Skip to main content

Delineating the role of osteoprotegerin as a marker of breast cancer risk among women with a BRCA1 mutation

Abstract

Women with a pathogenic germline mutation in the BRCA1 gene face a very high lifetime risk of developing breast cancer, estimated at 72% by age 80. Prophylactic bilateral mastectomy is the only effective way to lower their risk; however, most women with a mutation opt for intensive screening with annual MRI and mammography. Given that the BRCA1 gene was identified over 20 years ago, there is a need to identify a novel non-surgical approach to hereditary breast cancer prevention. Here, we provide a review of the emerging preclinical and epidemiologic evidence implicating the dysregulation of progesterone-mediated receptor activator of nuclear factor κB (RANK) signaling in the pathogenesis of BRCA1-associated breast cancer. Experimental studies have demonstrated that RANK inhibition suppresses Brca1-mammary tumorigenesis, suggesting a potential target for prevention. Data from studies conducted among women with a BRCA1 mutation further support this pathway in BRCA1-associated breast cancer development. Progesterone-containing (but not estrogen-alone) hormone replacement therapy is associated with an increased risk of breast cancer in women with a BRCA1 mutation. Furthermore, BRCA1 mutation carriers have significantly lower levels of circulating osteoprotegerin (OPG), the decoy receptor for RANK-ligand (RANKL) and thus endogenous inhibitor of RANK signaling. OPG levels may be associated with the risk of disease, suggesting a role of this protein as a potential biomarker of breast cancer risk. This may improve upon current risk prediction models, stratifying women at the highest risk of developing the disease, and further identify those who may be targets for anti-RANKL chemoprevention. Collectively, the evidence supports therapeutic inhibition of the RANK pathway for the primary prevention of BRCA1-associated breast cancer, which may generate unique prevention strategies (without prophylactic surgery) and enhance quality of life.

Peer Review reports

Clinical management of women with a BRCA1 mutation

Women who inherit a pathogenic germline mutation in the BRCA1 gene face a very high lifetime risk of developing breast cancer, estimated at 72% by age 80 compared to 11% among women in the general population [1, 2]. Current management of these women is limited to either preventive surgery (i.e., prophylactic bilateral mastectomy) or enhanced screening with MRI imaging and mammography [3, 4]. The goal of screening is early detection and whether this modality is a viable alternative to mastectomy has not been established as there are no studies that have compared mortality with MRI screening vs. preventive surgery specifically in this high-risk population. Chemoprevention with selective estrogen receptor modulators such as tamoxifen or aromatase inhibitors is also an option; however, this recommendation is based on data stemming from studies conducted among predominantly non-carriers and there have been no large-scale studies evaluating the effectiveness in the primary prevention of BRCA1-associated disease.

In an international study, Metcalfe and colleagues reported that among 6226 healthy women with a BRCA mutation, 80% were having regular breast screening, 28% had a preventive mastectomy and only 5% took tamoxifen [5]. This suggests that the rates of prophylactic mastectomy remain low and that the majority of BRCA1 mutation carriers opt for intensified screening instead [5,6,7]. Importantly, BRCA1 mutation carriers have strongly expressed their preference for breast cancer risk reduction and desire a novel prevention drug that is currently not available [8]. Given that the BRCA1 gene was identified over 20 years ago, that preventive mastectomy remains the gold standard, and that mutation carriers have strong preferences for chemoprevention, it is timely that an effective breast cancer risk reduction option be identified [9, 10].

In this work, we will review the existing experimental and epidemiological evidence implicating dysregulation of the receptor activator of nuclear factor-κB (RANK) signaling pathway in the predisposition to breast cancer among women with an inherited BRCA1 mutation. An emerging body of data suggests inhibition of RANK as a potential target for prevention in this high-risk population. Furthermore, we (and others) have hypothesized that quantification of circulating osteoprotegerin (OPG), the decoy receptor of RANK-ligand (RANKL), may serve as a potential biomarker of BRCA1-associated breast cancer risk that may not only improve upon current risk prediction models, stratifying women at the highest risk of developing the disease, but may further identify those who may be targets for anti-RANKL chemoprevention.

Emerging role of the RANK signaling pathway in the pathogenesis of BRCA1-associated breast cancer

RANK, RANKL, and, OPG are members of the tumor necrosis factor (TNF) and TNF receptor superfamily [11,12,13]. RANKL can bind to and activate RANK signaling whereas OPG acts as a soluble decoy receptor that binds to RANKL thereby antagonizing RANK/RANKL-mediated signaling [12,13,14]. The RANK pathway was originally identified as an essential regulator of bone resorption and remodeling [15] but is now known to be involved in physiological and pathological roles beyond bone remodeling, including mammary gland development and tumorigenesis [16,17,18,19].

Seminal studies have shown that progesterone-mediated upregulation of the RANK signaling pathway is involved in mammary epithelial proliferation and stem cell expansion and that it also drives tumorigenesis in Brca1 deficient mice [17,18,19,20,21]. An earlier study by Poole et al. showed that inhibition of progesterone signaling with the progesterone antagonist mifepristone prevented mammary tumorigenesis in Brca1-mutant mice [22]. In 2016, two key preclinical studies demonstrated that the inhibition of progesterone-mediated RANK signaling with pharmacological or genetic inactivation suppressed mammary tumor formation in experimental models [23, 24]. Upon examination of mammary tissue in a Brca1 knockout mouse model, Nolan et al. identified a highly proliferative subset of luminal progenitor cells comprising a larger proportion of the total in BRCA1-mutant vs. wildtype mammary tissue [23]. In human mammary tissue specimens, RANK expression was confined to luminal progenitor cells which had a transcriptional signature similar to that of basal-like breast cancers [23]. Furthermore, the authors demonstrated a reduction of progesterone-induced proliferation when RANKL signaling was inhibited with denosumab (a monoclonal antibody to RANKL that is widely used to treat osteoporosis and to prevent skeletal events in breast cancer patients with metastases [25,26,27]) in a 3D-organoid model derived from mammary cells with a mutation in BRCA1 and breast biopsies from BRCA1 mutation carriers [23]. When recombinant OPG (OPG-Fc), a RANKL inhibitor like denosumab, was used for inhibition of RANKL in mice with a Brca1 mutation, the authors also observed reduced mammary tumor growth [23].

In the same year, Sigl et al. demonstrated that genetic inactivation of RANK signaling in mammary epithelial cells reduced the proliferation of mammary progenitor cells and substantially delayed the onset of mammary tumorigenesis in Brca1/p53 mutant mice [24]. Remarkably, about 25% of Rank/Brca1/p53 triple-mutant mice never developed any breast tumors by around day 400 after birth, whereas all Brca1/p53 double-mutant mice developed tumors by around day 200 [24]. Additionally, the authors demonstrated that pharmacological inhibition of RANK signaling with RANK-Fc, which is similar to OPG-Fc, inhibited the development of pre-neoplastic mammary gland lesions in Brca1/p53 transgenic mice [24]. Sigl and colleagues also showed the effectiveness of RANKL blockade in human mammary progenitor activity using tissues of BRCA1 mutation carriers [24]. They reported a significant decrease in vitro clonogenic capacity of progenitor cells after denosumab treatment, highlighting the positive effects of RANKL inhibition to prevent breast cancer [24]. In summary, there is strong evidence indicating the essential role of progesterone-mediated RANK signaling in the expansion of a population of RANK+ luminal progenitors which are the likely cells of origin for BRCA1-associated basal-like breast cancers [28].

Along the same lines, evidence from epidemiological studies also supports progesterone-mediated upregulation of the RANK pathway in the predisposition to breast cancer among women with a BRCA1 mutation. In a study by Widschwendter and colleagues, the authors compared circulating levels of sex hormones (i.e., progesterone and estrogen) as well as OPG and soluble RANKL (sRANKL) in BRCA mutation carriers (n = 391) vs. non-carriers (n = 782). They observed 33% higher (p = 0.007) levels of luteal phase serum estrogen and 121% higher (p = 0.00037) levels of progesterone among BRCA mutation carriers, particularly in BRCA1 mutation carriers, compared with non-carriers across the menstrual cycle [29]. These data support those inherent aberrancies in the levels of sex hormones found in BRCA mutation carriers may be associated with the elevated risk of breast cancer. Moreover, serum OPG levels were inversely associated with luteal phase progesterone levels, particularly among BRCA1/2 mutation carriers (rho = − 0.216; p = 0.002) vs. controls (rho = − 0.098; p = 0.06) [30]. In macaques, administration of combined estrogen and progestin hormone replacement therapy (HRT), but not estrogen-only HRT, was associated with significantly lower levels of OPG in both breast and serum compared to the control animals not exposed to sex hormones [30]. Lower levels of serum OPG were also associated with increased mammary epithelial proliferation in these macaques (rho = − 0.545, p < 0.001), and increased (p = 0.01) levels of OPG were observed in postmenopause [30]. However, RANKL upregulation in mammary tissue samples in response to combination HRT was not reflected in the circulation [30]. These data support circulating OPG, but not necessarily RANKL, as a potential marker of local changes in RANK signaling at the breast tissue level and possibly breast cancer risk (see Section III below).

Findings from clinical trials have also supported the role of progesterone (rather than estrogen) signaling in development of breast cancer. Most notable are data from the Women’s Health Initiative, randomized trials of HRT, which found a positive correlation between combined estrogen-progestin therapy and breast cancer risk (hazard ratio, HR = 1.55; 95% CI 1.41–1.70) [31, 32]. However, no association was found between estrogen-alone HRT and breast cancer risk (HR = 0.77; 95% CI 0.62–0.95) [33]. This central role of progesterone breast cancer development was the conclusion of a recent meta-analysis of the worldwide evidence of HRT and breast cancer risk which summarized among current users, there were definite risks associated with the use of combined therapy vs. estrogen-alone during years 1–4 (estrogen plus progesterone relative risk, RR = 1.60, 95% CI 1.52–1.69; estrogen-only RR = 1.17, 1.10–1.26) [34]. These definite risks increased two-fold during years 5–14 (estrogen plus progesterone RR = 2.08, 2.02–2.15; estrogen-only RR = 1.33, 1.28–1.37) [34]. During years 5–14, the estrogen plus progesterone risks were greater with daily than with less frequent progesterone use (RR = 2.30, 95% CI 2.21–2.40 vs. RR = 1.93, 95% CI 1.84–2.01; heterogeneity p < 0.0001). Interestingly, for BRCA1 mutation carriers, breast cancer risk decreases after menopause when their sex hormones become substantially low [35, 36]. Importantly, in a prospective analysis by our group, we previously showed that combined HRT use following an oophorectomy has been reported to increase the incidence of breast cancer compared to estrogen-alone HRT among 872 BRCA1 mutation carriers who underwent bilateral oophorectomy [37]. The cumulative incidence among those who took progesterone HRT was 22% vs. 12% with the use of estrogen-alone (P-log rank = 0.04). These associations were stronger for women < 45 years at the time of prophylactic oophorectomy. In the same study cohort, we previously showed that oophorectomy was not associated with a reduced risk of breast cancer (primary and contralateral) substantiating less of a role of estrogen in the pathogenesis of BRCA1-associated breast cancer development [38]. Collectively, underlying mechanisms to describe the increased risk of breast cancer likely involve dysregulation of the progesterone-mediated RANK signaling.

Circulating levels of OPG as a potential marker of breast cancer risk: evidence from the general population and BRCA1 mutation carriers

Given the considerable experimental and preclinical data implicating RANK signaling in mammary tumorigenesis, there has been increasing interest in the quantification of either circulating sRANKL, OPG, or even the sRANKL/OPG ratio as potential biomarkers of cancer risk. Table 1 summarizes the key characteristics and findings of the five epidemiological studies that have evaluated the relationship between circulating OPG, sRANKL, or OPG/RANKL ratio and breast cancer risk in the general population. Although based on very few studies published to date, these limited data suggest the potential utility of these biomarkers for disease risk prediction.

Table 1 Overview of studies assessing the relationship between circulating concentrations of sRANKL or OPG and risk of breast cancer among healthy women without a BRCA mutation [28]

Briefly, a prospective study conducted by Vik et al. showed when stratified by age and sex, a significant inverse relationship between serum OPG and breast cancer risk in women under 60 years of age, but not in women above 60 years of age after adjustment, although the sample size was relatively small (upper vs. lower tertile RR = 0.24; 95% CI 0.10–0.61; ptrend = 0.002) [39]. While the authors observed a linear relationship between the overall cancer-related mortality and serum OPG (RR of cancer-related mortality increased by 25% per 1 standard deviation increase in serum OPG, RR 63% higher in upper vs. lower tertile), only 7 out of 6279 cases were related to breast cancer [39]. Interestingly, Vik et al. observed that high serum OPG levels were associated with higher risk of gastrointestinal cancer (HR = 1.79; 95% CI 1.19–2.67) [39].

Similarly, Fortner et al. conducted a case-control study nested in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort to investigate the association between circulating OPG levels and breast cancer risk by hormone receptor type and matched breast cancer cases with healthy controls [40]. The authors found that the high level of circulating OPG was a significant risk factor for estrogen receptor (ER)-negative breast cancer development (top vs. bottom tertile OPG RR ER-negative breast cancer = 1.93; 95% CI 1.24–3.02; ptrend = 0.03) but not for ER-positive breast cancer development (top vs. bottom tertile OPG RR ER-positive breast cancer = 0.84; 95% CI 0.68–1.04; ptrend = 0.18) [40].

In the same year, Kiechl et al. published a case-control study that assessed whether serum OPG and RANKL levels were associated with the risk of developing breast cancer for postmenopausal women without a BRCA mutation germline [41]. In this study, 98 women developed breast cancer and women with high ratio of RANKL/OPG serum levels and high progesterone exhibited a 5.33-fold higher risk of developing breast cancer (OR breast cancer in high RANKL/OPG ratio and high progesterone group = 5.33; 95% CI 1.5–25.4; p = 0.02) [28, 41]. In a subsequent analysis of sRANKL in the EPIC cohort, Sarink et al. observed an inverse association of serum sRANKL/OPG ratio with ER-negative breast cancer (5th vs. 1st quintile RR = 0.60; 95% CI 0.31–1.14; ptrend = 0.03). The authors also observed a positive association between serum sRANKL and ER-positive breast cancer (5th vs. 1st quintile RR = 1.28; 95% CI 1.01–1.63; ptrend = 0.20) but not ER-negative disease [42]. Additional studies are necessary to explain why these associations differ by breast cancer subtype.

Conversely, in a recent study conducted by Kotsopoulos et al., the authors observed no substantial evidence of an association between plasma OPG and breast cancer risk among premenopausal women in a case-control analysis nested within the Nurses’ Health Study II; however, the authors observed a suggestive inverse trend and their analyses may have been underpowered (highest vs. lowest quartile OR = 0.78; 95% CI 0.46–1.33; ptrend = 0.30) [43].

The three studies that have evaluated the relationship of OPG and/or sRANKL with breast cancer risk specifically among women with a BRCA1 or BRCA2 mutation are summarized in Table 2. In the previously described study by Widschwendter et al., the authors reported significantly lower serum OPG concentrations among premenopausal BRCA mutation carriers, particularly in BRCA1 mutation carriers (p = 0.018), compared with non-carriers [30]. They did not measure risk associated with levels of OPG but did find that higher levels of circulating OPG were associated with lower risk pathogenic germline mutations in BRCA1/2 mutation carriers (beta = − 0.058; 95% CI − 0.020, − 0.096; p = 0.003) [30]. In a prospective cohort study by Odén et al., the authors showed that high vs. low plasma OPG was associated with risk of developing breast cancer among BRCA1 and BRCA2 mutation carriers (n = 206; HR = 0.25; 95% CI 0.08–0.78; p = 0.02) [44]. However, there was no association between plasma RANKL and breast cancer risk when stratified into high vs. low plasma RANKL levels (HR = 1.06; 95% CI 0.34–3.28; p = 0.86) [45]. Although limited, the evidence points towards a role of dysregulated RANK signaling in the development of BRCA-associated breast cancer and thus the potential for chemoprevention with a RANKL inhibitor such as denosumab [30]. Furthermore, this suggests that the integration of circulating OPG levels (or the sRANKL/OPG ratio) into risk prediction models may have the potential to identify women who are at the highest risk of developing breast cancer.

Table 2 Summary of studies assessing the relationship between circulating concentrations of sRANKL and/or OPG and risk of breast cancer among BRCA1 and BRCA2 mutation carriers [28]

Non-genetic and genetic regulators of OPG and RANKL expression

Several studies have reported upon or evaluated how various hormonal factors, cytokines, growth factors may influence mRNA expression or protein levels of OPG and sRANKL. For example, estrogen, IL1α, TNFα, and TGFß upregulate OPG expression [46,47,48,49,50] whereas parathyroid hormone and parathyroid hormone-related prostaglandin E2 and glucocorticoids protein downregulate OPG expression [51,52,53,54,55,56,57,58,59,60]. The role of vitamin D3, cytokines and growth factors is less consistent and studies reported mixed results [55, 61,62,63,64,65,66,67,68,69,70]. Overall, OPG expression is upregulated by many suppressors of osteoclast differentiation (i.e., transforming growth factor β, estrogen) and downregulated by some pro-resorptive agents (i.e., parathyroid hormone, prostaglandins) [28, 71]. Most studies report reciprocal regulation of OPG and RANKL, where factors that downregulate expression of OPG will upregulate expression of RANKL, and vice-versa [28, 71]. This reciprocal regulation modulates pro-resorptive compounds to stimulate resorption via RANKL induction while inhibiting effects of OPG [28]. Estrogen, TGFß, and interferon-ɣ downregulate RANKL expression, whereas vitamin D3, parathyroid hormone, and parathyroid hormone-related prostaglandin E2, IL1α, IL1ß, IL11, IL17, TNFα, prostaglandin E2, and glucocorticoids upregulate RANKL expression [46, 47, 51,52,53,54, 57, 59, 64,65,66,67, 69]. Yet, there are some conflicting findings from one publication to another.

Epidemiological studies have also evaluated correlates of OPG. Notable, numerous studies have shown a strong positive correlation between OPG and age in women [40, 72,73,74,75]. The onset of menopause is highly related to age and decreased estrogen levels are one of the contributors to rapid bone loss. It is hypothesized that this decline in estrogen levels can decrease OPG production in osteoblasts and promote bone resorption [47, 48, 76]. Conversely, Sarink et al. found that OPG concentrations in healthy pre- and post-menopausal women in the general population are minimally impacted by hormonal lifestyle factors [77]. Other studies also found mixed, and often weak, associations with OPG levels in healthy women with circulating endogenous sex hormones, such as estradiol, menopause, and HRT use [40, 72, 78, 79].

For high-risk women, it appears that an inherited BRCA1 and BRCA2 are associated with significantly lower circulating OPG levels. As previously described, a BRCA1 or BRCA2 mutation is associated with decreased circulating serum OPG expression. Beyond the BRCA genes, a few studies investigated genetic factors associated with circulating OPG levels [80,81,82,83,84]. Recently, Kwan et al. conducted a meta-analysis of five genome-wide association studies comprising individuals from European and Asian origin (n = 10,336) [82]. The authors discovered two significant genome-wide significant loci (8q23-q24.1, located > 100 kb upstream of the gene that encodes OPG; and 17q11.2) and one locus (14q21.2) with near genome-wide significance associated with circulating OPG levels (mostly OPG serum levels) [82]. They estimated that over half of the heritability of age-adjusted OPG levels could be explained by all SNPs examined in their study [82]. Interestingly, Kwan et al. evaluated the association between a single nulceotide polymorphism (SNP) rs875525 in the ANKH gene and OPG levels however it did not reach genome-wide significance in their meta-analyses [82]. In contrast, Vistoropsky et al. found a significant association between a SNP rs875525 in the ANKH gene and plasma OPG levels from their family-based association study (n = 556) [84]. Collectively, the current literature demonstrates inconsistent findings regarding genetic regulators of OPG expression, which emphasizes the need for additional work on fine-mapping these regions and identifying other casual variants using a large sample size [82].

Overall, our current understanding of factors regulating OPG expression is limited and the conflicting findings among different studies make it challenging to come to a consensus. This is further confounded by the complexity of the RANK signaling pathway that can be regulated by the interaction of various factors and as the changes in serum OPG levels may not reflect the tissue of interest [85]. Nonetheless, OPG is a promising biomarker for BRCA1-associated breast cancer risk due to the association between BRCA1 mutation and OPG levels [30].

Conclusion and future directions

Here we have presented a brief overview of the experimental and epidemiologic evidence suggesting that dysregulation of the progesterone-mediated RANK signaling pathway plays a critical role in the pathogenesis of BRCA1-associated breast cancer. Based on observational data, BRCA1 mutation carriers have significantly lower concentrations of circulating OPG [30], and OPG levels may be associated with the risk of disease. This suggests the potential role of integrating OPG levels (or the sRANKL/OPG ratio) to improve upon cancer risk prediction models. Furthermore, the preclinical and experimental data support therapeutic inhibition of the RANK pathway for the primary prevention of BRCA1-associated breast cancer [23, 24].

Randomized controlled trials are needed to evaluate the clinical effectiveness of denosumab for BRCA1-associated breast cancer prevention in these high-risk women. Whether it is effective for the prevention of ovarian cancer or BRCA2 mutation carriers is not known. It is of interest to see whether this pathway will similarly apply to ovarian cancer. Ultimately, denosumab may provide a necessary, non-surgical, and effective risk reduction intervention for a population at substantial risk for developing breast (and potentially ovarian) cancer. Although a risk-reducing mastectomy is a highly effective procedure for women with a BRCA1 mutation, the low uptake and high opt-out rate for these women for screening instead underlies the need to develop new, non-invasive preventative agents and strategies for managing BRCA1-associated breast cancer risk.

Availability of data and materials

Not applicable.

Abbreviations

BRCA1:

Breast cancer type 1 susceptibility protein

BRCA2:

Breast cancer type 2 susceptibility protein

CI:

Confidence interval

EPIC:

The European Prospective Investigation into Cancer and Nutrition

ER:

Estrogen receptor

HR:

Hazard ratio

HRT:

Hormone replacement therapy

IL:

Interleukin

OPG:

Osteoprotegerin

OR:

Odds ratio

RANK:

Receptor activator of nuclear factor κB

RANKL:

Receptor activator of nuclear factor κB ligand

RR:

Relative risk

SNP:

Single nucleotide polymorphism

sRANKL:

Soluble receptor activator of nuclear factor κB ligand

TGF:

Transforming growth factor

TNF:

Tumor necrosis factor

References

  1. Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of breast, ovarian, and contralateral breast Cancer for BRCA1 and BRCA2 mutation carriers. JAMA. 2017;317(23):2402–16. https://doi.org/10.1001/jama.2017.7112.

    Article  CAS  PubMed  Google Scholar 

  2. Harbeck N, Penault-Llorca F, Cortes J, et al. Breast Cancer. 2019;5. https://doi.org/10.1038/s41572-019-0111-2.

  3. Guidelines NCCN. Breast Cancer (Version 1.2021). Published 2021. Accessed 2 Mar 2021. https://www.nccn.org/professionals/physician_gls/default.aspx#site

  4. Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, et al. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic, version 2.2021. JNCCN J Natl Compr Cancer Netw. 2021;19(1):77–102. https://doi.org/10.6004/JNCCN.2021.0001.

    Article  CAS  Google Scholar 

  5. Metcalfe K, Eisen A, Senter L, et al. International trends in the uptake of cancer risk reduction strategies in women with a BRCA1 or BRCA2 mutation. Br J Cancer. 2019;121(1):15–21. https://doi.org/10.1038/s41416-019-0446-1.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Hartmann LC, Thomas A, Schaid DJ, et al. Efficacy of bilateral. Cancer. 2001;93(21):1633–7.

    CAS  Google Scholar 

  7. Rebbeck TR, Friebel T, Lynch HT, Neuhausen SL, van ’t Veer L, Garber JE, et al. Bilateral prophylactic mastectomy reduces breast cancer risk in BRCA1 and BRCA2 mutation carriers: the PROSE study group. J Clin Oncol. 2004;22(6):1055–62. https://doi.org/10.1200/JCO.2004.04.188.

    Article  PubMed  Google Scholar 

  8. Liede A, Mansfield CA, Metcalfe KA, et al. Preferences for breast cancer risk reduction among BRCA1/BRCA2 mutation carriers: a discrete-choice experiment. Breast Cancer Res Treat. 2017;165(2):433–44. https://doi.org/10.1007/s10549-017-4332-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Easton DF, Narod SA, Ford D, Steel M. The genetic epidemiology of BRCA1. Breast Cancer Linkage Consortium. Lancet (London, England). 1994;344(8924):761. https://doi.org/10.1016/s0140-6736(94)92256-x.

    Article  CAS  Google Scholar 

  10. Tonin P, Serova O, Simard J, Lenoir G, Feunteun J, Morgan K, et al. The gene for hereditary breast-ovarian cancer, BRCA1, maps distal to EDH17B2 in chromosome region 17q12-q21. Hum Mol Genet. 1994;3(9):1679–82. https://doi.org/10.1093/hmg/3.9.1679.

    Article  CAS  PubMed  Google Scholar 

  11. Hanada R, Hanada T, Sigl V, Schramek D, Penninger JM. RANKL/RANK-beyond bones. J Mol Med (Berl). 2011;89(7):647–56. https://doi.org/10.1007/s00109-011-0749-z.

    Article  CAS  Google Scholar 

  12. Nagy V, Penninger JM. The RANKL-RANK Story. Gerontology. 2015;61(6):534–42. https://doi.org/10.1159/000371845.

    Article  CAS  PubMed  Google Scholar 

  13. Kotsopoulos J. BRCA mutations and breast cancer prevention. Cancers (Basel). 2018;10(12):1–15. https://doi.org/10.3390/cancers10120524.

    Article  CAS  Google Scholar 

  14. Kiesel L, Kohl A. Role of the RANK/RANKL pathway in breast cancer. Maturitas. 2016;86:10–6. https://doi.org/10.1016/j.maturitas.2016.01.001.

    Article  CAS  PubMed  Google Scholar 

  15. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Lüthy R, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997;89(2):309–19. https://doi.org/10.1016/S0092-8674(00)80209-3.

    Article  CAS  PubMed  Google Scholar 

  16. Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG system in immunity, bone, and beyond. Front Immunol. 2014;5(OCT):1–11. https://doi.org/10.3389/fimmu.2014.00511.

    Article  CAS  Google Scholar 

  17. Fata JE, Kong YY, Li J, Sasaki T, Irie-Sasaki J, Moorehead RA, et al. The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell. 2000;103(1):41–50. https://doi.org/10.1016/s0092-8674(00)00103-3.

    Article  CAS  PubMed  Google Scholar 

  18. Gonzalez-Suarez E, Jacob AP, Jones J, Miller R, Roudier-Meyer MP, Erwert R, et al. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature. 2010;468(7320):103–7. https://doi.org/10.1038/nature09495.

    Article  CAS  PubMed  Google Scholar 

  19. Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010;468(7320):98–102. https://doi.org/10.1038/nature09387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Joshi PA, Jackson HW, Beristain AG, di Grappa MA, Mote PA, Clarke CL, et al. Progesterone induces adult mammary stem cell expansion. Nature. 2010;465(7299):803–7. https://doi.org/10.1038/nature09091.

    Article  CAS  PubMed  Google Scholar 

  21. Asselin-Labat ML, Vaillant F, Sheridan JM, Pal B, Wu D, Simpson ER, et al. Control of mammary stem cell function by steroid hormone signalling. Nature. 2010;465(7299):798–802. https://doi.org/10.1038/nature09027.

    Article  CAS  PubMed  Google Scholar 

  22. Poole AJ, Li Y, Kim Y, Lin S-CJ, Lee W-H, Lee EY-HP. Prevention of Brca1-Mediated Mammary Tumorigenesis in Mice by a Progesterone Antagonist. Science (80- ). 2006;314(5804):1467–70. https://doi.org/10.1126/science.1130471.

    Article  CAS  Google Scholar 

  23. Nolan E, Vaillant F, Branstetter D, et al. RANK ligand as a potential target for breast cancer prevention in BRCA1-mutation carriers. Nat Med. 2016;22(8):933–9. https://doi.org/10.1038/nm.4118.

    Article  CAS  PubMed  Google Scholar 

  24. Sigl V, Owusu-Boaitey K, Joshi PA, Kavirayani A, Wirnsberger G, Novatchkova M, et al. RANKL/RANK control Brca1 mutation-driven mammary tumors. Cell Res. 2016;26(7):761–74. https://doi.org/10.1038/cr.2016.69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gnant M, Pfeiler G, Dubsky PC, et al. Adjuvant denosumab in breast cancer (ABCSG-18): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet (London, England). 2015;386(9992):433–43. https://doi.org/10.1016/S0140-6736(15)60995-3.

    Article  CAS  Google Scholar 

  26. Stopeck AT, Lipton A, Body J-J, Steger GG, Tonkin K, de Boer RH, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol Off J Am Soc Clin Oncol. 2010;28(35):5132–9. https://doi.org/10.1200/JCO.2010.29.7101.

    Article  CAS  Google Scholar 

  27. Hanley DA, Adachi JD, Bell A, Brown V. Denosumab: mechanism of action and clinical outcomes. Int J Clin Pract. 2012;66(12):1139–46. https://doi.org/10.1111/ijcp.12022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Uzelac A. Investigating the role of BRCA1 in the regulation of Osteoprotegerin. Published online. 2020.

  29. Widschwendter M, Rosenthal AN, Philpott S, Rizzuto I, Fraser L, Hayward J, et al. The sex hormone system in carriers of BRCA1/2 mutations: a case-control study. Lancet Oncol. 2013;14(12):1226–32. https://doi.org/10.1016/S1470-2045(13)70448-0.

    Article  CAS  PubMed  Google Scholar 

  30. Widschwendter M, Burnell M, Fraser L, Rosenthal AN, Philpott S, Reisel D, et al. Osteoprotegerin (OPG), the endogenous inhibitor of receptor activator of NF-κB ligand (RANKL), is dysregulated in BRCA mutation carriers. EBioMedicine. 2015;2(10):1331–9. https://doi.org/10.1016/j.ebiom.2015.08.037.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Chlebowski RT, Manson JE, Anderson GL, et al. Estrogen plus progestin and breast cancer incidence and mortality in the women’s health initiative observational study. J Natl Cancer Inst. 2013;105(8):526–35. https://doi.org/10.1093/jnci/djt043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lammert J, Lubinski J, Gronwald J, Huzarski T, Armel S, Eisen A, et al. Physical activity during adolescence and young adulthood and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat. 2018;169(3):561–71. https://doi.org/10.1007/s10549-018-4694-1.

    Article  CAS  PubMed  Google Scholar 

  33. Anderson GL, Chlebowski RT, Aragaki AK, Kuller LH, Manson JAE, Gass M, et al. Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women’s health initiative randomised placebo-controlled trial. Lancet Oncol. 2012;13(5):476–86. https://doi.org/10.1016/S1470-2045(12)70075-X.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Group C, Cancer B. Type and timing of menopausal hormone therapy and breast cancer risk: individual participant meta-analysis of the worldwide epidemiological evidence. Lancet. 2019;394(10204):1159–68. https://doi.org/10.1016/S0140-6736(19)31709-X.

    Article  Google Scholar 

  35. Kotsopoulos J, Lubinski J, Lynch HT, Neuhausen SL, Ghadirian P, Isaacs C, et al. Age at menarche and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Cancer Causes Control. 2005;16(6):667–74. https://doi.org/10.1007/s10552-005-1724-1.

    Article  PubMed  Google Scholar 

  36. Nolan E, Lindeman GJ, Visvader JE. Out-RANKing BRCA1 in mutation carriers. Cancer Res. 2017;77(3):595–600. https://doi.org/10.1158/0008-5472.CAN-16-2025.

    Article  CAS  PubMed  Google Scholar 

  37. Kotsopoulos J, Gronwald J, Karlan BY, Huzarski T, Tung N, Moller P, et al. Hormone replacement therapy after oophorectomy and breast Cancer risk among BRCA1 mutation carriers. JAMA Oncol. 2018;4(8):1059–65. https://doi.org/10.1001/jamaoncol.2018.0211.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Kotsopoulos J, Huzarski T, Gronwald J, et al. Bilateral Oophorectomy and Breast Cancer Risk in BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst. 2017;109(1). https://doi.org/10.1093/jnci/djw177.

  39. Vik A, Brodin EE, Mathiesen EB, Brox J, Jørgensen L, Njølstad I, et al. Serum osteoprotegerin and future risk of cancer and cancer-related mortality in the general population: the Tromsø study. Eur J Epidemiol. 2015;30(3):219–30. https://doi.org/10.1007/s10654-014-9975-3.

    Article  CAS  PubMed  Google Scholar 

  40. Fortner RT, Sarink D, Schock H, Johnson T, Tjønneland A, Olsen A, et al. Osteoprotegerin and breast cancer risk by hormone receptor subtype: a nested case-control study in the EPIC cohort. BMC Med. 2017;15(1):1–10. https://doi.org/10.1186/s12916-017-0786-8.

    Article  CAS  Google Scholar 

  41. Kiechl S, Schramek D, Widschwendter M, et al. Aberrant regulation of RANKL/OPG in women at high risk of developing breast cancer. Oncotarget. 2017;8(3):3811–3825. https://doi.org/10.18632/oncotarget.14013

  42. Sarink D, Schock H, Johnson T, Overvad K, Holm M, Tjønneland A, et al. Circulating RANKL and RANKL/OPG and breast cancer risk by ER and PR subtype: results from the EPIC cohort. Cancer Prev Res. 2017;10(9):525–34. https://doi.org/10.1158/1940-6207.CAPR-17-0125.

    Article  CAS  Google Scholar 

  43. Kotsopoulos J, McGee EE, Lozano-Esparza S, et al. Premenopausal plasma osteoprotegerin and breast cancer risk: a case-control analysis nested within the nurses’ health study II. Cancer Epidemiol Biomark Prev. 2020;29(6):1264–70. https://doi.org/10.1158/1055-9965.EPI-19-1154.

    Article  Google Scholar 

  44. Odén L, Akbari M, Zaman T, et al. Plasma osteoprotegerin and breast cancer risk in BRCA1 and BRCA2 mutation carriers. Oncotarget. 2016;7(52):86687–94. https://doi.org/10.18632/oncotarget.13417.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Zaman T, Sun P, Narod SA, Salmena L, Kotsopoulos J. Plasma RANKL levels are not associated with breast cancer risk in BRCA1 and BRCA2 mutation carriers. Oncotarget. 2019;10(25):2475–83. https://doi.org/10.18632/oncotarget.26810.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Bord S, Frith E, Ireland DC, Scott MA, Craig JIO, Compston JE. Synthesis of osteoprotegerin and RANKL by megakaryocytes is modulated by oestrogen. Br J Haematol. 2004;126(2):244–51. https://doi.org/10.1111/j.1365-2141.2004.05024.x.

    Article  CAS  PubMed  Google Scholar 

  47. Bord S, Ireland DC, Beavan SR, Compston JE. The effects of estrogen on osteoprotegerin, RANKL, and estrogen receptor expression in human osteoblasts. Bone. 2003;32(2):136–41. https://doi.org/10.1016/s8756-3282(02)00953-5.

    Article  CAS  PubMed  Google Scholar 

  48. Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC, Riggs BL. Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology. 1999;140(9):4367–70. https://doi.org/10.1210/endo.140.9.7131.

    Article  CAS  PubMed  Google Scholar 

  49. Liao EY, Luo XH, Su X. Comparison of the effects of 17beta-E2 and progesterone on the expression of osteoprotegerin in normal human osteoblast-like cells. J Endocrinol Investig. 2002;25(9):785–90. https://doi.org/10.1007/BF03345513.

    Article  CAS  Google Scholar 

  50. Saika M, Inoue D, Kido S, Matsumoto T. 17beta-estradiol stimulates expression of osteoprotegerin by a mouse stromal cell line, ST-2, via estrogen receptor-alpha. Endocrinology. 2001;142(6):2205–12. https://doi.org/10.1210/endo.142.6.8220.

    Article  CAS  PubMed  Google Scholar 

  51. Huang JC, Sakata T, Pfleger LL, Bencsik M, Halloran BP, Bikle DD, et al. PTH differentially regulates expression of RANKL and OPG. J Bone Miner Res Off J Am Soc Bone Miner Res. 2004;19(2):235–44. https://doi.org/10.1359/JBMR.0301226.

    Article  CAS  Google Scholar 

  52. Lee S-K, Lorenzo JA. Regulation of receptor activator of nuclear factor-kappa B ligand and osteoprotegerin mRNA expression by parathyroid hormone is predominantly mediated by the protein kinase a pathway in murine bone marrow cultures. Bone. 2002;31(1):252–9. https://doi.org/10.1016/s8756-3282(02)00804-9.

    Article  CAS  PubMed  Google Scholar 

  53. Ma YL, Cain RL, Halladay DL, Yang X, Zeng Q, Miles RR, et al. Catabolic effects of continuous human PTH (1--38) in vivo is associated with sustained stimulation of RANKL and inhibition of osteoprotegerin and gene-associated bone formation. Endocrinology. 2001;142(9):4047–54. https://doi.org/10.1210/endo.142.9.8356.

    Article  CAS  PubMed  Google Scholar 

  54. Thomas RJ, Guise TA, Yin JJ, Elliott J, Horwood NJ, Martin TJ, et al. Breast cancer cells interact with osteoblasts to support osteoclast formation. Endocrinology. 1999;140(10):4451–8. https://doi.org/10.1210/endo.140.10.7037.

    Article  CAS  PubMed  Google Scholar 

  55. Brändström H, Björkman T, Ljunggren O. Regulation of osteoprotegerin secretion from primary cultures of human bone marrow stromal cells. Biochem Biophys Res Commun. 2001;280(3):831–5. https://doi.org/10.1006/bbrc.2000.4223.

    Article  CAS  PubMed  Google Scholar 

  56. Vidal NO, Brändström H, Jonsson KB, Ohlsson C. Osteoprotegerin mRNA is expressed in primary human osteoblast-like cells: down-regulation by glucocorticoids. J Endocrinol. 1998;159(1):191–5. https://doi.org/10.1677/joe.0.1590191.

    Article  CAS  PubMed  Google Scholar 

  57. Humphrey EL, Williams JHH, Davie MWJ, Marshall MJ. Effects of dissociated glucocorticoids on OPG and RANKL in osteoblastic cells. Bone. 2006;38(5):652–61. https://doi.org/10.1016/j.bone.2005.10.004.

    Article  CAS  PubMed  Google Scholar 

  58. Brändström H, Jonsson KB, Ohlsson C, Vidal O, Ljunghall S, Ljunggren O. Regulation of osteoprotegerin mRNA levels by prostaglandin E2 in human bone marrow stroma cells. Biochem Biophys Res Commun. 1998;247(2):338–41. https://doi.org/10.1006/bbrc.1998.8783.

    Article  PubMed  Google Scholar 

  59. Mancini L, Paul-Clark MJ, Rosignoli G, et al. Calcitonin and prednisolone display antagonistic actions on bone and have synergistic effects in experimental arthritis. Am J Pathol. 2007;170(3):1018–27. https://doi.org/10.2353/ajpath.2007.060830.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, Spelsberg TC, et al. Stimulation of osteoprotegerin ligand and inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage cells: potential paracrine mechanisms of glucocorticoid-induced osteoporosis. Endocrinology. 1999;140(10):4382–9. https://doi.org/10.1210/endo.140.10.7034.

    Article  CAS  PubMed  Google Scholar 

  61. Hofbauer LC, Dunstan CR, Spelsberg TC, Riggs BL, Khosla S. Osteoprotegerin production by human osteoblast lineage cells is stimulated by vitamin D, bone morphogenetic protein-2, and cytokines. Biochem Biophys Res Commun. 1998;250(3):776–81. https://doi.org/10.1006/bbrc.1998.9394.

    Article  CAS  PubMed  Google Scholar 

  62. Schoppet M, Henser S, Ruppert V, Stübig T, al-Fakhri N, Maisch B, et al. Osteoprotegerin expression in dendritic cells increases with maturation and is NF-κB-dependent. J Cell Biochem. 2007;100(6):1430–9. https://doi.org/10.1002/jcb.21129.

    Article  CAS  PubMed  Google Scholar 

  63. Kondo T, Kitazawa R, Maeda S, Kitazawa S. 1 alpha,25 dihydroxyvitamin D3 rapidly regulates the mouse osteoprotegerin gene through dual pathways. J Bone Miner Res Off J Am Soc Bone Miner Res. 2004;19(9):1411–9. https://doi.org/10.1359/JBMR.040604.

    Article  CAS  Google Scholar 

  64. Quinn JM, Horwood NJ, Elliott J, Gillespie MT, Martin TJ. Fibroblastic stromal cells express receptor activator of NF-kappa B ligand and support osteoclast differentiation. J Bone Miner Res Off J Am Soc Bone Miner Res. 2000;15(8):1459–66. https://doi.org/10.1359/jbmr.2000.15.8.1459.

    Article  CAS  Google Scholar 

  65. Collin-Osdoby P, Rothe L, Anderson F, Nelson M, Maloney W, Osdoby P. Receptor activator of NF-kappa B and osteoprotegerin expression by human microvascular endothelial cells, regulation by inflammatory cytokines, and role in human osteoclastogenesis. J Biol Chem. 2001;276(23):20659–72. https://doi.org/10.1074/jbc.M010153200.

    Article  CAS  PubMed  Google Scholar 

  66. Hormdee D, Nagasawa T, Kiji M, Yashiro R, Kobayashi H, Koshy G, et al. Protein kinase-A-dependent osteoprotegerin production on interleukin-1 stimulation in human gingival fibroblasts is distinct from periodontal ligament fibroblasts. Clin Exp Immunol. 2005;142(3):490–7. https://doi.org/10.1111/j.1365-2249.2005.02937.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Collin-Osdoby P. Regulation of vascular calcification by osteoclast regulatory factors RANKL and osteoprotegerin. Circ Res. 2004;95(11):1046–57. https://doi.org/10.1161/01.RES.0000149165.99974.12.

    Article  CAS  PubMed  Google Scholar 

  68. Hofbauer LC, Lacey DL, Dunstan CR, Spelsberg TC, Riggs BL, Khosla S. Interleukin-1beta and tumor necrosis factor-alpha, but not interleukin-6, stimulate osteoprotegerin ligand gene expression in human osteoblastic cells. Bone. 1999;25(3):255–9. https://doi.org/10.1016/s8756-3282(99)00162-3.

    Article  CAS  PubMed  Google Scholar 

  69. Nakashima T, Kobayashi Y, Yamasaki S, Kawakami A, Eguchi K, Sasaki H, et al. Protein expression and functional difference of membrane-bound and soluble receptor activator of NF-kappaB ligand: modulation of the expression by osteotropic factors and cytokines. Biochem Biophys Res Commun. 2000;275(3):768–75. https://doi.org/10.1006/bbrc.2000.3379.

    Article  CAS  PubMed  Google Scholar 

  70. Thirunavukkarasu K, Miles RR, Halladay DL, Yang X, Galvin RJS, Chandrasekhar S, et al. Stimulation of osteoprotegerin (OPG) gene expression by transforming growth factor-beta (TGF-beta). Mapping of the OPG promoter region that mediates TGF-beta effects. J Biol Chem. 2001;276(39):36241–50. https://doi.org/10.1074/jbc.M104319200.

    Article  CAS  PubMed  Google Scholar 

  71. Kearns AE, Khosla S, Kostenuik PJ. Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulation of bone remodeling in health and disease. Endocr Rev. 2008;29(2):155–92. https://doi.org/10.1210/er.2007-0014.

    Article  CAS  PubMed  Google Scholar 

  72. Han KO, Choi JT, Choi HA, Moon IG, Yim CH, Park WK, et al. The changes in circulating osteoprotegerin after hormone therapy in postmenopausal women and their relationship with oestrogen responsiveness on bone. Clin Endocrinol. 2005;62(3):349–53. https://doi.org/10.1111/j.1365-2265.2005.02221.x.

    Article  CAS  Google Scholar 

  73. Khosla S, Arrighi HM, Melton LJ 3rd, et al. Correlates of osteoprotegerin levels in women and men. Osteoporos Int. 2002;13(5):394–9. https://doi.org/10.1007/s001980200045.

    Article  CAS  PubMed  Google Scholar 

  74. Samelson EJ, Broe KE, Demissie S, Beck TJ, Karasik D, Kathiresan S, et al. Increased plasma Osteoprotegerin concentrations are associated with indices of bone strength of the hip. J Clin Endocrinol Metab. 2008;93(5):1789–95. https://doi.org/10.1210/jc.2007-2492.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Shinkov AD, Borissova A-MI, Kovatcheva RD, Atanassova IB, Vlahov JD, Dakovska LN. Age and menopausal status affect osteoprotegerin and osteocalcin levels in women differently, irrespective of thyroid function. Clin Med Insights Endocrinol Diabetes. 2014;7:19–24. https://doi.org/10.4137/CMED.S15466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Rao S, Cronin SJF, Sigl V, Penninger JM. RANKL and RANK: from mammalian physiology to Cancer treatment. Trends Cell Biol. 2018;28(3):213–23. https://doi.org/10.1016/j.tcb.2017.11.001.

    Article  CAS  PubMed  Google Scholar 

  77. Sarink D, Yang J, Johnson T, Chang-Claude J, Overvad K, Olsen A, et al. Reproductive and lifestyle factors and circulating sRANKL and OPG concentrations in women: results from the EPIC cohort. Cancer Epidemiol Biomark Prev. 2019;28(10):1746–54. https://doi.org/10.1158/1055-9965.EPI-19-0241.

    Article  CAS  Google Scholar 

  78. Kudlacek S, Schneider B, Woloszczuk W, Pietschmann P, Willvonseder R. Serum levels of osteoprotegerin increase with age in a healthy adult population. Bone. 2003;32(6):681–6. https://doi.org/10.1016/s8756-3282(03)00090-5.

    Article  CAS  PubMed  Google Scholar 

  79. Rogers A, Saleh G, Hannon RA, Greenfield D, Eastell R. Circulating estradiol and osteoprotegerin as determinants of bone turnover and bone density in postmenopausal women. J Clin Endocrinol Metab. 2002;87(10):4470–5. https://doi.org/10.1210/jc.2002-020396.

    Article  CAS  PubMed  Google Scholar 

  80. Abrahamsen B, Hjelmborg JV, Kostenuik P, et al. Circulating amounts of osteoprotegerin and RANK ligand: genetic influence and relationship with BMD assessed in female twins. Bone. 2005;36(4):727–35. https://doi.org/10.1016/j.bone.2004.12.015.

    Article  CAS  PubMed  Google Scholar 

  81. Livshits G, Pantsulaia I, Trofimov S, Kobyliansky E. Genetic influences on the circulating cytokines involved in osteoclastogenesis. J Med Genet. 2004;41(6):e76. https://doi.org/10.1136/jmg.2003.014373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Kwan JSH, Hsu Y-H, Cheung C-L, Dupuis J, Saint-Pierre A, Eriksson J, et al. Meta-analysis of genome-wide association studies identifies two loci associated with circulating osteoprotegerin levels. Hum Mol Genet. 2014;23(24):6684–93. https://doi.org/10.1093/hmg/ddu386.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kwan JSH, Xiao S, Bow C, Cheung CL, Soong C, Lau KS, et al. Heritability of serum osteoprotegerin. Ann Hum Genet. 2011;75(5):584–8. https://doi.org/10.1111/j.1469-1809.2011.00661.x.

    Article  CAS  PubMed  Google Scholar 

  84. Vistoropsky Y, Malkin I, Kobyliansky E, Livshits G. Osteoprotegerin plasma levels are strongly associated with polymorphisms in human homologue of the mouse progressive ankylosis (ANKH) gene. Ann Hum Genet. 2007;71(Pt 3):302–7. https://doi.org/10.1111/j.1469-1809.2006.00331.x.

    Article  CAS  PubMed  Google Scholar 

  85. Goëb V, Anne-Priscille T. Receptor activator of nuclear factor-&amp;kappa;B ligand and osteoprotegerin: maintaining the balance to prevent bone loss. Clin Interv Aging. 2010;5:345. https://doi.org/10.2147/CIA.S10153.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Joanne Kotsopoulos is a recipient of a Tier II Canada Research Chair. Sarah Park is a recipient of an Enid Walker Graduate Award in Women’s Health Research from the Enid Walker Estate. Aleksandra Uzelac is a recipient of the Open Fellowship award from the University of Toronto and the Canada Graduate Scholarship Masters Award from the Canadian Institute of Health Research (CIHR). This work was supported by the Peter Gilgan Foundation.

Author information

Authors and Affiliations

Authors

Contributions

JK conceived the study. SP conducted the literature search and wrote the manuscript. AU conducted the literature search and contributed to the writing of the manuscript. JK participated in the critical revision of the manuscript and supervised the project. All authors approved the final version of the manuscript.

Corresponding author

Correspondence to Joanne Kotsopoulos.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors agree to the publication.

Competing interests

The authors declare no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, S.S., Uzelac, A. & Kotsopoulos, J. Delineating the role of osteoprotegerin as a marker of breast cancer risk among women with a BRCA1 mutation. Hered Cancer Clin Pract 20, 14 (2022). https://doi.org/10.1186/s13053-022-00223-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13053-022-00223-3

Keywords