1
|
Liang Y, Mi Z, Kuo PC. Differential MYC and PROM1 mRNA isoform expression in breast invasive carcinoma as biomarkers for subtyping and prognosis. Surgery 2025; 179:108798. [PMID: 39306567 DOI: 10.1016/j.surg.2024.07.056] [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: 05/01/2024] [Revised: 06/26/2024] [Accepted: 07/14/2024] [Indexed: 02/02/2025]
Abstract
BACKGROUND Cancer stem cells are a subpopulation of tumor cells with the ability to self-renew; evidence suggests that cancer stem cells are responsible for recurrence, metastasis, and resistance to therapy. MYC and CD133 (PROM1 gene) are clinical biomarkers for cancer stem cells, and their dysregulation is involved in the progression of many cancers. Alternative splicing of these genes may contribute to cancer stem cell differentiation. METHODS Transcriptional and clinical data of PROM1 and MYC mRNA isoforms in breast cancer samples were downloaded from the TCGA Splicing Variants Database site, a web-tool to explore mRNA alternative-splicing based on TCGA samples. Data include RSEM isoform expression, clinical sample types, survival data, and clinical receptor expression. Breast cancer subtypes (luminal A, luminal B, Her2 positive, triple negative) were assigned on the basis of estrogen, progesterone, and HER2 expression. RESULTS Expression of MYC isoforms uc003ysh.1 and uc003ysi.3 was significantly greater in triple-negative breast cancer compared with all other breast cancer subtypes (P < .001). Isoform uc003ysi.3 was associated with greater 5-year survival in luminal A breast cancer (hazard ratio, 0.79; 95% confidence interval, 0.65-0.96; P = .02). PROM1 isoforms uc003gop.2, uc003goq.3, uc003gos.2, and uc003gou.2 were expressed greatest in triple-negative breast cancer (P < .001). PROM1 isoform uc003gou.2 was associated with better 5-year survival in luminal A breast cancer (hazard ratio, 0.79; 95% confidence interval, 0.65-0.97; P = .02). CONCLUSIONS MYC and PROM1 isoforms are differentially expressed in breast cancer subtypes. Certain isoforms confer better survival prognosis. Further work should be done to study alternative splicing in cancer stem cells.
Collapse
Affiliation(s)
- Yifan Liang
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Zhiyong Mi
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Paul C Kuo
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL; Bay Pines Veterans Affairs Health Care System, Bay Pines, FL.
| |
Collapse
|
2
|
Kim H, Baek S, Han S, Kim GM, Sohn J, Rhee Y, Hong N, Kim MH. Low Skeletal Muscle Radiodensity Predicts Response to CDK4/6 Inhibitors Plus Aromatase Inhibitors in Advanced Breast Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e13666. [PMID: 39686815 DOI: 10.1002/jcsm.13666] [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: 05/24/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Recent evidence indicates that a dysregulated host metabolism influences treatment outcomes in patients with breast cancer. We investigated the association of computed tomography (CT)-derived body composition indices with therapeutic responses in patients with hormone receptor-positive, HER2-negative advanced breast cancer (ABC) on endocrine plus CDK4/6 inhibitor (CDK4/6i) treatment. METHODS The study involved a retrospective cohort of patients with ABC at the Yonsei Cancer Center who received CDK4/6i and aromatase inhibitors as first-line therapy between January 2017 and October 2020. Body composition parameters were estimated from the non-enhanced CT images of the third lumbar spine by commercialized deep learning software. Patients with low skeletal muscle radiodensity (SMD) were defined as patients with SMD of low tertile (≤ 28.7 Hounsfield Units). The primary outcome was progression-free survival (PFS). RESULTS Among the 247 female participants (median age, 53 years; mean body mass index [BMI], 23.7 kg/m2), 45.7% had disease progression or death during a median follow-up of 36.4 months. After adjusting for age and visceral metastasis, SMD was the only independent predictor among body composition parameters for worse PFS (adjusted hazard ratio [HR] = 1.20 per standard deviation decrement, 95% CI: 1.01-1.42, p = 0.041), whereas BMI, muscle area, and fat area were not. Participants with low SMD had a higher risk of progression than those without (PFS, 27.2 vs. 51.1 months, p = 0.009; adjusted HR 1.84, 95% CI: 1.22-2.76, p = 0.003). Strong associations between low SMD and poor PFS were observed in groups with pre-menopause status (HR, 3.04 vs. 1.19 in post-menopause; 95% CI: 1.54-5.99, p for interaction < 0.05) and without visceral metastases (HR, 2.95 vs. 1.19 in with visceral metastases; 95% CI: 1.59-5.49, p for interaction < 0.05). CONCLUSIONS CT-defined low SMD predicts poor treatment outcomes in patients with ABC undergoing first-line treatment with aromatase inhibitors and CDK4/6i.
Collapse
Affiliation(s)
- Hyunwook Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Seungjin Baek
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sookyeong Han
- Endocrine Research Institute, Severance Hospital, Seoul, South Korea
| | - Gun Min Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Joohyuk Sohn
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Yumie Rhee
- Department of Internal Medicine, Endocrine Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Namki Hong
- Department of Internal Medicine, Endocrine Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Hwan Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
3
|
Tsai YF, Lai JI, Liu CY, Hsi CN, Hsu CY, Huang CC, Feng CJ, Lin YS, Chao TC, Chiu JH, Tseng LM. Correlation Between PIK3R1 Expression and Cell Growth in Human Breast Cancer Cell Line BT-474 and Clinical Outcomes. World J Oncol 2025; 16:131-141. [PMID: 39850525 PMCID: PMC11750754 DOI: 10.14740/wjon1986] [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: 10/21/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Background While mutations in the PIK3CA gene play important roles in human breast carcinogenesis, PIK3R1 gene alterations are recognized as actionable mutations for clinical cancer treatment. We aimed to elucidate the role of PIK3R1 in cell proliferation on breast carcinoma and to correlate the PIK3R1 expression with patients' outcome using human tumor tissue arrays. Methods Using human BT-474 (estrogen receptor (ER)+/human epidermal growth factor receptor 2 (HER2)-high) breast carcinoma cell line as in vitro model, the role of PIK3R1 in cell proliferation was elucidated by knock-down of the PIK3R1 gene (ΔPIK3R1) in this cell line. Between January 2000 to December 2015, the records of a cohort of 440 patients in our hospital were retrospectively reviewed, including patients' survival. The correlations between PIK3R1 expression and patient prognosis, such as overall survival (OS) and disease-free survival (DFS), were elucidated by human breast cancer tumor tissue array immunostaining. Results After the PIK3R1 gene was silenced in the BT-474 line, there was an increased cell number and a decrease in the G0G1-fraction, and increased S-fraction and the S+G2M-fraction for the ΔPIK3R1-BT-474 cell line, as compared to their cell wild type (WT) line. Western blot analysis showed that decreased PIK3R1 protein levels were accompanied by an increase of the p-AKT and p-mTOR proteins in the ΔPIK3R1-BT-474 cell line, compared to the equivalent WT line. Using a human tumor tissue array, patients with high-expressed PIK3R1 protein had better outcomes in terms of DFS and OS, compared to those with low-expressed PIK3R1 protein, when breast cancer was at an early stage (stage I/II), but not across all stages of breast cancer in human patients. Conclusions We concluded that downregulated PIK3R1 in BT-474 cells resulted in an increased cell growth and upregulated AKT-mTOR signaling. Clinically, the high-expressed PIK3R1 protein in tumors correlates positively with patients' outcome in stage I and II breast cancer.
Collapse
Affiliation(s)
- Yi-Fang Tsai
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Jiun-I Lai
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chun-Yu Liu
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chieh-Ning Hsi
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chih-Yi Hsu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chi-Cheng Huang
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 100233, Taiwan
| | - Chin-Jung Feng
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yen-Shu Lin
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Ta-Chung Chao
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Cancer Prevention, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Jen-Hwey Chiu
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of General Surgery, Department of Surgery, Cheng-Hsin General Hospital, Taipei 112401, Taiwan
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- These authors contributed equally to this work
| |
Collapse
|
4
|
Atallah N, Makhlouf S, Li X, Zhang Y, Mongan NP, Rakha E. Prediction of Response to Anti-HER2 Therapy Using A Multigene Assay. Mod Pathol 2025; 38:100713. [PMID: 39826800 DOI: 10.1016/j.modpat.2025.100713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/22/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
HER2-positive breast cancer (BC), which constitutes 13% to 15% of cases, shows variable response to anti-HER2 therapies. HER2 positivity, defined as protein overexpression (immunohistochemistry [IHC] score 3+) or equivocal expression (IHC 2+) with evidence of HER2 gene amplification, determines the eligibility for anti-HER2 therapy. The MammaTyper assay (Cerca Biotech GmbH) is an RT-qPCR BC subtyping platform based on the micro RNA expression of ERBB2, ESR1, PGR, and MKI67. This study aimed to evaluate the accuracy of the MammaTyper assay in predicting the response of HER2-positive patients to therapy. A well-characterized HER2-positive BC cohort of 287 cases diagnosed at Nottingham University hospitals between 2006 and 2018 was included. The cohort was divided into 2 groups: a trastuzumab-treated group (n = 159) and a chemotherapy-only treated group (n = 128). Tumor clinicopathologic characteristics were matched between the 2 groups. Cases with discordant HER2 status were validated through staining of surgical excision specimens. ERBB2 micro RNA identified 251/287 (87.5%) cases as HER2-positive, 10.8% (31/287) as HER2 low and 1.7% (5/287) as HER2 negative. According to the MammaTyper assay, ERBB2-positive patients treated with anti-HER2 therapy had significantly prolonged 5-year disease-free survival and distant metastasis-free survival (hazard ratio = 0.56, P = .003 and hazard ratio = 0.62, P = .023, respectively). MammaTyper-defined HER2-enriched subtype showed a better response to anti-HER2 therapy compared with IHC-defined subtypes, with significant differences in both 5-year disease-free survival and BC-specific survival (P = .01 and < .001, respectively). Patients who were ERBB2 negative did not show a survival difference between the group of patients who were treated with trastuzumab and those who were treated with chemotherapy only (P > .05). Validation analysis revealed that 11/36 ERBB2-negative cases were IHC 2+/ISH positive with very low level of gene amplification and 25 cases were false classified as HER2-positive using current protocols. Combining the MammaTyper assay with IHC to assess HER2 status improves the identification of HER2-positive patients with BC who would benefit from anti-HER2 therapy.
Collapse
Affiliation(s)
- Nehal Atallah
- Translational Medical Science, School of Medicine, the University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; Department of Pathology, Faculty of Medicine, Menoufia University, Egypt
| | - Shorouk Makhlouf
- Translational Medical Science, School of Medicine, the University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; Department of Pathology, Faculty of Medicine, Assiut University, Egypt
| | | | | | - Nigel P Mongan
- Translational Medical Science, School of Medicine, the University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; Translational Medical Science, School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom; Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Emad Rakha
- Translational Medical Science, School of Medicine, the University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; Department of Pathology, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
5
|
Koon Sun Pat M, Manraj M, Manraj S. Breast cancer survival analysis in the Republic of Mauritius by age, stage at diagnosis and molecular subtype: A retrospective cohort study. Int J Cancer 2025; 156:331-338. [PMID: 39243396 DOI: 10.1002/ijc.35172] [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: 11/29/2023] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer is by far the leading cancer both in terms of incidence and mortality in the Republic of Mauritius, a Small Island Developing State (SIDS). However, few studies assessed its survival by age, stage at diagnosis and molecular subtype. We identified 1399 breast cancer cases newly diagnosed between 2017 and 2020 at the Central Health Laboratory, Victoria Hospital. Cancers were categorized into five molecular subtypes: (1) luminal A, (2) luminal B Her2 negative, (3) luminal B Her2 positive, (4) Her2 enriched and (5) Triple negative. The net 1 and 3-year survival were estimated for different age groups, staging at time of diagnosis and molecular subtype. We also estimated the excess hazards using a multivariate Cox proportional hazards model. While early stage at diagnosis (stage 1 [44.4%] and stage 2 [20.1%]) were most common compared to late presentation (Stage 3 [25.4%] and stage 4 [10.1%]), luminal B Her2 negative (36.7%) was the most frequent molecular subtype. The net 1- and 3-year breast cancer survival rates were 93.9% (92.3-95.4) and 83.4% (80.4-86.4), respectively. Breast cancer three-year survival rates were poorest among the youngest patients (<50 years), 77.1% (70.7-83.5), those diagnosed with stage 4 (28.5% [17.1-39.9]) and cancer with a triple negative molecular subtype (71.3% [63.3-79.3]). Emphasis on a national breast cancer screening programme, down staging breast cancer at diagnosis and systematic molecular subtyping of all breast tissues could be pivotal in improving breast cancer survival outcomes in the Republic of Mauritius.
Collapse
Affiliation(s)
- Marvin Koon Sun Pat
- Community Physician, Ministry of Health and Wellness, Port Louis, Mauritius
- Faculty of Medicine and Health Science, University of Mauritius, Reduit, Mauritius
| | - Meera Manraj
- Faculty of Medicine and Health Science, University of Mauritius, Reduit, Mauritius
| | - Shyam Manraj
- Central Health Laboratory, Victoria Hospital, Candos, Mauritius
| |
Collapse
|
6
|
Majumder S, Mishra S, Shinde N, Cuitino MC, Bauer M, Ahirwar D, Basree MM, Bharti V, Ormiston K, Mawalkar R, Alsammerai S, Sarathy G, Vilgelm AE, Zhang X, Ganju RK, Ramaswamy B. Divergent paths of mammary gland involution: unveiling the cellular dynamics in abruptly and gradually involuted mouse models. Breast Cancer Res 2025; 27:1. [PMID: 39754221 PMCID: PMC11697808 DOI: 10.1186/s13058-024-01933-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Epidemiological studies associate an increase in breast cancer risk, particularly triple-negative breast cancer (TNBC), with lack of breastfeeding. This is more prevalent in African American women, with significantly lower rate of breastfeeding compared to Caucasian women. Prolonged breastfeeding leads to gradual involution (GI), whereas short-term or lack of breastfeeding leads to abrupt involution (AI) of the breast. Our previous study utilizing a murine model demonstrated precancerous changes, specifically hyperplasia, a non-obligate precursor of breast cancer in the mammary glands of AI mice. Here we investigated mechanisms during early events of AI that prompts precancerous changes in mouse mammary glands. METHODS Uniparous FVB/N mice were randomized to AI and GI on postpartum day 7 when all pups were removed from AI dams. GI dams were allowed to nurse the pups till day 31. Cell death kinetics and gene expression were assessed by TUNEL assay and qPCR respectively. Immune cell changes were investigated by flow cytometry, cytokine array and multiplex immunofluorescence. 3D-organoid cultures were used for in vitro assay of luminal progenitor cells. RESULTS AI results in rapid cell death, DNA repair response, and immunosuppressive myeloid cells infiltration, leading to a chronically inflamed microenvironment. GI elicits a more controlled immune response and extended cell death. At the peak of cell death, AI glands harbored more immunosuppressive myeloid-derived suppressor cells (MDSCs) and CD206 + M2-like macrophages, known to promote oncogenic events, compared to GI glands. AI glands exhibit an enrichment of CCL9-producing MDSCs and CD206 + M2-like macrophages that promote expansion of ELF5 + /ERα- luminal cells, both in vitro and in vivo. Multiplex imaging of AI glands demonstrated an increase in ELF5 + /WNT5a + luminal cells alongside a reduction in the ELF5 + /ERα + population when involution appeared histologically complete. A significantly higher number of CD206 + cells in post involution AI gland attests to a chronically inflamed state induced by AI. CONCLUSIONS Our findings reveal significant disparities between AI and GI gland dynamics at the early phase of involution. CCL9, secreted by immune cells at the peak of cell death promotes expansion of Elf5 + /ERα- luminal progenitor cells, the putative precursors of TNBC connecting early events of AI with increased breast cancer risk.
Collapse
Affiliation(s)
- Sarmila Majumder
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| | - Sanjay Mishra
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Neelam Shinde
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maria C Cuitino
- Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA
| | - Morgan Bauer
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Dinesh Ahirwar
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Mustafa M Basree
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Vijaya Bharti
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Kate Ormiston
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Resham Mawalkar
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Sara Alsammerai
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gautam Sarathy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics and Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
7
|
Vaz-Goncalves L, Protani MM, Saunus JM, Colditz GA, Reeves MM. Continental differences in the association between excess body weight and prognosis in triple-negative breast cancer: a meta-analysis. Breast Cancer Res Treat 2025; 209:215-227. [PMID: 39476311 DOI: 10.1007/s10549-024-07538-w] [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: 01/19/2024] [Accepted: 10/22/2024] [Indexed: 02/02/2025]
Abstract
PURPOSE The association between obesity and triple-negative breast cancer (TNBC) prognosis has been equivocal, with considerable heterogeneity between and within studies. Recent meta-analyses report adverse associations with overall survival (OS) and disease-free survival (DFS) in TNBC. We update this evidence and examine study- and disease-specific sources of heterogeneity. METHODS A systematic search of four databases was conducted until February 22, 2023. Random-effects meta-analyses were used to pool hazard ratios (HR) for OS, DFS, and breast cancer-specific mortality (BCSM). Subgroup analyses examined sources of study heterogeneity. RESULTS In meta-analyses of included studies (n = 33), significant associations were observed between excess body weight and worse OS (n = 24; HR = 1.20; 95%CI 1.20-1.34), DFS (n = 26; HR = 1.15; 1.05-1.27), and BCSM (n = 9; HR = 1.13; 1.00-1.27). In subgroup meta-analyses, significant inter-study survival differences were observed for study location (OS, DFS), time period of diagnoses (DFS), menopausal status (OS), and body mass index cut points examined (OS). Asian and European studies reported significant associations with OS (HR = 1.31; 1.11-1.54 and HR = 1.38; 1.00-1.89, respectively) and DFS (HR = 1.28; 1.07-1.54 and HR = 1.44; 1.13-1.84, respectively); however, no association was observed between obesity and TNBC prognosis in North American studies (OS: HR = 1.03; 0.89-1.19; DFS: HR = 1.05; 0.95-1.15). Location subgroup differences remained robust after excluding poor-quality studies. Post hoc analysis in the subset of studies reporting predominantly (≥ 70%) White sample showed no statistically significant associations for OS (HR = 1.13; 95%CI 0.96, 1.34), DFS (HR = 1.03; 95%CI 0.86, 1.23), or BCSM (HR = 1.08; 95%CI 0.91, 1.27). CONCLUSION This study further confirms that obesity is associated with poor prognosis in TNBC and identified subgroups at higher risk. Ethnic differences in the association between excess body weight and TNBC are reported. Further exploration of study and patient characteristics is needed to properly understand the populations most at risk.
Collapse
Affiliation(s)
| | - Melinda M Protani
- School of Public Health, The University of Queensland, Brisbane, QLD, Australia
| | - Jodi M Saunus
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Graham A Colditz
- Division of Public Health Sciences, Department of Surgery, School of Medicine, Washington University, St Louis, MO, USA
| | - Marina M Reeves
- School of Public Health, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
8
|
Kumar RMR, Joghee S. A Review on Integrating Breast Cancer Clinical Data: A Unified Platform Perspective. Curr Treat Options Oncol 2025; 26:1-13. [PMID: 39752094 DOI: 10.1007/s11864-024-01285-2] [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] [Accepted: 12/04/2024] [Indexed: 01/04/2025]
Abstract
OPINION STATEMENT Integrating clinical datasets in breast cancer research emerges as a necessary tool for advancing our knowledge of the disease and enhancing patient outcomes. Synthesizing diverse datasets offers advantages, from facilitating evidence-based insights to enabling predictive analytics and precision medicine strategies. Crucially, effective integration of clinical datasets necessitates collaborative efforts, policy interventions, and technological advancements to elevate global standards of breast cancer care. This narrative review underscores the imperative and substantial benefits of dataset integration in advancing breast cancer research and optimizing patient management. First, integrating diverse datasets-encompassing patient demographics, tumor characteristics, treatment modalities, and clinical outcomes-can significantly enhance our understanding of the disease's complexities and treatment responses across diverse patient populations. Second, we suggest that regulatory approval processes should allow new treatments to be conditionally approved for patients who were not part of the initial trials. This approval would depend on evaluating how well these treatments perform in real-world situations before full approval is granted. Third, we emphasize the importance of incorporating high-quality real-world evidence into treatment guidelines to better inform patient counselling and optimize personalized treatment strategies.
Collapse
Affiliation(s)
- Ram Mohan Ram Kumar
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India.
| | - Suresh Joghee
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| |
Collapse
|
9
|
Han H, Yang M, Wen Z, Mei F, Chen Q, Ma Y, Lai X, Zhang Y, Fang R, Yin T, Sun S, Wang X, Qi J, Lin H, Yang Y. Trametinib and M17, a novel small molecule inhibitor of AKT, display a synergistic antitumor effect in triple negative breast cancer cells through the AKT/mTOR and MEK/ERK pathways. Bioorg Chem 2025; 154:107981. [PMID: 39591692 DOI: 10.1016/j.bioorg.2024.107981] [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: 09/12/2024] [Revised: 11/02/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024]
Abstract
Triple negative breast cancer (TNBC) is associated with a poor prognosis and limited response to traditional chemotherapy, necessitating the exploration of novel treatment approaches. Recent researches have highlighted the interconnected roles of the PI3K/AKT pathway and MAPK pathway in TNBC cells, contributing to the efficacy of treatments. Therefore, the concurrent inhibition of both pathways presents a potential new therapeutic strategy for TNBC patients. This study aimed to evaluate the antitumor efficacy of M17, an AKT allosteric inhibitor and a new synthesized shikonin derivative, both alone and in combination with the MEK inhibitor trametinib. We applied various cellular assays and a subcutaneous 4T1 tumor bearing BALB/c mice model were utilized to assess the in vitro and in vivo antitumor effects. Computational docking and Bio-Layer Interferometry (BLI) were employed to investigate the binding of M17 with AKT. Additionally, flow cytometry, transwell assays, western blotting, and tumor xenograft assays were conducted to explore the potential synergistic mechanisms of the combined therapy. The results demonstrated that M17 exhibited moderate antitumor activity against TNBC cells, but significantly enhanced the apoptotic effects and inhibited proliferation and migration when combined with trametinib. Furthermore, the combination of M17 and trametinib showed even more pronounced antitumor activity in vivo. Mechanistically, the dual therapy synergistically suppressed TNBC by targeting the AKT/mTOR and MEK/ERK signaling pathways and inhibiting epithelial-mesenchymal transition. In conclusion, the findings suggested that the combination of M17 and trametinib holds promise as a synergistic treatment option for TNBC patients.
Collapse
Affiliation(s)
- Hongwei Han
- School of Life Sciences and Chemical Engineering, Jiangsu Second Normal University, Nanjing 210013, China; State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Feng Mei
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Yudi Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaohui Lai
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Yahan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Rongjun Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Shucun Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xiaoming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Yonghua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
10
|
Liu L, Graff SL, Wang Y. New Emerging Therapies Targeting PI3K/AKT/mTOR/PTEN Pathway in Hormonal Receptor-Positive and HER2-Negative Breast Cancer-Current State and Molecular Pathology Perspective. Cancers (Basel) 2024; 17:16. [PMID: 39796647 PMCID: PMC11718791 DOI: 10.3390/cancers17010016] [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] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
In hormone receptor-positive and HER2-negative breast cancers, a growing number of revolutionary personalized therapies are in clinical use or trials, such as CDK4/6 inhibitors, immune checkpoint inhibitors, and PIK3CA inhibitors. Those treatment options are largely driven by the presence or absence of genomic alterations in the tumor. Therefore, molecular profiling is often performed during disease progression. The most encountered genomic alterations are in the PI3K/AKT/mTOR/PTEN pathway. This review discusses the genetic alterations associated with the PI3K/AKT/mTOR/PTEN pathway to help clinicians understand drug selection, resistance, or interaction from a molecular pathologist's perspective.
Collapse
Affiliation(s)
- Liu Liu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA;
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Stephanie L. Graff
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
- Division of Medical Oncology, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA;
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| |
Collapse
|
11
|
Bhattarai S, Sugita BM, Nunes-Souza E, Fonseca AS, Chandrashekar DS, Bhargava M, Cavalli LR, Aneja R. Dysregulated miRNA Expression and Androgen Receptor Loss in Racially Distinct Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:13679. [PMID: 39769441 PMCID: PMC11679545 DOI: 10.3390/ijms252413679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/23/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Androgen receptor (AR)-negative triple-negative breast cancer (TNBC), often termed quadruple-negative breast cancer (QNBC), disproportionately impacts women of African descent, leading to poorer overall survival (OS). MiRNAs regulate the expression of gene drivers involved in critical signaling pathways in TNBC, such as the AR gene, and their expression varies across races and breast cancer subtypes. This study investigates whether differentially expressed miRNAs influence AR transcription, potentially contributing to the observed disparities between African American (AA) and European American (EA) QNBC patients. Race-annotated TNBC samples (n = 129) were analyzed for AR expression status and revealed the prevalence of QNBC in AA patients compared to EA (76.6% vs. 57.7%) and a significant association of AR loss with poor survival among AAs. The Cancer Genome Atlas (TCGA) RNA-seq data showed that AAs with TNBC (n = 32) had lower AR mRNA levels than EAs (n = 67). Among TCGA patients in the AR-low group, AAs had significantly poorer OS than EAs. In our cohort, 46 miRNAs exhibited differential expression between AAs and EAs with QNBC. Ten of these miRNAs (miR-1185-5p, miR-1305, miR-3161, miR-3690, miR-494-3p, miR-509-3-5p, miR-619-3p, miR-628-3p, miR-873-5p, and miR-877-5p) were predicted to target the AR gene/signaling. The loss of AR expression is linked to poorer prognoses in AA women. The understanding of the specific miRNAs involved and their regulatory mechanisms on AR expression could provide valuable insights into why AA women are more prone to QNBC.
Collapse
Affiliation(s)
- Shristi Bhattarai
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Bruna M. Sugita
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (B.M.S.)
| | - Emanuelle Nunes-Souza
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (B.M.S.)
| | - Aline S. Fonseca
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (B.M.S.)
| | - Darshan Shimoga Chandrashekar
- Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Mahak Bhargava
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Luciane R. Cavalli
- Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (B.M.S.)
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
12
|
Giannoudis A, Sokol ES, Bhogal T, Ramkissoon SH, Razis ED, Bartsch R, Shaw JA, McGregor K, Clark A, Huang RSP, Palmieri C. Breast cancer brain metastases genomic profiling identifies alterations targetable by immune-checkpoint and PARP inhibitors. NPJ Precis Oncol 2024; 8:282. [PMID: 39706915 DOI: 10.1038/s41698-024-00761-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024] Open
Abstract
Understanding the genomic landscape of breast cancer brain metastases (BCBMs) is key to developing targeted treatments. In this study, targetable genomic profiling was performed on 822 BCBMs, 11,988 local breast cancer (BC) biopsies and 15,516 non-central nervous system (N-CNS) metastases (all unpaired samples) collected during the course of routine clinical care by Foundation Medicine Inc (Boston, MA). Clinically relevant genomic alterations were significantly enriched in BCBMs compared to local BCs and N-CNS metastases. Homologous recombination deficiency as measured by BRCA1/2 alteration prevalence and loss-of-heterozygosity and immune checkpoint inhibitor (ICI) biomarkers [Tumor mutation burden (TMB)-High, Microsatellite instability (MSI)-High, PD-L1/L2)] were significantly more prevalent in BCBM than local BC and N-CNS. High PD-L1 protein expression was observed in ER-negative/HER2-negative BCBMs (48.3% vs 50.0% in local BCs, 21.4% in N-CNS). Our data highlights that a high proportion of BCBMs are potentially amenable to treatment with targeted therapeutic agents including PARP inhibitors and ICIs.
Collapse
Affiliation(s)
- A Giannoudis
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - E S Sokol
- Foundation Medicine, Inc., Boston, MA, USA
| | - T Bhogal
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | | | - E D Razis
- Hygeia Hospital, 3rd Oncology Department, Marousi, Athens, Greece
| | - R Bartsch
- Medical University of Vienna, Department of Medicine I, Division of Oncology, Vienna, Austria
| | - J A Shaw
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - K McGregor
- Foundation Medicine, Inc., Boston, MA, USA
| | | | | | - C Palmieri
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
13
|
Arrey EN, GoPaul D, Anderson D, Okoli J, McKenzie-Johnson T. Addressing Breast Cancer Disparities: A Comprehensive Approach to Health Equity. J Surg Oncol 2024. [PMID: 39699972 DOI: 10.1002/jso.28011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/15/2024] [Accepted: 10/01/2024] [Indexed: 12/21/2024]
Abstract
This article addresses the persistent disparities in breast cancer outcomes across different racial, ethnic, and socioeconomic groups despite advancements in diagnosis and treatment. The disparities are rooted in various factors, including access to care, socioeconomic status, and cultural barriers. The article emphasizes the need for targeted interventions, such as expanding insurance coverage, mobile mammography units, and culturally tailored outreach programs to promote health equity. Achieving this requires comprehensive strategies addressing systemic and social determinants of health.
Collapse
Affiliation(s)
- Eliel N Arrey
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Darren GoPaul
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - David Anderson
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Joel Okoli
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Tamra McKenzie-Johnson
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
- General Surgery Section, Atlanta Veterans Affairs Health Care System, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Shams A. Impact of prolactin treatment on enhancing the cellular responses of MCF7 breast cancer cells to tamoxifen treatment. Discov Oncol 2024; 15:797. [PMID: 39692941 DOI: 10.1007/s12672-024-01701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024] Open
Abstract
Breast cancer remains one of the most challenging diseases to treat due to its heterogeneity, propensity to recur, capacity to spread to distant vital organs, and, ultimately, patient death. Estrogen receptor-positive illness comprises the most common breast cancer subtype. Preclinical progress is hampered by the scarcity of medication-naïve estrogen receptor-positive tumour models that recapitulate metastatic development and treatment resistance. It is becoming increasingly clear that loss of differentiation and increased cellular stemness and plasticity are important causes of cancer evolution, heterogeneity, recurrence, metastasis, and treatment failure. Therefore, it has been suggested that reprogramming cancer cell differentiation could offer an effective method of reversing cancer through terminal differentiation and maturation. In this context, the hormone prolactin is well recognized for its pivotal involvement in the development of the mammary glands lobuloalveolar tissue and the terminal differentiation that drives the production of the milk protein gene and lactation. Additionally, numerous studies have examined the engagement of prolactin in breast cancer as a differentiation player that resulted in the ablation of tumour growth and progression. Here, we showed that a pre-treatment of the estrogen-positive breast cancer cell line with prolactin led to a considerable improvement in the sensitivity of this cancer cell to Tamoxifen endocrine therapy. We also showed a favourable prognostic value of prolactin receptors/estrogen receptors 1 (or alpha) co-expression on breast cancer patients outcomes, and this co-expression is highly correlated with the well-differentiated breast tumour type. Our results revealed a fruitful aspect of the effects of prolactin in improving the responses of breast cancer cells to conventional endocrine therapy. Moreover, these findings further validated the ability of prolactin as a persuader of a more differentiated and less aggressive breast cancer phenotype. Hence, it suggested a potential implication of prolactin as a therapeutic candidate.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Taif, Saudi Arabia.
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research,, Taif University, Taif 26432, Taif, Saudi Arabia.
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Taif, Saudi Arabia.
| |
Collapse
|
15
|
Serrano García L, Jávega B, Llombart Cussac A, Gión M, Pérez-García JM, Cortés J, Fernández-Murga ML. Patterns of immune evasion in triple-negative breast cancer and new potential therapeutic targets: a review. Front Immunol 2024; 15:1513421. [PMID: 39735530 PMCID: PMC11671371 DOI: 10.3389/fimmu.2024.1513421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of progesterone and estrogen receptors and low (or absent) HER2 expression. TNBC accounts for 15-20% of all breast cancers. It is associated with younger age, a higher mutational burden, and an increased risk of recurrence and mortality. Standard treatment for TNBC primarily relies on cytotoxic agents, such as taxanes, anthracyclines, and platinum compounds for both early and advanced stages of the disease. Several targeted therapies, including bevacizumab and sunitinib, have failed to demonstrate significant clinical benefit in TNBC. The emergence of immune checkpoint inhibitors (ICI) has revolutionized cancer treatment. By stimulating the immune system, ICIs induce a durable anti-tumor response across various solid tumors. TNBC is a particularly promising target for treatment with ICIs due to the higher levels of tumor-infiltrating lymphocytes (TIL), increased PD-L1 expression, and higher mutational burden, which generates tumor-specific neoantigens that activate immune cells. ICIs administered as monotherapy in advanced TNBC yields only a modest response; however, response rates significantly improve when ICIs are combined with cytotoxic agents, particularly in tumors expressing PD-L1. Pembrolizumab is approved for use in both early and advanced TNBC in combination with standard chemotherapy. However, more research is needed to identify more potent biomarkers, and to better elucidate the synergism of ICIs with other targeted agents. In this review, we explore the challenges of immunotherapy in TNBC, examining the mechanisms of tumor progression mediated by immune cells within the tumor microenvironment, and the signaling pathways involved in both primary and acquired resistance. Finally, we provide a comprehensive overview of ongoing clinical trials underway to investigate novel immune-targeted therapies for TNBC.
Collapse
Affiliation(s)
- Lucía Serrano García
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| | - Beatriz Jávega
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| | - Antonio Llombart Cussac
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
- Grupo Oncología Traslacional, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-Centro de Estudios Universitarios (CEU), Alfara del Patriarca, Spain
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
| | - María Gión
- Medical Oncology Department, Hospital Ramon y Cajal, Madrid, Spain
| | - José Manuel Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
| | - Javier Cortés
- Medica Scientia Innovation Research (MEDSIR), Oncoclínicas & Co., Jersey City, NJ, United States
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - María Leonor Fernández-Murga
- Medical Oncology Department, Hospital Arnau de Vilanova, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Valencia, Spain
| |
Collapse
|
16
|
Lu J, Yang L, Yang X, Chen B, Liu Z. Investigating the clinical significance of OAS family genes in breast cancer: an in vitro and in silico study. Hereditas 2024; 161:50. [PMID: 39633486 PMCID: PMC11619215 DOI: 10.1186/s41065-024-00353-9] [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: 09/17/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy among women worldwide, characterized by complex molecular and cellular heterogeneity. Despite advances in diagnosis and treatment, there is an urgent need to identify reliable biomarkers and therapeutic targets to improve early detection and personalized therapy. The OAS (2'-5'-oligoadenylate synthetase) family genes, known for their roles in antiviral immunity, have emerged as potential regulators in cancer biology. This study aimed to explore the diagnostic and functional relevance of OAS family genes in breast cancer. METHODOLOGY Breast cancer cell lines and controls were cultured under specific conditions, and DNA and RNA were extracted for downstream analyses. RT-qPCR, bisulfite sequencing, and Western blotting were employed to assess gene expression, promoter methylation, and knockdown efficiency of OAS family genes. Functional assays, including CCK-8, colony formation, and wound healing, evaluated cellular behaviors, while bioinformatics tools (UALCAN, GEPIA, HPA, OncoDB, cBioPortal, and others) validated findings and explored correlations with clinical data. RESULTS The OAS family genes (OAS1, OAS2, OAS3, and OASL) were found to be significantly upregulated in breast cancer cell lines and tissues compared to normal controls. This overexpression was strongly associated with reduced promoter methylation. Receiver operating characteristic (ROC) analysis demonstrated high diagnostic accuracy, with area under the curve (AUC) values exceeding 0.93 for all four genes. Increased OAS expression correlated with advanced cancer stages and poor overall survival in breast cancer patients. Functional analysis revealed their involvement in critical biological processes, including immune modulation and oncogenic pathways. Silencing OAS genes in breast cancer cells significantly inhibited cell proliferation and colony formation, while unexpectedly enhancing migratory capacity. Additionally, correlations with immune cell infiltration, molecular subtypes, and drug sensitivity highlighted their potential roles in the tumor microenvironment and therapeutic response. CONCLUSION The findings of this study established OAS family genes as potential biomarkers and key players in breast cancer progression, offering promise as diagnostic biomarkers and therapeutic targets to address unmet clinical needs.
Collapse
Affiliation(s)
- Jinjun Lu
- Department of General Surgery, Nantong Haimen People's Hospital, NanTong, JiangSu, 226100, China
| | - Lu Yang
- Department of Clinical Laboratory, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Xinghai Yang
- Department of General Surgery, Nantong Haimen People's Hospital, NanTong, JiangSu, 226100, China
| | - Bin Chen
- Department of General Surgery, Nantong Haimen People's Hospital, NanTong, JiangSu, 226100, China
| | - Zheqi Liu
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310000, China.
| |
Collapse
|
17
|
Reid S, Fan R, Venton L, Weidner A, Tezak A, Roberson ML, Vadaparampil S, Wang X, Yoder S, Rosa M, Hirbo J, Whisenant JG, Pietenpol J, Rajagopal PS, Lehmann B, Ye F, Pal T. West African Genetic Ancestry and Breast Cancer Outcomes Among Black Women. JAMA Netw Open 2024; 7:e2449798. [PMID: 39652347 PMCID: PMC11629124 DOI: 10.1001/jamanetworkopen.2024.49798] [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: 05/23/2024] [Accepted: 10/16/2024] [Indexed: 12/12/2024] Open
Abstract
Importance Young Black women bear a disproportionate burden of breast cancer deaths compared with White women, yet they remain underrepresented in genomic studies. Objective To evaluate the association of biological factors, including West African genetic ancestry, and nonbiological factors with disease-free survival (DFS) among young Black women with breast cancer. Design, Setting, and Participants This observational cohort study included Black women diagnosed with invasive breast cancer between January 1, 2005, and December 31, 2016. Participants diagnosed with breast cancer at age 50 years or younger were recruited through the Florida and Tennessee state cancer registries. The final analysis was completed between June and September 2024. Exposure West African genetic ancestry. Main Outcomes and Measures A multivariable model was developed to evaluate the association between West African genetic ancestry and breast cancer DFS, adjusting for immunohistochemistry subtype, lymph node (LN) status, and full-time employment. Results This study included 687 Black women with early-stage invasive breast cancer. Their median age at diagnosis was 44 years (IQR, 38-47 years), and the median follow-up was 10 years (IQR, 7-11 years). In multivariable analysis, triple-negative breast cancer (TNBC) and LN involvement were associated with shorter breast cancer DFS (hazard ratio, 1.81 [95% CI, 1.20-2.73] and 1.77 [95% CI, 1.30-2.41], respectively), whereas full-time employment was associated with improved outcomes (hazard ratio, 0.44 [95% CI, 0.30-0.63]). Among the 551 participants for whom global genetic ancestry could be assessed, having a higher percentage of West African genetic ancestry was associated with shorter breast cancer DFS among 246 participants in the hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (ERBB2 [formerly HER2])-negative subgroup (hazard ratio, 1.45 [95% CI, 1.04-2.04]). Of the 369 participants (53.7%) with PAM50 data available, basal (133 [36.0%]) and luminal B (107 [29.0%]) subtypes were the most common. Among the 179 patients with HR-positive/ERBB2-negative disease and PAM50 data available, luminal B and basal subtypes combined were also overrepresented (81 [45.3%] and 24 [13.4%], respectively) compared with luminal A (70 [39.1%]). Conclusions and Relevance In this study of young Black women with breast cancer, having a higher percentage of West African genetic ancestry, TNBC, and LN involvement were associated with shorter breast cancer DFS. Interestingly, full-time employment was associated with improved breast cancer DFS. These findings highlight the importance of considering genetic ancestry beyond self-reported race and accounting for social determinants of health, in efforts to improve survival outcomes among Black women with breast cancer.
Collapse
Affiliation(s)
- Sonya Reid
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Run Fan
- Department of Biostatistics and Bioinformatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lindsay Venton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anne Weidner
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ann Tezak
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mya L. Roberson
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill
| | | | | | | | | | - Jibril Hirbo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Jennifer Pietenpol
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Brian Lehmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fei Ye
- Department of Biostatistics and Bioinformatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tuya Pal
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
18
|
Anampa JD, Alvarez Soto A, Bernal AM, Acuna-Villaorduna A. Racial disparities in treatment and outcomes between Hispanic and non-Hispanic black women with triple-negative breast cancer. Breast Cancer Res Treat 2024:10.1007/s10549-024-07565-7. [PMID: 39589609 DOI: 10.1007/s10549-024-07565-7] [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: 09/06/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is an aggressive breast cancer (BC) subtype with higher incidence and mortality rates in non-Hispanic Black (NHB) women than non-Hispanic Whites. Studies assessing disparities between NHB and Hispanic women, the two largest US racial/ethnic minorities, are lacking. This study evaluates disparities in the treatment and outcomes between NHB and Hispanic women with non-metastatic TNBC. METHODS This observational, population-based study using the SEER database included adult, female patients diagnosed with non-metastatic TNBC between 2010 and 2015 and identified as NHB or Hispanic. Logistic regression analysis was used to examine the adjusted odds of receiving breast cancer-directed treatment. Kaplan-Meier and cumulative incidence of death curves were plotted to assess overall survival (OS) and risk of breast cancer-related death, respectively. Multivariate regression analyses with Cox and Fine-Gray methods were calculated to assess factors associated with OS and breast cancer-related death, respectively. RESULTS There were 3426 Hispanic and 5419 NHB patients with non-metastatic TNBC. Hispanics had better 5-year OS relative to NHB (76% vs. 72%). No differences in the odds of receiving chemotherapy or surgery between cohorts was seen. However, the odds of undergoing breast-conserving surgery (BCS) and receiving radiation was higher in NHB than Hispanics, (OR, 1.22; 95% CI, 1.10-1.36) and (OR, 1.50; 95% CI, 1.36-1.66), respectively. Lack of radiation therapy was associated with increased BC-related death in NHB relative to Hispanics (sHR, 1.40; 95% CI, 1.19-1.65). Nevertheless, this difference was not seen when radiation was given, (sHR, 1.03; 95% CI, 0.87-1.23). CONCLUSIONS We found racial disparities in treatment and outcomes between NHB and Hispanics. NHB were more likely to receive radiation therapy and have BCS. Still, after adjusting for demographic and treatment-related factors, NHB had worse OS and BCSS relative to Hispanics. Additional research is needed to understand the drivers of these disparities.
Collapse
Affiliation(s)
- Jesus D Anampa
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, 1695 Eastchester Rd, 2nd Floor, Bronx, NY, 10461, USA.
| | - Alvaro Alvarez Soto
- Department of Medicine, Hematology/Oncology, Carole and Ray Neag Comprehensive Cancer Center, UCONN Health, Farmington, CT, USA
| | - Ana M Bernal
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, 1695 Eastchester Rd, 2nd Floor, Bronx, NY, 10461, USA
| | - Ana Acuna-Villaorduna
- Department of Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
19
|
Chernosky NM, Tamagno I, Polak KL, Chan ER, Yuan X, Jackson MW. Toll-Like receptor 3-mediated interferon-β production is suppressed by oncostatin m and a broader epithelial-mesenchymal transition program. Breast Cancer Res 2024; 26:167. [PMID: 39593161 PMCID: PMC11590466 DOI: 10.1186/s13058-024-01918-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Patients with Triple Negative Breast Cancer (TNBC) currently lack targeted therapies, and consequently face higher mortality rates when compared to patients with other breast cancer subtypes. The tumor microenvironment (TME) cytokine Oncostatin M (OSM) reprograms TNBC cells to a more stem-like/mesenchymal state, conferring aggressive cancer cell properties such as enhanced migration and invasion, increased tumor-initiating capacity, and intrinsic resistance to the current standards of care. In contrast to OSM, Interferon-β (IFN-β) promotes a more differentiated, epithelial cell phenotype in addition to its role as an activator of anti-tumor immunity. Importantly, OSM suppresses the production of IFN-β, although the mechanism of IFN-β suppression has not yet been elucidated. METHODS IFN-β production and downstream autocrine signaling were assessed via quantitative real-time PCR (qRT-PCR) and Western blotting in TNBC cells following exposure to OSM. RNA-sequencing (RNA-seq) was used to assess an IFN-β metagene signature, and to assess the expression of innate immune sensors, which are upstream activators of IFN-β. Cell migration was assessed using an in vitro chemotaxis assay. Additionally, TNBC cells were exposed to TGF-β1, Snail, and Zeb1, and IFN-β production and downstream autocrine signaling were assessed via RNA-seq, qRT-PCR, and Western blotting. RESULTS Here, we identify the repression of Toll-like Receptor 3 (TLR3), an innate immune sensor, as the key molecular event linking OSM signaling and the repression of IFN-β transcription, production, and autocrine IFN signaling. Moreover, we demonstrate that additional epithelial-mesenchymal transition-inducing factors, such as TGF-β1, Snail, and Zeb1, similarly suppress TLR3-mediated IFN-β production and signaling. CONCLUSIONS Our findings provide a novel insight into the regulation of TLR3 and IFN-β production in TNBC cells, which are known indicators of treatment responses to DNA-damaging therapies. Furthermore, strategies to stimulate TLR3 in order to increase IFN-β within the TME may be ineffective in stem-like/mesenchymal cells, as TLR3 is strongly repressed. Rather, we propose that therapies targeting OSM or OSM receptor would reverse the stem-like/mesenchymal program and restore TLR3-mediated IFN-β production within the TME, facilitating improved responses to current therapies.
Collapse
Affiliation(s)
- Noah M Chernosky
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Ilaria Tamagno
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Kelsey L Polak
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - E Ricky Chan
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, 44106, USA
| | - Xueer Yuan
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - Mark W Jackson
- Department of Pathology Case, Western Reserve University, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
20
|
Yau AWN, Chu SYC, Yap WH, Wong CL, Chia AYY, Tang YQ. Phage display screening in breast cancer: From peptide discovery to clinical applications. Life Sci 2024; 357:123077. [PMID: 39332485 DOI: 10.1016/j.lfs.2024.123077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Breast cancer is known as the most common type of cancer found in women and a leading cause of cancer death in women, with the global incidence only increasing. Breast cancer in Malaysia is also unfortunately the most prevalent in Malaysian women. Many treatment options are available for breast cancer, but there is increasing resistance developed against treatment and increased recurrence risk, emphasizing the need for new treatment options. This review will focus on the applications of phage display screening in the context of breast cancer. Phage display screening can facilitate the drug discovery process by providing rapid screening and isolation of peptides that bind to targets of interest with high specificity. Peptides derived from phage display target various types of proteins involved in breast cancer, including HER2, C5AR1, p53 and PRDM14, either for therapeutic or diagnostic purposes. Different approaches were employed as well to produce potential peptides using radiolabelling and conjugation techniques. Promising results were reported for in vitro and in vivo studies utilizing peptides derived from phage display screening. Further optimization of the protocols and factors to consider are required to mitigate the challenges involved with phage display screening of peptides for breast cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ashlyn Wen Ning Yau
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Sylvester Yee Chun Chu
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Wei Hsum Yap
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Chuan Loo Wong
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Digital Health and Medical Advancement Impact lab, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Adeline Yoke Yin Chia
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Digital Health and Medical Advancement Impact lab, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Yin-Quan Tang
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia; Digital Health and Medical Advancement Impact lab, Taylor's University, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
21
|
Torres JM, Sodipo MO, Hopkins MF, Chandler PD, Warner ET. Racial Differences in Breast Cancer Survival Between Black and White Women According to Tumor Subtype: A Systematic Review and Meta-Analysis. J Clin Oncol 2024; 42:3867-3879. [PMID: 39288352 PMCID: PMC11540747 DOI: 10.1200/jco.23.02311] [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: 10/24/2023] [Revised: 05/08/2024] [Accepted: 06/24/2024] [Indexed: 09/19/2024] Open
Abstract
PURPOSE Despite effective early-detection approaches and innovative treatments, Black women in the United States have higher breast cancer mortality rates compared with White women. The purpose of this systematic review and meta-analysis is to determine the extent of disparities in breast cancer survival between Black and White women according to tumor subtype. METHODS A comprehensive database search was performed for full-text, English-language articles published from January 1, 2000, to December 31, 2022. Included studies compared survival between Black and White female patients with breast cancer within subtypes defined by hormone receptor and human epidermal growth factor receptor 2 (HER2)/neu (HER2; now known as ERBB2) status. Random-effects models were used to combine study-specific results and generate pooled relative risks (RRs) and 95% CIs for breast cancer-specific or overall survival (OS). A protocol for this review was registered in PROSPERO (CRD42021268212). RESULTS Eighteen studies including 228,885 (34,262 Black; 182,466 White) patients with breast cancer were identified. Compared with White women, Black women had a higher risk of breast cancer death for all tumor subtypes. The summary risk of breast cancer death was 50% higher among hormone receptor-positive HER2-negative [HER2-] tumors (RR, 1.50 [95% CI, 1.30 to 1.72]), 34% higher for hormone receptor+/HER2+ (RR, 1.34 [95% CI, 1.10 to 1.64]), 20% higher for hormone receptor-negative (-)/HER2+ (RR, 1.29 [95% CI, 1.00 to 1.43]), and 17% higher among individuals with hormone receptor-/HER2- tumors (hazard ratio, 1.17; 95% CI, 1.10 to 1.25). Black women also had poorer OS than White women for all subtypes. CONCLUSION These results suggest there are both subtype-specific and subtype-independent mechanisms that contribute to disparities in breast cancer survival between Black and White women, which require multilevel interventions to address and achieve health equity.
Collapse
Affiliation(s)
| | - Michelle O. Sodipo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Margaret F. Hopkins
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paulette D. Chandler
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Pfizer, Inc., Cambridge, MA, USA
| | - Erica T. Warner
- Clinical Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston MA
| |
Collapse
|
22
|
Zuo X, Zhao X, Zhang X, Li Q, Jiang X, Huang S, Chen X, Chen X, Jia W, Zou H, Shi D, Qian X. PTPN20 promotes metastasis through activating NF-κB signaling in triple-negative breast cancer. Breast Cancer Res 2024; 26:155. [PMID: 39506852 PMCID: PMC11542355 DOI: 10.1186/s13058-024-01910-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: 07/22/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Cancer metastasis remains a major challenge in the clinical management of triple-negative breast cancer (TNBC). The NF-κB signaling pathway has been implicated as a crucial factor in the development of metastases, but the underlying molecular mechanisms remain largely unclear. METHODS PTPN20 expression was evaluated using data from the Sweden Cancerome Analysis Network-Breast and The Cancer Genome Atlas database, as well as by western blotting and immunohistochemistry in 88 TNBC patients. The ability of PTPN20 to activate NF-κB was assessed by luciferase reporter assays. The effects of PTPN20 overexpression and knockdown via short hairpin RNA were examined in TNBC cell lines by wound healing and transwell matrix penetration assays. Additionally, we analyzed the growth and metastasis abilitiy of 4T1 xenograft tumors in nude mice. RESULTS PTPN20 levels were elevated in TNBC cell lines and patient samples compared to controls, and higher protein levels correlated with metastasis-free survival. Overexpression of PTPN20 enhanced migration and invasion in vitro, and promoted lung metastasis in vivo. Our finding revealed that PTPN20 activates NF-κB signaling by dephosphorylating p65 at Ser468, preventing its binding to COMMD1, thereby protecting p65 from degradation. Downregulation of PTPN20 effectively inhibit, while p65 S468A mutant restored the migratory and invasive abilities of TNBC cells. CONCLUSIONS Collectively, our results demonstrate that PTPN20 plays a critical role in TNBC metastasis through the activation of NF-κB signaling. We propose that PTPN20 may serve as a novel prognostic marker and potential therapeutic target for the treatment of TNBC.
Collapse
Affiliation(s)
- Xiaoxiao Zuo
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaohan Zhao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Xiaofei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qingyuan Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Xingyu Jiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xuwei Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Weihua Jia
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
- Department of Biobank, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Hequn Zou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Dongni Shi
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China.
| | - Xueke Qian
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
23
|
Arzuk E, Birim D, Armağan G. Celecoxib inhibits NLRP1 inflammasome pathway in MDA-MB-231 Cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9191-9202. [PMID: 38990306 PMCID: PMC11522188 DOI: 10.1007/s00210-024-03286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
NLRP1 is predominantly overexpressed in breast cancer tissue, and the evaluated activation of NLRP1 inflammasome is associated with tumor growth, angiogenesis, and metastasis. Therefore, targeting NLRP1 activation could be a crucial strategy in anticancer therapy. In this study, we investigated the hypothesis that NLRP1 pathway may contribute to the cytotoxic effects of celecoxib and nimesulide in MDA-MB-231 cells. First of all, IC50 values and inhibitory effects on the colony-forming ability of drugs were evaluated in cells. Then, the alterations in the expression levels of NLRP1 inflammasome components induced by drugs were investigated. Subsequently, the release of inflammatory cytokine IL-1β and the activity of caspase-1 in drug-treated cells were measured. According to our results, celecoxib and nimesulide selectively inhibited the viability of MDA-MB-231 cells. These drugs remarkably inhibited the colony-forming ability of cells. The expression levels of NLRP1 inflammasome components decreased in celecoxib-treated cells, accompanied by decreased caspase-1 activity and IL-1β release. In contrast, nimesulide treatment led to the upregulation of the related protein expressions with unchanged caspase-1 activity and increased IL-1β secretion. Our results indicated that the NLRP1 inflammasome pathway might contribute to the antiproliferative effects of celecoxib in MDA-MB-231 cells but is not a crucial mechanism for nimesulide.
Collapse
Affiliation(s)
- Ege Arzuk
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ege University, Bornova, 35040, Izmir, Turkey.
| | - Derviş Birim
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| | - Güliz Armağan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
24
|
Wu S, Zheng Y, Olopade OI. The convergence of genomic medicine and translational omics in transforming breast cancer patient care. J Clin Invest 2024; 134:e187520. [PMID: 39484719 PMCID: PMC11527438 DOI: 10.1172/jci187520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Affiliation(s)
- Sulin Wu
- Section of Hematology and Oncology, Department of Medicine and
| | - Yonglan Zheng
- Section of Hematology and Oncology, Department of Medicine and
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Olufunmilayo I. Olopade
- Section of Hematology and Oncology, Department of Medicine and
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
25
|
Moreno-Ulloa A, Zárate-Córdova VL, Ramírez-Sánchez I, Cruz-López JC, Perez-Ortiz A, Villarreal-Garza C, Díaz-Chávez J, Estrada-Mena B, Antonio-Aguirre B, López-Almanza PX, Lira-Romero E, Estrada-Mena FJ. Evaluation of a Proteomics-Guided Protein Signature for Breast Cancer Detection in Breast Tissue. J Proteome Res 2024; 23:4907-4923. [PMID: 39412830 DOI: 10.1021/acs.jproteome.4c00295] [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] [Indexed: 11/02/2024]
Abstract
The distinction between noncancerous and cancerous breast tissues is challenging in clinical settings, and discovering new proteomics-based biomarkers remains underexplored. Through a pilot proteomic study (discovery cohort), we aimed to identify a protein signature indicative of breast cancer for subsequent validation using six published proteomics/transcriptomics data sets (validation cohorts). Sequential window acquisition of all theoretical (SWATH)-based mass spectrometry revealed 370 differentially abundant proteins between noncancerous tissue and breast cancer. Protein-protein interaction-based networks and enrichment analyses revealed dysregulation in pathways associated with extracellular matrix organization, platelet degranulation, the innate immune system, and RNA metabolism in breast cancer. Through multivariate unsupervised analysis, we identified a four-protein signature (OGN, LUM, DCN, and COL14A1) capable of distinguishing breast cancer. This dysregulation pattern was consistently verified across diverse proteomics and transcriptomics data sets. Dysregulation magnitude was notably higher in poor-prognosis breast cancer subtypes like Basal-Like and HER2 compared to Luminal A. Diagnostic evaluation (receiver operating characteristic (ROC) curves) of the signature in distinguishing breast cancer from noncancerous tissue revealed area under the curve (AUC) ranging from 0.87 to 0.9 with predictive accuracy of 80% to 82%. Upon stratifying, to solely include the Basal-Like/Triple-Negative subtype, the ROC AUC increased to 0.922-0.959 with predictive accuracy of 84.2%-89%. These findings suggest a potential role for the identified signature in distinguishing cancerous from noncancerous breast tissue, offering insights into enhancing diagnostic accuracy.
Collapse
Affiliation(s)
- Aldo Moreno-Ulloa
- Laboratorio MS2, Departamento de Innovación Biomédica, CICESE, Ensenada 22860, Baja California, México
| | - Vareska L Zárate-Córdova
- Laboratorio MS2, Departamento de Innovación Biomédica, CICESE, Ensenada 22860, Baja California, México
- Posgrado en Ciencias de la Vida, CICESE, Ensenada 22860, Baja California, México
| | - Israel Ramírez-Sánchez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, IPN, Ciudad de México 11340, México
| | - Juan Carlos Cruz-López
- Hospital Puebla, Puebla 72197, Pue., México
- Hospital General Zona Norte SSEP, Puebla 72200, Pue., México
| | - Andric Perez-Ortiz
- Escuela de Medicina, Universidad Panamericana, Ciudad de México 03920, México
- Departamento de Cirugía, Centro Médico ABC, Ciudad de México 05348, México
| | - Cynthia Villarreal-Garza
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, Monterrey 66260, Nuevo León, México
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Ciudad de México 14080, México
| | - Benito Estrada-Mena
- Escuela de Enfermería, Universidad Panamericana, Ciudad de México 03920, México
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | | | | | | | | |
Collapse
|
26
|
Ormiston K, Kulkarni A, Sarathy G, Alsammerai S, Shankar E, Majumder S, Stanford KI, Ganju RK, Ramaswamy B. Obesity and lack of breastfeeding: a perfect storm to augment risk of breast cancer? Front Oncol 2024; 14:1432208. [PMID: 39525621 PMCID: PMC11543574 DOI: 10.3389/fonc.2024.1432208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer with higher rates of recurrence and distant metastasis, as well as decreased 5-year survival rates. Racial disparities are evident in the incidence and mortality rates of triple negative breast cancer particularly increased in young African American women. Concurrently, young African American women have multiple risk factors for TNBC including higher rates of premenopausal abdominal obesity (higher waist-hip ratio) and lower rates of breastfeeding with higher parity, implicating these factors as potentially contributors to poor outcomes. By understanding the mechanisms of how premenopausal obesity and lack of breastfeeding may be associated with increased risk of triple negative breast cancer, we can determine the best strategies for intervention and awareness to improve outcomes in TNBC.
Collapse
Affiliation(s)
- Kate Ormiston
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Anagh Kulkarni
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Gautam Sarathy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Sara Alsammerai
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Eswar Shankar
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Sarmila Majumder
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Kristin I. Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Ramesh K. Ganju
- Department of Pathology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
27
|
Rivas FWS, Gonçalves R, Mota BS, Sorpreso ICE, Toporcov TN, Filassi JR, Lopes EDT, Schio LR, Comtesse YLP, Baracat EC, Soares Júnior JM. Comprehensive diagnosis of advanced-stage breast cancer: exploring detection methods, molecular subtypes, and demographic influences - A cross-sectional study. Clinics (Sao Paulo) 2024; 79:100510. [PMID: 39413498 PMCID: PMC11530810 DOI: 10.1016/j.clinsp.2024.100510] [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: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Brazil faces notable Breast Cancer (BC) mortality despite lower incidence rates versus developed countries. Despite guidelines from medical societies, Brazilian public policy recommends biennial mammographic screening for women aged 50 to 69. This study investigates sociodemographic and clinical factors related to BC detection methods and clinical stage at diagnosis. METHODS The authors conducted a cross-sectional study at a São Paulo tertiary hospital. Patients were divided into 'symptomatic' and 'mammographic' detection groups. Bivariate analyses by detection method and clinical stage compared groups' profiles in terms of sociodemographic and clinical characteristics. Poisson regression analyses assessed sociodemographic and molecular subtypes´ influence on "mammographic detection" prevalence and "advanced-stage BC", reporting prevalence ratios and 95 % Confidence Intervals. RESULTS The authors studied 1,536 BC patients admitted from January 2016 to December 2017. The "mammographic detection" group had a higher proportion of patients aged 50‒69 years (62.9 % vs. 44.1 %), white race (63.3 % vs. 51.6 %), Catholic religion (58.2 % vs. 51.1 %), and Luminal A subtype (25.2 % vs. 13.2 %) compared to the "symptomatic detection" group. Patients with early-stage disease were more likely to have higher education levels (8.1 % vs. 5.5 %) and be married (39.8 % vs. 46.6 %) compared to those with advanced-stage. Molecular subtypes were significantly associated with the detection method and stage. The prevalence of advanced-stage disease in "mammographic" (n=313) and "symptomatic" (n=1191) groups was 18.5 % and 55 %, respectively . Mammographic detection significantly reduced advanced-stage BC prevalence (PR = 0.40, 95 % CI 0.31‒0.51). CONCLUSION Mammographic detection reduces advanced-stage breast cancer prevalence in Brazil, emphasizing the importance of regular screenings, especially among at-risk sociodemographic groups. Enhancing mammographic screening accessibility, lowering the starting age to 40, and extending coverage to include annual mammograms can significantly lower breast cancer mortality in Brazil, benefiting public health and patient outcomes.
Collapse
Affiliation(s)
- Fernando Wladimir Silva Rivas
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rodrigo Gonçalves
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Setor de Mastologia da Disciplina de Ginecologia, Instituto de Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Bruna Salani Mota
- Setor de Mastologia da Disciplina de Ginecologia, Instituto de Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Isabel Cristina Esposito Sorpreso
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Tatiana Natasha Toporcov
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, SP, Brazil
| | - José Roberto Filassi
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Setor de Mastologia da Disciplina de Ginecologia, Instituto de Câncer do Estado de São Paulo, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Edia di Tullio Lopes
- Registro Hospitalar de Câncer, Serviço de Arquivo Médico, Instituto de Câncer do Estado de São Paulo, São Paulo, SP, Brazil
| | - Laura Raíssa Schio
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Yann-Luc Patrick Comtesse
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Edmund Chada Baracat
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - José Maria Soares Júnior
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
28
|
Sasaki T, Liyanage A, Bansil S, Silva A, Pagano I, Hidalgo EY, Jones C, Ueno NT, Takahashi Y, Fukui J. Differences in Breast Cancer Subtypes among Racial/Ethnic Groups. Cancers (Basel) 2024; 16:3462. [PMID: 39456556 PMCID: PMC11506832 DOI: 10.3390/cancers16203462] [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: 09/07/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Differences in the incidence of breast cancer subtypes among racial/ethnic groups have been evaluated as a contributing factor in disparities seen in breast cancer prognosis. We evaluated new breast cancer cases in Hawai'i to determine if there were subtype differences according to race/ethnicity that may contribute to known disparities. METHODS We reviewed 4591 cases of women diagnosed with breast cancer from two large tumor registries between 2015 and 2022. We evaluated breast cancer cases according to age at diagnosis, self-reported race, breast cancer subtype (ER, PR, and HER2 receptor status), histology, county, and year. RESULTS We found both premenopausal and postmenopausal Native Hawaiian women were less likely to be diagnosed with triple-negative breast cancer (OR = 0.26, 95% CI 0.12-0.58 p = 0.001; OR = 0.54, 95% CI 0.36, 0.80 p = 0.002, respectively). CONCLUSIONS The results of our study support that there are racial/ethnic differences in breast cancer subtypes among our population, which may contribute to differences in outcomes. Further evaluation of clinical and pathological features in each breast cancer subtype may help improve the understanding of outcome disparities seen among different racial/ethnic groups.
Collapse
Affiliation(s)
- Tamlyn Sasaki
- John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA; (T.S.); (N.T.U.)
| | - Akash Liyanage
- John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA; (T.S.); (N.T.U.)
| | - Surbhi Bansil
- John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA; (T.S.); (N.T.U.)
| | - Anthony Silva
- John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA; (T.S.); (N.T.U.)
| | - Ian Pagano
- Translational and Clinical Research Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA; (I.P.); (Y.T.)
| | - Elena Y. Hidalgo
- Queen’s Medical Center Oncology Data Registry, Honolulu, HI 96813, USA
| | - Corinne Jones
- Kapi’olani Medical Center for Women and Children, Honolulu, HI 96822, USA;
| | - Naoto T. Ueno
- John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA; (T.S.); (N.T.U.)
- Translational and Clinical Research Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA; (I.P.); (Y.T.)
- Kapi’olani Medical Center for Women and Children, Honolulu, HI 96822, USA;
- The Queen’s Health Systems, Honolulu, HI 96813, USA
| | - Yoko Takahashi
- Translational and Clinical Research Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA; (I.P.); (Y.T.)
| | - Jami Fukui
- John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA; (T.S.); (N.T.U.)
- Translational and Clinical Research Program, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA; (I.P.); (Y.T.)
- Kapi’olani Medical Center for Women and Children, Honolulu, HI 96822, USA;
- The Queen’s Health Systems, Honolulu, HI 96813, USA
| |
Collapse
|
29
|
Klayech Z, Moussa A, Souid M, Hadhri R, Miled S, Gabbouj S, Remadi Y, Faleh R, Bouaouina N, Zakhama A, Hassen E. Prognostic Significance of Combining Cytokeratin-19, E-Cadherin and Ki-67 Analysis in Triple-Negative Breast Cancer with Basal-Like and Non-Basal-Like Phenotype. Cancer Invest 2024; 42:769-781. [PMID: 39435793 DOI: 10.1080/07357907.2024.2416166] [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: 01/25/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
Triple-negative breast cancer (TNBC) is known to have the worst outcome compared to the other forms of breast cancer. Moreover, molecular markers identified basal-like breast cancer (BLBC) phenotypes to be also related to a worse prognosis. In this study, we evaluated by immunohistochemistry (IHC) the prognostic significance of combining Cytokeratin-19 (CK19), E-cadherin, and Ki-67 tissue expression in triple-negative breast cancer (TNBC) cases presenting a basal-like (BLBC) or a non-basal-like (n-BLBC) phenotype to improve the selection and the monitoring of BC patients with a more aggressive outcome. Herein, when compared to n-BLBC, patients with BLBC showed a positive correlation with lymph node metastasis occurrence and lower survival rates. Immunohistochemistry analysis revealed significantly lower E-cadherin prevalence and higher prevalence of both CK19 and Ki-67 in BLBC when compared to n-BLBC. Spearman correlation showed that E-cadherin is negatively and significantly correlated to CK19 and Ki-67 expressions. Moreover, in BLBC, expressing both CK19 and Ki-67 combined with E-cadherin loss was associated with the worst relapse-free and overall survival. In conclusion, TNBC/BLBC phenotypes simultaneously losing E-cadherin and overexpressing CK19 and Ki-67 markers are the most aggressive forms. This combined analysis could be a predictive marker of poor prognosis.
Collapse
Affiliation(s)
- Zahra Klayech
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| | - Adnene Moussa
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Anatomy and Pathologic Cytology, Fattouma Bourguiba University Hospital, Monastir University, Monastir, Tunisia
| | - Moufida Souid
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| | - Rim Hadhri
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Anatomy and Pathologic Cytology, Fattouma Bourguiba University Hospital, Monastir University, Monastir, Tunisia
| | - Souad Miled
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Anatomy and Pathologic Cytology, Fattouma Bourguiba University Hospital, Monastir University, Monastir, Tunisia
| | - Sallouha Gabbouj
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
| | - Yassmine Remadi
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
| | - Raja Faleh
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Gynecology and Obstetrics, Fattouma Bourguiba University Hospital, Monastir University, Monastir, Tunisia
| | - Noureddine Bouaouina
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Ibn Khaldoun Medical Center of Cancerology, Sousse, Tunisia
| | - Abdelfattah Zakhama
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Anatomy and Pathologic Cytology, Fattouma Bourguiba University Hospital, Monastir University, Monastir, Tunisia
| | - Elham Hassen
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| |
Collapse
|
30
|
Posani SH, Gillis NE, Lange CA. Glucocorticoid receptors orchestrate a convergence of host and cellular stress signals in triple negative breast cancer. J Steroid Biochem Mol Biol 2024; 243:106575. [PMID: 38950871 PMCID: PMC11344665 DOI: 10.1016/j.jsbmb.2024.106575] [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: 11/06/2023] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks expression of the nuclear steroid receptors that bind estrogens (ER) and progestogens (PRs) and does not exhibit HER2 (Human epidermal growth factor 2) receptor overexpression. Even in the face of initially effective chemotherapies, TNBC patients often relapse. One primary cause for therapy-resistant tumor progression is the activation of cellular stress signaling pathways. The glucocorticoid receptor (GR), a corticosteroid-activated transcription factor most closely related to PR, is a mediator of both endocrine/host stress and local tumor microenvironment (TME)-derived and cellular stress responses. Interestingly, GR expression is associated with a good prognosis in ER+ breast cancer but predicts poor prognosis in TNBC. Classically, GR's transcriptional activity is regulated by circulating glucocorticoids. Additionally, GR is regulated by ligand-independent signaling events. Notably, the stress-activated protein kinase, p38 MAP kinase, phosphorylates GR at serine 134 (Ser134) in response to TME-derived growth factors and cytokines, including HGF and TGFβ1. Phospho-Ser134-GR (p-Ser134-GR) associates with cytoplasmic and nuclear signaling molecules, including 14-3-3ζ, aryl hydrocarbon receptors (AhR), and hypoxia-inducible factors (HIFs). Phospho-GR/HIF-containing transcriptional complexes upregulate gene sets whose protein products include the components of inducible oncogenic signaling pathways (PTK6) that further promote cancer cell survival, chemoresistance, altered metabolism, and migratory/invasive behavior in TNBC. Recent studies have implicated liganded p-Ser134-GR (p-GR) in dexamethasone-mediated upregulation of genes related to TNBC cell motility and dysregulated metabolism. Herein, we review the tumor-promoting roles of GR and discuss how both ligand-dependent and ligand-independent/stress signaling-driven inputs to p-GR converge to orchestrate metastatic TNBC progression.
Collapse
Affiliation(s)
- Sai Harshita Posani
- Molecular Pharmacology and Therapeutics Program, University of Minnesota, Minneapolis 55455, United States; Department of Pharmacology, University of Minnesota, Minneapolis 55455, United States
| | - Noelle E Gillis
- Masonic Cancer Center, University of Minnesota, Minneapolis 55455, United States
| | - Carol A Lange
- Masonic Cancer Center, University of Minnesota, Minneapolis 55455, United States; Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis 55455, United States; Department of Pharmacology, University of Minnesota, Minneapolis 55455, United States.
| |
Collapse
|
31
|
Sun X, Kennedy LC, Gonzalez-Ericsson PI, Sanchez V, Sanders M, Perou CM, Troester MA, Balko JM, Reid SA. Associations of Immune Checkpoint Predictive Biomarkers (MHC-I and MHC-II) with Clinical and Molecular Features in a Diverse Breast Cancer Cohort. Clin Cancer Res 2024; 30:4077-4081. [PMID: 39007881 PMCID: PMC11398978 DOI: 10.1158/1078-0432.ccr-24-1286] [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: 04/23/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE Immunotherapy (IO) in triple-negative breast cancer (TNBC) has improved survival outcomes, with promising improvements in pCR rates among early high-risk hormone receptor (HR)+/HER2- breast cancers. However, biomarkers are needed to select patients likely to benefit from IO. MHC-I and tumor-specific MHC-II (tsMHC-II) expression are candidate biomarkers for PD-(L)1 checkpoint inhibition but existing data from clinical trials included limited racial/ethnic diversity. EXPERIMENTAL DESIGN We performed multiplexed immunofluorescence assays in the Carolina Breast Cancer Study (CBCS; n = 1,628, 48% Black, 52% non-Black). Intrinsic subtype and P53 mutant-like status were identified using RNA-based multigene assays. We ranked participants based on tumoral MHC-I intensity (top 33% categorized as "MHC-Ihigh") and MHC-II+ (≥5% of tumor cells as tsMHC-II+). MHC-I/II were evaluated in association with clinicopathological features by race. RESULTS Black participants had higher frequency of TNBC (25% vs. 12.5%, P ≤ 0.001) and basal-like (30% vs. 14%, P ≤ 0.001) tumors overall, and higher frequency of basal-like (11% vs. 5.5%, P = 0.002) and TP53 mutant tumors (26% vs. 17%, P = 0.002) among HR+/HER2-. The frequency of tsMHC-II+ was higher in HR+/HER2- Black participants (7.9% vs. 4.9%, P = 0.04). Black participants also had higher frequency of MHC-Ihigh (38.7% vs. 28.2%, P < 0.001), which was significant among HR+/HER2- (28.2% vs. 22.1%, P = 0.02). CONCLUSIONS In this diverse study population, MHC-I and MHC-II tumor cell expression were more highly expressed in HR+/HER2- tumors from Black women, underscoring the importance of diverse and equitable enrollment in future IO trials.
Collapse
Affiliation(s)
- Xiaopeng Sun
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Laura C Kennedy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Paula I Gonzalez-Ericsson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Violeta Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Melinda Sanders
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | | | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sonya A Reid
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
32
|
Lopes LCP, Medeiros GA, Gualberto IJN, Gut TB, Ferrazini RVS, Negrato CA. Relationship between early age at menarche, older age at menopause and subtypes of breast cancer: a scoping review. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2024; 46:e-rbgo50. [PMID: 39380584 PMCID: PMC11460410 DOI: 10.61622/rbgo/2024rbgo50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/08/2024] [Indexed: 10/10/2024] Open
Abstract
Objective To determine the relationship between early age at menarche, late age at menopause with specific subtypes of breast cancer (BC). Methods A literature search was conducted in Embase, Lilacs, PubMed, Scopus, and Scielo databases, following the Joanna Briggs Institute scoping review protocol and answering the question "How early age at menarche or late age at menopause are related to different breast cancer subtypes?". Results A number of 4,003 studies were identified, of which 17 were selected. Most of the included articles found a clear relationship between early menarche, late menopause and some subtypes of BC, mainly, PR+, ER+, luminal, and HER-2 tumors. However, some studies have found a contradictory relationship and one study didn't find any relationship between them. Conclusion A relationship between early age at menarche and advanced age at menopause was observed with some subtypes of breast cancer, since other factors must be considered in its understanding.
Collapse
Affiliation(s)
- Lucas Casagrande Passoni Lopes
- Faculdade de MedicinaUniversidade de São PauloBauruSPBrazil Faculdade de Medicina, Universidade de São Paulo, Bauru, SP, Brazil.
| | - Gabriel Araújo Medeiros
- Faculdade de MedicinaUniversidade de São PauloBauruSPBrazil Faculdade de Medicina, Universidade de São Paulo, Bauru, SP, Brazil.
| | - Igor José Nogueira Gualberto
- Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrazil Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Thales Baptista Gut
- Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrazil Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | | | - Carlos Antonio Negrato
- Faculdade de MedicinaUniversidade de São PauloBauruSPBrazil Faculdade de Medicina, Universidade de São Paulo, Bauru, SP, Brazil.
| |
Collapse
|
33
|
Strand SH, Houlahan KE, Branch V, Lynch T, Rivero-Guitiérrez B, Harmon B, Couch F, Gallagher K, Kilgore M, Wei S, DeMichele A, King T, McAuliffe P, Curtis C, Owzar K, Marks JR, Colditz GA, Hwang ES, West RB. Analysis of ductal carcinoma in situ by self-reported race reveals molecular differences related to outcome. Breast Cancer Res 2024; 26:127. [PMID: 39223670 PMCID: PMC11367816 DOI: 10.1186/s13058-024-01885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a non-obligate precursor to invasive breast cancer (IBC). Studies have indicated differences in DCIS outcome based on race or ethnicity, but molecular differences have not been investigated. METHODS We examined the molecular profile of DCIS by self-reported race (SRR) and outcome groups in Black (n = 99) and White (n = 191) women in a large DCIS case-control cohort study with longitudinal follow up. RESULTS Gene expression and pathway analyses suggested that different genes and pathways are involved in diagnosis and ipsilateral breast outcome (DCIS or IBC) after DCIS treatment in White versus Black women. We identified differences in ER and HER2 expression, tumor microenvironment composition, and copy number variations by SRR and outcome groups. CONCLUSIONS Our results suggest that different molecular mechanisms drive initiation and subsequent ipsilateral breast events in Black versus White women.
Collapse
MESH Headings
- Adult
- Aged
- Female
- Humans
- Middle Aged
- Biomarkers, Tumor/genetics
- Black or African American/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/ethnology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/ethnology
- Case-Control Studies
- DNA Copy Number Variations
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Receptors, Estrogen/metabolism
- Self Report
- Tumor Microenvironment/genetics
- White/genetics
Collapse
Affiliation(s)
- Siri H Strand
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kathleen E Houlahan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Genetics, Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Vernal Branch
- National Breast Cancer Coalition, 2001 L Street NW, Suite 500 PMB#50111, Washington, DC, 20036, USA
| | - Thomas Lynch
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27708, USA
| | | | - Bryan Harmon
- Department of Pathology, Montefiore Medical Center, New York City, NY, USA
| | - Fergus Couch
- Department of Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kristalyn Gallagher
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark Kilgore
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Angela DeMichele
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tari King
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Christina Curtis
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Genetics, Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27708, USA
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Jeffrey R Marks
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Graham A Colditz
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
34
|
Karmach A, Beaudoin IS, Navina S, Di Como JA. Triple-Negative Papillary Carcinoma of the Breast: A Case Report. Cureus 2024; 16:e69020. [PMID: 39385920 PMCID: PMC11463877 DOI: 10.7759/cureus.69020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Papillary carcinoma is a rare form of breast malignancy, representing only a small percentage of newly diagnosed breast cancers. Bloody nipple discharge is the most consistent symptom reported among patients. These lesions are visualized histologically as fibrovascular cores lined with proliferating neoplastic epithelial cells. Papillary breast carcinomas are characterized by estrogen receptor (ER), progesterone receptor (PR), and/or human epidermal growth factor receptor 2 (HER2) positivity, allowing for targeted therapeutic approaches with favorable outcomes. Triple-negative papillary carcinoma (TNPC) is a rare variant that lacks this characteristic hormone receptor expression, creating a unique challenge in diagnosis and management. Here, we highlight the case of a 43-year-old asymptomatic female with TNPC following an abnormal screening mammogram that revealed a suspicious mass in the left breast. Surgical excision with clear margins remains the cornerstone of treatment, with adjuvant chemotherapy considered for high-risk cases. As there is limited evidence on the efficacy of targeted therapies and hormone-based treatments, this case analyzes the diagnostic criteria, therapeutic options, and prognosis of TNPC to prompt further investigation into specific treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Joseph A Di Como
- Breast Surgical Oncology, Ironwood Cancer and Research Centers, Scottsdale, USA
| |
Collapse
|
35
|
Schneider BP, Zhao F, Ballinger TJ, Garcia SF, Shen F, Virani S, Cella D, Bales C, Jiang G, Hayes L, Miller N, Srinivasiah J, Stringer-Reasor EM, Chitalia A, Davis AA, Makower DF, Incorvati J, Simon MA, Mitchell EP, DeMichele A, Miller KD, Sparano JA, Wagner LI, Wolff AC. ECOG-ACRIN EAZ171: Prospective Validation Trial of Germline Predictors of Taxane-Induced Peripheral Neuropathy in Black Women With Early-Stage Breast Cancer. J Clin Oncol 2024; 42:2899-2907. [PMID: 38828938 PMCID: PMC11670807 DOI: 10.1200/jco.24.00526] [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: 03/14/2024] [Revised: 04/16/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
PURPOSE Black women experience higher rates of taxane-induced peripheral neuropathy (TIPN) compared with White women when receiving adjuvant once weekly paclitaxel for early-stage breast cancer, leading to more dose reductions and higher recurrence rates. EAZ171 aimed to prospectively validate germline predictors of TIPN and compare rates of TIPN and dose reductions in Black women receiving (neo)adjuvant once weekly paclitaxel and once every 3 weeks docetaxel for early-stage breast cancer. METHODS Women with early-stage breast cancer who self-identified as Black and had intended to receive (neo)adjuvant once weekly paclitaxel or once every 3 weeks docetaxel were eligible, with planned accrual to 120 patients in each arm. Genotyping was performed to determine germline neuropathy risk. Grade 2-4 TIPN by Common Terminology Criteria for Adverse Events (CTCAE) v5.0 was compared between high- versus low-risk genotypes and between once weekly paclitaxel versus once every 3 weeks docetaxel within 1 year. Patient-rated TIPN and patient-reported outcomes were compared using patient-reported outcome (PRO)-CTCAE and Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity. RESULTS Two hundred and forty of 249 enrolled patients had genotype data, and 91 of 117 (77.8%) receiving once weekly paclitaxel and 87 of 118 (73.7%) receiving once every 3 weeks docetaxel were classified as high-risk. Physician-reported grade 2-4 TIPN was not significantly different in high- versus low-risk genotype groups with once weekly paclitaxel (47% v 35%; P = .27) or with once every 3 weeks docetaxel (28% v 19%; P = .47). Grade 2-4 TIPN was significantly higher in the once weekly paclitaxel versus once every 3 weeks docetaxel arm by both physician-rated CTCAE (45% v 29%; P = .02) and PRO-CTCAE (40% v 24%; P = .03). Patients receiving once weekly paclitaxel required more dose reductions because of TIPN (28% v 9%; P < .001) or any cause (39% v 25%; P = .02). CONCLUSION Germline variation did not predict risk of TIPN in Black women receiving (neo)adjuvant once weekly paclitaxel or once every 3 weeks docetaxel. Once weekly paclitaxel was associated with significantly more grade 2-4 TIPN and required more dose reductions than once every 3 weeks docetaxel.
Collapse
Affiliation(s)
| | - Fengmin Zhao
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center
| | | | - Sofia F Garcia
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center
| | - Fei Shen
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | | | - David Cella
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center
| | - Casey Bales
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | - Guanglong Jiang
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | | | | | | | | | | | - Andrew A. Davis
- Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Melissa A. Simon
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center
| | | | | | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | | | | | | |
Collapse
|
36
|
Fortunato A, Mallo D, Cisneros L, King LM, Khan A, Curtis C, Ryser MD, Lo JY, Hall A, Marks JR, Hwang ES, Maley CC. Evolutionary Measures Show that Recurrence of DCIS is Distinct from Progression to Breast Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.15.24311949. [PMID: 39185534 PMCID: PMC11343254 DOI: 10.1101/2024.08.15.24311949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Progression from pre-cancers like ductal carcinoma in situ (DCIS) to invasive disease (cancer) is driven by somatic evolution and is altered by clinical interventions. We hypothesized that genetic and/or phenotypic intra-tumor heterogeneity would predict clinical outcomes for DCIS since it serves as the substrate for natural selection among cells. We profiled two samples from two geographically distinct foci from each DCIS in both cross-sectional (N = 119) and longitudinal cohorts (N = 224), with whole exome sequencing, low-pass whole genome sequencing, and a panel of immunohistochemical markers. In the longitudinal cohorts, the only statistically significant predictors of time to non-invasive DCIS recurrence were the combination of treatment (lumpectomy only vs mastectomy or lumpectomy with radiation, HR = 12.13, p = 0.003, Wald test with FDR correction), ER status (HR = 0.16 for ER+ compared to ER-, p = 0.0045), and divergence in SNVs between the two samples (HR = 1.33 per 10% divergence, p = 0.018). SNV divergence also distinguished between pure DCIS and DCIS synchronous with invasive disease in the cross-sectional cohort. In contrast, the only statistically significant predictors of time to progression to invasive disease were the combination of the width of the surgical margin (HR = 0.67 per mm, p = 0.043) and the number of mutations that were detectable at high allele frequencies (HR = 1.30 per 10 SNVs, p = 0.02). These results imply that recurrence with DCIS is a clinical and biological process different from invasive progression.
Collapse
Affiliation(s)
- Angelo Fortunato
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ 85287, USA
| | - Diego Mallo
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ 85287, USA
| | - Luis Cisneros
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ 85281, USA
| | | | - Aziz Khan
- Department of Medicine, Genetics, and Biomedical Data Science Stanford School of Medicine, Stanford, CA 94305
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA 94305
| | - Christina Curtis
- Department of Medicine, Genetics, and Biomedical Data Science Stanford School of Medicine, Stanford, CA 94305
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA 94305
- Chan Zuckerberg Biohub, San Francisco, CA
| | - Marc D. Ryser
- Duke University School of Medicine, Durham, NC 27710, USA
| | - Joseph Y. Lo
- Duke University School of Medicine, Durham, NC 27710, USA
| | - Allison Hall
- Duke University School of Medicine, Durham, NC 27710, USA
| | | | | | - Carlo C. Maley
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ 85287, USA
| |
Collapse
|
37
|
Deng Y, Lu Y, Li X, Zhu Y, Zhao Y, Ruan Z, Mei N, Yin B, Liu L. Prediction of human epidermal growth factor receptor 2 (HER2) status in breast cancer by mammographic radiomics features and clinical characteristics: a multicenter study. Eur Radiol 2024; 34:5464-5476. [PMID: 38276982 DOI: 10.1007/s00330-024-10607-9] [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: 06/05/2023] [Revised: 12/11/2023] [Accepted: 12/23/2023] [Indexed: 01/27/2024]
Abstract
OBJECTIVES To preoperatively evaluate the human epidermal growth factor 2 (HER2) status in breast cancer using mammographic radiomics features and clinical characteristics on a multi-vendor and multi-center basis. METHODS This multi-center study included a cohort of 1512 Chinese female with invasive ductal carcinoma of no special type (IDC-NST) from two different hospitals and five devices (1332 from Institution A, used for training and testing the models, and 180 women from Institution B, as the external validation cohort). The Gradient Boosting Machine (GBM) was employed to establish radiomics and multiomics models. Model efficacy was evaluated by the area under the curve (AUC). RESULTS The number of HER2-positive patients in the training, testing, and external validation cohort were 245(26.3%), 105 (26.3.8%), and 51(28.3%), respectively, with no statistical differences among the three cohorts (p = 0.842, chi-square test). The radiomics model, based solely on the radiomics features, achieved an AUC of 0.814 (95% CI, 0.784-0.844) in the training cohort, 0.776 (95% CI, 0.727-0.825) in the testing cohort, and 0.702 (95% CI, 0.614-0.790) in the external validation cohort. The multiomics model, incorporated radiomics features with clinical characteristics, consistently outperformed the radiomics model with AUC values of 0.838 (95% CI, 0.810-0.866) in the training cohort, 0.788 (95% CI, 0.741-0.835) in the testing cohort, and 0.722 (95% CI, 0.637-0.811) in the external validation cohort. CONCLUSIONS Our study demonstrates that a model based on radiomics features and clinical characteristics has the potential to accurately predict HER2 status of breast cancer patients across multiple devices and centers. CLINICAL RELEVANCE STATEMENT By predicting the HER2 status of breast cancer reliably, the presented model built upon radiomics features and clinical characteristics on a multi-vendor and multi-center basis can help in bolstering the model's applicability and generalizability in real-world clinical scenarios. KEY POINTS • The mammographic presentation of breast cancer is closely associated with the status of human epidermal growth factor receptor 2 (HER2). • The radiomics model, based solely on radiomics features, exhibits sub-optimal performance in the external validation cohort. • By combining radiomics features and clinical characteristics, the multiomics model can improve the prediction ability in external data.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yiping Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xuanxuan Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yuqi Zhu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yajing Zhao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhuoying Ruan
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Nan Mei
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Li Liu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
38
|
McDaniel JM, Morrissey RL, Dibra D, Patel LR, Xiong S, Zhang Y, Chau GP, Su X, Qi Y, El-Naggar AK, Lozano G. p53R172H and p53R245W Hotspot Mutations Drive Distinct Transcriptomes in Mouse Mammary Tumors Through a Convergent Transcriptional Mediator. CANCER RESEARCH COMMUNICATIONS 2024; 4:1991-2007. [PMID: 38994678 PMCID: PMC11310746 DOI: 10.1158/2767-9764.crc-24-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/11/2024] [Accepted: 07/09/2024] [Indexed: 07/13/2024]
Abstract
Aggressive breast cancers harbor TP53 missense mutations. Tumor cells with TP53 missense mutations exhibit enhanced growth and survival through transcriptional rewiring. To delineate how TP53 mutations in breast cancer contribute to tumorigenesis and progression in vivo, we created a somatic mouse model driven by mammary epithelial cell-specific expression of Trp53 mutations. Mice developed primary mammary tumors reflecting the human molecular subtypes of luminal A, luminal B, HER2-enriched, and triple-negative breast cancer with metastases. Transcriptomic analyses comparing MaPR172H/- or MaPR245W/- mammary tumors to MaP-/- tumors revealed (1) differences in cancer-associated pathways activated in both p53 mutants and (2) Nr5a2 as a novel transcriptional mediator of distinct pathways in p53 mutants. Meta-analyses of human breast tumors corroborated these results. In vitro assays demonstrate mutant p53 upregulates specific target genes that are enriched for Nr5a2 response elements in their promoters. Co-immunoprecipitation studies revealed p53R172H and p53R245W interact with Nr5a2. These findings implicate NR5A2 as a novel mediator of mutant p53 transcriptional activity in breast cancer. SIGNIFICANCE Our findings implicate NR5A2 as a novel mediator of mutant p53 transcriptional activity in breast cancer. NR5A2 may be an important therapeutic target in hard-to-treat breast cancers such as endocrine-resistant tumors and metastatic triple-negative breast cancers harboring TP53 missense mutations.
Collapse
Affiliation(s)
- Joy M. McDaniel
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Rhiannon L. Morrissey
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas.
| | - Denada Dibra
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Lalit R. Patel
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Shunbin Xiong
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Yun Zhang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas.
| | - Gilda P. Chau
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Adel K. El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
39
|
Yang C, Liu H, Feng X, Shi H, Jiang Y, Li J, Tan J. Research hotspots and frontiers of neoadjuvant therapy in triple-negative breast cancer: a bibliometric analysis of publications between 2002 and 2023. Int J Surg 2024; 110:4976-4992. [PMID: 39143709 PMCID: PMC11326012 DOI: 10.1097/js9.0000000000001586] [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: 01/22/2024] [Accepted: 04/25/2024] [Indexed: 08/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer with poor prognosis, and neoadjuvant therapy (NAT) has emerged as an important component in managing advanced-stage patients by providing surgical opportunities and improving survival outcomes. A search of publications on NAT for TNBC from 2002 to 2023 was conducted through the Web of Science core collection. A comprehensive bibliometric analysis was conducted on the data using CiteSpace, VOSviewer, and Bibliometrix. The analysis revealed a continuous and steady growth in the number of articles published in this field over the past 20 years. The United States has made significant contributions to this field, with The University of Texas MD Anderson Cancer Center publishing the most articles. Loibl, S. from Germany was found to be the most published author with 54 articles. Analysis of the journals showed that the Journal of Clinical Oncology is the most cited journal. Combined with the keyword co-occurrence analysis and clustering analysis, current research topic focuses on treatment regimens and disease prognosis. Dual-map overlay of the journals indicates that the research trend is gradually shifting from molecular biology and genetics to immunology and clinical research. Combination therapy, including immunotherapy, may be the future direction for NAT treatment of TNBC. Overall, this study provides valuable insights into the current research status, latest advancements, and emerging development trend of NAT for TNBC.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Hui Liu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University
| | - Xing Feng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
- Department of Hepatobiliary, Breast and Thyroid Surgery, The People’s Hospital of Liangping District, Chongqing, China
| | - Han Shi
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Yuchan Jiang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Junfeng Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Jinxiang Tan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| |
Collapse
|
40
|
Golijanin D, Radovanović Z, Radovanović D, Đermanović A, Starčević S, Đermanović M. Molecular subtype and risk of local recurrence after nipple‑sparing mastectomy for breast cancer. Oncol Lett 2024; 28:389. [PMID: 38966584 PMCID: PMC11223028 DOI: 10.3892/ol.2024.14522] [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: 01/14/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
The present study aimed to investigate whether local recurrence (LR) after nipple-sparing mastectomy (NSM) and reconstruction was associated with i) Ki67 values and molecular subtypes of the initial lesions, and ii) the size of the initial tumor and the size of the implant. A total of 156 patients with breast cancer with a mean age of 51.58 years (age range, 26-75 years) who underwent NSM with primary implant breast reconstruction were analyzed. After surgery, the mean follow-up time was 59.26 months (range, 17-85 months). Molecular subtypes, Ki67 values, estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) status were recorded for each patient. Additionally, information regarding the size of the implant and the initial tumor size were collected. The information was used to assess LR. For univariate analyses of risk factors, χ2 test, Fisher's exact test, Mann-Whitney U test and Student's t-test for independent samples were used. For multivariate analyses, a Cox proportional-hazards model was used. NSM was the primary treatment for breast cancer in 34/156 patients (21.8%), while 122/156 (78.2%) of patients received neoadjuvant chemotherapy followed by surgery. Luminal B was the most frequent molecular subtype, detected in 82/156 patients (52.6%), whereas the luminal A subtype was detected in 37 patients (23.7%) and the HER2-enriched subtype was detected in 17/156 patients (10.9%). Ki67 expression was low in 13/156 patients (8.3%), while medium expression was detected in 78/156 patients (50.0%) and high expression was present in 58/156 patients (37.2%). LR was noted in 17/156 patients (10.9%). As determined by univariate analysis, lower ER (P=0.010) and PR (P=0.008) expression were indicated to be significant risk factors for LR. In conclusion, in the present patient cohort, low ER and PR expression were risk factors for LR of breast cancer, whereas Ki67 status and molecular subtype were not statistically significant risk factors for LR. Additionally, the size of the initial tumor and the size of the implant were not risk factors for LR. These findings are consistent with the current literature, and should be utilized when discussing treatment options and potential clinical outcomes with patients prior to surgical management.
Collapse
Affiliation(s)
- Danica Golijanin
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Republic of Serbia
- Clinic for Surgical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Republic of Serbia
| | - Zoran Radovanović
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Republic of Serbia
- Clinic for Surgical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Republic of Serbia
| | - Dragana Radovanović
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Republic of Serbia
- Department for Anesthesiology with Reanimatology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Republic of Serbia
| | - Aleksandar Đermanović
- Clinic for Surgical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Republic of Serbia
| | - Sanja Starčević
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Republic of Serbia
- Department for Anesthesiology with Reanimatology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Republic of Serbia
| | - Marija Đermanović
- Department for Neonatology, Institute for Child and Youth Health Care of Vojvodina, 21000 Novi Sad, Republic of Serbia
| |
Collapse
|
41
|
González-Woge M, Contreras-Espinosa L, García-Gordillo JA, Aguilar-Villanueva S, Bargallo-Rocha E, Cabrera-Galeana P, Vasquez-Mata T, Cervantes-López X, Vargas-Lías DS, Montiel-Manríquez R, Bautista-Hinojosa L, Rebollar-Vega R, Castro-Hernández C, Álvarez-Gómez RM, De La Rosa-Velázquez IA, Díaz-Chávez J, Jiménez-Trejo F, Arriaga-Canon C, Herrera LA. The Expression Profiles of lncRNAs Are Associated with Neoadjuvant Chemotherapy Resistance in Locally Advanced, Luminal B-Type Breast Cancer. Int J Mol Sci 2024; 25:8077. [PMID: 39125649 PMCID: PMC11311431 DOI: 10.3390/ijms25158077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/06/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
lncRNAs are noncoding transcripts with tissue and cancer specificity. Particularly, in breast cancer, lncRNAs exhibit subtype-specific expression; they are particularly upregulated in luminal tumors. However, no gene signature-based laboratory tests have been developed for luminal breast cancer identification or the differential diagnosis of luminal tumors, since no luminal A- or B-specific genes have been identified. Particularly, luminal B patients are of clinical interest, since they have the most variable response to neoadjuvant treatment; thus, it is necessary to develop diagnostic and predictive biomarkers for these patients to optimize treatment decision-making and improve treatment quality. In this study, we analyzed the lncRNA expression profiles of breast cancer cell lines and patient tumor samples from RNA-Seq data to identify an lncRNA signature specific for luminal phenotypes. We identified an lncRNA signature consisting of LINC01016, GATA3-AS1, MAPT-IT1, and DSCAM-AS1 that exhibits luminal subtype-specific expression; among these lncRNAs, GATA3-AS1 is associated with the presence of residual disease (Wilcoxon test, p < 0.05), which is related to neoadjuvant chemotherapy resistance in luminal B breast cancer patients. Furthermore, analysis of GATA3-AS1 expression using RNA in situ hybridization (RNA ISH) demonstrated that this lncRNA is detectable in histological slides. Similar to estrogen receptors and Ki67, both commonly detected biomarkers, GATA3-AS1 proves to be a suitable predictive biomarker for clinical application in breast cancer laboratory tests.
Collapse
Affiliation(s)
- Miguel González-Woge
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
| | - Laura Contreras-Espinosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C. P. 04510, Mexico;
| | - José Antonio García-Gordillo
- Departamento de Oncología Médica de Mama, Instituto Nacional de Cancerología, Tlalpan, Mexico City C. P. 14080, Mexico; (J.A.G.-G.); (P.C.-G.)
| | - Sergio Aguilar-Villanueva
- Departamento de Tumores Mamarios, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (S.A.-V.); (E.B.-R.); (D.S.V.-L.)
| | - Enrique Bargallo-Rocha
- Departamento de Tumores Mamarios, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (S.A.-V.); (E.B.-R.); (D.S.V.-L.)
| | - Paula Cabrera-Galeana
- Departamento de Oncología Médica de Mama, Instituto Nacional de Cancerología, Tlalpan, Mexico City C. P. 14080, Mexico; (J.A.G.-G.); (P.C.-G.)
| | - Tania Vasquez-Mata
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
| | - Ximena Cervantes-López
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
| | - Diana Sofía Vargas-Lías
- Departamento de Tumores Mamarios, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (S.A.-V.); (E.B.-R.); (D.S.V.-L.)
| | - Rogelio Montiel-Manríquez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
| | - Luis Bautista-Hinojosa
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C. P. 04510, Mexico;
| | - Rosa Rebollar-Vega
- Genomics Laboratory, Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, Tlalpan, Mexico City C. P. 14080, Mexico;
| | - Clementina Castro-Hernández
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
| | - Rosa María Álvarez-Gómez
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico;
| | | | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey C. P. 64710, Mexico
| | - Francisco Jiménez-Trejo
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C, Coyoacán, Mexico City C. P. 04530, Mexico;
| | - Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey C. P. 64710, Mexico
| | - Luis Alonso Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C. P. 14080, Mexico; (M.G.-W.); (L.C.-E.); (T.V.-M.); (X.C.-L.); (R.M.-M.); (C.C.-H.); (J.D.-C.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey C. P. 64710, Mexico
| |
Collapse
|
42
|
Varghese S, Jisha M, Rajeshkumar K, Gajbhiye V, Alrefaei AF, Jeewon R. Endophytic fungi: A future prospect for breast cancer therapeutics and drug development. Heliyon 2024; 10:e33995. [PMID: 39091955 PMCID: PMC11292557 DOI: 10.1016/j.heliyon.2024.e33995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Globally, breast cancer is a primary contributor to cancer-related fatalities and illnesses among women. Consequently, there is a pressing need for safe and effective treatments for breast cancer. Bioactive compounds from endophytic fungi that live in symbiosis with medicinal plants have garnered significant interest in pharmaceutical research due to their extensive chemical composition and prospective medicinal attributes. This review underscores the potentiality of fungal endophytes as a promising resource for the development of innovative anticancer agents specifically tailored for breast cancer therapy. The diversity of endophytic fungi residing in medicinal plants, success stories of key endophytic bioactive metabolites tested against breast cancer and the current progress with regards to in vivo studies and clinical trials on endophytic fungal metabolites in breast cancer research forms the underlying theme of this article. A thorough compilation of putative anticancer compounds sourced from endophytic fungi that have demonstrated therapeutic potential against breast cancer, spanning the period from 1990 to 2022, has been presented. This review article also outlines the latest trends in endophyte-based drug discovery, including the use of artificial intelligence, machine learning, multi-omics approaches, and high-throughput strategies. The challenges and future prospects associated with fungal endophytes as substitutive sources for developing anticancer drugs targeting breast cancer are also being highlighted.
Collapse
Affiliation(s)
- Sherin Varghese
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India
| | - M.S. Jisha
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India
| | - K.C. Rajeshkumar
- National Fungal Culture Collection of India (NFCCI), Biodiversity and Palaeobiology (Fungi) Gr., Agharkar Research Institute, G.G. Agharkar Road, Pune, 411 004, Maharashtra, India
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, G.G. Agharkar Road, Pune, 411 004, Maharashtra, India
| | - Abdulwahed Fahad Alrefaei
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Rajesh Jeewon
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Reduit, Mauritius
| |
Collapse
|
43
|
Van Alsten SC, Vohra SN, Ivory JM, Hamilton AM, Gao X, Kirk EL, Butler EN, Earp HS, Reeder-Hayes KE, Hoadley KA, Carey LA, Troester MA. Differences in 21-Gene and PAM50 Recurrence Scores in Younger and Black Women With Breast Cancer. JCO Precis Oncol 2024; 8:e2400137. [PMID: 39013134 PMCID: PMC11555617 DOI: 10.1200/po.24.00137] [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: 03/05/2024] [Revised: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
PURPOSE Genomic tests, such as the Oncotype Dx 21-gene and Prosigna risk of recurrence (ROR-P) assay, are commonly used for breast cancer prognostication. Emerging data suggest variability between assays, but this has not been compared in diverse populations. MATERIALS AND METHODS RNA sequencing was performed on 647 previously untreated stage I-III estrogen receptor-positive/human epidermal growth factor receptor 2-negative tumors in the Carolina Breast Cancer Study, which oversampled Black and younger women (age <50 years at diagnosis), using research versions of two common RNA-based prognostic assays: ROR-PR and the 21-gene recurrence score (RSR). Relative frequency differences and 95% CIs were estimated for associations with race and age, and hazards of 5-year local or distant recurrence were modeled with Cox regression. Proliferation and estrogen module scores from each assay, representing broad activity of genes in those pathways, were examined to guide interpretation of differences between tests. RESULTS Among both younger and older individuals, Black women had higher frequency of intermediate and high ROR-PR scores than non-Black women. Race was not significantly associated with RSR in either age group. High (hazard ratio [HR], 4.67 [95% CI, 1.73 to 12.70]) and intermediate (HR, 2.12 [95% CI, 0.98 to 4.62]) ROR-PR scores were associated with greater risk of recurrence, but RSR did not predict recurrence. RSR emphasized estrogen over proliferation modules, whereas ROR-PR emphasized proliferation. Higher proliferation scores were associated with younger age and Black race in both assays. Modifications to the RSR algorithm that increased emphasis on proliferation improved prognostication in this diverse population. CONCLUSION ROR-PR and the 21-gene RSR differentially emphasize estrogen-related and proliferative biology. The emphasis of 21-gene RS on estrogen-related biology and lower endocrine therapy initiation among Black women may contribute to poorer prognostic ability in heterogeneously treated populations.
Collapse
Affiliation(s)
- Sarah C. Van Alsten
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sanah N. Vohra
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joannie M. Ivory
- Division of Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Alina M. Hamilton
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Xiaohua Gao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Erin L. Kirk
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Eboneé N. Butler
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Katherine E. Reeder-Hayes
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Lisa A. Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Melissa A. Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
44
|
Abbas MA, Al-Kabariti AY, Sutton C. Comprehensive understanding of the role of GPER in estrogen receptor-alpha negative breast cancer. J Steroid Biochem Mol Biol 2024; 241:106523. [PMID: 38636681 DOI: 10.1016/j.jsbmb.2024.106523] [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: 09/03/2023] [Revised: 11/01/2023] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
G protein-coupled estrogen receptor (GPER) plays a prominent role in facilitating the rapid, non-genomic signaling of estrogens in breast cancer cells. Herein, a comprehensive overview of the role of GPER in ER-ɑ-negative breast cancer is provided. Activation of GPER affected proliferation, metastasis and epithelial mesenchymal transition in ER-ɑ negative breast cancer cells. Clinical studies have demonstrated that GPER positivity was strongly correlated with larger tumor size and advanced clinical stage, suggesting that GPER/ERK signaling may play a role in promoting tumor progression. Strong evidence existed that environmental contaminants like bisphenol A have a carcinogenic potential mediated by GPER activation. The complexity of the cross talk between GPER and other receptors including ER-β, ER-α36, Estrogen-related receptor α (ERRα) and androgen receptor has been discussed. The potential utility of small molecules and phytoestrogens targeting GPER, adds valuable insights into its therapeutic potential. This review holds promises in advancing our understanding of GPER role in ER-ɑ-negative breast cancer. Overall, the consequences of GPER activation are still an area of active research and the implication are not entirely clear.
Collapse
Affiliation(s)
- Manal A Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Chris Sutton
- School of Chemistry and Biosciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
45
|
Sandoval-Ato R, Coral-Gonzales P, Coronel-Arias S, Espinoza-Mantilla L, Terrones-Chaparro G, Serna-Alarcón V. Clinical and radiological manifestations associated with triple-negative breast cancer in women from northern Peru. A case-control study. Ecancermedicalscience 2024; 18:1720. [PMID: 39021537 PMCID: PMC11254400 DOI: 10.3332/ecancer.2024.1720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Indexed: 07/20/2024] Open
Abstract
Objective Triple-negative breast cancer (TNBC) has an aggressive clinical behaviour, with advanced stages at initial diagnostic evaluation, early recurrences and poor survival, so the purpose was to determine the clinical and radiological manifestations associated with TNBC. Materials and methods A case-control study in women diagnosed with breast cancer from January 2015 to August 2022 at the 'Instituto Regional de Enfermedades Neoplásicas del Norte'. We classified cases (Triple Negative subtype) and controls (Luminal A, Luminal B and HER2) according to immunohistochemistry ical analysis. Bivariate and multivariate logistic regression models were used to calculate the odds ratio (OR) with their respective 95% confidence intervals (CIs). Results The medical reports of 88 cases and 236 controls were reviewed. Cases were more likely to report pain (p = 0.001), nodules on ultrasound (p = 0.01) and mammography (p = 0.003), superior median size (p < 0.05), posterior enhancement (p = 0.001) and moderate density (p = 0.003). Multivariate analysis identified that TNBC was more likely to have a nodular type lesion by ultrasound (OR: 9.73, 95% CI: 1.10-86.16; p = 0.04), ultrasound lesion larger than 36 mm (OR: 4.99, 95% CI: 1.75-14.17; p = 0.003) and moderate density (OR: 3.83, 95% CI: 1.44-10.14; p = 0.007). Conclusion There are particular clinical and imaging manifestations of TNBC, showing that radiological lesions that presented characteristics in ultrasound as nodular type lesions larger than 36 mm and in mammography moderate grade density, were associated with this subtype of breast tumours in a Peruvian population.
Collapse
Affiliation(s)
- Raúl Sandoval-Ato
- Escuela de Posgrado, Facultad de Medicina, Universidad Privada Antenor Orrego, Trujillo 13008, Perú
- Unidad de Investigación Clínica, Scientia Clinical and Epidemiological Research Institute, Trujillo 13001, Perú
- https://orcid.org/0000-0001-8666-7188
| | - Patricia Coral-Gonzales
- Servicio de Radiodiagnóstico, Instituto Regional de Enfermedades Neoplásicas Norte, Trujillo 13008, Perú
- Escuela Profesional de Medicina, Facultad de Medicina, Universidad Privada Antenor Orrego, Trujillo 13008, Perú
- https://orcid.org/0000-0002-8734-4687
| | - Sebastian Coronel-Arias
- Servicio de Radiodiagnóstico, Instituto Regional de Enfermedades Neoplásicas Norte, Trujillo 13008, Perú
- https://orcid.org/0000-0002-2607-7191
| | - Luisa Espinoza-Mantilla
- Servicio de Radiodiagnóstico, Instituto Regional de Enfermedades Neoplásicas Norte, Trujillo 13008, Perú
- https://orcid.org/0000-0002-5465-7775
| | - Grace Terrones-Chaparro
- Servicio de Radiodiagnóstico, Instituto Regional de Enfermedades Neoplásicas Norte, Trujillo 13008, Perú
- https://orcid.org/0000-0001-6938-1401
| | - Victor Serna-Alarcón
- Escuela Profesional de Medicina, Facultad de Medicina, Universidad Privada Antenor Orrego, Trujillo 13008, Perú
- https://orcid.org/0000-0002-9803-6217
| |
Collapse
|
46
|
Rujchanarong D, Spruill L, Sandusky GE, Park Y, Mehta AS, Drake RR, Ford ME, Nakshatri H, Angel PM. Spatial N-glycomics of the normal breast microenvironment reveals fucosylated and high-mannose N-glycan signatures related to BI-RADS density and ancestry. Glycobiology 2024; 34:cwae043. [PMID: 38869882 PMCID: PMC11193881 DOI: 10.1093/glycob/cwae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/25/2024] [Accepted: 06/12/2024] [Indexed: 06/14/2024] Open
Abstract
Higher breast cancer mortality rates continue to disproportionally affect black women (BW) compared to white women (WW). This disparity is largely due to differences in tumor aggressiveness that can be related to distinct ancestry-associated breast tumor microenvironments (TMEs). Yet, characterization of the normal microenvironment (NME) in breast tissue and how they associate with breast cancer risk factors remains unknown. N-glycans, a glucose metabolism-linked post-translational modification, has not been characterized in normal breast tissue. We hypothesized that normal female breast tissue with distinct Breast Imaging and Reporting Data Systems (BI-RADS) categories have unique microenvironments based on N-glycan signatures that varies with genetic ancestries. Profiles of N-glycans were characterized in normal breast tissue from BW (n = 20) and WW (n = 20) at risk for breast cancer using matrix assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI). A total of 176 N-glycans (32 core-fucosylated and 144 noncore-fucosylated) were identified in the NME. We found that certain core-fucosylated, outer-arm fucosylated and high-mannose N-glycan structures had specific intensity patterns and histological distributions in the breast NME dependent on BI-RADS densities and ancestry. Normal breast tissue from BW, and not WW, with heterogeneously dense breast densities followed high-mannose patterns as seen in invasive ductal and lobular carcinomas. Lastly, lifestyles factors (e.g. age, menopausal status, Gail score, BMI, BI-RADS) differentially associated with fucosylated and high-mannose N-glycans based on ancestry. This study aims to decipher the molecular signatures in the breast NME from distinct ancestries towards improving the overall disparities in breast cancer burden.
Collapse
Affiliation(s)
- Denys Rujchanarong
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29425, United States
| | - Laura Spruill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 96 Jonathan Lucas St. Ste. 601, MSC 617, Charleston, SC 29425, United States
| | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, 340 West 10th Street Fairbanks Hall, Suite 6200 Indianapolis, IN 46202-3082, United States
| | - Yeonhee Park
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Warf Office Bldg, 610 Walnut St Room 201, Madison, WI 53726, United States
| | - Anand S Mehta
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29425, United States
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29425, United States
| | - Marvella E Ford
- Department of Public Health Sciences, Medical University of South Carolina, 35 Cannon Street, Charleston, SC 29425, United States
| | - Harikrishna Nakshatri
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Dr, Indianapolis, IN 46202, United States
- Department of Surgery, Indiana University School of Medicine, 545 Barnhill Dr, Indianapolis, IN 46202, United States
| | - Peggi M Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29425, United States
| |
Collapse
|
47
|
Li Y, Van Alsten SC, Lee DN, Kim T, Calhoun BC, Perou CM, Wobker SE, Marron JS, Hoadley KA, Troester MA. Visual Intratumor Heterogeneity and Breast Tumor Progression. Cancers (Basel) 2024; 16:2294. [PMID: 39001357 PMCID: PMC11240824 DOI: 10.3390/cancers16132294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
High intratumoral heterogeneity is thought to be a poor prognostic indicator. However, the source of heterogeneity may also be important, as genomic heterogeneity is not always reflected in histologic or 'visual' heterogeneity. We aimed to develop a predictor of histologic heterogeneity and evaluate its association with outcomes and molecular heterogeneity. We used VGG16 to train an image classifier to identify unique, patient-specific visual features in 1655 breast tumors (5907 core images) from the Carolina Breast Cancer Study (CBCS). Extracted features for images, as well as the epithelial and stromal image components, were hierarchically clustered, and visual heterogeneity was defined as a greater distance between images from the same patient. We assessed the association between visual heterogeneity, clinical features, and DNA-based molecular heterogeneity using generalized linear models, and we used Cox models to estimate the association between visual heterogeneity and tumor recurrence. Basal-like and ER-negative tumors were more likely to have low visual heterogeneity, as were the tumors from younger and Black women. Less heterogeneous tumors had a higher risk of recurrence (hazard ratio = 1.62, 95% confidence interval = 1.22-2.16), and were more likely to come from patients whose tumors were comprised of only one subclone or had a TP53 mutation. Associations were similar regardless of whether the image was based on stroma, epithelium, or both. Histologic heterogeneity adds complementary information to commonly used molecular indicators, with low heterogeneity predicting worse outcomes. Future work integrating multiple sources of heterogeneity may provide a more comprehensive understanding of tumor progression.
Collapse
Affiliation(s)
- Yao Li
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.L.); (T.K.); (J.S.M.)
| | - Sarah C. Van Alsten
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Dong Neuck Lee
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Taebin Kim
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.L.); (T.K.); (J.S.M.)
| | - Benjamin C. Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (B.C.C.); (C.M.P.); (S.E.W.)
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Charles M. Perou
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (B.C.C.); (C.M.P.); (S.E.W.)
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sara E. Wobker
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (B.C.C.); (C.M.P.); (S.E.W.)
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - J. S. Marron
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.L.); (T.K.); (J.S.M.)
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Katherine A. Hoadley
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Melissa A. Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (B.C.C.); (C.M.P.); (S.E.W.)
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
48
|
Aquino-Acevedo AN, Orengo-Orengo JA, Cruz-Robles ME, Saavedra HI. Mitotic kinases are emerging therapeutic targets against metastatic breast cancer. Cell Div 2024; 19:21. [PMID: 38886738 PMCID: PMC11184769 DOI: 10.1186/s13008-024-00125-x] [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: 12/21/2023] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
This review aims to outline mitotic kinase inhibitors' roles as potential therapeutic targets and assess their suitability as a stand-alone clinical therapy or in combination with standard treatments for advanced-stage solid tumors, including triple-negative breast cancer (TNBC). Breast cancer poses a significant global health risk, with TNBC standing out as the most aggressive subtype. Comprehending the role of mitosis is crucial for understanding how TNBC advances from a solid tumor to metastasis. Chemotherapy is the primary treatment used to treat TNBC. Some types of chemotherapeutic agents target cells in mitosis, thus highlighting the need to comprehend the molecular mechanisms governing mitosis in cancer. This understanding is essential for devising targeted therapies to disrupt these mitotic processes, prevent or treat metastasis, and improve patient outcomes. Mitotic kinases like Aurora kinase A, Aurora Kinase B, never in mitosis gene A-related kinase 2, Threonine-Tyrosine kinase, and Polo-kinase 1 significantly impact cell cycle progression by contributing to chromosome separation and centrosome homeostasis. When these kinases go awry, they can trigger chromosome instability, increase cell proliferation, and activate different molecular pathways that culminate in a transition from epithelial to mesenchymal cells. Ongoing clinical trials investigate various mitotic kinase inhibitors as potential biological treatments against advanced solid tumors. While clinical trials against mitotic kinases have shown some promise in the clinic, more investigation is necessary, since they induce severe adverse effects, particularly affecting the hematopoietic system.
Collapse
Affiliation(s)
- Alexandra N Aquino-Acevedo
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA
| | - Joel A Orengo-Orengo
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA
| | - Melanie E Cruz-Robles
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA
| | - Harold I Saavedra
- Department of Basic Sciences, Ponce Health Sciences University-Ponce Research Institute, 388 Luis Salas Zona Industrial Reparada 2, P.O. Box 7004, Ponce, Puerto Rico, 00716-2347, USA.
| |
Collapse
|
49
|
Yanguela J, Jackson BE, Reeder-Hayes KE, Roberson ML, Rocque GB, Kuo TM, LeBlanc MR, Baggett CD, Green L, Laurie-Zehr E, Wheeler SB. Simulating the population impact of interventions to reduce racial gaps in breast cancer treatment. J Natl Cancer Inst 2024; 116:902-910. [PMID: 38281076 PMCID: PMC11160503 DOI: 10.1093/jnci/djae019] [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: 08/28/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Inequities in guideline-concordant treatment receipt contribute to worse survival in Black patients with breast cancer. Inequity-reduction interventions (eg, navigation, bias training, tracking dashboards) can close such treatment gaps. We simulated the population-level impact of statewide implementation of inequity-reduction interventions on racial breast cancer inequities in North Carolina. METHODS Using registry-linked multipayer claims data, we calculated inequities between Black and White patients receiving endocrine therapy (n = 12 033) and chemotherapy (n = 1819). We then built cohort-stratified (endocrine therapy and chemotherapy) and race-stratified Markov models to simulate the potential increase in the proportion of patients receiving endocrine therapy or chemotherapy and subsequent improvements in breast cancer outcomes if inequity-reducing intervention were implemented statewide. We report uncertainty bounds representing 95% of simulation results. RESULTS In total, 75.6% and 72.1% of Black patients received endocrine therapy and chemotherapy, respectively, over the 2006-2015 and 2004-2015 periods (vs 79.3% and 78.9% of White patients, respectively). Inequity-reduction interventions could increase endocrine therapy and chemotherapy receipt among Black patients to 89.9% (85.3%, 94.6%) and 85.7% (80.7%, 90.9%). Such interventions could also decrease 5-year and 10-year breast cancer mortality gaps from 3.4 to 3.2 (3.0, 3.3) and from 6.7 to 6.1 (5.9, 6.4) percentage points in the endocrine therapy cohorts and from 8.6 to 8.1 (7.7, 8.4) and from 8.2 to 7.8 (7.3, 8.1) percentage points in the chemotherapy cohorts. CONCLUSIONS Inequity-focused interventions could improve cancer outcomes for Black patients, but they would not fully close the racial breast cancer mortality gap. Addressing other inequities along the cancer continuum (eg, screening, pre- and postdiagnosis risk factors) is required to achieve full equity in breast cancer outcomes.
Collapse
Affiliation(s)
- Juan Yanguela
- Department of Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Bradford E Jackson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine E Reeder-Hayes
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mya L Roberson
- Department of Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle B Rocque
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tzy-Mey Kuo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew R LeBlanc
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher D Baggett
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Laura Green
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Erin Laurie-Zehr
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie B Wheeler
- Department of Health Policy and Management, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
50
|
Garlapati C, Joshi S, Yang C, Chandrashekar DS, Rida P, Aneja R. A novel role for KIFC1-MYH9 interaction in triple-negative breast cancer aggressiveness and racial disparity. Cell Commun Signal 2024; 22:312. [PMID: 38902769 PMCID: PMC11188183 DOI: 10.1186/s12964-024-01664-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
African American (AA) women are twice as likely to develop triple-negative breast cancer (TNBC) as women of European descent. Additionally, AA women with TNBC present a much more aggressive disease course than their European American (EA) counterparts. Thus, there is an unmet clinical need to identify race-specific biomarkers and improve survival outcomes in AA patients with TNBC. The minus-end directed microtubule motor protein kinesin family member C1 (KIFC1) promotes centrosome clustering and chromosomal instability and is often overexpressed in TNBC. Previous findings suggest that KIFC1 plays a role in cell proliferation and migration in TNBC cells from AAs and that the levels of nuclear KIFC1 (nKIFC1) are particularly high in AA patients with TNBC. The nuclear localization of KIFC1 in interphase may underlie its previously unrecognized race-specific association. In this study, we found that in TNBC cells derived from AAs, nKIFC1 interacted with the tumor suppressor myosin heavy chain 9 (MYH9) over EA cells. Treatment of AA TNBC cells with commercial inhibitors of KIFC1 and MYH9 disrupted the interaction between KIFC1 and MYH9. To characterize the racial differences in the KIFC1-MYH9-MYC axis in TNBC, we established homozygous KIFC1 knockout (KO) TNBC cell lines. KIFC1 KO significantly inhibited proliferation, migration, and invasion in AA TNBC cells but not in EA TNBC cells. RNA sequencing analysis showed significant downregulation of genes involved in cell migration, invasion, and metastasis upon KIFC1 KO in TNBC cell lines from AAs compared to those from EAs. These data indicate that mechanistically, the role of nKIFC1 in driving TNBC progression and metastasis is stronger in AA patients than in EA patients, and that KIFC1 may be a critical therapeutic target for AA patients with TNBC.
Collapse
Affiliation(s)
- Chakravarthy Garlapati
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Alkermes Inc, Waltham, MA, 02451, USA
| | - Shriya Joshi
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Alkermes Inc, Waltham, MA, 02451, USA
- Small molecule drug discovery, Bristol Myers Squibb, Cambridge, MA, 02141, USA
| | - Chunhua Yang
- Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | | | | | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|