Erika Hamilton, Timothy Pluard, Judy S Wang, Aki Morikawa, Stephen Johnston, E Claire Dees, Christos Vaklavas, Anne Armstrong, Pamela Munster, Nisha Unni, Gail S Wright, Fadi Kayali, Tingting Song, Yuanxin Rong, Kohei Yamaguchi, Dejan Juric
{"title":"Phase 1/2 study of H3B-6545 in women with locally advanced/metastatic estrogen receptor-positive, HER2-negative breast cancer.","authors":"Erika Hamilton, Timothy Pluard, Judy S Wang, Aki Morikawa, Stephen Johnston, E Claire Dees, Christos Vaklavas, Anne Armstrong, Pamela Munster, Nisha Unni, Gail S Wright, Fadi Kayali, Tingting Song, Yuanxin Rong, Kohei Yamaguchi, Dejan Juric","doi":"10.1186/s13058-025-02069-8","DOIUrl":"10.1186/s13058-025-02069-8","url":null,"abstract":"","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"151"},"PeriodicalIF":5.6,"publicationDate":"2025-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12363031/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144884168","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Katherine W Reeves, Youssef Oulhote, Philippe Grandjean, Flemming Nielsen
{"title":"Associations of perfluoroalkyl substances (PFAS) with terminal ductal lobular unit involution of the normal breast.","authors":"Katherine W Reeves, Youssef Oulhote, Philippe Grandjean, Flemming Nielsen","doi":"10.1186/s13058-025-02103-9","DOIUrl":"10.1186/s13058-025-02103-9","url":null,"abstract":"<p><strong>Background: </strong>Per- and polyfluoroalkyl substances (PFAS) may be carcinogenic, and animal studies demonstrate their harmful effects on mammary gland development. Terminal ductal lobular units (TDLUs) are the structures that produce milk following childbirth, and involution of TDLUs normally occurs with aging. Most breast cancers arise from TDLUs, and a greater degree of TDLU involution may be associated with lower breast cancer risk. We estimated associations between PFAS concentrations and TDLU involution in normal breast tissue samples.</p><p><strong>Methods: </strong>Concentrations of seven PFAS were measured in serum provided by a subset of 263 cancer-free volunteer participants from the Susan G. Komen for the Cure Tissue Bank (KTB) who were postmenopausal, not currently using hormone therapy, and had available TDLU measurements made by a trained pathologist examining H&E stained section of a core biopsy sample of tissue from the outer upper quadrant of a single breast. Bayesian kernel machine regression and quantile-G computation were used to estimate covariate-adjusted associations between the PFAS mixture and measures of TDLU involution (presence of TDLUs, number of observed TDLUs, and median TDLU span) within this population and with stratification on parity and breastfeeding history.</p><p><strong>Results: </strong>TDLUs were observed in breast tissue samples of 40.3% (N = 106) of the study population, with similar PFAS concentrations between participants with and without observed TDLUs. No strong, statistically significant associations were observed between individual PFAS and presence of observed TDLUs. The overall effect of the PFAS mixture suggested an inverted U-shaped association with odds of observed TDLUs, although this was not statistically significant (β = 0.03 95% CI -2.75, 2.81; p = 0.98). Among the subgroup of parous women, stratified analyses suggested a positive association between the PFAS mixture and observed TDLUs among those who had ever breastfed, but a slightly negative association among those who had never breastfed.</p><p><strong>Conclusions: </strong>Overall, our analysis does not support meaningful effects of PFAS on TDLU involution, although we note that these findings are not applicable to premenopausal women or to postmenopausal women using hormone therapy.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"148"},"PeriodicalIF":5.6,"publicationDate":"2025-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12363039/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144876375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Matthew Dankner, Veronique Ouellet, Jakob Lafon, Rima Ezzeddine, Matthew G Annis, Afnan Abu-Thuraia, Peter M Siegel
{"title":"Cold-inducible RNA-binding protein is associated with subtype-specific breast cancer patient outcomes.","authors":"Matthew Dankner, Veronique Ouellet, Jakob Lafon, Rima Ezzeddine, Matthew G Annis, Afnan Abu-Thuraia, Peter M Siegel","doi":"10.1186/s13058-025-02098-3","DOIUrl":"10.1186/s13058-025-02098-3","url":null,"abstract":"<p><strong>Background: </strong>Cold-inducible RNA-binding protein (CIRBP) is a stress-induced mRNA-binding protein associated with clinical outcomes in a variety of human disease states. The role of CIRBP as a role as a prognostic biomarker in breast cancer (BC) has yet to be established.</p><p><strong>Findings: </strong>We describe a clinically annotated tissue micro-array cohort of 1406 hormone receptor positive (HR +) and 281 triple negative primary breast cancers (TNBC) stained by immunohistochemistry (IHC) for CIRBP. Statistical analyses were performed with the Kaplan-Meier estimator, as well as univariate and multivariate Cox proportional-hazards models. Multivariate models incorporated tumor size, lymph node status, grade and CIRBP expression levels. Co-primary endpoints were overall survival (OS) and progression-free survival (PFS). In N = 281 primary TNBCs, high levels of CIRBP expression by IHC was associated with poor prognosis in multivariate analysis (OS: adjusted hazard ratio (aHR) 2.05, 95% confidence interval (CI) 1.24-3.41, P = 0.005. PFS: aHR 2.46, 95% CI 1.33-4.57, P = 0.004). However, in N = 1406 HR + primary BC, CIRBP expression was correlated with favorable prognosis (OS: aHR 0.927, 95% CI 0.88-0.98, P = 0.005. PFS: aHR 0.904, 95% CI 0.85-0.96, P = 0.002).</p><p><strong>Conclusions: </strong>CIRBP expression is associated with poor prognosis in TNBC but not HR + BC patients. This finding highlights the prognostic significance of CIRBP in TNBC and suggests differential underlying mRNA targets bound and modulated by CIRBP in TNBC and HR + BC, respectively.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"149"},"PeriodicalIF":5.6,"publicationDate":"2025-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12363121/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144876376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ruchi Patel, Elizabeth Weil, Sara Bugamelli, Emma Carroll, Kylie Steinke, Erinn Stockhausen, John Burfeind, Colin Mooney, Janet Retseck, Deepika Sriram, Jutta Deininger, Angela Halbach, Maressa Sweeney, Sailaja Kamaraju, Lubna N Chaudhary, Yee Chung Cheng
{"title":"Evaluation and management of incomplete ovarian function suppression in premenopausal breast cancer patients receiving anti-hormone therapy.","authors":"Ruchi Patel, Elizabeth Weil, Sara Bugamelli, Emma Carroll, Kylie Steinke, Erinn Stockhausen, John Burfeind, Colin Mooney, Janet Retseck, Deepika Sriram, Jutta Deininger, Angela Halbach, Maressa Sweeney, Sailaja Kamaraju, Lubna N Chaudhary, Yee Chung Cheng","doi":"10.1186/s13058-025-01979-x","DOIUrl":"10.1186/s13058-025-01979-x","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer is the most common malignancy among females. The Tamoxifen and Exemestane Trial (TEXT) and the Suppression of Ovarian Function Trial (SOFT) were two landmark studies, which showed adjuvant therapy with the aromatase inhibitor (AI), exemestane, or tamoxifen combined with ovarian function suppression (OFS) improved disease-free survival (DFS) among premenopausal women (Pagani et al. in N Engl J Med 371(2):107-118, 2014). However, data has since emerged regarding incomplete OFS with Gonadotropin-releasing hormone (GnRH) agonists in some premenopausal patients. The SOFT Estrogen Substudy (SOFT-EST) was a prospective substudy of SOFT, which evaluated estradiol (E2) levels to determine if patients on exemestane and triptorelin experienced suboptimal ovarian function suppression. In this study, 17% of patients had an E2 level greater than 2.72 pg/mL at each time point, which was determined to be incomplete OFS. This study called into question whether E2 levels should routinely be monitored in women on GnRH agonists (Bellet et al. in J Clin Oncol 34(14):1584-1593, 2016). The National Comprehensive Cancer Network (NCCN) and the American Society of Clinical Oncology (ASCO) provide recommendations to monitor E2 levels in premenopausal patients on an aromatase inhibitor (AI) due to the concern of incomplete OFS for patients receiving GnRH agonist therapy (Burstein et al. in J Clin Oncol 37(5):423-438, 2019, National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Breast Cancer. Version 2.2024. www.nccn.org ). The purpose of our study is to develop and implement a protocol for monitoring E2 levels at Froedtert & MCW based on the NCCN and ASCO recommendations.</p><p><strong>Methods: </strong>This study implemented an OFS monitoring guideline in combination with a collaborative practice agreement at Froedtert & MCW. Pharmacists were responsible for ordering and monitoring E2 levels monthly on eligible patients based on a specified protocol. An E2 level ≤ 2.72 pg/mL while receiving an AI or ≤ 21 pg/mL while receiving tamoxifen in addition to a GnRH agonist injection was defined as complete OFS. E2 levels were retrospectively analyzed to evaluate if patients had incomplete OFS and if clinical management changed based on those levels. The primary outcome was proportion of patients identified per the E2 monitoring protocol that fail to achieve OFS for two consecutive levels over an 18-month period. Nominal data were compared using the chi-squared or Fisher's exact test, and continuous data using the Mann Whitney U or student's t-test, where appropriate.</p><p><strong>Results: </strong>A total of 85 patients were reviewed at the time of analysis. Fifty-three patients (62.4%) achieved complete OFS (three consecutive E2 levels within goal), compared to 17 patients (20%) demonstrating incomplete OFS. Seven patients (8.2%) did not have enough consecutive levels demonstrating complete or incomplet","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"147"},"PeriodicalIF":5.6,"publicationDate":"2025-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12360005/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144876377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Miriam Pearl Klahr, Khadija Faheem, Rohit R Raghunathan, Margaux Wooster, Dawn L Hershman, Melissa K Accordino
{"title":"Association between hyperglycemia and the development of chemotherapy induced peripheral neuropathy among patients with breast cancer in the control trial.","authors":"Miriam Pearl Klahr, Khadija Faheem, Rohit R Raghunathan, Margaux Wooster, Dawn L Hershman, Melissa K Accordino","doi":"10.1186/s13058-025-01984-0","DOIUrl":"10.1186/s13058-025-01984-0","url":null,"abstract":"<p><strong>Background: </strong>Poorly controlled diabetes is a predictor of chemotherapy induced peripheral neuropathy (CIPN) and hyperglycemia may mediate CIPN risk. We evaluated the association between hyperglycemia and CIPN development.</p><p><strong>Findings: </strong>Methods: This was a secondary analysis of the CONTRoL trial, which randomized patients with Stage I-III breast cancer receiving taxane chemotherapy to cryotherapy, compression therapy, or placebo. CIPN data was obtained from the FACT-NTX survey which patients completed at baseline, week-12, and week-24. Hyperglycemia (glucose ≥ 140 mmol/L) was assessed using random glucose values obtained throughout trial duration. We included patients who completed FACT-NTX at baseline and week-12. Patients were divided into two groups based on CIPN development. For each group we calculated the proportion of patients with hyperglycemia and mean glucose values; 95% confidence intervals were constructed.</p><p><strong>Results: </strong>Fifty-nine patients met inclusion criteria; 34 patients developed CIPN and 25 did not. Hyperglycemia occurred in 47.1% vs. 36.0% of patients who developed CIPN vs. those who did not (p = 0.56). Mean glucose was numerically but not significantly higher in patients with CIPN compared to those without at all timepoints, most notably at week-12 (132.2 mg/dL vs. 122.9 mg/dL p = 0.47).</p><p><strong>Conclusions: </strong>In this analysis, hyperglycemia affected almost half of patients with CIPN versus about a third of patients without CIPN. Patients with CIPN also had higher mean glucose levels compared to those without CIPN, though the differences were not significant. No conclusions can be drawn from these results and further research is needed to assess these trends in a larger prospective patient population.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"150"},"PeriodicalIF":5.6,"publicationDate":"2025-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12363088/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144876374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lis Victoria Ravani, Zahra Bagheri, Adam M Brufsky, Isabella Michellon, Ruth O'Regan, Maryam Lustberg, Hyo Han, Ming Wang, Seth A Wander
{"title":"Comparative efficacy of first- versus second-line CDK4/6 inhibition in hormone receptor-positive, HER2-negative metastatic breast cancer.","authors":"Lis Victoria Ravani, Zahra Bagheri, Adam M Brufsky, Isabella Michellon, Ruth O'Regan, Maryam Lustberg, Hyo Han, Ming Wang, Seth A Wander","doi":"10.1186/s13058-025-02095-6","DOIUrl":"10.1186/s13058-025-02095-6","url":null,"abstract":"<p><strong>Purpose: </strong>The greater progression-free survival (PFS) improvements observed in first-line (1L) versus second-line (2L) CDK4/6 inhibitor (CDK4/6i) trials underpin current guideline recommendations establishing these agents as standard 1L therapy in hormone receptor-positive, HER2-negative (HR+/HER2-) metastatic breast cancer (mBC). While earlier CDK4/6i use is associated with increased cumulative toxicity and costs, comparative survival data of earlier versus deferred use remain scarce.</p><p><strong>Methods: </strong>We performed a systematic review and meta-analysis, searching PubMed, Embase, Cochrane, and conference proceedings for observational studies and randomized clinical trials (RCTs) including patients treated with first- and/or 2L CDK4/6i. Patients who received CDK4/6 inhibition as part of 1L therapy were included in the 1 L group, while those who did not receive CDK4/6i in the 1L setting and deferred it until the 2L setting were assigned to the 2 L group. Pooled analysis of Kaplan-Meier-derived individual patient data was conducted for PFS2, defined as time from randomization to progression on 2L therapy, and overall survival (OS). Both outcomes were measured from the start of 1L treatment to progression on 2L treatment for both 1L and 2L groups. Sensitivity analysis by study design was also conducted.</p><p><strong>Results: </strong>Nine studies (5 RCTs and 4 observational studies) comprising 7,602 patients with mBC were included. Of these, 6,475 (85.1%) received CDK4/6i in 1L, and 1,127 (14.8%) in the 2 L setting. Overall, 1L CDK4/6i therapy was associated with significantly longer PFS2 compared to 2L treatment (HR 2.08; 95%CI 1.90-2.27), a trend not observed in sensitivity analysis of RCTs alone (HR 1.10; 95%CI 0.94-1.30). No significant differences in OS were observed between 1L and 2L CDK4/6i regimens (HR 1.09; 95%CI 1.00-1.18), or in sensitivity analysis of only RCTs (HR 1.03; 95%CI 0.84-1.26).</p><p><strong>Conclusion: </strong>This extensive data pool suggests that deferring CDK4/6is to 2L may be associated with worse PFS2 but comparable OS when compared to early use in 1L, challenging the assumption that shifting therapies from 2L to 1L universally improves outcomes despite increased toxicity and costs.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"146"},"PeriodicalIF":5.6,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12351955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144849376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bo Huang, Yichao Zhang, Zhanjun Chen, Yuanyuan Yuan, Jianshan Lin
{"title":"Exosome-delivered METTL14 drives hypoxia-induced proliferation, metastasis, and glycolysis of breast cancer cells through regulating TRIM16-mediated FGF7 ubiquitination.","authors":"Bo Huang, Yichao Zhang, Zhanjun Chen, Yuanyuan Yuan, Jianshan Lin","doi":"10.1186/s13058-025-02099-2","DOIUrl":"10.1186/s13058-025-02099-2","url":null,"abstract":"<p><strong>Background: </strong>Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer and has a poor prognosis. Previous studies have indicated that Fibroblast Growth Factor 7 (FGF7) plays a vital role in the development and progression of breast cancer. However, the role and molecular mechanisms of FGF7 in TNBC remain largely unclear under hypoxia.</p><p><strong>Methods: </strong>FGF7 and Methyltransferase-like 14 (METTL14) levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). FGF7, tripartite motif-containing protein 16 (TRIM16), METTL14, METTL3, YTHDF1, WTAP, FTO, CD63, CD81, and TSG101 protein levels were examined by western blot. Cell viability, proliferation, invasion, and migration were determined using MTT, EdU, transwell, and wound healing assays. Glucose consumption, lactate production, and ATP levels were assessed using relevant kits. After STRING database analysis, the interaction between TRIM16 and FGF7 was verified using a Co-immunoprecipitation (CoIP) assay. Interaction between METTL14 and TRIM16 was validated using methylated RNA immunoprecipitation (MeRIP), RIP, and dual-luciferase reporter assays. The biological role of hypoxia-induced exosomal METTL14 on breast cancer tumor growth was assessed using the xenograft tumor model in vivo.</p><p><strong>Results: </strong>FGF7 was highly expressed in TNBC patients and cell lines. Moreover, FGF7 expression was increased in the hypoxia group compared with the normoxia group. Functionally, FGF7 knockdown suppressed hypoxia-induced TNBC cell proliferation, metastasis, and glycolysis. Mechanistically, TRIM16 triggered the ubiquitination of FGF7 and promoted its degradation. METTL14 enhanced TRIM16 mRNA stability and expression by m6A methylation. Hypoxia-induced exosomal METTL14 knockdown repressed tumor growth in vivo.</p><p><strong>Conclusion: </strong>Hypoxia-induced exosomal METTL14 supports the proliferation, metastasis, and glycolysis of TNBC cells through regulating TRIM16-mediated FGF7 ubiquitination, providing a promising therapeutic target for TNBC treatment.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"145"},"PeriodicalIF":5.6,"publicationDate":"2025-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12345010/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144838394","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
George W Jr Sledge, Joanne Xiu, Jeffrey Peter Solzak, Jennifer Ribeiro, Reshma L Mahtani, Maryam B Lustberg, Matthew J Oberley, Milan Radovich, David Spetzler
{"title":"Comparison of trastuzumab deruxtecan and sacituzumab govitecan in HER2-negative metastatic breast cancer: a large real-world data analysis.","authors":"George W Jr Sledge, Joanne Xiu, Jeffrey Peter Solzak, Jennifer Ribeiro, Reshma L Mahtani, Maryam B Lustberg, Matthew J Oberley, Milan Radovich, David Spetzler","doi":"10.1186/s13058-025-02094-7","DOIUrl":"10.1186/s13058-025-02094-7","url":null,"abstract":"","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"144"},"PeriodicalIF":5.6,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12341277/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144823008","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Non-enzymatic SETD1A activity drives breast cancer cell proliferation via cyclin K.","authors":"Kanako Hayashi, Takayuki Hoshii, Meng Ning, Makoto Matsumoto, Shintaro Izumi, Masaki Fukuyo, Bahityar Rahmutulla, Masahiko Tanabe, Atsushi Kaneda","doi":"10.1186/s13058-025-02101-x","DOIUrl":"10.1186/s13058-025-02101-x","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer is the most common cancer in women worldwide. SETD1A, a histone H3 lysine-4 methyltransferase, is associated with poor prognosis in breast cancer. While both its enzymatic and non-enzymatic functions are implicated in cancer progression, the specific role of SETD1A in breast cancer remains unclear. This study aimed to elucidate the molecular mechanisms underlying SETD1A dependency in breast cancer.</p><p><strong>Methods: </strong>SETD1A-high breast cancer cell lines were identified using TCGA and DepMap databases, along with SETD1A knockdown experiments. A CRISPR knockout of SETD1A was performed in a doxycycline-inducible Cas9-expressing KPL-1 breast cancer cell line. RNA-seq, ChIP-seq, CRISPR-tiling screening, and rescue experiments with exogenous SETD1A mutants were performed to investigate the roles of SETD1A in breast cancer.</p><p><strong>Results: </strong>SETD1A is highly expressed in estrogen receptor-positive / human epidermal growth factor receptor 2-negative (ER<sup>+</sup>HER2<sup>-</sup>) breast cancer and is associated with shorter overall survival in luminal-type breast cancer patients. We found that SETD1A is required for cell cycle progression from G1 to S phase in KPL-1 breast cancer cells, but its catalytic domain is dispensable. SETD1A disruption reduces the expression of DNA repair-associated genes, including RPA3 and PRIM1. Exogenous expression of both genes restores defective cell proliferation following SETD1A knockout. The non-catalytic function of SETD1A in breast cancer cells depends on its cyclin K-associated FLOS domain. SETD1A disruption also leads to defective transcriptional elongation in downregulated genes. Treatment with the cyclin K degrader CR8 recapitulates the phenotypes observed in SETD1A knockout cells. Both SETD1A knockout and CR8 were also effective in triple negative breast cancer (TNBC) cells.</p><p><strong>Conclusions: </strong>SETD1A promotes breast cancer cell replication through its non-enzymatic role via cyclin K, suggesting that the SETD1A-cyclin K axis could be a potential therapeutic target in breast cancer.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"142"},"PeriodicalIF":5.6,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12337470/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144823010","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Amadeo M Parissenti, Laura B Pritzker, Maria Aanesland Dahle, Hedda von der Lippe Gythfeldt, Twinkle Masilamani, Gabriel Theriault, Renée St-Onge, Lavina D'costa, Ole Christian Lingjaerde, Mads Haugland Haugen, Olav Engebraaten
{"title":"High mid-treatment tumour RNA disruption in patients with HER2-negative breast cancer is associated with improved disease-free survival after neoadjuvant chemotherapy.","authors":"Amadeo M Parissenti, Laura B Pritzker, Maria Aanesland Dahle, Hedda von der Lippe Gythfeldt, Twinkle Masilamani, Gabriel Theriault, Renée St-Onge, Lavina D'costa, Ole Christian Lingjaerde, Mads Haugland Haugen, Olav Engebraaten","doi":"10.1186/s13058-025-02092-9","DOIUrl":"10.1186/s13058-025-02092-9","url":null,"abstract":"","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":"27 1","pages":"143"},"PeriodicalIF":5.6,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12337404/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144823009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}