, Orlando, FL 32804, USA
| | - Sabrina Pavri
- Division of Plastic Surgery, Department of Surgery, AdventHealth Hospital, Orlando, FL 32804, USA;
| |
Collapse
|
16
|
Valachis A, Karihtala P, Geisler J, Tuxen MK. Metastatic triple-negative breast cancer - current treatment strategies in the Nordics: a modified Delphi study. Acta Oncol 2025; 64:349-357. [PMID: 40045533 PMCID: PMC11898304 DOI: 10.2340/1651-226x.2025.42733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND AND PURPOSE This study aimed to assess current treatment strategies for metastatic triple-negative breast cancer (mTNBC) and the perceptions of clinical experts in Sweden, Denmark, Norway, Finland, and Iceland, comparing them to international guidelines to provide insights into how these therapies are implemented and adapted to national Nordic guidelines. METHODS A three-round modified Delphi method was followed with consensus defined as 70% agreement. A steering committee selected 20 experienced oncologists as panellists and developed the questionnaires. Questions included items related to treatment preferences in different treatment lines with different clinical scenarios in mTNBC patients. RESULTS In the first round, eight out of 33 questions on clinical treatment reached consensus with 14 out of 27 in the second round reaching consensus. In round three, eight out of eight questions reached consensus. The preferred treatment for mTNBC patients with PD-L1 positive was checkpoint inhibitors (CPI) in combination with chemotherapy. For patients with germline BRCA mutation and PD-L1 negative disease, PARP-inhibitors were preferred as 1L and sacituzumab govitecan (SG) in both 2L and later lines. Disagreement was observed for chemotherapy in later lines where evidence is sparse or lacking. INTERPRETATION The high level of consensus for new treatment strategies, such as CPI and PARP-inhibitors in 1L and SG in 2L or later lines, in comparison with the limited consensus for older treatments, such as chemotherapy, may reflect the growing academic evidence for different treatment strategies. Understanding the treatment patterns across different countries contributes to gaining consensus on the upcoming therapeutic advances.
Collapse
Affiliation(s)
- Antonis Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Örebro, Sweden.
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Centre and University of Helsinki, Helsinki, Finland
| | - Jürgen Geisler
- Department of Oncology, Division of Medicine, Akershus University Hospital (AHUS), Lørenskog, Norway & Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Campus AHUS, Norway
| | - Malgorzata K Tuxen
- Department of Oncology, Herlev and Gentofte University Hospital, Herlev, Denmark
| |
Collapse
|
17
|
Maselli-Schoueri JH, De Carvalho LEW, De Melo Sette CV, de Abreu LC, Fonseca FLA, Adami F, da Silva Paiva L. How are we allocating physicians to deal with breast cancer in men and women in Brazil? BMC Cancer 2025; 25:392. [PMID: 40038653 DOI: 10.1186/s12885-025-13742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/14/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Female breast cancer (FBC) is a well-known public health issue worldwide. However, male breast cancer (MBC), though rare, may be overlooked by both public health authorities and clinicians. Both diseases exhibit similarities, and understanding their behavior over time is crucial to grasping their annual impact on many citizens. Furthermore, analyzing if medical personnel are well allocated and influence disease outcomes in a limited setting such as the Public Health System (PHS) is of utmost importance. METHODS This ecological study utilized secondary data from 2008 to 2020 to explore the relationship between the number of doctors per 100,000 inhabitants and mortality from FBC and MBC in Brazil. All data were sourced from Brazil's PHS. Mortality rates were analyzed by age and standardized according to the World Health Organization's population figures. The number of physicians was calculated per 100,000 inhabitants. A linear regression analysis was performed using a stepwise selection/backward elimination approach. RESULTS Between 2008 and 2020, Brazil recorded 195,969 breast cancer-related deaths among adults, including 2,220 male victims. The majority of these deaths occurred in the Southeast region among patients older than 50 years. Although both MBC and FBC demonstrated increasing trends over the study period, no correlation was found between the number of physicians and mortality rates for MBC. Conversely, an increase in primary care physicians over the years was positively correlated with mortality rates for FBC (p < 0.05). In addition, the number of physicians in the PHS (β = -0.163; 95% CI: -0.240 to -0.085; p = 0.002), oncologists (β = -0.507; 95% CI: -0.881 to -0.134; p = 0.015), and radiotherapists (β = -6.402; 95% CI: -12.357 to -0.446; p = 0.039) all showed an inverse association with FBC mortality. CONCLUSIONS The increasing trends in FBC and MBC underscore the need for urgent monitoring. Lower FBC mortality correlates with higher numbers of physicians and specialized care, highlighting the critical role of healthcare workforce capacity and the strategic allocation of specialized personnel in enhancing patient outcomes.
Collapse
Affiliation(s)
- Jean Henri Maselli-Schoueri
- Laboratório de Epidemiologia e Análise de Dados, Centro Universitário Saúde ABC, Av. Lauro Gomes 2000, Vila Sacadura Cabral, Santo André, São Paulo, 09060-870, SP, Brazil.
| | | | | | - Luiz Carlos de Abreu
- province of Munster, University of Limerick - Sarsfield Ave. Limerick, Limerick, Ireland
| | | | - Fernando Adami
- Laboratório de Epidemiologia e Análise de Dados, Centro Universitário Saúde ABC, Av. Lauro Gomes 2000, Vila Sacadura Cabral, Santo André, São Paulo, 09060-870, SP, Brazil
| | - Laercio da Silva Paiva
- Laboratório de Epidemiologia e Análise de Dados, Centro Universitário Saúde ABC, Av. Lauro Gomes 2000, Vila Sacadura Cabral, Santo André, São Paulo, 09060-870, SP, Brazil
| |
Collapse
|
18
|
Ogata N, Barnett BG, Sharp NJH, Fujii T, Iwase T, Dunn SE, Ueno NT. Efficiency of Fulvestrant Monotherapy After CDK4/6 Inhibitor Exposure: Is This a Viable Choice? Cancers (Basel) 2025; 17:884. [PMID: 40075731 PMCID: PMC11898757 DOI: 10.3390/cancers17050884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Guidelines for the first-line treatment of Hormone Receptor-positive, HER2-negative advanced or recurrent breast cancer have shifted to combination therapies of a CDK4/6 inhibitor and endocrine therapy. However, determining an optimal subsequent therapy following CDK4/6 inhibitor progression remains challenging, especially for tumors lacking actionable mutations. Real-world data suggest that fulvestrant monotherapy is frequently selected in this post-CDK4/6 inhibitor setting. This review examines its therapeutic potential in this evolving landscape. A systematic literature search using PubMed and ClinicalTrials.gov identified 153 clinical trials published between 2017 and November 2024, from which ten studies met our strict inclusion criteria, focusing solely on fulvestrant monotherapy. These trials encompassed 1038 patients who had prior exposure to CDK4/6 inhibitors. The selected studies were categorized into three groups: monotherapy trials (EMERALD, SERENA-2, AMEERA-3, and ELAINE-1), combination therapy trials (CAPItello-291 and VERONICA), and CDK4/6 inhibitor rechallenge trials (post-MONARCH, PACE, PALMIRA, and MAINTAIN). The median progression-free survival for fulvestrant monotherapy was 3.18 months (range 1.9-5.3 months). Factors affecting the efficacy of fulvestrant monotherapy in second-line therapy include prior treatments, treatment duration, and genetic mutations. Given that the efficacy of fulvestrant was short-lived in the second or subsequent lines, participating in clinical trials is a vital option until a novel alternative treatment choice becomes available.
Collapse
Affiliation(s)
- Nanae Ogata
- Translational and Clinical Research Program, University of Hawaiʻi Cancer Center, Honolulu, HI 96813, USA
- Cancer Biology Program, University of Hawaiʻi Cancer Center, Honolulu, HI 96813, USA
| | - Brian G Barnett
- Phoenix Molecular Designs Ltd., 1-8755 Ash St, Vancouver, BC V6P 6T3, Canada
| | | | - Takeo Fujii
- Translational and Clinical Research Program, University of Hawaiʻi Cancer Center, Honolulu, HI 96813, USA
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Toshiaki Iwase
- Translational and Clinical Research Program, University of Hawaiʻi Cancer Center, Honolulu, HI 96813, USA
| | - Sandra E. Dunn
- Phoenix Molecular Designs Ltd., 1-8755 Ash St, Vancouver, BC V6P 6T3, Canada
| | - Naoto T. Ueno
- Translational and Clinical Research Program, University of Hawaiʻi Cancer Center, Honolulu, HI 96813, USA
- Cancer Biology Program, University of Hawaiʻi Cancer Center, Honolulu, HI 96813, USA
| |
Collapse
|
19
|
Villacampa G, Llop-Guevara A, Filmann N, Herencia-Ropero A, Fasching PA, Karn T, Marmé F, Klare P, Müller V, Stefek A, Schem C, Uleer C, Fehm T, Doering G, Stickeler E, van Mackelenbergh M, Felder B, Nekljudova V, Balmaña J, Denkert C, Loibl S, Serra V. RAD51 Testing in Patients with Early HER2-Negative Breast Cancer and Homologous Recombination Deficiency: A Post Hoc Analysis of the GeparOLA Trial. Clin Cancer Res 2025; 31:808-814. [PMID: 39786436 DOI: 10.1158/1078-0432.ccr-24-3148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE The randomized GeparOLA trial reported comparable pathologic complete response (pCR) rates with neoadjuvant treatment containing olaparib versus carboplatin. In this study, we evaluate the association between functional homologous recombination deficiency (HRD) by RAD51 foci and pCR and the potential of improving patient selection by combining RAD51 and stromal tumor-infiltrating lymphocytes. PATIENTS AND METHODS This is a post hoc blinded biomarker analysis from the randomized GeparOLA trial. Patients with early-stage HER2-negative breast cancer and HRD assessed by Myriad MyChoice or BRCA1/BRCA2 mutations were randomized 1:1 to receive (i) paclitaxel plus olaparib or (ii) paclitaxel plus carboplatin, both followed by epirubicin/cyclophosphamide. Functional HRD was predefined as a RAD51 score ≤10% (RAD51-low). RESULTS Overall, 90 of 97 (92.8%) samples were evaluable for RAD51 testing, and 72 of 90 (80.0%) were RAD51-low. The pCR rate in patients with RAD51-low tumors was 66.7% (48/72), whereas it decreased to 22.2% (4/18) in those with RAD51-high. In the multivariable model including clinicopathologic factors and treatment, the RAD51 score remained significantly associated with pCR (OR = 12.03; 95% confidence interval, 2.60-55.73; P = 0.002). Patients with RAD51-low and high stromal tumor-infiltrating lymphocytes in their tumors achieved a pCR rate of 75.0% (27/36). Similar results were observed for olaparib or carboplatin. In the exploratory disease-free survival analysis, no differences were observed between RAD51 groups (high vs. low: HR = 0.85; 95% confidence interval, 0.25-2.97). CONCLUSIONS In a preselected population with HRD, according to a genetic test, RAD51 testing identifies patients with different pCR rates under PARP inhibitor-based or platinum-based therapies. Future biomarker-driven studies should consider this information to refine stratification factors and to improve patient selection.
Collapse
Affiliation(s)
| | | | - Natalie Filmann
- German Breast Group (GBG) Forschungs GmbH, Neu-Isenburg, Germany
| | | | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Thomas Karn
- Department of Gynecology and Obstetrics, University of Frankfurt, Frankfurt, Germany
| | - Frederik Marmé
- Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Volkmar Müller
- Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Stefek
- Johanniter-Krankenhaus Genthin-Stendal, Stendal, Germany
| | - Christian Schem
- Mammazentrum Hamburg am Krankenhaus Jerusalem, Hamburg, Germany
| | | | - Tanja Fehm
- Universitaetsklinikum Düsseldorf, Düsseldorf, Germany
| | | | - Elmar Stickeler
- Klinik für Gynäkologie und Geburtsmedizin, Uniklinik Aachen, RWTH Aachen, Aachen, Germany
| | - Marion van Mackelenbergh
- Universitätsklinikum Schleswig-Holstein, Klinik für Gynäkologie und Geburtshilfe, Schleswig-Holstein, Germany
| | - Bärbel Felder
- German Breast Group (GBG) Forschungs GmbH, Neu-Isenburg, Germany
| | | | - Judith Balmaña
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Carsten Denkert
- Institut für Pathologie, Philipps Universität Marburg und Universitätsklinikum Marburg (UKGM), Marburg, Germany
| | - Sibylle Loibl
- German Breast Group (GBG) Forschungs GmbH, Neu-Isenburg, Germany
| | - Violeta Serra
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
20
|
Moser SC, Jonkers J. Thirty Years of BRCA1: Mechanistic Insights and Their Impact on Mutation Carriers. Cancer Discov 2025; 15:461-480. [PMID: 40025950 PMCID: PMC11893084 DOI: 10.1158/2159-8290.cd-24-1326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 03/04/2025]
Abstract
SIGNIFICANCE Here, we explore the impact of three decades of BRCA1 research on the lives of mutation carriers and propose strategies to improve the prevention and treatment of BRCA1-associated cancer.
Collapse
Affiliation(s)
- Sarah C. Moser
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Muzzana M, Broggini M, Damia G. The Landscape of PARP Inhibitors in Solid Cancers. Onco Targets Ther 2025; 18:297-317. [PMID: 40051775 PMCID: PMC11884256 DOI: 10.2147/ott.s499226] [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: 10/15/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
PARP inhibitors are a class of agents that have shown significant preclinical activity in models defective in homologous recombination (HR). The identification of synthetic lethality between HR defects and PARP inhibition led to several clinical trials in tumors with known HR defects (initially mutations in BRCA1/2 genes and subsequently in other genes involved in HR). These studies demonstrated significant responses in breast and ovarian cancers, which are known to have a significant proportion of patients with HR defects. Since the approval of the first PARP inhibitor (PARPi), olaparib, several other inhibitors have been developed, expanding the armamentarium available to clinicians in this setting. The positive results obtained in breast and ovarian cancer have expanded the use of PARPi in other solid tumors with HR defects, including prostate and pancreatic cancer in which these defects have been identified. The clinical trials have demonstrated responses to PARPi which are now also available for the subset of patients with prostate and pancreatic cancer with HR defects. This review summarizes the results obtained in solid tumors with PARPi and their potential use when combined with other agents, including immune checkpoint inhibitors that are likely to further increase the survival of these patients which still needs a dramatic improvement.
Collapse
Affiliation(s)
- Marta Muzzana
- Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimo Broggini
- Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanna Damia
- Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
22
|
Bonnefoi H, Lerebours F, Pulido M, Arnedos M, Tredan O, Dalenc F, Guiu S, Teixeira L, Mollon D, Levy C, Verret B, Dawood H, Deiana L, Mouret Reynier MA, Augereau P, Canon JL, Huchet N, Guyonneau C, Lemonnier J, MacGrogan G, Gonçalves A, Darbo E, Iggo R. Darolutamide or capecitabine in triple-negative, androgen receptor-positive, advanced breast cancer (UCBG 3-06 START): a multicentre, non-comparative, randomised, phase 2 trial. Lancet Oncol 2025; 26:355-366. [PMID: 39978376 DOI: 10.1016/s1470-2045(24)00737-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND We proposed in 2005 that androgens replace oestrogens as the driver steroids in a subgroup of triple-negative breast cancer (TNBC) with androgen receptor (AR) expression called molecular apocrine (MA) or luminal androgen receptor (LAR). Here, we report the analysis of a clinical trial evaluating the antitumour activity of the anti-androgen darolutamide in MA breast cancer. Our aim was to assess the clinical benefit in patients with AR-positive TNBCs defined by immunohistochemistry and by RNA profiling. METHODS In this multicentre, non-comparative, randomised, phase 2 trial, women aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0-1 and with advanced TNBC that was previously treated with a maximum of one line of chemotherapy were recruited from 45 hospitals in France. After central confirmation of TNBC status and AR positivity (≥10%; SP107 antibody), participants were randomly assigned (2:1) to receive darolutamide 600 mg orally twice daily or capecitabine minimum 1000 mg/m2 twice daily for 2 weeks on and 1 week off, until disease progression, unacceptable toxicity, lost to follow-up, or withdrawal of consent. Randomisation was done centrally using the minimisation procedure and was stratified according to the number of previous lines of chemotherapy. Transcriptomic analysis was used to classify tumours into groups with high and low AR activity (MAhigh and MAlow). The primary clinical endpoint was clinical benefit rate at 16 weeks (confirmed complete response, partial response, or stable disease). The primary translational endpoint was clinical benefit rate in the darolutamide group in MAhigh tumours versus all other tumours. Analyses were done per protocol. This trial is registered with ClinicalTrials.gov (NCT03383679), and is closed to recruitment. FINDINGS Between April 9, 2018, and July 20, 2021, 254 women were screened and 94 were randomly assigned to darolutamide (n=61) or capecitabine (n=33), of whom 90 were evaluable for efficacy analyses. Median follow-up at the data cutoff on July 20, 2022, was 22·5 months (IQR 16·5-30·5). The clinical benefit rate was 29% (17 of 58; 90% CI 19-39) with darolutamide and 59% (19 of 32; 90% CI 45-74) with capecitabine. In patients treated with darolutamide, the clinical benefit rate was 57% (12 of 21; 95% CI 36-78) in MAhigh tumours, and 16% (five of 31; 95% CI 3-29; p=0·0020) in other tumours. The most common grade 3 adverse events were palmar-plantar erythrodysaesthesia syndrome (none of 60 in the darolutamide group vs two [6%] of 33 in the capecitabine group), and headache (three [5%] vs none). No grade 4 or 5 adverse events were observed. Drug-related serious adverse events occurred in three (5%) patients in the darolutamide group and three (9%) in the capecitabine group, which were toxicoderma (n=1) and headache (n=2) in the darolutamide group, and diarrhoea, general physical deterioration, and hepatic cytolysis in the capecitabine group (n=1 each). INTERPRETATION This study did not reach its prespecified endpoint for darolutamide activity in patients with triple-negative breast cancer selected on the basis of immunohistochemistry for AR. Further studies selecting patients based on RNA profiling might allow better identification of tumours sensitive to anti-androgens. FUNDING Bayer and Fondation Bergonié.
Collapse
Affiliation(s)
- Hervé Bonnefoi
- Department of Medical Oncology, Institut Bergonié, INSERM U1312 BRIC, Université de Bordeaux Collège Sciences de la Santé, Bordeaux, France.
| | | | - Marina Pulido
- Clinical and Epidemiological Research Unit, Institut Bergonié, INSERM CIC1401, Bordeaux, France
| | - Monica Arnedos
- Department of Medical Oncology, Institut Bergonié, INSERM U1312 BRIC, Bordeaux, France
| | - Olivier Tredan
- Department of Medical Oncology, Centre Léon Bérard, Cancer Research Center of Lyon (UMR Inserm 1052 - CNRS 5286), Lyon, France
| | - Florence Dalenc
- Department of Medical Oncology, Oncopole Claudius Regaud, IUCT-Oncopole, Université de Toulouse, Toulouse, France
| | - Séverine Guiu
- Department of Medical Oncology, Institut du Cancer Montpellier - Val d'Aurelle Montpellier, France
| | - Luis Teixeira
- Department of Senologie, Hôpital Saint Louis, INSERM U976, Université Paris Cité, Paris, France
| | - Delphine Mollon
- Department of Medical Oncology, Centre Hospitalier de Cornouaille, Quimper, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Benjamin Verret
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Heba Dawood
- Department of Medical Oncology, Centre Hospitalier Régional, Orléans, France
| | - Laura Deiana
- Department of Medical Oncology, Centre Hospitalier Universitaire, Brest, France
| | | | - Paule Augereau
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest - Paul Papin, Angers, France
| | - Jean-Luc Canon
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Noémie Huchet
- Clinical and Epidemiological Research Unit, Institut Bergonié, Bordeaux, France
| | | | | | | | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, INSERM, CNRS, CRCM, Aix Marseille Univ, Marseille, France
| | - Elodie Darbo
- INSERM U1312, Université de Bordeaux, Bordeaux, France
| | - Richard Iggo
- INSERM U1312, Department of Medical and Biological Sciences, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
23
|
Borgmann K, Krug D. BRCAness Identifies Synthetic Cytotoxicity Between Cisplatin and Radiation Therapy. Int J Radiat Oncol Biol Phys 2025; 121:780-782. [PMID: 39909612 DOI: 10.1016/j.ijrobp.2024.11.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/23/2024] [Indexed: 02/07/2025]
Affiliation(s)
- Kerstin Borgmann
- Department of Radiotherapy & Radiation Oncology, Hubertus Wald Tumor Center-University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - David Krug
- Department of Radiotherapy & Radiation Oncology, Hubertus Wald Tumor Center-University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
24
|
Merkuryev AV, Egorov VV. Role of PARP-1 structural and functional features in PARP-1 inhibitors development. Bioorg Chem 2025; 156:108188. [PMID: 39855113 DOI: 10.1016/j.bioorg.2025.108188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is the key enzyme among other PARPs for post-translational modification of DNA repair proteins. It has four functional domains for DNA-binding, automodification and enzymatic activity. PARP-1 participates in poly-ADP-ribosylation of itself or other proteins during DNA damage response. It recruits reparation machinery proteins that restore native DNA sequence. PARP-1 participates in chromatin structure organization and gene expression regulation. It was shown that PARP-1-dependent regulation mechanisms affect on possible risk of carcinogenesis. Therefore, PARP-1 was proposed as a novel target for cancer treatment. Three generations of PARP-1 inhibitors had been developed depending on pharmacophore structure. To date, four PARP-1 inhibitors have been approved for cancer treatment as a chemotherapy potentiators or as a stand-alone therapy. However, different cytotoxicity effects of specific PARP-1 inhibitors were observed due to diverse PARP-1 activity in cellular processes. Moreover, cancer cells can develop resistance to PARP-1 inhibitors and decrease chemotherapy efficacy. There are promising strategies how to avoid these disadvantages including dual-targeted inhibitors and combination therapy.
Collapse
|
25
|
García-Saenz JÁ, Rodríguez-Lescure Á, Cruz J, Albanell J, Alba E, Llombart A. Second-Line Treatment Options for Patients with Metastatic Triple-Negative Breast Cancer: A Review of the Clinical Evidence. Target Oncol 2025; 20:191-213. [PMID: 39806129 PMCID: PMC11933194 DOI: 10.1007/s11523-024-01125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Metastatic triple-negative breast cancer has a poor prognosis and poses significant therapeutic challenges. Until recently, limited therapeutic options have been available for patients with advanced disease after failure of first-line chemotherapy. The aim of this review is to assess the current evidence supporting second-line treatment options in patients with metastatic triple-negative breast cancer. Evidence was reviewed from controlled clinical trials in which eribulin, vinorelbine, capecitabine, gemcitabine, gemcitabine plus carboplatin, fam-trastuzumab-deruxtecan, sacituzumab govitecan, olaparib, and talazoparib were used in the second-line treatment for metastatic breast cancer, either as study drugs or as comparators. The benefit of treatment was evaluated using the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale. Based on the evidence review, sacituzumab govitecan was identified as the preferred second-line treatment option for patients with metastatic triple-negative breast cancer, supported by clinical evidence and consensus across international clinical guidelines. Olaparib and talazoparib are of use in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer and germline BRCA1/2 mutations. Exploratory data for fam-trastuzumab-deruxtecan suggest a survival benefit in human epidermal growth factor receptor 2-low, hormone-receptor-negative patients, but further solid evidence is required. Other chemotherapies with lower European Society for Medical Oncology-Magnitude of Clinical Benefit Scale scores may continue to be useful in highly selected patients.
Collapse
Affiliation(s)
- José Ángel García-Saenz
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Calle Profesor Martín Lagos, S/N, 28040, Madrid, Spain.
| | | | - Josefina Cruz
- Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Joan Albanell
- Hospital del Mar Research Institute, Barcelona, Spain
- Pompeu Fabra University, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| | - Emilio Alba
- Medical Oncology Unit, Universitary Hospital Virgen de la Victoria, CIBERONC, IBIMA, Malaga, Spain
| | | |
Collapse
|
26
|
Villette CC, Dupuy N, Brightman FA, Zimmermann A, Lignet F, Zenke FT, Terranova N, Bolleddula J, El Bawab S, Chassagnole C. Semi-mechanistic efficacy model for PARP + ATR inhibitors-application to rucaparib and talazoparib in combination with gartisertib in breast cancer PDXs. Br J Cancer 2025; 132:481-491. [PMID: 39875558 PMCID: PMC11876674 DOI: 10.1038/s41416-024-02935-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Promising cancer treatments, such as DDR inhibitors, are often challenged by the heterogeneity of responses in clinical trials. The present work aimed to build a computational framework to address those challenges. METHODS A semi-mechanistic pharmacokinetic-pharmacodynamic model of tumour growth inhibition was developed to investigate the efficacy of PARP and ATR inhibitors as monotherapies, and in combination. Key features of the DNA damage response were incorporated into the model to allow the emergence of synthetic lethality, including redundant DNA repair pathways that may be impaired due to genetic mutations, and due to PARP and ATR inhibition. Model parameters were calibrated using preclinical in vivo data for PARP inhibitors rucaparib and talazoparib and the ATR inhibitor gartisertib. RESULTS The model successfully captured the monotherapy efficacies of rucaparib and talazoparib, as well as the combination efficacy with gartisertib. The model was evaluated against multiple tumour xenografts with diverse genetic backgrounds and was able to capture the observed heterogeneity of response profiles. CONCLUSIONS By enabling simulation of in vivo tumour growth inhibition with PARP and ATR inhibitors for specific tumour types, the model provides a rational approach to support the optimisation of dosing regimens to stratified populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Frank T Zenke
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Nadia Terranova
- Quantitative Pharmacology, Ares Trading S.A. (An Affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | | | | | |
Collapse
|
27
|
Hage Chehade C, Gebrael G, Sayegh N, Ozay ZI, Narang A, Crispino T, Golan T, Litton JK, Swami U, Moore KN, Agarwal N. A pan-tumor review of the role of poly(adenosine diphosphate ribose) polymerase inhibitors. CA Cancer J Clin 2025; 75:141-167. [PMID: 39791278 PMCID: PMC11929130 DOI: 10.3322/caac.21870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/03/2024] [Indexed: 01/12/2025] Open
Abstract
Poly(adenosine diphosphate ribose) polymerase (PARP) inhibitors, such as olaparib, talazoparib, rucaparib, and niraparib, comprise a therapeutic class that targets PARP proteins involved in DNA repair. Cancer cells with homologous recombination repair defects, particularly BRCA alterations, display enhanced sensitivity to these agents because of synthetic lethality induced by PARP inhibitors. These agents have significantly improved survival outcomes across various malignancies, initially gaining regulatory approval in ovarian cancer and subsequently in breast, pancreatic, and prostate cancers in different indications. This review offers a comprehensive clinical overview of PARP inhibitor approvals, emphasizing their efficacy across different cancers based on landmark phase 3 clinical trials.
Collapse
Affiliation(s)
- Chadi Hage Chehade
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zeynep Irem Ozay
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Arshit Narang
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Tony Crispino
- UsTOO Prostate Cancer Support and Education Las Vegas Chapter, Las Vegas, Nevada, USA
| | - Talia Golan
- Division of Medical Oncology, Sheba Medical Center, Tel Aviv Medical University, Tel Aviv, Israel
| | - Jennifer K Litton
- Division of Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Kathleen N Moore
- Division of Gynecologic Oncology, Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
28
|
Wu S, Thawani R. Tumor-Agnostic Therapies in Practice: Challenges, Innovations, and Future Perspectives. Cancers (Basel) 2025; 17:801. [PMID: 40075649 PMCID: PMC11899253 DOI: 10.3390/cancers17050801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
This review comprehensively analyzes the current landscape of tumor-agnostic therapies in oncology. Tumor-agnostic therapies are designed to target specific molecular alterations rather than the primary site of the tumor, representing a shift in cancer treatment. We discuss recent approvals by regulatory agencies such as the FDA and EMA, highlighting therapies that have demonstrated efficacy across multiple cancer types sharing common alterations. We delve into the trial methodologies that underpin these approvals, emphasizing innovative designs such as basket trials and umbrella trials. These methodologies present unique advantages, including increased efficiency in patient recruitment and the ability to assess drug efficacy in diverse populations rapidly. However, they also entail certain challenges, including the need for robust biomarkers and the complexities of regulatory requirements. Moreover, we examine the promising prospects for developing therapies for rare cancers that exhibit common molecular targets typically associated with more prevalent malignancies. By synthesizing these insights, this review underscores the transformative potential of tumor-agnostic therapies in oncology. It offers a pathway for personalized cancer treatment that transcends conventional histology-based classification.
Collapse
Affiliation(s)
| | - Rajat Thawani
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
29
|
Weddle CJ, Blancard M, Uche N, Pongpamorn P, Cejas RB, Burridge PW. Examining patient-specific responses to PARP inhibitors in a novel, human induced pluripotent stem cell-based model of breast cancer. NPJ Precis Oncol 2025; 9:53. [PMID: 40000798 PMCID: PMC11862011 DOI: 10.1038/s41698-025-00837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Preclinical models of breast cancer that better predict patient-specific drug responses are critical for expanding the clinical utility of targeted therapies, including for inhibitors of poly(ADP-ribose) polymerase (PARP). Reprogramming primary cancer cells into human induced pluripotent stem cells (hiPSCs) recently emerged as a powerful tool to model drug response phenotypes, but its use to date has been limited to hematopoietic malignancies. We designed an optimized reprogramming methodology to generate breast cancer-derived hiPSCs (BC-hiPSCs) from nine patients representing all major subtypes of breast cancer. BC-hiPSCs retain patient-specific oncogenic variants, including variants unique to individual tumor subclones. Additionally, we developed a protocol to differentiate BC-hiPSCs into mammary epithelial cells and mammary-like organoids for in vitro disease modeling, including drug response phenotyping. Using these tools, we demonstrated that BC-hiPSCs can be used to screen for differential sensitivity to PARP inhibitors and mechanistically investigated the causal genetic variant driving drug sensitivity in one patient.
Collapse
Affiliation(s)
- Carly J Weddle
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Malorie Blancard
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nnamdi Uche
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Praeploy Pongpamorn
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Romina B Cejas
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
30
|
Sweatman E, Bayley R, Selemane R, Higgs MR. SETD1A-dependent EME1 transcription drives PARPi sensitivity in HR deficient tumour cells. Br J Cancer 2025:10.1038/s41416-025-02963-0. [PMID: 39994444 DOI: 10.1038/s41416-025-02963-0] [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] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cells deficient in DNA repair factors breast cancer susceptibility 1/2 (BRCA1/2) or ataxia-telangiectasia mutated (ATM) are sensitive to poly-ADP ribose polymerase (PARP) inhibitors. Building on our previous findings, we asked how the lysine methyltransferase SETD1A contributed to PARP inhibitor-mediated cell death in these contexts and determined the mechanisms responsible. METHODS We used cervical, breast, lung and ovarian cancer cells bearing mutations in BRCA1 or ATM and depleted SETD1A using siRNA or CRISPR/Cas9. We assessed the effects of the PARPi Olaparib on cell viability, homologous recombination, and DNA repair. We assessed underlying transcriptional perturbations using RNAseq. We used The Cancer Genomics Atlas (TCGA) and DepMap to investigate patient survival and cancer cell characteristics. RESULTS Loss of SETD1A from both BRCA1-deficient and ATM-deficient cancer cells was associated with resistance to Olaparib, explained by partial restoration of homologous recombination. Mechanistically, SETD1A-dependent transcription of the crossover junction endonuclease EME1 correlated with sensitivity to Olaparib in these cells. Accordingly, when SETD1A or EME1 was lost, BRCA1 or ATM-mutated cells became resistant to Olaparib, and homologous recombination was partially restored. CONCLUSIONS Loss of SETD1A or EME1 drives cellular resistance to Olaparib in certain genetic contexts and may help explain why patients develop resistance to PARP inhibitors in the clinic.
Collapse
Affiliation(s)
- Ellie Sweatman
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Rachel Bayley
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Richad Selemane
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Martin R Higgs
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
31
|
Randall Armel S, Malcolmson J, Volenik A, Maganti M, Watkins N, Charames GS, McCuaig J. Genetic counseling referral rates and genetic testing outcomes in women with young breast cancer: a 20-year Canadian review. Breast Cancer Res Treat 2025:10.1007/s10549-025-07646-1. [PMID: 39985623 DOI: 10.1007/s10549-025-07646-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
PURPOSE Despite guidelines recommending genetic testing for all cases of very young breast cancer (VYBC), poor uptake has been reported. This study aimed to examine genetic testing referral rates and outcomes over a 20-year period within the Canadian context. METHODS A retrospective chart review of all incident VYBC cases (at or below 35 years of age) between January 1, 2000 and December 31, 2019 was conducted. Descriptive statistics were used to summarize demographic factors and logistic regression analyses were performed to identify the predictors associated with referral for genetic counseling and positive genetic test results. RESULTS 628 women were identified with VYBC. Most women presented with stage 2 (42%), hormone receptor-positive (HR +) and HER2-negative (54%) invasive ductal carcinoma (94%). Over the study period, referral rates increased from 44 to 84%. Of women initially tested for BRCA1/BRCA2, only 21% were referred for updated panel testing. Among those tested, 19% had a pathogenic variant, 21% of whom reported no family history of cancer. Predictors of referral included stage 0-2 disease while predictors of positive test results included a second breast cancer diagnosis and positive family history. CONCLUSION Despite guidelines based on age alone, barriers to referral persist. Results of this study suggest the need for new models of care that ensure equitable access to genetic testing for all women diagnosed with VYBC regardless of family history, ethnicity, or disease stage. As genetic testing criteria evolve, protocols must address these barriers to prevent missed opportunities for testing.
Collapse
Affiliation(s)
- Susan Randall Armel
- Bhalwani Familial Cancer Clinic, Princess Margaret Cancer Centre, Toronto, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.
| | - Janet Malcolmson
- Bhalwani Familial Cancer Clinic, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Alexandra Volenik
- Bhalwani Familial Cancer Clinic, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Manjula Maganti
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, Canada
| | - Nicholas Watkins
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Canada
| | - George S Charames
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Jeanna McCuaig
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
32
|
Mitranovici MI, Caravia LG, Moraru L, Pușcașiu L. Targeting Cancer Stemness Using Nanotechnology in a Holistic Approach: A Narrative Review. Pharmaceutics 2025; 17:277. [PMID: 40142941 PMCID: PMC11945010 DOI: 10.3390/pharmaceutics17030277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Increasing evidence shows that a very small population of cancer stem cells (CSCs) is responsible for cancer recurrence, drug resistance, and metastasis. CSCs usually reside in hypoxic tumor regions and are characterized by high tumorigenicity. Their inaccessible nature allows them to avoid the effects of conventional treatments such as chemotherapy, radiotherapy, and surgery. In addition, conventional chemo- and radiotherapy is potentially toxic and could help CSCs to spread and survive. New therapeutic targets against CSCs are sought, including different signaling pathways and distinct cell surface markers. Recent advances in nanotechnology have provided hope for the development of new therapeutic avenues to eradicate CSCs. In this review, we present newly discovered nanoparticles that can be co-loaded with an apoptosis-inducing agent or differentiation-inducing agent, with high stability, cellular penetration, and drug release. We also summarize the molecular characteristics of CSCs and the signaling pathways responsible for their survival and maintenance. Controlled drug release targeting CSCs aims to reduce stemness-related drug resistance, suppress tumor growth, and prevent tumor relapse and metastases.
Collapse
Affiliation(s)
- Melinda-Ildiko Mitranovici
- Department of Anatomy, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| | - Laura Georgiana Caravia
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Liviu Moraru
- Department of Anatomy, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| | - Lucian Pușcașiu
- Department of Anatomy, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| |
Collapse
|
33
|
Gao R, Wu P, Yin X, Zhuang L, Meng X. Deep analysis of the trials and major challenges in the first-line treatment for patients with extensive-stage small cell lung cancer. Int Immunopharmacol 2025; 148:114116. [PMID: 39847950 DOI: 10.1016/j.intimp.2025.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The median overall survival (OS) is approximately 10 months when chemotherapy alone is the first-line treatment for extensive-stage small cell lung cancer (ES-SCLC). The approval of the two PD-L1 inhibitors, atezolizumab and durvalumab, marked the beginning of the immunotherapy era for ES-SCLC. Serplulimab, as the first PD-1 inhibitor to achieve success in the first-line treatment of ES-SCLC, has not only demonstrated significant improvements in patient survival outcomes but also ushered in a new era for PD-1 inhibitors in the treatment of ES-SCLC. Recently, antiangiogenic agents with chemo-immunotherapy have achieved breakthroughs in first-line ES-SCLC treatment. Improving the clinical benefits of individualized treatment for patients with ES-SCLC remains challenging. Challenges include identifying biomarkers for targeted therapy, exploring new treatments, developing new medicines, and classifying SCLC molecular subtypes. This review provides an in-depth analysis of research on first-line ES-SCLC treatment. Additionally, it discusses advances in ES-SCLC treatment.
Collapse
Affiliation(s)
- Ran Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Peizhu Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Lulu Zhuang
- Cheeloo College of Cancer Center, Shandong University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China.
| |
Collapse
|
34
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [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: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
35
|
Zhang H, Yang F, Xu Y, Zhao S, Jiang YZ, Shao ZM, Xiao Y. Multimodal integration using a machine learning approach facilitates risk stratification in HR+/HER2- breast cancer. Cell Rep Med 2025; 6:101924. [PMID: 39848244 DOI: 10.1016/j.xcrm.2024.101924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 08/11/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
Hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer is the most common type of breast cancer, with continuous recurrence remaining an important clinical issue. Current relapse predictive models in HR+/HER2- breast cancer patients still have limitations. The integration of multidimensional data represents a promising alternative for predicting relapse. In this study, we leverage our multi-omics cohort comprising 579 HR+/HER2- breast cancer patients (200 patients with complete data across 7 modalities) and develop a machine-learning-based model, namely CIMPTGV, which integrates clinical information, immunohistochemistry, metabolomics, pathomics, transcriptomics, genomics, and copy number variations to predict recurrence risk of HR+/HER2- breast cancer. This model achieves concordance indices (C-indices) of 0.871 and 0.869 in the train and test sets, respectively. The risk population predicted by the CIMPTGV model encompasses those identified by single-modality models. Feature analysis reveals that synergistic and complementary effects exist in different modalities. Simultaneously, we develop a simplified model with a mean area under the curve (AUC) of 0.840, presenting a useful approach for clinical applications.
Collapse
Affiliation(s)
- Hang Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Fan Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Shen Zhao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China.
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R.China.
| |
Collapse
|
36
|
Wu X, Yuan F, Guo L, Gao D, Zheng W, Chen C, Zheng H, Liu J. Intraductal chemotherapy for triple-negative breast cancer: a pathway to minimally invasive clinical treatment. BMC Cancer 2025; 25:285. [PMID: 39966717 PMCID: PMC11837698 DOI: 10.1186/s12885-025-13693-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is traditionally treated with systemic chemotherapy, often resulting in significant off-target toxicity. In this study, we assess the efficacy of intraductal chemotherapeutic delivery, aimed at reducing systemic side effects. Using an in situ TNBC model, created by intraductal injection of 4T1-luc cells, we identified day 3 post-tumor implantation as an optimal early intervention point. Echocardiographic analysis confirmed that intraductal administration of eribulin (ERI) or doxorubicin (DOX) did not cause cardiac dysfunction or apoptosis. Our results demonstrate that intraductal delivery of ERI and DOX significantly enhances anti-tumor and anti-metastatic effects. Mechanistically, ERI followed by DOX increased intratumoral perfusion, improved drug concentration, reversed epithelial-mesenchymal transition, and inhibited tumor cell invasion and metastasis. Additionally, this approach triggered immunogenic cell death and activated a systemic anti-tumor immune response. These findings underscore the potential of intraductal chemotherapy as a safe, highly effective approach, offering a preclinical foundation for minimally invasive TNBC therapies.
Collapse
Affiliation(s)
- Xinhong Wu
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China
| | - Feng Yuan
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China
| | - Liantao Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Dongcheng Gao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou City, Henan Province, China
| | - Weijie Zheng
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuchang District, Wuhan, 430060, People's Republic of China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuchang District, Wuhan, 430060, People's Republic of China.
| | - Hongmei Zheng
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China.
| | - Jianhua Liu
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China.
| |
Collapse
|
37
|
Ma H, Li J. Impact of HER2-targeting antibody drug conjugates in treatment strategies for patients with breast cancer. Heliyon 2025; 11:e41590. [PMID: 39916839 PMCID: PMC11799954 DOI: 10.1016/j.heliyon.2024.e41590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 02/09/2025] Open
Abstract
Antibody drug conjugates (ADCs) are novel drugs that exert specific cytotoxicity against breast cancer. Although ADCs such as trastuzumab emtansine and trastuzumab deruxtecan have significantly improved survival for patients with breast cancer expressing HER2, there is still controversy over options after ADCs. The radiotherapy and ablation should also be used as an effective strategy for oligoprogressions. Herein, we conducted a review of ADCs, and then discussed several strategies to maximize the potential benefit to patients with HER2 expression breast cancer.
Collapse
Affiliation(s)
- Hanghang Ma
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
- Outpatient Department of the 55th Retired Cadre Rest Center in Haidian District, Beijing, China
| | - Jianbin Li
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Cătană A, Iordănescu I, Filip GG, Filip S, Militaru MS, Pătrășcanu AA, Pîrlog LM. Quality-of-Life Assessment in Patients Undergoing Mastectomy and Breast Reconstruction for Moderate-Penetrance Gene-Related Breast Cancer. J Clin Med 2025; 14:1140. [PMID: 40004670 PMCID: PMC11856230 DOI: 10.3390/jcm14041140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background. Breast cancer remains a leading cause of cancer-related death among women, with genetic mutations playing a key role. While high-penetrance mutations are well-studied, moderate-to-low-penetrance mutations, which present challenges in clinical decision-making and patient outcomes, are less understood. This study explores the quality of life of breast cancer patients with moderate-penetrance mutations, focusing on the psychosocial and physical consequences of mastectomy and reconstruction to improve patient-centered care. Materials and Methods. A cohort of 620 breast cancer patients treated at Regina Maria Private Health Network, Bucharest, between January 2022 and July 2024 was identified. From this group, 61 patients were selected based on the following criteria: (1) meeting NCCN genetic testing guidelines, (2) carrying moderate-to-low-penetrance mutations, (3) undergoing bilateral mastectomy with double reconstruction, and (4) agreeing to complete a modified version of the BREAST-Q questionnaire. Genetic testing was performed using a 125-gene next-generation sequencing panel. Statistical analyses included non-parametric tests to examine group differences and correlations. Results. Significant correlations were found between several factors. Emotional distress was positively correlated with concerns for family, while couple relationships and financial burden showed a strong positive association. Negative correlations were found between couple relationships and self-concept. Distress levels varied, with "Interference with personal relationships" causing more distress than "Impact on employment", and financial burden causing more distressing than impact on sexuality. Conclusions. Prophylactic mastectomy significantly reduces cancer risk for women with moderate-penetrance mutations. This study highlights the relationship between surgical choices and quality-of-life factors, advancing personalized prevention strategies and emphasizing patient-centered care.
Collapse
Affiliation(s)
- Andreea Cătană
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (M.S.M.)
- Department of Oncogenetics, Institute of Oncology, “Prof. Dr. I. Chiricuță”, 400015 Cluj-Napoca, Romania
- Regional Laboratory Cluj-Napoca, Department of Medical Genetics, Regina Maria Health Network, 400363 Cluj-Napoca, Romania
| | - Irina Iordănescu
- Genetic Centre Laboratory, Department of Medical Genetics, Regina Maria Health Network, 011376 Bucharest, Romania;
| | | | - Simona Filip
- Ponderas Academic Hospital Bucharest, 014142 Bucharest, Romania; (G.G.F.); (S.F.)
| | - Mariela Sanda Militaru
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (M.S.M.)
- Regional Laboratory Cluj-Napoca, Department of Medical Genetics, Regina Maria Health Network, 400363 Cluj-Napoca, Romania
| | - Andrada-Adelaida Pătrășcanu
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (M.S.M.)
| | - Lorin-Manuel Pîrlog
- Department of Molecular Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania; (A.C.); (M.S.M.)
| |
Collapse
|
39
|
Shao C, Zhou H, Chen C, Dettman EJ, Ren Y, Cristescu R, Gozman A, Jin F. Prevalence of Homologous Recombination Repair Mutations and Association with Clinical Outcomes in Patients with Solid Tumors: A Study Using the AACR Project GENIE Dataset. Cancers (Basel) 2025; 17:577. [PMID: 40002171 PMCID: PMC11852487 DOI: 10.3390/cancers17040577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Mutations in BRCA1 and/or BRCA2 (BRCAm) and other homologous recombination repair genes (HRRm) are associated with several cancers. We evaluated the prevalence and association with overall survival (OS) of somatic BRCAm and HRRm among patients with advanced solid tumors. METHODS We used deidentified data from the AACR GENIE Biopharma Collaborative dataset derived from patients with tumors genotyped using next-generation sequencing between 1 January 2014 and 31 December 2017. Eligible patients were aged ≥18 years diagnosed with non-small-cell lung, colorectal, breast, bladder, prostate, or pancreatic cancer, with documented BRCA/HRR somatic mutation status. The primary analysis was OS (censored at the start of poly[ADP ribose] polymerase inhibitors [PARPi]/immunotherapy) after initiation of second-line therapy since most patients had sequencing after first-line therapy. RESULTS Among eligible patients, 242/7022 (3.4%) had BRCAm and 477/5474 (8.7%) had HRRm. Adjusted OS HRs (95% CI) for the primary analysis (using the initiation of second-line therapy as index date) were 0.79 (0.61-1.03) with/without BRCAm (n = 116/n = 3394) and 0.83 (0.69-0.99) with/without HRRm (n = 247/n = 2656); in sensitivity analysis of patients with stage IV disease, HRs were 0.97 (0.68-1.38) with/without BRCAm (n = 58/n = 1847) and 0.92 (0.73-1.18) with/without HRRm (n = 132/n = 1488). CONCLUSIONS Overall, BRCAm and HRRm did not show a strong association with OS, with a trend toward longer OS among patients receiving standard second-line therapies excluding PARPi/immunotherapy.
Collapse
Affiliation(s)
- Changxia Shao
- Merck & Co., Inc., Rahway, NJ 07065, USA; (H.Z.); (C.C.); (E.J.D.); (Y.R.); (R.C.); (A.G.); (F.J.)
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [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: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
41
|
Xiao M, Tang R, Pan H, Yang J, Tong X, Xu H, Guo Y, Lei Y, Wu D, Lei Y, Han Y, Ma Z, Wang W, Xu J, Yu X, Shi S. TPX2 serves as a novel target for expanding the utility of PARPi in pancreatic cancer through conferring synthetic lethality. Gut 2025; 74:410-423. [PMID: 39500552 PMCID: PMC11874363 DOI: 10.1136/gutjnl-2024-332782] [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: 05/03/2024] [Accepted: 10/14/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND PARP inhibitors (PARPi) have been licensed for the maintenance therapy of patients with metastatic pancreatic cancer carrying pathogenic germline BRCA1/2 mutations. However, mutations in BRCA1/2 are notably rare in pancreatic cancer. OBJECTIVE There is a significant unmet clinical need to broaden the utility of PARPi. DESIGN RNA sequencing was performed to screen potential targets for PARPi sensitivity. The synthetic lethal effects were verified in patient-derived xenograft (PDX), xenograft and patient-derived organoid models. Mechanisms were explored via LC‒MS/MS, coimmunoprecipitation, laser microirradiation, immunofluorescence, the homologous recombination (HR) or non-homologous end joining (NHEJ) reporter system, in situ proximity ligation assay and live-cell time-lapse imaging analyses. RESULTS Targeting protein for Xenopus kinesin-like protein 2 (TPX2) is an exploitable vulnerability. TPX2 was downregulated in PDX models sensitive to PARPi, and TPX2 inhibition conferred synthetic lethality to PARPi both in vitro and in vivo. Mechanistically, TPX2 functions in a cell cycle-dependent manner. In the S/G2 phase, ATM-mediated TPX2 S634 phosphorylation promotes BRCA1 recruitment to double-strand breaks (DSBs) for HR repair, whereas non-phosphorylated TPX2 interacts with 53BP1 to recruit it for NHEJ. The balance between phosphorylated and non-phosphorylated TPX2 determines the DSB repair pathway choice. During mitosis, TPX2 phosphorylation enhances Aurora A activity, promoting mitotic progression and chromosomal stability. Targeting TPX2 S634 phosphorylation with a cell-penetrating peptide causes genomic instability and mitotic catastrophe and enhances PARPi sensitivity. Additionally, the inhibition of TPX2 or S634 phosphorylation combined with gemcitabine further sensitised pancreatic cancer to PARPi. CONCLUSIONS Our findings revealed the dual-functional significance of TPX2 in controlling DNA DSB repair pathway choice and mitotic progression, suggesting a potential therapeutic strategy involving PARPi for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Mingming Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Haoqi Pan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Jing Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Xuhui Tong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - He Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Yanmei Guo
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Yalan Lei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Di Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Yubin Lei
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yamei Han
- Department of Biochemistry and Molecular Biology,Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Shanghai Pancreatic Center Institute, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
- Pancreatic Center Institute, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Nakamura S, Kojima Y, Takeuchi S. Causative Genes of Homologous Recombination Deficiency (HRD)-Related Breast Cancer and Specific Strategies at Present. Curr Oncol 2025; 32:90. [PMID: 39996890 PMCID: PMC11854191 DOI: 10.3390/curroncol32020090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Recently, homologous recombination deficiency (HRD) has become a new target for hereditary cancers. Molecular-based approaches for hereditary cancers in the clinical setting have been reviewed. In particular, the efficacy of the PARP inhibitor has been considered by several clinical trials for various kinds of hereditary cancers. This indicates that the PARP inhibitor can be effective for any kind of BRCA mutated cancers, regardless of the organ-specific cancer. Homologous recombination deficiency (HRD) has become a new target for hereditary cancers, indicating the necessity to confirm the status of HRD-related genes. ARID1A, ATM, ATRX, PALB2, BARD1, RAD51C and CHEK2 are known as HRD-related genes for which simultaneous examination as part of panel testing is more suitable. Both surgical and medical oncologists should learn the basis of genetics including HRD. An understanding of the basic mechanism of homologous repair recombination (HRR) in BRCA-related breast cancer is mandatory for all surgical or medical oncologists because PARP inhibitors may be effective for these cancers and a specific strategy of screening for non-cancers exists. The clinical behavior of each gene should be clarified based on a large-scale database in the future, or, in other words, on real-world data. Firstly, HRD-related genes should be examined when the hereditary nature of a cancer is placed in doubt after an examination of the relevant family history. Alternatively, HRD score examination is a solution by which to identify HRD-related genes at the first step. If lifetime risk is estimated at over 20%, an annual breast MRI is necessary for high-risk screening. However, there are limited data to show its benefit compared with BRCA. Therefore, a large-scale database, including clinical information and a long-term follow-up should be established, after which a periodical assessment is mandatory. The clinical behavior of each gene should be clarified based on a large-scale database, or, in other words, real-world data.
Collapse
Affiliation(s)
- Seigo Nakamura
- Institute for Clinical Genetics and Genomics, Showa University, Tokyo 142-8555, Japan; (Y.K.); (S.T.)
- Division of Breast Surgical Oncology, Department of Surgery, Showa University, Tokyo 142-8666, Japan
| | - Yasuyuki Kojima
- Institute for Clinical Genetics and Genomics, Showa University, Tokyo 142-8555, Japan; (Y.K.); (S.T.)
- Division of Breast Surgical Oncology, Department of Surgery, Showa University, Tokyo 142-8666, Japan
| | - Sayoko Takeuchi
- Institute for Clinical Genetics and Genomics, Showa University, Tokyo 142-8555, Japan; (Y.K.); (S.T.)
| |
Collapse
|
43
|
Xu L, Xu P, Wang J, Ji H, Zhang L, Tang Z. Advancements in clinical research and emerging therapies for triple-negative breast cancer treatment. Eur J Pharmacol 2025; 988:177202. [PMID: 39675457 DOI: 10.1016/j.ejphar.2024.177202] [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: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Triple-negative breast cancer (TNBC), defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor 2 (HER2) expression, is acknowledged as the most aggressive form of breast cancer (BC), comprising 15%-20% of all primary cases. Despite the prevalence of TNBC, effective and well-tolerated targeted therapies remain limited, with chemotherapy continuing to be the mainstay of treatment. However, the horizon is brightened by recent advancements in immunotherapy and antibody-drug conjugates (ADCs), which have garnered the U.S. Food and Drug Administration (FDA) approval for various stages of TNBC. Poly (ADP-ribose) polymerase inhibitors (PARPi), particularly for TNBC with BRCA mutations, present a promising avenue, albeit with the challenge of resistance that must be addressed. The success of phosphoinositide-3 kinase (PI3K) pathway inhibitors in hormone receptor (HR)-positive BC suggests potential applicability in TNBC, spurring optimism within the research community. This review endeavors to offer a comprehensive synthesis of both established and cutting-edge targeted therapies for TNBC. We delve into the specifics of PARPi, androgen receptor (AR) inhibitors, Cancer stem cells (CSCs), PI3K/Protein Kinase B (AKT)/mammalian target of rapamycin (mTOR), the transforming growth factor-beta (TGF-β), Ntoch, Wnt/β-catenin, hedgehog (Hh) pathway inhibitors, Epigenetic target-mediated drug delivery, ADCs, immune checkpoint inhibitors (ICIs)and novel immunotherapeutic solutions, contextualizing TNBC within current treatment paradigms. By elucidating the mechanisms of these drugs and their prospective clinical applications, we aim to shed light on the challenges and underscore the beacon of hope that translational research and innovative therapies represent for the oncology field.
Collapse
Affiliation(s)
- Lili Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Pengtao Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, 628000, China
| | - Hui Ji
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
44
|
Berton Giachetti PPM, Morganti S, Gandini S, Giudici F, Marra A, Nicolò E, Zattarin E, Corti C, Boldrini L, Verrazzo A, Sposetti C, Razeti MG, Carnevale Schianca A, Scafetta R, Taurelli Salimbeni B, Esposito A, Zagami P, Trapani D, Malagutti B, Caputo R, Vernieri C, Munzone E, Scagnoli S, Botticelli A, Lambertini M, Giuliano M, De Laurentiis M, Viale G, Bianchini G, Curigliano G, De Angelis C, Criscitiello C. Survival Following CDK4/6 Inhibitor Therapy for Hormone Receptor-Positive, ERBB2-Negative Metastatic Breast Cancer. JAMA Netw Open 2025; 8:e2461067. [PMID: 39982725 PMCID: PMC11846014 DOI: 10.1001/jamanetworkopen.2024.61067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/09/2024] [Indexed: 02/22/2025] Open
Abstract
Importance Endocrine therapy (ET) combined with cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) agents is the standard first-line treatment for patients with hormone receptor-positive, ERBB2 (formerly HER2 or HER2/neu)-negative metastatic breast cancer. However, optimal therapy after tumor progression to ET plus CDK4/6i remains unclear. Objective To evaluate progression-free survival (PFS) and overall survival (OS) in the clinical practice setting in patients with hormone receptor-positive, ERBB2-negative metastatic breast cancer following progression with ET plus CDK4/6i. Design, Setting, and Participants The multicenter retrospective cohort study included 506 patients diagnosed with hormone receptor-positive, ERBB2-negative metastatic breast cancer between April 22, 2015, and January 31, 2023, and who received ET-based or chemotherapy (CT)-based treatment following progression during ET plus CDK4/6i. Outcomes were analyzed based on treatment type, clinicopathologic features, and the duration of prior CDK4/6i therapy. Main Outcomes and Measures The primary end point was PFS in the clinical practice setting, defined as the time between the initiation of the first systemic treatment on tumor progression to ET plus CDK4/6i treatment and the detection of disease progression or patient death from any cause. The secondary end point was OS in the clinical practice setting, defined as the time interval between tumor progression during ET plus CDK4/6i treatment and patient death from any cause. Results In 506 women (median age at diagnosis, 52.4 [IQR, 44.6-62.8] years) diagnosed with hormone receptor-positive, ERBB2-negative metastatic breast cancer progressing during ET plus CDK4/6i, independent factors associated with poorer PFS outcomes were visceral metastases (hazard ratio [HR], 1.45; 95% CI, 1.17-1.80; P = .008) and de novo metastatic disease (HR, 1.25; 95% CI, 1.01-1.54; P = .04). A longer duration of CDK4/6i therapy (OS HR, 0.55; 95% CI, 0.41-0.73; P < .001) and an older age (PFS HR, 0.99; 95% CI 0.98-1.00; P = .03) were associated with better outcomes. Compared with oral CT, both intravenous CT- and ET-based treatments were associated with shorter PFS (intravenous CT: hazard ratio [HR], 1.45; 95% CI, 1.11-1.89; P = .006; everolimus plus exemestane: HR, 1.38; 95% CI, 1.06-1.78; P = .02; ET only: HR, 1.38; 95% CI, 1.05-1.89; P = .02). A duration of CDK4/6i treatment exceeding 12 months was associated with longer OS (HR, 0.55; 95% CI, 0.41-0.73; P < .001). Among patients with visceral metastases, intravenous CT was associated with shorter OS compared with oral CT (HR, 1.52; 95% CI, 1.03-2.24; P = .04). Conclusions and Relevance In this cohort study, the duration of tumor control achieved with CDK4/6i-based therapy and the presence of visceral metastases emerged as key factors that may affect treatment decision. Oral CT may offer potential benefits for specific patient subgroups.
Collapse
Affiliation(s)
- Pier Paolo Maria Berton Giachetti
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Stefania Morganti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Fabiola Giudici
- Cancer Epidemiology Unit, Centro di Riferimento Oncologico di Aviano IRCCS, Aviano, Italy
| | - Antonio Marra
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Eleonora Nicolò
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, New York
| | - Emma Zattarin
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Corti
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Laura Boldrini
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Annarita Verrazzo
- Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Pascale, Naples, Italy
| | - Caterina Sposetti
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Grazia Razeti
- Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Ambra Carnevale Schianca
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Roberta Scafetta
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128, Roma, Italy
| | | | - Angela Esposito
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Paola Zagami
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Dario Trapani
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Bianca Malagutti
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Roberta Caputo
- Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Pascale, Naples, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Elisabetta Munzone
- Division of Medical Senology, Research Unit in Medical Senology, European Institute of Oncology IRCCS, Milan, Italy
| | - Simone Scagnoli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Andrea Botticelli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Michelino De Laurentiis
- Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Pascale, Naples, Italy
| | - Giulia Viale
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giampaolo Bianchini
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giuseppe Curigliano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Carmen Criscitiello
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
45
|
Kaseki S, Sonehara R, Motooka Y, Tanaka H, Nakamura T, Osuka S, Akatsuka S, Kajiyama H, Mashimo T, Imaoka T, Toyokuni S. Susceptibility of Brca1 (L63X/+) rat to ovarian reserve dissipation by chemotherapeutic agents to breast cancer. Cancer Sci 2025. [PMID: 39901592 DOI: 10.1111/cas.16412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 02/05/2025] Open
Abstract
BRCA1 is one of the causative genes for hereditary breast and ovarian cancer syndrome with a high risk of early-onset breast cancer. Whereas olaparib (OLA), an inhibitor of poly-ADP-ribose polymerase, has been applied as adjuvant therapy to those cancer patients, its effect on ovarian reproductive function remains unelucidated. Recently, a rat model (MUT; Brca1(L63X/+) mutation) mimicking a human BRCA1 pathogenic variant has been established. Using this model, we evaluated the effects of OLA on ovarian reproductive function in comparison with the wild-type (WT) rats. MUT showed a significantly reduced number of primordial follicles and subfertility in accordance with aging. Oxidative stress was significantly elevated in the young MUT granulosa cells (GCs) accompanied by increased mTOR but decreased PTEN signals. OLA administration in MUT further decreased primordial follicles, with gene set enrichment analysis, indicating upregulated DNA repair pathways. Furthermore, a combination of OLA and cyclophosphamide (CPA) induced empty primordial follicles, recognized as CPA-induced severe ovarian toxicity. Whereas OLA + CPA caused greater reduction in primordial follicles both in MUT and WT in comparison with CPA alone, MUT ovaries were more susceptible to oxidative stress, potentially depleting primordial follicles via activation of GCs and inducing oocyte death due to accumulated DNA damage by OLA treatment. Our findings in this preclinical model underscore the importance of evaluating ovarian reserve prior to chemotherapy by performing reproductive consultation with female patients with BRCA1 pathogenic variants.
Collapse
Affiliation(s)
- Satoshi Kaseki
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reina Sonehara
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yashiro Motooka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideaki Tanaka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Nakamura
- Department of Maternal and Perinatal Medicine, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Akatsuka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Genome Engineering, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
- Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| |
Collapse
|
46
|
Demarest K, Anantharajah A, Maxwell KN, Rohanizadegan M, Bradbury A, Nathanson KL, McCarthy AM, Domchek SM, Nayak A, Shah PD. Pathogenic Germline Variants in Patients With Metaplastic Breast Cancer. JAMA Netw Open 2025; 8:e2460312. [PMID: 39964682 PMCID: PMC11836754 DOI: 10.1001/jamanetworkopen.2024.60312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/13/2024] [Indexed: 02/21/2025] Open
Abstract
Importance Metaplastic breast cancer (MpBC) is a rare, heterogeneous disease often associated with inferior outcomes. A growing body of literature describes the clinical and molecular features of MpBC, yet limited data describe the pathogenic germline variants (PGVs) in breast cancer susceptibility genes among affected individuals. Objective To examine the frequency and types of PGVs in breast cancer genes among patients with MpBC. Design, Setting, and Participants This is a descriptive retrospective cohort study of patients who received a diagnosis of MpBC at the University of Pennsylvania between January 2010 and May 2023. Electronic medical records were reviewed for demographic, clinicopathologic, and germline genetic testing information. Germline variant status was independently confirmed by a licensed genetic counselor and a physician with expertise in genetics. MpBC diagnosis and subtype were confirmed by a breast pathologist. Participants were identified via query of an institutional pathology database for reports signed between January 2010 and May 2023 including the term metaplastic. Among 320 initially obtained reports, 272 individuals had confirmed MpBC and were included in the study. Exposure Germline genetic testing to investigate the presence of PGVs in breast cancer susceptibility genes. Main Outcomes and Measures The primary outcome measurement was the prevalence of PGVs in breast cancer susceptibility genes among participants. The hypothesis that individuals with MpBC have an enrichment of PGVs in genes associated with inherited breast cancer risk was formulated before data collection. Results The total sample size was 272 women, and the median age at diagnosis was 58 years (range, 20-102 years); all were biological female patients; 143 of 272 (52.6%) had documentation of germline genetic testing; and participants with testing were significantly younger than those without (median age, 53 years [range, 20-79 years] vs 63 years [range, 29-102 years]; P < .001). Of the 143 patients, 24 (16.8%) had a PGV in a breast cancer susceptibility gene (BRCA1, n = 17; BRCA2, n = 5; PALB2, n = 1; CHEK2, n = 1). Patients with PGV-associated MpBC received a diagnosis at a younger age than those with sporadic disease, but there were no significant differences in hormone receptor positivity, ERBB2 status, or metaplastic subtype. Conclusions and Relevance In this cohort study of patients with MpBC, a substantial proportion of clinically tested patients had a PGV in a breast cancer susceptibility gene, most commonly BRCA1. Germline testing was high yield in patients with MpBC, many of whom would be included in current germline testing eligibility criteria.
Collapse
Affiliation(s)
- Kaitlin Demarest
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
| | | | - Kara N. Maxwell
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
- Department of Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Mersedeh Rohanizadegan
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
- Department of Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
- Department of Medicine, Division of Translational Medicine & Human Genetics, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
| | - Angela Bradbury
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
- Department of Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Katherine L. Nathanson
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
- Department of Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Anne Marie McCarthy
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia
| | - Susan M. Domchek
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
- Department of Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
| | - Payal D. Shah
- Basser Center for BRCA, University of Pennsylvania, Philadelphia
- Department of Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia
- Abramson Cancer Center, University of Pennsylvania, Philadelphia
| |
Collapse
|
47
|
Milella M, Orsi G, di Marco M, Salvatore L, Procaccio L, Noventa S, Bozzarelli S, Garajova I, Vasile E, Giordano G, Macchini M, Cavaliere A, Gaule M, Bergamo F, Chiaravalli M, Palloni A, Carloni R, Bittoni A, Niger M, Rapposelli IG, Rodriquenz MG, Scartozzi M, Mosconi S, Giommoni E, Bernardini I, Paratore C, Spallanzani A, Bencardino K, Forti L, Tamburini E, Lonardi S, Scarpa A, Cascinu S, Tortora G, Sperduti I, Reni M. Real-World Impact of Olaparib Exposure in Advanced Pancreatic Cancer Patients Harboring Germline BRCA1-2 Pathogenic Variants. Cancer Med 2025; 14:e70364. [PMID: 39861955 PMCID: PMC11761426 DOI: 10.1002/cam4.70364] [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: 01/14/2024] [Revised: 04/07/2024] [Accepted: 10/13/2024] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION Pancreatic cancer arising in the context of BRCA predisposition may benefit from poly(ADP-ribose) polymerase inhibitors. We analyzed real-world data on the impact of olaparib on survival in metastatic pancreatic cancer patients harboring germline BRCA mutations in Italy, where olaparib is not reimbursed for this indication. METHODS Clinico/pathological data of pancreatic cancer patients with documented BRCA1-2 germline pathogenic variants who had received first-line chemotherapy for metastatic disease were collected from 23 Italian oncology departments and the impact of olaparib exposure on overall survival (OS) was analyzed. RESULTS Of 114, 53 BRCA-mutant pancreatic cancer patients had received olaparib for metastatic disease. OS was significantly longer in patients who were exposed to olaparib (hazard ratio [HR] 0.568, 95% confidence interval [CI] 0.351-0.918, log-rank p = 0.02) in any setting/line of treatment; similar results were obtained for patients who received olaparib as maintenance treatment (in any line of treatment), patients who had stage IV disease at diagnosis, and patients who did not experience progressive disease as their best response to first-line chemotherapy. Exposure to olaparib in the first-line maintenance setting after platinum-based chemotherapy, however, did not significantly impact survival. At multivariate analysis, CA19.9 levels at diagnosis and response to first-line chemotherapy were independently prognostic; however, when response to chemotherapy was excluded, any exposure to olaparib was a significant independent predictor of longer OS, together with CA19.9 levels. CONCLUSION The real-world data presented here support the use of olaparib for metastatic disease in germline BRCA-mutant pancreatic cancer patients, as it may significantly prolong survival.
Collapse
|
48
|
Blondeaux E, Boni L, Chilà G, Dri A, Caputo R, Poggio F, Fabi A, Arpino G, Pravisano F, Geuna E, Delucchi V, Ruelle T, Giannubilo I, De Laurentiis M, Puglisi F, Bighin C, Lambertini M, Montemurro F, Del Mastro L. Factors associated with first-to-second-line attrition among patients with metastatic breast cancer in the real world. ESMO Open 2025; 10:104125. [PMID: 39842245 PMCID: PMC11794064 DOI: 10.1016/j.esmoop.2024.104125] [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: 08/30/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Estimating patient attrition across lines of treatment (i.e. the probability that upon treatment failure the patient will not be able to receive a subsequent treatment) may be a valuable tool for optimizing treatment sequencing. We sought to describe the first-to-second-line attrition rate and factors associated with attrition in a real-world cohort of patients with metastatic breast cancer. METHODS The Gruppo Italiano Mammella (GIM)14/BIO-META (NCT02284581) is an ongoing, ambispective observational multicenter study enrolling patients with metastatic breast cancer receiving first-line therapy. In patients experiencing disease progression, attrition was defined as no further anticancer treatment and death within 6 months from the end of first-line therapy. The attrition rate from the first-to-second line was studied by descriptive analyses and univariate and multivariable logistic models were used to explore potentially predictive factors. RESULTS From January 2000 to December 2021, 3109 patients with metastatic breast cancer were enrolled in the GIM14/BIO-META study. Among them, 2498 patients experienced first-line treatment failure. Overall, first-to-second line attrition was 9.0% (95% confidence interval 7.9% to 10.1%), with similar attrition for patients with hormone receptor-positive/HER2-negative (8.5%) and HER2-positive (7.1%) breast cancer. Patients with triple-negative disease experienced the highest attrition (13.0%). Age, menopausal status, disease-free interval from primary tumor diagnosis, type of metastatic spread, and tumor subtype independently predicted first-to-second-line attrition. CONCLUSIONS These findings could inform treatment decisions and guide clinical research on treatment sequencing. For instance, patients with the lowest risk of attrition may be the ideal candidates for trials exploring de-escalated first-line regimens, followed by more aggressive treatments upon progression.
Collapse
Affiliation(s)
- E Blondeaux
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova.
| | - L Boni
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova
| | - G Chilà
- Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo
| | - A Dri
- Department of Medical Oncology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine; Department of Medicine (DAME), University of Udine, Udine
| | - R Caputo
- Breast Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli
| | - F Poggio
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova
| | - A Fabi
- U.O. Oncologia Medica 1, IRCCS Regina Elena National Cancer Institute, Rome; Precision Medicine Unit in Breast, Department of Woman and Child Health and Public Health, IRCCS Fondazione Policlinico Universitario A. Gemelli, Roma
| | - G Arpino
- Department of Medical Oncology, Università di Napoli Federico II, Napoli
| | - F Pravisano
- Department of Medical Oncology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine; Department of Medicine (DAME), University of Udine, Udine
| | - E Geuna
- Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo
| | - V Delucchi
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova
| | - T Ruelle
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova
| | - I Giannubilo
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova
| | - M De Laurentiis
- Breast Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli
| | - F Puglisi
- Department of Medicine (DAME), University of Udine, Udine; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano IRCCS, Aviano
| | - C Bighin
- U.O. Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Genova
| | - M Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova; Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova
| | - F Montemurro
- Breast Unit, Candiolo Cancer Institute, FPO-IRCCs, Candiolo, Italy
| | - L Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova; Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova
| |
Collapse
|
49
|
Shiri I, Salimi Y, Mohammadi Kazaj P, Bagherieh S, Amini M, Saberi Manesh A, Zaidi H. Deep Radiogenomics Sequencing for Breast Tumor Gene-Phenotype Decoding Using Dynamic Contrast Magnetic Resonance Imaging. Mol Imaging Biol 2025; 27:32-43. [PMID: 39815134 PMCID: PMC11805855 DOI: 10.1007/s11307-025-01981-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE We aim to perform radiogenomic profiling of breast cancer tumors using dynamic contrast magnetic resonance imaging (MRI) for the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) genes. METHODS The dataset used in the current study consists of imaging data of 922 biopsy-confirmed invasive breast cancer patients with ER, PR, and HER2 gene mutation status. Breast MR images, including a T1-weighted pre-contrast sequence and three post-contrast sequences, were enrolled for analysis. All images were corrected using N4 bias correction algorithms. Based on all images and tumor masks, a bounding box of 128 × 128 × 68 was chosen to include all tumor regions. All networks were implemented in 3D fashion with input sizes of 128 × 128 × 68, and four images were input to each network for multi-channel analysis. Data were randomly split into train/validation (80%) and test set (20%) with stratification in class (patient-wise), and all metrics were reported in 20% of the untouched test dataset. RESULTS For ER prediction, SEResNet50 achieved an AUC mean of 0.695 (CI95%: 0.610-0.775), a sensitivity of 0.564, and a specificity of 0.787. For PR prediction, ResNet34 achieved an AUC mean of 0.658 (95% CI: 0.573-0.741), a sensitivity of 0.593, and a specificity of 0.734. For HER2 prediction, SEResNext101 achieved an AUC mean of 0.698 (95% CI: 0.560-0.822), a sensitivity of 0.750, and a specificity of 0.625. CONCLUSION The current study demonstrated the feasibility of imaging gene-phenotype decoding in breast tumors using MR images and deep learning algorithms with moderate performance.
Collapse
Affiliation(s)
- Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Yazdan Salimi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | | | - Sara Bagherieh
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Amini
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Abdollah Saberi Manesh
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva, Switzerland.
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
- Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
- University Research and Innovation Center, Óbuda University, Budapest, Hungary.
| |
Collapse
|
50
|
Lopetegui-Lia N, Varma R, Abraham J, Roesch E. Current and Novel Treatment Options in Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer. JCO Oncol Pract 2025; 21:145-154. [PMID: 39167745 DOI: 10.1200/op.23.00830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Metastatic breast cancer (mBC) remains an incurable disease, and most patients will experience disease progression during their treatment course. Although endocrine therapy remains the mainstay of treatment for hormone receptor-positive/human epidermal growth factor receptor 2-negative mBC, significant progress has been and continues to be made in the treatment of this BC subtype. The discovery of molecular markers, mutations in key cellular pathways, and genomic signatures have led to the development of novel and targeted agents, such as antibody-drug conjugates, oral selective estrogen receptor downregulators, and inhibitors of the PI3K/AKT/mTOR pathway. This has resulted in significant improvements in the survival and quality of life of patients. With the increasing number of treatment options for patients, appropriate drug sequencing remains a challenge. Treatment discussions should involve patient-physician shared decision making, with consideration of genomic data, previous lines of therapy, side effect profiles, and clinical trial enrollment.
Collapse
|