1
|
Das GM, Oturkar CC, Menon V. Interaction between Estrogen Receptors and p53: A Broader Role for Tamoxifen? Endocrinology 2025; 166:bqaf020. [PMID: 39891710 PMCID: PMC11837209 DOI: 10.1210/endocr/bqaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Tamoxifen is one of the most widely used anticancer drugs in the world. It is a safe drug with generally well-tolerated side effects and has been prescribed for the treatment of early-stage and advanced-stage or metastatic estrogen receptor α (ERα/ESR1)-positive breast cancer. Tamoxifen therapy also provides a 38% reduction of the risk of developing breast cancer in women at high risk. With the advent of newer medications targeting ERα-positive breast cancer, tamoxifen is now mainly used as adjuvant therapy for lower-risk premenopausal breast cancer and cancer prevention. It is widely accepted that tamoxifen as a selective estrogen receptor modulator exerts its therapeutic effect by competitively binding to ERα, leading to the recruitment of corepressors and inhibition of transcription of genes involved in the proliferation of breast cancer epithelium. As such, expression of ERα in breast tumors has been considered necessary for tumors to be responsive to tamoxifen therapy. However, ERα-independent effects of tamoxifen in various in vitro and in vivo contexts have been reported over the years. Importantly, the recent discovery that ERα and estrogen receptor β (ERβ/ESR2) can bind tumor suppressor protein p53 with functional consequences has provided new insights into the mechanisms underlying response to tamoxifen therapy and resistance. Furthermore, these findings have paved the way for broadening the use of tamoxifen by potentially repurposing it to treat triple negative (negative for ERα, human epidermal growth factor receptor 2, and progesterone receptor) breast cancer. Herein, we summarize these developments and discuss their mechanistic underpinnings and clinical implications.
Collapse
Affiliation(s)
- Gokul M Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chetan C Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Vishnu Menon
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
2
|
Bhutta ZA, Choi KC. Canine mammary tumors as a promising adjunct preclinical model for human breast cancer research: similarities, opportunities, and challenges. Arch Pharm Res 2025; 48:43-61. [PMID: 39752109 DOI: 10.1007/s12272-024-01524-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: 07/23/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Despite significant progress in the field of human breast cancer research and treatment, there is a consistent increase in the incidence rate of 0.5 percent annually, posing challenges in the development of effective novel therapeutic strategies. The failure rate of drugs in clinical trials stands at approximately 95%, primarily attributed to the limitations and lack of reliability of existing preclinical models, such as mice, which do not mimic human tumor biology. This article examines the potential utility of canine mammary tumors as an adjunct preclinical model for investigating human breast cancer. Given the numerous similarities between canine and human breast cancer, canines present a promising alternative model. The discussion delves into the intricate molecular and clinical aspects of human breast cancer and canine mammary tumors, shedding light on the tumors' molecular profiles, identifying specific molecular markers, and the application of radiological imaging modalities. Furthermore, the manuscript addresses the current constraints of preclinical cancer studies, the benefits of using canines as models, and the obstacles linked to the canine mammary tumors model. By concentrating on these elements, this review aims to highlight the viability of canine models in enhancing our understanding and management of human breast cancer.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
3
|
Marzouk M, Greco S, Gbahou F, Küblbeck J, Labani N, Jockers R, Holzgrabe U, Wiesmüller L, Zlotos DP. Cancer Cells Show Higher Sensitivity to Melatonin-Tamoxifen Drug Conjugates than to Combination of Melatonin and Tamoxifen. ACS OMEGA 2024; 9:47857-47871. [PMID: 39651096 PMCID: PMC11618438 DOI: 10.1021/acsomega.4c08881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 12/11/2024]
Abstract
Drug conjugates of tamoxifen and melatonin linked through the amide side chain of melatonin (4a,4b) were reported as promising agents for future treatment of breast cancer, possibly reversing the adverse effects of tamoxifen. Here, we report the synthesis and pharmacological evaluation of a novel series of anticancer drug conjugates linking melatonin with tamoxifen through polymethylene spacers through the ether oxygen of melatonin (16a-c, 19a-c, 21) and compare them to the previously reported amide-linked analogues 4a and 4b. All hybrid ligands are antagonists of estrogen receptor alpha and agonists of the melatonin MT1 receptor with variable potencies. Several drug conjugates including the (CH2)4-linked analogues 4a and 16a and the (CH2)6-linked compound 16c showed higher potency to inhibit cell viability than the combination of melatonin and tamoxifen on at least one cancer cell line including MCF-7, MDA-MB-231, and HT-1080.
Collapse
Affiliation(s)
- Mohamed
Akmal Marzouk
- Institute
of Pharmacy and Food Chemistry, University
of Würzburg, Würzburg 97074, Germany
| | - Sara Greco
- Department
of Obstetrics and Gynecology, Ulm University, Prittwitzstrasse 43, Ulm 89075, Germany
| | - Florence Gbahou
- Université
Paris Cité, Institut Cochin, INSERM, CNRS, Paris 75014, France
| | - Jenni Küblbeck
- A.I. Virtanen
Institute for Molecular Sciences, University
of Eastern Finland, P.O. Box 1627, Kuopio FI-70210, Finland
- School of
Pharmacy, University of Eastern Finland, P.O. Box 1627, Kuopio FI-70210, Finland
| | - Nedjma Labani
- Université
Paris Cité, Institut Cochin, INSERM, CNRS, Paris 75014, France
| | - Ralf Jockers
- Université
Paris Cité, Institut Cochin, INSERM, CNRS, Paris 75014, France
| | - Ulrike Holzgrabe
- Institute
of Pharmacy and Food Chemistry, University
of Würzburg, Würzburg 97074, Germany
| | - Lisa Wiesmüller
- Department
of Obstetrics and Gynecology, Ulm University, Prittwitzstrasse 43, Ulm 89075, Germany
| | - Darius P. Zlotos
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, The German University in Cairo, New Cairo City, Cairo 11835, Egypt
| |
Collapse
|
4
|
Rangel N, Sánchez IL, Valbuena DS, Rondón-Lagos M. ZNF217 Gene Copy Number as a Marker of Response to Standard Therapy Drugs According to ERα Status in Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:127-139. [PMID: 38505863 PMCID: PMC10950081 DOI: 10.2147/bctt.s445753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/24/2024] [Indexed: 03/21/2024]
Abstract
Purpose The therapeutic decision for the management of breast cancer (BC) patients is based on the evaluation of prognostic factors alongside clinical and pathological parameters. Despite the use of standard biomarkers, response and resistance to therapy represent a challenge for clinicians. Among the new potential biomarkers for BC the ZNF217 gene have gained importance in recent years. However, while associations between ZNF217 gene copy number and clinicopathological characteristics have been established, its correlation with treatment response remains unclear. Patients and Methods This study aimed to evaluate the ZNF217 gene copy number and establish its associations with treatment response in estrogen receptor positive (ERα+) and ERα negative (ERα-) BC cell lines. In addition, a validation of the relationship between ZNF217 gene copy number and its prognostic value was performed using datasets of BC patients retrieved from the cBioPortal public database. Results Our data show that in ERα+ cells, ZNF217 gene copy number increase (amplification), while cell proliferation decreases in response to standard drug treatments. In contrast, both ZNF217 gene copy number (gain) and cell proliferation increases in response to standard drug treatments in ERα- cells. The results obtained align with findings from the cBioPortal database analysis, demonstrating that ERα+/HER2- low proliferation patients, exhibiting ZNF217 gene amplification or gain, have a significantly higher survival probability after treatment, compared to ERα-/HER2- and HER2+ patients. Conclusion Our results suggest that in ERα+ BC cells, ZNF217 gene amplification could be indicative of a favorable response, while in ERα- BC cells, ZNF217 gene gain could be postulated as a potential predictor of treatment resistance. A broader understanding of the role of ZNF217 gene in treatment response, together with prospective studies in BC patients, could contribute to confirming our data, as well as optimizing existing treatments and exploring novel approaches to improve overall cancer treatment outcomes.
Collapse
Affiliation(s)
- Nelson Rangel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, 110231, Colombia
| | - Iris Lorena Sánchez
- School of Biological Sciences, Universidad Pedagógica Y Tecnológica de Colombia, Tunja, 150003, Colombia
| | - Duván Sebastián Valbuena
- School of Biological Sciences, Universidad Pedagógica Y Tecnológica de Colombia, Tunja, 150003, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica Y Tecnológica de Colombia, Tunja, 150003, Colombia
| |
Collapse
|
5
|
Kirkby M, Popatia AM, Lavoie JR, Wang L. The Potential of Hormonal Therapies for Treatment of Triple-Negative Breast Cancer. Cancers (Basel) 2023; 15:4702. [PMID: 37835396 PMCID: PMC10571841 DOI: 10.3390/cancers15194702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is considered one of the most aggressive forms of breast cancer with poor survival rates compared to other breast cancer subtypes. TNBC is characterized by the absence of the estrogen receptor alpha, progesterone receptor, and the human epidermal growth factor receptor 2, limiting those viable treatment options available to patients with other breast cancer subtypes. Furthermore, due to the particularly high heterogeneity of TNBC, conventional treatments such as chemotherapy are not universally effective, leading to drug resistance and intolerable side effects. Thus, there is a pressing need to discover new therapies beneficial to TNBC patients. This review highlights current findings regarding the roles of three steroid hormone receptors, estrogen receptor beta, the androgen receptor, and the glucocorticoid receptor, in the progression of TNBC. In addition, we discussed several ongoing and completed clinical trials targeting these hormone receptors in TNBC patients.
Collapse
Affiliation(s)
- Melanie Kirkby
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (M.K.); (A.M.P.)
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- The Centre for Infection, Immunity, and Inflammation (CI3), University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Alyanna M. Popatia
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (M.K.); (A.M.P.)
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- The Centre for Infection, Immunity, and Inflammation (CI3), University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Jessie R. Lavoie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (M.K.); (A.M.P.)
- Centre for Oncology, Radiopharmaceuticals and Research, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (M.K.); (A.M.P.)
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- The Centre for Infection, Immunity, and Inflammation (CI3), University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
6
|
Takano EA, Younes MM, Meehan K, Spalding L, Yan M, Allan P, Fox SB, Redfern A, Clouston D, Giles GG, Christie EL, Anderson RL, Zethoven M, Phillips KA, Gorringe K, Britt KL. Estrogen receptor beta expression in triple negative breast cancers is not associated with recurrence or survival. BMC Cancer 2023; 23:459. [PMID: 37208678 DOI: 10.1186/s12885-023-10795-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/31/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Triple negative BCa (TNBC) is defined by a lack of expression of estrogen (ERα), progesterone (PgR) receptors and human epidermal growth factor receptor 2 (HER2) as assessed by protein expression and/or gene amplification. It makes up ~ 15% of all BCa and often has a poor prognosis. TNBC is not treated with endocrine therapies as ERα and PR negative tumors in general do not show benefit. However, a small fraction of the true TNBC tumors do show tamoxifen sensitivity, with those expressing the most common isoform of ERβ1 having the most benefit. Recently, the antibodies commonly used to assess ERβ1 in TNBC have been found to lack specificity, which calls into question available data regarding the proportion of TNBC that express ERβ1 and any relationship to clinical outcome. METHODS To confirm the true frequency of ERβ1 in TNBC we performed robust ERβ1 immunohistochemistry using the specific antibody CWK-F12 ERβ1 on 156 primary TNBC cancers from patients with a median of 78 months (range 0.2-155 months) follow up. RESULTS We found that high expression of ERβ1 was not associated with increased recurrence or survival when assessed as percentage of ERβ1 positive tumor cells or as Allred > 5. In contrast, the non-specific PPG5-10 antibody did show an association with recurrence and survival. CONCLUSIONS Our data indicate that ERβ1 expression in TNBC tumours does not associate with prognosis.
Collapse
Affiliation(s)
- Elena A Takano
- Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Melissa M Younes
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, Research Division 305 Grattan St, Melbourne, VIC, 3000, Australia
| | - Katie Meehan
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- The University of Western Australia (M504), 35 Stirling Highway, Perth, 6009, Australia
| | - Lisa Spalding
- The University of Western Australia (M504), 35 Stirling Highway, Perth, 6009, Australia
| | - Max Yan
- South Eastern Area Laboratory Services, Randwick, NSW, Australia
| | - Prue Allan
- Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Stephen B Fox
- Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Andy Redfern
- The University of Western Australia (M504), 35 Stirling Highway, Perth, 6009, Australia
| | - David Clouston
- TissuPath, 32 Ricketts Rd, Mount Waverley, VIC, 3149, Australia
| | - Graham G Giles
- 7a Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, 3004, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, University of Melbourne, Parkville, VIC, 3012, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, 3168, Australia
| | - Elizabeth L Christie
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Peter MacCallum Cancer Centre Melbourne, Victoria, 3000, Australia
| | - Robin L Anderson
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Magnus Zethoven
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Peter Mac, Bioinformatics Core Facility, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Kelly-Anne Phillips
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| | - Kylie Gorringe
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Precision Cancer Medicine Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, Australia
| | - Kara L Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, Research Division 305 Grattan St, Melbourne, VIC, 3000, Australia.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
7
|
Scarpetti L, Oturkar CC, Juric D, Shellock M, Malvarosa G, Post K, Isakoff S, Wang N, Nahed B, Oh K, Das GM, Bardia A. Therapeutic Role of Tamoxifen for Triple-Negative Breast Cancer: Leveraging the Interaction Between ERβ and Mutant p53. Oncologist 2023; 28:358-363. [PMID: 36772966 PMCID: PMC10078911 DOI: 10.1093/oncolo/oyac281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/30/2022] [Indexed: 02/12/2023] Open
Abstract
The absence of effective therapeutic targets and aggressive nature of triple-negative breast cancer (TNBC) renders this disease subset difficult to treat. Although estrogen receptor beta (ERβ) is expressed in TNBC, studies on its functional role have yielded inconsistent results. However, recently, our preclinical studies, along with other observations, have shown the potential therapeutic utility of ERβ in the context of mutant p53 expression. The current case study examines the efficacy of the selective estrogen receptor modulator tamoxifen in p53-mutant TNBC with brain metastases. Significant increase in ERβ protein expression and anti-proliferative interaction between mutant p53 and ERβ were observed after cessation of tamoxifen therapy, with significant regression of brain metastases. This case study provides supporting evidence for the use of tamoxifen in p53-mutant, ERβ+TNBC, especially in the setting of brain metastasis.
Collapse
Affiliation(s)
- Lauren Scarpetti
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Dejan Juric
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Maria Shellock
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Giuliana Malvarosa
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kathryn Post
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Steven Isakoff
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Nancy Wang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Brian Nahed
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Oh
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Gokul M Das
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Blum KM, Mirhaidari GJM, Zbinden JC, Breuer C, Barker JC. Tamoxifen reduces silicone implant capsule formation in a mouse model. FASEB Bioadv 2022; 4:638-647. [PMID: 36238364 PMCID: PMC9536088 DOI: 10.1096/fba.2022-00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/19/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Capsular contracture as a result of the foreign body response (FBR) is a common issue after implant-based breast reconstruction, affecting up to 20% of patients. New evidence suggests that tamoxifen may mitigate the FBR. C57BL/6 female mice were treated with daily tamoxifen or control injections and implanted with bilateral silicone implants in the submammary glandular plane. Implants were removed en bloc after 2 weeks and the implant capsules were evaluated histologically. Tamoxifen treatment decreased capsule thickness, decreased the number of αSMA+ cells (477 ± 156 cells/mm control vs 295 ± 121 cells/mm tamoxifen, p = 0.005 unpaired t test), and decreased CD31+ cells (173.9 ± 96.1 cells/mm2 control vs 106.3 ± 51.8 cells/mm2 tamoxifen, p = 0.043 unpaired t test). There were similar amounts of pro- and anti-inflammatory macrophages (iNOS 336.1 ± 226.3 cells/mm control vs 290.6 ± 104.2 cells/mm tamoxifen, p > 0.999 Mann-Whitney test and CD163 136.6 ± 76.4 cells/mm control vs 94.1 ± 45.9 cells/mm tamoxifen, p = 0.108 unpaired t test). Tamoxifen treatment in the mouse silicone breast implant model decreased capsule formation through modulation of myofibroblasts, neovascularization, and collagen deposition. Tamoxifen may be useful for reducing or preventing capsule formation in clinical breast implantations.
Collapse
Affiliation(s)
- Kevin M. Blum
- Center for Regenerative Medicine, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Gabriel J. M. Mirhaidari
- Center for Regenerative Medicine, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
- Biological Sciences Graduate ProgramThe Ohio State UniversityColumbusOhioUSA
| | - Jacob C. Zbinden
- Center for Regenerative Medicine, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Christopher K. Breuer
- Center for Regenerative Medicine, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
| | - Jenny C. Barker
- Center for Regenerative Medicine, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
- Department of Plastic and Reconstructive Surgery, Wexner Medical CenterThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
9
|
Nance RL, Sajib AM, Smith BF. Canine models of human cancer: Bridging the gap to improve precision medicine. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:67-99. [PMID: 35595353 DOI: 10.1016/bs.pmbts.2021.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dogs are remarkable, adaptable, and dependable creatures that have evolved alongside humans while contributing tremendously to our survival. Our canine companions share many similarities to human disease, particularly cancer. With the advancement of next-generation sequencing technology, we are beginning to unravel the complexity of cancer and the vast intra- and intertumoral heterogeneity that makes treatment difficult. Consequently, precision medicine has emerged as a therapeutic approach to improve patient survival by evaluating and classifying an individual tumor's molecular profile. Many canine and human cancers share striking similarities in terms of genotypic, phenotypic, clinical, and histological presentations. Dogs are superior to rodent models of cancer because they are a naturally heterogeneous population in which tumors occur spontaneously, are exposed to similar environmental conditions, and show more similarities in key modulators of tumorigenesis and clinical response, including the immune system, drug metabolism, and gut microbiome. In this chapter, we will explore various canine models of human cancers and emphasize the dog's critical role in advancing precision medicine and improving the survival of both man and man's best friend.
Collapse
Affiliation(s)
- Rebecca L Nance
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States; Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Abdul Mohin Sajib
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Bruce F Smith
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States; Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL, United States.
| |
Collapse
|
10
|
Calhoun S, Duan L, Maki CG. Acetyl-CoA synthetases ACSS1 and ACSS2 are 4-hydroxytamoxifen responsive factors that promote survival in tamoxifen treated and estrogen deprived cells. Transl Oncol 2022; 19:101386. [PMID: 35263700 PMCID: PMC8904238 DOI: 10.1016/j.tranon.2022.101386] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/22/2022] Open
Affiliation(s)
- Sarah Calhoun
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac Suite 507, Chicago, IL 60612, USA
| | - Lei Duan
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac Suite 507, Chicago, IL 60612, USA
| | - Carl G Maki
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac Suite 507, Chicago, IL 60612, USA.
| |
Collapse
|
11
|
ERβ Isoforms Have Differential Clinical Significance in Breast Cancer Subtypes and Subgroups. Curr Issues Mol Biol 2022; 44:1564-1586. [PMID: 35723365 PMCID: PMC9164084 DOI: 10.3390/cimb44040107] [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: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
ERβ, an ER subtype first identified in 1996, is highly expressed in different types of BCa including ERα-negative BCa and TNBC. Many studies on ERβ expression investigated mostly on ERβ1 protein expression in ERα-positive and ERα-negative BCa combined. The results are conflicting. This may be due to the complexity of ERβ isoforms, subject heterogeneity, and various study designs targeting different ERβ isoforms and either ERβ protein or mRNA expression, as well as to the lack of a standardized testing protocol. Herein, we simultaneously investigated both mRNA and protein expression of ERβ isoforms 1, 2, and 5 in different BCa subtypes and clinical characteristics. Patient samples (138) and breast cancer cell lines (BCC) reflecting different types of BCa were tested for ERα and ERβ mRNA expression using quantitative real-time PCR, as well as for protein expression of ERα, ERβ1, ERβ2, and ERβ5 isoforms, PR, HER2/neu, Ki-67, CK 5/6, and p53 using immunohistochemistry. Associations of ERβ isoform expression with clinical characteristics and overall survival (OS) were analyzed. ERβ1, 2, and 5 isoforms are differentially expressed in different BCa subtypes including ERα-negative and TNBC. Each ERβ isoform seemingly plays a distinct role and is associated with clinical tumor characteristics and patient outcomes. ERβ isoform expression is significantly associated with >15% Ki-67 positivity and poor prognostic markers, and it predicts poorer OS, mostly in the subgroups. High ERβ2 and 5 isoform expression in ERα-negative BCa and TNBC is predictive of poor OS. Further investigation of ERβ isoforms in a larger cohort of BCa subgroups is needed to evaluate the role of ERβ for the potential usefulness of ERβ as a prognostic and predictive marker and for therapeutic use. The inconsistent outcomes of ERβ isoform mRNA or protein expression in many studies suggest that the standardization of ERβ testing would facilitate the use of ERβ in a clinical setting.
Collapse
|
12
|
Dalal H, Dahlgren M, Gladchuk S, Brueffer C, Gruvberger-Saal SK, Saal LH. Clinical associations of ESR2 (estrogen receptor beta) expression across thousands of primary breast tumors. Sci Rep 2022; 12:4696. [PMID: 35304506 PMCID: PMC8933558 DOI: 10.1038/s41598-022-08210-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/03/2022] [Indexed: 12/31/2022] Open
Abstract
Estrogen receptor alpha (ERα, encoded by ESR1) is a well-characterized transcription factor expressed in more than 75% of breast tumors and is the key biomarker to direct endocrine therapies. On the other hand, much less is known about estrogen receptor beta (ERβ, encoded by ESR2) and its importance in cancer. Previous studies had some disagreement, however most reports suggested a more favorable prognosis for patients with high ESR2 expression. To add further clarity to ESR2 in breast cancer, we interrogated a large population-based cohort of primary breast tumors (n = 3207) from the SCAN-B study. RNA-seq shows ESR2 is expressed at low levels overall with a slight inverse correlation to ESR1 expression (Spearman R = −0.18, p = 2.2e−16), and highest ESR2 expression in the basal- and normal-like PAM50 subtypes. ESR2-high tumors had favorable overall survival (p = 0.006), particularly in subgroups receiving endocrine therapy (p = 0.03) and in triple-negative breast cancer (p = 0.01). These results were generally robust in multivariable analyses accounting for patient age, tumor size, node status, and grade. Gene modules consistent with immune response were associated to ESR2-high tumors. Taken together, our results indicate that ESR2 is generally expressed at low levels in breast cancer but associated with improved overall survival and may be related to immune response modulation.
Collapse
Affiliation(s)
- Hina Dalal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University Cancer Center, Lund University, Medicon Village 404-B2, 22381, Lund, Sweden.,Lund University Cancer Center, Medicon Village, Lund, Sweden
| | - Malin Dahlgren
- Division of Oncology, Department of Clinical Sciences Lund, Lund University Cancer Center, Lund University, Medicon Village 404-B2, 22381, Lund, Sweden.,Lund University Cancer Center, Medicon Village, Lund, Sweden
| | - Sergii Gladchuk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University Cancer Center, Lund University, Medicon Village 404-B2, 22381, Lund, Sweden.,Lund University Cancer Center, Medicon Village, Lund, Sweden
| | - Christian Brueffer
- Division of Oncology, Department of Clinical Sciences Lund, Lund University Cancer Center, Lund University, Medicon Village 404-B2, 22381, Lund, Sweden.,Lund University Cancer Center, Medicon Village, Lund, Sweden
| | - Sofia K Gruvberger-Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University Cancer Center, Lund University, Medicon Village 404-B2, 22381, Lund, Sweden.,Section for Molecular Diagnostics, Skåne University Hospital, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University Cancer Center, Lund University, Medicon Village 404-B2, 22381, Lund, Sweden. .,Lund University Cancer Center, Medicon Village, Lund, Sweden.
| |
Collapse
|
13
|
Langendonk M, de Jong MRW, Smit N, Seiler J, Reitsma B, Ammatuna E, Glaudemans AWJM, van den Berg A, Huls GA, Visser L, van Meerten T. Identification of the estrogen receptor beta as a possible new tamoxifen-sensitive target in diffuse large B-cell lymphoma. Blood Cancer J 2022; 12:36. [PMID: 35256592 PMCID: PMC8901714 DOI: 10.1038/s41408-022-00631-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 01/14/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma subtype. Despite the proven efficacy of combined immunochemotherapy (R-CHOP) in the majority of patients, ~40% of DLBCL patients do not respond or will relapse and consequently have a very poor prognosis. The development of targeted therapies has not improved patient survival, underscoring the need for new treatment approaches. Using an unbiased genome-wide CD20 guilt-by-association approach in more than 1800 DLBCL patients, we previously identified the estrogen receptor beta (ERβ) as a new target in DLBCL. Here, we demonstrate that ERβ is expressed at significantly higher levels in DLBCL compared to normal B cells, and ERβ plays a role in the protection against apoptosis in DLBCL. Targeting of the ERβ with the selective estrogen receptor modulator tamoxifen reduces cell viability in all tested DLBCL cell lines. Tamoxifen-induced cell death was significantly decreased in an ERβ knock-out cell line. The activity of tamoxifen was confirmed in a xenograft human lymphoma model, as tumor growth decreased, and survival significantly improved. Finally, tamoxifen-treated breast cancer (BC) patients showed a significantly reduced risk of 38% for DLBCL compared to BC patients who did not receive tamoxifen. Our findings provide a rationale to investigate tamoxifen, a hormonal drug with a good safety profile, in DLBCL patients.
Collapse
Affiliation(s)
- Myra Langendonk
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Mathilde R W de Jong
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Nienke Smit
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Jonas Seiler
- University of Groningen, ERIBA, Genomic Instability in Development and Disease, Groningen, the Netherlands
| | - Bart Reitsma
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Emanuele Ammatuna
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Andor W J M Glaudemans
- University of Groningen, University Medical center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| | - Anke van den Berg
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Gerwin A Huls
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands
| | - Lydia Visser
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Tom van Meerten
- University of Groningen, University Medical Center Groningen, Department of Hematology, Groningen, the Netherlands.
| |
Collapse
|
14
|
Brandão DC, Lima PMAP, Martins IC, Cordeiro CS, Cordeiro AO, Vecchi L, Guerra JFC, Orsolin PC, Gazolla MC, Costa DS, da Silva Filho AA, Araújo TG. Arrabidaea chica chloroform extract modulates estrogen and androgen receptors on luminal breast cancer cells. BMC Complement Med Ther 2022; 22:18. [PMID: 35057779 PMCID: PMC8773405 DOI: 10.1186/s12906-022-03506-3] [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: 08/17/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast Cancer (BC) is the most common cancer in women worldwide and, although 70% of patients are responsive to selective Estrogen Receptor (ER) modulators such as Tamoxifen (Tam), patients' survival is comprised by resistance to endocrine therapy. Brazilian flora, especially the Amazon biome, is one of the richest global sources of native species with potentially bioactive compounds. Arrabidaea chica is a plant native to the Amazon that has been used in the treatment of different diseases. However, its action on BC remains unclear. METHODS Herein the biological effects of the chloroform extract of A. chica (CEAC) were evaluated on BC cells and in in vivo model. After confirmation of CEAC antioxidant capacity, cells were treated with CEAC and Tam, alone and with CEAC+Tam. The cell viability was evaluated by MTT and hormone receptor transcripts levels were assessed (ESR1, ESR2 and AR). Finally, anticarcinogenicity of CEAC was recorded in Drosophila melanogaster through Epithelial Tumor Test (ETT). RESULTS The study confirmed the antioxidant activity of CEAC. CEAC was selective for MCF-7, downregulating ESR2 and AR transcripts and upregulating ESR2 expression. The modulatory effects of CEAC on ERs did not differ between cells treated with Tam and with CEAC+Tam. Interestingly, previous treatment with CEAC, followed by treatment with Tam promoted a significant decrease in cell viability. The extract also presented anticarcinogenic effect in in vivo assay. CONCLUSION The bioassays on breast tumor cells demonstrated the antiproliferative activity of the extract, which modulated the expression of hormone receptors and sensitized luminal tumor cells to Tam. These results suggest that CEAC could be a complementary treatment for BC.
Collapse
Affiliation(s)
- Douglas C. Brandão
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
| | - Paula M. A. P. Lima
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
- Laboratory of Cytogenetic and Mutagenesis, University Center of Patos de Minas, Patos de Minas, MG Brazil
| | - Isabella C. Martins
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
| | - Carina S. Cordeiro
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
| | - Antonielle O. Cordeiro
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG Brazil
| | - Lara Vecchi
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG Brazil
| | - Joyce F. C. Guerra
- Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas, MG Brazil
| | - Priscila C. Orsolin
- Laboratory of Cytogenetic and Mutagenesis, University Center of Patos de Minas, Patos de Minas, MG Brazil
| | - Matheus C. Gazolla
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG Brazil
| | - Danilo S. Costa
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG Brazil
| | - Ademar A. da Silva Filho
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG Brazil
| | - Thaise G. Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Rua Major Jerônimo, 566, Sala 601, Patos de Minas, MG 38700-002 Brazil
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG Brazil
| |
Collapse
|
15
|
Choi Y. Estrogen Receptor β Expression and Its Clinical Implication in Breast Cancers: Favorable or Unfavorable? J Breast Cancer 2022; 25:75-93. [PMID: 35380018 PMCID: PMC9065353 DOI: 10.4048/jbc.2022.25.e9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 02/03/2022] [Indexed: 11/30/2022] Open
Abstract
There are two estrogen receptor (ER) genes (ESR1/ERα and ESR2/ERβ) in humans. Of those. ERβ, the second ER isotype identified in 1996, is differentially expressed in different phenotypes and molecular subtypes of breast cancer (BCa), and is highly expressed in ERα-negative BCa and triple-negative BCa (TNBC). This review summarizes the potential clinical relevance of ERβ in BCa and the challenges associated with studies on the role of ERβ in BCa. The experimental and clinical studies evaluating clinical outcomes and associations with clinical characteristics and responses to endocrine therapy on targeting ERβ reviewed herein indicate that ERβ is a clinically important biomarker in BCa. The reviewed studies also suggest that each ERβ isoform has a distinct role in BCa subtypes and the potential of novel- targeted therapies in BCa, especially ERα-negative BCa and TNBC. However, the findings of many studies on ERβ are inconsistent, and the exact role of ERβ in BCa remains elusive; this may potentially be attributed to the complexity of ERβ isoforms, but also to the lack of standardized testing protocol. Thus, successful clinical application of ERβ requires the development of standardized, reproducible, and objective measurement methods for ERβ that can be widely and routinely applied in clinical setting.
Collapse
Affiliation(s)
- Young Choi
- Department of Pathology, Yale School of Medicine, Hartsdale, NY, USA
| |
Collapse
|
16
|
Nzegwu MA, Nwokoro O, Nnamani C, Enemuo VC, Nzegwu VI, Nwoye O, Edeh A, Nwankwo K. TP 53 status and estrogen receptor-beta in triple negative breast cancer management in Africa: Time to rethink regime management of triple negative breast cancer and save more lives in Nigeria. Rare Tumors 2021; 13:20363613211050355. [PMID: 34659670 PMCID: PMC8511903 DOI: 10.1177/20363613211050355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
| | - Onyekachi Nwokoro
- Department of Morbid Anatomy, University of Nigeria, Nsukka, Nigeria
| | - Christian Nnamani
- Department of Morbid Anatomy, University of Nigeria, Nsukka, Nigeria
| | | | | | - Ogochukwu Nwoye
- Department of Morbid Anatomy, University of Nigeria, Nsukka, Nigeria
| | - Anthony Edeh
- Department of Radiation Medicine, University of Nigeria, Nsukka, Nigeria
| | - Kenneth Nwankwo
- Department of Surgery, Enugu State University of Science and Technology, Enugu, Nigeria
| |
Collapse
|
17
|
van Barele M, Heemskerk-Gerritsen BAM, Louwers YV, Vastbinder MB, Martens JWM, Hooning MJ, Jager A. Estrogens and Progestogens in Triple Negative Breast Cancer: Do They Harm? Cancers (Basel) 2021; 13:2506. [PMID: 34063736 PMCID: PMC8196589 DOI: 10.3390/cancers13112506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancers (TNBC) occur more frequently in younger women and do not express estrogen receptor (ER) nor progesterone receptor (PR), and are therefore often considered hormone-insensitive. Treatment of premenopausal TNBC patients almost always includes chemotherapy, which may lead to premature ovarian insufficiency (POI) and can severely impact quality of life. Hormone replacement therapy (HRT) is contraindicated for patients with a history of hormone-sensitive breast cancer, but the data on safety for TNBC patients is inconclusive, with a few randomized trials showing increased risk-ratios with wide confidence intervals for recurrence after HRT. Here, we review the literature on alternative pathways from the classical ER/PR. We find that for both estrogens and progestogens, potential alternatives exist for exerting their effects on TNBC, ranging from receptor conversion, to alternative receptors capable of binding estrogens, as well as paracrine pathways, such as RANK/RANKL, which can cause progestogens to indirectly stimulate growth and metastasis of TNBC. Finally, HRT may also influence other hormones, such as androgens, and their effects on TNBCs expressing androgen receptors (AR). Concluding, the assumption that TNBC is completely hormone-insensitive is incorrect. However, the direction of the effects of the alternative pathways is not always clear, and will need to be investigated further.
Collapse
Affiliation(s)
- Mark van Barele
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Bernadette A. M. Heemskerk-Gerritsen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Yvonne V. Louwers
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Mijntje B. Vastbinder
- Department of Internal Medicine, Ijsselland Hospital, Prins Constantijnweg 2, 2906 ZC Capelle aan den IJssel, The Netherlands;
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| |
Collapse
|
18
|
Rossini E, Tamburello M, Abate A, Beretta S, Fragni M, Cominelli M, Cosentini D, Hantel C, Bono F, Grisanti S, Poliani PL, Tiberio GAM, Memo M, Sigala S, Berruti A. Cytotoxic Effect of Progesterone, Tamoxifen and Their Combination in Experimental Cell Models of Human Adrenocortical Cancer. Front Endocrinol (Lausanne) 2021; 12:669426. [PMID: 33981288 PMCID: PMC8108132 DOI: 10.3389/fendo.2021.669426] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Progesterone (Pg) and estrogen (E) receptors (PgRs and ERs) are expressed in normal and neoplastic adrenal cortex, but their role is not fully understood. In literature, Pg demonstrated cytotoxic activity on AdrenoCortical Carcinoma (ACC) cells, while tamoxifen is cytotoxic in NCI-H295R cells. Here, we demonstrated that in ACC cell models, ERs were expressed in NCI-H295R cells with a prevalence of ER-β over the ER-α.Metastasis-derived MUC-1 and ACC115m cells displayed a very weak ER-α/β signal, while PgR cells were expressed, although at low level. Accordingly, these latter were resistant to the SERM tamoxifen and scarcely sensitive to Pg, as we observed a lower potency compared to NCI-H295R cells in cytotoxicity (IC50: MUC-1 cells: 67.58 µM (95%CI: 63.22-73.04), ACC115m cells: 51.76 µM (95%CI: 46.45-57.67) and cell proliferation rate. Exposure of NCI-H295R cells to tamoxifen induced cytotoxicity (IC50: 5.43 µM (95%CI: 5.18-5.69 µM) mainly involving ER-β, as their nuclear localization increased after tamoxifen: Δ A.U. treated vs untreated: 12 h: +27.04% (p < 0.01); 24 h: +36.46% (p < 0.0001). This effect involved the SF-1 protein reduction: Pg: -36.34 ± 9.26%; tamoxifen: -46.25 ± 15.68% (p < 0.01). Finally, in a cohort of 36 ACC samples, immunohistochemistry showed undetectable/low level of ERs, while PgR demonstrated a higher expression. In conclusion, ACC experimental cell models expressed PgR and low levels of ER in line with data obtained in patient tissues, thus limiting the possibility of a clinical approach targeting ER. Interestingly, Pg exerted cytotoxicity also in metastatic ACC cells, although with low potency.
Collapse
Affiliation(s)
- Elisa Rossini
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Mariangela Tamburello
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Andrea Abate
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Silvia Beretta
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Martina Fragni
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Manuela Cominelli
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Deborah Cosentini
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Federica Bono
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Pietro Luigi Poliani
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Guido A. M. Tiberio
- Surgical Clinic, Department of Clinical and Experimental Sciences, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Sandra Sigala
- Department of Molecular and Translational Medicine, Section of Pharmacology, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| | - Alfredo Berruti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|
19
|
Shalabi MG, Abbas AM, Mills J, Kheirelseid MA, Elderdery AY. The Prognostic Value of Estrogen Receptor β Isoform With Correlation of Estrogen Receptor α Among Sudanese Breast Cancer Patients. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2021; 15:1178223421998354. [PMID: 33716507 PMCID: PMC7917412 DOI: 10.1177/1178223421998354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 02/02/2021] [Indexed: 11/16/2022]
Abstract
Two estrogen receptor isoforms (ERα and ERβ) have been characterized with variable and sometimes contrasting responses to estrogens, partially explained by different receptor signaling pathways in estrogen-sensitive tissues. This is a retrospective, descriptive, cross-sectional study, aiming to evaluate the expression pattern of ERβ, employing immunohistochemical techniques using specific monoclonal antibody for ERβ, to correlate its expression with that of ERα in a Sudanese population. Two-hundred and fifty formalin-fixed paraffin-wax-embedded breast tissue blocks were used in this study. Of these, 200 were taken from breast cancer patients ascertained as study cases, and the remaining 50 were noninvolved surgical margin considered as normal breast tissue. Receptor expression was demonstrated using immunohistochemical techniques. The immune expression of ERβ was detected in 57.5% of breast cancers. It was differentially expressed in breast tissues encompassing normal, noninvasive, as well as invasive carcinoma (P = .02). There was no evidence of a significant relationship between ERβ and ERα expression. Among the ERα-negative tumor, 60.4% expressed ERβ. The expression of ERβ among this subgroup was significantly associated with good clinicopathological parameters such as negative Her2/neu, lower grade, and negative lymph node metastasis (P = .002). This study concludes that ERβ was commonly expressed among Sudanese patients with breast cancer, either co-expressed with ERα or expressed alone. In the ERα-negative subgroup, it was associated with better tumor outcomes suggesting ERβ should be included in the diagnostic protocol as an independent marker for favorable prognosis.
Collapse
Affiliation(s)
- Manar G Shalabi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Jouf University, Sakakah, Saudi Arabia.,Medical Laboratory Sciences Department, Nahda College, Khartoum, Sudan
| | - Anass M Abbas
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Jouf University, Sakakah, Saudi Arabia.,Medical Laboratory Program, School of Medicine, Alyarmouk Medical College, Khartoum, Sudan
| | - Jeremy Mills
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Mohamed A Kheirelseid
- Pathology Department, Faculty of Medicine, Omdurman Islamic University, Omdurman, Sudan
| | - Abozer Y Elderdery
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Jouf University, Sakakah, Saudi Arabia.,Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti, Sudan
| |
Collapse
|
20
|
Božović A, Mandušić V, Todorović L, Krajnović M. Estrogen Receptor Beta: The Promising Biomarker and Potential Target in Metastases. Int J Mol Sci 2021; 22:ijms22041656. [PMID: 33562134 PMCID: PMC7914503 DOI: 10.3390/ijms22041656] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/24/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
The discovery of the Estrogen Receptor Beta (ERβ) in 1996 opened new perspectives in the diagnostics and therapy of different types of cancer. Here, we present a review of the present research knowledge about its role in endocrine-related cancers: breast, prostate, and thyroid, and colorectal cancers. We also discuss the reasons for the controversy of its role in carcinogenesis and why it is still not in use as a biomarker in clinical practice. Given that the diagnostics and therapy would benefit from the introduction of new biomarkers, we suggest ways to overcome the contradictions in elucidating the role of ERβ.
Collapse
|
21
|
CHRNA5 belongs to the secondary estrogen signaling network exhibiting prognostic significance in breast cancer. Cell Oncol (Dordr) 2021; 44:453-472. [PMID: 33469842 DOI: 10.1007/s13402-020-00581-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2020] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Cholinergic signals can be important modulators of cellular signaling in cancer. We recently have shown that knockdown of nicotinic acetylcholine receptor subunit alpha 5, CHRNA5, diminishes the proliferative potential of breast cancer cells. However, modulation of CHRNA5 expression in the context of estrogen signaling and its prognostic implications in breast cancer remained unexplored. METHODS Meta-analyses of large breast cancer microarray cohorts were used to evaluate the association of CHRNA5 expression with estrogen (E2) treatment, estrogen receptor (ER) status and patient prognosis. The results were validated through RT-qPCR analyses of multiple E2 treated cell lines, CHRNA5 depleted MCF7 cells and across a breast cancer patient cDNA panel. We also calculated a predicted secondary (PS) score representing direct/indirect induction of gene expression by E2 based on a public dataset (GSE8597). Co-expression analysis was performed using a weighted gene co-expression network analysis (WGCNA) pipeline. Multiple other publicly available datasets such as CCLE, COSMIC and TCGA were also analyzed. RESULTS Herein we found that CHRNA5 expression was induced by E2 in a dose- and time-dependent manner in breast cancer cell lines. ER- breast tumors exhibited higher CHRNA5 expression levels than ER+ tumors. Independent meta-analysis for survival outcome revealed that higher CHRNA5 expression was associated with a worse prognosis in untreated breast cancer patients. Furthermore, CHRNA5 and its co-expressed gene network emerged as secondarily induced targets of E2 stimulation. These targets were largely downregulated by exposure to CHRNA5 siRNA in MCF7 cells while the response of primary ESR1 targets was dependent on the direction of the PS-score. Moreover, primary and secondary target genes were uncoupled and clustered distinctly based on multiple public datasets. CONCLUSION Our findings strongly associate increased expression of CHRNA5 and its co-expression network with secondary E2 signaling and a worse prognosis in breast cancer.
Collapse
|
22
|
Daraei A, Izadi P, Khorasani G, Nafissi N, Naghizadeh MM, Meysamie A, Mansoori Y, Nariman-Saleh-Fam Z, Bastami M, Saadatian Z, Roshan SJ, Bayani N, Tavakkoly-Bazzaz J. A methylation signature at the CpG island promoter of estrogen receptor beta (ER-β) in breasts of women may be an early footmark of lack of breastfeeding and nulliparity. Pathol Res Pract 2020; 218:153328. [PMID: 33422777 DOI: 10.1016/j.prp.2020.153328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 11/24/2022]
Abstract
Although little is known regarding the mechanisms behind the onset of breast cancer (BC) through reproductive risk factors, new researches have highlighted some early tumor-related methylation footmarks in the breast tissue of apparently clinically healthy women as their potential epigenetic mechanism. Previous evidence supports that the estrogen receptor beta (ER-β), whose anti-cancer roles had already been revealed in BC, is downregulated in the breasts of healthy nulliparous women. Nevertheless, data on such a link about its methylation alterations have not been reported. The goal of current study was to determine possible methylation alterations at CpG island promoter of the ER-β gene, including promoter 0 N and exon 0 N, in relation to aspects of reproductive history in the healthy breasts. The DNA was extracted from the breasts of 120 subjects undergoing cosmetic mammoplasty. Thereafter, the methylation levels of targeted regions in ER-β gene were determined by using MeDIP-qPCR assay. The results revealed that ER-β exon 0 N had no methylation in 84.2 % of the women, whereas the rest, comprising 2.5 % and 13.3 % of the samples, showed a lower and higher of its methylation, respectively. Interestingly, nulliparous women were found to have an elevated methylation level of the ER-β exon 0 N than parous women (P = 0.036). Moreover, we observed a high methylation of the ER-β exon 0 N in the breasts of non-breastfeeding women compared to breastfeeding subgroup (P = 0.048). Likewise, the non-breastfeeding subgroup showed exon 0N high methylation in comparison to women with breastfeeding >24 months (P = 0.023). Finally, although we found that 6.67 % of the samples had a high methylation level at the promoter 0N, no any relationship was found between its methylation and reproductive history. These results may provide key clues to revealing the epigenetic mechanism through which the nulliparity and lack of breastfeeding influencing the risk factor of BC as well as introducing the potential new early prediction and prevention strategies. Although further investigations need to be done in order to gain a better understanding the roles of these epigenetic signatures.
Collapse
Affiliation(s)
- Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasemali Khorasani
- Division of Plastic and Reconstructive Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Nafissi
- Surgical Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alipasha Meysamie
- Community and Preventive Medicine Department, Medical Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Zahra Saadatian
- Department of Physiology, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Samaneh Jafari Roshan
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Niloofar Bayani
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Zattarin E, Leporati R, Ligorio F, Lobefaro R, Vingiani A, Pruneri G, Vernieri C. Hormone Receptor Loss in Breast Cancer: Molecular Mechanisms, Clinical Settings, and Therapeutic Implications. Cells 2020; 9:cells9122644. [PMID: 33316954 PMCID: PMC7764472 DOI: 10.3390/cells9122644] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/02/2020] [Accepted: 12/05/2020] [Indexed: 12/14/2022] Open
Abstract
Hormone receptor-positive breast cancer (HR+ BC) accounts for approximately 75% of new BC diagnoses. Despite the undisputable progresses obtained in the treatment of HR+ BC in recent years, primary or acquired resistance to endocrine therapies still represents a clinically relevant issue, and is largely responsible for disease recurrence after curative surgery, as well as for disease progression in the metastatic setting. Among the mechanisms causing primary or acquired resistance to endocrine therapies is the loss of estrogen/progesterone receptor expression, which could make BC cells independent of estrogen stimulation and, consequently, resistant to estrogen deprivation or the pharmacological inhibition of estrogen receptors. This review aims at discussing the molecular mechanisms and the clinical implications of HR loss as a result of the therapies used in the neoadjuvant setting or for the treatment of advanced disease in HR+ BC patients.
Collapse
Affiliation(s)
- Emma Zattarin
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
| | - Rita Leporati
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
| | - Francesca Ligorio
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
| | - Riccardo Lobefaro
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
| | - Andrea Vingiani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
| | - Giancarlo Pruneri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
- Department of Oncology and Haematology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Claudio Vernieri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy; (E.Z.); (R.L.); (F.L.); (R.L.); (A.V.); (G.P.)
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
- Correspondence: ; Tel.: +39-02-2390-3650
| |
Collapse
|
24
|
Björk Gunnarsdottir F, Auoja N, Bendahl PO, Rydén L, Fernö M, Leandersson K. Co-localization of CD169 + macrophages and cancer cells in lymph node metastases of breast cancer patients is linked to improved prognosis and PDL1 expression. Oncoimmunology 2020; 9:1848067. [PMID: 33299660 PMCID: PMC7714471 DOI: 10.1080/2162402x.2020.1848067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Breast cancer is the most common form of cancer in women worldwide. Although the survival among breast cancer patients has improved, there is still a large group of patients with dismal prognosis. One of the most important prognostic factors for poor prognosis is lymph node metastasis. Increasing knowledge concerning the lymph nodes of breast cancer patients indicates that they are affected by the primary tumor. In this study we show that presence of CD169+ subcapsular sinus macrophages in contact with lymph node metastases in breast cancer patients, is related to better prognosis after adjuvant tamoxifen treatment, but only in patients with PDL1+ primary tumors. This is in contrast to the prognostic effect of CD169+ primary tumor-associated macrophages (TAMs). We further show that CD169+ macrophages were spatially associated with expression of PDL1 on nearby cells, both in primary tumors and metastatic lymph node, although PDL1 expression in metastatic lymph node as such did not have further prognostic impact. Our data suggest that CD169+ resident lymph node macrophages have a unique function in targeting immune responses against breast cancer and should be further investigated in detail.
Collapse
Affiliation(s)
| | - Nathalie Auoja
- Cancer Immunology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Pär-Ola Bendahl
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lisa Rydén
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Mårten Fernö
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
25
|
Vargas-Rondón N, Pérez-Mora E, Villegas VE, Rondón-Lagos M. Role of chromosomal instability and clonal heterogeneity in the therapy response of breast cancer cell lines. Cancer Biol Med 2020; 17:970-985. [PMID: 33299647 PMCID: PMC7721098 DOI: 10.20892/j.issn.2095-3941.2020.0028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/20/2020] [Indexed: 01/08/2023] Open
Abstract
Objective Chromosomal instability (CIN) is a hallmark of cancer characterized by cell-to-cell variability in the number or structure of chromosomes, frequently observed in cancer cell populations and is associated with poor prognosis, metastasis, and therapeutic resistance. Breast cancer (BC) is characterized by unstable karyotypes and recent reports have indicated that CIN may influence the response of BC to chemotherapy regimens. However, paradoxical associations between extreme CIN and improved outcome have been observed. Methods This study aimed to 1) evaluate CIN levels and clonal heterogeneity (CH) in MCF7, ZR-751, MDA-MB468, BT474, and KPL4 BC cells treated with low doses of tamoxifen (TAM), docetaxel (DOC), doxorubicin (DOX), Herceptin (HT), and combined treatments (TAM/DOC, TAM/DOX, TAM/HT, HT/DOC, and HT/DOX) by using fluorescence in situ hybridization (FISH), and 2) examine the association with response to treatments by comparing FISH results with cell proliferation. Results Intermediate CIN was linked to drug sensitivity according to three characteristics: estrogen receptor α (ERα) and HER2 status, pre-existing CIN level in cancer cells, and the CIN induced by the treatments. ERα+/HER2- cells with intermediate CIN were sensitive to treatment with taxanes (DOC) and anthracyclines (DOX), while ERα-/HER2-, ERα+/HER2+, and ERα-/HER2+ cells with intermediate CIN were resistant to these treatments. Conclusions A greater understanding of CIN and CH in BC could assist in the optimization of existing therapeutic regimens and/or in supporting new strategies to improve cancer outcomes.
Collapse
Affiliation(s)
- Natalia Vargas-Rondón
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | - Erika Pérez-Mora
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | - Victoria E. Villegas
- Biology Program, Faculty of Natural Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| |
Collapse
|
26
|
Ilg MM, Stafford SJ, Mateus M, Bustin SA, Carpenter MJ, Muneer A, Bivalacqua TJ, Ralph DJ, Cellek S. Phosphodiesterase Type 5 Inhibitors and Selective Estrogen Receptor Modulators Can Prevent But Not Reverse Myofibroblast Transformation in Peyronie's Disease. J Sex Med 2020; 17:1848-1864. [PMID: 32771352 DOI: 10.1016/j.jsxm.2020.06.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Myofibroblast transformation is a key step in the pathogenesis of Peyronie's disease (PD). Phosphodiesterase type 5 inhibitors (PDE5is) and selective estrogen receptor modulators (SERMs) can prevent the formation of fibrosis in in vitro and in vivo models of PD. However, it is unknown whether these drugs can also reverse established fibrosis. AIM To investigate whether PDE5is and SERMs can reverse transforming growth factor beta 1 (TGF-β1)-induced myofibroblast transformation and determine the point of no return. METHODS In-Cell enzyme-linked immunosorbent assay was used to quantify TGF-β1-induced myofibroblast transformation of human primary fibroblasts isolated from tunica albuginea (TA) of patients undergoing surgery for treatment of PD. Extracellular matrix production and collagen contraction assays were used as secondary assays. Reverse transcription-quantitative polymerase chain reaction and In-Cell enzyme-linked immunosorbent assay were used to measure drug target expression. PDE5i (vardenafil) and SERM (tamoxifen) were applied at various time points after TGF-β1. OUTCOMES Reversibility of myofibroblast transformation and drug target expression were investigated in a time-dependent manner in TA-derived fibroblasts. RESULTS Vardenafil or tamoxifen could not reverse the myofibroblast traits of alpha-smooth muscle actin expression and extracellular matrix production, whereas only tamoxifen affected collagen contraction after 72 hours of TGF-β1 treatment. Phosphodiesterase 5A and estrogen receptor (ER)-β were downregulated after 72 hours, and estrogen receptor -α protein could not be quantified. Tamoxifen could prevent myofibroblast transformation until 36 hours after TGF-β1 treatment, whereas vardenafil could prevent only 24 hours after TGF-β1 treatment. This was mirrored by downregulation of drug targets on mRNA and protein level. Furthermore, antifibrotic signaling pathways, peroxisome proliferator-activated receptor gamma and betaglycan (TGFB receptor III), were significantly downregulated after 36 hours of TGF-β1 exposure, as opposed to upregulation of profibrotic thrombospondin-1 at the same time point. CLINICAL TRANSLATION This study suggests that using PDE5is and SERMs might only help for early-phase PD and further highlights the need to test drugs at the appropriate stage of the disease based on their mechanism of action. STRENGTHS & LIMITATIONS The study uses primary human TA-derived fibroblasts that enhances translatability of the results. Limitations include that only 1 example of PDE5i- and SERM-type drug was tested. Time course experiments were only performed for marker expression experiments and not for functional assays. CONCLUSION This is the first study to demonstrate that timing for administration of drugs affecting myofibroblast transformation appears to be vital in in vitro models of PD, where 36 hours of TGF-β1 treatment can be suggested as a "point of no return" for myofibroblast transformation. Ilg MM, Stafford SJ, Mateus M, et al. Phosphodiesterase Type 5 Inhibitors and Selective Estrogen Receptor Modulators Can Prevent But Not Reverse Myofibroblast Transformation in Peyronie's Disease. J Sex Med 2020;17:1848-1864.
Collapse
Affiliation(s)
- Marcus M Ilg
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK.
| | - Simon J Stafford
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Marta Mateus
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Stephen A Bustin
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Michael J Carpenter
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Asif Muneer
- Department of Urology, University College London, London, UK; NIHR Biomedical Research Centre, University College London, London, UK
| | - Trinity J Bivalacqua
- James Buchanan Brady Urologic Institute, John Hopkins University, Baltimore, MD, USA
| | - David J Ralph
- Department of Urology, University College London, London, UK
| | - Selim Cellek
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| |
Collapse
|
27
|
Sellitto A, D’Agostino Y, Alexandrova E, Lamberti J, Pecoraro G, Memoli D, Rocco D, Coviello E, Giurato G, Nassa G, Tarallo R, Weisz A, Rizzo F. Insights into the Role of Estrogen Receptor β in Triple-Negative Breast Cancer. Cancers (Basel) 2020; 12:cancers12061477. [PMID: 32516978 PMCID: PMC7353068 DOI: 10.3390/cancers12061477] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptors (ERα and ERβ) are ligand-activated transcription factors that play different roles in gene regulation and show both overlapping and specific tissue distribution patterns. ERβ, contrary to the oncogenic ERα, has been shown to act as an oncosuppressor in several instances. However, while the tumor-promoting actions of ERα are well-known, the exact role of ERβ in carcinogenesis and tumor progression is not yet fully understood. Indeed, to date, highly variable and even opposite effects have been ascribed to ERβ in cancer, including for example both proliferative and growth-inhibitory actions. Recently ERβ has been proposed as a potential target for cancer therapy, since it is expressed in a variety of breast cancers (BCs), including triple-negative ones (TNBCs). Because of the dependence of TNBCs on active cellular signaling, numerous studies have attempted to unravel the mechanism(s) behind ERβ-regulated gene expression programs but the scenario has not been fully revealed. We comprehensively reviewed the current state of knowledge concerning ERβ role in TNBC biology, focusing on the different signaling pathways and cellular processes regulated by this transcription factor, as they could be useful in identifying new diagnostic and therapeutic approaches for TNBC.
Collapse
Affiliation(s)
- Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Ylenia D’Agostino
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Jessica Lamberti
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Giovanni Pecoraro
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Domenico Memoli
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Domenico Rocco
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Elena Coviello
- Genomix4Life, via S. Allende 43/L, 84081 Baronissi (SA), Italy;
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
- CRGS (Genome Research Center for Health), University of Salerno Campus of Medicine, 84081 Baronissi (SA), Italy
- Correspondence: (A.W.); (F.R.); Tel.: (39+)-089-965043 (A.W.); Tel.: (39+)-089-965221 (F.R.)
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Italy; (A.S.); (Y.D.); (E.A.); (J.L.); (G.P.); (D.M.); (D.R.); (G.G.); (G.N.); (R.T.)
- CRGS (Genome Research Center for Health), University of Salerno Campus of Medicine, 84081 Baronissi (SA), Italy
- Correspondence: (A.W.); (F.R.); Tel.: (39+)-089-965043 (A.W.); Tel.: (39+)-089-965221 (F.R.)
| |
Collapse
|
28
|
Badve SS, Gökmen-Polar Y. TP53 Status and Estrogen Receptor-Beta in Triple-Negative Breast Cancer: Company Matters. J Natl Cancer Inst 2020; 111:1118-1119. [PMID: 30989234 DOI: 10.1093/jnci/djz052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/05/2019] [Indexed: 01/21/2023] Open
|
29
|
van Barele M, Heemskerk-Gerritsen BAM, van Doorn HC, Schmidt MK, Hooning MJ, Jager A. The impact of menstruation persistence or recovery after chemotherapy on survival in young patients with hormone receptor negative breast cancer. Breast 2020; 52:102-109. [PMID: 32497987 PMCID: PMC7375582 DOI: 10.1016/j.breast.2020.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Hormone replacement therapy can diminish hormone depletion-related complaints in postmenopausal women, but is contraindicated for postmenopausal breast cancer (BC) patients. Recovery of menstruation after chemotherapy-induced amenorrhea in young hormone receptor-negative BC patients however, is accepted. To determine the safety of this strategy, we investigated the effect of recovery of menstruation on disease-free survival (DFS) and overall survival (OS) in young hormone receptor-negative BC patients treated with (neo)adjuvant chemotherapy. Methods We selected 636 patients from a single-center cohort with early stage hormone receptor-negative BC and under the age of 50 years when treated with chemotherapy. Sufficient data on course of menstruation in medical records was retrospectively found for 397 patients, of whom 299 patients (75%) had a recovery of menstruation after chemotherapy. We used Cox proportional hazards models to estimate hazard ratios (HR) for the effect of recovery of menstruation on DFS and OS. Results Patients with recovery of menstruation after chemotherapy less frequently had lymph node involvement at diagnosis (45% vs 66%, p = 0.001). After a median follow-up of 6.7 years, the adjusted hazard ratios were 1.45 (95% CI: 0.83–2.54) for DFS and 1.19 (95% CI: 0.71–1.98) for OS. Conclusion No significantly increased recurrence risk was found for hormone receptor-negative BC patients with recovery of menstruation after chemotherapy. However, the outcome of the multivariable model is not reassuring and a potentially increased recurrence risk cannot be excluded. The results need to be validated in a larger prospective study for a more definitive answer. Menstruation often recovers after chemotherapy in young breast cancer patients. This is currently accepted for those with a hormone receptor negative subtype. The cell type such tumors arise from is possibly sensitive to paracrine stimulation. Recovery of menstruation may still negatively affect outcome in these patients.
Collapse
Affiliation(s)
- Mark van Barele
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Helena C van Doorn
- Department of Gynecological Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands; Department of Epidemiology, Leiden University, Leiden, the Netherlands
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
30
|
Bhattacharjee A, Hossain MU, Chowdhury ZM, Rahman SMA, Bhuyan ZA, Salimullah M, Keya CA. Insight of druggable cannabinoids against estrogen receptor β in breast cancer. J Biomol Struct Dyn 2020; 39:1688-1697. [PMID: 32116130 DOI: 10.1080/07391102.2020.1737233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Breast cancer (BC) is the second most prevalent cancer worldwide. Estrogen receptor beta (ERβ) is an essential protein of breast cells to suppress estrogen-induced uncontrolled proliferation. Thus, small molecules that can modulate and enhance ERβ expression would be an effective agent to suppress BC development. Studies showed that cannabinoid (CB), specifically delta-9-tetrahydrocannabinol (Del9THC), can increase the expression of ERβ and inhibits BC cell proliferation. In this study, less psychoactive and structurally similar analogs of Del9THC were chosen as drug candidates and ERβ was targeted as a therapeutic receptor. Delta-8-tetrahydrocannabinol (Del8THC) and delta-4-isotetrahydrocannabinol (Del4isoTHC) were the drug candidates selected on the basis of literature reports, absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties, medicinal chemistry profile, and physicochemical features. Molecular docking simulations were carried out to determine ligand receptor interactions and binding affinity based on free binding energy. To get a better drug, the structural modification was done on Del8THC and generated a new CB analog called Cannabinoid A. Finally, molecular interaction analysis revealed that two CBs and one of their analog interact with the active site residues of ERβ. Therefore, this study revealed a new way to discover novel drug(s) for BC patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Arittra Bhattacharjee
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Zeshan Mahmud Chowdhury
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| | - S M Adiat Rahman
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| | - Zaied Ahmed Bhuyan
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka, Bangladesh
| |
Collapse
|
31
|
Le Cornet C, Walter B, Sookthai D, Johnson TS, Kühn T, Herpel E, Kaaks R, Fortner RT. Circulating 27-hydroxycholesterol and breast cancer tissue expression of CYP27A1, CYP7B1, LXR-β, and ERβ: results from the EPIC-Heidelberg cohort. Breast Cancer Res 2020; 22:23. [PMID: 32075687 PMCID: PMC7031866 DOI: 10.1186/s13058-020-1253-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background Experimental and epidemiological studies demonstrate a role for 27-hydroxycholesterol (27HC) in breast cancer development, though results are conflicting. Cholesterol 27-hydroxylase (CYP27A1) and oxysterol 7-alpha-hydroxylase (CYP7B1) regulate 27HC concentrations, while differential expression of the liver X receptor (LXR) and estrogen receptor beta (ERβ) may impact the association between 27HC and breast cancer risk. Methods We evaluated correlates of tumor tissue expression of CYP27A1, CYP7B1, LXR-β, and ERβ and the association between circulating prediagnostic 27HC concentrations and breast cancer risk by marker expression in a nested case-control study within the European Prospective Investigation into Cancer and Nutrition (EPIC)-Heidelberg cohort including 287 breast cancer cases with tumor tissue available. Tumor protein expression was evaluated using immunohistochemistry, and serum 27HC concentrations quantified using liquid chromatography–mass spectrometry. Conditional logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Results A higher proportion of CYP7B1-positive cases were progesterone receptor (PR)-positive, relative to CYP7B1-negative cases, whereas a higher proportion of ERβ-positive cases were Bcl-2 low, relative to ERβ-negative cases. No differences in tumor tissue marker positivity were observed by reproductive and lifestyle factors. We observed limited evidence of heterogeneity in associations between circulating 27HC and breast cancer risk by tumor tissue expression of CYP27A1, CYP7B1, LXR-β, and ERβ, with the exception of statistically significant heterogeneity by LXR-β status in the subgroup of women perimenopausal at blood collection (p = 0.02). Conclusion This exploratory study suggests limited associations between tumor marker status and epidemiologic or breast cancer characteristics. Furthermore, the association between circulating 27HC and breast cancer risk may not vary by tumor expression of CYP27A1, CYP7B1, LXR-β, or ERβ. Electronic supplementary material The online version of this article (10.1186/s13058-020-1253-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte Le Cornet
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Britta Walter
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Disorn Sookthai
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Theron S Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ester Herpel
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Renée T Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
32
|
Wu ZH, Yun-Tang, Cheng Q. Data Mining Identifies Six Proteins that Can Act as Prognostic Markers for Head and Neck Squamous Cell Carcinoma. Cell Transplant 2020; 29:963689720929308. [PMID: 32452220 PMCID: PMC7563932 DOI: 10.1177/0963689720929308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a malignant tumor of the upper aerodigestive tract affecting the oral cavity, lips, paranasal sinuses, larynx, and nasopharynx. Proteogenomics combines proteomics and genomics and employs mass spectrometry and high-throughput sequencing technologies to identify novel peptides. The aim of this study was to identify potential protein biomarkers for clinical treatment of HNSCC. To achieve this, we utilized two sets of data, one on proteins from The Cancer Proteome Atlas (TCPA) and the other on gene expression from The Cancer Genome Atlas (TCGA) database, to evaluate dysfunctional proteogenomics microenvironment. Univariate Cox regression analysis was performed to examine the relationship between protein signatures and prognosis. A total of 19 proteins were significantly associated with overall survival (OS) of patients, of which E2F transcription factor 1 (E2F1; HR = 4.557, 95% CI = 1.810 to 11.469) and enhancer of zeste homolog 2 (EZH2; HR = 0.430, 95% CI = 0.187 to 0.984) were the most differentially expressed between patients with longer and shorter OS, respectively. Furthermore, multivariate Cox regression analysis on six proteins (ERALPHA, HER3, BRAF, P27, RAPTOR, and E2F1) was performed to build the prognostic model. The receiver operating characteristic curves were used to determine whether the expression pattern of survival-related proteins could provide an early prediction of the occurrence of HNSCC. Herein, we found an AUC of 0.720. Based on an online database, we identified novel protein markers for the prognosis of HNSCC. The findings of the present study may provide new insights into the development of new and reliable tools for precise cancer intervention.
Collapse
Affiliation(s)
- Zeng-hong Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun-Tang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Cheng
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
33
|
Barupal DK, Gao B, Budczies J, Phinney BS, Perroud B, Denkert C, Fiehn O. Prioritization of metabolic genes as novel therapeutic targets in estrogen-receptor negative breast tumors using multi-omics data and text mining. Oncotarget 2019; 10:3894-3909. [PMID: 31231467 PMCID: PMC6570467 DOI: 10.18632/oncotarget.26995] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/13/2019] [Indexed: 01/12/2023] Open
Abstract
Estrogen-receptor negative (ERneg) breast cancer is an aggressive breast cancer subtype in the need for new therapeutic options. We have analyzed metabolomics, proteomics and transcriptomics data for a cohort of 276 breast tumors (MetaCancer study) and nine public transcriptomics datasets using univariate statistics, meta-analysis, Reactome pathway analysis, biochemical network mapping and text mining of metabolic genes. In the MetaCancer cohort, a total of 29% metabolites, 21% proteins and 33% transcripts were significantly different (raw p <0.05) between ERneg and ERpos breast tumors. In the nine public transcriptomics datasets, on average 23% of all genes were significantly different (raw p <0.05). Specifically, up to 60% of the metabolic genes were significantly different (meta-analysis raw p <0.05) across the transcriptomics datasets. Reactome pathway analysis of all omics showed that energy metabolism, and biosynthesis of nucleotides, amino acids, and lipids were associated with ERneg status. Text mining revealed that several significant metabolic genes and enzymes have been rarely reported to date, including PFKP, GART, PLOD1, ASS1, NUDT12, FAR1, PDE7A, FAHD1, ITPK1, SORD, HACD3, CDS2 and PDSS1. Metabolic processes associated with ERneg tumors were identified by multi-omics integration analysis of metabolomics, proteomics and transcriptomics data. Overall results suggested that TCA anaplerosis, proline biosynthesis, synthesis of complex lipids and mechanisms for recycling substrates were activated in ERneg tumors. Under-reported genes were revealed by text mining which may serve as novel candidates for drug targets in cancer therapies. The workflow presented here can also be used for other tumor types.
Collapse
Affiliation(s)
- Dinesh Kumar Barupal
- West Coast Metabolomics Center, University of California, Davis, CA, USA.,Co-first authors and contributed equally to this work
| | - Bei Gao
- West Coast Metabolomics Center, University of California, Davis, CA, USA.,Co-first authors and contributed equally to this work
| | - Jan Budczies
- Institute of Pathology, Charité University Hospital, Berlin, Germany
| | - Brett S Phinney
- UC Davis Genome Center, University of California, Davis, CA, USA
| | - Bertrand Perroud
- UC Davis Genome Center, University of California, Davis, CA, USA
| | - Carsten Denkert
- Institute of Pathology, Charité University Hospital, Berlin, Germany.,German Institute of Pathology, Philipps-University Marburg, Marburg, Germany
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| |
Collapse
|
34
|
Mukhopadhyay UK, Oturkar CC, Adams C, Wickramasekera N, Bansal S, Medisetty R, Miller A, Swetzig WM, Silwal-Pandit L, Børresen-Dale AL, Creighton CJ, Park JH, Konduri SD, Mukhopadhyay A, Caradori A, Omilian A, Bshara W, Kaipparettu BA, Das GM. TP53 Status as a Determinant of Pro- vs Anti-Tumorigenic Effects of Estrogen Receptor-Beta in Breast Cancer. J Natl Cancer Inst 2019; 111:1202-1215. [PMID: 30990221 PMCID: PMC6855950 DOI: 10.1093/jnci/djz051] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 12/28/2018] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anti-tumorigenic vs pro-tumorigenic roles of estrogen receptor-beta (ESR2) in breast cancer remain unsettled. We investigated the potential of TP53 status to be a determinant of the bi-faceted role of ESR2 and associated therapeutic implications for triple negative breast cancer (TNBC). METHODS ESR2-TP53 interaction was analyzed with multiple assays including the in situ proximity ligation assay. Transcriptional effects on TP53-target genes and cell proliferation in response to knocking down or overexpressing ESR2 were determined. Patient survival according to ESR2 expression levels and TP53 mutation status was analyzed in the basal-like TNBC subgroup in the Molecular Taxonomy of Breast Cancer International Consortium (n = 308) and Roswell Park Comprehensive Cancer Center (n = 46) patient cohorts by univariate Cox regression and log-rank test. All statistical tests are two-sided. RESULTS ESR2 interaction with wild-type and mutant TP53 caused pro-proliferative and anti-proliferative effects, respectively. Depleting ESR2 in cells expressing wild-type TP53 resulted in increased expression of TP53-target genes CDKN1A (control group mean [SD] = 1 [0.13] vs ESR2 depletion group mean [SD] = 2.08 [0.24], P = .003) and BBC3 (control group mean [SD] = 1 [0.06] vs ESR2 depleted group mean [SD] = 1.92 [0.25], P = .003); however, expression of CDKN1A (control group mean [SD] = 1 [0.21] vs ESR2 depleted group mean [SD] = 0.56 [0.12], P = .02) and BBC3 (control group mean [SD] = 1 [0.03] vs ESR2 depleted group mean [SD] = 0.55 [0.09], P = .008) was decreased in cells expressing mutant TP53. Overexpressing ESR2 had opposite effects. Tamoxifen increased ESR2-mutant TP53 interaction, leading to reactivation of TP73 and apoptosis. High levels of ESR2 expression in mutant TP53-expressing basal-like tumors is associated with better prognosis (Molecular Taxonomy of Breast Cancer International Consortium cohort: log-rank P = .001; hazard ratio = 0.26, 95% confidence interval = 0.08 to 0.84, univariate Cox P = .02). CONCLUSIONS TP53 status is a determinant of the functional duality of ESR2. Our study suggests that ESR2-mutant TP53 combination prognosticates survival in TNBC revealing a novel strategy to stratify TNBC for therapeutic intervention potentially by repurposing tamoxifen.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gokul M Das
- Correspondence to: Gokul M. Das, PhD, Department of Pharmacology and Therapeutics, Center for Genetics and Pharmacology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 (e-mail: )
| |
Collapse
|
35
|
Gandhi N, Das GM. Metabolic Reprogramming in Breast Cancer and Its Therapeutic Implications. Cells 2019; 8:E89. [PMID: 30691108 PMCID: PMC6406734 DOI: 10.3390/cells8020089] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 12/22/2022] Open
Abstract
Current standard-of-care (SOC) therapy for breast cancer includes targeted therapies such as endocrine therapy for estrogen receptor-alpha (ERα) positive; anti-HER2 monoclonal antibodies for human epidermal growth factor receptor-2 (HER2)-enriched; and general chemotherapy for triple negative breast cancer (TNBC) subtypes. These therapies frequently fail due to acquired or inherent resistance. Altered metabolism has been recognized as one of the major mechanisms underlying therapeutic resistance. There are several cues that dictate metabolic reprogramming that also account for the tumors' metabolic plasticity. For metabolic therapy to be efficacious there is a need to understand the metabolic underpinnings of the different subtypes of breast cancer as well as the role the SOC treatments play in targeting the metabolic phenotype. Understanding the mechanism will allow us to identify potential therapeutic vulnerabilities. There are some very interesting questions being tackled by researchers today as they pertain to altered metabolism in breast cancer. What are the metabolic differences between the different subtypes of breast cancer? Do cancer cells have a metabolic pathway preference based on the site and stage of metastasis? How do the cell-intrinsic and -extrinsic cues dictate the metabolic phenotype? How do the nucleus and mitochondria coordinately regulate metabolism? How does sensitivity or resistance to SOC affect metabolic reprogramming and vice-versa? This review addresses these issues along with the latest updates in the field of breast cancer metabolism.
Collapse
Affiliation(s)
- Nishant Gandhi
- Department of Pharmacology and Therapeutics, Center for Genetics & Pharmacology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Gokul M Das
- Department of Pharmacology and Therapeutics, Center for Genetics & Pharmacology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
36
|
Expression of estrogen receptor beta correlates with adverse prognosis in resected pancreatic adenocarcinoma. BMC Cancer 2018; 18:1049. [PMID: 30373552 PMCID: PMC6206939 DOI: 10.1186/s12885-018-4973-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/18/2018] [Indexed: 02/08/2023] Open
Abstract
Background The relevance of estrogen receptor (ER) expression in pancreatic ductal adenocarcinoma (PDAC) is largely unknown. Clinical trials targeting ER with selective estrogen receptor modulators in pancreatic cancer did not show any benefit. Here, we analyze the impact of recently characterized ER isoform beta on survival in a cohort of patients with resected PDAC. Methods Eighty-four patients having undergone pancreatic resection for PDAC at a single institution were identified. Tissue microarrays were constructed of archival tumor specimens. The expression of ER beta was determined by immunohistochemistry and quantified by a system established for estrogen receptor expression in breast cancer. ER beta expression was then correlated with clinicopathological parameters, and univariate and multivariate survival analyses were performed. Results Nuclear expression of ER beta was found in 31% of tumors. No significant correlation was found between ER beta expression and TNM status, tumor grade, age or sex. Univariate analysis revealed nodal metastasis and the expression of ER beta as factors correlating with a shorter overall survival and disease free survival. When comparing ER beta expression in patients surviving more than 24 months with those who died from the tumor within 12 or 24 months, respectively, a significantly lower ER beta expression was found in the long term survivors. In multivariate analysis, ER beta expression was demonstrated to be an independent predictor of shorter overall survival. Conclusions In resected PDAC, expression of ER beta seems to correlate with poor prognosis. These data may help to identify patients who may benefit from additional systemic therapy including selective estrogen receptor modulators.
Collapse
|
37
|
Oyenihi OR, Krygsman A, Verhoog N, de Beer D, Saayman MJ, Mouton TM, Louw A. Chemoprevention of LA7-Induced Mammary Tumor Growth by SM6Met, a Well-Characterized Cyclopia Extract. Front Pharmacol 2018; 9:650. [PMID: 29973879 PMCID: PMC6019492 DOI: 10.3389/fphar.2018.00650] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/31/2018] [Indexed: 12/25/2022] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related deaths in women. Chemoprevention of BC by using plant extracts is gaining attention. SM6Met, a well-characterized extract of Cyclopia subternata with reported selective estrogen receptor subtype activity, has shown tumor suppressive effects in a chemically induced BC model in rats, which is known to be estrogen responsive. However, there is no information on the estrogen sensitivity of the relatively new orthotopic model of LA7 cell-induced mammary tumors. In the present study, the potential chemopreventative and side-effect profile of SM6Met on LA7 cell-induced tumor growth was evaluated, as was the effects of 17β-estradiol and standard-of-care (SOC) endocrine therapies, such as tamoxifen (TAM), letrozole (LET), and fulvestrant (FUL). Tumor growth was observed in the tumor-vehicle control group until day 10 post tumor induction, which declined afterward on days 12-14. SM6Met suppressed tumor growth to the same extent as TAM, while LET, but not FUL, also showed substantial anti-tumor effects. Short-term 17β-estradiol treatment reduced tumor volume on days prior to day 10, whereas tumor promoting effects were observed during long-term treatment, which was especially evident at later time points. Marked elevation in serum markers of liver injury, which was further supported by histological evaluation, was observed in the vehicle-treated tumor control, TAM, LET, and long-term 17β-estradiol treatment groups. Alterations in the lipid profiles were also observed in the 17β-estradiol treatment groups. In contrast, SM6Met did not augment the increase in serum levels of liver injury biomarkers caused by tumor induction and no effect was observed on lipid profiles. In summary, the results from the current study demonstrate the chemopreventative effect of SM6Met on mammary tumor growth, which was comparable to that of TAM, without eliciting the negative side-effects observed with this SOC endocrine therapy. Furthermore, the results of this study also showed some responsiveness of LA7-induced tumors to estrogen and SOC endocrine therapies. Thus, this model may be useful in evaluating potential endocrine therapies for hormone responsive BC.
Collapse
Affiliation(s)
- Omolola R. Oyenihi
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Annadie Krygsman
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Nicolette Verhoog
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Dalene de Beer
- Post-Harvest and Agro-Processing Technologies, Agricultural Research Council of South Africa, Infruitec-Nietvoorbij, Stellenbosch, South Africa
- Department of Food Science, Stellenbosch University, Stellenbosch, South Africa
| | - Michael J. Saayman
- Department of Biomedical Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Thys M. Mouton
- Department of Biomedical Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Ann Louw
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
38
|
Dall GV, Hawthorne S, Seyed-Razavi Y, Vieusseux J, Wu W, Gustafsson JA, Byrne D, Murphy L, Risbridger GP, Britt KL. Estrogen receptor subtypes dictate the proliferative nature of the mammary gland. J Endocrinol 2018; 237:323-336. [PMID: 29636363 DOI: 10.1530/joe-17-0582] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Estrogen induces proliferation of breast epithelial cells and is responsible for breast development at puberty. This tightly regulated control is lost in estrogen-receptor-positive (ER+) breast cancers, which comprise over 70% of all breast cancers. Currently, breast cancer diagnosis and treatment considers only the α isoform of ER; however, there is a second ER, ERβ. Whilst ERα mediates estrogen-driven proliferation of the normal breast in puberty and breast cancers, ERβ has been shown to exert an anti-proliferative effect on the normal breast. It is not known how the expression of each ER (alone or in combination) correlates with the ability of estrogen to induce proliferation in the breast. We assessed the levels of each ER in normal mouse mammary glands subdivided into proliferative and non-proliferative regions. ERα was most abundant in the proliferative regions of younger mice, with ERβ expressed most abundantly in old mice. We correlated this expression profile with function by showing that the ability of estrogen to induce proliferation was reduced in older mice. To show that the ER profile associated with breast cancer risk, we assessed ER expression in parous mice which are known to have a reduced risk of developing ERα breast cancer. ERα expression was significantly decreased yet co-localization analysis revealed ERβ expression increased with parity. Parous mice had less unopposed nuclear ERα expression and increased levels of ERβ. These changes suggest that the nuclear expression of ERs dictates the proliferative nature of the breast and may explain the decreased breast cancer risk with parity.
Collapse
Affiliation(s)
| | - Samuel Hawthorne
- Department of Anatomy and Developmental BiologyMonash University, Melbourne, Australia
| | - Yashar Seyed-Razavi
- Department of Anatomy and Developmental BiologyMonash University, Melbourne, Australia
| | | | - Wanfu Wu
- Department of Biology and BiochemistryUniversity of Houston, Houston, Texas, USA
| | - Jan-Ake Gustafsson
- Department of Biology and BiochemistryUniversity of Houston, Houston, Texas, USA
| | - David Byrne
- Department of PathologyPeter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Gail P Risbridger
- Peter MacCallum Cancer CentreMelbourne, Australia
- Department of Anatomy and Developmental BiologyMonash University, Melbourne, Australia
| | - Kara L Britt
- Peter MacCallum Cancer CentreMelbourne, Australia
- The Sir Peter MacCallumDepartment of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
39
|
Abdelmegeed SM, Mohammed S. Canine mammary tumors as a model for human disease. Oncol Lett 2018; 15:8195-8205. [PMID: 29928319 PMCID: PMC6004712 DOI: 10.3892/ol.2018.8411] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/12/2018] [Indexed: 12/13/2022] Open
Abstract
Animal models for examining human breast cancer (HBC) carcinogenesis have been extensively studied and proposed. With the recent advent of immunotherapy, significant attention has been focused on the dog as a model for human cancer. Dogs develop mammary tumors and other cancer types spontaneously with an intact immune system, which exhibit a number of clinical and molecular similarities to HBC. In addition to the spontaneous tumor presentation, the clinical similarities between human and canine mammary tumors (CMT) include the age at onset, hormonal etiology and course of the diseases. Furthermore, factors that affect the disease outcome, including tumor size, stage and lymph node invasion, are similar in HBC and CMT. Similarly, the molecular characteristics of steroid receptor, epidermal growth factor, proliferation marker, metalloproteinase and cyclooxygenase expression, and the mutation of the p53 tumor suppressor gene in CMT, mimic HBC. Furthermore, ductal carcinomas in situ in human and canine mammary glands are particularly similar in their pathological, molecular and visual characteristics. These CMT characteristics and their similarities to HBC indicate that the dog could be an excellent model for the study of human disease. These similarities are discussed in detail in the present review, and are compared with the in vitro and other in vivo animal models available.
Collapse
Affiliation(s)
- Somaia M Abdelmegeed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Sulma Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
40
|
Mishra AK, Abrahamsson A, Dabrosin C. Fulvestrant inhibits growth of triple negative breast cancer and synergizes with tamoxifen in ERα positive breast cancer by up-regulation of ERβ. Oncotarget 2018; 7:56876-56888. [PMID: 27486755 PMCID: PMC5302959 DOI: 10.18632/oncotarget.10871] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 07/16/2016] [Indexed: 12/17/2022] Open
Abstract
The estrogen receptor-alpha (ERα) is used as a predictive marker for anti-estrogen therapy in breast cancer patients. In addition to aromatase inhibitors, ERα can be targeted at the receptor level using the receptor modulator tamoxifen or by the pure anti-estrogen fulvestrant. The role of the second ER, ER-beta (ERβ), as a therapeutic target or prognostic marker in breast cancer is still elusive. Hitherto, it is not known if ERα+/ERβ+ breast cancers would benefit from a treatment strategy combining tamoxifen and fulvestrant or if fulvestrant exert any therapeutic effects in ERα-/ERβ+ breast cancer. Here, we report that fulvestrant up-regulated ERβ in ERα+/ERβ+ breast cancer and in triple negative ERβ+ breast cancers (ERα-/ERβ+). In ERα+/ERβ+ breast cancer, a combination therapy of tamoxifen and fulvestrant significantly reduced tumor growth compared to either treatment alone both in vivo and in vitro. In ERα-/ERβ+ breast cancer fulvestrant had potent effects on cancer growth, in vivo as well as in vitro, and this effect was dependent on intrinsically expressed levels of ERβ. The role of ERβ was further confirmed in cells where ERβ was knocked-in or knocked-down. Inhibition of DNA methyltransferase (DNMT) increased the levels of ERβ and fulvestrant exerted similar potency on DNMT activity as the DNMT inhibitor decitabine. We conclude that fulvestrant may have therapeutic potential in additional groups of breast cancer patients; i) in ERα+/ERβ+ breast cancer where fulvestrant synergizes with tamoxifen and ii) in triple negative/ERβ+ breast cancer patients, a subgroup of breast cancer patients with poor prognosis.
Collapse
Affiliation(s)
- Ameet K Mishra
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Annelie Abrahamsson
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
41
|
Dong W, Li J, Li J, Zhang P, Wang Z, Sun W, Zhang H. Reduced expression of oestrogen receptor-β is associated with tumour invasion and metastasis in oestrogen receptor-α-negative human papillary thyroid carcinoma. Int J Exp Pathol 2018; 99:15-21. [PMID: 29655286 PMCID: PMC5917391 DOI: 10.1111/iep.12266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/20/2018] [Indexed: 12/11/2022] Open
Abstract
Oestrogens play an important role in the development and progression of papillary thyroid carcinoma (PTC) through oestrogen receptor (ER)-α and -β, which may exert different or even opposing actions in PTC. The roles of ERβ in ERα-negative PTC are still not clear. This study investigated the expression dynamics of ERβ1 (wild-type ERβ) and its clinical significance in female ERα-negative PTC patients. ERβ1 expression was detected in thyroid tissues of 136 female patients diagnosed with PTC. The relationships between ERβ1 expression and clinicopathological/biological factors were also analysed in female ERα-negative PTC patients. The total score for ERβ1 was significantly lower in female ERα-negative PTC patients with LNM or ETE when compared to those without LNM or ETE (Z = -2.923, P = 0.003 and Z = -3.441, P = 0.001). Accordingly, the total score for ERβ1 was significantly higher in ERα-negative PTC patients expressing E-cadherin compared to patients negative for E-cadherin expression (Z = -2.636, P = 0.008). The total score was lower in ERα-negative PTC patients positive for VEGF expression compared to those negative for VEGF expression (Z = -1.914, P = 0.056). This preliminary study indicates that reduced expression of ERβ1 in female ERα-negative PTC patients is associated with greater progression of the disease. This may provide insights into the underlying molecular mechanisms of ERβ1 and could help design targeted approaches for treating or even preventing this disease.
Collapse
Affiliation(s)
- Wen‐Wu Dong
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Jian Li
- Department of SurgeryJinqiu Hospital of LiaoningShenyangChina
| | - Jing Li
- Department of Endocrinology and MetabolismInstitute of EndocrinologyLiaoning Provincial Key Laboratory of Endocrine DiseasesThe First Hospital of China Medical UniversityShenyangChina
| | - Ping Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Zhi‐Hong Wang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Wei Sun
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Hao Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
42
|
Girgert R, Emons G, Gründker C. Estrogen Signaling in ERα-Negative Breast Cancer: ERβ and GPER. Front Endocrinol (Lausanne) 2018; 9:781. [PMID: 30687231 PMCID: PMC6333678 DOI: 10.3389/fendo.2018.00781] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/12/2018] [Indexed: 01/22/2023] Open
Abstract
Estrogen receptors are important regulators of the growth of breast tumors. Three different receptors for estrogens have been identified in breast tumors, two nuclear receptors, ERα and ERβ, and a G-protein coupled estrogen receptor 1 (GPER) that initiates non-genomic effects of estrogens in the cytosol. Recent findings show that the stimulation of cytoplasmic ERα and ERβ also triggers non-genomic signaling pathways. The treatment of breast cancer with anti-estrogens depends on the presence of ERα. About 40% of all breast cancers, however, do not express ERα. One subgroup of these tumors overexpress Her-2, another important group is designated as triple-negative breast cancer, as they neither express ERα, nor progesterone receptors, nor do they overexpress Her-2. This review addresses the signaling of ERβ and GPER in ERα-negative breast tumors. In addition to the well-established EGF-receptor transactivation pathways of GPER, more recent findings of GPER-dependent activation of FOXO3a, the Hippo-pathway, and HOTAIR-activation are summarized.
Collapse
|
43
|
Dalasanur Nagaprashantha L, Adhikari R, Singhal J, Chikara S, Awasthi S, Horne D, Singhal SS. Translational opportunities for broad-spectrum natural phytochemicals and targeted agent combinations in breast cancer. Int J Cancer 2017; 142:658-670. [PMID: 28975625 DOI: 10.1002/ijc.31085] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/18/2017] [Accepted: 09/12/2017] [Indexed: 12/17/2022]
Abstract
Breast cancer (BC) prevention and therapy in the context of life-style risk factors and biological drivers is a major focus of developmental therapeutics in oncology. Obesity, alcohol, chronic estrogen signaling and smoking have distinct BC precipitating and facilitating effects that may act alone or in combination. A spectrum of signaling events including enhanced oxidative stress and changes in estrogen-receptor (ER)-dependent and -independent signaling drive the progression of BC. Breast tumors modulate ERα/ERβ ratio, upregulate proliferative pathways driven by ERα and HER2 with a parallel loss and/or downregulation of tumor suppressors such as TP53 and PTEN which together impact the efficacy of therapeutic strategies and frequently lead to emergence of drug resistance. Natural phytochemicals modulate oxidative stress, leptin, integrin, HER2, MAPK, ERK, Wnt/β-catenin and NFκB signaling along with regulating ERα and ERβ, thereby presenting unique opportunities for both primary and combinatorial interventions in BC. In this regard, this article focuses on critical analyses of the evidence from multiple studies on the efficacy of natural phytochemicals in BC. In addition, areas in which the combinations of such effective natural phytochemicals with approved and/or developing anticancer agents can be translationally beneficial are discussed to derive evidence-based inference for addressing challenges in BC control and therapy.
Collapse
Affiliation(s)
| | | | - Jyotsana Singhal
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Shireen Chikara
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Sanjay Awasthi
- Texas Tech University Health Sciences Center, Lubbock, TX
| | - David Horne
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Sharad S Singhal
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
44
|
Rangel N, Villegas VE, Rondón-Lagos M. Profiling of gene expression regulated by 17β-estradiol and tamoxifen in estrogen receptor-positive and estrogen receptor-negative human breast cancer cell lines. BREAST CANCER-TARGETS AND THERAPY 2017; 9:537-550. [PMID: 29033607 PMCID: PMC5614746 DOI: 10.2147/bctt.s146247] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One area of great importance in breast cancer (BC) research is the study of gene expression regulated by both estrogenic and antiestrogenic agents. Although many studies have been performed in this area, most of them have only addressed the effects of 17β-estradiol (E2) and tamoxifen (TAM) on MCF7 cells. This study aimed to determine the effect of low doses of E2 and TAM on the expression levels of 84 key genes, which are commonly involved in breast carcinogenesis, in four BC cell lines differentially expressing estrogen receptor (ER) α and HER2 (MCF7, T47D, BT474, and SKBR3). The results allowed us to determine the expression patterns modulated by E2 and TAM in ERα+ and ERα− cell lines, as well as to identify differences in expression patterns. Although the MCF7 cell line is the most frequently used model to determine gene expression profiles in response to E2 and TAM, the changes in gene expression patterns identified in ERα+ and ERα− cell lines could reflect distinctive properties of these cells. Our results could provide important markers to be validated in BC patient samples, and subsequently used for predicting the outcome in ERα+ and ERα− tumors after TAM or hormonal therapy. Considering that BC is a molecularly heterogeneous disease, it is important to understand how well, and which cell lines, best model that diversity.
Collapse
Affiliation(s)
- Nelson Rangel
- Department of Medical Sciences, University of Turin, Turin, Italy.,Faculty of Natural Sciences and Mathematics, Universidad del Rosario, Bogotá, Colombia
| | - Victoria E Villegas
- Faculty of Natural Sciences and Mathematics, Universidad del Rosario, Bogotá, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| |
Collapse
|
45
|
Ma R, Karthik GM, Lövrot J, Haglund F, Rosin G, Katchy A, Zhang X, Viberg L, Frisell J, Williams C, Linder S, Fredriksson I, Hartman J. Estrogen Receptor β as a Therapeutic Target in Breast Cancer Stem Cells. J Natl Cancer Inst 2017; 109:1-14. [PMID: 28376210 PMCID: PMC5441302 DOI: 10.1093/jnci/djw236] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 09/20/2016] [Indexed: 12/14/2022] Open
Abstract
Background Breast cancer cells with tumor-initiating capabilities (BSCs) are considered to maintain tumor growth and govern metastasis. Hence, targeting BSCs will be crucial to achieve successful treatment of breast cancer. Methods We characterized mammospheres derived from more than 40 cancer patients and two breast cancer cell lines for the expression of estrogen receptors (ERs) and stem cell markers. Mammosphere formation and proliferation assays were performed on cells from 19 cancer patients and five healthy individuals after incubation with ER-subtype selective ligands. Transcriptional analysis was performed to identify pathways activated in ERβ-stimulated mammospheres and verified using in vitro experiments. Xenograft models (n = 4 or 5 per group) were used to study the role of ERs during tumorigenesis. Results We identified an absence of ERα but upregulation of ERβ in BSCs associated with phenotypic stem cell markers and responsible for the proliferative role of estrogens. Knockdown of ERβ caused a reduction of mammosphere formation in cell lines and in patient-derived cancer cells (40.7%, 26.8%, and 39.1%, respectively). Gene set enrichment analysis identified glycolysis-related pathways (false discovery rate < 0.001) upregulated in ERβ-activated mammospheres. We observed that tamoxifen or fulvestrant alone was insufficient to block proliferation of patient-derived BSCs while this could be accomplished by a selective inhibitor of ERβ (PHTPP; 53.7% in luminal and 45.5% in triple-negative breast cancers). Furthermore, PHTPP reduced tumor initiation in two patient-derived xenografts (75.9% and 59.1% reduction in tumor volume, respectively) and potentiated tamoxifen-mediated inhibition of tumor growth in MCF7 xenografts. Conclusion We identify ERβ as a mediator of estrogen action in BSCs and a novel target for endocrine therapy.
Collapse
Affiliation(s)
- Ran Ma
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Govindasamy-Muralidharan Karthik
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - John Lövrot
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Felix Haglund
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| | - Gustaf Rosin
- Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Anne Katchy
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Xiaonan Zhang
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Viberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Jan Frisell
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.,Science for Life Laboratory, Department of Proteomics, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Stig Linder
- Department of Medical and Health Sciences, Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - Irma Fredriksson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| |
Collapse
|
46
|
Witusik-Perkowska M, Zakrzewska M, Sikorska B, Papierz W, Jaskolski DJ, Szemraj J, Liberski PP. Glioblastoma-derived cells in vitro unveil the spectrum of drug resistance capability - comparative study of tumour chemosensitivity in different culture systems. Biosci Rep 2017; 37:BSR20170058. [PMID: 28522553 PMCID: PMC5964726 DOI: 10.1042/bsr20170058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/05/2017] [Accepted: 05/18/2017] [Indexed: 12/28/2022] Open
Abstract
Resistance to cancer drugs is a complex phenomenon which could be influenced by in vitro conditions. However, tumour-derived cell cultures are routinely used for studies related to mechanisms of drug responsiveness or the search for new therapeutic approaches. The purpose of our work was to identify the potential differences in drug resistance and response to treatment of glioblastoma with the use of three in vitro models: traditional adherent culture, serum-free spheroid culture and novel adherent serum-free culture.The experimental models were evaluated according to 'stemness state' and epithelial-to-mesenchymal transition (EMT) status, invasion capability and their expression pattern of genes related to the phenomenon of tumour drug resistance. Additionally, the response to drug treatments of three different culture models was compared with regard to the type of cell death.Multi-gene expression profiling revealed differences between examined culture types with regard to the expression pattern of the selected genes. Functionally, the examined genes were related to drug resistance and metabolism, DNA damage and repair and cell cycle control, and included potential therapeutic targets.Cytotoxicity analyses confirmed that environmental factors can influence not only the molecular background of glioblastoma drug-resistance and efficiency of treatment, but also the mechanisms/pathways of cell death, which was reflected by a distinct intensification of apoptosis and autophagy observed in particular culture models. Our results suggest that parallel exploitation of different in vitro experimental models can be used to reveal the spectrum of cancer cell resistance capability, especially regarding intra-heterogeneous glioblastomas.
Collapse
Affiliation(s)
- Monika Witusik-Perkowska
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Magdalena Zakrzewska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Beata Sikorska
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Wielislaw Papierz
- Department of Pathomorphology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| | - Dariusz J Jaskolski
- Department of Neurosurgery and Oncology of Central Nervous System, Medical University of Lodz, Barlicki University Hospital, Kopcińskiego 22, Lodz 90-153, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Pawel P Liberski
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland
| |
Collapse
|
47
|
de Andrade Natal R, Derchain SF, Pavanello M, Paiva GR, Sarian LO, Vassallo J. Expression of unusual immunohistochemical markers in mucinous breast carcinoma. Acta Histochem 2017; 119:327-336. [PMID: 28336164 DOI: 10.1016/j.acthis.2017.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Mucinous breast carcinoma is characterized by the production of variable amounts of mucin. Some studies have addressed immunohistochemical characterization of mucinous breast carcinoma using a limited set of antibodies. However, the purpose of the present study was to investigate a larger panel of markers not widely used in daily practice and to determine their pathological implications. METHODS Forty patients diagnosed with mucinous breast carcinoma were enrolled. An immunohistochemical study was performed on whole sections of paraffin embedded tissue, using antibodies for the following markers: estrogen receptor alpha and beta, progesterone receptor, androgen receptor, HER2, EGFR, Ki-67, E-cadherin, β-catenin, p53, chromogranin, synaptophysin, GCDFP15, mammaglobin, and CDX2. RESULTS The pure mucinous type was more prevalent in older patients and more frequently expressed GCDFP15. Capella type B presented more frequently with a high Ki-67 index and neuroendocrine differentiation. Although there was a lower frequency of vascular invasion and lymph node metastases in the pure type, the difference was not statistically significant. No case expressed CDX2 (a marker for gastrointestinal tumors), while 85% of the cases expressed at least one of the two typical breast markers (GCDFP15 and mammaglobin), suggesting that these markers may be reliably used for differential diagnosis. Expression of estrogen receptor beta was related to the presence of mucin cell producing lymph node metastasis, with potential prognostic and predictive value. CONCLUSION our findings support the immunohistochemical homogeneity of mucinous breast carcinomas because only minor differences were found when subgrouping them into Capella types A and B or into types pure and mixed.
Collapse
|
48
|
Nelson AW, Groen AJ, Miller JL, Warren AY, Holmes KA, Tarulli GA, Tilley WD, Katzenellenbogen BS, Hawse JR, Gnanapragasam VJ, Carroll JS. Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity. Mol Cell Endocrinol 2017; 440:138-150. [PMID: 27889472 PMCID: PMC5228587 DOI: 10.1016/j.mce.2016.11.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/01/2016] [Accepted: 11/20/2016] [Indexed: 11/20/2022]
Abstract
Estrogen Receptor-β (ERβ) has been implicated in many cancers. In prostate and breast cancer its function is controversial, but genetic studies implicate a role in cancer progression. Much of the confusion around ERβ stems from antibodies that are inadequately validated, yet have become standard tools for deciphering its role. Using an ERβ-inducible cell system we assessed commonly utilized ERβ antibodies and show that one of the most commonly used antibodies, NCL-ER-BETA, is non-specific for ERβ. Other antibodies have limited ERβ specificity or are only specific in one experimental modality. ERβ is commonly studied in MCF-7 (breast) and LNCaP (prostate) cancer cell lines, but we found no ERβ expression in either, using validated antibodies and independent mass spectrometry-based approaches. Our findings question conclusions made about ERβ using the NCL-ER-BETA antibody, or LNCaP and MCF-7 cell lines. We describe robust reagents, which detect ERβ across multiple experimental approaches and in clinical samples.
Collapse
Affiliation(s)
- Adam W Nelson
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK; Academic Urology Group, Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK; Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Arnoud J Groen
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Jodi L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Anne Y Warren
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Kelly A Holmes
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Gerard A Tarulli
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute Building, School of Medicine, Faculty of Health Sciences, The University of Adelaide, SA 5005, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute Building, School of Medicine, Faculty of Health Sciences, The University of Adelaide, SA 5005, Australia
| | - Benita S Katzenellenbogen
- Departments of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Vincent J Gnanapragasam
- Academic Urology Group, Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK; Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK.
| |
Collapse
|
49
|
Ehinger A, Malmström P, Bendahl PO, Elston CW, Falck AK, Forsare C, Grabau D, Rydén L, Stål O, Fernö M. Histological grade provides significant prognostic information in addition to breast cancer subtypes defined according to St Gallen 2013. Acta Oncol 2017; 56:68-74. [PMID: 27762648 DOI: 10.1080/0284186x.2016.1237778] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The St Gallen surrogate definition of the intrinsic subtypes of breast cancer consist of five subgroups based on estrogen receptor (ER), progesterone receptor (PgR), human epidermal growth factor receptor type 2 (HER2), and Ki-67. PgR and Ki-67 are used for discriminating between the 'Luminal A-like' and 'Luminal B-like (HER2-negative)' subtypes. Histological grade (G) has prognostic value in breast cancer; however, its relationship to the St Gallen subtypes is not clear. Based on a previous pilot study, we hypothesized that G could be a primary discriminator for ER-positive/HER2-negative breast cancers that were G1 or G3, whereas Ki-67 and PgR could provide additional prognostic information specifically for patients with G2 tumors. To test this hypothesis, a larger patient cohort was examined. PATIENTS AND METHODS Six hundred seventy-one patients (≥35 years of age, pT1-2, pN0-1) with ER-positive/HER2-negative breast cancer and complete data for PgR, Ki-67, G, lymph node status, tumor size, age, and distant disease-free survival (DDFS; median follow-up 9.2 years) were included. RESULTS 'Luminal A-like' tumors were mostly G1 or G2 (90%) whereas 'Luminal B-like' tumors were mostly G2 or G3 (87%) and corresponded with good and poor DDFS, respectively. In 'Luminal B-like' tumors that were G1 (n = 23), no metastasis occurred, whereas 14 of 40 'Luminal A-like' tumors that were G3 metastasized. In the G2 subgroup, low PgR and high Ki-67 were associated with an increased risk of distant metastases, hazard ratio (HR) and 95% confidence interval (CI) 1.8 (0.95-3.4) and 1.5 (0.80-2.8), respectively. CONCLUSIONS Patients with ER-positive/HER2-negative/G1 breast cancer have a good prognosis, similar to that of 'Luminal A-like', while those with ER-positive/HER2-negative/G3 breast cancer have a worse prognosis, similar to that of 'Luminal B-like', when assessed independently of PgR and Ki-67. Therapy decisions based on Ki-67 and PgR might thus be restricted to the subgroup G2.
Collapse
Affiliation(s)
- Anna Ehinger
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
- Department of Pathology and Cytology, Blekinge County Hospital, Karlskrona, Sweden
| | - Per Malmström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Pär-Ola Bendahl
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
| | - Christopher W. Elston
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Anna-Karin Falck
- Department of Surgery, Helsingborg Hospital, Helsingborg, Sweden
| | - Carina Forsare
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
| | - Dorthe Grabau
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
- Department of Pathology, Skåne University Hospital, Lund, Sweden
| | - Lisa Rydén
- Division of Surgery, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Olle Stål
- Division of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Mårten Fernö
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund Cancer Center at Medicon Village, Lund University, Lund, Sweden
| | | |
Collapse
|
50
|
Elebro K, Borgquist S, Rosendahl AH, Markkula A, Simonsson M, Jirström K, Rose C, Ingvar C, Jernström H. High Estrogen Receptor β Expression Is Prognostic among Adjuvant Chemotherapy-Treated Patients-Results from a Population-Based Breast Cancer Cohort. Clin Cancer Res 2016; 23:766-777. [PMID: 27810901 DOI: 10.1158/1078-0432.ccr-16-1095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/07/2016] [Accepted: 10/11/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Isoform-specific tumor estrogen receptor β (ERβ) expression may hold prognostic information in breast cancer, especially among endocrine-treated breast cancer patients. The study's purpose was to evaluate ERβ isoform 1 (ERβ1) expression in relation to tumor characteristics, ESR2 genotypes, and prognosis in different treatment groups. EXPERIMENTAL DESIGN A population-based prospective cohort of 1,026 patients diagnosed with primary invasive breast cancer in Lund, Sweden, between October 2002 and June 2012 was followed until June 2014 (median 5 years). Associations between immunohistochemical ERβ1 expression, patient and tumor characteristics, as well as outcome within treatment groups were analyzed. RESULTS Tumor ERβ1 expression was available for 911 patients (89%) and was not associated with ESR2 genotypes. ERβ1 positivity, defined as >75% (ERβ175+, 72.7%), was positively associated with established favorable tumor characteristics. Overall, ERβ175+ was associated with lower risk of breast cancer events [HRadj = 0.60; 95% confidence interval (CI), 0.41-0.89]. The magnitude of the association was larger in patients with ERα- tumors (HRadj = 0.30; 95% CI, 0.12-0.76), compared with ERα+ tumors (HRadj = 0.66; 95% CI, 0.42-1.03). Among the 232 chemotherapy-treated patients, ERβ175+ tumors were associated with lower risk of breast cancer events compared with ERβ175- tumors (HRadj = 0.31; 95% CI, 0.15-0.64). Among the 671 chemonaïve patients, ERβ175 status was not associated with the outcome. CONCLUSIONS High ERβ1 expression was a favorable prognostic marker in this breast cancer cohort, especially in chemotherapy-treated patients, but not in endocrine therapy-treated patients. These results warrant confirmation, preferably via a biomarker study in a previously conducted randomized trial. Clin Cancer Res; 23(3); 766-77. ©2016 AACR.
Collapse
Affiliation(s)
- Karin Elebro
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Plastic and Reconstructive Surgery, Skåne University Hospital, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Oncology and Hematology, Skåne University Hospital, Sweden
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Andrea Markkula
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Maria Simonsson
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Carsten Rose
- CREATE Health and Department of Immunotechnology, Faculty of Engineering, Lund University, Lund, Sweden
| | - Christian Ingvar
- Department of Clinical Sciences Lund, Surgery, Faculty of Medicine, Lund University, and Skåne University Hospital, Sweden
| | - Helena Jernström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Faculty of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|