Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Am Heart Assoc ; 8(7): e010744, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30890022

ABSTRACT

Background Asian-American subgroups (Asian-Indian, Chinese, Filipino, Korean, Japanese, and Vietnamese) display varied cardiovascular disease mortality patterns, especially at younger ages. This study aims to examine the years of potential life lost because of ischemic heart disease and cerebrovascular disease among the 6 largest Asian-American subgroups compared with non-Hispanic whites. Methods and Results We used National Center for Health Statistics Multiple Causes of Death mortality files from 2003 to 2012 to calculate race-specific life expectancy, mean years of potential life lost, and years of potential life lost per 100 000 population for each Asian subgroup and non-Hispanic whites. Asian-American subgroups display heterogeneity in cardiovascular disease burden. Asian-Indians had a high burden of ischemic heart disease; Asian-Indian men lost 724 years per 100 000 population in 2012 and a mean of 17 years to ischemic heart disease. Respectively, Vietnamese and Filipino men and women lost a mean of 17 and 16 years of life to cerebrovascular disease; Filipino men lost 352 years per 100 000 population in 2012. All Asian subgroups for both sexes had higher years of life lost to cerebrovascular disease compared with non-Hispanic whites. Conclusions Cardiovascular disease burden varies among Asian subgroups, and contributes to greater premature mortality in certain subgroups. Asian-Indian and Filipino populations have the highest years of life lost because of ischemic heart disease and Filipino and Vietnamese have the highest years of life lost because of cerebrovascular disease. Analysis of risk factors and development of subgroup-specific interventions are required to address these health disparities.


Subject(s)
Asian/statistics & numerical data , Cerebrovascular Disorders/ethnology , Life Expectancy/ethnology , Myocardial Ischemia/ethnology , Aged , Aged, 80 and over , Cardiovascular Diseases/ethnology , Cardiovascular Diseases/mortality , Cause of Death , Cerebrovascular Disorders/mortality , China , Female , Humans , India , Japan , Male , Middle Aged , Myocardial Ischemia/mortality , Philippines , Republic of Korea , Sex Factors , United States , Vietnam , White People
2.
Circulation ; 138(1): e1-e34, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29794080

ABSTRACT

South Asians (from Bangladesh, Bhutan, India, the Maldives, Nepal, Pakistan, and Sri Lanka) make up one quarter of the world's population and are one of the fastest-growing ethnic groups in the United States. Although native South Asians share genetic and cultural risk factors with South Asians abroad, South Asians in the United States can differ in socioeconomic status, education, healthcare behaviors, attitudes, and health insurance, which can affect their risk and the treatment and outcomes of atherosclerotic cardiovascular disease (ASCVD). South Asians have higher proportional mortality rates from ASCVD compared with other Asian groups and non-Hispanic whites, in contrast to the finding that Asian Americans (Asian Indian, Chinese, Filipino, Japanese, Korean, and Vietnamese) aggregated as a group are at lower risk of ASCVD, largely because of the lower risk observed in East Asian populations. Literature relevant to South Asian populations regarding demographics and risk factors, health behaviors, and interventions, including physical activity, diet, medications, and community strategies, is summarized. The evidence to date is that the biology of ASCVD is complex but is no different in South Asians than in any other racial/ethnic group. A majority of the risk in South Asians can be explained by the increased prevalence of known risk factors, especially those related to insulin resistance, and no unique risk factors in this population have been found. This scientific statement focuses on how ASCVD risk factors affect the South Asian population in order to make recommendations for clinical strategies to reduce disease and for directions for future research to reduce ASCVD in this population.


Subject(s)
American Heart Association , Asian People , Atherosclerosis/ethnology , Atherosclerosis/therapy , Culturally Competent Care/standards , Emigrants and Immigrants , Asia, Western/ethnology , Atherosclerosis/diagnosis , Atherosclerosis/mortality , Comorbidity , Evidence-Based Medicine/standards , Health Status , Humans , Incidence , Indian Ocean Islands/ethnology , Life Style/ethnology , Prevalence , Prognosis , Risk Assessment , Risk Factors , United States/epidemiology
3.
Endocrinology ; 157(4): 1341-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26934299

ABSTRACT

Patients with breast cancer (BCa) frequently have preexisting vitamin D deficiency (low serum 25-hydroxyvitamin D) when their cancer develops. A number of epidemiological studies show an inverse association between BCa risk and vitamin D status in humans, although some studies have failed to find an association. In addition, several studies have reported that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic indicators. However, it is unknown whether this association is mechanistically causative and, if so, whether it results from systemic or tumor autonomous effects of vitamin D signaling. We found that ablation of vitamin D receptor expression within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We show that vitamin D signaling inhibits the expression of the tumor progression gene Id1, and this pathway is abrogated in vitamin D deficiency in vivo in 2 murine models of BCa. These findings are relevant to humans, because we discovered that the mechanism of VDR regulation of Inhibitor of differentiation 1 (ID1) is conserved in human BCa cells, and there is a negative correlation between serum 25-hydroxyvitamin D levels and the level of ID1 in primary tumors from patients with BCa.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction , Vitamin D Deficiency/metabolism , Animals , Blotting, Western , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/genetics , Mice, Inbred BALB C , Neoplasm Metastasis , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D/metabolism , Vitamin D Deficiency/complications , Vitamin D Deficiency/genetics
4.
Endocr Relat Cancer ; 23(4): 251-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26817629

ABSTRACT

Obesity is an established risk factor for postmenopausal breast cancer (BCa), insulin resistance, and vitamin D deficiency, and all contribute to increased synthesis of mammary estrogens, the drivers of estrogen receptor-positive (ER+) BCa growth. As both dietary vitamin D and calcitriol treatments inhibit breast estrogen synthesis and signaling, we hypothesized that vitamin D would be especially beneficial in mitigating the adverse effects of obesity on ER+BCa. To assess whether obesity exerted adverse effects on BCa growth and whether vitamin D compounds could reduce these unfavorable effects, we employed a diet-induced obesity (DIO) model in ovariectomized C57BL/6 mice. Breast tumor cells originally from syngeneic Mmtv-Wnt1 transgenic mice were then implanted into the mammary fat pads of lean and obese mice. DIO accelerated the initiation and progression of the mammary tumors. Treatments with either calcitriol or dietary vitamin D reduced the adverse effects of obesity causing a delay in tumor appearance and inhibiting continued tumor growth. Beneficial actions of treatments with vitamin D or calcitriol on BCa and surrounding adipose tissue included repressed Esr1, aromatase, and Cox2 expression; decreased tumor-derived estrogen and PGE2; reduced expression of leptin receptors; and increased adiponectin receptors. We demonstrate that vitamin D treatments decreased insulin resistance, reduced leptin, and increased adiponectin signaling and also regulated the LKB1/AMPK pathway contributing to an overall decrease in local estrogen synthesis in the obese mice. We conclude that calcitriol and dietary vitamin D, acting by multiple interrelated pathways, mitigate obesity-enhanced BCa growth in a postmenopausal setting.


Subject(s)
Dietary Supplements , Mammary Neoplasms, Experimental/metabolism , Obesity/metabolism , Vitamin D/pharmacology , AMP-Activated Protein Kinases/metabolism , Adiponectin/blood , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Aromatase/genetics , Calcium/blood , Cyclooxygenase 2/genetics , Diet, High-Fat , Dinoprostone/metabolism , Estradiol/metabolism , Estrogens/metabolism , Estrone/metabolism , Female , Humans , Leptin/blood , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/complications , Obesity/pathology , Ovariectomy , RNA, Messenger/metabolism , Tumor Burden , Vitamin D/blood
5.
Mol Cancer Ther ; 14(8): 1951-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25934710

ABSTRACT

The anticancer actions of vitamin D and its hormonally active form, calcitriol, have been extensively documented in clinical and preclinical studies. However, the mechanisms underlying these actions have not been completely elucidated. Here, we examined the effect of dietary vitamin D and calcitriol on mouse breast tumor-initiating cells (TICs, also known as cancer stem cells). We focused on MMTV-Wnt1 mammary tumors, for which markers for isolating TICs have previously been validated. We confirmed that these tumors expressed functional vitamin D receptors and estrogen receptors (ER) and exhibited calcitriol-induced molecular responses including ER downregulation. Following orthotopic implantation of MMTV-Wnt1 mammary tumor cells into mice, calcitriol injections or a vitamin D-supplemented diet caused a striking delay in tumor appearance and growth, whereas a vitamin D-deficient diet accelerated tumor appearance and growth. Calcitriol inhibited TIC tumor spheroid formation in a dose-dependent manner in primary cultures and inhibited TIC self-renewal in secondary passages. A combination of calcitriol and ionizing radiation inhibited spheroid formation more than either treatment alone. Further, calcitriol significantly decreased TIC frequency as evaluated by in vivo limiting dilution analyses. Calcitriol inhibition of TIC spheroid formation could be overcome by the overexpression of ß-catenin, suggesting that the inhibition of Wnt/ß-catenin pathway is an important mechanism mediating the TIC inhibitory activity of calcitriol in this tumor model. Our findings indicate that vitamin D compounds target breast TICs reducing tumor-initiating activity. Our data also suggest that combining vitamin D compounds with standard therapies may enhance anticancer activity and improve therapeutic outcomes.


Subject(s)
Calcitriol/pharmacology , Neoplastic Stem Cells/drug effects , Vitamin D/pharmacology , Animals , Body Weight , Calcium/blood , Cell Line, Tumor , Estrogens/metabolism , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/pathology , Mice , Neoplastic Stem Cells/metabolism , Receptors, Calcitriol/metabolism , Receptors, Estrogen/metabolism , Tumor Burden , Vitamin D/metabolism , Wnt Signaling Pathway/drug effects
6.
Nat Rev Cancer ; 14(5): 342-57, 2014 May.
Article in English | MEDLINE | ID: mdl-24705652

ABSTRACT

Vitamin D is not really a vitamin but the precursor to the potent steroid hormone calcitriol, which has widespread actions throughout the body. Calcitriol regulates numerous cellular pathways that could have a role in determining cancer risk and prognosis. Although epidemiological and early clinical trials are inconsistent, and randomized control trials in humans do not yet exist to conclusively support a beneficial role for vitamin D, accumulating results from preclinical and some clinical studies strongly suggest that vitamin D deficiency increases the risk of developing cancer and that avoiding deficiency and adding vitamin D supplements might be an economical and safe way to reduce cancer incidence and improve cancer prognosis and outcome.


Subject(s)
Calcitriol/physiology , Neoplasms/pathology , Neoplasms/prevention & control , Vitamin D/physiology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Breast Neoplasms/metabolism , Cholecalciferol/physiology , Colonic Neoplasms/metabolism , Disease Progression , Endocrine System , Female , Humans , Male , Neoplastic Stem Cells/cytology , Polymorphism, Genetic , Prognosis , Prostatic Neoplasms/metabolism , Randomized Controlled Trials as Topic , Risk , Signal Transduction , Steroid Hydroxylases/metabolism , Vitamin D Deficiency/complications , Vitamin D3 24-Hydroxylase
7.
Endocr Relat Cancer ; 20(4): 565-77, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23744764

ABSTRACT

Calcitriol (1,25-dihydroxyvitamin D3), the hormonally active metabolite of vitamin D, exerts its anti-proliferative activity in breast cancer (BCa) cells by multiple mechanisms including the downregulation of the expression of estrogen receptor α (ER). We analyzed an ∼3.5 kb ER promoter sequence and demonstrated the presence of two potential negative vitamin D response elements (nVDREs), a newly identified putative nVDRE upstream at -2488 to -2473 bp (distal nVDRE) and a previously published sequence (proximal nVDRE) at -94 to -70 bp proximal to the P1 start site. Transactivation analysis using ER promoter deletion constructs and heterologous promoter-reporter constructs revealed that both nVDREs functioned to mediate calcitriol transrepression. In the electrophoretic mobility shift assay, the vitamin D receptor (VDR) showed strong binding to both nVDREs in the presence of calcitriol, and the chromatin immunoprecipitation assay demonstrated the recruitment of the VDR to the distal nVDRE site. Mutations in the 5' hexameric DNA sequence of the distal nVDRE resulted in the loss of calcitriol-mediated transrepression and the inhibition of protein-DNA complex formation, demonstrating the importance of these nucleotides in VDR DNA binding and transrepression. A putative nuclear factor-Y (NFY) binding site, identified within the distal nVDRE, led to the findings that NFY bound to the distal nVDRE site interfered with the binding of the VDR at the site and reduced calcitriol-mediated transrepression. In conclusion, the ER promoter region contains two negative VDREs that act in concert to bind to the VDR and both nVDREs are required for the maximal inhibition of ER expression by calcitriol. The suppression of ER expression and estrogen-mediated signaling by calcitriol in BCa cells suggests that vitamin D may be useful in the treatment of ER+ BCa.


Subject(s)
Breast Neoplasms/genetics , Calcitriol/pharmacology , Receptors, Estrogen/genetics , Vitamin D Response Element/genetics , Vitamins/pharmacology , Breast Neoplasms/metabolism , CCAAT-Binding Factor/metabolism , Cell Line, Tumor , Female , Humans , Promoter Regions, Genetic , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Retinoid X Receptors/metabolism
8.
J Steroid Biochem Mol Biol ; 136: 289-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22939886

ABSTRACT

Calcitriol [1,25(OH)2D3], the hormonally active form of vitamin D exerts anti-proliferative, pro-apoptotic, anti-inflammatory effects and other anticancer actions in breast cancer (BCa) cell cultures and animal models of BCa. Our research is focused on investigating the potential beneficial effects of dietary vitamin D3 compared to calcitriol and the underlying mechanisms in BCa treatment and chemoprevention. We recently found that dietary vitamin D3 exhibits significant tumor inhibitory effects in xenograft models of BCa that are equivalent to those elicited by the administration of the active hormone calcitriol. At the easily achievable dose tested in our studies, dietary vitamin D3 exhibited substantial tumor inhibitory activity and, unlike calcitriol, did not cause hypercalcemia demonstrating its relative safety. We found elevations in circulating calcitriol as well as increased CYP27B1 expression in the tumor and the intestine in tumor-bearing mice ingesting a vitamin D3-supplemented diet. We hypothesize that the elevation in circulating 25(OH)D induced by dietary vitamin D3 supplements stimulates local synthesis of calcitriol in the mammary tumor microenvironment and the ensuing paracrine/autocrine actions play a major role in the anticancer activity of dietary vitamin D3. Our findings suggest that the endocrine activity of calcitriol derived from tumor and other extra-renal sources such as the intestine, probably also plays a role in mediating the anticancer effects of dietary vitamin D3. Thus it appears that multiple sites of 1α-hydroxylation contribute to the anticancer effects of dietary vitamin D3. Our data strongly suggest that dietary vitamin D will be useful in the chemoprevention and treatment of BCa since it is a safe, economical and easily available nutritional agent that is equivalent to calcitriol in exerting anticancer effects, at least in mouse models. Furthermore, adequate vitamin D nutrition and avoidance of vitamin D deficiency appear to be important in reducing BCa risk. These findings warrant clinical trials in BCa patients and in women at high risk for BCa to evaluate the benefits of dietary vitamin D3 supplementation. This article is part of a Special Issue entitled 'Vitamin D Workshop'.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/physiology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/therapeutic use , Calcitriol/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/prevention & control , Vitamin D/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Calcitriol/therapeutic use , Dietary Supplements , Female , Humans , Mammary Neoplasms, Experimental/enzymology , Mice , Therapeutic Equivalency , Tumor Microenvironment , Vitamin D/therapeutic use
9.
Steroids ; 77(11): 1107-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22801352

ABSTRACT

Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active form of vitamin D, inhibits the growth of many malignant cells including breast cancer (BCa) cells. The mechanisms of calcitriol anticancer actions include cell cycle arrest, stimulation of apoptosis and inhibition of invasion, metastasis and angiogenesis. In addition we have discovered new pathways of calcitriol action that are especially relevant in inhibiting the growth of estrogen receptor positive (ER+) BCa cells. Calcitriol suppresses COX-2 expression and increases that of 15-PGDH thereby reducing the levels of inflammatory prostaglandins (PGs). Our in vitro and in vivo studies show that calcitriol decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis selectively in BCa cells and in the mammary adipose tissue surrounding BCa, by a direct repression of aromatase transcription via promoter II as well as an indirect effect due to the reduction in the levels of PGs, which are major stimulator of aromatase transcription through promoter II. Calcitriol down-regulates the expression of ERα and thereby attenuates estrogen signaling in BCa cells including the proliferative stimulus provided by estrogens. Thus the inhibition of estrogen synthesis and signaling by calcitriol and its anti-inflammatory actions will play an important role in inhibiting ER+BCa. We hypothesize that dietary vitamin D would exhibit similar anticancer activity due to the presence of the enzyme 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1) in breast cells ensuring conversion of circulating 25-hydroxyvitamin D to calcitriol locally within the breast micro-environment where it can act in a paracrine manner to inhibit BCa growth. Cell culture and in vivo data in mice strongly suggest that calcitriol and dietary vitamin D would play a beneficial role in the prevention and/or treatment of ER+BCa in women.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Receptors, Estrogen/metabolism , Vitamin D/pharmacology , Vitamins/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Calcitriol/pharmacology , Calcitriol/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Dietary Supplements , Estrogens/metabolism , Female , Humans , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/prevention & control , Vitamin D/administration & dosage , Vitamin D/therapeutic use , Vitamins/administration & dosage , Vitamins/therapeutic use
10.
Rheum Dis Clin North Am ; 38(1): 161-78, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22525850

ABSTRACT

Considerable data described in the first part of this review suggest that there is a role for vitamin D in cancer therapy and prevention. Although the preclinical data are persuasive and the epidemiologic data intriguing, no well-designed clinical trial of optimal administration of vitamin D as a cancer therapy has ever been conducted. Had there been the opportunity and insight to develop calcitriol as any other cancer drug, the following studies would have been completed: 1. Definition of the MTD. 2. Definition of a phase II dose, as a single agent and in combination with cytotoxic agents. 3. Studies to define a biologically optimal dose. 4. Phase II (probably randomized phase II) studies of calcitriol alone and chemotherapy ± calcitriol. 5. Then, randomized phase III trials would be conducted and designed such that the only variable was the administration of calcitriol. Prerequisites 1 to 5 have not been completed for calcitriol. Preclinical data provide considerable rationale for continued development of vitamin D analogue-based cancer therapies. However, design of future studies should be informed by good clinical trials design principles and the mistakes of the past not repeated. Such studies may finally provide compelling data to prove whether or not there is a role for vitamin D analogues in cancer therapy.

11.
Prostate ; 72(15): 1628-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22457201

ABSTRACT

BACKGROUND: The potential role of vitamin D and soy in prostate cancer (PCa) prevention/treatment has gained much attention in recent years. In this study, we evaluated the anticancer activity of calcitriol, the active form of vitamin D, dietary soy, and their combinations in a mouse model of PCa. METHODS: Athymic male nude mice bearing PC-3 human PCa xenografts received diets containing 10 or 20 kcal% soy, calcitriol injections, or a combination of dietary soy and calcitriol. Changes in tumor growth, serum levels of 1,25(OH)(2)D and calcium, and regulation of tumor gene expression were examined. RESULTS: The combination treatments resulted in substantially greater inhibition of tumor growth than either agent alone. Soy diets alone caused a modest elevation in serum 1,25(OH)(2)D, whereas the calcitriol-soy combinations led to substantially elevated serum 1,25(OH)(2) D, hypercalcemia, and in some cases lethal toxicity. The combinations enhanced calcitriol activity in regulating target gene expression, including greater up-regulation of anti-proliferative (p21, IGFBP-3) and pro-apoptotic (Bax) genes, increased inhibition of anti-apoptotic (Bcl-2) and cell cycle promoting (cyclin D1) genes, and suppression of prostaglandin (PG) synthesis and signaling (COX-2, 15-PGDH, PG receptors). Increases in serum calcium were accompanied by elevated expression of intestinal calcium absorption genes (TRPV6, calbindin-9k). CONCLUSIONS: Soy increases the bioavailability of endogenous and administered calcitriol, thereby enhancing its anticancer effects and risk of hypercalcemia. Since both agents are easily available as dietary supplements, the increased potential for hypercalcemic toxicity becomes an important factor when considering the combined use of vitamin D and soy in PCa therapy.


Subject(s)
Adenocarcinoma/drug therapy , Calcitriol/therapeutic use , Hypercalcemia/chemically induced , Prostatic Neoplasms/drug therapy , Soybean Proteins/administration & dosage , Vitamins/therapeutic use , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Apoptosis/genetics , Calcitriol/adverse effects , Calcitriol/blood , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Dietary Supplements , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypercalcemia/pathology , Male , Mice , Mice, Nude , Prostaglandins/biosynthesis , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Soybean Proteins/adverse effects , Vitamins/adverse effects , Xenograft Model Antitumor Assays
12.
Endocrinology ; 153(6): 2576-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22454149

ABSTRACT

1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3) or calcitriol], the hormonally active vitamin D metabolite, exhibits anticancer actions in models of breast cancer and prostate cancer. Because CYP27B1 (1α-hydroxylase), the enzyme catalyzing 1,25(OH)(2)D(3) formation in the kidney, is also expressed in extrarenal tissues, we hypothesize that dietary vitamin D(3) will be converted to 25(OH)D(3) in the body and then to 1,25(OH)(2)D(3) locally in the cancer microenvironment in which it will exert autocrine/paracrine anticancer actions. Immunocompromised mice bearing MCF-7 breast cancer xenografts showed significant tumor shrinkage (>50%) after ingestion of a vitamin D(3)-supplemented diet (5000 IU/kg) compared with a control diet (1000 IU/kg). Dietary vitamin D(3) inhibition of tumor growth was equivalent to administered calcitriol (0.025, 0.05, or 0.1 µg/mouse, three times a week). Both treatments equivalently inhibited PC-3 prostate cancer xenograft growth but to a lesser extent than the MCF-7 tumors. Calcitriol at 0.05 µg and 0.1 µg caused modest but statistically significant increases in serum calcium levels indicating that the dietary vitamin D(3) comparison was to a maximally safe calcitriol dose. Dietary vitamin D(3) did not increase serum calcium, demonstrating its safety at the concentration tested. The vitamin D(3) diet raised circulating 1,25 dihydroxyvitamin D levels and did not alter CYP27B1 mRNA in the kidney but increased it in the tumors, suggesting that extrarenal sources including the tumors contributed to the elevated circulating 1,25 dihydroxyvitamin D(3). Both calcitriol and dietary vitamin D(3) were equipotent in suppressing estrogen synthesis and signaling and other proinflammatory and growth signaling pathways. These preclinical data demonstrate the potential utility of dietary vitamin D(3) supplementation in cancer prevention and therapy.


Subject(s)
Breast Neoplasms/drug therapy , Calcitriol/pharmacology , Cholecalciferol/pharmacology , Prostatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , Animals , Body Weight/drug effects , Breast Neoplasms/pathology , Calcitriol/administration & dosage , Calcium/blood , Cell Line, Tumor , Cholecalciferol/administration & dosage , Dietary Supplements , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Ovariectomy , Prostatic Neoplasms/pathology , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroid Hydroxylases/genetics , Tumor Burden/drug effects , Vitamin D3 24-Hydroxylase , Vitamins/pharmacology
13.
Horm Cancer ; 2(3): 190-202, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21686077

ABSTRACT

Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active metabolite of vitamin D, exerts many anticancer effects in breast cancer (BCa) cells. We have previously shown using cell culture models that calcitriol acts as a selective aromatase modulator (SAM) and inhibits estrogen synthesis and signaling in BCa cells. We have now examined calcitriol effects in vivo on aromatase expression, estrogen signaling, and tumor growth when used alone and in combination with aromatase inhibitors (AIs). In immunocompromised mice bearing MCF-7 xenografts, increasing doses of calcitriol exhibited significant tumor inhibitory effects (~50% to 70% decrease in tumor volume). At the suboptimal doses tested, anastrozole and letrozole also caused significant tumor shrinkage when used individually. Although the combinations of calcitriol and the AIs caused a statistically significant increase in tumor inhibition in comparison to the single agents, the cooperative interaction between these agents appeared to be minimal at the doses tested. Calcitriol decreased aromatase expression in the xenograft tumors. Importantly, calcitriol also acted as a SAM in the mouse, decreasing aromatase expression in the mammary adipose tissue, while increasing it in bone marrow cells and not altering it in the ovaries and uteri. As a result, calcitriol significantly reduced estrogen levels in the xenograft tumors and surrounding breast adipose tissue. In addition, calcitriol inhibited estrogen signaling by decreasing tumor ERα levels. Changes in tumor gene expression revealed the suppressive effects of calcitriol on inflammatory and growth signaling pathways and demonstrated cooperative interactions between calcitriol and AIs to modulate gene expression. We hypothesize that cumulatively these calcitriol actions would contribute to a beneficial effect when calcitriol is combined with an AI in the treatment of BCa.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/biosynthesis , Calcitriol/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Vitamins/pharmacology , Anastrozole , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogens/metabolism , Female , Humans , Immunohistochemistry , Letrozole , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Nude , Nitriles/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Triazoles/pharmacology , Xenograft Model Antitumor Assays
14.
Mol Cell Endocrinol ; 347(1-2): 61-9, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-21664249

ABSTRACT

Prostate cancer (PCa) is the second most common cancer in men worldwide. Epidemiological, molecular, and cellular studies have implicated vitamin D deficiency as a risk factor for the development and/or progression of PCa. Studies using cell culture systems and animal models suggest that vitamin D acts to reduce the growth of PCa through regulation of cellular proliferation and differentiation. However, although preclinical studies provide a strong indication for anti-cancer activity, proof of therapeutic benefits in men is still lacking. The anti-proliferative and pro-differentiating properties of vitamin D have been attributed to calcitriol [1,25(OH)(2)D(3)], the hormonally active form of vitamin D, acting through the vitamin D receptor (VDR). Metabolism of vitamin D in target tissues is mediated by two key enzymes: 1α-hydroxylase (CYP27B1), which catalyzes the synthesis of calcitriol from 25(OH)D and 24-hydroxylase (CYP24), which catalyzes the initial step in the conversion of calcitriol to less active metabolites. Many factors affect the balance of calcitriol synthesis and catabolism and several maneuvers, like combination therapy of calcitriol with other drugs, have been explored to treat PCa and reduce its risk. The current paper is an overview addressing some of the key factors that influence the biological actions of vitamin D and its metabolites in the treatment and/or prevention of PCa.


Subject(s)
Prostatic Neoplasms/metabolism , Vitamin D Deficiency/complications , Vitamin D/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Calcitriol/administration & dosage , Calcitriol/therapeutic use , Cholecalciferol/administration & dosage , Cholecalciferol/therapeutic use , Clinical Trials as Topic , Drug Therapy, Combination , Humans , Isoflavones/administration & dosage , Isoflavones/therapeutic use , Male , Mice , Prostatic Neoplasms/complications , Prostatic Neoplasms/prevention & control , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/physiology , Risk Factors , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Steroid Hydroxylases/physiology , Vitamin D/physiology , Vitamin D3 24-Hydroxylase
15.
Annu Rev Pharmacol Toxicol ; 51: 311-36, 2011.
Article in English | MEDLINE | ID: mdl-20936945

ABSTRACT

Calcitriol, the hormonally active form of vitamin D, is being evaluated in clinical trials as an anti-cancer agent. Calcitriol exerts multiple anti-proliferative, pro-apoptotic, and pro-differentiating actions on various malignant cells and retards tumor growth in animal models of cancer. Calcitriol also exhibits several anti-inflammatory effects including suppression of prostaglandin (PG) action, inhibition of p38 stress kinase signaling, and the subsequent production of pro-inflammatory cytokines and inhibition of NF-κB signaling. Calcitriol also decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis in breast cancer, both by a direct transcriptional repression and indirectly by reducing PGs, which are major stimulators of aromatase transcription. Other important effects include the suppression of tumor angiogenesis, invasion, and metastasis. These calcitriol actions provide a basis for its potential use in cancer therapy and chemoprevention. We summarize the status of trials involving calcitriol and its analogs, used alone or in combination with known anti-cancer agents.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Calcitriol/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Calcitriol/therapeutic use , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Neoplasms/drug therapy , Neoplasms/physiopathology , Neoplasms/prevention & control
16.
Endocrinol Metab Clin North Am ; 39(2): 401-18, table of contents, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20511060

ABSTRACT

Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active form of vitamin D, exerts growth inhibitory and prodifferentiating effects on many malignant cells and retards tumor growth in animal models. Calcitriol is being evaluated as an anticancer agent in several human cancers. The mechanisms underlying the anticancer effects of calcitriol include inhibition of cell proliferation, stimulation of apoptosis, suppression of inflammation, and inhibition of tumor angiogenesis, invasion, and metastasis. This review discusses some of the molecular pathways mediating these anticancer actions of calcitriol and the preclinical data in cell culture and animal models. The clinical trials evaluating the use of calcitriol and its analogues in the treatment of patients with cancer are described. The reasons for the lack of impressive beneficial effects in clinical trials compared with the substantial efficacy seen in preclinical models are discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcitriol/therapeutic use , Neoplasms/drug therapy , Vitamin D/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/pharmacology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Clinical Trials as Topic , Dual-Specificity Phosphatases/metabolism , Humans , Male , Mice , Mitogen-Activated Protein Kinase Phosphatases/metabolism , NF-kappa B/metabolism , Neoplasms/prevention & control , Neovascularization, Pathologic/drug therapy , Prostaglandins/metabolism , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D/toxicity
17.
J Steroid Biochem Mol Biol ; 121(1-2): 343-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20156557

ABSTRACT

Calcitriol (1,25-dihydroxyvitamin D3), the hormonally active metabolite of vitamin D, inhibits the growth and induces the differentiation of many malignant cells including breast cancer (BCa) cells. Calcitriol exerts its anti-proliferative activity in BCa cells by inducing cell cycle arrest and stimulating apoptosis. Calcitriol also inhibits invasion, metastasis and tumor angiogenesis in experimental models of BCa. Our recent studies show additional newly discovered pathways of calcitriol action to inhibit the growth of BCa cells. Calcitriol suppresses COX-2 expression and increases that of 15-PGDH thereby reducing the levels and biological activity of prostaglandins (PGs). Calcitriol decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis selectively in BCa cells and the breast adipose tissue surrounding BCa, by a direct repression of aromatase transcription via promoter II as well as an indirect effect due to the reduction in the levels and biological activity of PGE2, which is a major stimulator of aromatase transcription through promoter II in BCa. Calcitriol down-regulates the expression of estrogen receptor alpha and thereby attenuates estrogen signaling in BCa cells including the proliferative stimulus provided by estrogens. We hypothesize that the inhibition of estrogen synthesis and signaling by calcitriol and its anti-inflammatory actions will play an important role in the use of calcitriol for the prevention and/or treatment of BCa.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Antagonists/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Vitamin D/metabolism , Adipose Tissue/metabolism , Animals , Apoptosis , Breast/pathology , Calcitriol/metabolism , Cyclooxygenase 2/metabolism , Female , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Neoplasm Metastasis , Neovascularization, Pathologic , Signal Transduction
18.
Endocrinology ; 151(1): 32-42, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19906814

ABSTRACT

Aromatase, the enzyme that catalyzes estrogen synthesis, is critical for the progression of estrogen receptor-positive breast cancer (BCa) in postmenopausal women. We show that calcitriol, the hormonally active form of vitamin D, regulates the expression of aromatase in a tissue-selective manner. Calcitriol significantly decreased aromatase expression in human BCa cells and adipocytes and caused substantial increases in human osteosarcoma cells (a bone cell model exhibiting osteoblast phenotype in culture) and modest increases in ovarian cancer cells. Calcitriol administration to immunocompromised mice bearing human BCa xenografts decreased aromatase mRNA levels in the tumors and the surrounding mammary adipose tissue but did not alter ovarian aromatase expression. In BCa cells, calcitriol also reduced the levels of prostaglandins (PGs), major stimulators of aromatase transcription, by suppressing the expression of cyclooxygenase-2 (which catalyzes PG synthesis) and increasing that of 15-hydroxyprostaglandin dehydrogenase (which catalyzes PG degradation). The mechanism of aromatase down-regulation by calcitriol in BCa cells is therefore 2-fold: a direct repression of aromatase transcription via promoter II through the vitamin D-response elements identified in this promoter and an indirect suppression by reducing the levels of PGs. Combinations of calcitriol with three different aromatase inhibitors (AIs) caused enhanced inhibition of BCa cell growth. The combination of calcitriol and an AI may have potential benefits for BCa therapy. In addition to augmenting the ability of AIs to inhibit BCa growth, calcitriol acting as a selective aromatase modulator that increases aromatase expression in bone would reduce the estrogen deprivation in bone caused by the AIs, thus ameliorating the AI-induced side effect of osteoporosis.


Subject(s)
Aromatase/genetics , Breast Neoplasms/drug therapy , Calcitriol/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , 3T3-L1 Cells , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Animals , Aromatase/metabolism , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , COS Cells , Calcitriol/administration & dosage , Chlorocebus aethiops , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Organ Specificity/drug effects , Organ Specificity/genetics , Osteoporosis/chemically induced , Osteoporosis/prevention & control , Prostaglandins/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Endocr Relat Cancer ; 17(1): R19-38, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19926709

ABSTRACT

Calcitriol, the hormonally active form of vitamin D, exerts multiple anti-proliferative and pro-differentiating actions including cell cycle arrest and induction of apoptosis in many malignant cells, and the hormone is currently being evaluated in clinical trials as an anti-cancer agent. Recent research reveals that calcitriol also exhibits multiple anti-inflammatory effects. First, calcitriol inhibits the synthesis and biological actions of pro-inflammatory prostaglandins (PGs) by three mechanisms: i) suppression of the expression of cyclooxygenase-2, the enzyme that synthesizes PGs; ii) up-regulation of the expression of 15-hydroxyprostaglandin dehydrogenase, the enzyme that inactivates PGs; and iii) down-regulation of the expression of PG receptors that are essential for PG signaling. The combination of calcitriol and nonsteroidal anti-inflammatory drugs results in a synergistic inhibition of the growth of prostate cancer (PCa) cells and offers a potential therapeutic strategy for PCa. Second, calcitriol increases the expression of mitogen-activated protein kinase phosphatase 5 in prostate cells resulting in the subsequent inhibition of p38 stress kinase signaling and the attenuation of the production of pro-inflammatory cytokines. Third, calcitriol also exerts anti-inflammatory activity in PCa through the inhibition of nuclear factor-kappaB signaling that results in potent anti-inflammatory and anti-angiogenic effects. Other important direct effects of calcitriol as well as the consequences of its anti-inflammatory effects include the inhibition of tumor angiogenesis, invasion, and metastasis. We hypothesize that these anti-inflammatory actions, in addition to the other known anti-cancer effects of calcitriol, play an important role in its potential use as a therapeutic agent for PCa. Calcitriol or its analogs may have utility as chemopreventive agents and should be evaluated in clinical trials in PCa patients with early or precancerous disease.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Calcitriol/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/prevention & control , Adenocarcinoma/etiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Calcitriol/therapeutic use , Cell Line, Tumor/drug effects , Clinical Trials as Topic , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/genetics , Enzyme Induction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Hydroxyprostaglandin Dehydrogenases/genetics , Male , Mitogen-Activated Protein Kinase Phosphatases/biosynthesis , Mitogen-Activated Protein Kinase Phosphatases/genetics , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostaglandins/metabolism , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatitis/complications , Prostatitis/drug therapy , Receptors, Prostaglandin/biosynthesis , Receptors, Prostaglandin/genetics
20.
Int J Cancer ; 124(9): 2050-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19127598

ABSTRACT

Soy and its constituent isoflavone genistein inhibit the development and progression of prostate cancer (PCa). Our study in both cultured cells and PCa patients reveals a novel pathway for the actions of genistein, namely the inhibition of the synthesis and biological actions of prostaglandins (PGs), known stimulators of PCa growth. In the cell culture experiments, genistein decreased cyclooxygenase-2 (COX-2) mRNA and protein expression in both human PCa cell lines (LNCaP and PC-3) and primary prostate epithelial cells and increased 15-hydroxyprostaglandin dehydrogenase (15-PGDH) mRNA levels in primary prostate cells. As a result genistein significantly reduced the secretion of PGE(2) by these cells. EP4 and FP PG receptor mRNA were also reduced by genistein, providing an additional mechanism for the suppression of PG biological effects. Further, the growth stimulatory effects of both exogenous PGs and endogenous PGs derived from precursor arachidonic acid were attenuated by genistein. We also performed a pilot randomised double blind clinical study in which placebo or soy isoflavone supplements were given to PCa patients in the neo-adjuvant setting for 2 weeks before prostatectomy. Gene expression changes were measured in the prostatectomy specimens. In PCa patients ingesting isoflavones, we observed significant decreases in prostate COX-2 mRNA and increases in p21 mRNA. There were significant correlations between COX-2 mRNA suppression, p21 mRNA stimulation and serum isoflavone levels. We propose that the inhibition of the PG pathway contributes to the beneficial effect of soy isoflavones in PCa chemoprevention and/or treatment.


Subject(s)
Anticarcinogenic Agents/pharmacology , Dinoprostone/metabolism , Genistein/pharmacology , Isoflavones/pharmacology , Prostatic Neoplasms/drug therapy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Double-Blind Method , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Luciferases/metabolism , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Pilot Projects , Promoter Regions, Genetic , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Signal Transduction , Soy Foods , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...