1
|
Voutsadakis IA. Breast Cancers With Intermediate Estrogen Receptor Expression: Characteristics, Prognosis and Treatment. Clin Breast Cancer 2025; 25:214-222. [PMID: 39710525 DOI: 10.1016/j.clbc.2024.11.020] [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: 06/21/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
In the era of personalized oncology biomarkers that identify subgroups of specific cancers and help predict response to specific therapies are critical tools for prognosis determination and therapeutic decisions. The Estrogen Receptor (ER) had been one of the first biomarkers used in breast cancer and has helped advance the field of breast oncology by contributing to the success of hormonal therapies for the ER positive subgroup of the disease. Expression of the receptor in 1% or more of tumor cells in immunohistochemical sections define currently the ER positive subgroup of breast cancers, which may be treated with regimens that include hormonal inhibitors. The highest sensitivity and benefit of hormonal therapies is observed in cancers with robust ER expression (in 90% to 100% of tumor cells). However, it has become clear that the subgroup of breast cancers with low ER expression (in 1% to 10% of tumor cells) behaves similarly to ER negative breast cancers and has an inferior response to hormonal therapies. The behavior of the rest of ER positive breast cancers with an intermediate ER expression between these 2 extremes (ER expression between 10% and 90%) is less well described and their response to estrogen targeting therapies is less clear. Breast cancers with intermediate ER expression represent a small subgroup of ER positive breast cancers and the wide range of expressions suggests heterogeneity. This review will discuss this subgroup of ER positive breast cancers and examine their genomic landscape and therapeutic repercussions.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste Marie, Ontario, Canada; Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, Ontario, Canada.
| |
Collapse
|
2
|
Jayachandran P, Deshmukh SK, Wu S, Ribeiro JR, Kang I, Xiu J, Farrell A, Battaglin F, Spicer DV, Soni S, Zhang W, Ashouri K, Millstein J, Ma CX, Graff SL, Radovich M, Sledge GW, Lenz HJ, Roussos Torres ET. Association of Androgen Receptor Expression With Tumor Immune Landscape and Treatment Outcomes of Patients With Breast Cancer. JCO Precis Oncol 2025; 9:e2400459. [PMID: 40294352 DOI: 10.1200/po-24-00459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
PURPOSE Although estrogen receptor is well studied in breast cancer (BC), the role of androgen receptor (AR) in prognosis and therapy response is less understood. Here, we characterized the clinicopathologic and molecular features of AR gene expression in BC subtypes. METHODS Ten thousand seven hundred twenty-eight BC samples were tested by next-generation DNA sequencing, whole-transcriptome sequencing, and immunohistochemistry at Caris Life Sciences (Phoenix, AZ). Tumors with AR-high and AR-low RNA expression were stratified by top and bottom quartiles, respectively. Treatment-associated survival was obtained from insurance claims and calculated from treatment start to last contact using Kaplan-Meier estimates. Statistical significance was determined by chi-square and Mann-Whitney U test with P values adjusted for multiple comparisons (q < .05). RESULTS AR-low was associated with basal-like tumors. AR-high tumors were associated with increased mutation rates in several genes-namely PIK3CA and CDH1-across all subtypes, while other associations such as RB1 and MAP3K1 were subtype-dependent. The immune landscape was differentially affected by AR expression in each subtype, but these differences did not correspond to differential responses to immune checkpoint blockade. Patients with AR-high tumors had a longer therapy response for most subtypes, but those with AR-high tumors that were human epidermal growth factor receptor 2-enriched and luminal B trended toward worse chemotherapy or hormone therapy response, respectively. CONCLUSION Our data suggest a unique molecular profile of AR-high BC that is subtype-specific and generally associated with improved outcomes. Exploration of specific mutations and immune-oncology markers associated with AR-high may aid in molecularly selected clinical trial design for patients with advanced BC.
Collapse
Affiliation(s)
- Priya Jayachandran
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | | | | | - Irene Kang
- Department of Medical Oncology & Therapeutics Research, City of Hope Orange County, Lennar Foundation Cancer Center, Irvine, CA
| | | | | | - Francesca Battaglin
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Darcy V Spicer
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Shivani Soni
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Wu Zhang
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Karam Ashouri
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Cynthia X Ma
- Division of Oncology, Washington University, St Louis, MO
| | - Stephanie L Graff
- Legorreta Cancer Center, Brown University, Providence, RI
- Lifespan Cancer Institute, Providence, RI
| | | | | | - Heinz-Josef Lenz
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Evanthia T Roussos Torres
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
3
|
Hu Y, Bian J, Chen W, Shi J, Wei X, Du Y, Zhang W. Androgen receptor-induced lncRNA SOX2-OT promotes triple-negative breast cancer tumorigenesis via targeting miR-320a-5p-CCR5 axis. J Biol Chem 2025; 301:108428. [PMID: 40118451 PMCID: PMC12017981 DOI: 10.1016/j.jbc.2025.108428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/13/2025] [Accepted: 03/13/2025] [Indexed: 03/23/2025] Open
Abstract
Our previous study showed that androgen receptor (AR) promotes triple-negative breast cancer (TNBC) cell tumorigenesis, but the underlying mechanisms remain unclear. Herein, using microarray analysis of long noncoding RNA expression profiles, we identified an AR-related long noncoding RNA SOX2-OT in TNBC. We found that AR could promote TNBC tumorigenesis by acting as a transcription factor to activate the expression of SOX2-OT. Mechanistic analysis demonstrated that SOX2-OT serves as a molecular sponge for miR-320a-5p to regulate the expression of CCR5. In addition, SOX2-OT promotes TNBC cell proliferation and inhibits apoptosis in an miR-320a-5p-dependent manner. Using a xenograft mouse model, we found that SOX2-OT-CCR5 axis could promote TNBC tumorigenesis in vivo. Importantly, the AR-SOX2-OT-miR-320a-5p-CCR5 axis is manifested in the tissues of 165 TNBC patients. Collectively, our results suggest that SOX2-OT can regulate AR-induced TNBC tumorigenesis through the miR-320a-5p-CCR5 signaling axis and reveal the great potential of targeting SOX2-OT for the treatment of TNBC patients.
Collapse
Affiliation(s)
- Yixuan Hu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Bian
- Department of Medical Oncology of PLA Cancer Center, Jinling Hospital, Nanjing, China
| | - Weiwei Chen
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junfeng Shi
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaowei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yueyao Du
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Ingle JN, Suman VJ, Solanki MH, Passow MR, Campbell JD, Wang L, Goetz MP. Evaluation of the androgen receptor in patients with ERα-positive early breast cancer treated with adjuvant tamoxifen ± fluoxymesterone. Breast Cancer Res 2025; 27:40. [PMID: 40108635 PMCID: PMC11924783 DOI: 10.1186/s13058-025-01992-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Our goal was to evaluate the impact of level of androgen receptor (AR) expression on outcomes in women with estrogen receptor α (ER) positive breast cancer. We sought to corroborate our preclinical findings that AR-agonists were efficacious in patients with ER-positive tumors that also expressed high levels of AR. METHODS Tissue microarrays (TMAs) were prepared from primary tumor blocks from patients entered on a prospective randomized adjuvant trial of tamoxifen (Tam) alone or combined with fluoxymesterone (Flu), an AR-agonist, (NCCTG 89-30-52). TMAs were stained for ER and AR and expression examined in decile increments (0-100%) of positive invasive tumor nuclei. The primary endpoint was relapse-free survival (RFS). RESULTS 301 (59%) of the 514 patients had sufficient tissue to determine ER and AR expression, where nuclear staining of > 70% was considered "enriched" and nuclear staining of ≤ 70% was considered "poor/moderate". Eleven (4%) of these patients had poor/moderate ER staining and were excluded from these analyses. The proportion of the ER-enriched tumors that also had AR-enriched expression levels was 56.3% in the Tam arm and 51.8% in the Tam + Flu arm. Within the AR-enriched patients, the cumulative incidence of RFS events showed an advantage for Tam + Flu over Tam alone that reached significance (Gray's test p = 0.0472). CONCLUSIONS Our findings suggest that an AR-agonist may be of value in AR-enriched, ER-enriched breast cancers and should be studied in future trials because of the availability of new, more tolerable AR-agonists.
Collapse
Affiliation(s)
- James N Ingle
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| | - Vera J Suman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Malvika H Solanki
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Marie R Passow
- Cytopathology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Jordan D Campbell
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Matthew P Goetz
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
5
|
Spurnić I, Šušnjar S, Jovanić I, Medić-Miljić N, Milovanović Z, Popović Krneta M, Bukumirić Z, Gavrilović D, Rajšić S, Marković I. The Diagnostics of Disease Relapse Within Five-Year Follow-Up and the Role of Androgen Receptor Expression in Patients with Early Triple-Negative Breast Cancer. Diagnostics (Basel) 2025; 15:692. [PMID: 40150035 PMCID: PMC11941219 DOI: 10.3390/diagnostics15060692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Triple-negative breast cancer (TNBC) is characterized by the absence of the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2. As there are no specific targeted therapies, TNBC patients often face an aggressive clinical course. The expression of the androgen receptor (AR) has been found in up to 30% of TNBC cases, but the association between the AR status and survival rates in TNBC remains controversial. The aim of this study was to explore the association of AR expression with the disease outcome in patients with early TNBC within a 5-year follow-up. Methods: AR expression was determined by immunohistochemistry in a cohort of 124 early-TNBC patients treated at the Institute for Oncology and Radiology of Serbia. The cut-off value used for the positive AR status was >10% tumor cells. The association of the AR status with clinicopathological factors (age, stage, tumor diameter, lymph node invasion, metastatic spread, Ki-67 score, EGFR score, and cytokeratin 5/6 score) and the disease outcome (disease-free survival-DFS-and overall survival-OS) was investigated. Results: Our analysis showed that the AR-positive status was associated with a significantly lower Ki-67 score compared to the AR-negative samples. A univariate analysis indicated that the age, tumor size, nodal status, and EGFR score significantly influenced both 5-year DFS and OS. Multivariate Cox analysis suggested that a smaller tumor size, lower nodal status, and AR expression were independent predictors of longer survival rates in TNBC patients. Conclusions: The results of this study suggest that the positive AR status may be a favorable prognostic factor in TNBC patients within the first five years after surgery.
Collapse
Affiliation(s)
- Igor Spurnić
- Surgical Oncology Clinic, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Snežana Šušnjar
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
| | - Irena Jovanić
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.J.); (N.M.-M.); (Z.M.)
| | - Nataša Medić-Miljić
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.J.); (N.M.-M.); (Z.M.)
| | - Zorka Milovanović
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.J.); (N.M.-M.); (Z.M.)
| | - Marina Popović Krneta
- Department of Nuclear Medicine, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia;
| | - Zoran Bukumirić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Institute of Medical Statistics and Informatics, 11000 Belgrade, Serbia
| | - Dušica Gavrilović
- Data Center, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia;
| | - Saša Rajšić
- Department of Anesthesiology and Intensive Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Ivan Marković
- Surgical Oncology Clinic, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
6
|
Turova P, Kushnarev V, Baranov O, Butusova A, Menshikova S, Yong ST, Nadiryan A, Antysheva Z, Khorkova S, Guryleva MV, Bagaev A, Lennerz JK, Chernyshov K, Kotlov N. The Breast Cancer Classifier refines molecular breast cancer classification to delineate the HER2-low subtype. NPJ Breast Cancer 2025; 11:19. [PMID: 39979291 PMCID: PMC11842814 DOI: 10.1038/s41523-025-00723-0] [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: 04/19/2024] [Accepted: 01/19/2025] [Indexed: 02/22/2025] Open
Abstract
Current breast cancer classification methods, particularly immunohistochemistry and PAM50, face challenges in accurately characterizing the HER2-low subtype, a therapeutically relevant entity with distinct biological features. This notable gap can lead to misclassification, resulting in inappropriate treatment decisions and suboptimal patient outcomes. Leveraging RNA-seq and machine-learning algorithms, we developed the Breast Cancer Classifier (BCC), a unique transcriptomic classifier for more precise breast cancer subtyping, specifically by delineating and incorporating HER2-low as a distinct subtype. BCC also redefined the PAM50 Normal subtype into other subtypes, disputing its classification as a unique molecular group. Our statistical analysis not only confirmed the reproducibility and accuracy of BCC, but also revealed similarities in prognostic characteristics between the HER2-low and Basal subtypes. Addressing this gap in breast cancer classification is clinically significant because it not only improves treatment stratification, but also uncovers novel molecular and immunohistochemical features associated with the HER2-low and HER2-high subtypes, thereby advancing our understanding of breast cancer heterogeneity and providing guidance in precision oncology.
Collapse
|
7
|
Yao N, Han L, Sun H, Zhou L, Wei Z. Androgen receptor expression and clinical significance in breast cancer. World J Surg Oncol 2025; 23:48. [PMID: 39934842 PMCID: PMC11816761 DOI: 10.1186/s12957-025-03673-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: 09/14/2024] [Accepted: 01/19/2025] [Indexed: 02/13/2025] Open
Abstract
PURPOSE This study aimed to investigate the expression and clinical relevance of the androgen receptor (AR) in breast cancer. METHODS This retrospective study examined the expression of AR in breast cancer and its correlation with patients' clinicopathological and immunohistochemical characteristics. A total of 521 patient records were gathered and assessed. Patients were categorized as either positive or negative for AR expression, and statistical analyses were conducted using the chi-square test, logistic regression in SPSS 26.0, and Kaplan-Meier analysis. RESULTS AR was detected in 83.7% of the 521 patients studied. There was a statistically significant difference in the prevalence of AR positivity among different molecular subtypes, estrogen receptor (ER) status, progesterone receptor (PR) status, human epidermal growth factor receptor 2 (HER2) status, and epidermal growth factor receptor (EGFR) (P < 0.05). Logistic regression analysis further revealed that ER and PR positivity were identified as risk factors for AR expression, and Kaplan-Meier curve analysis demonstrated the potential of AR as a prognostic indicator for breast cancer outcomes. Additionally, AR positivity was associated with a favorable prognosis. CONCLUSIONS The results suggest a strong correlation between AR expression and ER and PR co-expression in breast cancer. Additionally, AR positivity in the absence of ER and PR expression is associated with a favorable prognosis, indicating potential therapeutic value as a novel target in breast cancer treatment. Particularly in endocrine resistance or triple-negative breast cancer (TNBC), AR may serve as a significant prognostic indicator, warranting further investigation.
Collapse
MESH Headings
- Humans
- Receptors, Androgen/metabolism
- Female
- Retrospective Studies
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Middle Aged
- Prognosis
- Biomarkers, Tumor/metabolism
- Receptors, Progesterone/metabolism
- Receptors, Estrogen/metabolism
- Receptor, ErbB-2/metabolism
- Follow-Up Studies
- Adult
- Aged
- Survival Rate
- ErbB Receptors/metabolism
- Aged, 80 and over
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Clinical Relevance
Collapse
Affiliation(s)
- Ningning Yao
- Biobank, Linyi People's Hospital, Linyi, 276000, China
| | - Lei Han
- Biobank, Linyi People's Hospital, Linyi, 276000, China
| | - Han Sun
- Department of Laboratory Medicine, Linyi Peoples' Hospital, Linyi, 276000, China
| | - Liangjian Zhou
- Department of Scientific Research Management, Linyi People's Hospital, Linyi, 276000, China.
| | - Zhiyong Wei
- Biobank, Linyi People's Hospital, Linyi, 276000, China.
- Department of Pathology, Linyi People's Hospital, Linyi, 276000, China.
| |
Collapse
|
8
|
Dotto GP, Buckinx A, Özdemir BC, Simon C. Androgen receptor signalling in non-prostatic malignancies: challenges and opportunities. Nat Rev Cancer 2025; 25:93-108. [PMID: 39587300 PMCID: PMC11947662 DOI: 10.1038/s41568-024-00772-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 11/27/2024]
Abstract
The androgen receptor (AR) signalling pathway has been intensively studied in the context of prostate cancer, where androgen deprivation therapy is part of the standard of care for metastatic disease. By contrast, fewer studies have investigated the impact and translational potential of targeting AR in other cancer types where it is also expressed and functional. In this Review, we discuss the current understanding of AR in non-prostatic cancer types and summarize ongoing AR-directed clinical trials. While different androgen levels contribute to sexual dimorphism in cancer, targeting the AR system could benefit both sexes and help overcome resistance to targeted therapies. However, a bimodal function of AR signalling, which suppresses stromal changes associated with the early stages of cancer development, also needs to be considered. Future research is necessary to scrutinize cellular and molecular mechanisms of action of AR in cancer cells and the tumour microenvironment, to develop selective modulators of AR activity, and to identify patients with non-prostatic cancer who might benefit from targeting this pathway. AR-directed manipulation of host immune cells may offer a promising therapeutic approach for many types of cancers.
Collapse
Affiliation(s)
- G Paolo Dotto
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
- Service d'Oto-rhino-laryngologie et chirurgie cervical faciale, Centre Hospitalier Universitaire Vaudois (CHUV), Université de Lausanne (UNIL), Lausanne, Switzerland.
- International Cancer Prevention Institute, Epalinges, Switzerland.
| | - An Buckinx
- International Cancer Prevention Institute, Epalinges, Switzerland
| | - Berna C Özdemir
- Department of Medical Oncology, Inselspital Bern, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christian Simon
- Service d'Oto-rhino-laryngologie et chirurgie cervical faciale, Centre Hospitalier Universitaire Vaudois (CHUV), Université de Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
9
|
Srivastava TP, Dhar R, Karmakar S. Looking beyond the ER, PR, and HER2: what's new in the ARsenal for combating breast cancer? Reprod Biol Endocrinol 2025; 23:9. [PMID: 39833837 PMCID: PMC11744844 DOI: 10.1186/s12958-024-01338-z] [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: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BrCa) is a complex and heterogeneous disease with diverse molecular subtypes, leading to varied clinical outcomes and posing significant treatment challenges. The increasing global burden of BrCa, particularly in low- and middle-income countries, underscores the urgent need for more effective therapeutic strategies. The androgen receptor (AR), expressed in a substantial proportion of breast cancer cases, has emerged as a potential biomarker and therapeutic target. In breast cancer, AR exhibits diverse functions across subtypes, often interacting with other hormone receptors, thereby influencing tumor progression and treatment responses. This intricate interplay is further complicated by the presence of constitutively expressed AR splice variants (AR-Vs) that drive resistance to AR-targeting therapies through structural rearrangements in the domains and activation of aberrant signaling pathways. Although AR-targeting drugs, initially developed for prostate cancer (PCa), have shown promise in AR-positive breast cancer, significant gaps remain in understanding AR's precise functions and therapeutic potential. The systemic management of breast cancer is guided primarily by theranostic biomarkers; ER, PR, HER2, and Ki67 which also dictate the breast cancer classification. The ubiquitous expression of AR in BrCa and the emergence of AR-Vs can assist the management of disease complementing the standard of care. This article provides a comprehensive overview of AR and its splice variants in the context of breast cancer, highlighting their prognostic and predictive value across different subtypes looking beyond the conventional ER, PR, and HER2 status. This review also raises the possibility of using AR splice variants in predicting tumor aggressiveness. From the settings of developing nations, this may provide useful insight by integrating recent advances in AR-targeted therapies and exploring their translational potential, emphasizing the critical need for further research to optimize AR-based therapeutic strategies for breast cancer management.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/therapy
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Female
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
Collapse
Affiliation(s)
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
10
|
Pataccini G, Elia A, Sequeira G, Ambrosio L, Coianis M, Lamb CA, Rojas PA, Martínez Vázquez P, Burruchaga J, Spengler E, Vanzulli SI, Abba M, Lanari C. Steroid hormone receptors, exome sequencing and treatment responsiveness of breast cancer patient-derived xenografts originated in a South American country. Sci Rep 2025; 15:2415. [PMID: 39827242 PMCID: PMC11742900 DOI: 10.1038/s41598-025-86389-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BC) patient-derived xenografts (PDX) are relevant models for precision medicine. However, there are no collections derived from South American BC patients. Since ethnicity significantly impacts clinical outcomes, it is necessary to develop PDX models from different lineages. Our goals were to a) develop BC PDX from our population; b) characterize the expression of estrogen (ER), progesterone (PR), androgen (AR) and glucocorticoid (GR) receptors, basal and luminal cytokeratins, EGFR and HER2; c) identify PDX mutations; d) evaluate the response to treatments selected based on their biological and genetic features, and e) perform BC tissue cultures (BCTC) from PDX tissues and compare in vivo and ex vivo results. Surgical fragments were maintained in a culture medium and inoculated subcutaneously into untreated NSG female mice, or treated with estradiol pellets. Other fragments were fixed in formalin for diagnosis and immunohistochemistry, and a third piece was frozen at -80°C for molecular studies or whole exome-sequencing. Tumors were serially transplanted into NSG mice. Once the PDX was established, in vivo and ex vivo drug responses were evaluated. Eight PDX were established: two ER + [BC-AR685 (PR +) and BC-AR707 (PR-)], one from a triple-negative (TN) recurrence whose primary tumor was ER + (BC-AR485), one HER2 + (BC-AR474) and four TN primary tumors (BC-AR553, BC-AR546, BC-AR631 and BC-AR687). BC-AR685 had higher levels of PR isoform A than isoform B and was sensitive to mifepristone, tamoxifen, and palbociclib. BC-AR707 was inhibited by tamoxifen and testosterone. BC-AR474 was inhibited by trastuzumab and trastuzumab emtansine. BC-AR485 was sensitive to doxorubicin and resistant to paclitaxel in vivo and ex vivo. BC-AR687 carried a PIK3CA (C420R) mutation and was sensitive to alpelisib and mTOR inhibitors. All PDX expressed AR with varying intensities. GR and AR were co-expressed in the ER + tumors and in 3 TN PDX. We report the first PDX originated from South American countries that were genetically and biologically characterized and may be used in precision medicine studies. PDX expressing AR and/or GR are powerful tools to evaluate different endocrine treatment combinations even in TN tumors.
Collapse
Affiliation(s)
- Gabriela Pataccini
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Andrés Elia
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Gonzalo Sequeira
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Luisa Ambrosio
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Marcela Coianis
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Caroline A Lamb
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Paola A Rojas
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | | | - Javier Burruchaga
- Hospital de Agudos Magdalena V. de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Eunice Spengler
- Hospital de Agudos Magdalena V. de Martínez, General Pacheco, Buenos Aires, Argentina
| | | | - Martin Abba
- Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Kiraz U, Rewcastle E, Fykse SK, Lundal I, Gudlaugsson EG, Skaland I, Søiland H, Baak JPA, Janssen EAM. Dual Functions of Androgen Receptor Overexpression in Triple-Negative Breast Cancer: A Complex Prognostic Marker. Bioengineering (Basel) 2025; 12:54. [PMID: 39851328 PMCID: PMC11761274 DOI: 10.3390/bioengineering12010054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
A subset of triple-negative breast cancer (TNBC) expresses the androgen receptor (AR), but thresholds for AR positivity and its clinical significance vary. We hypothesize that objective assessment outperforms subjective methods, and that high AR negatively impacts prognosis. In a population-based TNBC cohort (n = 198) with long follow-up (4-383 months), AR expression was evaluated via subjective scoring (AR-Manual) and automated digital image analysis (AR-DIA). A 10% cut-off value via AR-DIA was the strongest negative prognostic threshold for distant metastases (p = 0.008). High AR-DIA correlated with lower grade (p = 0.014), and lower proliferation (p = 0.004) but also with larger tumors (p = 0.047), distant metastasis (p = 0.052), and lymph node (LN) positivity (p < 0.001), highlighting its dual roles. Multivariate analysis revealed interaction between LN status and AR-DIA (p < 0.001) as the strongest prognostic factor, followed by fibrotic focus (FF; p = 0.009), mitotic activity index (MAI; p = 0.018), and stromal tumor-infiltrating lymphocytes (sTILs; p = 0.041). AR-DIA had no additional prognostic value in favorable subgroups but was significant in unfavorable subgroups. In high AR-DIA patients with unfavorable characteristics, ACT did not improve survival, and patients may benefit from AR-targeted therapy. Overall, the DIA method provides reproducibility, high AR-DIA (≥10%) shows opposing survival effects in different TNBC subgroups, and AR evaluation is crucial for prognosis and AR-targeted therapies.
Collapse
Affiliation(s)
- Umay Kiraz
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway
| | - Emma Rewcastle
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Silja K. Fykse
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Ingrid Lundal
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Einar G. Gudlaugsson
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Ivar Skaland
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Håvard Søiland
- Department of Research, Stavanger University Hospital, 4011 Stavanger, Norway
| | - Jan P. A. Baak
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway
- Institute for Biomedicine and Glycomics, Griffith University, Queensland, QLD 4215, Australia
| |
Collapse
|
12
|
Thakkar NH, Osama MA, Dhawan S. Analyzing Androgen Receptor Expression in Breast Cancer: Insights into Histopathological Parameters and Hormone Receptor Status Among Indian Women. Indian J Surg Oncol 2024; 15:789-795. [PMID: 39555351 PMCID: PMC11564589 DOI: 10.1007/s13193-024-01997-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/19/2024] [Indexed: 11/19/2024] Open
Abstract
Breast cancer, an exceptionally hormone-dependent tumor, exhibits a diverse clinical profile. Its therapeutic categorization relies on the expression of key receptors, namely, estrogen receptor (ER), progesterone receptor (PR), and Her2neu. The androgen receptor (AR), a member of the nuclear receptor superfamily, is a biomarker gaining attention in breast cancer research, particularly for triple-negative breast cancers. We conducted an analysis of AR expression in 113 primary breast cancer cases, using a cutoff criterion of ≥ 10% tumor cell positivity. ER, PR, and Her2neu statuses were determined based on the 2023 ASCO-CAP criteria. AR expression was then correlated with various clinicopathological factors, including age, menopausal status, centricity, histological type, grade, tumor size, nodal status, lymphovascular and perineural invasion, and ER, PR, and HER2neu statuses. Among the 113 cases, 57 (50.4%) showed positive AR expression. No statistically significant associations were found between AR expression and age, menopausal status, histological type, histological grade, nodal status, or ER and PR expression. Notably, all multicentric tumors (n = 7, 100%) were AR negative. AR expression was linked to smaller tumor sizes. Positive AR cases exhibited an association with Her2neu overexpression, particularly in ER and PR-negative tumors. Of note, 35% of triple-negative tumors displayed AR positivity. AR emerges as a promising marker in breast cancers, particularly in triple-negative cases. Larger-scale studies are warranted to comprehensively assess the relationship between AR expression and histopathological parameters, as well as other immunohistochemical markers.
Collapse
Affiliation(s)
| | - Md Ali Osama
- Department of Pathology, Lady Hardinge Medical College, New Delhi, India
| | - Shashi Dhawan
- Department of Histopathology, Sir Gangaram Hospital, New Delhi, India
| |
Collapse
|
13
|
Adam N, Wieder R. AI Survival Prediction Modeling: The Importance of Considering Treatments and Changes in Health Status over Time. Cancers (Basel) 2024; 16:3527. [PMID: 39456622 PMCID: PMC11505986 DOI: 10.3390/cancers16203527] [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: 09/18/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Deep learning (DL)-based models for predicting the survival of patients with local stages of breast cancer only use time-fixed covariates, i.e., patient and cancer data at the time of diagnosis. These predictions are inherently error-prone because they do not consider time-varying events that occur after initial diagnosis. Our objective is to improve the predictive modeling of survival of patients with localized breast cancer to consider both time-fixed and time-varying events; thus, we take into account the progression of a patient's health status over time. METHODS We extended four DL-based predictive survival models (DeepSurv, DeepHit, Nnet-survival, and Cox-Time) that deal with right-censored time-to-event data to consider not only a patient's time-fixed covariates (patient and cancer data at diagnosis) but also a patient's time-varying covariates (e.g., treatments, comorbidities, progressive age, frailty index, adverse events from treatment). We utilized, as our study data, the SEER-Medicare linked dataset from 1991 to 2016 to study a population of women diagnosed with stage I-III breast cancer (BC) enrolled in Medicare at 65 years or older as qualified by age. We delineated time-fixed variables recorded at the time of diagnosis, including age, race, marital status, breast cancer stage, tumor grade, laterality, estrogen receptor (ER), progesterone receptor (PR), and human epidermal receptor 2 (HER2) status, and comorbidity index. We analyzed six distinct prognostic categories, cancer stages I-III BC, and each stage's ER/PR+ or ER/PR- status. At each visit, we delineated the time-varying covariates of administered treatments, induced adverse events, comorbidity index, and age. We predicted the survival of three hypothetical patients to demonstrate the model's utility. MAIN OUTCOMES AND MEASURES The primary outcomes of the modeling were the measures of the model's prediction error, as measured by the concordance index, the most commonly applied evaluation metric in survival analysis, and the integrated Brier score, a metric of the model's discrimination and calibration. RESULTS The proposed extended patients' covariates that include both time-fixed and time-varying covariates significantly improved the deep learning models' prediction error and the discrimination and calibration of a model's estimates. The prediction of the four DL models using time-fixed covariates in six different prognostic categories all resulted in approximately a 30% error in all six categories. When applying the proposed extension to include time-varying covariates, the accuracy of all four predictive models improved significantly, with the error decreasing to approximately 10%. The models' predictive accuracy was independent of the differing published survival predictions from time-fixed covariates in the six prognostic categories. We demonstrate the utility of the model in three hypothetical patients with unique patient, cancer, and treatment variables. The model predicted survival based on the patient's individual time-fixed and time-varying features, which varied considerably from Social Security age-based, and stage and race-based breast cancer survival predictions. CONCLUSIONS The predictive modeling of the survival of patients with early-stage breast cancer using DL models has a prediction error of around 30% when considering only time-fixed covariates at the time of diagnosis and decreases to values under 10% when time-varying covariates are added as input to the models, regardless of the prognostic category of the patient groups. These models can be used to predict individual patients' survival probabilities based on their unique repertoire of time-fixed and time-varying features. They will provide guidance for patients and their caregivers to assist in decision making.
Collapse
Affiliation(s)
- Nabil Adam
- Phalcon, LLC, Manhasset, NY 11030, USA;
- Newark Campus, Rutgers University, Newark, NJ 07102, USA
| | - Robert Wieder
- Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
14
|
Sheva K, Roy Chowdhury S, Kravchenko-Balasha N, Meirovitz A. Molecular Changes in Breast Cancer Induced by Radiation Therapy. Int J Radiat Oncol Biol Phys 2024; 120:465-481. [PMID: 38508467 DOI: 10.1016/j.ijrobp.2024.03.019] [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: 06/13/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Breast cancer treatments are based on prognostic clinicopathologic features that form the basis for therapeutic guidelines. Although the utilization of these guidelines has decreased breast cancer-associated mortality rates over the past three decades, they are not adequate for individualized therapy. Radiation therapy (RT) is the backbone of breast cancer treatment. Although a highly successful therapeutic modality clinically, from a biological perspective, preclinical studies have shown RT to have the potential to alter tumor cell phenotype, immunogenicity, and the surrounding microenvironment, potentially changing the behavior of cancer cells and resulting in a significant variation in RT response. This review presents the recent advances in revealing the complex molecular changes induced by RT in the treatment of breast cancer and highlights the complexities of translating this information into clinically relevant tools for improved prognostic insights and the revelation of novel approaches for optimizing RT. METHODS AND MATERIALS Current literature was reviewed with a focus on recent advances made in the elucidation of tumor-associated radiation-induced molecular changes across molecular, genetic, and proteomic bases. This review was structured with the aim of providing an up-to-date overview over the very broad and complex subject matter of radiation-induced molecular changes and radioresistance, familiarizing the reader with the broader issue at hand. RESULTS The subject of radiation-induced molecular changes in breast cancer has been broached from various physiological focal points including that of the immune system, immunogenicity and the abscopal effect, tumor hypoxia, breast cancer classification and subtyping, molecular heterogeneity, and molecular plasticity. It is becoming increasingly apparent that breast cancer clinical subtyping alone does not adequately account for variation in RT response or radioresistance. Multiple components of the tumor microenvironment and immune system, delivered RT dose and fractionation schedules, radiation-induced bystander effects, and intrinsic tumor physiology and heterogeneity all contribute to the resultant RT outcome. CONCLUSIONS Despite recent advances and improvements in anticancer therapies, tumor resistance remains a significant challenge. As new analytical techniques and technologies continue to provide crucial insight into the complex molecular mechanisms of breast cancer and its treatment responses, it is becoming more evident that personalized anticancer treatment regimens may be vital in overcoming radioresistance.
Collapse
Affiliation(s)
- Kim Sheva
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka University Medical Center, Ben Gurion University of the Negev, Faculty of Medicine, Be'er Sheva, Israel.
| | - Sangita Roy Chowdhury
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Amichay Meirovitz
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka University Medical Center, Ben Gurion University of the Negev, Faculty of Medicine, Be'er Sheva, Israel.
| |
Collapse
|
15
|
Keerthana Devi D, Pavithra V, Joseph LD, Challa CB. Correlation of Androgen Receptor Expression With Ki67 Proliferative Index and Other Clinicopathological Characteristics in Invasive Mammary Carcinomas. Cureus 2024; 16:e70867. [PMID: 39497884 PMCID: PMC11534436 DOI: 10.7759/cureus.70867] [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: 10/04/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction The clinical importance of androgen receptor (AR) status in breast cancer is uncertain. The existing knowledge regarding the association of androgen receptor expression with clinicopathological factors of breast cancer is also limited. The main aim of this study is to evaluate the AR expression in breast cancer and to correlate it with the Ki67 proliferative index and other clinicopathological prognostic factors. Methods The expression of AR was evaluated in 192 invasive mammary carcinoma cases and the expression patterns were correlated with various clinicopathological prognostic factors such as age, tumor size, pathological primary tumor (pT) stage, nodal status, histological grade, estrogen receptor (ER) expression, progesterone receptor (PgR) expression, human epidermal growth factor receptor 2 (Her2) status, molecular subtype, and Ki67 labeling index. Immunohistochemistry was performed to assess AR, ER, PgR, Her2, and Ki67 expression. The clinicopathological data required for the analysis were obtained from the laboratory information system. Results Out of the 192 breast carcinoma cases, 139 (72.4%) showed AR-positive expression. The average age of the AR-positive cases was slightly higher than the AR-negative cases. AR-positive tumors tended to have a lower histological grade and positive ER and PgR expression. The expression of AR did not correlate with tumor size, pT stage, nodal status, Her2 status, and Ki67 labeling index. Conclusion The expression of AR is noted in a significant proportion of breast carcinoma cases. AR expression may be related to good prognostic factors such as ER expression, PgR expression, and lower histologic grade. We also observed that AR expression did not have any association with the Ki67 proliferative index.
Collapse
Affiliation(s)
- D Keerthana Devi
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - V Pavithra
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Leena D Joseph
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Chithra Bhanu Challa
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| |
Collapse
|
16
|
Asemota S, Effah W, Holt J, Johnson D, Cripe L, Ponnusamy S, Thiyagarajan T, Khosrosereshki Y, Hwang DJ, He Y, Grimes B, Fleming MD, Pritchard FE, Hendrix A, Fan M, Jain A, Choi HY, Makowski L, Hayes DN, Miller DD, Pfeffer LM, Santhanam B, Narayanan R. A molecular switch from tumor suppressor to oncogene in ER+ve breast cancer: Role of androgen receptor, JAK-STAT, and lineage plasticity. Proc Natl Acad Sci U S A 2024; 121:e2406837121. [PMID: 39312663 PMCID: PMC11459127 DOI: 10.1073/pnas.2406837121] [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: 04/10/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Cancers develop resistance to inhibitors of oncogenes mainly due to target-centric mechanisms such as mutations and splicing. While inhibitors or antagonists force targets to unnatural conformation contributing to protein instability and resistance, activating tumor suppressors may maintain the protein in an agonistic conformation to elicit sustainable growth inhibition. Due to the lack of tumor suppressor agonists, this hypothesis and the mechanisms underlying resistance are not understood. In estrogen receptor (ER)-positive breast cancer (BC), androgen receptor (AR) is a druggable tumor suppressor offering a promising avenue for this investigation. Spatial genomics suggests that the molecular portrait of AR-expressing BC cells in tumor microenvironment corresponds to better overall patient survival, clinically confirming AR's role as a tumor suppressor. Ligand activation of AR in ER-positive BC xenografts reprograms cistromes, inhibits oncogenic pathways, and promotes cellular elasticity toward a more differentiated state. Sustained AR activation results in cistrome rearrangement toward transcription factor PROP paired-like homeobox 1, transformation of AR into oncogene, and activation of the Janus kinase/signal transducer (JAK/STAT) pathway, all culminating in lineage plasticity to an aggressive resistant subtype. While the molecular profile of AR agonist-sensitive tumors corresponds to better patient survival, the profile represented in the resistant phenotype corresponds to shorter survival. Inhibition of activated oncogenes in resistant tumors reduces growth and resensitizes them to AR agonists. These findings indicate that persistent activation of a context-dependent tumor suppressor may lead to resistance through lineage plasticity-driven tumor metamorphosis. Our work provides a framework to explore the above phenomenon across multiple cancer types and underscores the importance of factoring sensitization of tumor suppressor targets while developing agonist-like drugs.
Collapse
Affiliation(s)
- Sarah Asemota
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Wendy Effah
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Jeremiah Holt
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Daniel Johnson
- Molecular Bioinformatics Core, University of Tennessee Health Science Center, Memphis, TN38163
| | - Linnea Cripe
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Suriyan Ponnusamy
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Thirumagal Thiyagarajan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Yekta Khosrosereshki
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN38163
| | - Yali He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN38163
| | - Brandy Grimes
- West Cancer Center and Research Institute, Memphis, TN38120
| | - Martin D. Fleming
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Frances E. Pritchard
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Ashley Hendrix
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Meiyun Fan
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Abhinav Jain
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX77030
| | - Hyo Young Choi
- University of Tennessee Health Science Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN38163
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN38163
| | - Liza Makowski
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
- University of Tennessee Health Science Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN38163
| | - D. Neil Hayes
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
- University of Tennessee Health Science Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN38163
| | - Duane D. Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN38163
- University of Tennessee Health Science Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN38163
| | - Lawrence M. Pfeffer
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
- University of Tennessee Health Science Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN38163
| | - Balaji Santhanam
- Center of Excellence for Data Driven Discovery and Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Ramesh Narayanan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN38163
- University of Tennessee Health Science Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN38163
| |
Collapse
|
17
|
Senga SS, Bisson WH, Colacci A. Key characteristics of carcinogens meet hallmarks for prevention-cutting the Gordian knot. Front Oncol 2024; 14:1420687. [PMID: 39435286 PMCID: PMC11491790 DOI: 10.3389/fonc.2024.1420687] [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: 04/20/2024] [Accepted: 08/05/2024] [Indexed: 10/23/2024] Open
Abstract
The complexity of cancer requires a comprehensive approach to understand its diverse manifestations and underlying mechanisms. Initially outlined by Hanahan and Weinberg in 2000 and updated in 2010, the hallmarks of cancer provide a conceptual basis for understanding inherent variability in cancer biology. Recent expansions have further elucidated additional hallmarks, including phenotypic plasticity and senescent cells. The International Agency for Research on Cancer (IARC) has identified the key characteristics of carcinogens (KCCs) to evaluate their carcinogenic potential. We analyzed chemicals of concern for environmental exposure that interact with specific receptors to induce genomic instability, epigenetic alterations, immune suppression, and receptor-mediated effects, thereby contributing to chronic inflammation. Despite their varying degrees of carcinogenicity, these chemicals have similar KCC profiles. Our analysis highlights the pivotal role of receptor binding in activating most other KCCs, underscoring their significance in cancer initiation. Although KCCs are associated with early molecular or cellular events, they do not encompass processes directly linked to full cellular malignancy. Thus, there is a need to integrate clear endpoints that anchor KCCs to the acquisition of a complete malignant phenotype into chemical testing. From the perspective of toxicology and cancer research, an all-encompassing strategy that incorporates both existing and novel KCCs and cancer hallmarks is essential to enable the targeted identification of prevalent carcinogens and facilitate zone-specific prevention strategies. To achieve this goal, collaboration between the KCC and cancer hallmarks communities becomes essential.
Collapse
Affiliation(s)
- Sasi S. Senga
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - William H. Bisson
- Integrative Toxicology and Cancer Prevention, Durham, NC, United States
| | - Annamaria Colacci
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Bologna, Italy
- Alma Mater Institute on Healthy Planet – University of Bologna, Bologna, Italy
| |
Collapse
|
18
|
Gao H, Wei L, Indulkar S, Nguyen TTL, Liu D, Ho MF, Zhang C, Li H, Weinshilboum RM, Ingle JN, Wang L. Androgen receptor-mediated pharmacogenomic expression quantitative trait loci: implications for breast cancer response to AR-targeting therapy. Breast Cancer Res 2024; 26:111. [PMID: 38965614 PMCID: PMC11225427 DOI: 10.1186/s13058-024-01861-2] [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: 11/17/2023] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies. METHODS We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog. RESULTS We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs. CONCLUSIONS We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.
Collapse
Affiliation(s)
- Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Shreya Indulkar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Thanh Thanh L Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA.
| |
Collapse
|
19
|
Shukla N, Shah K, Rathore D, Soni K, Shah J, Vora H, Dave H. Androgen receptor: Structure, signaling, function and potential drug discovery biomarker in different breast cancer subtypes. Life Sci 2024; 348:122697. [PMID: 38710280 DOI: 10.1016/j.lfs.2024.122697] [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: 01/09/2024] [Revised: 04/28/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
The Androgen Receptor (AR) is emerging as an important factor in the pathogenesis of breast cancer (BC), which is the most common malignancy worldwide. >70 % of AR expression in primary and metastatic breast tumors has been observed which suggests that AR may be a new marker and a potential therapeutic target among AR-positive BC patients. Biological insight into AR-positive breast cancer reveals that AR may cross-talk with several vital signaling pathways, including key molecules and receptors. Downstream signaling of AR might also affect many clinically important pathways that are emerging as clinical targets in BC. AR exhibits different behaviors depending on the breast cancer molecular subtype. Preliminary clinical research using AR-targeted drugs, which have already been FDA-approved for prostate cancer (PC), has given promising results for AR-positive breast cancer patients. However, since AR positivity's prognostic and predictive value remains uncertain, it is difficult to identify and stratify patients who would benefit from AR-targeted therapies alone. Thus, the need of the hour is to target the androgen receptor as a monotherapy or in combination with other conventional therapies which has proven to be an effective clinical strategy for the treatment of prostate cancer patients, and these therapeutic strategies are increasingly being investigated in breast cancer. Therefore, in this manuscript, we review the role of AR in various cellular processes that promote tumorigenesis and aggressiveness, in different subtypes of breast cancer, as well as discuss ongoing efforts to target AR for the more effective treatment and prevention of breast cancer.
Collapse
Affiliation(s)
- Nirali Shukla
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Kanisha Shah
- Division of Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Deepshikha Rathore
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Kinal Soni
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Jigna Shah
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Hemangini Vora
- The Gujarat Cancer & Research Institute, Ahmedabad, Gujarat 380016, India
| | - Heena Dave
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
20
|
Keigley QJ, Fowler AM, O'Brien SR, Dehdashti F. Molecular Imaging of Steroid Receptors in Breast Cancer. Cancer J 2024; 30:142-152. [PMID: 38753748 PMCID: PMC11101139 DOI: 10.1097/ppo.0000000000000715] [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] [Indexed: 05/18/2024]
Abstract
ABSTRACT Steroid receptors regulate gene expression for many important physiologic functions and pathologic processes. Receptors for estrogen, progesterone, and androgen have been extensively studied in breast cancer, and their expression provides prognostic information as well as targets for therapy. Noninvasive imaging utilizing positron emission tomography and radiolabeled ligands targeting these receptors can provide valuable insight into predicting treatment efficacy, staging whole-body disease burden, and identifying heterogeneity in receptor expression across different metastatic sites. This review provides an overview of steroid receptor imaging with a focus on breast cancer and radioligands for estrogen, progesterone, and androgen receptors.
Collapse
Affiliation(s)
- Quinton J Keigley
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | - Sophia R O'Brien
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Farrokh Dehdashti
- Division of Nuclear Medicine, Edward Mallinckrodt Institute of Radiology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
21
|
Tateishi Y, Webb SN, Li B, Liu L, Lindsey Rose K, Leser M, Patel P, Guengerich FP. Proteomics, modeling, and fluorescence assays delineate cytochrome b 5 residues involved in binding and stimulation of cytochrome P450 17A1 17,20-lyase. J Biol Chem 2024; 300:105688. [PMID: 38280431 PMCID: PMC10878793 DOI: 10.1016/j.jbc.2024.105688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024] Open
Abstract
Cytochrome b5 (b5) is known to stimulate some catalytic activities of cytochrome P450 (P450, CYP) enzymes, although mechanisms still need to be defined. The reactions most strongly enhanced by b5 are the 17,20-lyase reactions of P450 17A1 involved in steroid biosynthesis. We had previously used a fluorescently labeled human b5 variant (Alexa 488-T70C-b5) to characterize human P450 17A1-b5 interactions, but subsequent proteomic analyses indicated that lysines in b5 were also modified with Alexa 488 maleimide in addition to Cys-70, due to disulfide dimerization of the T70C mutant. A series of b5 variants were constructed with Cys replacements for the identified lysine residues and labeled with the dye. Fluorescence attenuation and the function of b5 in the steroid lyase reaction depended on the modified position. Apo-b5 (devoid of heme group) studies revealed the lack of involvement of the b5 heme in the fluorescence attenuation. A structural model of b5 with P450 17A1 was predicted using AlphaFold-Multimer algorithms/Rosetta docking, based upon the individual structures, which predicted several new contacts not previously reported, that is, interactions of b5 Glu-48:17A1 Arg-347, b5 Glu-49:17A1 Arg-449, b5 Asp-65:17A1 Arg-126, b5 Asp-65:17A1 Arg-125, and b5 Glu-61:17A1 Lys-91. Fluorescence polarization assays with two modified b5 variants yielded Kd values (for b5-P450 17A1) of 120 to 380 nM, the best estimate of binding affinity. We conclude that both monomeric and dimeric b5 can bind to P450 17A1 and stimulate activity. Results with the mutants indicate that several Lys residues in b5 are sensitive to the interaction with P450 17A1, including Lys-88 and Lys-91.
Collapse
Affiliation(s)
- Yasuhiro Tateishi
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Stephany N Webb
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Bian Li
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lu Liu
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kristie Lindsey Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Micheal Leser
- Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Purvi Patel
- Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
22
|
Hosseinzadeh L, Kikhtyak Z, Laven-Law G, Pederson SM, Puiu CG, D'Santos CS, Lim E, Carroll JS, Tilley WD, Dwyer AR, Hickey TE. The androgen receptor interacts with GATA3 to transcriptionally regulate a luminal epithelial cell phenotype in breast cancer. Genome Biol 2024; 25:44. [PMID: 38317241 PMCID: PMC10840202 DOI: 10.1186/s13059-023-03161-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND The androgen receptor (AR) is a tumor suppressor in estrogen receptor (ER) positive breast cancer, a role sustained in some ER negative breast cancers. Key factors dictating AR genomic activity in a breast context are largely unknown. Herein, we employ an unbiased chromatin immunoprecipitation-based proteomic technique to identify endogenous AR interacting co-regulatory proteins in ER positive and negative models of breast cancer to gain new insight into mechanisms of AR signaling in this disease. RESULTS The DNA-binding factor GATA3 is identified and validated as a novel AR interacting protein in breast cancer cells irrespective of ER status. AR activation by the natural ligand 5α-dihydrotestosterone (DHT) increases nuclear AR-GATA3 interactions, resulting in AR-dependent enrichment of GATA3 chromatin binding at a sub-set of genomic loci. Silencing GATA3 reduces but does not prevent AR DNA binding and transactivation of genes associated with AR/GATA3 co-occupied loci, indicating a co-regulatory role for GATA3 in AR signaling. DHT-induced AR/GATA3 binding coincides with upregulation of luminal differentiation genes, including EHF and KDM4B, established master regulators of a breast epithelial cell lineage. These findings are validated in a patient-derived xenograft model of breast cancer. Interaction between AR and GATA3 is also associated with AR-mediated growth inhibition in ER positive and ER negative breast cancer. CONCLUSIONS AR and GATA3 interact to transcriptionally regulate luminal epithelial cell differentiation in breast cancer regardless of ER status. This interaction facilitates the tumor suppressor function of AR and mechanistically explains why AR expression is associated with less proliferative, more differentiated breast tumors and better overall survival in breast cancer.
Collapse
Affiliation(s)
- Leila Hosseinzadeh
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Zoya Kikhtyak
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Geraldine Laven-Law
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Stephen M Pederson
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Caroline G Puiu
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Clive S D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Elgene Lim
- Garvan Institute of Medical Research, University of New South Wales, Sydney, Australia
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Amy R Dwyer
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
23
|
Xu F, Xu K, Fan L, Li X, Liu Y, Yang F, Zhu C, Guan X. Estrogen receptor beta suppresses the androgen receptor oncogenic effects in triple-negative breast cancer. Chin Med J (Engl) 2024; 137:338-349. [PMID: 38105538 PMCID: PMC10836903 DOI: 10.1097/cm9.0000000000002930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer associated with poor prognosis and limited treatment options. The androgen receptor (AR) has emerged as a potential therapeutic target for luminal androgen receptor (LAR) TNBC. However, multiple studies have claimed that anti-androgen therapy for AR-positive TNBC only has limited clinical benefits. This study aimed to investigate the role of AR in TNBC and its detailed mechanism. METHODS Immunohistochemistry and TNBC tissue sections were applied to investigate AR and nectin cell adhesion molecule 4 (NECTIN4) expression in TNBC tissues. Then, in vitro and in vivo assays were used to explore the function of AR and estrogen receptor beta (ERβ) in TNBC. Chromatin immunoprecipitation sequencing (ChIP-seq), co-immunoprecipitation (co-IP), molecular docking method, and luciferase reporter assay were performed to identify key molecules that affect the function of AR. RESULTS Based on the TNBC tissue array analysis, we revealed that ERβ and AR were positive in 21.92% (32/146) and 24.66% (36/146) of 146 TNBC samples, respectively, and about 13.70% (20/146) of TNBC patients were ERβ positive and AR positive. We further demonstrated the pro-tumoral effects of AR on TNBC cells, however, the oncogenic biology was significantly suppressed when ERβ transfection in LAR TNBC cell lines but not in AR-negative TNBC. Mechanistically, we identified that NECTIN4 promoter -42 bp to -28 bp was an AR response element, and that ERβ interacted with AR thus impeding the AR-mediated NECTIN4 transcription which promoted epithelial-mesenchymal transition in tumor progression. CONCLUSIONS This study suggests that ERβ functions as a suppressor mediating the effect of AR in TNBC prognosis and cell proliferation. Therefore, our current research facilitates a better understanding of the role and mechanisms of AR in TNBC carcinogenesis.
Collapse
Affiliation(s)
- Feng Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Kun Xu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Lingling Fan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xintong Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yiqiu Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Fang Yang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Chengjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
24
|
Tien AH, Sadar MD. Treatments Targeting the Androgen Receptor and Its Splice Variants in Breast Cancer. Int J Mol Sci 2024; 25:1817. [PMID: 38339092 PMCID: PMC10855698 DOI: 10.3390/ijms25031817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/15/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Breast cancer is a major cause of death worldwide. The complexity of endocrine regulation in breast cancer may allow the cancer cells to escape from a particular treatment and result in resistant and aggressive disease. These breast cancers usually have fewer treatment options. Targeted therapies for cancer patients may offer fewer adverse side effects because of specificity compared to conventional chemotherapy. Signaling pathways of nuclear receptors, such as the estrogen receptor (ER), have been intensively studied and used as therapeutic targets. Recently, the role of the androgen receptor (AR) in breast cancer is gaining greater attention as a therapeutic target and as a prognostic biomarker. The expression of constitutively active truncated AR splice variants in breast cancer is a possible mechanism contributing to treatment resistance. Therefore, targeting both the full-length AR and AR variants, either through the activation or suppression of AR function, depending on the status of the ER, progesterone receptor, or human epidermal growth factor receptor 2, may provide additional treatment options. Studies targeting AR in combination with other treatment strategies are ongoing in clinical trials. The determination of the status of nuclear receptors to classify and identify patient subgroups will facilitate optimized and targeted combination therapies.
Collapse
Affiliation(s)
- Amy H. Tien
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Marianne D. Sadar
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| |
Collapse
|
25
|
Ilenič P, Herman A, Langerholc E, Gazić B, Šeruga B. Association of androgen receptor and tumour-infiltrating lymphocytes with bone recurrence in triple-negative breast cancer. J Bone Oncol 2024; 44:100518. [PMID: 38374890 PMCID: PMC10874935 DOI: 10.1016/j.jbo.2023.100518] [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: 10/26/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 02/21/2024] Open
Abstract
Background As compared to endocrine responsive breast cancer bone is less frequent site of distant recurrence in triple-negative breast cancer (TNBC). A biomarker which predicts bone recurrence would allow a more personalized treatment approach with adjuvant bisphosphonates in TNBC. Here we hypothesised that tumour expression of androgen receptor (AR) is associated with bone recurrence in TNBC. Materials and methods Patients with operable TNBC who were treated at the Institute of Oncology Ljubljana between 2005 and 2015 and developed distant recurrence were included into our study. Nuclear expression of AR in the tissue of primary tumours was determined immunohistochemically by using the Androgen Receptor (SP107) Rabbit Monoclonal Antibody. We applied a logistic regression model to test the association between expression of AR and development of bone metastases. The model was adjusted for selected known prognostic factors and possible confounders in TNBC, including the level of the stromal tumour-infiltrating lymphocytes (sTILs). Results At recurrence 45 (45 %) out of 100 patients presented with bone metastases. Additionally, seven (7 %) developed bone metastases metachronously. AR was expressed in primary tumours of 35 (35 %) women and 19 (54.3 %) developed bone recurrence. In 25 (25 %) patients sTILs were absent. Neither the proportion of AR positive cancer cells (OR = 1.00; 95 % CI 0.96-1.03; p = 1.00) nor the intensity of AR positive reaction (OR = 0.71; 95 % CI 0.02-21.4; p = 1.00) were significantly associated with bone recurrence. However, women with at least mild level of the sTILs were at significantly lower risk for bone recurrence as compared to those without any sTILs (OR = 0.01; 95 % CI < 0.01-0.08; p = 0.01). Conclusions Expression of AR is not significantly associated with the development of bone metastases in TNBC. However, patients with absent sTILs in their primary tumours are highly susceptible for recurrence in the bone and might particularly benefit from adjuvant bisphosphonates.
Collapse
Affiliation(s)
- Petra Ilenič
- University Medical Centre Ljubljana, Zaloška cesta 2, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Ajda Herman
- University Medical Centre Ljubljana, Zaloška cesta 2, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Erik Langerholc
- Institute for Biostatistics and Medical Informatics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Barbara Gazić
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
- Department of Pathology, Institute of Oncology Ljubljana, Zaloška cesta 2, Ljubljana, Slovenia
| | - Boštjan Šeruga
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
- Division of Medical Oncology, Institute of Oncology Ljubljana, Zaloška cesta 2, Ljubljana, Slovenia
| |
Collapse
|
26
|
Sato S, Imada S, Hayami R, Arai K, Kosugi R, Tsuneizumi M, Matsunuma R. Complexities in Adjuvant Endocrine Therapy for Breast Cancer in Female-to-Male Transgender Patients. Case Rep Oncol 2024; 17:208-216. [PMID: 38327829 PMCID: PMC10849748 DOI: 10.1159/000536212] [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: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Introduction Managing breast cancer in female-to-male (FtM) transgender patients is complicated and challenging. Androgens play a crucial role in the development of secondary sexual identity in FtM transgender patients, but their effectiveness in breast cancer remains unclear. Furthermore, the considerations for adjuvant endocrine therapy in this population are highly intricate and warrant thorough discussion. Case Presentation We describe the case of a 44-year-old FtM transgender diagnosed with breast cancer 3 years after initiating androgen receptor agonist therapy as part of his gender identity transition. After mastectomy, adjuvant endocrine therapy was initiated, consisting of a combination of an aromatase inhibitor and a gonadotropin-releasing hormone agonist, along with a cross-sex hormone. Conclusion Estradiol levels were significantly reduced, and male-typical levels of sex hormones were attained.
Collapse
Affiliation(s)
- Shoko Sato
- Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, Japan
| | - Sae Imada
- Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, Japan
| | - Ryosuke Hayami
- Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, Japan
| | - Kazumori Arai
- Department of Pathology, Shizuoka General Hospital, Shizuoka, Japan
| | - Rieko Kosugi
- Center for Diabetes, Endocrinology and Metabolism, Shizuoka General Hospital, Shizuoka, Japan
| | - Michiko Tsuneizumi
- Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, Japan
| | - Ryoichi Matsunuma
- Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, Japan
| |
Collapse
|
27
|
Sharifi MN, O'Regan RM, Wisinski KB. Is the Androgen Receptor a Viable Target in Triple Negative Breast Cancer in 5 Years? Clin Breast Cancer 2023; 23:813-824. [PMID: 37419745 DOI: 10.1016/j.clbc.2023.06.009] [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: 04/13/2023] [Revised: 06/09/2023] [Accepted: 06/17/2023] [Indexed: 07/09/2023]
Abstract
Triple negative breast cancer (TNBC) is characterized by high rates of disease recurrence after definitive therapy, and median survival of less than 18 months in the metastatic setting. Systemic therapy options for TNBC consist primarily of cytotoxic chemotherapy-containing regimens, and while recently FDA-approved chemo-immunotherapy combinations and antibody-drug conjugates such as Sacituzumab govitecan have improved clinical outcomes, there remains an unmet need for more effective and less toxic therapies. A subset of TNBC expresses the androgen receptor (AR), a nuclear hormone steroid receptor that activates an androgen-responsive transcriptional program, and gene expression profiling has revealed a TNBC molecular subtype with AR expression and luminal and androgen responsive features. Both preclinical and clinical data suggest biologic similarities between luminal AR (LAR) TNBC and ER+ luminal breast cancer, including lower proliferative activity, relative chemoresistance, and high rates of oncogenic activating mutations in the phosphatidylinositol-3-kinase (PI3K) pathway. Preclinical LAR-TNBC models are sensitive to androgen signaling inhibitors (ASIs), and particularly given the availability of FDA-approved ASIs with robust efficacy in prostate cancer, there has been great interest in targeting this pathway in AR+ TNBC. Here, we review the underlying biology and completed and ongoing androgen-targeted therapy studies in early stage and metastatic AR+ TNBC.
Collapse
Affiliation(s)
- Marina N Sharifi
- UW Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI.
| | - Ruth M O'Regan
- Department of Medicine, University of Rochester, Rochester, NY
| | - Kari B Wisinski
- UW Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI
| |
Collapse
|
28
|
Keup C, Kimmig R, Kasimir-Bauer S. The Diversity of Liquid Biopsies and Their Potential in Breast Cancer Management. Cancers (Basel) 2023; 15:5463. [PMID: 38001722 PMCID: PMC10670968 DOI: 10.3390/cancers15225463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Analyzing blood as a so-called liquid biopsy in breast cancer (BC) patients has the potential to adapt therapy management. Circulating tumor cells (CTCs), extracellular vesicles (EVs), cell-free DNA (cfDNA) and other blood components mirror the tumoral heterogeneity and could support a range of clinical decisions. Multi-cancer early detection tests utilizing blood are advancing but are not part of any clinical routine yet. Liquid biopsy analysis in the course of neoadjuvant therapy has potential for therapy (de)escalation.Minimal residual disease detection via serial cfDNA analysis is currently on its way. The prognostic value of blood analytes in early and metastatic BC is undisputable, but the value of these prognostic biomarkers for clinical management is controversial. An interventional trial confirmed a significant outcome benefit when therapy was changed in case of newly emerging cfDNA mutations under treatment and thus showed the clinical utility of cfDNA analysis for therapy monitoring. The analysis of PIK3CA or ESR1 variants in plasma of metastatic BC patients to prescribe targeted therapy with alpesilib or elacestrant has already arrived in clinical practice with FDA-approved tests available and is recommended by ASCO. The translation of more liquid biopsy applications into clinical practice is still pending due to a lack of knowledge of the analytes' biology, lack of standards and difficulties in proving clinical utility.
Collapse
Affiliation(s)
- Corinna Keup
- Department of Gynecology and Obstetrics, University Hospital of Essen, 45147 Essen, Germany
| | | | | |
Collapse
|
29
|
Sidhu S, Kwatra KS, Kinsley PA. Androgen Receptor Expression in ER and PR Negative Breast Cancer-A Study from a Tertiary Hospital in Northern India. South Asian J Cancer 2023; 12:319-325. [PMID: 38130277 PMCID: PMC10733066 DOI: 10.1055/s-0043-1768920] [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] [Indexed: 12/23/2023] Open
Abstract
Sumeet SidhuObjectives Breast cancer is the leading cause of cancer-related deaths in women. Estrogen (ER) and progesterone receptor (PR) status and Her2 overexpression are major determinants in therapeutic decision making. Triple-negative breast cancers (TNBCs) have limited treatment options. Androgen receptor (AR) expression opens up therapeutic avenues for these patients. The aim of this article was to study the immunohistochemical expression of ARs in ER and PR Negative breast carcinomas and to correlate AR expression with various clinical, histopathological, and other immunohistochemical parameters. Materials and Methods It is a cross-sectional study including 105 ER and PR Negative cases of breast carcinoma. Clinical parameters, histopathology, and immunohistochemical expression of AR, Her2, and Ki67 were analyzed in all cases. Results AR expression was observed in 63.8% of ER and PR Negative breast cancers. In this group, AR expression was strongly associated with Her2 co-expression (89.2%) as compared to TNBCs (45.8%); p -value = 0.0002. Significant correlation was also observed between AR expression and tumor necrosis ( p -value = 0.034) and postmenopausal status ( p = 0.007). Conclusion Our study shows that significant proportion of ER and PR Negative breast carcinomas (ER- PR- Her2+ and TNBCs) show AR expression. We strongly recommend routine evaluation of all hormone receptor-negative breast carcinomas for AR status by immunohistochemistry.
Collapse
Affiliation(s)
- Sumeet Sidhu
- Christian Medical College and Hospital Ludhiana, Ludhiana, Punjab, India
| | | | | |
Collapse
|
30
|
Zhang Z, Zhang R, Li D. Molecular Biology Mechanisms and Emerging Therapeutics of Triple-Negative Breast Cancer. Biologics 2023; 17:113-128. [PMID: 37767463 PMCID: PMC10520847 DOI: 10.2147/btt.s426392] [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: 07/12/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is conventionally characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2), accounting for approximately 15-20% of all breast cancers. Compared to other molecular phenotypes, TNBC is typically associated with high malignancy and poor prognosis. Cytotoxic agents have been the mainstay of treatment for the past few decades due to the lack of definitive targets and limited therapeutic interventions. However, recent developments have demonstrated that TNBC has peculiar molecular classifications and biomarkers, which provide the possibility of evolving treatment from basic cytotoxic chemotherapy to an expanding domain of targeted therapies. This review presents a framework for understanding the current clinical experience surrounding molecular biology mechanisms in TNBC (Figure 1). Including immunotherapy, polymerase (PARP) and PI3K/AKT pathway inhibitors, antibody-drug conjugates, and androgen receptor (AR) blockade. Additionally, the role of miRNA therapeutics targeting TNBC and potential strategies targeting cancer stem cells (CSCs) are discussed and highlighted. As more and more treatments arise on the horizon, we believe that patients with TNBC will have a new sense of hope.
Collapse
Affiliation(s)
- Zhiying Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Rui Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Donghai Li
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| |
Collapse
|
31
|
Laface C, Giuliani F, Melaccio A, Pappagallo MN, Santoro AN, Perrone M, De Santis P, Guarini C, Carrozzo D, Fedele P. The Treatment Landscape of Elderly Patients with Hormone Receptor-Positive Her2 Negative Advanced Breast Cancer: Current Perspectives and Future Directions. J Clin Med 2023; 12:6012. [PMID: 37762952 PMCID: PMC10532156 DOI: 10.3390/jcm12186012] [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: 08/07/2023] [Revised: 08/26/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) in elderly women is an increasing health issue due to demographic changes. BC tends to present later and may receive less than standard treatment options. More often, BC in elderly patients is endocrine-positive (HR+). The treatment of elderly patients with metastatic BC (mBC) represents a therapeutic challenge. In recent years, the treatment landscape of patients that are HR+/Her2-negative has changed due to the introduction in clinical practice of new targeted drugs, which have improved patient outcomes. Elderly patients are a small percentage of all patients enrolled in clinical trials and, to date, there are no standardized guidelines that define the best treatment option for this patient population. This can lead to undertreatment or overtreatment, impacting patient morbidity and mortality. Geriatric Assessment tools to tailor the treatment in elderly patients are underused because they are long and difficult to apply in a busy routine clinical practice. For all these reasons, there is an urgent need to produce data about the best treatment for elderly patients with HR+ mBC. Herein, we report data from randomized clinical trials and real-world evidence on the therapeutic options for HR+ Her2-negative mBC elderly patients and explore future treatment directions.
Collapse
Affiliation(s)
- Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| | - Francesco Giuliani
- Medical Oncology, San Paolo Hospital, ASL Bari, 70123 Bari, Italy; (F.G.); (A.M.); (M.N.P.)
| | - Assunta Melaccio
- Medical Oncology, San Paolo Hospital, ASL Bari, 70123 Bari, Italy; (F.G.); (A.M.); (M.N.P.)
| | - Maria Nicla Pappagallo
- Medical Oncology, San Paolo Hospital, ASL Bari, 70123 Bari, Italy; (F.G.); (A.M.); (M.N.P.)
| | - Anna Natalizia Santoro
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| | - Martina Perrone
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| | - Pierluigi De Santis
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| | - Chiara Guarini
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| | - Daniela Carrozzo
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| | - Palma Fedele
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy (A.N.S.); (M.P.); (P.D.S.); (C.G.); (D.C.)
| |
Collapse
|
32
|
Li N, Wu J, Qi X, Yang Q, Liu R, Yang Y, Li C, Huang M, Lin M, Wang C, Cha Z, Hou X, Luo L, Ge F, Chen W. Correlation between androgen receptor expression and pathological response rate in pre-operative HER2-positive breast cancer patients. J Cancer Res Clin Oncol 2023; 149:10109-10117. [PMID: 37264265 DOI: 10.1007/s00432-023-04904-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/20/2023] [Indexed: 06/03/2023]
Abstract
PURPOSE In this study, we aimed to explore the potential significance of AR expression in HER2-positive breast cancer patients who underwent neoadjuvant targeted therapy. Specifically, we investigated the correlation between AR expression levels and pathological complete response (pCR) rates. Our objective was to determine whether there were significant differences in pCR rates among HER2-positive breast cancer patients with different levels of AR expression. METHODS We conducted a retrospective analysis of 258 HER-2 positive breast cancer patients who underwent neoadjuvant dual-blocked standard therapy (following the NCCN Guideline 2021) at three breast cancer centers in southwest China. We analyzed the clinicopathological features and pCR rates of these patients. The cut-off value for AR expression level was calculated as the median value of 70%. We used the chi-square test to investigate the correlation between AR expression level and pCR rate, as well as other clinicopathological features. RESULTS Out of the 258 patients analyzed, 154 (59.69%) achieved pCR. Based on the cut-off value of 70%, AR expression level was classified as low (AR ≤ 70%) or high (AR > 70%) expression. Our analysis revealed a significant correlation between AR expression level and pCR rate in HER2-positive breast cancer patients (P = 0.031). We also found a significant association between pCR rate and clinical stage (P = 0.033) and chemotherapy regimen (P = 0.034). Furthermore, subgroup analyses showed that the pCR rate was higher in patients with high AR expression levels compared to those with low AR expression levels. Additionally, we observed that patients with an ER/AR ratio of less than 1 had a higher pCR rate than those with an ER/AR ratio greater than 1 (P = 0.038). CONCLUSION Our study findings suggest that HER2-positive breast cancer patients with high AR expression levels may achieve higher pCR rates when treated with neoadjuvant dual-blocked therapy. Overall, our results support the idea that AR expression levels have a significant correlation with pCR rates in HER2-positive breast cancer patients receiving this particular form of treatment.
Collapse
Affiliation(s)
- Na Li
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Jingxuan Wu
- Department of Breast Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qianxi Yang
- First Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Ruimin Liu
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Yang Yang
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Chenxi Li
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Mingqing Huang
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Mengping Lin
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Chenxi Wang
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Zhuocen Cha
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Xiaochen Hou
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Lin Luo
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China
| | - Fei Ge
- Department of Breast Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| | - Wenlin Chen
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, 650118, Yunnan, China.
| |
Collapse
|
33
|
Jinna ND, Van Alsten S, Rida P, Seewaldt VL, Troester MA. Molecular features of androgen-receptor low, estrogen receptor-negative breast cancers in the Carolina breast cancer study. Breast Cancer Res Treat 2023; 201:171-181. [PMID: 37438515 PMCID: PMC10361868 DOI: 10.1007/s10549-023-07014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/19/2023] [Indexed: 07/14/2023]
Abstract
PURPOSE Androgen receptor (AR) expression is absent in 40-90% of estrogen receptor (ER)-negative breast cancers. The prognostic value of AR in ER-negative patients and therapeutic targets for patients absent in AR remains poorly explored. METHODS We used an RNA-based multigene classifier to identify AR-low and AR-high ER-negative participants in the Carolina Breast Cancer Study (CBCS; N = 669) and The Cancer Genome Atlas (TCGA; N = 237). We compared AR-defined subgroups by demographics, tumor characteristics, and established molecular signatures [PAM50 risk of recurrence (ROR), homologous recombination deficiency (HRD), and immune response]. RESULTS AR-low tumors were more prevalent among younger (RFD = + 10%, 95% CI = 4% to 16%) participants in CBCS and were associated with HER2 negativity (RFD = - 35%, 95% CI = - 44% to - 26%), higher grade (RFD = + 17%, 95% CI = 8% to 26%), and higher risk of recurrence scores (RFD = + 22%, 95% CI = 16.1% to 28%), with similar results in TCGA. The AR-low subgroup was strongly associated with HRD in CBCS (RFD = + 33.3%, 95% CI = 23.8% to 43.2%) and TCGA (RFD = + 41.5%, 95% CI = 34.0% to 48.6%). In CBCS, AR-low tumors had high adaptive immune marker expression. CONCLUSION Multigene, RNA-based low AR expression is associated with aggressive disease characteristics as well as DNA repair defects and immune phenotypes, suggesting plausible precision therapies for AR-low, ER-negative patients.
Collapse
Affiliation(s)
- Nikita D Jinna
- Department of Population Sciences, City of Hope Beckman Research Institute, Duarte, CA, 91010, USA.
| | - Sarah Van Alsten
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Padmashree Rida
- Department of Science, Rowland Hall, Salt Lake City, UT, 84102, USA
| | - Victoria L Seewaldt
- Department of Population Sciences, City of Hope Beckman Research Institute, Duarte, CA, 91010, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
34
|
Santos MMD, Frasson AL, Silva VDD, Maciel ADCA, Watte G, Werutsky G, Reinert T, Fay AP. Core Needle Biopsy Accuracy for Androgen Receptor Expression in Invasive Breast Cancer. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2023; 45:e535-e541. [PMID: 37846186 PMCID: PMC10579921 DOI: 10.1055/s-0043-1772486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 05/04/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVE Breast cancer (BC) biomarkers, such as hormone receptors expression, are crucial to guide therapy in BC patients. Antiandrogens have been studied in BC; however, limited data are available on androgen receptor (AR) expression test methodology. We aim to report the core needle biopsy (CNB) accuracy for AR expression in BC. METHODS Patients diagnosed with stage I-III invasive BC from a single institution were included. Androgen receptor expression was evaluated by immunohistochemistry (IHC) using 1 and 10% cutoff and the AR expression in surgical specimens (SS) was the gold standard. Kappa coefficients were used to evaluate the intraprocedural agreement. RESULTS A total of 72 patients were included, with a mean age of 61 years old and 84% were Luminal A or B tumors. The prevalence of AR expression in all BC samples was 87.5% using a cutoff ≥ 10% in SS. With a cutoff value ≥ 1%, CNB had an accuracy of 95.8% (Kappa value = 0.645; 95% confidence interval [CI]: 0.272-1.000; p < 0.001) and 86.1% (Kappa value = 0.365; 95% CI: 0.052-0.679; p < 0.001) when ≥ 10% cutoff was used for AR positivity. Androgen receptor expression in CNB (cutoff ≥ 1%) had a sensitivity of 98.5%, specificity of 60%, positive predictive value of 97.0%, and a negative predictive value of 76.9% in the detection of AR expression in SS. CONCLUSION Core needle biopsy has good accuracy in evaluating AR expression in BC. The accuracy of CNB decreases with higher cutoff values for AR positivity.
Collapse
Affiliation(s)
- Marcelle Morais dos Santos
- Department of Breast Surgery, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Antonio Luiz Frasson
- Department of Breast Surgery, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | - Guilherme Watte
- Department of Medical Oncology, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gustavo Werutsky
- School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tomás Reinert
- School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Oncoclínicas, Porto Alegre, RS, Brazil
| | - André Poisl Fay
- School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Oncoclínicas, Porto Alegre, RS, Brazil
| |
Collapse
|
35
|
Li L, Zheng S, Chen M, Chi W, Xue J, Wu J. The Prognostic Values of Androgen Receptor in Breast Cancer. Arch Pathol Lab Med 2023; 147:1075-1085. [PMID: 36508355 DOI: 10.5858/arpa.2021-0590-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 09/01/2023]
Abstract
CONTEXT.— Whether androgen receptor (AR) expression can predict prognosis in breast cancer is under debate. OBJECTIVE.— To analyze, retrospectively, the prognostic and treatment-predictive ability of AR status in breast cancer. DESIGN.— A total of 5765 patients diagnosed with primary invasive breast cancer without distant metastasis in the adjuvant setting were analyzed. The propensity score-matching method was used to develop a new cohort of 3978 patients (1989 patients each) in which important prognostic factors were balanced. RESULTS.— Positive AR expression is an independent prognostic factor for disease-free survival and overall survival. Estrogen receptor (ER)+ and progesterone receptor (PR)+ AR+ breast cancer patients had the longest survival, whereas ER-PR-AR- breast cancer patients had the shortest survival. The ER/PR/AR combinations could not predict the treatment effects for adjuvant trastuzumab but could be used for adjuvant chemotherapy and endocrine therapy selection. The worst survival was found in ER+PR-AR- patients receiving toremifene, ER+PR-AR+ patients receiving exemestane, ER+PR+AR- patients receiving anthracycline, and ER-PR-AR+ patients receiving taxanes. ER+PR-AR-, ER-PR-AR+, and ER-PR-AR- patients were associated with the worst survival among those who received radiotherapy and anthracycline plus taxanes. CONCLUSIONS.— AR in combination with ER and PR could predict the prognosis and treatment effects of chemotherapy, endocrine therapy, and radiotherapy in the adjuvant setting.
Collapse
Affiliation(s)
- Lun Li
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China (Li)
| | - Shuyue Zheng
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Ming Chen
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Weiru Chi
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Jingyan Xue
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Jiong Wu
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Collaborative Innovation Center for Cancer Medicine, Shanghai, China (Wu)
| |
Collapse
|
36
|
Kim H, Wisniewska K, Regner MJ, Thennavan A, Spanheimer PM, Franco HL. Single-Cell Transcriptional and Epigenetic Profiles of Male Breast Cancer Nominate Salient Cancer-Specific Enhancers. Int J Mol Sci 2023; 24:13053. [PMID: 37685859 PMCID: PMC10487538 DOI: 10.3390/ijms241713053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Male breast cancer represents about 1% of all breast cancer diagnoses and, although there are some similarities between male and female breast cancer, the paucity of data available on male breast cancer makes it difficult to establish targeted therapies. To date, most male breast cancers (MBCs) are treated according to protocols established for female breast cancer (FBC). Thus, defining the transcriptional and epigenetic landscape of MBC with improved resolution is critical for developing better avenues for therapeutic intervention. In this study, we present matched transcriptional (scRNA-seq) and epigenetic (scATAC-seq) profiles at single-cell resolution of two treatment naïve MBC tumors processed immediately after surgical resection. These data enable the detection of differentially expressed genes between male and female breast tumors across immune, stromal, and malignant cell types, to highlight several genes that may have therapeutic implications. Notably, MYC target genes and mTORC1 signaling genes were significantly upregulated in the malignant cells of MBC compared to the female counterparts. To understand how the regulatory landscape of MBC gives rise to these male-specific gene expression patterns, we leveraged the scATAC-seq data to systematically link changes in chromatin accessibility to changes in gene expression within each cell type. We observed cancer-specific rewiring of several salient enhancers and posit that these enhancers have a higher regulatory load than lineage-specific enhancers. We highlight two examples of previously unannotated cancer-cell-specific enhancers of ANXA2 and PRDX4 gene expression and show evidence for super-enhancer regulation of LAMB3 and CD47 in male breast cancer cells. Overall, this dataset annotates clinically relevant regulatory networks in male breast tumors, providing a useful resource that expands our current understanding of the gene expression programs that underlie the biology of MBC.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew J. Regner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Aatish Thennavan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Oral and Craniofacial Biomedicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Surgical Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hector L. Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
37
|
Sawant M, Wilson A, Sridaran D, Mahajan K, O'Conor CJ, Hagemann IS, Luo J, Weimholt C, Li T, Roa JC, Pandey A, Wu X, Mahajan NP. Epigenetic reprogramming of cell cycle genes by ACK1 promotes breast cancer resistance to CDK4/6 inhibitor. Oncogene 2023; 42:2263-2277. [PMID: 37330596 PMCID: PMC10348910 DOI: 10.1038/s41388-023-02747-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/16/2023] [Accepted: 06/08/2023] [Indexed: 06/19/2023]
Abstract
Hormone receptor-positive, HER2-negative advanced breast cancers exhibit high sensitivity to CDK4/6 inhibitors such as palbociclib. However, most patients inevitably develop resistance, thus identification of new actionable therapeutic targets to overcome the recurrent disease is an urgent need. Immunohistochemical studies of tissue microarray revealed increased activation of non-receptor tyrosine kinase, ACK1 (also known as TNK2) in most of the breast cancer subtypes, independent of their hormone receptor status. Chromatin immunoprecipitation studies demonstrated that the nuclear target of activated ACK1, pY88-H4 epigenetic marks, were deposited at cell cycle genes, CCNB1, CCNB2 and CDC20, which in turn initiated their efficient transcription. Pharmacological inhibition of ACK1 using its inhibitor, (R)-9b dampened CCNB1, CCNB2 and CDC20 expression, caused G2/M arrest, culminating in regression of palbociclib-resistant breast tumor growth. Further, (R)-9b suppressed expression of CXCR4 receptor, which resulted in significant impairment of metastasis of breast cancer cells to lung. Overall, our pre-clinical data identifies activated ACK1 as an oncogene that epigenetically controls the cell cycle genes governing the G2/M transition in breast cancer cells. ACK1 inhibitor, (R)-9b could be a novel therapeutic option for the breast cancer patients that have developed resistance to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Mithila Sawant
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Audrey Wilson
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Dhivya Sridaran
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Christopher J O'Conor
- Department of Pathology and Immunology, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Jingqin Luo
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Cody Weimholt
- Department of Pathology and Immunology, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA
| | - Tiandao Li
- Bioinformatics Research Core, Center of Regenerative Medicine, Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Juan Carlos Roa
- Department of Pathology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xinyan Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nupam P Mahajan
- Department of Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA.
- Division of Urologic Surgery, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St. Louis, Cancer Research Building, 660 Euclid Ave., St. Louis, MO, 63110, USA.
| |
Collapse
|
38
|
Cai GX, Kong WY, Liu Y, Zhong SY, Liu Q, Deng YF, Ye GL. Nuclear transport maintenance of USP22-AR by Importin-7 promotes breast cancer progression. Cell Death Discov 2023; 9:211. [PMID: 37391429 DOI: 10.1038/s41420-023-01525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 06/11/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
The translocation of biological macromolecules between cytoplasm and nucleus is of great significance to maintain various life processes in both normal and cancer cells. Disturbance of transport function likely leads to an unbalanced state between tumor suppressors and tumor-promoting factors. In this study, based on the unbiased analysis of protein expression differences with a mass spectrometer between human breast malignant tumors and benign hyperplastic tissues, we identified that Importin-7, a nuclear transport factor, is highly expressed in breast cancer (BC) and predicts poor outcomes. Further studies showed that Importin-7 promotes cell cycle progression and proliferation. Mechanistically, through co-immunoprecipitation, immunofluorescence, and nuclear-cytoplasmic protein separation experiments, we discovered that AR and USP22 can bind to Importin-7 as cargoes to promote BC progression. In addition, this study provides a rationale for a therapeutic strategy to restream the malignant progression of AR-positive BC by inhibiting the high expression state of Importin-7. Moreover, the knockdown of Importin-7 increased the responsiveness of BC cells to the AR signaling inhibitor, enzalutamide, suggesting that targeting Importin-7 may be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Geng-Xi Cai
- Department of Breast Surgery, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China
| | - Wei-Yao Kong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, Guangdong, China
| | - Yuan Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, Guangdong, China
| | - Shu-Yi Zhong
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, 511436, Guangzhou, Guangdong, China
| | - Qing Liu
- Department of Pathology, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China
| | - Yuan-Fei Deng
- Department of Pathology, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China.
| | - Guo-Lin Ye
- Department of Breast Surgery, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China.
| |
Collapse
|
39
|
Wróbel TM, Jørgensen FS, Pandey AV, Grudzińska A, Sharma K, Yakubu J, Björkling F. Non-steroidal CYP17A1 Inhibitors: Discovery and Assessment. J Med Chem 2023; 66:6542-6566. [PMID: 37191389 DOI: 10.1021/acs.jmedchem.3c00442] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
CYP17A1 is an enzyme that plays a major role in steroidogenesis and is critically involved in the biosynthesis of steroid hormones. Therefore, it remains an attractive target in several serious hormone-dependent cancer diseases, such as prostate cancer and breast cancer. The medicinal chemistry community has been committed to the discovery and development of CYP17A1 inhibitors for many years, particularly for the treatment of castration-resistant prostate cancer. The current Perspective reflects upon the discovery and evaluation of non-steroidal CYP17A1 inhibitors from a medicinal chemistry angle. Emphasis is placed on the structural aspects of the target, key learnings from the presented chemotypes, and design guidelines for future inhibitors.
Collapse
Affiliation(s)
- Tomasz M Wróbel
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Flemming Steen Jørgensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Amit V Pandey
- Pediatric Endocrinology, Department of Pediatrics, University Children's Hospital, Inselspital, Bern and Translational Hormone Research Program, Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Angelika Grudzińska
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
| | - Katyayani Sharma
- Pediatric Endocrinology, Department of Pediatrics, University Children's Hospital, Inselspital, Bern and Translational Hormone Research Program, Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Jibira Yakubu
- Pediatric Endocrinology, Department of Pediatrics, University Children's Hospital, Inselspital, Bern and Translational Hormone Research Program, Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
40
|
Fan KY, Chehade R, Qazi M, Moravan V, Nofech-Mozes S, Jerzak KJ. Androgen Receptor Is Expressed in the Majority of Breast Cancer Brain Metastases and Is Subtype-Dependent. Cancers (Basel) 2023; 15:2748. [PMID: 37345085 PMCID: PMC10216529 DOI: 10.3390/cancers15102748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/27/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
We aimed to evaluate the expression of the "targetable" androgen receptor (AR) in breast cancer brain metastases (BrM). An established, retrospective 57-patient cohort with metastatic breast cancer who underwent surgery for BrM at the Sunnybrook Odette Cancer Centre between 1999-2013 was studied. AR expression in BrM samples was assessed in triplicate using immunohistochemistry (IHC). AR positive status was defined as nuclear AR expression ≥ 10% by IHC using the SP107 antibody. The median age of patients was 52 years (range 32-85 years). 28 (49%) of BrM were HER2+, 17 (30%) were hormone receptor positive (HR+)/HER2-, and 12 (21%) were triple negative breast cancers (TNBCs). 56% (n = 32/57) of BrM were AR positive, and median AR expression was 20% (CI 1.6-38.3%). AR expression was different across breast cancer subtypes; AR was most frequently expressed in HER2+ (n = 21/28), followed by HR+/HER2- (n = 9/17), and lowest in TNBC (n = 2/12) BrM (p = 0.003). Patients with AR positive versus AR negative BrM had similar overall survival (12.5 vs. 7.9 months, p = 0.6), brain-specific progression-free survival (8.0 vs. 5.1 months, p = 0.95), and time from breast cancer diagnosis to BrM diagnosis (51 vs. 29 months, p = 0.16). AR is expressed in the majority of breast cancer BrM and represents a potential therapeutic target.
Collapse
Affiliation(s)
- Kevin Yijun Fan
- Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Rania Chehade
- Department of Medical Oncology, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Maleeha Qazi
- Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada
| | | | - Sharon Nofech-Mozes
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G1X5, Canada
| | - Katarzyna J. Jerzak
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON M4N3M5, Canada
| |
Collapse
|
41
|
Leo J, Dondossola E, Basham KJ, Wilson NR, Alhalabi O, Gao J, Kurnit KC, White MG, McQuade JL, Westin SN, Wellberg EA, Frigo DE. Stranger Things: New Roles and Opportunities for Androgen Receptor in Oncology Beyond Prostate Cancer. Endocrinology 2023; 164:bqad071. [PMID: 37154098 PMCID: PMC10413436 DOI: 10.1210/endocr/bqad071] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The androgen receptor (AR) is one of the oldest therapeutic targets in oncology and continues to dominate the treatment landscape for advanced prostate cancer, where nearly all treatment regimens include some form of AR modulation. In this regard, AR remains the central driver of prostate cancer cell biology. Emerging preclinical and clinical data implicate key roles for AR in additional cancer types, thereby expanding the importance of this drug target beyond prostate cancer. In this mini-review, new roles for AR in other cancer types are discussed as well as their potential for treatment with AR-targeted agents. Our understanding of these additional functions for AR in oncology expand this receptor's potential as a therapeutic target and will help guide the development of new treatment approaches.
Collapse
Affiliation(s)
- Javier Leo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Nathaniel R Wilson
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katherine C Kurnit
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Michael G White
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth A Wellberg
- Department of Pathology, Harold Hamm Diabetes Center, and Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
42
|
Zhang W, Li E, Wang L, Lehmann BD, Chen XS. Transcriptome Meta-Analysis of Triple-Negative Breast Cancer Response to Neoadjuvant Chemotherapy. Cancers (Basel) 2023; 15:2194. [PMID: 37190123 PMCID: PMC10137141 DOI: 10.3390/cancers15082194] [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: 03/04/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease with varying responses to neoadjuvant chemotherapy (NAC). The identification of biomarkers to predict NAC response and inform personalized treatment strategies is essential. In this study, we conducted large-scale gene expression meta-analyses to identify genes associated with NAC response and survival outcomes. The results showed that immune, cell cycle/mitotic, and RNA splicing-related pathways were significantly associated with favorable clinical outcomes. Furthermore, we integrated and divided the gene association results from NAC response and survival outcomes into four quadrants, which provided more insights into potential NAC response mechanisms and biomarker discovery.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Emma Li
- California Academy of Mathematics and Science, 1000 E Victoria St, Carson, CA 90747, USA
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian D. Lehmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - X. Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
43
|
Jinna N, Van Alsten S, Rida P, Seewaldt V, Troester M. Molecular Features of Androgen-Receptor Low, Estrogen Receptor-Negative Breast Cancers in the Carolina Breast Cancer Study. RESEARCH SQUARE 2023:rs.3.rs-2693555. [PMID: 36993425 PMCID: PMC10055609 DOI: 10.21203/rs.3.rs-2693555/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
PURPOSE Androgen receptor (AR) expression is absent in 40-90% of estrogen receptor (ER)-negative breast cancers. The prognostic value of AR in ER-negative patients and therapeutic targets for patients absent in AR remains poorly explored. METHODS We used an RNA-based multigene classifier to identify AR-low and AR-high ER-negative participants in the Carolina Breast Cancer Study (CBCS; n=669) and The Cancer Genome Atlas (TCGA; n=237). We compared AR-defined subgroups by demographics, tumor characteristics, and established molecular signatures [PAM50 risk of recurrence (ROR), homologous recombination deficiency (HRD), and immune response]. RESULTS AR-low tumors were more prevalent among Black (relative frequency difference (RFD) = +7%, 95% CI = 1% to 14%) and younger (RFD = +10%, 95% CI = 4% to 16%) participants in CBCS and were associated with HER2-negativity (RFD = -35%, 95% CI = -44% to -26%), higher grade (RFD = +17%, 95% CI = 8% to 26%), and higher risk of recurrence scores (RFD = +22%, 95% CI = 16.1% to 28%), with similar results in TCGA. The AR-low subgroup was strongly associated with HRD in CBCS (RFD = +33.3%, 95% CI = 23.8% to 43.2%) and TCGA (RFD = +41.5%, 95% CI = 34.0% to 48.6%). In CBCS, AR-low tumors had high adaptive immune marker expression. CONCLUSION Multigene, RNA-based low AR expression is associated with aggressive disease characteristics as well as DNA repair defects and immune phenotypes, suggesting plausible precision therapies for AR-low, ER-negative patients.
Collapse
Affiliation(s)
| | | | | | | | - Melissa Troester
- UNC-Chapel Hill: The University of North Carolina at Chapel Hill
| |
Collapse
|
44
|
Wei L, Gao H, Yu J, Zhang H, Nguyen TTL, Gu Y, Passow MR, Carter JM, Qin B, Boughey JC, Goetz MP, Weinshilboum RM, Ingle JN, Wang L. Pharmacological Targeting of Androgen Receptor Elicits Context-Specific Effects in Estrogen Receptor-Positive Breast Cancer. Cancer Res 2023; 83:456-470. [PMID: 36469363 PMCID: PMC9896025 DOI: 10.1158/0008-5472.can-22-1016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Androgen receptor (AR) is expressed in 80% to 90% of estrogen receptor α-positive (ER+) breast cancers. Accumulated evidence has shown that AR is a tumor suppressor and that its expression is associated with improved prognosis in ER+ breast cancer. However, both a selective AR agonist (RAD140) and an AR inhibitor (enzalutamide, ENZ) have shown a therapeutic effect on ER+ breast cancer, so the potential for clinical application of AR-targeting therapy for ER+ breast cancer is still in dispute. In this study, we evaluated the efficacy of ENZ and RAD140 in vivo and in vitro in AR+/ER+ breast cancer models, characterizing the relationship of AR and ER levels to response to AR-targeting drugs and investigating the alterations of global gene expression and chromatin binding of AR and ERα after ENZ treatment. In the AR-low setting, ENZ directly functioned as an ERα antagonist. Cell growth inhibition by ENZ in breast cancer with low AR expression was independent of AR and instead dependent on ER. In AR-high breast cancer models, AR repressed ERα signaling and ENZ promoted ERα signaling by antagonizing AR. In contrast, RAD140 activated AR signaling and suppressed AR-high tumor growth by deregulating ERα expression and blocking ERα function. Overall, analysis of the dynamic efficacies and outcomes of AR agonist, and antagonist in the presence of different AR and ERα levels reveals regulators of response and supports the clinical investigation of ENZ in selected ER+ tumors with a low AR/ER ratio and AR agonists in tumors with a high AR/ER ratio. SIGNIFICANCE The ratio of androgen receptor to estrogen receptor in breast cancer dictates the response to AR-targeted therapies, providing guidelines for developing AR-directed treatment strategies for patients with breast cancer.
Collapse
Affiliation(s)
- Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Huan Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Thanh Thanh L. Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Yayun Gu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Marie R. Passow
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Bo Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Matthew P. Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - James N. Ingle
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
45
|
Elbalka SS, Metwally IH, Hassan A, Eladl AE, Shoman AM, Jawad M, Shahda E, Abdelkhalek M. Prognostic value of androgen receptor expression in different molecular types of breast cancer in women. Breast Dis 2023; 41:495-502. [PMID: 36641656 DOI: 10.3233/bd-220037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Breast cancer is a common women's disease. Usually, oestrogen is blamed in the aetiology and correlated with the prognosis; however, androgens are recently raising concern about its role in the breast cancer treatment and prognosis. METHODS In this study we retrieved archival paraffin blocks of breast cancer patients and stained it for androgen. Thereafter, we compared clinico-epidemiologic parameters, histopathology, neoadjuvant response and recurrence rate and pattern among patients with and without androgen receptor (AR) expression. RESULTS In total, 119 patients fulfilled enrolment criteria; AR expression were present in 77.3% of the patients. AR expression was associated with less grade III (6.8% versus 36.4%), and less triple negative (6.2% versus 25%), but similar overall recurrence rate (25% versus 22.2%). However, distant recurrence was significantly higher in androgen positive patients (91.3% versus 33.3% of all recurrences). CONCLUSION Androgen expression appears to be common among breast cancer, but with no clear implication in tumour aggressiveness or effect on the rate of recurrence. However, being commonly associated with distant spread may have an impact on survival of the patients.
Collapse
Affiliation(s)
- Saleh S Elbalka
- Surgical Oncology Department, Oncology Center Mansoura University (OCMU), Mansoura, Egypt
| | - Islam H Metwally
- Surgical Oncology Department, Oncology Center Mansoura University (OCMU), Mansoura, Egypt
| | - Amany Hassan
- Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed E Eladl
- Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed M Shoman
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Jawad
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Shahda
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Abdelkhalek
- Surgical Oncology Department, Oncology Center Mansoura University (OCMU), Mansoura, Egypt
| |
Collapse
|
46
|
Falato C, Schettini F, Pascual T, Brasó-Maristany F, Prat A. Clinical implications of the intrinsic molecular subtypes in hormone receptor-positive and HER2-negative metastatic breast cancer. Cancer Treat Rev 2023; 112:102496. [PMID: 36563600 DOI: 10.1016/j.ctrv.2022.102496] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
Traditionally, the classification of breast cancer relies on the expression of immunohistochemical (IHC) biomarkers readily available in clinical practice. Using highly standardized and reproducible assays across patient cohorts, intrinsic molecular subtypes of breast cancer - also called "intrinsic subtypes" (IS) - have been identified based on the expression of 50 genes. Although IHC-based subgroups and IS moderately correlate to each other, they are not superimposable. In fact, non-luminal biology has been detected in a substantial proportion (5-20%) of hormone receptor-positive (HoR+) tumors, has prognostic value, and identifies reduced and increased sensitivity to endocrine therapy and chemotherapy, respectively. During tumor progression, a shift toward a non-luminal estrogen-independent and more aggressive phenotype has been demonstrated. Intrinsic genomic instability and cell plasticity, alone or combined with external constraints deriving from treatment selective pressure or interplay with the tumor microenvironment, may represent the determinants of such biological diversity between primary and metastatic disease, and during metastatic tumor evolution. In this review, we describe the distribution and the clinical behavior of IS as the disease progresses, focusing on HoR+/HER2-negative advanced breast cancer. In addition, we provide an overview of the ongoing clinical trials aiming to validate the predictive and prognostic value of IS towards their incorporation into routine care.
Collapse
Affiliation(s)
- Claudette Falato
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cancer Research Group, Barcelona, Spain; Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden.
| | - Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain; Faculty of Medicine, University of Barcelona, Barcelona, Spain.
| | - Tomás Pascual
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cancer Research Group, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain.
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Faculty of Medicine, University of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain.
| |
Collapse
|
47
|
Kolyvas EA, Caldas C, Kelly K, Ahmad SS. Androgen receptor function and targeted therapeutics across breast cancer subtypes. Breast Cancer Res 2022; 24:79. [PMID: 36376977 PMCID: PMC9664788 DOI: 10.1186/s13058-022-01574-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant progress in breast cancer (BC) therapy, it is globally the most commonly diagnosed cancer and leads to the death of over 650,000 women annually. Androgen receptor (AR) is emerging as a potential new therapeutic target in BC. While the role of AR is well established in prostate cancer (PCa), its function in BC remains incompletely understood. Emerging data show that AR's role in BC is dependent on several factors including, but not limited to, disease subtype, tumour microenvironment, and levels of circulating oestrogens and androgens. While targeting AR in PCa is becoming increasingly effective, these advances have yet to make any significant impact on the care of BC patients. However, this approach is increasingly being evaluated in BC and it is clear that improvements in our understanding of AR's role in BC will increase the likelihood of success for AR-targeted therapies. This review summarizes our current understanding of the function of AR across BC subtypes. We highlight limitations in our current knowledge and demonstrate the importance of categorizing BC subtypes effectively, in relation to determining AR activity. Further, we describe the current state of the art regarding AR-targeted approaches for BC as monotherapy or in combination with radiotherapy.
Collapse
Affiliation(s)
- Emily A Kolyvas
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- NIH-Oxford-Cambridge Scholars Program, Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Breast Cancer Programme, CRUK Cambridge Centre, Cambridge, CB2 0RE, UK
- Cambridge Breast Cancer Research Unit, NIHR Cambridge Biomedical Research Centre and Cambridge Experimental Cancer Medicine Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Saif S Ahmad
- Cancer Research UK Cambridge Institute, Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, CB2 0RE, UK.
- Department of Oncology, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK.
| |
Collapse
|
48
|
Walsh EM, Gucalp A, Patil S, Edelweiss M, Ross DS, Razavi P, Modi S, Iyengar NM, Sanford R, Troso-Sandoval T, Gorsky M, Bromberg J, Drullinsky P, Lake D, Wong S, DeFusco PA, Lamparella N, Gupta R, Tabassum T, Boyle LA, Arumov A, Traina TA. Adjuvant enzalutamide for the treatment of early-stage androgen-receptor positive, triple-negative breast cancer: a feasibility study. Breast Cancer Res Treat 2022; 195:341-351. [PMID: 35986801 PMCID: PMC10506398 DOI: 10.1007/s10549-022-06669-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/29/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Chemotherapy with or without immunotherapy remains the mainstay of treatment for triple-negative breast cancer (TNBC). A subset of TNBCs express the androgen receptor (AR), representing a potential new therapeutic target. This study assessed the feasibility of adjuvant enzalutamide, an AR antagonist, in early-stage, AR-positive (AR +) TNBC. METHODS This study was a single-arm, open-label, multicenter trial in which patients with stage I-III, AR ≥ 1% TNBC who had completed standard-of-care therapy were treated with enzalutamide 160 mg/day orally for 1 year. The primary objective of this study was to evaluate the feasibility of 1 year of adjuvant enzalutamide, defined as the treatment discontinuation rate of enzalutamide due to toxicity, withdrawal of consent, or other events related to tolerability. Secondary endpoints included disease-free survival (DFS), overall survival (OS), safety, and genomic features of recurrent tumors. RESULTS Fifty patients were enrolled in this study. Thirty-five patients completed 1 year of therapy, thereby meeting the prespecified trial endpoint for feasibility. Thirty-two patients elected to continue with an optional second year of treatment. Grade ≥ 3 treatment-related adverse events were uncommon. The 1-year, 2-year, and 3-year DFS were 94%, 92% , and 80%, respectively. Median OS has not been reached. CONCLUSION This clinical trial demonstrates that adjuvant enzalutamide is a feasible and well-tolerated regimen in patients with an early-stage AR + TNBC. Randomized trials in the metastatic setting may inform patient selection through biomarker development; longer follow-up is needed to determine the effect of anti-androgens on DFS and OS in this patient population.
Collapse
Affiliation(s)
- Elaine M Walsh
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA.
| | - Ayca Gucalp
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Sujata Patil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcia Edelweiss
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Neil M Iyengar
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Rachel Sanford
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Tiffany Troso-Sandoval
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Mila Gorsky
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Jacqueline Bromberg
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Pamela Drullinsky
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Diana Lake
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Serena Wong
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | | | | | - Ranja Gupta
- Lehigh Valley Health Network Cancer Institute, Allentown, PA, USA
| | - Tasmila Tabassum
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Leigh Ann Boyle
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Artavazd Arumov
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Tiffany A Traina
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| |
Collapse
|
49
|
Long M, You C, Song Q, Hu LXJ, Guo Z, Yao Q, Hou W, Sun W, Liang B, Zhou X, Liu Y, Hu T. AR Expression Correlates with Distinctive Clinicopathological and Genomic Features in Breast Cancer Regardless of ESR1 Expression Status. Int J Mol Sci 2022; 23:ijms231911468. [PMID: 36232774 PMCID: PMC9570294 DOI: 10.3390/ijms231911468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Androgen receptor (AR) expression is frequently observed in breast cancer, but its association with estrogen receptor (ER) expression in breast cancer remains unclear. This study analyzed the clinicopathological and molecular features associated with AR negativity in both ER-positive and ER-negative breast cancer, trying to elucidate the molecular correlation between AR and ER. Our results showed that AR negativity was associated with different clinicopathological characteristics and molecular features in ER-positive and ER-negative breast cancer. Moreover, AR-positive breast cancer has better clinicopathological features than AR-negative breast cancer, especially in the ER-negative subtype. These results suggest that the role of AR in ER-negative breast cancer is distinctive from that in ER-positive breast cancer.
Collapse
Affiliation(s)
- Mengping Long
- Department of Pathology, Peking University Cancer Hospital, Beijing 100083, China
| | - Chong You
- Chongqing Research Institute of Big Data, Peking University, Chongqing 401121, China
| | - Qianqian Song
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Lina X. J. Hu
- Department of Pathology, Alaska Native Medical Center, Anchorage, AK 99501, USA
| | - Zhaorong Guo
- Department of Breast Center, Peking University Cancer Hospital, Beijing 100083, China
| | - Qian Yao
- Department of Pathology, Peking University Cancer Hospital, Beijing 100083, China
| | - Wei Hou
- Department of Pathology, Peking University Cancer Hospital, Beijing 100083, China
| | - Wei Sun
- Department of Pathology, Peking University Cancer Hospital, Beijing 100083, China
| | - Baosheng Liang
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Xiaohua Zhou
- Chongqing Research Institute of Big Data, Peking University, Chongqing 401121, China
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Yiqiang Liu
- Department of Pathology, Peking University Cancer Hospital, Beijing 100083, China
- Correspondence: (Y.L.); (T.H.)
| | - Taobo Hu
- Chongqing Research Institute of Big Data, Peking University, Chongqing 401121, China
- Department of Breast Surgery, Peking University People’s Hospital, Beijing 100044, China
- Correspondence: (Y.L.); (T.H.)
| |
Collapse
|
50
|
Racial Disparity in Quadruple Negative Breast Cancer: Aggressive Biology and Potential Therapeutic Targeting and Prevention. Cancers (Basel) 2022; 14:cancers14184484. [PMID: 36139643 PMCID: PMC9497140 DOI: 10.3390/cancers14184484] [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: 06/30/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Quadruple negative breast cancer (QNBC), a subgroup of triple negative BC, has emerged as a highly aggressive BC subtype that disproportionately afflicts and impacts Black/African-American (AA) women. In this article, we review molecular distinctions in Black/AA and White/European-American (EA) QNBC biology as well as address potential non-genetic risk factors that could be underlying this racially disparate burden. We aim to provide deeper insight and provide a framework for novel discovery of actionable therapeutic targets and identify lifestyle changes to improve outcomes for Black/AA QNBC patients. Abstract Black/African-American (AA) women, relative to their White/European-American (EA) counterparts, experience disproportionately high breast cancer mortality. Central to this survival disparity, Black/AA women have an unequal burden of aggressive breast cancer subtypes, such as triple-negative breast cancer (ER/PR-, HER2-wild type; TNBC). While TNBC has been well characterized, recent studies have identified a highly aggressive androgen receptor (AR)-negative subtype of TNBC, quadruple-negative breast cancer (ER/PR-, HER2-wildtype, AR-; QNBC). Similar to TNBC, QNBC disproportionately impacts Black/AA women and likely plays an important role in the breast cancer survival disparities experienced by Black/AA women. Here, we discuss the racial disparities of QNBC and molecular signaling pathways that may contribute to the aggressive biology of QNBC in Black/AA women. Our immediate goal is to spotlight potential prevention and therapeutic targets for Black/AA QNBC; ultimately our goal is to provide greater insight into reducing the breast cancer survival burden experienced by Black/AA women.
Collapse
|