Clinical studies backed by research in animal models suggest that vitamin D may protect against the development of breast cancer, implicating vitamin D as a promising candidate for breast cancer prevention. However, despite clear preclinical evidence showing protective roles for vitamin D, broadly targeted clinical trials of vitamin D supplementation have yielded conflicting findings, highlighting the complexity of translating preclinical data to efficacy in humans. While vitamin D supplementation targeted to high-risk populations is a strategy anticipated to increase prevention efficacy, a complimentary approach is to target transient, developmental windows of elevated breast cancer risk. Postpartum mammary gland involution represents a developmental window of increased breast cancer promotion that may be poised for vitamin D supplementation. Targeting the window of involution with short-term vitamin D intervention may offer a simple, cost-effective approach for the prevention of breast cancers that develop postpartum. In this review, we highlight epidemiologic and preclinical studies linking vitamin D deficiency with breast cancer development. We discuss the underlying mechanisms through which vitamin D deficiency contributes to cancer development, with an emphasis on the anti-inflammatory activity of vitamin D. We also discuss current evidence for vitamin D as an immunotherapeutic agent and the potential for vitamin D as a preventative strategy for young woman's breast cancer.

Breast cancer is the most commonly diagnosed cancer in young women worldwide and this diagnosis carries a high morbidity and mortality burden (1). Young women are more likely to present with poor prognostic disease and have worse clinical outcomes compared with older women (2). Since the early 1990s, advances in treatment strategies have led to a decline in breast cancer mortality for young women (3, 4). However, breast cancer incidence in young women continues to rise globally (3), with a steady 1%–2% increase per year in the United States, leading to an overall 35% increase over the last four decades (1). Similar trends in incidence are observed worldwide (3). With approximately 26,000 early-onset breast cancer cases, defined here as ≤45 years of age, diagnosed each year in the United States alone (5), there are ongoing efforts to identify risk factors for early-onset disease, identify high risk populations, and develop new preventative therapies (6).

The anticancer nature of vitamin D and the potential for vitamin D as a breast cancer preventative agent has attracted considerable interest. Vitamin D is the biologically inactive precursor to the steroid hormone calcitriol (1,25-dihydroxyvitamin D; 1,25(OH)2D), a hormone that has been shown to exert anticancer effects in various tissues, including the breast. In women, there is substantial evidence supporting a protective role for vitamin D against breast cancer development (7–9), with causal links identified in rodents (10–17). While these and other studies provide a compelling argument for further investigation of vitamin D supplementation as a simple, nontoxic, and cost-effective approach to protect against breast cancer, whether vitamin D might be particularly efficacious against young women's breast cancer has yet to be explored.

Here, we review how downregulation of vitamin D signaling can contribute to breast cancer development and progression. We discuss the underlying biological mechanisms through which vitamin D deficiency contributes to cancer development, with an emphasis on the anti-inflammatory activity of vitamin D, and highlight the current clinical evidence for the use of vitamin D as an immunotherapeutic agent. We also discuss physiology unique to younger women, pregnancy, lactation, and weaning-induced breast involution, which exacerbate vitamin D deficiency, as well as present opportunities for future research to develop preventative strategies for young women's breast cancer.

The metabolism of vitamin D is a tightly regulated process (summarized in Fig. 1), which is consistent with its known role as a key transcription factor that regulates varied and complex developmental and physiologic responses (18). The actions of vitamin D are mediated through the binding of its active form 1,25(OH)2D to the vitamin D receptor (VDR), a transcription factor belonging to the steroid hormone receptor superfamily. Binding of 1,25(OH)2D to VDR stimulates heterodimerization with the retinoid X receptor (RXR). The VDR-RXR heterodimer subsequently binds vitamin D-responsive DNA elements and regulates expression of VDR target genes (19).

Figure 1.

Overview of the metabolism and function of vitamin D. Vitamin D is primarily obtained through exposure to sunlight, with additional sources of vitamin D obtained through dietary intake. Vitamin D is metabolized in the liver by CYP2R1 (also known as 25-hydroxylase) into 25-hydroxyvitamin D (25(OH)D), the form of the hormone found circulating in the blood. Hydroxylation of 25(OH)D is catalyzed primarily in the kidneys by CYP27B1 (also known as 1α-hydroxylase) to yield the active hormone 1,25(OH)2D. CYP24A1 catalyzes the degradation of 1,25(OH)2D into inactive metabolites. Metabolism of vitamin D is self-regulated; 1,25(OH)2D inhibits expression of CYP27B1 to prevent new synthesis of 1,25(OH)2D, while simultaneously inducing expression of CYP24A1 to promote 1,25(OH)2D degradation. Extrarenal sites of vitamin D metabolism include the breast, where CYP24A1 and CYP27B1 are expressed by epithelial, stromal, and immune cells, which regulate 1,25(OH)2D concentrations locally. The numerous biological functions of vitamin D, including mineral homeostasis, immune modulation, and mammary gland development and remodeling, are illustrated. Dysregulation in these signaling pathways can drive disease progression and metastasis.

Figure 1.

Overview of the metabolism and function of vitamin D. Vitamin D is primarily obtained through exposure to sunlight, with additional sources of vitamin D obtained through dietary intake. Vitamin D is metabolized in the liver by CYP2R1 (also known as 25-hydroxylase) into 25-hydroxyvitamin D (25(OH)D), the form of the hormone found circulating in the blood. Hydroxylation of 25(OH)D is catalyzed primarily in the kidneys by CYP27B1 (also known as 1α-hydroxylase) to yield the active hormone 1,25(OH)2D. CYP24A1 catalyzes the degradation of 1,25(OH)2D into inactive metabolites. Metabolism of vitamin D is self-regulated; 1,25(OH)2D inhibits expression of CYP27B1 to prevent new synthesis of 1,25(OH)2D, while simultaneously inducing expression of CYP24A1 to promote 1,25(OH)2D degradation. Extrarenal sites of vitamin D metabolism include the breast, where CYP24A1 and CYP27B1 are expressed by epithelial, stromal, and immune cells, which regulate 1,25(OH)2D concentrations locally. The numerous biological functions of vitamin D, including mineral homeostasis, immune modulation, and mammary gland development and remodeling, are illustrated. Dysregulation in these signaling pathways can drive disease progression and metastasis.

Close modal

Vitamin D is primarily obtained through exposure to sunlight, where ultraviolet-B in the skin converts 7-dehydrocholesterol to vitamin D. Additional sources of vitamin D can be obtained through dietary intake; however, few foods naturally contain significant amounts of this hormone (20). Vitamin D is metabolized to its active 1,25(OH)2D hormonal form through two hydroxylation steps. The first hydroxylation step occurs in the liver, where CYP2R1 catalyzes the conversion of vitamin D to 25-hydroxyvitamin D (25(OH)D). This form of vitamin D is the form of the hormone found circulating in blood and is used clinically to monitor vitamin D status (21). The second hydroxylation step occurs in the kidneys, where 25(OH)D is hydroxylated by CYP27B1 to yield the active hormone 1,25(OH)2D. In addition to homeostatic control of active hormone synthesis, inactivation of vitamin D metabolites is tightly regulated by the catalytic enzyme CYP24A1. This tight regulation of vitamin D metabolism reflects the importance of maintaining vitamin D concentrations within an appropriate range for optimal function. While there is some controversy surrounding optimal 25(OH)D concentrations (22), current clinical guidelines recommended that serum concentrations should be maintained within the range of 30 nmol/L–50 nmol/L for optimal health (23). Patients with serum concentrations of 25(OH)D below 30 nmol/L are defined as vitamin D deficient (22, 23).

Much of what is known about the physiology of vitamin D is elucidated from studies in the bone, due to a primary function of vitamin D in calcium and phosphate homeostasis (20, 24). Importantly, this primary bone function of vitamin D may yield insight into breast cancer risk in young women; a topic discussed later in this review. For bone health, vitamin D maintains optimal circulating concentrations of calcium and phosphate by acting directly on intestinal cells to modulate absorption (25). Vitamin D also facilitates transcellular calcium and phosphate absorption by stimulating expression of calcium-binding proteins, including calbindin-D9k (26, 27), calcium ion channels including TRPV5/6 (26, 27), and the sodium-dependent phosphate cotransporter NaPi-IIb (28, 29). Vitamin D also promotes calcium and phosphate absorption through passive paracellular transport mechanisms, by modulating expression of cell-junction proteins, including claudins (26, 30), cadherins (26), and aquaporins (26). The precise coordination of multiple tiers of vitamin-D regulation assures mineral homeostasis and highlights the importance of vitamin D in bone health. However, there is also strong evidence for vitamin D signaling beyond bone homeostasis.

The vitamin D receptor, VDR, is expressed in most tissues throughout the body, including immune, nervous, muscle, reproductive, and glandular tissue (31). This widespread expression profile broadly implicates vitamin D signaling in the maintenance of cell and tissue health. Indeed, impaired vitamin D signaling is implicated in the development and progression of various diseases, including inflammation and autoimmune disorders (32), chronic kidney disease (33), cardiovascular disease (34), hypertension (35), obesity (36), diabetes mellitus (37), as well as various types of cancer (38). Together, these studies reflect the numerous biologic functions of vitamin D, including mineral homeostasis, immune regulation, and epithelial cell proliferation, differentiation, and apoptosis. Importantly, many of these vitamin D functions interface with the hallmarks of cancer (39).

In the field of breast cancer, there is considerable evidence supporting a protective role for vitamin D. In women, increased circulating concentrations of vitamin D correlate with a decreased risk of breast cancer development (7–9); observations that have been recapitulated in rodent models (10–17). Moreover, expression of the VDR, through which vitamin D exerts its effects, is observed in approximately 80%–90% of invasive human breast tumors, and is implicated as a biomarker for good patient prognosis. Specifically, VDR expression in breast cancer samples positively associates with favorable tumor characteristics, such as smaller size, lower grade, lower proliferation, and steroid hormone receptor positivity (40–42). Furthermore, increased expression of VDR in breast tumors correlates with reduced disease recurrence, metastatic incidence, and mortality (41, 43). Preclinical studies using VDR-knockout mice corroborate these findings, where loss of VDR expression results in shorter time to tumor development, and increased tumor incidence and burden (44, 45). Interestingly, loss of only one copy of VDR is sufficient to increase tumorigenicity in mice, suggesting that partial reduction in VDR signaling is sufficient to promote tumor growth (46).

Vitamin D has been suggested to protect against breast cancer development through multiple potentially related mechanisms. Numerous studies provide evidence that vitamin D has potent antiproliferative effects in the breast. Treatment of breast cancer cell lines with physiologically relevant doses of vitamin D reduces cell proliferation in vitro (16, 17, 47–51) and in vivo (13, 16, 52, 53); effects observed in both hormone receptor–dependent and -independent breast cancer cell lines. Vitamin D inhibits cell proliferation by inducing cell-cycle arrest in the G1 phase, downregulating expression of cyclins and cyclin-dependent kinases (CDK), upregulating expression of CDK inhibitors, and stimulating hypophosphorylation of the retinoblastoma protein (54–56).

In parallel with its antiproliferative activity, vitamin D also has proapoptotic effects. Treatment of breast cancer cell lines with physiologically relevant doses of vitamin D induces cell apoptosis in vitro (49–51) and in vivo (53). Moreover, vitamin D promotes cell differentiation and reduces the stem cell potential of breast cancer cells in vitro (57–59)—actions that are associated with downstream proapoptotic effects. These findings are supported by studies using carcinogen-induced and transgenic mouse models of breast cancer, which report that vitamin D inhibits the growth and progression of mammary tumors (12, 15, 48), and promotes tumor cell apoptosis to stimulate tumor regression (17).

The enzymes involved in the anabolism and catabolism of vitamin D, CYP27B1 and CYP24A1 respectively, have also been implicated in breast cancer development and progression (Fig. 1). CYP27B1 and CYP24A1 are expressed in all major cell types within the mammary gland, suggesting that 1,25(OH)2D levels are regulated locally within the mammary microenvironment. Thus, dysregulation in local vitamin D production and degradation may contribute to breast cancer development. In the context of cancer, decreased expression of the anabolic enzyme CYP27B1, and thus reduced 1,25(OH)2D synthesis, are observed in breast cancer tissue compared with adjacent normal breast tissue (60). Conversely, increased expression of the catabolic enzyme CYP24A1, coupled with increased inactivation of 1,25(OH)2D, is observed in breast cancer samples, compared with normal breast tissue (61, 62). Corroborating data have been obtained from rodent models. In the PyMT-MMTV mouse model of breast cancer, knockout of CYP27B1 in mammary epithelial cells inhibits local synthesis of 1,25(OH)2D and results in accelerated mammary cancer development and increased tumor burden (46). Conversely, inhibiting degradation of 1,25(OH)2D through CYP24A1 knockout leads to sustained levels of 1,25(OH)2D and suppression of tumorigenicity in xenograft models of breast cancer (63, 64)—observations consistent with an oncogenic role for CYP24A1 in breast cancer (65). Combined, these human and preclinical data show that reduced vitamin D signaling, through either vitamin D deficiency, reduced VDR expression, or the impaired anabolism and catabolism of 1,25(OH)2D, is likely to promote breast cancer development and progression.

In addition to the potential of vitamin D as a chemopreventative agent, preclinical data suggest a role for vitamin D in the prevention of disease progression and metastasis. Vitamin D modulates various aspects of the metastatic process, including invasion, migration, and establishment at distant sites. In vitro, vitamin D treatment reduces the invasiveness and migration of breast cancer cells (13, 57, 66–68), through increasing protein expression of E-cadherin (57, 67, 69) and focal adhesions (50, 69), while simultaneously downregulating N-cadherin (67, 69), P-cadherin (69), and matrix metalloproteinase (50, 68) expression. Corroborating data have been obtained from rodent models. In xenograft models of breast cancer, vitamin D deficiency (10, 52) and loss of VDR expression (13, 40) increase breast cancer metastasis, through promotion of angiogenesis and vascularization (66). In the MMTV-PyMT mouse model, dietary vitamin D deficiency increases metastatic burden in the lung (10, 70), an effect likely mediated through increased CXCL12/CXCR4 signaling within the metastatic niche (70). Together, these observations suggest that vitamin D deficiency may establish a proangiogenic environment supportive of tumor cell dissemination, metastasis, and establishment at the secondary site; and implicate vitamin D as a potential therapeutic for the prevention and possible management of advanced, metastatic disease.

In addition to direct antiproliferative, proapoptotic effects on tumor cells, another important mechanism through which vitamin D may exert its anticancer properties is by influencing the immune microenvironment. It is well established that vitamin D plays important roles in regulating inflammation and immune response in various tissues. The VDR is expressed by most cells of the immune system, including macrophages (71, 72), T cells (71, 73), B cells (73), dendritic cells (74), neutrophils (75), and natural killer cells (76). Furthermore, immune cells express CYP27B1 and CYP24A1, thus can regulate local metabolism of 1,25(OH)2D (72, 74). However, while there is extensive research into the anti-inflammatory effects of vitamin D in various disease models, including inflammatory bowel disease (77), diet-induced obesity (36), collagen-induced arthritis (78), and chemical-induced liver toxicity (79); there is currently little information on the effects of vitamin D on total or specific subpopulations of immune cells within the normal breast microenvironment, or in the context of breast cancer.

In cell culture, vitamin D stimulation influences macrophage phenotype by shifting the polarization from the proinflammatory M1 phenotype to the anti-inflammatory M2 phenotype (80). This phenotype shift is associated with reductions in expression of several proinflammatory cytokines including IL6 (80, 81), IL8 (82), IL12 (83), and TNFα (80–82), coupled with increased expression of anti-inflammatory cytokines IL10 (80) and IL1β (80, 84). Similarly, in cultured neutrophils, vitamin D inhibits expression of TNFα, while enhancing the production of IL8 and IL1β (84). Vitamin D also influences T-cell polarization by shifting T-cell responses from a proinflammatory Th1 to an anti-inflammatory Th2 phenotype. Specifically, vitamin D inhibits the expression of proinflammatory cytokines IL12 and IFNγ (85–87) while promoting Th2 cell development and the production of anti-inflammatory cytokines IL4, IL5, and IL13 (83, 85). In parallel, vitamin D suppresses the differentiation of naïve T cells into proinflammatory Th17 cells (88), thus inhibiting IL17 production (87, 88), while simultaneously promoting differentiation into protumor FOXP3+ regulatory T cells (Treg; refs. 86, 87). Vitamin D also influences T-cell response by impairing the migration and maturation of dendritic cells, an action that results in reduced antigen presentation capacity and reduced activation of T cells (89, 90).

Together, these in vitro study observations suggest that vitamin D exerts anti-inflammatory effects by reducing proinflammatory Th1, Th17, and M1 polarization, while promoting anti-inflammatory Th2 and M2 polarization. Within the context of normal tissue, the ability of vitamin D to suppress a Th1-skewed inflammatory environment is consistent with cancer prevention, where chronic inflammation associates with increased cancer risk (91). However, in the context of existing cancer, Th1 suppression and Th2 induction are associated with loss of tumor cell immune surveillance and poor prognosis (92). While this yin yang nature of the immune system in cancer is widely recognized (93), it adds significant complexity to understanding how best to incorporate an immunomodulatory agent such as vitamin D into the breast cancer prevention setting.

One limitation to our understanding of vitamin D as an immune-modulatory agent is that current research has been conducted primarily in vitro, using immune cell lines and isolated human immune cells in the absence of tissue context cues, including cancer. These reductionist models are unable to capture the in vivo interactions that occur between cancer cells and the immune microenvironment, which is an important requirement for cancer prevention research. Importantly, the use of in vivo models has shed considerable light into how anti-inflammatory agents, including vitamin D, might remodel a protumor immune milieu (i.e., Th2-skewed) into one of antitumor (Th1-skewed), possibly reducing concerns that vitamin D supplementation might promote existing cancers. A recent study using an immunocompetent mouse model of breast cancer demonstrated that vitamin D supplementation promoted infiltration of activated T cells (as measured by CD44 expression) into the mammary tumor, and reduced the infiltration of F4/80+CD11b+ macrophages in the peripheral tissue; an immune profile consistent with active tumor cell immune surveillance (94). Thus, in the context of a mouse model with an intact immune system, the anti-inflammatory activity of vitamin D appears linked to the suppression of myeloid cells, which releases inhibition on cytotoxic T cells. Of note, similar results are seen with nonsteroidal anti-inflammatory agents (NSAID) in immunocompetent mice, where ibuprofen promoted maturation of tumor-associated myeloid cells, resulting in macrophage maturation, infiltration of cytotoxic T cells, and tumor suppression (95).

Additional context-dependent complexity has been observed between vitamin D and its antitumor activity in the context of obesity. In contrast to healthy weight mice, vitamin D treatment in obese mice resulted in increased tumor volume and reduced immune cell infiltrate. Similar data have been obtained from observational clinical studies. A recent randomized clinical trial of 25,254 participants reported that while vitamin D supplementation associates with a significant reduction in advanced cancer incidence, when stratified by body mass index (BMI), the protective effect of vitamin D only persists for patients with a normal BMI (96). These observations introduce additional complexity into the use of vitamin D as a preventative agent, as patients with high BMI also exhibit increased rates of vitamin D deficiency (97).

In obesity, the tumor microenvironment is characterized by an increased abundance of adipose tissue. Critically, adipocytes express VDR, including those within the breast (98, 99). Adipocytes play important roles in regulating inflammation and immune response following vitamin D stimulation through the modulation of inflammatory cytokine expression (100). Consequently, the effects of vitamin D on the immune response in the breast may be influenced by the abundance and/or metabolic state of adipocytes. Together, these studies demonstrate how immune-modulatory agents, including vitamin D, can be either pro- or antitumor depending on the local immune milieu, and highlight the need for well characterized, immunocompetent models to advance understanding of vitamin D as a breast cancer preventive agent.

Despite strong preclinical evidence suggesting vitamin D supplementation could offer protection against the development of breast cancer, phase III randomized clinical trials with primary outcomes assessing breast cancer biomarker endpoints or incidence have yet to be implemented. However, some large-scale vitamin D trials with other disease endpoints have had secondary analyses for breast cancer outcomes, and report that vitamin D supplementation associates with an approximate 18% reduction in breast cancer risk [HRs = 0.82 (0.70–0.97); 0.82 (0.68–0.99); refs. 101, 102]. However, other large-scale studies report no association (103–107).

The most recent promising, yet indirect, evidence comes from results of the VITamin D and OmegA-3 TriaL (VITAL), a randomized clinical trial of 25,871 participants that measured incidence of any cancer and cardiovascular disease as the primary outcomes (108). Participants received either 2,000 IU of vitamin D or placebo daily, for an average length of 5.3 years. While results from VITAL reported that vitamin D supplementation did not reduce total cancer incidence or mortality, when data from the first year was omitted to account for tumor latency, vitamin D supplementation was associated with a 21% reduction in cancer-associated mortality [HR = 0.79 (0.63–0.99); ref. 109]. Unfortunately for breast cancer cases, similar subanalyses omitting first year of data were not performed. In a secondary analysis of VITAL, vitamin D supplementation was shown to associate with a significant reduction in metastatic cancer incidence [HR = 0.83 (0.69–0.99)]; however, site-specific case numbers for breast cancer were too small to be analyzed (96).

Numerous meta-analyses have been conducted to address the potential for vitamin D supplementation in breast cancer prevention (110–112). Most recently, a meta-analysis of eight randomized control trials of 72,275 participants compared vitamin D supplementation versus placebo for the prevention of breast cancer (111). Of the eight trials included in the meta-analysis, study methods varied significantly, with dosages of vitamin D supplementation ranging from 400 to 3704 IU/day and mean follow-up periods ranging from 1 to 11.9 years. Findings from the meta-analysis show that current evidence does not support a protective role for vitamin D in the prevention of breast cancer.

As described above, many vitamin D intervention studies have been limited to small samples sizes with short follow-up times and varied significantly in the dose, frequency, and duration of vitamin D supplementation. These studies were also limited by a lack of data surrounding the patient's vitamin D status at baseline, preventing analysis of outcomes in light of initial and change in vitamin D status. As it is anticipated that the protective effect of vitamin D supplementation may be influenced by whether the patient is vitamin D–deficient or -sufficient at the time of study commencement, it is important for future studies to stratify participants by baseline vitamin D status. Furthermore, given the heterogeneity of breast cancer, different breast cancer subtypes may be differentially responsive to vitamin D treatment, which could further cloud any potential protective effects of vitamin D. Consistent with this possibility, a recent meta-analysis suggests that the protective effect of vitamin D may be more pronounced for estrogen receptor (ER)-negative breast tumors, compared with ER+ tumors (113). Currently, data from large-scale randomized trials assessing vitamin D supplementation on breast cancer incidence are lacking, and there remains insufficient evidence to make recommendations for incorporating vitamin D supplementation as a general strategy to prevent breast cancer.

While trials of vitamin D supplementation for breast cancer prevention have been underwhelming, there is some evidence that vitamin D supplementation targeted to higher risk populations may be a more effective approach for breast cancer prevention. Observational studies suggest that the association between vitamin D deficiency and increased breast cancer risk is stronger in younger women, compared with older women (7, 114). A meta-analysis of 68 studies of 91,594 patients identified an inverse relationship between vitamin D status and breast cancer risk; women with high serum vitamin D concentrations were at a lower risk for developing breast cancer [OR = 0.65 (0.56–0.76)]. Critically, when these results were analyzed by menopausal status, the protective effect of vitamin D persisted only for premenopausal women [premenopausal OR = 0.67 (0.49–0.92); postmenopausal OR = 0.97 (0.82–1.14); ref. 7]. Consistent with these observations, vitamin D deficiency during times of breast development is suggested to be more influential on breast cancer risk compared with vitamin D status later in life. A population-based case–control study of 2,217 women reported that vitamin D deficiency at the time of adolescent breast development was more strongly associated with breast cancer risk, compared with vitamin D status later in life (114).

However, not all studies have found a protective effect of vitamin D against breast cancer for premenopausal women (115, 116). A population-based case–control study of 2,101 women reported that increased vitamin D serum concentrations were associated with an approximate 54% reduction in breast cancer risk for postmenopausal women [OR = 0.92 (0.89–0.96)], whereas no significant reduction in risk was observed for premenopausal women [OR = 0.97 (0.92–1.02); ref. 115]. Consistent with this, a nested case–control study of 100 pregnant and recently pregnant patients with breast cancer reported no association between serum concentrations of vitamin D and risk of breast cancer during pregnancy. Critically, the authors instead reported that higher circulating concentration of vitamin D during pregnancy were associated with a 2- to 4-fold increased risk of developing breast cancer within one year following delivery (116).

Clinical trials assessing the effect of vitamin D supplementation on breast cancer biomarker endpoints have yielded similar indefinite results. A recent randomized control trial of 208 premenopausal women at high risk for breast cancer assessed the effect of 20,000 IU/week vitamin D supplementation on mammographic breast density, a strong predictor of breast cancer risk (117). This trial reported that vitamin D supplementation did not reduce mammographic breast density at either 12 or 24 months follow-up and concluded that there is insufficient evidence to support the use of vitamin D for reducing breast cancer risk (117). However, the majority of women enrolled in this study were sufficient for vitamin D, and subgroup analyses were not performed to assess whether the protective effects of vitamin D were influenced by vitamin D status at baseline. Furthermore, the potential protective effect of vitamin D may not be mediated by changes in mammographic breast density and may instead be a result of changes in other breast cancer biomarkers.

An ongoing prospective clinical trial is anticipated to provide additional data on the effect of vitamin D supplementation on breast cancer biomarkers (118). In this trial, 300 premenopausal women have been randomized to receive either 2,000 IU of vitamin D or placebo daily for 12 months. The majority of women enrolled were deficient for vitamin D at baseline (62%, serum 25(OH)D < 30 nmol/L), which may permit subset analyses on the effects of vitamin D supplementation on biomarker expression in light of deficient or sufficient serum 25(OH)D at baseline. Primary outcomes will assess the change in mammographic density at 12 months, with secondary outcomes assessing changes in other breast cancer biomarkers, including atypia, cell proliferation, and serum IGF-1. Results from this trial are awaited.

Vitamin D as a potential preventative therapeutic for postpartum breast cancer

While vitamin D supplementation targeted to premenopausal women is a strategy anticipated to increase prevention efficacy, a complimentary approach is to specifically target transient, developmental windows of elevated breast cancer risk. The window of weaning-induced breast involution represents a key developmental window that contributes to breast cancer risk, and which may be particularly poised for vitamin D supplementation. In support of a prevention strategy targeted to the postpartum window, studies consistently find a transient increased risk for breast cancer following childbirth. The peak incidence has been reported at 5 years postpartum (119), with a long tail of increased risk persisting up to 15 years postpartum (120). It has been proposed that postpartum breast cancers account for approximately 50% of all young women's breast cancer cases (121,122). Furthermore, these cancers have worse prognosis compared with age-, stage-matched cases in nulliparous women. A breast cancer diagnosis within 5–10 years of a recent pregnancy independently associates with a 2- to 3-fold increased risk of death, for both ER+ and ER disease (123, 124). Conversely, a breast cancer diagnosis during pregnancy is not associated with poorer outcomes (125). Combined, these studies implicate the existence of a postpartum event that negatively impacts breast cancer.

In women, the postpartum window coincides with weaning-induced breast involution. During involution, the mammary gland is characterized by a unique microenvironment that shares similarities with wound-healing, inflammation, and desmoplasia (126–128). The inflammatory microenvironment of the involuting gland has been demonstrated to promote breast cancer progression and tumor cell dissemination in rodent models (129–131), and strongly associate with the high rate of metastases observed in postpartum patients with breast cancer (122, 124, 132). Importantly, a recent study reports that the weaning-induced mammary involution programs observed in rodents are mirrored in the human breast (133). Specifically, in breast tissue of recently lactating women, weaning associates with transient epithelial cell death, immune cell infiltrate, and stromal hallmarks of wound healing, including proinflammatory cyclooxygenase-2 (COX-2) expression (133). Importantly, these wound healing like tissue attributes are largely resolved by 3 months post wean, potentially identifying a very narrow window of therapeutic preventative intervention. These human data, combined with preclinical rodent studies, support the hypothesis that breast cancer risk might be mitigated by anti-inflammatory strategies, such as vitamin D, when targeted to the post-wean window of breast involution.

Pregnancy, lactation, and weaning as unique windows of vitamin D deficiency

There are heightened demands for vitamin D during a reproductive cycle that emphasize vitamin D deficiency in the postpartum period and implicate the window of breast involution as a viable target for vitamin D supplementation. Changes in the metabolism of vitamin D and calcium occur to meet the increased demands during pregnancy and lactation. During pregnancy, the unique demand for vitamin D is for proper fetal skeletal growth. Renal production of 1,25(OH)2D increases 2-fold during pregnancy to promote intestinal absorption of calcium and returns to prepregnancy levels following parturition (134, 135). The unique demand for vitamin D in the postpartum period occurs during lactation, where the increased metabolic demand of milk production requires calcium. During lactation, the demand for calcium is met primarily by the increased resorption of calcium from the bone. This effect likely occurs via vitamin D–dependent mechanisms (136, 137) and is mediated by the secretion of parathyroid hormone-related protein from the lactating breast (138, 139).

It is also possible that reductions in vitamin D synthesis occur during postlactational involution, further depleting vitamin D levels during this critical window of increased breast cancer risk. In rats and mice, it has recently been reported that the normal liver undergoes weaning-induced involution. Similar to the mammary gland, weaning-induced liver involution is characterized by epithelial cell death (i.e., hepatocytes), stromal remodeling, and immune cell influx (132). Critically, the rodent involuting liver also exhibits metabolic signatures of protein catabolism and oxidative stress. In postpartum women, indirect evidence of liver involution comes from a recent study, which demonstrates that liver size increases with pregnancy, before returning to normal size post wean (140). As the liver is a primary site of vitamin D synthesis, it is possible that during weaning-induced involution the liver is compromised in its ability to hydroxylase vitamin D. Albeit an untested hypothesis, this could result in reduced circulating concentrations of 25(OH)D during involution—a hallmark of vitamin D deficiency.

The importance of vitamin D in breast health across a reproductive cycle is further exemplified by the observation that key vitamin D genes, VDR, CYP24A1, and CYP27B1, are expressed in all major cell types in the breast (42, 61, 62, 72, 74, 99) and that their expression is dynamically regulated in the mammary gland throughout a pregnancy, lactation, and wean reproductive cycle (Fig. 2; refs. 126, 141). Expression of VDR is upregulated during pregnancy, with increased expression persisting throughout lactation (142). Expression of VDR remains high during early involution, where it regulates apoptosis and glandular remodeling, before returning to low levels in the nonpregnant, quiescent breast (Fig. 2; refs. 98, 141–143;). Of note, expression of CYP24A1 and CYP27B1 peak during lactation and early involution, before returning to low levels in the quiescent breast (Fig. 2; refs. 126, 141, 142, 144).

Figure 2.

Change in vitamin D receptor (VDR), CYP27B1, and CYP24A1 mRNA expression in the murine mammary gland during pregnancy, lactation, and involution. Gene expression of VDR (A), and the enzymes involved in the anabolism (CYP27B1) (B) and catabolism (CYP24A1) (C) of vitamin D throughout the reproductive cycle. Gene expression data were obtained from previously published microarray data. Data obtained from Rudolph and colleagues (2003; 141) and Stein and colleagues (2004; 126) are normalized to gene expression at day 1 of pregnancy (P1); data obtained from Clarkson and colleagues (2004; 144) are normalized to gene expression in virgin mice (V). All data are presented as mean ± SEM. V, virgin; P, pregnancy; L, lactation; Inv, involution.

Figure 2.

Change in vitamin D receptor (VDR), CYP27B1, and CYP24A1 mRNA expression in the murine mammary gland during pregnancy, lactation, and involution. Gene expression of VDR (A), and the enzymes involved in the anabolism (CYP27B1) (B) and catabolism (CYP24A1) (C) of vitamin D throughout the reproductive cycle. Gene expression data were obtained from previously published microarray data. Data obtained from Rudolph and colleagues (2003; 141) and Stein and colleagues (2004; 126) are normalized to gene expression at day 1 of pregnancy (P1); data obtained from Clarkson and colleagues (2004; 144) are normalized to gene expression in virgin mice (V). All data are presented as mean ± SEM. V, virgin; P, pregnancy; L, lactation; Inv, involution.

Close modal

In sum, the increased demand for vitamin D during pregnancy and lactation, in combination with potentially reduced vitamin D synthesis within the involuting liver, is anticipated to enhance vitamin D deficiency for postpartum women. Indeed, vitamin D deficiency is exceptionally common among postpartum women (145–148). Recent meta-analyses report that 18%–97% of pregnant and recently pregnant women are deficient for vitamin D, depending on the country and population studied (145, 146). In the United States, vitamin D deficiency is observed in up to 72% of pregnant and recently pregnant women (147, 148).

We speculate that the accumulated vitamin D deficiency of pregnancy, lactation, and involution that is prevalent post wean may exaggerate the tumor promotional attributes specific to the involuting breast and increase breast cancer risk in some postpartum women. Restoring vitamin D to optimal levels during pregnancy, lactation, and involution may offer a new therapeutic approach for the management of postpartum breast cancer. Support for transiently targeting weaning-induced mammary gland involution with prevention strategies comes from rodent studies, which demonstrate that short-term administration of NSAIDs targeted only to the window of weaning-induced mammary gland involution significantly reduces postpartum mammary cancer incidence and slows disease progression (95, 130).

While both vitamin D and NSAIDs show potential as preventative agents, neither agent completely abrogates tumor growth in animal models. The combination of vitamin D with NSAIDs may be a more efficacious approach to reduce postpartum breast cancer incidence in at-risk women. Indeed, preliminary in vitro studies in multiple cancer cell lines demonstrate that cotreatment with vitamin D and NSAIDs synergistically inhibit tumor proliferation, compared with treatment with either agent alone (149, 150). Together, these observations justify further investigation into whether vitamin D intervention, possibly in combination with other immune-modulatory agents such as NSAIDs, may offer protection against the development of postpartum breast cancer.

Establishing a clinical trial to assess the cancer preventative potential of vitamin D in postpartum women presents unique challenges. Significant problems with clinical trials to date stem from the variable doses, frequencies, and durations of vitamin D supplementation, as well as the targeting of relatively undefined populations with respect to breast cancer risk. For postpartum women, determination of the optimal dosages of vitamin D supplementation for cancer prevention presents challenges, as data on the ideal range of 25(OH)D concentrations required for pregnant and recently pregnant women are unclear (22). Additional complexity in dose determination is introduced by data from observational studies, which suggest that a U-shaped relationship exists between serum 25(OH)D and cancer incidence, where both lowest and highest 25(OH)D concentrations associate with increased cancer risk (151). Additional limitations with the clinical trials to date lie in their small sample sizes, short follow-up times, and lack of data on vitamin D status at baseline. To address these limitations, future trials in postpartum women must measure serum 25(OH)D at baseline, and be significantly powered with sufficient follow-up time to analyze results by vitamin D–deficient and -sufficient subgroups. Finally, the incidence of young women's breast cancer is relatively low, with approximately 1 in 65 women developing breast cancer by age 40 (1, 5). Thus, in a PPBC prevention trial, the use of cancer incidence as the trial endpoint is likely not feasible and surrogate endpoints would be required. In sum, while there is strong rationale to warrant a clinical trial to assess the cancer preventative potential of vitamin D for postpartum women, development of such a clinical trial requires additional preclinical model data, as well as unique considerations for successful implementation and analyses before such a trial would be compelling.

There is strong evidence that downregulation of vitamin D signaling contributes to the development and progression of breast cancer. Epidemiologic studies have linked vitamin D deficiency with an increased risk of breast cancer development; observations that are well supported by research in animal models. However, while the potential for vitamin D as a breast cancer preventative agent is well backed by preclinical data, clinical trials of vitamin D supplementation have produced modest and/or conflicting results. Vitamin D supplementation during transient, developmental windows of elevated risk may instead offer a more effective approach for the prevention of breast cancer. Postlactational involution represents a key developmental window that may be poised for vitamin D supplementation. The unique demands for vitamin D during pregnancy and lactation emphasize vitamin D deficiency during the postpartum period, which in the background of the proinflammatory microenvironment of the involuting gland may enhance the tumor promotional attributes of involution. Attenuation of the inflammatory microenvironment of the vitamin D–deficient involuting breast with short-term vitamin D supplementation may offer protection against the development of postpartum breast cancer. Further research into the protective role of vitamin D against postpartum breast cancer development and progression is warranted.

No disclosures were reported.

The authors would like to thank Alexandra Bartlett for constructive review of the manuscript and Weston Anderson for valuable support in review and editing of the manuscript. This work was supported by the NCI R01CA169175. The authors regret that not all references could not be cited in this review due to space constraints, and apologize for any omissions.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2020
.
CA Cancer J Clin
2020
;
70
:
7
30
.
2.
Johansson
ALV
,
Trewin
CB
,
Hjerkind
KV
,
Ellingjord-Dale
M
,
Johannesen
TB
,
Ursin
G
. 
Breast cancer-specific survival by clinical subtype after 7 years follow-up of young and elderly women in a nationwide cohort
.
Int J Cancer
2019
;
144
:
1251
61
.
3.
Bray
F
,
McCarron
P
,
Parkin
DM
. 
The changing global patterns of female breast cancer incidence and mortality
.
Breast Cancer Res
2004
;
6
:
229
39
.
4.
Jatoi
I
,
Miller
AB
. 
Why is breast-cancer mortality declining?
Lancet Oncol
2003
;
4
:
251
4
.
5.
SEER*Explorer
.
An interactive website for SEER cancer statistics [Internet]
.
Surveillance Research Program, National Cancer Institute
.
Available from
: https://seer.cancer.gov/explorer/.
6.
Nelson
HD
,
Zakher
B
,
Cantor
A
,
Fu
R
,
Griffin
J
,
O'Meara
ES
, et al
Risk factors for breast cancer for women aged 40 to 49 years: a systematic review and meta-analysis
.
Ann Intern Med
2012
;
156
:
635
48
.
7.
Estebanez
N
,
Gomez-Acebo
I
,
Palazuelos
C
,
Llorca
J
. 
Dierssen-sotos T. Vitamin D exposure and risk of breast cancer: a meta-analysis
.
Sci Rep
2018
;
8
:
9039
.
8.
Hossain
S
,
Beydoun
MA
,
Beydoun
HA
,
Chen
X
,
Zonderman
AB
,
Wood
RJ
. 
Vitamin D and breast cancer: a systematic review and meta-analysis of observational studies
.
Clin Nutr ESPEN
2019
;
30
:
170
84
.
9.
Kim
Y
,
Je
Y
. 
Vitamin D intake, blood 25(OH)D levels, and breast cancer risk or mortality: a meta-analysis
.
Br J Cancer
2014
;
110
:
2772
84
.
10.
Rossdeutscher
L
,
Li
J
,
Luco
AL
,
Fadhil
I
,
Ochietti
B
,
Camirand
A
, et al
Chemoprevention activity of 25-hydroxyvitamin D in the MMTV-PyMT mouse model of breast cancer
.
Cancer Prev Res
2015
;
8
:
120
8
.
11.
Jacobson
EA
,
James
KA
,
Newmark
HL
,
Carroll
KK
. 
Effects of dietary fat, calcium, and vitamin D on growth and mammary tumorigenesis induced by 7,12-dimethylbenz(a)anthracene in female Sprague-Dawley rats
.
Cancer Res
1989
;
49
:
6300
3
.
12.
Iino
Y
,
Yoshida
M
,
Sugamata
N
,
Maemura
M
,
Ohwada
S
,
Yokoe
T
, et al
1 alpha-hydroxyvitamin D3, hypercalcemia, and growth suppression of 7,12-dimethylbenz[a]anthracene-induced rat mammary tumors
.
Breast Cancer Res Treat
1992
;
22
:
133
40
.
13.
Williams
JD
,
Aggarwal
A
,
Swami
S
,
Krishnan
AV
,
Ji
L
,
Albertelli
MA
, et al
Tumor autonomous effects of vitamin d deficiency promote breast cancer metastasis
.
Endocrinology
2016
;
157
:
1341
7
.
14.
Colston
KW
,
Berger
U
,
Coombes
RC
. 
Possible role for vitamin D in controlling breast cancer cell proliferation
.
Lancet
1989
;
1
:
188
91
.
15.
Peng
X
,
Hawthorne
M
,
Vaishnav
A
,
St-Arnaud
R
,
Mehta
RG
. 
25-Hydroxyvitamin D3 is a natural chemopreventive agent against carcinogen induced precancerous lesions in mouse mammary gland organ culture
.
Breast Cancer Res Treat
2009
;
113
:
31
41
.
16.
Abe
J
,
Nakano
T
,
Nishii
Y
,
Matsumoto
T
,
Ogata
E
,
Ikeda
K
. 
A novel vitamin D3 analog, 22-oxa-1,25-dihydroxyvitamin D3, inhibits the growth of human breast cancer in vitro and in vivo without causing hypercalcemia
.
Endocrinology
1991
;
129
:
832
7
.
17.
James
SY
,
Mackay
AG
,
Colston
KW
. 
Effects of 1,25 dihydroxyvitamin D3 and its analogues on induction of apoptosis in breast cancer cells
.
J Steroid Biochem Mol Biol
1996
;
58
:
395
401
.
18.
Evans
RM
. 
The steroid and thyroid hormone receptor superfamily
.
Science
1988
;
240
:
889
95
.
19.
Haussler
MR
,
Haussler
CA
,
Jurutka
PW
,
Thompson
PD
,
Hsieh
JC
,
Remus
LS
, et al
The vitamin D hormone and its nuclear receptor: molecular actions and disease states
.
J Endocrinol
1997
;
154
:
S57
73
.
20.
Holick
MF
. 
Vitamin D deficiency
.
N Engl J Med
2007
;
357
:
266
81
.
21.
Holick
MF
,
Binkley
NC
,
Bischoff-Ferrari
HA
,
Gordon
CM
,
Hanley
DA
,
Heaney
RP
, et al
Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline
.
J Clin Endocrinol Metab
2011
;
96
:
1911
30
.
22.
Roth
DE
,
Abrams
SA
,
Aloia
J
,
Bergeron
G
,
Bourassa
MW
,
Brown
KH
, et al
Global prevalence and disease burden of vitamin D deficiency: a roadmap for action in low- and middle-income countries
.
Ann N Y Acad Sci
2018
;
1430
:
44
79
.
23.
(IOM) IoM
. 
Dietary reference intakes for calcium and vitamin D
.
In
:
Ross
AC
,
Taylor
CL
,
Yaktine
AL
,
Del Valle
HB
,
editors
.
Dietary reference intakes for calcium and vitamin D, The National Academies Collection: reports funded by National Institutes of Health
.
Washington D.C.
:
The National Academies Press
; 
2011
.
24.
Bouillon
R
,
Marcocci
C
,
Carmeliet
G
,
Bikle
D
,
White
JH
,
Dawson-Hughes
B
, et al
Skeletal and extraskeletal actions of vitamin D: Current evidence and outstanding questions
.
Endocr Rev
2019
;
40
:
1109
51
.
25.
Heaney
RP
. 
Vitamin D in health and disease
.
Clin J Am Soc Nephrol
2008
;
3
:
1535
41
.
26.
Kutuzova
GD
,
Deluca
HF
. 
Gene expression profiles in rat intestine identify pathways for 1,25-dihydroxyvitamin D (3) stimulated calcium absorption and clarify its immunomodulatory properties
.
Arch Biochem Biophys
2004
;
432
:
152
66
.
27.
Song
Y
,
Peng
X
,
Porta
A
,
Takanaga
H
,
Peng
JB
,
Hediger
MA
, et al
Calcium transporter 1 and epithelial calcium channel messenger ribonucleic acid are differentially regulated by 1,25 dihydroxyvitamin D3 in the intestine and kidney of mice
.
Endocrinology
2003
;
144
:
3885
94
.
28.
Williams
KB
,
DeLuca
HF
. 
Characterization of intestinal phosphate absorption using a novel in vivo method
.
Am J Physiol Endocrinol Metab
2007
;
292
:
E1917
21
.
29.
Marks
J
,
Srai
SK
,
Biber
J
,
Murer
H
,
Unwin
RJ
,
Debnam
ES
. 
Intestinal phosphate absorption and the effect of vitamin D: a comparison of rats with mice
.
Exp Physiol
2006
;
91
:
531
7
.
30.
Fujita
H
,
Sugimoto
K
,
Inatomi
S
,
Maeda
T
,
Osanai
M
,
Uchiyama
Y
, et al
Tight junction proteins claudin-2 and -12 are critical for vitamin D-dependent Ca2+ absorption between enterocytes
.
Mol Biol Cell
2008
;
19
:
1912
21
.
31.
Marino
R
,
Misra
M
. 
Extra-skeletal effects of vitamin D
.
Nutrients
2019
;
11
:
1460
.
32.
Dankers
W
,
Colin
EM
,
van Hamburg
JP
,
Lubberts
E
. 
Vitamin D in autoimmunity: molecular mechanisms and therapeutic potential
.
Front Immunol
2016
;
7
:
697
.
33.
Nakashima
A
,
Yokoyama
K
,
Yokoo
T
,
Urashima
M
. 
Role of vitamin D in diabetes mellitus and chronic kidney disease
.
World J Diabetes
2016
;
7
:
89
100
.
34.
Kheiri
B
,
Abdalla
A
,
Osman
M
,
Ahmed
S
,
Hassan
M
,
Bachuwa
G
. 
Vitamin D deficiency and risk of cardiovascular diseases: a narrative review
.
Clin Hypertens
2018
;
24
:
9
.
35.
Jeong
HY
,
Park
KM
,
Lee
MJ
,
Yang
DH
,
Kim
SH
,
Lee
SY
. 
Vitamin D and hypertension
.
Electrolyte Blood Press
2017
;
15
:
1
11
.
36.
Farhangi
MA
,
Mesgari-Abbasi
M
,
Hajiluian
G
,
Nameni
G
,
Shahabi
P
. 
Adipose tissue inflammation and oxidative stress: the ameliorative effects of vitamin D
.
Inflammation
2017
;
40
:
1688
97
.
37.
Penckofer
S
,
Kouba
J
,
Wallis
DE
,
Emanuele
MA
. 
Vitamin D and diabetes: let the sunshine in
.
Diabetes Educ
2008
;
34
:
939
40
.
38.
Welsh
J
. 
Cellular and molecular effects of vitamin D on carcinogenesis
.
Arch Biochem Biophys
2012
;
523
:
107
14
.
39.
Hanahan
D
,
Weinberg
RA
. 
The hallmarks of cancer
.
Cell
2000
;
100
:
57
70
.
40.
Zhang
Y
,
Guo
Q
,
Zhang
Z
,
Bai
N
,
Liu
Z
,
Xiong
M
, et al
VDR status arbitrates the prometastatic effects of tumor-associated macrophages
.
Mol Cancer Res
2014
;
12
:
1181
91
.
41.
Huss
L
,
Butt
ST
,
Borgquist
S
,
Elebro
K
,
Sandsveden
M
,
Rosendahl
A
, et al
Vitamin D receptor expression in invasive breast tumors and breast cancer survival
.
Breast Cancer Res
2019
;
21
:
84
.
42.
Al-Azhri
J
,
Zhang
Y
,
Bshara
W
,
Zirpoli
G
,
McCann
SE
,
Khoury
T
, et al
Tumor expression of vitamin D receptor and breast cancer histopathological characteristics and prognosis
.
Clin Cancer Res
2017
;
23
:
97
103
.
43.
Ditsch
N
,
Toth
B
,
Mayr
D
,
Lenhard
M
,
Gallwas
J
,
Weissenbacher
T
, et al
The association between vitamin D receptor expression and prolonged overall survival in breast cancer
.
J Histochem Cytochem
2012
;
60
:
121
9
.
44.
Zinser
GM
,
Welsh
J
. 
Vitamin D receptor status alters mammary gland morphology and tumorigenesis in MMTV-neu mice
.
Carcinogenesis
2004
;
25
:
2361
72
.
45.
Zinser
GM
,
Welsh
J
. 
Effect of Vitamin D3 receptor ablation on murine mammary gland development and tumorigenesis
.
J Steroid Biochem Mol Biol
2004
;
89–90
:
433
6
.
46.
Li
J
,
Luco
AL
,
Ochietti
B
,
Fadhil
I
,
Camirand
A
,
Reinhardt
TA
, et al
Tumoral vitamin D synthesis by CYP27B1 1-alpha-Hydroxylase delays mammary tumor progression in the PyMT-MMTV mouse model and its action involves NF-kappaB modulation
.
Endocrinology
2016
;
157
:
2204
16
.
47.
Frampton
RJ
,
Omond
SA
,
Eisman
JA
. 
Inhibition of human cancer cell growth by 1,25-dihydroxyvitamin D3 metabolites
.
Cancer Res
1983
;
43
:
4443
7
.
48.
Colston
KW
,
Chander
SK
,
Mackay
AG
,
Coombes
RC
. 
Effects of synthetic vitamin D analogues on breast cancer cell proliferation in vivo and in vitro
.
Biochem Pharmacol
1992
;
44
:
693
702
.
49.
Simboli-Campbell
M
,
Narvaez
CJ
,
van Weelden
K
,
Tenniswood
M
,
Welsh
J
. 
Comparative effects of 1,25(OH)2D3 and EB1089 on cell cycle kinetics and apoptosis in MCF-7 breast cancer cells
.
Breast Cancer Res Treat
1997
;
42
:
31
41
.
50.
Lee
HJ
,
Liu
H
,
Goodman
C
,
Ji
Y
,
Maehr
H
,
Uskokovic
M
, et al
Gene expression profiling changes induced by a novel Gemini Vitamin D derivative during the progression of breast cancer
.
Biochem Pharmacol
2006
;
72
:
332
43
.
51.
Swami
S
,
Raghavachari
N
,
Muller
UR
,
Bao
YP
,
Feldman
D
. 
Vitamin D growth inhibition of breast cancer cells: gene expression patterns assessed by cDNA microarray
.
Breast Cancer Res Treat
2003
;
80
:
49
62
.
52.
Ooi
LL
,
Zhou
H
,
Kalak
R
,
Zheng
Y
,
Conigrave
AD
,
Seibel
MJ
, et al
Vitamin D deficiency promotes human breast cancer growth in a murine model of bone metastasis
.
Cancer Res
2010
;
70
:
1835
44
.
53.
VanWeelden
K
,
Flanagan
L
,
Binderup
L
,
Tenniswood
M
,
Welsh
J
. 
Apoptotic regression of MCF-7 xenografts in nude mice treated with the vitamin D3 analog, EB1089
.
Endocrinology
1998
;
139
:
2102
10
.
54.
Escaleira
MT
,
Brentani
MM
. 
Vitamin D3 receptor (VDR) expression in HC-11 mammary cells: regulation by growth-modulatory agents, differentiation, and Ha-ras transformation
.
Breast Cancer Res Treat
1999
;
54
:
123
33
.
55.
Wu
G
,
Fan
RS
,
Li
W
,
Ko
TC
,
Brattain
MG
. 
Modulation of cell cycle control by vitamin D3 and its analogue, EB1089, in human breast cancer cells
.
Oncogene
1997
;
15
:
1555
63
.
56.
Verlinden
L
,
Verstuyf
A
,
Convents
R
,
Marcelis
S
,
Van Camp
M
,
Bouillon
R
. 
Action of 1,25(OH)2D3 on the cell cycle genes, cyclin D1, p21 and p27 in MCF-7 cells
.
Mol Cell Endocrinol
1998
;
142
:
57
65
.
57.
Wang
Q
,
Lee
D
,
Sysounthone
V
,
Chandraratna
RAS
,
Christakos
S
,
Korah
R
, et al
1,25-dihydroxyvitamin D3 and retonic acid analogues induce differentiation in breast cancer cells with function- and cell-specific additive effects
.
Breast Cancer Res Treat
2001
;
67
:
157
68
.
58.
Shan
NL
,
Minden
A
,
Furmanski
P
,
Bak
MJ
,
Cai
L
,
Wernyj
R
, et al
Analysis of the transcriptome: regulation of cancer stemness in breast ductal carcinoma in situ by vitamin D compounds
.
Cancer Prev Res
2020
;
13
:
673
86
.
59.
Shan
NL
,
Wahler
J
,
Lee
HJ
,
Bak
MJ
,
Gupta
SD
,
Maehr
H
, et al
Vitamin D compounds inhibit cancer stem-like cells and induce differentiation in triple negative breast cancer
.
J Steroid Biochem Mol Biol
2017
;
173
:
122
9
.
60.
Zhalehjoo
N
,
Shakiba
Y
,
Panjehpour
M
. 
Gene expression profiles of CYP24A1 and CYP27B1 in malignant and normal breast tissues
.
Mol Med Rep
2017
;
15
:
467
73
.
61.
Lopes
N
,
Sousa
B
,
Martins
D
,
Gomes
M
,
Vieira
D
,
Veronese
LA
, et al
Alterations in Vitamin D signalling and metabolic pathways in breast cancer progression: a study of VDR, CYP27B1 and CYP24A1 expression in benign and malignant breast lesions
.
BMC Cancer
2010
;
10
:
483
.
62.
Townsend
K
,
Banwell
CM
,
Guy
M
,
Colston
KW
,
Mansi
JL
,
Stewart
PM
, et al
Autocrine metabolism of vitamin D in normal and malignant breast tissue
.
Clin Cancer Res
2005
;
11
:
3579
86
.
63.
Osanai
M
,
Lee
GH
. 
CYP24A1-induced vitamin D insufficiency promotes breast cancer growth
.
Oncol Rep
2016
;
36
:
2755
62
.
64.
Sheng
L
,
Turner
AG
,
Barratt
K
,
Kremer
R
,
Morris
HA
,
Callen
DF
, et al
Mammary-specific ablation of Cyp24a1 inhibits development, reduces proliferation and increases sensitivity to vitamin D
.
J Steroid Biochem Mol Biol
2019
;
189
:
240
7
.
65.
Albertson
DG
,
Ylstra
B
,
Segraves
R
,
Collins
C
,
Dairkee
SH
,
Kowbel
D
, et al
Quantitative mapping of amplicon structure by array CGH identifies CYP24 as a candidate oncogene
.
Nat Genet
2000
;
25
:
144
6
.
66.
Mantell
DJ
,
Owens
PE
,
Bundred
NJ
,
Mawer
EB
,
Canfield
AE
. 
1 alpha,25-dihydroxyvitamin D(3) inhibits angiogenesis in vitro and in vivo
.
Circ Res
2000
;
87
:
214
20
.
67.
Wilmanski
T
,
Barnard
A
,
Parikh
MR
,
Kirshner
J
,
Buhman
K
,
Burgess
J
, et al
1alpha,25-Dihydroxyvitamin D inhibits the metastatic capability of MCF10CA1a and MDA-MB-231 cells in an in vitro model of breast to bone metastasis
.
Nutr Cancer
2016
;
68
:
1202
9
.
68.
Koli
K
,
Keski-Oja
J
. 
1alpha,25-dihydroxyvitamin D3 and its analogues down-regulate cell invasion-associated proteases in cultured malignant cells
.
Cell Growth Differ
2000
;
11
:
221
9
.
69.
Pendas-Franco
N
,
Gonzalez-Sancho
JM
,
Suarez
Y
,
Aguilera
O
,
Steinmeyer
A
,
Gamallo
C
, et al
Vitamin D regulates the phenotype of human breast cancer cells
.
Differentiation
2007
;
75
:
193
207
.
70.
Li
J
,
Luco
AL
,
Camirand
A
,
St-Arnaud
R
,
Kremer
R
. 
Vitamin D regulates CXCL12/CXCR4 and epithelial-to-mesenchymal transition in a model of breast cancer metastasis to lung
.
Endocrinology
2021
;
162
:
bqab049
.
71.
Veldman
CM
,
Cantorna
MT
,
DeLuca
HF
. 
Expression of 1,25-dihydroxyvitamin D(3) receptor in the immune system
.
Arch Biochem Biophys
2000
;
374
:
334
8
.
72.
Kreutz
M
,
Andreesen
R
,
Krause
SW
,
Szabo
A
,
Ritz
E
,
Reichel
H
. 
1,25-dihydroxyvitamin D3 production and vitamin D3 receptor expression are developmentally regulated during differentiation of human monocytes into macrophages
.
Blood
1993
;
82
:
1300
7
.
73.
Provvedini
DM
,
Tsoukas
CD
,
Deftos
LJ
,
Manolagas
SC
. 
1,25-dihydroxyvitamin D3 receptors in human leukocytes
.
Science
1983
;
221
:
1181
3
.
74.
Hewison
M
,
Freeman
L
,
Hughes
SV
,
Evans
KN
,
Bland
R
,
Eliopoulos
AG
, et al
Differential regulation of vitamin D receptor and its ligand in human monocyte-derived dendritic cells
.
J Immunol
2003
;
170
:
5382
90
.
75.
Takahashi
K
,
Nakayama
Y
,
Horiuchi
H
,
Ohta
T
,
Komoriya
K
,
Ohmori
H
, et al
Human neutrophils express messenger RNA of vitamin D receptor and respond to 1alpha,25-dihydroxyvitamin D3
.
Immunopharmacol Immunotoxicol
2002
;
24
:
335
47
.
76.
Balogh
G
,
de Boland
AR
,
Boland
R
,
Barja
P
. 
Effect of 1,25(OH)(2)-vitamin D(3) on the activation of natural killer cells: role of protein kinase C and extracellular calcium
.
Exp Mol Pathol
1999
;
67
:
63
74
.
77.
Takada
I
,
Makishima
M
. 
Control of inflammatory bowel disease and colorectal cancer by synthetic vitamin D receptor ligands
.
Curr Med Chem
2017
;
24
:
868
75
.
78.
Zhou
L
,
Wang
J
,
Li
J
,
Li
T
,
Chen
Y
,
June
RR
, et al
1,25-Dihydroxyvitamin D3 ameliorates collagen-induced arthritis via suppression of Th17 cells through miR-124 mediated inhibition of IL-6 signaling
.
Front Immunol
2019
;
10
:
178
.
79.
El-Boshy
M
,
BaSalamah
MA
,
Ahmad
J
,
Idris
S
,
Mahbub
A
,
Abdelghany
AH
, et al
Vitamin D protects against oxidative stress, inflammation and hepatorenal damage induced by acute paracetamol toxicity in rat
.
Free Radic Biol Med
2019
;
141
:
310
21
.
80.
Liang
S
,
Cai
J
,
Li
Y
,
Yang
R
. 
1,25DihydroxyVitamin D3 induces macrophage polarization to M2 by upregulating Tcell Igmucin3 expression
.
Mol Med Rep
2019
;
19
:
3707
13
.
81.
Kankova
M
,
Luini
W
,
Pedrazzoni
M
,
Riganti
F
,
Sironi
M
,
Bottazzi
B
, et al
Impairment of cytokine production in mice fed a vitamin D3-deficient diet
.
Immunology
1991
;
73
:
466
71
.
82.
Eleftheriadis
T
,
Antoniadi
G
,
Liakopoulos
V
,
Kartsios
C
,
Stefanidis
I
,
Galaktidou
G
. 
Paricalcitol reduces basal and lipopolysaccharide-induced (LPS) TNF-alpha and IL-8 production by human peripheral blood mononuclear cells
.
Int Urol Nephrol
2010
;
42
:
181
5
.
83.
Mattner
F
,
Smiroldo
S
,
Galbiati
F
,
Muller
M
,
Di Lucia
P
,
Poliani
PL
, et al
Inhibition of Th1 development and treatment of chronic-relapsing experimental allergic encephalomyelitis by a non-hypercalcemic analogue of 1,25-dihydroxyvitamin D (3)
.
Eur J Immunol
2000
;
30
:
498
508
.
84.
Chen
L
,
Eapen
MS
,
Zosky
GR
. 
Vitamin D both facilitates and attenuates the cellular response to lipopolysaccharide
.
Sci Rep
2017
;
7
:
45172
.
85.
Boonstra
A
,
Barrat
FJ
,
Crain
C
,
Heath
VL
,
Savelkoul
HF
,
O'Garra
A
. 
1alpha,25-Dihydroxyvitamin d3 has a direct effect on naive CD4(+) T cells to enhance the development of Th2 cells
.
J Immunol
2001
;
167
:
4974
80
.
86.
Barrat
FJ
,
Cua
DJ
,
Boonstra
A
,
Richards
DF
,
Crain
C
,
Savelkoul
HF
, et al
In vitro generation of interleukin 10-producing regulatory CD4(+) T cells is induced by immunosuppressive drugs and inhibited by T helper type 1 (Th1)- and Th2-inducing cytokines
.
J Exp Med
2002
;
195
:
603
16
.
87.
Jeffery
LE
,
Burke
F
,
Mura
M
,
Zheng
Y
,
Qureshi
OS
,
Hewison
M
, et al
1,25-Dihydroxyvitamin D3 and IL-2 combine to inhibit T cell production of inflammatory cytokines and promote development of regulatory T cells expressing CTLA-4 and FoxP3
.
J Immunol
2009
;
183
:
5458
67
.
88.
Tang
J
,
Zhou
R
,
Luger
D
,
Zhu
W
,
Silver
PB
,
Grajewski
RS
, et al
Calcitriol suppresses antiretinal autoimmunity through inhibitory effects on the Th17 effector response
.
J Immunol
2009
;
182
:
4624
32
.
89.
Piemonti
L
,
Monti
P
,
Sironi
M
,
Fraticelli
P
,
Leone
BE
,
Dal Cin
E
, et al
Vitamin D3 affects differentiation, maturation, and function of human monocyte-derived dendritic cells
.
J Immunol
2000
;
164
:
4443
51
.
90.
Almerighi
C
,
Sinistro
A
,
Cavazza
A
,
Ciaprini
C
,
Rocchi
G
,
Bergamini
A
. 
1Alpha,25-dihydroxyvitamin D3 inhibits CD40L-induced pro-inflammatory and immunomodulatory activity in human monocytes
.
Cytokine
2009
;
45
:
190
7
.
91.
Grivennikov
SI
,
Greten
FR
,
Karin
M
. 
Immunity, inflammation, and cancer
.
Cell
2010
;
140
:
883
99
.
92.
Tosolini
M
,
Kirilovsky
A
,
Mlecnik
B
,
Fredriksen
T
,
Mauger
S
,
Bindea
G
, et al
Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, Treg, th17) in patients with colorectal cancer
.
Cancer Res
2011
;
71
:
1263
71
.
93.
Mantovani
A
,
Allavena
P
,
Sica
A
,
Balkwill
F
. 
Cancer-related inflammation
.
Nature
2008
;
454
:
436
44
.
94.
Karkeni
E
,
Morin
SO
,
Bou Tayeh
B
,
Goubard
A
,
Josselin
E
,
Castellano
R
, et al
Vitamin D controls tumor growth and CD8+ T cell infiltration in breast cancer
.
Front Immunol
2019
;
10
:
1307
.
95.
Pennock
ND
,
Martinson
HA
,
Guo
Q
,
Betts
CB
,
Jindal
S
,
Tsujikawa
T
, et al
Ibuprofen supports macrophage differentiation, T cell recruitment, and tumor suppression in a model of postpartum breast cancer
.
J Immunother Cancer
2018
;
6
:
98
.
96.
Chandler
PD
,
Chen
WY
,
Ajala
ON
,
Hazra
A
,
Cook
N
,
Bubes
V
, et al
Effect of vitamin D3 supplements on development of advanced cancer: a secondary analysis of the VITAL randomized clinical trial
.
JAMA Netw Open
2020
;
3
:
e2025850
.
97.
Kremer
R
,
Campbell
PP
,
Reinhardt
T
,
Gilsanz
V
. 
Vitamin D status and its relationship to body fat, final height, and peak bone mass in young women
.
J Clin Endocrinol Metab
2009
;
94
:
67
73
.
98.
Zinser
G
,
Packman
K
,
Welsh
J
. 
Vitamin D(3) receptor ablation alters mammary gland morphogenesis
.
Development
2002
;
129
:
3067
76
.
99.
Ching
S
,
Kashinkunti
S
,
Niehaus
MD
,
Zinser
GM
. 
Mammary adipocytes bioactivate 25-hydroxyvitamin D(3) and signal via vitamin D(3) receptor, modulating mammary epithelial cell growth
.
J Cell Biochem
2011
;
112
:
3393
405
.
100.
Karkeni
E
,
Marcotorchino
J
,
Tourniaire
F
,
Astier
J
,
Peiretti
F
,
Darmon
P
, et al
Vitamin D limits chemokine expression in adipocytes and macrophage migration in vitro and in male mice
.
Endocrinology
2015
;
156
:
1782
93
.
101.
Cauley
JA
,
Chlebowski
RT
,
Wactawski-Wende
J
,
Robbins
JA
,
Rodabough
RJ
,
Chen
Z
, et al
Calcium plus vitamin D supplementation and health outcomes five years after active intervention ended: the Women's Health Initiative
.
J Womens Health
2013
;
22
:
915
29
.
102.
Bolland
MJ
,
Grey
A
,
Gamble
GD
,
Reid
IR
. 
Calcium and vitamin D supplements and health outcomes: a reanalysis of the Women's Health Initiative (WHI) limited-access data set
.
Am J Clin Nutr
2011
;
94
:
1144
9
.
103.
Manson
JE
,
Cook
NR
,
Lee
IM
,
Christen
W
,
Bassuk
SS
,
Mora
S
, et al
Vitamin D supplements and prevention of cancer and cardiovascular disease
.
N Engl J Med
2019
;
380
:
33
44
.
104.
Chlebowski
RT
,
Johnson
KC
,
Kooperberg
C
,
Pettinger
M
,
Wactawski-Wende
J
,
Rohan
T
, et al
Calcium plus vitamin D supplementation and the risk of breast cancer
.
J Natl Cancer Inst
2008
;
100
:
1581
91
.
105.
Trivedi
DP
,
Doll
R
,
Khaw
KT
. 
Effect of four monthly oral vitamin D3 (cholecalciferol) supplementation on fractures and mortality in men and women living in the community: randomised double blind controlled trial
.
BMJ
2003
;
326
:
469
.
106.
Avenell
A
,
MacLennan
GS
,
Jenkinson
DJ
,
McPherson
GC
,
McDonald
AM
,
Pant
PR
, et al
Long-term follow-up for mortality and cancer in a randomized placebo-controlled trial of vitamin D(3) and/or calcium (RECORD trial)
.
J Clin Endocrinol Metab
2012
;
97
:
614
22
.
107.
Lappe
J
,
Watson
P
,
Travers-Gustafson
D
,
Recker
R
,
Garland
C
,
Gorham
E
, et al
Effect of vitamin D and calcium supplementation on cancer incidence in older women: a randomized clinical trial
.
JAMA
2017
;
317
:
1234
43
.
108.
Manson
JE
,
Cook
NR
,
Lee
IM
,
Christen
W
,
Bassuk
SS
,
Mora
S
, et al
Marine n-3 fatty acids and prevention of cardiovascular disease and cancer
.
N Engl J Med
2019
;
380
:
23
32
.
109.
Manson
JE
,
Bassuk
SS
,
Buring
JE
,
Group
VR
. 
Principal results of the VITamin D and OmegA-3 TriaL (VITAL) and updated meta-analyses of relevant vitamin D trials
.
J Steroid Biochem Mol Biol
2020
;
198
:
105522
.
110.
Sperati
F
,
Vici
P
,
Maugeri-Sacca
M
,
Stranges
S
,
Santesso
N
,
Mariani
L
, et al
Vitamin D supplementation and breast cancer prevention: a systematic review and meta-analysis of randomized clinical trials
.
PLoS One
2013
;
8
:
e69269
.
111.
Zhou
L
,
Chen
B
,
Sheng
L
,
Turner
A
. 
The effect of vitamin D supplementation on the risk of breast cancer: a trial sequential meta-analysis
.
Breast Cancer Res Treat
2020
;
182
:
1
8
.
112.
Yin
L
,
Grandi
N
,
Raum
E
,
Haug
U
,
Arndt
V
,
Brenner
H
. 
Meta-analysis: serum vitamin D and breast cancer risk
.
Eur J Cancer
2010
;
46
:
2196
205
.
113.
Tommie
JL
,
Pinney
SM
,
Nommsen-Rivers
LA
. 
Serum vitamin D status and breast cancer risk by receptor status: a systematic review
.
Nutr Cancer
2018
;
70
:
804
20
.
114.
Knight
JA
,
Lesosky
M
,
Barnett
H
,
Raboud
JM
,
Vieth
R
. 
Vitamin D and reduced risk of breast cancer: a population-based case-control study
.
Cancer Epidemiol Biomarkers Prev
2007
;
16
:
422
9
.
115.
Crew
KD
,
Gammon
MD
,
Steck
SE
,
Hershman
DL
,
Cremers
S
,
Dworakowski
E
, et al
Association between plasma 25-hydroxyvitamin D and breast cancer risk
.
Cancer Prev Res
2009
;
2
:
598
604
.
116.
Agborsangaya
CB
,
Surcel
HM
,
Toriola
AT
,
Pukkala
E
,
Parkkila
S
,
Tuohimaa
P
, et al
Serum 25-hydroxyvitamin D at pregnancy and risk of breast cancer in a prospective study
.
Eur J Cancer
2010
;
46
:
467
70
.
117.
Crew
KD
,
Anderson
GL
,
Hershman
DL
,
Terry
MB
,
Tehranifar
P
,
Lew
DL
, et al
Randomized double-blind placebo-controlled biomarker modulation study of Vitamin D supplementation in premenopausal women at high risk for breast cancer (SWOG S0812)
.
Cancer Prev Res
2019
;
12
:
481
90
.
118.
Apoe
O
,
Jung
SH
,
Liu
H
,
Seisler
DK
,
Charlamb
J
,
Zekan
P
, et al
Effect of vitamin D supplementation on breast cancer biomarkers: CALGB 70806 (Alliance) study design and baseline data
.
Am J Hematol Oncol
2016
;
12
:
4
9
.
119.
Nichols
HB
,
Schoemaker
MJ
,
Cai
J
,
Xu
J
,
Wright
LB
,
Brook
MN
, et al
Breast cancer risk after recent childbirth: a pooled analysis of 15 prospective studies
.
Ann Intern Med
2019
;
170
:
22
30
.
120.
Lambe
M
,
Hsieh
C
,
Trichopoulos
D
,
Ekbom
A
,
Pavia
M
,
Adami
HO
. 
Transient increase in the risk of breast cancer after giving birth
.
N Engl J Med
1994
;
331
:
5
9
.
121.
Borges
VF
,
Lyons
TR
,
Germain
D
,
Schedin
P
. 
Postpartum Involution and cancer: an opportunity for targeted breast cancer prevention and treatments?
Cancer Res
2020
;
80
:
1790
8
.
122.
Callihan
EB
,
Gao
D
,
Jindal
S
,
Lyons
TR
,
Manthey
E
,
Edgerton
S
, et al
Postpartum diagnosis demonstrates a high risk for metastasis and merits an expanded definition of pregnancy-associated breast cancer
.
Breast Cancer Res Treat
2013
;
138
:
549
59
.
123.
Hartman
EK
,
Eslick
GD
. 
The prognosis of women diagnosed with breast cancer before, during and after pregnancy: a meta-analysis
.
Breast Cancer Res Treat
2016
;
160
:
347
60
.
124.
Goddard
ET
,
Bassale
S
,
Schedin
T
,
Jindal
S
,
Johnston
J
,
Cabral
E
, et al
Association between postpartum breast cancer diagnosis and metastasis and the clinical features underlying risk
.
JAMA Netw Open
2019
;
2
:
e186997
.
125.
Amant
F
,
von Minckwitz
G
,
Han
SN
,
Bontenbal
M
,
Ring
AE
,
Giermek
J
, et al
Prognosis of women with primary breast cancer diagnosed during pregnancy: results from an international collaborative study
.
J Clin Oncol
2013
;
31
:
2532
9
.
126.
Stein
T
,
Morris
JS
,
Davies
CR
,
Weber-Hall
SJ
,
Duffy
MA
,
Heath
VJ
, et al
Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3
.
Breast Cancer Res
2004
;
6
:
R75
91
.
127.
O'Brien
J
,
Lyons
T
,
Monks
J
,
Lucia
MS
,
Wilson
RS
,
Hines
L
, et al
Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species
.
Am J Pathol
2010
;
176
:
1241
55
.
128.
Martinson
HA
,
Jindal
S
,
Durand-Rougely
C
,
Borges
VF
,
Schedin
P
. 
Wound healing-like immune program facilitates postpartum mammary gland involution and tumor progression
.
Int J Cancer
2015
;
136
:
1803
13
.
129.
Bemis
LT
,
Schedin
P
. 
Reproductive state of rat mammary gland stroma modulates human breast cancer cell migration and invasion
.
Cancer Res
2000
;
60
:
3414
8
.
130.
Lyons
TR
,
O'Brien
J
,
Borges
VF
,
Conklin
MW
,
Keely
PJ
,
Eliceiri
KW
, et al
Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2
.
Nat Med
2011
;
17
:
1109
15
.
131.
McDaniel
SM
,
Rumer
KK
,
Biroc
SL
,
Metz
RP
,
Singh
M
,
Porter
W
, et al
Remodeling of the mammary microenvironment after lactation promotes breast tumor cell metastasis
.
Am J Pathol
2006
;
168
:
608
20
.
132.
Goddard
ET
,
Hill
RC
,
Nemkov
T
,
D'Alessandro
A
,
Hansen
KC
,
Maller
O
, et al
The rodent liver undergoes weaning-induced involution and supports breast cancer metastasis
.
Cancer Discov
2017
;
7
:
177
87
.
133.
Jindal
S
,
Narasimhan
J
,
Borges
V
,
Schedin
P
. 
Characterization of weaning-induced breast involution in women: implications for young women's breast cancer.
NPJ Breast Cancer
2020
;
6
:
55
.
134.
Park
H
,
Brannon
PM
,
West
AA
,
Yan
J
,
Jiang
X
,
Perry
CA
, et al
Vitamin D metabolism varies among women in different reproductive states consuming the same intakes of vitamin D and related nutrients
.
J Nutr
2016
;
146
:
1537
45
.
135.
Mulligan
ML
,
Felton
SK
,
Riek
AE
,
Bernal-Mizrachi
C
. 
Implications of vitamin D deficiency in pregnancy and lactation
.
Am J Obstet Gynecol
2010
;
202
:
429 e1–9
.
136.
Gillies
BR
,
Ryan
BA
,
Tonkin
BA
,
Poulton
IJ
,
Ma
Y
,
Kirby
BJ
, et al
Absence of calcitriol causes increased lactational bone loss and lower milk calcium but does not impair post-lactation bone recovery in Cyp27b1 null mice
.
J Bone Miner Res
2018
;
33
:
16
26
.
137.
Van Cromphaut
SJ
,
Rummens
K
,
Stockmans
I
,
Van Herck
E
,
Dijcks
FA
,
Ederveen
AG
, et al
Intestinal calcium transporter genes are upregulated by estrogens and the reproductive cycle through vitamin D receptor-independent mechanisms
.
J Bone Miner Res
2003
;
18
:
1725
36
.
138.
DeMauro
S
,
Wysolmerski
J
. 
Hypercalcemia in breast cancer: an echo of bone mobilization during lactation?
J Mammary Gland Biol Neoplasia
2005
;
10
:
157
67
.
139.
VanHouten
JN
,
Dann
P
,
Stewart
AF
,
Watson
CJ
,
Pollak
M
,
Karaplis
AC
, et al
Mammary-specific deletion of parathyroid hormone-related protein preserves bone mass during lactation
.
J Clin Invest
2003
;
112
:
1429
36
.
140.
Bartlett
AQ
,
Vesco
KK
,
Purnell
JQ
,
Francisco
M
,
Goddard
E
,
DeBarber
A
, et al
Pregnancy and weaning regulate human maternal liver size and function
.
bioRxiv
2021
Available from
: https://www.biorxiv.org/content/10.1101/2021.02.18.431862v2.
141.
Rudolph
MC
,
McManaman
JL
,
Hunter
L
,
Phang
T
,
Neville
MC
. 
Functional development of the mammary gland: use of expression profiling and trajectory clustering to reveal changes in gene expression during pregnancy, lactation, and involution
.
J Mammary Gland Biol Neoplasia
2003
;
8
:
287
307
.
142.
Zinser
GM
,
Welsh
J
. 
Accelerated mammary gland development during pregnancy and delayed postlactational involution in vitamin D3 receptor null mice
.
Mol Endocrinol
2004
;
18
:
2208
23
.
143.
Colston
KW
,
Berger
U
,
Wilson
P
,
Hadcocks
L
,
Naeem
I
,
Earl
HM
, et al
Mammary gland 1,25-dihydroxyvitamin D3 receptor content during pregnancy and lactation
.
Mol Cell Endocrinol
1988
;
60
:
15
22
.
144.
Clarkson
RW
,
Wayland
MT
,
Lee
J
,
Freeman
T
,
Watson
CJ
. 
Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression
.
Breast Cancer Res
2004
;
6
:
R92
109
.
145.
Saraf
R
,
Morton
SM
,
Camargo
CA
 Jr
,
Grant
CC
. 
Global summary of maternal and newborn vitamin D status - a systematic review
.
Matern Child Nutr
2016
;
12
:
647
68
.
146.
Hilger
J
,
Friedel
A
,
Herr
R
,
Rausch
T
,
Roos
F
,
Wahl
DA
, et al
A systematic review of vitamin D status in populations worldwide
.
Br J Nutr
2014
;
111
:
23
45
.
147.
Ginde
AA
,
Sullivan
AF
,
Mansbach
JM
,
Camargo
CA
 Jr
. 
Vitamin D insufficiency in pregnant and nonpregnant women of childbearing age in the United States
.
Am J Obstet Gynecol
2010
;
202
:
436
.
148.
Gernand
AD
,
Simhan
HN
,
Klebanoff
MA
,
Bodnar
LM
. 
Maternal serum 25-hydroxyvitamin D and measures of newborn and placental weight in a U.S. multicenter cohort study
.
J Clin Endocrinol Metab
2013
;
98
:
398
404
.
149.
Thill
M
,
Reichert
K
,
Woeste
A
,
Polack
S
,
Fischer
D
,
Hoellen
F
, et al
Combined treatment of breast cancer cell lines with vitamin D and COX-2 inhibitors
.
Anticancer Res
2015
;
35
:
1189
95
.
150.
Moreno
J
,
Krishnan
AV
,
Swami
S
,
Nonn
L
,
Peehl
DM
,
Feldman
D
. 
Regulation of prostaglandin metabolism by calcitriol attenuates growth stimulation in prostate cancer cells
.
Cancer Res
2005
;
65
:
7917
25
.
151.
Grant
WB
,
Karras
SN
,
Bischoff-Ferrari
HA
,
Annweiler
C
,
Boucher
BJ
,
Juzeniene
A
, et al
Do studies reporting ‘U’-shaped serum 25-hydroxyvitamin D-health outcome relationships reflect adverse effects?
Dermatoendocrinol
2016
;
8
:
e1187349
.
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs International 4.0 License.