1
|
Kim J, Munster PN. Estrogens and breast cancer. Ann Oncol 2025; 36:134-148. [PMID: 39522613 DOI: 10.1016/j.annonc.2024.10.824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Estrogens have been associated with an increase in breast cancer risk. Yet emerging clinical and experimental evidence points to progestogens [endogenous progesterone or synthetic progesterone (progestin)] as the primary hormonal driver underlying seemingly estrogen-associated breast cancer risk. Estrogens may contribute to breast cancer risk indirectly by induction of the progesterone receptor and thus amplifying progesterone signaling. Large studies of hormonal contraceptives suggest that the small increase in breast cancer risk from hormonal contraceptives is mainly attributable to progestins, not estrogens. Estrogen-plus-progestin hormone replacement therapy (HRT) has consistently shown an increase in breast cancer risk among postmenopausal women, whereas estrogen-alone HRT has little impact on breast cancer risk in naturally or surgically menopausal women. In particular, the long-term follow-up of the Women's Health Initiative (WHI) randomized trials suggests a benefit of estrogen alone. Recent data further indicate that endogenously elevated estrogen during assisted reproductive technology (ART) exhibits little adverse effect on or potentially a reduction in breast cancer risk and recurrence. Also, accumulating evidence suggests that inhibition of progesterone signaling is a critical mechanism underlying the risk-reducing and therapeutic effects of antiestrogens. Estrogen HRT has shown an array of proven benefits, including ameliorating menopausal symptoms and improving bone health. Collective evidence thus suggests that estrogen HRT is likely to offer health benefits to perimenopausal or postmenopausal women, including breast cancer survivors, as well as young BRCA1/2 carriers with prophylactic oophorectomy for ovarian cancer prevention.
Collapse
Affiliation(s)
- J Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, USA.
| | - P N Munster
- Department of Medicine, Center for BRCA Research, UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, USA
| |
Collapse
|
2
|
Arnone AA, Tsai YT, Cline JM, Wilson AS, Westwood B, Seger ME, Chiba A, Howard-McNatt M, Levine EA, Thomas A, Soto-Pantoja DR, Cook KL. Endocrine-targeting therapies shift the breast microbiome to reduce estrogen receptor-α breast cancer risk. Cell Rep Med 2025; 6:101880. [PMID: 39742868 PMCID: PMC11866439 DOI: 10.1016/j.xcrm.2024.101880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/14/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Studies indicate that breast tissue has a distinct modifiable microbiome population. We demonstrate that endocrine-targeting therapies, such as tamoxifen, reshape the non-cancerous breast microbiome to influence tissue metabolism and reduce tumorigenesis. Using 16S sequencing, we found that tamoxifen alters β-diversity and increases Firmicutes abundance, including Lactobacillus spp., in mammary glands (MGs) of mice and non-human primates. Immunohistochemistry showed that lipoteichoic acid (LTA)-positive bacteria were elevated in tamoxifen-treated breast tissue. In B6.MMTV-PyMT mice, intra-nipple probiotic bacteria injections reduced tumorigenesis, altered metabolic gene expression, and decreased tumor proliferation. Probiotic-conditioned media selectively reduced viability in estrogen receptor-positive (ER+) breast cancer cells and altered mitochondrial metabolism in non-cancerous epithelial cells. Human tumor samples revealed that LTA-positive bacteria negatively correlated with Ki67, suggesting that endocrine therapies influence tumor-associated microbiota to regulate proliferation. Our data indicate that endocrine-targeting therapies modify the breast microbiome, corresponding with a shift in tissue metabolism to potentially reduce ER+ breast cancer risk.
Collapse
Affiliation(s)
- Alana A Arnone
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Yu-Ting Tsai
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian Westwood
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Meghan E Seger
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Akiko Chiba
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Durham VA Medical Center, Department of Surgery, Durham, NC 27705, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Marissa Howard-McNatt
- Department of General Surgery, Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem NC, 27157, USA
| | - Edward A Levine
- Department of General Surgery, Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem NC, 27157, USA
| | - Alexandra Thomas
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
3
|
Arnone AA, Cline JM, Soto-Pantoja DR, Cook KL. Investigating the role of endogenous estrogens, hormone replacement therapy, and blockade of estrogen receptor-α activity on breast metabolic signaling. Breast Cancer Res Treat 2021; 190:53-67. [PMID: 34448090 PMCID: PMC8557185 DOI: 10.1007/s10549-021-06354-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022]
Abstract
Purpose Menopause is associated with an increased risk of estrogen receptor-positive (ER +) breast cancer. To characterize the metabolic shifts associated with reduced estrogen bioavailability on breast tissue, metabolomics was performed from ovary-intact and ovariectomized (OVX) female non-human primates (NHP). The effects of exogenous estrogen administration or estrogen receptor blockade (tamoxifen treatment) on menopause-induced metabolic changes were also investigated. Methods Bilateral ovariectomies were performed on female cynomolgus macaques (Macaca fascicularis) to model menopause. OVX NHP were then divided into untreated (n = 13), conjugated equine estrogen (CEE)-treated (n= 13), or tamoxifen-treated (n = 13) subgroups and followed for 3 years. Aged-matched ovary-intact female NHP (n = 12) were used as a premenopausal comparison group. Metabolomics was performed on snap-frozen breast tissue. Results Changes in several different metabolic biochemicals were noted, particularly in glucose and fatty acid metabolism. Specifically, glycolytic, Krebs cycle, acylcarnitines, and phospholipid metabolites were elevated in breast tissue from ovary-intact NHP and OVX + CEE in relation to the OVX and OVX + tamoxifen group. In contrast, treatment with CEE and tamoxifen decreased several cholesterol metabolites, compared to the ovary-intact and OVX NHP. These changes were accompanied by elevated bile acid metabolites in the ovary-intact group. Conclusion Alterations in estrogen bioavailability are associated with changes in the mammary tissue metabolome, particularly in glucose and fatty acid metabolism. Changes in these pathways may represent a bioenergetic shift in gland metabolism at menopause that may affect breast cancer risk. Supplementary Information The online version contains supplementary material available at 10.1007/s10549-021-06354-w.
Collapse
Affiliation(s)
- Alana A Arnone
- Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA.,Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA. .,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA. .,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
4
|
Dewi FN, Cline JM. Nonhuman primate model in mammary gland biology and neoplasia research. Lab Anim Res 2021; 37:3. [PMID: 33397518 PMCID: PMC7784333 DOI: 10.1186/s42826-020-00053-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
Research on breast cancer pathogenesis, prevention and drug development remains an important field as this disease is still one of the leading causes of cancer death worldwide. Nonhuman primates, particularly macaque species, may serve as a highly translational animal model in breast cancer studies due to their similarity with humans in genetics, anatomy, reproductive and endocrine physiology including mammary gland development profile. The use of nonhuman primates in biomedical research, however, requires high ethical standards and an increasing expectation to improve strategies to replace, reduce and refine their use. Here, we discuss some key features of nonhuman primate mammary gland biology relevant to their strengths and limitations as models in studies of breast development and cancer risk.
Collapse
Affiliation(s)
- Fitriya N Dewi
- Primate Research Center at IPB University, Jl. Lodaya II No.5, Bogor, West Java, 16151, Indonesia.
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| |
Collapse
|
5
|
Lundström E, Virijevic I, Söderqvist G. Progestogen addition with low-dose levonorgestrel intrauterine system in menopausal hormone treatment gives less normal breast tissue proliferation than oral norethisterone acetate or medroxyprogesterone acetate. Horm Mol Biol Clin Investig 2020; 41:hmbci-2019-0051. [PMID: 32735552 DOI: 10.1515/hmbci-2019-0051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/05/2020] [Indexed: 11/15/2022]
Abstract
Background The impact of hormones on the development of breast cancer is despite extensive studies, incompletely understood. Combined estrogen-progestogen treatment augments the risk for breast cancer beyond that of estrogen alone, according to numerous studies. The role of breast cell proliferation as a promoter in the development and growth of breast cancer is well recognized. Materials and methods Seventy-nine patients from three randomised trials were subject to a re-analysis of breast cell proliferation: (1) 22 women received continuous combined treatment with oral estradiol (E2) 2 mg/norethisterone acetate (NETA) 1 mg once daily for 3 months. (2) Thirty-seven women received 2 months of sequential treatment with oral conjugated equine estrogens (CEE) 0.625 mg daily combined with medroxyprogesterone acetate (MPA) 5 mg for 14/28 days of each cycle. (3) Twenty women received oral estradiol-valerate (E2V) 2 mg daily combined with levonorgestrel (LNG) intrauterine system (IUS), 20 μg/24 h for 2 months. Fine needle aspiration (FNA) (studies 1 and 3) and core needle biopsy (CNB) (study 2) were used for the assessment of breast cell proliferation. Results There were no baseline proliferation differences, but at the end of treatment there was a highly significant between-group difference for E2V/LNG IUS versus the other two groups (p = 0.0025). E2/NETA and CEE treatments gave a 4-7-old increase in proliferation during treatment (p = 0.04) and (p = 0.007), respectively, which was absent in the E2V/LNG group, showing a significant correlation with insulin-like growth factor binding protein-3 (IGFBP-3) serum levels. Conclusion E2V in combination with very low serum concentrations of LNG in the IUS gives no increase in proliferation in the normal breast.
Collapse
Affiliation(s)
- Eva Lundström
- Division for Obstetrics and Gynecology, Department of Children's and Women's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Ivana Virijevic
- Department of Obstetrics and Gynecology, Västerås Hospital, Västerås, Sweden
| | - Gunnar Söderqvist
- Division for Obstetrics and Gynecology, Department of Children's and Women's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 76, Stockholm, Sweden.,Division for Obstetrics and Gynecology, Department of Children's and Women's Health, Karolinska Institutet, SE-171 76 Stockholm, Sweden, Phone: +46 73 833 82 22
| |
Collapse
|
6
|
Wang Y, Wei S, Gao T, Yang Y, Lu X, Zhou X, Li H, Wang T, Qian L, Zhao Y, Zou W. Anti-Inflammatory Effect of a TCM Formula Li-Ru-Kang in Rats With Hyperplasia of Mammary Gland and the Underlying Biological Mechanisms. Front Pharmacol 2018; 9:1318. [PMID: 30524280 PMCID: PMC6262177 DOI: 10.3389/fphar.2018.01318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
Li-Ru-Kang (LRK), a formula of eight traditional Chinese medicines (TCM), has been used to treat hyperplasia of mammary gland (HMG) in TCM clinics. However, how LRK works in HMG patients is unclear. To explore the possible mechanisms of LRK against HMG, the network pharmacology was used to screen the potential targets and possible pathways that involved in LRK treated HMG. Rat HMG model induced by estrogen and progesterone was used to further verify the effects of the key molecules of LRK selected from the enriched pathways on HMG. Nipple heights and diameters were measured and uterus index was calculated. The histopathological changes of mammary gland tissue were detected by hematoxylin-eosin (H&E) staining. Western blot was used to detect the phosphorylation of ERK, JNK, and P38. And immunohistochemistry staining was performed to evaluate the levels of estrogen receptor α (ERα), progesterone receptor (PR), nuclear factor-(NF-)κB (p65), interleukin-1β (IL-1β), tumor necrosis factor α (TNF-α), cyclooxygenases 2 (COX-2), inducible nitric oxide synthase (iNOS), 8-hydroxy-2'deoxyguanosine (8-OHdG), and nitrotyrosine (NT). Our results indicate that LRK treatment rescues significantly nipples height and diameter, decreases uterus index and ameliorates HMG. LRK treatment also markedly attenuates the over-expression of IL-1β, TNF-α, COX-2, and iNOS, and suppressed the formation of 8-OHdG and NT. Furthermore, LRK treatment significantly inhibits the phosphorylation of JNK, ERK, and p38 and expression of NF-κB (p65), interestingly, LRK treatment has no effect on the expression of ERα and PR. Our data suggest that the LRK treatment protects the mammary glands from the damage of oxidative stress and inflammation induced by estrogen and progesterone, via suppresses of MAPK/NF-κB signaling pathways without affecting on the expression of ERα and PR.
Collapse
Affiliation(s)
- Yingying Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Shizhang Wei
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Tian Gao
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuxue Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Xiaohua Lu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Xuelin Zhou
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Haotian Li
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Tao Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Liqi Qian
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yanling Zhao
- Department of Pharmacy, 302 Military Hospital of China, Beijing, China
| | - Wenjun Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Jayo MJ, Register TC, Hughes CL, Blas-Machado U, Sulistiawati E, Borgerink H, Johnson CS. Effects of an Oral Contraceptive Combination With or Without Androgen on Mammary Tissues: A Study in Rats. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760000700411] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Manuel J. Jayo
- Pathology Associates International, Advance, North Carolina; the Department of Comparative Medicine, Wake Forest University Medical School, Winston-Salem, North Carolina; Cedars-Sinai Medical Center, Center for Women's Health, Los Angeles, California; the Department of Anatomy, Pathology, and Pharmacology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma; Primate Research Center, Bogor Agricultural University, Bogor, Indonesia
| | | | | | | | | | | | - Christopher S. Johnson
- Pathology Associates International, Advance, North Carolina; the Department of Comparative Medicine, Wake Forest University Medical School, Winston-Salem, North Carolina; Cedars-Sinai Medical Center, Center for Women's Health, Los Angeles, California; the Department of Anatomy, Pathology, and Pharmacology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma; Primate Research Center, Bogor Agricultural University, Bogor, Indonesia
| |
Collapse
|
8
|
Wood CE, Usborne AL, Starost MF, Tarara RP, Hill LR, Wilkinson LM, Geisinger KR, Feiste EA, Cline JM. Hyperplastic and Neoplastic Lesions of the Mammary Gland in Macaques. Vet Pathol 2016; 43:471-83. [PMID: 16846989 DOI: 10.1354/vp.43-4-471] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Macaques provide an important animal model for the study of hormonal agents and their effects on risk biomarkers for breast cancer. A common criticism of this model is that spontaneous breast cancer has rarely been described in these animals. In this report, we characterize 35 mammary gland lesions ranging from ductal hyperplasia to carcinoma in situ and invasive ductal carcinoma in cynomolgus and rhesus macaques. Based on a retrospective analysis, we estimated the lifetime incidence of mammary gland neoplasia in aged female macaques to be about 6%. Hyperplastic lesions (n = 19) occurred segmentally along ducts and included such features as columnar alteration, micropapillary atypia, and fibroadenomatous change. in situ carcinomas (n = 8) included solid, comedo, cribriform, and micropapillary elements, encompassing 4 of the major architectural patterns seen in human lesions. invasive ductal carcinomas (n = 8) were generally solid, with prominent central necrosis and mineralization, often on a background of micropapillary ductal hyperplasia and in situ carcinoma. Cytologic changes of invasive lesions included increased mitoses, nuclear pleomorphism, extensive microinvasion, and stromal desmoplasia. Axillary lymph-node metastases were confirmed in 5 of the 8 invasive carcinomas. on immunohistochemistry, intraductal and invasive carcinomas had increased Ki67/MIB1 and HER2 expression and selective loss of estrogen and progesterone receptors. These findings suggest that breast cancer is an underreported lesion in macaques and highlight unique morphologic and molecular similarities in breast cancer between human and macaque species.
Collapse
MESH Headings
- Animals
- Carcinoma in Situ/genetics
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/pathology
- Carcinoma in Situ/veterinary
- Carcinoma, Ductal/genetics
- Carcinoma, Ductal/metabolism
- Carcinoma, Ductal/pathology
- Carcinoma, Ductal/veterinary
- Female
- Gene Expression
- Genes, erbB-2
- Immunohistochemistry/veterinary
- Ki-67 Antigen/metabolism
- Macaca fascicularis
- Macaca mulatta
- Male
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Monkey Diseases/genetics
- Monkey Diseases/metabolism
- Monkey Diseases/pathology
- Oncogenes
- Receptors, Estrogen/biosynthesis
- Receptors, Progesterone/biosynthesis
- Retrospective Studies
Collapse
Affiliation(s)
- C E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Eigeliene N, Erkkola R, Härkönen P. Comparison of the Effects of the Selective Estrogen Receptor Modulators Ospemifene, Raloxifene, and Tamoxifen on Breast Tissue in Ex Vivo Culture. Methods Mol Biol 2016; 1366:327-336. [PMID: 26585146 DOI: 10.1007/978-1-4939-3127-9_25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Explant tissue culture provides a model for studying the direct effects of steroid hormones, their analogs, and novel hormonally active compounds on normal freshly isolated human breast tissues (HBTs). For this purpose, pre- and postmenopausal HBTs can be maintained in this culture system. The results demonstrate that the morphological integrity of HBT explants can be maintained in tissue culture up to 2 weeks and expression of differentiation markers, steroid hormone receptors, proliferation and apoptosis ratios can be evaluated as a response to hormonal stimulation. This chapter describes an ex vivo culture model that we have applied to study the effects of various hormonally active substances, including 17β-estradiol and selective estrogen receptor modulators (SERMs), on normal human breast tissues.
Collapse
Affiliation(s)
- Natalija Eigeliene
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, Turku, 20520, Finland.
| | - Risto Erkkola
- Department of Obstetrics and Gynecology, Turku University Central Hospital, 20520, Turku, Finland
| | - Pirkko Härkönen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, Turku, 20520, Finland
| |
Collapse
|
10
|
Holtorf K. The Bioidentical Hormone Debate: Are Bioidentical Hormones (Estradiol, Estriol, and Progesterone) Safer or More Efficacious than Commonly Used Synthetic Versions in Hormone Replacement Therapy? Postgrad Med 2015; 121:73-85. [DOI: 10.3810/pgm.2009.01.1949] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
Effects of bazedoxifene, conjugated equine estrogens, and a tissue-selective estrogen complex containing both bazedoxifene and conjugated equine estrogens on cerebral artery atherosclerosis in postmenopausal monkeys. Menopause 2014; 21:8-14. [PMID: 23676638 DOI: 10.1097/gme.0b013e31829370e5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aims to evaluate the effects of a new selective estrogen receptor modulator (bazedoxifene acetate [BZA]) and a tissue-specific estrogen complex (BZA combined with conjugated equine estrogens [CEE]) on the extent and severity of cerebral artery atherosclerosis. METHODS Ninety-eight surgically postmenopausal monkeys (Macaca fascicularis) were fed a moderately atherogenic diet and randomized to receive no treatment or women's equivalent doses of BZA (20 mg/d), CEE (0.45 mg/d), or BZA + CEE. After an experimental period of 20 months (approximately equivalent to 5 years of participant experience), the extent and severity of atherosclerosis in the common carotid artery, carotid bifurcation, internal carotid artery, and basilar artery were determined. Lesion severity was determined using the American Heart Association grading system (grades 0-V). RESULTS BZA had no consistent adverse effects on the extent and severity of atherosclerosis in the cerebral arteries and did not attenuate the beneficial effects of CEE on the severity of common carotid artery atherosclerosis. Although CEE had only modest beneficial effects on the extent of carotid bifurcation atherosclerosis, the severity of lesions and the number of affected cases in the common carotid artery were reduced with CEE treatment. As reported previously, plasma lipid profiles did not differ among the treatment groups. CONCLUSIONS In this long-term (equivalent to 5 human patient-years) nonhuman primate trial, BZA shows no consistent adverse effect on cerebral artery atherosclerosis and does not attenuate the modest beneficial effect of CEE on the common carotid artery. Furthermore, CEE inhibits the development of complicated plaques in the common carotid artery.
Collapse
|
12
|
Penagos Tabares F, Bedoya Jaramillo JV, Ruiz-Cortés ZT. Pharmacological overview of galactogogues. Vet Med Int 2014; 2014:602894. [PMID: 25254141 PMCID: PMC4165197 DOI: 10.1155/2014/602894] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/31/2014] [Indexed: 02/08/2023] Open
Abstract
Galactogogues are substances used to induce, maintain, and increase milk production, both in human clinical conditions (like noninfectious agalactias and hypogalactias) and in massification of production in the animal dairy industry. This paper aims to report the state of the art on the possible mechanisms of action, effectiveness, and side effects of galactogogues, including potential uses in veterinary and human medicine. The knowledge gaps in veterinary clinical practice use of galactogogues, especially in the standardization of the lactogenic dose in some pure drugs and herbal preparations, are reviewed.
Collapse
Affiliation(s)
- Felipe Penagos Tabares
- Biogenesis Research Group, Agrarian Sciences Faculty, University of Antioquia, Medellin, Colombia
| | | | | |
Collapse
|
13
|
Effects of bazedoxifene acetate with and without conjugated equine estrogens on the breast of postmenopausal monkeys. Menopause 2013; 19:1242-52. [PMID: 23103754 DOI: 10.1097/gme.0b013e318252e46d] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Concerns about increased breast cancer risk with estrogen and progestin therapy have led to an increased interest in progestin alternatives. The main objective of this study was to determine if bazedoxifene acetate (BZA), a new selective estrogen receptor modulator, will antagonize the proliferative and transcriptional effects of conjugated equine estrogens (CEE) in the breast. METHODS As part of a 20-month preclinical trial, 95 ovariectomized cynomolgus macaques (Macaca fascicularis) were randomized to receive no treatment or treatment with BZA (20 mg/d), CEE (0.45 mg/d), or BZA and CEE in combination (women's daily equivalent doses). The data presented here include breast effects after 6 months of treatment. Endpoints included histomorphometry, histopathological evaluations, gene microarray assays, polymerase chain reaction quantification of specific estrogen receptor α (ER-α) activity markers, and immunohistochemical detection of sex steroid receptors, and the proliferation marker Ki67. RESULTS BZA + CEE and BZA resulted in significantly less total epithelial density, lobular enlargement, and Ki67 immunolabeling in the terminal ducts compared with CEE alone (P < 0.05 for all). The addition of BZA to CEE antagonized the expression of ER-α-regulated genes such as GREB1 and TFF1 (P < 0.01 for both), whereas BZA alone had minimal effects on ER-α-mediated transcriptional activity. BZA and BZA + CEE did not significantly up-regulate genes related to cell cycle progression and proliferation. BZA with and without CEE also resulted in less lobular and terminal duct ER-α immunolabeling compared with control and CEE (P < 0.0001 for all). CONCLUSIONS These findings demonstrate that BZA given at a clinically relevant dose is an estrogen antagonist in the breast, supporting the idea that CEE + BZA may provide a lower breast cancer risk profile compared with traditional estrogen + progestin therapies.
Collapse
|
14
|
Progestogen levels, progesterone receptor gene polymorphisms, and mammographic density changes: results from the Postmenopausal Estrogen/Progestin Interventions Mammographic Density Study. Menopause 2012; 19:302-10. [PMID: 22105149 DOI: 10.1097/gme.0b013e3182310f9f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Estrogen plus progestin therapy (EPT) in postmenopausal women increases breast cancer risk and mammographic density to a higher extent than does estrogen therapy alone. Data from the randomized placebo-controlled Postmenopausal Estrogen/Progestinv Interventions trial showed that EPT-induced increases in serum estrone and estrone sulfate levels were positively correlated with increases in mammographic density. Here, after adjusting for serum estrone and estrone sulfate levels, we investigated the roles of posttreatment serum progestogen increase and of progesterone receptor gene (PGR) genetic variations on changes in mammographic density. METHODS We measured the percent mammographic density and serum progestogen levels in 280 Postmenopausal Estrogen/Progestin Interventions trial participants randomized to EPT treatment. Analyses of genetic variations in PGR were limited to 260 white women for whom we successfully obtained PGR genotypes. We used linear regression analyses to determine how an increase in progestogen levels and PGR genetic variation influenced mammographic density change after EPT. RESULTS The increase in posttreatment serum progestogen level was positively associated with greater increases in mammographic density after adjustment for covariates (P trend = 0.044). Compared with women in the lowest quartile of serum progestogen level, women in the highest quartile experienced a 3.5% greater increase in mammographic density (P = 0.046). We did not find a strong indication that genetic variation in PGR was associated with mammographic density increase or modified the association with serum progestogen; however, confidence in these null findings is constrained by our small sample size. CONCLUSIONS Our results suggest that higher serum progestogen levels resulting from EPT treatment lead to greater increases in mammographic density.
Collapse
|
15
|
Schwartz E, Holtorf K. Hormone replacement therapy in the geriatric patient: current state of the evidence and questions for the future. Estrogen, progesterone, testosterone, and thyroid hormone augmentation in geriatric clinical practice: part 1. Clin Geriatr Med 2012; 27:541-59. [PMID: 22062440 DOI: 10.1016/j.cger.2011.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This article presents an up-to-date review of the literature on hormone augmentation in the elderly to help primary care physicians better evaluate and utilize hormone replacement and optimization strategies to benefit their patients. The scientific literature suggests that hormone supplementation with estrogen, progesterone, testosterone, growth hormone, and thyroid hormone has the potential to improve quality of life and to prevent, or reverse, the many symptoms and conditions associated with aging, including fatigue, depression, weight gain,frailty, osteoporosis, loss of libido, and heart disease. Possible long-term side effects are also considered.
Collapse
Affiliation(s)
- Erika Schwartz
- Age Management Institute, 200 West 57 Street, New York, NY 10019, USA.
| | | |
Collapse
|
16
|
Distribution of cyclooxygenases 1 and 2 in the uterus and breast of cynomolgus monkeys-effects of hormone treatment. Menopause 2011; 18:1001-9. [PMID: 21540754 DOI: 10.1097/gme.0b013e3182127c9b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE The aim of this study was to compare the distribution and immunoreactivity of cyclooxygenase (COX) 1 and COX-2 in normal uterus and breast after long-term hormone therapy in postmenopausal monkeys. METHODS Female adult cynomolgus macaques were bilaterally ovariectomized 3 months before the initiation of hormone treatment. The animals were either treated (experiment 1) with conjugated equine estrogens (CEE), medroxyprogesterone acetate (MPA), CEE + MPA, or tamoxifen or designated as controls (C). In experiment 2, the animals were either treated with CEE, CEE + MPA, or tibolone or designated as C. Breast tissue and uteri were collected, fixed, and paraffin embedded. Immunohistochemistry assays for COX-1 and COX-2 were performed. RESULTS COX-1 immunostaining was decreased by tamoxifen and CEE treatment in the endometrial stroma and by CEE + MPA in the myometrium. COX-1 immunostaining of the breast epithelia was down-regulated by CEE + MPA, whereas other cell types in the breast seem to be less affected by hormone treatment.COX-2 immunoreactivity in the endometrial stroma was increased by CEE + MPA. In the glandular epithelium, CEE + MPA and tibolone treatment increased COX-2 immunostaining compared with CEE treatment only and no treatment at all (C). No effect from hormone treatment on COX-2 immunostaining was found in the myometrium. COX-2 immunostaining in the glandular epithelium of the breast was, in experiment 2, increased after CEE treatment compared with no treatment (C). No other effects by hormone therapy on COX-2 expression were found in the breast. CONCLUSIONS Our results show that COX-1 and COX-2 are differently distributed and regulated by hormones in the normal uterus and breast of ovariectomized macaques. COX-1 is prevailing in the uterus, whereas COX-2 is dominant in the mammary gland.
Collapse
|
17
|
Allred DC. Molecular Biomarkers of Risk in Premalignancy and Breast Cancer Prevention: Figure 1. Cancer Prev Res (Phila) 2011; 4:1947-52. [DOI: 10.1158/1940-6207.capr-11-0478] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Wood CE, Stavisky RC, Nowak J, Kaplan JR. Stimulatory Adrenocortical Effects of a Selective Estrogen Receptor Modulator in Ovariectomized Female Macaques. Toxicol Pathol 2011; 40:55-61. [DOI: 10.1177/0192623311425509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Here, we report the effects of estrogen and the selective estrogen receptor modulator (SERM) levormeloxifene on adrenocortical measures in ovariectomized female cynomolgus monkeys ( Macaca fascicularis). Animals were randomized into one of five treatment groups, each containing 23 to 26 animals: (1) placebo, (2) 0.016 mg/kg 17β-estradiol (E2), (3) 0.5 mg/kg levormeloxifene (L1), (4) 1.0 mg/kg levormeloxifene (L2), and (5) 5.0 mg/kg levormeloxifene (L3). Treatments were administered orally each day for 18 mo. All doses of levormeloxifene resulted in adrenal weights at least 50% greater than placebo ( p < .0001 for all). The target dose of levormeloxifene (L2) resulted in higher serum concentrations of cortisol (+63%), dehydroepiandrosterone-sulfate (+73%), and androstenedione (+37%) compared with the placebo group ( p < .05 for all). In contrast, E2 resulted in no significant differences in adrenal weight or adrenocortical steroids. Oral E2 and all SERM doses resulted in similar reductions in serum gonadotropins and at least threefold greater uterine weight versus placebo ( p < .0001 for all). Results indicate that the SERM levormeloxifene, in contrast to E2, may have robust stimulatory effects on adrenocortical hormones in a postmenopausal model. These findings warrant further investigation into long-term SERM effects on adrenocortical function.
Collapse
Affiliation(s)
- Charles E. Wood
- Department of Pathology (Section on Comparative Medicine), Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ronda C. Stavisky
- Department of Pathology (Section on Comparative Medicine), Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Jay R. Kaplan
- Department of Pathology (Section on Comparative Medicine), Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
19
|
Hill GD, Moore AB, Kissling GE, Flagler ND, Ney E, Cline JM, Dixon D. Effects of hormonally active agents on steroid hormone receptor expression and cell proliferation in the myometrium of ovariectomized macaques. Toxicol Pathol 2011; 39:508-15. [PMID: 21411722 DOI: 10.1177/0192623311401045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hormone replacement therapy and selective estrogen receptor modulators have been controversial treatment options for postmenopausal women because of their potential health benefits and/or risks. In this study, we determine the effects of the hormonally active compounds, conjugated equine estrogens (CEE), medroxyprogesterone acetate (MPA), CEE + MPA, and tamoxifen (TAM) on the myometrium of ovariectomized macaques. Immunoexpression of estrogen receptor-α (ERα), progesterone receptor (PR), and Ki-67 in the myometrium is assessed. We found no significant difference in ERα myometrial expression in the CEE, MPA, and CEE + MPA treatment groups, but there was a significant decrease in expression in animals administered TAM versus controls. Conjugated equine estrogen-, TAM-, and CEE + MPA-treated animals had significantly increased expression of PR in myometrial cells and there was no difference in PR expression in cells from MPA-treated animals versus control animals. Myometrial cell proliferation did not significantly differ between the controls and any of the treatment groups, although normalized Ki-67 values were somewhat higher in the CEE and TAM groups. These data suggest that ERα and PR expression in the myometrium is influenced by treatment with hormonally active agents.
Collapse
Affiliation(s)
- Georgette D Hill
- Integrated Laboratory Systems (ILS), Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Söderqvist G. Mechanisms for differential effects between natural progesterone and synthetic progestogens on normal breast tissue. Horm Mol Biol Clin Investig 2010; 3:437-40. [DOI: 10.1515/hmbci.2010.067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 11/10/2010] [Indexed: 11/15/2022]
Abstract
AbstractBoth epidemiological studies and experimental data on normal breast tissue suggest increased cancer risk, proliferation and mammographic breast density (MD) during hormone therapy (HT) containing synthetic progestogens in traditional doses, and the relative risk or RR is approximately 1.5–3 (for women treated vs. untreated with the above therapies), proliferation levels of normal breast epithelial cells of around 10% and increase in MD in up to around 50% of women during treatment. Dose-response relationships have been inferred by correlations between progestogens as levonorgestrel, norethisterone acetate and medroxyprogesterone acetate on the one hand and proliferation and/or MD on the other hand, and of indications of lower relative risk of breast cancer with modern low or ultra-low dose HT. In contrast, natural progesterone endogenously during the menstrual cycle has a weak effect and exogenous estrogen in combination with oral micronized progesterone in HT has shown to yield an indifferent effect on proliferation. Furthermore, in epidemiological studies such as the French E3N cohort, these combinations have not shown any risk increase for breast cancer for at least 5 years of treatment. Experimental data supporting or not supporting the view that the main proliferative mechanism for natural progesterone is through binding to its nascent progesterone receptors is discussed as well as the pros and cons that the non-physiological higher proliferation levels induced by synthetic progestogens is mainly mediated through interaction with potent growth factors and their paracrine and/or cell signaling pathways.
Collapse
|
21
|
Clarkson TB, Mehaffey MH. Coronary heart disease of females: lessons learned from nonhuman primates. Am J Primatol 2009; 71:785-93. [PMID: 19382155 DOI: 10.1002/ajp.20693] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The cynomolgus monkey model has contributed to significant advances regarding the understanding of coronary artery atherosclerosis of females. There are currently 8 million women in the United States living with heart disease, necessitating further study and understanding of this leading cause of morbidity and mortality for postmenopausal women. Specifically, studies involving the monkey model have allowed greater understanding of the effect of the stage of reproductive life, time since menopause, and the extent of subclinical atherosclerosis as determinates of estrogen-mediated effects on arteries. Utilizing the commonalities among monkeys and human beings, these studies have shown that postmenopausal atherosclerosis is associated with the premenopausal reproductive timeframe. In addition, monkey studies have shown that estrogen deficiency during the premenopausal stage is extremely relevant regarding the progression of atherosclerosis. After several postmenopausal years, however, studies have shown that estrogen has no beneficial effects on atherosclerosis progression and may, in fact, be deleterious. Studies using the monkey model are currently underway to investigate further uses and possibilities of postmenopausal hormone therapy for treating menopausal symptoms while protecting the breast and uterus and inhibiting the progression of coronary artery atherosclerosis. These studies will hopefully clarify the role of estrogen and eliminate the need for the possibly harmful progestin effects through the use of a highly selective estrogen receptor modulator.
Collapse
Affiliation(s)
- Thomas B Clarkson
- Wake Forest University Primate Center, Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1040, USA.
| | | |
Collapse
|
22
|
Abstract
Soy isoflavones are phytoestrogenic components of dietary soy, which are widely consumed for their potential health benefits. Soy isoflavones appear to decrease breast and endometrial cancer risk in human observational studies, but paradoxically stimulate growth of breast cancer cells in culture and uterine enlargement in rodents. We have shown that these compounds are not estrogenic in cynomolgus monkeys even at relatively high doses, but that they reduce estrogen-induced proliferative responses of the breast and endometrium. This effect may be mediated through estrogen receptor interactions and/or modulation of endogenous estrogen metabolism. Interindividual variation in isoflavone absorption and metabolism contributes to the degree of estrogen antagonistic effect. Our recent studies have also shown that individual isoflavone metabolites such as glyceollins may have unique selective estrogen receptor modulator-like activity, acting as tissue-specific antagonists without agonist activity. Rodent studies and human epidemiologic data suggest that timing of exposure and dose relative to endogenous estrogen concentrations are important determinants of effect, and studies of dietary soy on breast development and pubertal maturation are under way. Because soy isoflavones are both abundant in standard monkey chow diets and widely available as dietary supplements for human beings, these findings have broad relevance to the health of human and nonhuman primates.
Collapse
Affiliation(s)
- J Mark Cline
- Wake Forest University Primate Center, Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1040, USA.
| | | |
Collapse
|
23
|
Wood CE, Lees CJ, Cline JM. Mammary gland and endometrial effects of testosterone in combination with oral estradiol and progesterone. Menopause 2009; 16:466-76. [PMID: 19265727 PMCID: PMC2755604 DOI: 10.1097/gme.0b013e318191747a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The goal of this pilot study was to evaluate the effects of testosterone (T) cotherapy on mammary gland and endometrial measures in a postmenopausal primate model. METHODS Twenty-five surgically postmenopausal cynomolgus monkeys were randomized by social group to receive daily treatment with (1) placebo, (2) oral micronized 17beta-estradiol (1 mg/d equivalent in women) + progesterone (200 mg/d equivalent in women) (E + P), or (3) E + P with T administered via subcutaneous pellets for 8 weeks at a high dose (15 mg) followed by 8 weeks at a low dose (1.5 mg) (E + P + T). The main outcome measures were breast and endometrial epithelial proliferation, as measured by Ki67/MIB1 immunolabeling. RESULTS Intralobular breast proliferation did not differ significantly among groups after 8 weeks of treatment but was marginally higher (P = 0.03) in the E + P + T group after 16 weeks of treatment. No significant increase in proliferation was seen for E + P alone. Comparable changes in mammary gland markers of estrogen-receptor activity were seen for the E + P and E + P + T groups. In the endometrium, the addition of T did not increase endometrial glandular proliferation or estrogen-receptor activity or result in any distinct histologic changes. CONCLUSIONS The findings of this study do not support the idea that T antagonizes the effects of combined hormone therapy on breast proliferation or markers of estrogen-receptor activity. Overall, the short-term effects of T cotherapy on the mammary gland and endometrium were minimal.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA.
| | | | | |
Collapse
|
24
|
Abstract
This review describes the normal biology and physiology of the mammary gland in macaques, including the typical histologic appearance across the life span (development, reproductive maturity, lactation, and senescence). The molecular events regulating breast morphogenesis are described, as well as systemic and local hormonal regulators of mammary gland proliferation, differentiation, and function. Similarities and differences to the human breast are described. Regulatory events are illuminated by discussion of genetically modified mouse models. Tissue response markers, including immunohistochemical markers of proliferation and other hormonally induced changes and studies to date, regarding the effects of exogenous hormones, are briefly summarized. In general, estrogens stimulate progesterone receptor expression and proliferation in the mammary gland, and combinations of estrogens and progestogens cause greater proliferation than estrogens alone. Evaluation of novel chemical agents in macaques requires careful evaluation of age and hormonal context to avoid the confounding effects of mammary gland development, past reproductive history, and other influences on mammary gland morphology. The expression of proliferation markers and progesterone receptors may be used as biomarkers to measure chemically induced hormonal effects.
Collapse
Affiliation(s)
- J Mark Cline
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
25
|
Ma L, Hofling M, Masironi B, von Schoultz B, Cline JM, Sahlin L. Effects of tibolone and conventional HRT on the expression of estrogen and progesterone receptors in the breast. Maturitas 2008; 61:345-9. [PMID: 18980817 DOI: 10.1016/j.maturitas.2008.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 09/12/2008] [Accepted: 09/15/2008] [Indexed: 11/27/2022]
Abstract
OBJECTIVE There is evidence that long-term hormone replacement therapy (HRT) is associated with an increased breast cancer risk. The aim of this study was to assess the effects of tibolone on estrogen and progesterone receptors in comparison to the effects of conventional HRT in the breast of surgically postmenopausal macaques. METHOD Sixty macaques were bilaterally ovariectomized 3 months before hormonal treatment was initiated. The animals were randomized into four treatment groups, including tibolone (TIB), conjugated equine estrogens (CEE), conjugated equine estrogens+medroxyprogesterone acetate (CEE+MPA) and control animals (C). After 2 years treatment, breast tissues were collected, fixed and paraffin embedded. Immunohistochemistry assays with monoclonal antibodies for estrogen receptors (ERalpha and ERbeta) and progesterone receptors (PRA and PRB) were performed. RESULTS The expression of ERalpha was markedly decreased in the CEE+MPA group as compared to C and TIB groups. The TIB group was not different from the C and CEE groups. No significant differences were found for ERbeta immunostaining. The expression of PRA was strongly increased in the TIB group as compared to the C and CEE+MPA groups. Immunostaining of PRB was increased in the CEE and TIB treated animals as compared to both C and CEE+MPA groups. CONCLUSIONS Tibolone increased the expression of both PRA and PRB, without affecting ERalpha and ERbeta expression in the macaque breast. These findings indicate that the effects of tibolone in breast tissue could be mediated via differential regulation of PRA and PRB isoforms and therefore distinct from those observed with conventional HRT.
Collapse
Affiliation(s)
- L Ma
- Division for Reproductive Endocrinology, Department of Woman and Child Health, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
26
|
Comparative effects of oral conjugated equine estrogens and micronized 17β-estradiol on breast proliferation. Menopause 2008; 15:890-8. [DOI: 10.1097/gme.0b013e318168f0ad] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
27
|
Wood CE, Hester JM, Appt SE, Geisinger KR, Cline JM. Estrogen effects on epithelial proliferation and benign proliferative lesions in the postmenopausal primate mammary gland. J Transl Med 2008; 88:938-48. [PMID: 18607345 PMCID: PMC2691895 DOI: 10.1038/labinvest.2008.64] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Proliferative lesions of the mammary gland are risk markers and potential precursors for the development of breast cancer in postmenopausal women. In this study we evaluated mammary epithelial proliferation and proliferative lesions in a group of 63 aged postmenopausal macaques randomized by social group to receive one of three experimental diets for 8 months: (1) control; (2) control with 17beta-estradiol (E2) at the human equivalent dose of 1.0 mg per day; and (3) control with the soy phytoestrogen equol (EQ) at the human equivalent dose of 105 mg per day. In normal mammary epithelium, treatment with E2 but not EQ resulted in greater proliferation, epithelial area, and progesterone receptor expression (P<0.05 for all). Mammary lesions included columnar cell change (26/63), columnar cell hyperplasia with and without atypia (13/63), atypical ductal hyperplasia (6/63), and atypical lobular hyperplasia (3/63). Lesions were most common within terminal ductal lobular units. The prevalence of columnar cell hyperplasia (total and atypical cases) was higher in animals treated with E2 compared to control (P<0.05 for both). Compared to normal mammary epithelium, columnar cell lesions (CCLs) showed greater constitutive expression of estrogen receptor-alpha across all groups (P<0.001) and greater expression of progesterone receptor in response to E2 (P<0.01). Independent of treatment, animals with CCLs on histology had greater gene expression of estrogen receptor-alpha and markers of estrogen receptor activity (trefoil factor 1) and proliferation (gene for Ki67 antigen) at a site contralateral to the CCL (P<0.05 for all). These findings demonstrate that the terminal ductal lobular units of the postmenopausal mammary gland contain morphologically distinct cell populations that may hyperrespond to E2 exposure, resulting in specific types of hyperplastic lesions.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1040, USA.
| | | | | | | | | |
Collapse
|
28
|
Cline JM, Botts S, Lees CJ, Brommage R. Effects of lasofoxifene on the uterus, vagina, and breast in ovariectomized cynomolgus monkeys (Macaca fascicularis). Am J Obstet Gynecol 2008; 199:158.e1-8. [PMID: 18501325 DOI: 10.1016/j.ajog.2008.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 11/21/2007] [Accepted: 02/08/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The purpose of this study was to assess the effects of lasofoxifene on the reproductive system in ovariectomized nonhuman primates. STUDY DESIGN This was a 2-year, randomized study. Adult female macaques (Macaca fascicularis) were assigned randomly into 5 groups: ovariectomized, placebo-treated controls (n = 22); sham-ovariectomized, placebo-treated controls (n = 24); ovariectomized animals given 0.021 mg Premarin kg/d (conjugated equine estrogen; n = 25); lasofoxifene at 1.0 mg/kg/d (n = 23); or lasofoxifene at 5.0 mg/kg/d (n = 25). Outcomes included organ weights and histopathologic findings. RESULTS Lasofoxifene did not increase uterine weight or endometrial thickness and did not change mammary, vaginal, or cervical histologic condition. Mild endometrial fibrosis and cystic change were seen in lasofoxifene-treated animals, in contrast to significant uterine weight increases and endometrial hyperplasia induced by conjugated equine estrogen. CONCLUSION Lasofoxifene did not increase uterine weight and produced minor histologic uterine changes at the doses that were given and had no effect on the breast, vagina, or cervix.
Collapse
|
29
|
Rachoń D, Menche A, Vortherms T, Seidlová-Wuttke D, Wuttke W. Effects of dietary equol administration on the mammary gland in ovariectomized Sprague-Dawley rats. Menopause 2008; 15:340-5. [PMID: 17693898 DOI: 10.1097/gme.0b013e318093df58] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate the effects of dietary equol, a metabolite of soy-derived daidzein or formononetin present in red clover, on the mammary gland of ovariectomized Sprague-Dawley rats. DESIGN Sixty ovariectomized rats were divided into five groups (n = 12) and fed soy-free chow with the addition of equol (50 mg/kg chow and 400 mg/kg chow) or estradiol-3 benzoate (E2B) (4.3 mg/kg chow and 17.3 mg/kg chow). The control group received soy-free chow only. After 3 months animals were killed, blood was collected, and the mammary glands were removed for histological and immunohistochemical evaluation. RESULTS Equol and E2B treatment significantly increased serum equol and 17beta-estradiol concentrations, respectively. Serum prolactin in animals treated with high-dose equol was also significantly higher than in the controls. Animals treated with high-dose equol had a significantly higher number of terminal ducts and type II lobules compared with controls. This was also apparent in animals treated with low- and high-dose E2B, but a higher number of type I lobules also was seen. Compared with controls, animals treated with high-dose equol had a significantly higher percentage of proliferating cell nuclear antigen-positive cells in terminal ducts and type II lobules. The percentage of progesterone receptor-positive cells in animals treated with high-dose equol was significantly higher only in type II lobules. In animals treated with low- and high-dose E2B, the percentage of proliferating cell nuclear antigen- and progesterone receptor-positive cells was significantly higher in all the mammary structures. Low-dose equol did not have any effects on the parameters listed above. CONCLUSIONS High-dose dietary equol administration to ovariectomized rats exerts clear mammotropic effects.
Collapse
Affiliation(s)
- Dominik Rachoń
- Department of Clinical and Experimental Endocrinology, University of Göttingen, Göttingen, Germany.
| | | | | | | | | |
Collapse
|
30
|
Engman M, Skoog L, Soderqvist G, Gemzell-Danielsson K. The effect of mifepristone on breast cell proliferation in premenopausal women evaluated through fine needle aspiration cytology. Hum Reprod 2008; 23:2072-9. [DOI: 10.1093/humrep/den228] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
31
|
Crabtree JS, Peano BJ, Zhang X, Komm BS, Winneker RC, Harris HA. Activity of three selective estrogen receptor modulators on hormone-dependent responses in the mouse uterus and mammary gland. Mol Cell Endocrinol 2008; 287:40-6. [PMID: 18367319 DOI: 10.1016/j.mce.2008.01.027] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 01/18/2008] [Accepted: 01/22/2008] [Indexed: 11/27/2022]
Abstract
Selective estrogen receptor modulators (SERMs) have the unique potential to provide estrogenic effects in the skeletal and cardiovascular system, while minimizing/eliminating side effects on reproductive organs. However, despite the unifying characteristic of mixed estrogen receptor (ER) agonist/antagonist activity, compounds within this class are not interchangeable. In order to define and compare the effects of SERMs on different hormone-responsive tissues, we evaluated effects of bazedoxifene acetate (BZA), lasofoxifene (LAS) and raloxifene (RAL) in the mammary gland and uterus of the ovariectomized mouse. Endpoints measured included those regulated by estradiol alone (uterine wet weight, uterine G protein-coupled receptor 105 (GPR105) mRNA expression and mammary gland indoleamine-pyrrole 2,3 dioxygenase (INDO) mRNA expression) as well as others that required the combination of estradiol and progesterone (uterine serine protease inhibitor Kazal type 3 (Spink3) mRNA expression, mammary gland morphology and mammary gland defensin beta1 (Defbeta1) mRNA expression). The three SERMs tested had variable agonist and antagonist activity on these endpoints. In the uterus, the SERMs were mixed agonists/antagonists on estradiol-induced wet weight increase, whereas all three SERMs were estrogen receptor antagonists on GPR105 mRNA expression. However, in the presence of progesterone, BZA and RAL were agonists on Spink3 expression, while LAS was primarily an antagonist. In the mammary gland, BZA and RAL were predominantly agonists on the endpoint of mammary morphology and all three SERMs were clear agonists on Defbeta1 mRNA expression, an E+P-dependent marker. Finally, LAS and RAL had mixed agonist/antagonist activity on INDO mRNA expression, while BZA had only antagonist activity. These results demonstrate that compounds with small structural differences can elicit distinct biological responses, and that in general, SERMs tended to behave more as antagonists on endpoints requiring estrogen alone and agonists on endpoints requiring the combination of estrogen and progesterone.
Collapse
Affiliation(s)
- Judy S Crabtree
- Endocrinology & Reproductive Disorders Division, Women's Health and Musculoskeletal Biology, Wyeth Research, Collegeville, PA 19426, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Transcriptional profiles of progestogen effects in the postmenopausal breast. Breast Cancer Res Treat 2008; 114:233-42. [DOI: 10.1007/s10549-008-0003-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 04/01/2008] [Indexed: 10/22/2022]
|
33
|
Rodriguez GC, Rimel B, Watkin W, Turbov JM, Barry C, Du H, Maxwell GL, Cline J. Progestin Treatment Induces Apoptosis and Modulates Transforming Growth Factor- in the Uterine Endometrium. Cancer Epidemiol Biomarkers Prev 2008; 17:578-84. [DOI: 10.1158/1055-9965.epi-07-0551] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
34
|
Stute P, Szuwart T, Schlueter M, Götte M, Packeisen J, Kiesel L. Effects of hormone therapy on estrogen synthesis from E1S in the mammary gland of postmenopausal women. Maturitas 2008; 59:163-73. [DOI: 10.1016/j.maturitas.2007.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 12/07/2007] [Accepted: 12/13/2007] [Indexed: 10/22/2022]
|
35
|
Stute P, Register TC, Blair RM, Cline JM. Effects of tibolone on estrogen biosynthesis in the mammary tissue of postmenopausal monkeys. Menopause 2008; 13:232-40. [PMID: 16645537 DOI: 10.1097/01.gme.0000198487.55456.0e] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To evaluate the long-term effects of tibolone on estrone sulfate (E1S)-sulfatase activity in breast tissue in a primate model (Macaca fascicularis) in comparison with conventional hormone therapies. DESIGN Ovariectomized female animals (n = 112) were randomized into five groups and treated for 2 years. Treatment included tibolone at 0.05 mg/kg (LoTib, n = 23) or 0.2 mg/kg (HiTib, n = 23), conjugated equine estrogens at 0.042 mg/kg (CEE, n = 24), CEE + medroxyprogesterone acetate at 0.167 mg/kg (CEE+MPA, n = 21), or placebo (controls, n = 21). E1S-sulfatase activity was evaluated by incubating homogenized breast tissue with [H]-E1S. Thin-layer chromatography was performed to separate the products estrone (E1) and estradiol (E2). Histomorphometry was performed to measure the amount of epithelial and fat tissue in the mammary gland. RESULTS Significantly more E2 than E1 was produced in all groups. E1S-sulfatase activity did not differ among the groups. E1S-sulfatase activity was highest in HiTib animals with less fatty breasts (5.9 fmol total estrogen/mg of protein/min; P < or =0.05) and lowest in HiTib animals with more fatty breasts (2.8 fmol total estrogen/mg of protein/min; P = 0.004 relative to less fatty breasts). CONCLUSIONS We conclude that tibolone had a differential effect on local estrogen biosynthesis in animals with high and low breast fat content. Therefore, breast tissue composition affects the steroidogenic response to hormonal treatment.
Collapse
Affiliation(s)
- Petra Stute
- Department of Obstetrics and Gynecology, University of Münster, Münster, Germany
| | | | | | | |
Collapse
|
36
|
Wood CE, Hester JM, Cline JM. Mammary gland development in early pubertal female macaques. Toxicol Pathol 2008; 35:795-805. [PMID: 17943653 DOI: 10.1080/01926230701584213] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Ductal morphogenesis of the mammary gland occurs largely in the early pubertal period under the influence of ovarian sex hormones. In this study we characterized developing ductal structures in breast tissue of 14 female prepubertal or early pubertal rhesus monkeys (average age, 2.6 +/- 0.1 years). Serum estradiol concentrations ranged from < 5.0 to 61.2 pg/ml, while progesterone concentrations were low in all animals (< 0.6 ng/ml). No corpora lutea were seen on ovarian histology. The most immature mammary glands contained multilayered ducts frequently ending in distinctive terminal end bud structures. Transitional ducts had variably stratified columnar epithelium, often with prominent rounded myoepithelial cells and loose periductal stroma. Mature ducts had simple cuboidal luminal epithelium, flattened myoepithelium, and associated lobular development, typical of the premenopausal breast. Expression of estrogen receptor alpha was significantly higher in immature ducts compared to mature ducts, while progesterone receptor expression, epithelial proliferation, and cytokeratin expression did not show a relationship to the maturity of the ducts. These findings identify dynamic morphologic changes occurring in the primate breast during early puberty and suggest that estradiol (rather than progesterone) is the predominant hormonal stimulus for early ductal and lobular development.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | |
Collapse
|
37
|
Wood CE, Sitruk-Ware RL, Tsong YY, Register TC, Lees CJ, Cline JM. Effects of estradiol with oral or intravaginal progesterone on risk markers for breast cancer in a postmenopausal monkey model. Menopause 2007; 14:639-47. [PMID: 17224855 DOI: 10.1097/01.gme.0000247017.41007.80] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate the effects of oral estradiol given with either oral or intravaginal micronized progesterone (P4) on risk biomarkers for breast cancer in a postmenopausal monkey model. DESIGN This experiment was a two-way crossover study in which 20 ovariectomized adult female cynomolgus macaques were treated (in equivalent doses for women) with oral estradiol (1 mg/d) + oral micronized P4 (200 mg/d) or intravaginal P4 delivered by Silastic rings (6- to 10-mg/d release rate). Hormone treatments lasted 2 months and were separated by a 1-month washout period. The primary outcome measure was breast epithelial proliferation. RESULTS Serum P4 concentrations were significantly greater in subjects receiving oral P4 (10.9 ng/mL) compared with intravaginal P4 (3.8 ng/mL) at 2 to 3 hours after oral dosing (P<0.0001) but not at 24 to 28 hours after oral dosing (2.9 ng/mL for oral P4 vs 3.2 ng/mL for intravaginal P4 at 2 months, P=0.19). Serum estradiol concentrations were significantly lower after oral P4 than after intravaginal P4 (P<0.05 for all time points). Oral P4 resulted in significantly decreased body weight (-2.5%) compared with intravaginal P4 (+3.6%) (P=0.0001). Markers of breast proliferation, sex steroid receptor expression, and endometrial area did not differ significantly between oral P4 and intravaginal P4 treatments (P>0.1 for all). CONCLUSIONS Despite different pharmacodynamic profiles, oral and intravaginal P4 had similar effects on biomarkers in the postmenopausal breast.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Cline JM. Assessing the mammary gland of nonhuman primates: effects of endogenous hormones and exogenous hormonal agents and growth factors. ACTA ACUST UNITED AC 2007; 80:126-46. [PMID: 17443713 DOI: 10.1002/bdrb.20112] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This review provides a summary of the normal biology, development, and morphology of the breast in nonhuman primates (macaques), and of the major published work addressing hormonally-induced changes in the breast of these animals. The mammary glands of macaques are anatomically, developmentally, and physiologically similar to the human breast, with similar expression of sex steroid receptors (estrogen receptors alpha and beta, progesterone receptor A and B, androgen receptors), estrogen dependent markers, and steroid metabolizing enzymes. Genetic similarity between human beings and macaques is high, varying from 95-99% depending on the sequence evaluated. Macaques develop hyperplastic and cancerous lesions of the breast spontaneously, which are similar in type and prevalence to those of human beings. They have a reproductive physiology typical of anthropoid primates, including a distinct menarche and menopause, and a 28-day menstrual cycle. These similarities give unique value to the macaque model for evaluation of the effectiveness and safety of hormonal agents. Such agents considered in this review include estrogens and progestogens, combined therapies such as oral contraceptives and post-menopausal hormone therapies, androgens, selective estrogen receptor modulators, phytoestrogens, prolactin, somatotropin, epidermal growth factor, and other novel agents with hormonal or growth factor-like activity. This review also includes a consideration of selected background changes and typical strategies and markers used for evaluation of experimentally-induced changes, including biopsy-based strategies designed to control for inter-individual variability and minimize numbers of animals used.
Collapse
Affiliation(s)
- J Mark Cline
- Comparative Medicine Clinical Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1040, USA.
| |
Collapse
|
39
|
Wood CE, Register TC, Lees CJ, Chen H, Kimrey S, Cline JM. Effects of estradiol with micronized progesterone or medroxyprogesterone acetate on risk markers for breast cancer in postmenopausal monkeys. Breast Cancer Res Treat 2007; 101:125-34. [PMID: 16841178 DOI: 10.1007/s10549-006-9276-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The addition of the synthetic progestin medroxyprogesterone acetate (MPA) to postmenopausal estrogen therapy significantly increases breast cancer risk. Whether this adverse effect is specific to MPA or characteristic of all progestogens is not known. The goal of this study was to compare the effects of oral estradiol (E2) given with either MPA or micronized progesterone (P4) on risk biomarkers for breast cancer in a postmenopausal primate model. For this randomized crossover trial, twenty-six ovariectomized adult female cynomolgus macaques were divided into social groups and rotated randomly through the following treatments (expressed as equivalent doses for women): (1) placebo; (2) E2 (1 mg/day); (3) E2 + P4 (200 mg/day); and (4) E2 + MPA (2.5 mg/day). Hormones were administered orally, and all animals were individually dosed. Treatments lasted two months and were separated by a one-month washout period. The main outcome measure was breast epithelial proliferation, as measured by Ki67 expression. Compared to placebo, E2 + MPA resulted in significantly greater breast proliferation in lobular (P < 0.01) and ductal (P < 0.01) epithelium, while E2 + P4 did not. Intramammary gene expression of the proliferation markers Ki67 and cyclin B1 was also higher after treatment with E2 + MPA (P < 0.01) but not E2 + P4. Both progestogens significantly attenuated E2 effects on body weight, endometrium, and the TFF1 marker of estrogen receptor activity in the breast. These findings suggest that oral micronized progesterone has a more favorable effect on risk biomarkers for postmenopausal breast cancer than medroxyprogesterone acetate.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Conner P. Breast response to menopausal hormone therapy--aspects on proliferation, apoptosis and mammographic density. Ann Med 2007; 39:28-41. [PMID: 17364449 DOI: 10.1080/07853890601039842] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Breast cancer is the major malignancy among women in the Western world. The breast is clearly a target organ for sex steroid hormones and hormonal treatments have been associated with an increased risk of breast cancer. The balance between proliferation and apoptosis is important for breast cell homeostasis. Mammographic breast density has been identified as a strong and independent risk factor for breast cancer. It seems clear that there is a difference between various hormonal treatments with regard to their effects on breast density and cell proliferation. Also, not all women respond similarly to the same treatment. Combined estrogen and progestogen therapy generally will enhance density and proliferation more than treatment with estrogen alone. Certain constitutional and hormonal factors appear to be predictive of breast reactivity. Older women with a low body mass index respond more strongly to treatment. Estrogen levels have a positive and androgens a negative association to increase in density and proliferation. A combination of increased proliferation and decreased apoptosis could be one mechanism to explain the excess risk of breast cancer during combined estrogen/progestogen treatment. Tibolone seems to have less impact on breast response than conventional hormone therapy. Efforts should be made to identify those women with an adverse response to treatment as well as therapeutic principles with the least possible influence on the breast.
Collapse
Affiliation(s)
- Peter Conner
- Department of Obstetrics and Gynecology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
41
|
Eigėlienė N, Härkönen P, Erkkola R. Effects of estradiol and medroxyprogesterone acetate on morphology, proliferation and apoptosis of human breast tissue in organ cultures. BMC Cancer 2006; 6:246. [PMID: 17044944 PMCID: PMC1624847 DOI: 10.1186/1471-2407-6-246] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Accepted: 10/18/2006] [Indexed: 11/21/2022] Open
Abstract
Background Human breast tissue undergoes phases of proliferation, differentiation and regression regulated by changes of the levels of circulating sex hormones during the menstrual cycle or aging. Ovarian hormones also likely play a key role in the etiology and biology of breast cancer. Reports concerning the proliferative effects of steroid hormones on the normal epithelium of human breast have been conflicting. Some studies have shown that steroid hormones may predispose breast epithelial cells to malignant changes by stimulating their proliferation, which is known to be regulated tightly by stromal cells. The aim of this study was to investigate the effects of 17β-estradiol and medroxyprogesterone acetate on proliferation, apoptosis, expression of differentiation markers and steroid hormone receptors in breast epithelium using an in vitro model of freshly isolated human breast tissue, in which a proper interaction of breast epithelium and stroma has been maintained. Methods Human breast tissues were obtained from women undergoing surgery for breast tumours. Peritumoral tissues were excised and explants were cultured for 3 weeks in medium supplemented with E2 or MPA or with E2+MPA. Endpoints included histopathological, histomorphometric and immunohistochemical assessment of the breast explants. Results Culture of breast explants for 14 or 21 days with steroid hormones increased proliferative activity and the thickness of acinar and ductal epithelium. E2-treatment led to hyperplastic epithelial morphology, MPA to hypersecretory single-layered epithelium and E2+MPA to multilayered but organised epithelium. The proliferative response to E2 in comparison to control (p < 0.001) was more pronounced than to MPA (p < 0.05) or E2+MPA (p < 0.05) at 7 and 14 days for Ki-67 and PCNA. E2 treatment also decreased the proportion of apoptotic cells after 7 (p < 0.01) and 14 (p < 0.01) days. In addition, the relative number of ERα, ERβ and PR positive epithelial cells was decreased by all hormonal treatments. Conclusion Organ culture system provides a model for studying the direct effects of steroid hormones and their analogues on postmenopausal human breast tissue. Addition of E2 or MPA or E2+MPA to breast explants caused characteristic changes in morphology, stimulated epithelial proliferation, lowered apoptosis ratio and decreased the relative number of epithelial cells expressing ERα, ERβ and PR.
Collapse
Affiliation(s)
- Natalija Eigėlienė
- Department of Obstetrics and Gynecology, Turku University Central Hospital, 20520 Turku, Finland
- Department of Anatomy, Institute of Biomedicine, University of Turku, 20520 Turku, Finland
- Kaunas University of Medicine, 44307 Kaunas, Lithuania
| | - Pirkko Härkönen
- Department of Anatomy, Institute of Biomedicine, University of Turku, 20520 Turku, Finland
- Department of Laboratory Medicine, Lund University, MASUniversity Hospital, CRS, 20502Malmö, Sweden
| | - Risto Erkkola
- Department of Obstetrics and Gynecology, Turku University Central Hospital, 20520 Turku, Finland
| |
Collapse
|
42
|
Wood CE, Appt SE, Clarkson TB, Franke AA, Lees CJ, Doerge DR, Cline JM. Effects of high-dose soy isoflavones and equol on reproductive tissues in female cynomolgus monkeys. Biol Reprod 2006; 75:477-86. [PMID: 16723506 DOI: 10.1095/biolreprod.106.052142] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Soy isoflavonoids have well-established estrogenic properties in cell culture and rodent models, raising concerns that high isoflavonoid intake may promote development of uterine and breast cancers. To address this concern we evaluated the effects of high-dose isoflavonoid supplements on reproductive tissues in a postmenopausal primate model. Thirty adult female ovariectomized monkeys (Macaca fascicularis) were randomized to receive a control diet 1) alone, 2) with 509 mg/day of the soy isoflavones genistein and daidzein (IF), or 3) with 1020 mg/day of racemic equol (EQ), an isoflavan, for approximately 1 mo. Doses are expressed in aglycone units as calorically scaled human equivalents. Total serum isoflavonoid levels 4 h postfeeding were <20 nmol/L, 2570.7 nmol/L, and 6944.8 nmol/L for control, IF, and EQ groups, respectively. Equol was the predominant serum isoflavonoid in both IF (72.5%) and EQ (99.7%) groups. Aglycones represented 0.9% (IF) and 0.5% (EQ) of total serum isoflavonoids. Histologically, uteri and mammary glands were diffusely atrophic in all groups. Uterine weight, endometrial thickness, glandular area, and epithelial proliferation in the uterus were not significantly different among treatment groups (ANOVA P > 0.1 for all). Endometrial progesterone receptor gene expression was significantly increased in the IF group (P = 0.02), while protein expression was not altered (ANOVA P > 0.1). Within the mammary gland, proliferation and indicators of estrogen exposure did not differ among treatment groups (ANOVA P > 0.1 for all). These findings indicate that high doses of dietary soy isoflavonoids have minimal uterotrophic or mammotrophic effects in an established primate model.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1040, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
McNaught J, Reid RL. Progesterone-Only and Non-Hormonal Contraception in the Breast Cancer Survivor: Joint Review and Committee Opinion of the Society of Obstetricians and Gynaecologists of Canada and the Society of Gynecologic Oncologists of Canada. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2006; 28:616-626. [PMID: 16924781 DOI: 10.1016/s1701-2163(16)32195-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
44
|
McNaught J, Reid RL, Provencher DM, Lea RH, Jeffrey JF, Oza A, Reid RL, Swenerton KD. Contraception hormonale ne contenant qu’un progestatif et contraception non hormonale chez les survivantes du cancer du sein : Une analyse et une opinion de comité communes de la Société des obstétriciens et gynécologues du Canada et de la Société des gynécologues oncologues du Canada. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2006. [DOI: 10.1016/s1701-2163(16)32196-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
45
|
Wood CE, Kaplan JR, Stute P, Cline JM. Effects of soy on the mammary glands of premenopausal female monkeys. Fertil Steril 2006; 85 Suppl 1:1179-86. [PMID: 16616090 DOI: 10.1016/j.fertnstert.2005.08.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Revised: 08/15/2005] [Accepted: 08/15/2005] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To evaluate the interactive effects of dietary soy and menstrual cycle phase on breast proliferation, apoptosis, and sex steroid receptor expression in premenopausal female cynomolgus monkeys. DESIGN Animals were randomized to receive diets with either casein lactalbumin or soy as the protein source for 12 months. Paired follicular and luteal mammary gland biopsies were taken before and after treatment and evaluated for epithelial proliferation and area, apoptosis, and sex steroid receptor expression. SETTING Academic research environment. ANIMAL(S) Fifty-two adult female cynomolgus monkeys (Macaca fascicularis). INTERVENTION(S) Soy protein with the human equivalent of 129 mg/d of isoflavones. MAIN OUTCOME MEASURE(S) Breast proliferation and epithelial area. RESULT(S) Follicular and luteal assessments were taken on cycle day (+/- SE) 5.3 +/- 0.1 and 22.0 +/- 0.1, respectively. Serum E2, serum P, breast proliferation, and vaginal maturation were significantly higher in the luteal vs. follicular phase. Soy treatment resulted in significantly higher serum isoflavone concentrations compared with the casein lactalbumin group but did not significantly affect breast proliferation, epithelial area, apoptosis, or progesterone receptor expression; uterine size; or vaginal maturation in either the follicular or luteal phase. CONCLUSION(S) Results indicate that dietary levels of soy intake have no detectable hormonal effects on the premenopausal breast.
Collapse
Affiliation(s)
- Charles E Wood
- Section on Comparative Medicine, The Comparative Medicine Clinical Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1040, USA.
| | | | | | | |
Collapse
|
46
|
Noh JJ, Maskarinec G, Pagano I, Cheung LWK, Stanczyk FZ. Mammographic densities and circulating hormones: A cross-sectional study in premenopausal women. Breast 2006; 15:20-8. [PMID: 16000251 DOI: 10.1016/j.breast.2005.04.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 03/10/2005] [Accepted: 04/13/2005] [Indexed: 11/19/2022] Open
Abstract
Progestogens appear to influence breast density more than estrogens in postmenopausal women taking hormone replacement therapy (HRT), but little is known about the effect of circulating hormones on mammographic density among premenopausal women. This cross-sectional study explores the relationship of body weight and sex steroids with breast density. Luteal serum samples were analyzed for progesterone, estrone, estradiol, and sex hormone-binding globulin (SHBG). Mammograms were assessed for density using a computer-assisted method. We performed mediation tests using multiple linear regression models. Significant associations of SHBG and estradiol with percentage density disappeared after adjustment for body weight and other covariates, whereas the relationship between progesterone and breast density remained borderline significant. The mediation tests indicated that progesterone has a direct and an indirect effect on mammographic density. Our finding that progesterone shows a stronger association with percentage of mammographic density than estrogen agrees with clinical reports describing denser mammographic patterns among women taking HRT, although these women differ in menopausal status.
Collapse
Affiliation(s)
- Jihae J Noh
- Cancer Research Center of Hawaii, 1236 Lauhala Street, Honolulu, HI 96813, USA
| | | | | | | | | |
Collapse
|
47
|
Wood CE, Register TC, Franke AA, Anthony MS, Cline JM. Dietary soy isoflavones inhibit estrogen effects in the postmenopausal breast. Cancer Res 2006; 66:1241-9. [PMID: 16424064 DOI: 10.1158/0008-5472.can-05-2067] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Soy isoflavones are promising dietary agents for prevention of breast cancer. Isoflavones bind estrogen receptors (ER) and may variably act as either estrogen agonists or antagonists depending on the estrogen environment. In this study, we used a postmenopausal primate model to evaluate interactive effects of dietary soy isoflavones and estrogen on risk markers for breast cancer. The experiment followed a randomized factorial design in which 31 ovariectomized adult female cynomolgus monkeys were divided into social groups of three to four animals each and rotated through eight different diets containing the human equivalent of 0, 60, 120, or 240 mg/d soy isoflavones with a dose of oral micronized 17beta-estradiol (E(2)) corresponding to either a low (0.09 mg/d) or a high (0.5 mg/d) postmenopausal estrogen environment. Treatment periods lasted 4 months with a 1-month washout period between diets. The highest isoflavone dose resulted in significantly lower breast proliferation and uterine size in the high-estrogen environment. These effects were accompanied by divergent changes in breast markers of ER activation in which pS2 expression was significantly lower and progesterone receptor expression was significantly higher following the 240 mg isoflavone dose. All isoflavone doses resulted in lower serum estrone and E(2) concentrations in the high-estrogen environment. In contrast, isoflavone treatment had no significant estrogen agonist effects and minimal antagonistic effects in the lower-estrogen environment. These findings show that in the presence of estrogen higher doses of dietary soy isoflavones may alter ER signaling and induce selective antagonistic effects in the breast.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | | | | | | | | |
Collapse
|
48
|
Wood CE, Clarkson TB, Appt SE, Franke AA, Boue SM, Burow ME, McCoy T, Cline JM. Effects of soybean glyceollins and estradiol in postmenopausal female monkeys. Nutr Cancer 2006; 56:74-81. [PMID: 17176220 DOI: 10.1207/s15327914nc5601_10] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Glyceollins are a novel class of soybean phytoalexins with potential cancer-protective antiestrogenic effects. The purpose of this study was to evaluate the estrogen-antagonist effects of glyceollin-enriched soy protein on biomarkers for breast cancer risk. Thirty female postmenopausal cynomolgus macaques were randomized to one of three dietary treatments for 3 wk: 1) estradiol (E2, 1 mg/day) + casein/lactalbumin (control); 2) E2 + soy protein isolate (SPI) containing 194 mg/day isoflavonoids; and 3) E2 + glyceollin-enriched soy protein (GLY) containing 189 mg/day isoflavonoids + 134 mg/day glyceollins. Doses are expressed in calorically scaled human equivalents. Mean serum glyceollin concentrations at 4 h postfeeding were 134.2 +/- 34.6 nmol/L in the GLY group and negligible in the SPI group (P = 0.0007). Breast proliferation was significantly increased in the control group (+237%, P = 0.01) but not in the SPI group (+198%, P = 0.08) or GLY group (+36%, P = 0.18). Gene expression of trefoil factor 1 and progesterone receptor, two markers of estrogen receptor activity in breast epithelium, were also significantly higher in the control (P < 0.05 for both) but not in the GLY group. These preliminary findings suggest that soybean glyceollins are natural compounds with potential estrogen-modulating properties in the breast.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology/Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Clarkson TB, Appt SE. Controversies about HRT--lessons from monkey models. Maturitas 2005; 51:64-74. [PMID: 15883111 DOI: 10.1016/j.maturitas.2005.02.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 01/18/2005] [Accepted: 01/21/2005] [Indexed: 10/25/2022]
Abstract
Lessons from monkey models contribute significantly to a better understanding of the controversies in reconciling the differences in postmenopausal hormone treatment outcomes between observational and randomized trial data. Monkey studies brought attention to premenopausal estrogen deficiency with resulting premature coronary artery atherosclerosis. Recently, those monkey studies were confirmed for premenopausal women in the NHLBI-sponsored Women's Ischemia Syndrome Evaluation (WISE) Study. Monkey studies have provided convincing evidence for the primary prevention of coronary artery atherosclerosis when estrogens are administered soon after the development of estrogen deficiency. Equally convincing are the data from monkey studies indicating the total loss of these estrogens beneficial effects if treatment is delayed for a period equal to six postmenopausal years for women. An attempt has been made using the monkey model to identify the hormone treatment regimen most effective in preventing the progression of coronary artery atherosclerosis. By a substantial margin, the most effective approach is that of using estrogen containing oral contraceptive during the perimenopausal transition, followed directly by hormone replacement therapy postmenopausally. Because of similarities between human and nonhuman breast, monkeys have had a major role in clarifying controversies surrounding the breast cancer risk of estrogen only versus estrogen plus progestin therapies. The results of monkey studies suggest little or no effects of estrogen only treatment; whereas, estrogen+progestin clearly increases breast cancer risk.
Collapse
Affiliation(s)
- Thomas B Clarkson
- Comparative Medicine Clinical Research Center, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157-1040, USA.
| | | |
Collapse
|
50
|
Conner P, Register TC, Skoog L, Tani E, von Schoultz B, Cline JM. Expression of p53 and markers for apoptosis in breast tissue during long-term hormone therapy in cynomolgus monkeys. Am J Obstet Gynecol 2005; 193:58-63. [PMID: 16021059 DOI: 10.1016/j.ajog.2004.11.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The purpose of this study was to analyze the expression of p53 and markers for apoptosis in breast tissue during long-term hormone therapy in cynomolgus monkeys. STUDY DESIGN Sixty oophorectomized macaques were randomized to receive tibolone, conjugated equine estrogens (CEE), CEE+medroxyprogesterone acetate (MPA), or no hormonal treatment. Breast tissue was collected at necropsy after 2 years, and stained for p53, p21, and cleaved caspase-3. RESULTS Differences were seen between treatment groups. The expression of p53 and caspase-3 was significantly lower in tibolone-treated animals (P<.01). Levels of p53 and caspase-3 were lower in the combined treatment group (CEE+MPA) when compared to both estrogen only and control groups. CONCLUSION Alternative regimens for hormonal therapy seem to have different effects on p53 expression and apoptotic activity within the breast. A combined effect of increased proliferation and decreased apoptosis could be one possible mechanism to explain an increased risk for breast cancer.
Collapse
Affiliation(s)
- Peter Conner
- Department of Obstetrics, Karolinska Hospital, Stockholm, Sweden and Comparative Medicine Clinical Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | | | | | | | | | | |
Collapse
|