30 juni 2012: Het volledige studierapport van Vitamin D, sunlight and prostate cancer risk is gratis in te zien op de website van het NCBI. Onderaan referentielijst toegevoegd van studies gerelateerd aan dit onderwerp.

December 2008: Bron: Pubmed.

Hier de abstracten van enkele studies (ook een overzichtstudie van gerandomiserde studies, die onderzochten wat en hoe hoog en op wat voor manier de vitamine D gehaltes in bloed in de winter kunnen worden beïnlvoed door bv. suppletie. Vooral hormoon gerelateerde vormen van kanker zoals prostaatkanker, en borstkanker lijken hierdoor te worden beïnvloed. Voor leken die goed engels begrijpen is hier een video te zien met een lezing van dr. Mercola over de rol van vitamine D voor onze gezondheid en ook als preventie van kanker.

1: Am J Clin Nutr. 2008 Dec;88(6):1528-34.

Efficacy of food fortification on serum 25-hydroxyvitamin D concentrations: systematic review.

Clinical Epidemiology Program, Ottawa Health Research Institute, Ottawa, Canada.

BACKGROUND: Many residents of the United States and Canada depend on dietary sources of vitamin D to help maintain vitamin D status. Because few natural food sources contain vitamin D, fortified foods may be required. OBJECTIVE: We aimed to determine the effects of vitamin D-fortified foods on serum 25-hydroxyvitamin D [25(OH)D] concentrations.

DESIGN: We searched MEDLINE (1966 to June Week 3 2006), Embase, CINAHL, AMED, Biological Abstracts, and the Cochrane Central Register of Controlled Trials for randomized controlled trials (RCTs) comparing vitamin D-fortified foods with a control and reporting serum 25(OH)D concentrations. Two reviewers independently determined study eligibility, assessed trial quality, and extracted relevant data. Disagreements were resolved by consensus. Meta-analyses of absolute mean change in 25(OH)D were conducted by using a random-effects model, with evaluation of heterogeneity.

RESULTS: Nine RCTs (n = 889 subjects) were included, of which 8 consistently showed a significant beneficial effect of food fortification on 25(OH)D concentrations. Although 7 RCTs (n = 585 subjects) potentially were meta-analyzable, we were unable to combine the overall results because of significant heterogeneity. The individual treatment effects ranged from 14.5 (95% CIs: 10.6, 18.4) nmol/L to 34.5 (17.64, 51.36) nmol/L (3.4-25 microg vitamin D/d). Subgroup analyses showed a reduction in heterogeneity and significant treatment effect when 4 trials that used milk as the fortified food source were combined. CONCLUSION: Most trials were small in size and inadequately reported allocation concealment, but results showed that vitamin D-fortified foods improved vitamin D status in adults.

PMID: 19064512 [PubMed - in process]

 

1: Am J Clin Nutr. 2008 Dec;88(6):1535-42.

 

 

Estimation of the dietary requirement for vitamin D in healthy adults.

Department of Food and Nutritional Sciences, University College, Cork, Ireland. k.cashman@ucc.ie

BACKGROUND: Knowledge gaps have contributed to considerable variation among international dietary recommendations for vitamin D.

OBJECTIVE: We aimed to establish the distribution of dietary vitamin D required to maintain serum 25-hydroxyvitamin D [25(OH)D] concentrations above several proposed cutoffs (ie, 25, 37.5, 50, and 80 nmol/L) during wintertime after adjustment for the effect of summer sunshine exposure and diet. DESIGN: A randomized, placebo-controlled, double-blind 22-wk intervention study was conducted in men and women aged 20-40 y (n = 238) by using different supplemental doses (0, 5, 10, and 15 microg/d) of vitamin D(3) throughout the winter. Serum 25(OH)D concentrations were measured by using enzyme-linked immunoassay at baseline (October 2006) and endpoint (March 2007).

RESULTS: There were clear dose-related increments (P < 0.0001) in serum 25(OH)D with increasing supplemental vitamin D(3). The slope of the relation between vitamin D intake and serum 25(OH)D was 1.96 nmol x L(-1) x microg(-1) intake. The vitamin D intake that maintained serum 25(OH)D concentrations of >25 nmol/L in 97.5% of the sample was 8.7 microg/d. This intake ranged from 7.2 microg/d in those who enjoyed sunshine exposure, 8.8 microg/d in those who sometimes had sun exposure, and 12.3 microg/d in those who avoided sunshine. Vitamin D intakes required to maintain serum 25(OH)D concentrations of >37.5, >50, and >80 nmol/L in 97.5% of the sample were 19.9, 28.0, and 41.1 microg/d, respectively. CONCLUSION: The range of vitamin D intakes required to ensure maintenance of wintertime vitamin D status [as defined by incremental cutoffs of serum 25(OH)D] in the vast majority (>97.5%) of 20-40-y-old adults, considering a variety of sun exposure preferences, is between 7.2 and 41.1 microg/d.

PMID: 19064513 [PubMed - in process]

 

11 november 2004: Bron: Pubmed

Bepaalde vormen van vitamine D zorgen voor inkapseling/doden van prostaatkankercellen in dierproeven. Al in 1996 toonden laboratoriumstudies aan dat vitamine D. in vitro kankercellen van prostaat, leukemie en borst doodde zonder enige bijwerkingen. Excuses maar we zijn geen medisch deskundige en kunnen niet precies aangeven welke vormen van vitamine D. hier zijn gebruikt, maar uw arts kan dat ongetwijfeld wel. Hier twee studies met meest recente bovenaan gepubliceerd. Deze studies komen uit Pubmed-Medline, waar nog meer studies aangeven dat bepaalde vormen van vitamine D. effectief werken in een behandeling van kanker.

Anticancer Res. 2003 Jan-Feb;23(1A):283-9.

Ability of potent vitamin D3 analogs to inhibit growth of prostate cancer cells in vivo.
Vegesna V, O'Kelly J, Said J, Uskokovic M, Binderup L, Koeffle HP. Cedars-Sinai Medical Center/UCLA School of Medicine, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
,br> BACKGROUND: Studies have identified analogs of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], which in vitro are 10- to 3,000-fold more active than 1,25(OH)2D3. We compared in vivo the anti-cancer activity of three potent vitamin D3 analogs and 1,25(OH)2D3 at near to each of their maximal tolerated dose (MTD).

MATERIALS AND METHODS: Human LNCaP prostate cancer xenografts were grown in nude mice and the animals were treated with intraperitoneal injections of either diluant; 1,25(OH)2D3; 1,25-Dihydroxy-20epi-22-oxa-24,26,27-trisho-mocholecalciferol (KH 1060); 1,25-Dihydroxy-22E,24E-diene-24,26,27-trishomocholecalciferol (EB 1039); and 1,25-Dihydroxy-16-ene-24-oxo-19-norcholecalciferol (RO 26-9114). Tumor sizes were measured weekly and tumor weights were measured at autopsy on the 12th week.

RESULTS: Each of the analogs equally and markedly inhibited growth of the prostate cancer xenografts. The 1,25(OH)2D3 initially inhibited growth but, by the time of sacrifice, the tumors were nearly the same size as diluant controls. The histological examination of the tumors showed that the analogs produced tumor necrosis and microcalcification. None of the mice developed hypercalcemia, which is the major toxicity of vitamin D3 compounds.

CONCLUSION: The MTD of the analogs varied by 400-fold but each had similar efficacy suggesting that, when choosing a vitamin D analog for clinical study, overall efficacy without toxicity is more important than the total amount of the compound that can be administered. In summary, we have identified three vitamin D analogs that show marked potency in vivo to inhibit growth of human prostate cancer xenografts; each had no detectable toxicity. This study should help lay the foundation for clinical studies.

PMID: 12680225 [PubMed - indexed for MEDLINE]

Hier het abstract van de in vitro studie uit 1998

Cancer Res. 1998 Aug 1;58(15):3370-5.

19-nor-26,27-bishomo-vitamin D3 analogs: a unique class of potent inhibitors of proliferation of prostate, breast, and hematopoietic cancer cells.

Kubota T, Koshizuka K, Koike M, Uskokovic M, Miyoshi I, Koeffler HP. Division of Hematology/Oncology, Cedars-Sinai Research Institute, University of California at Los Angeles School of Medicine, 90048, USA.

Vitamin D3 [1,25-dihydroxyvitamin-D3 (1,25(OH)2D3)] modulates the proliferation and differentiation of many cell types. Analogs of 1,25(OH)2D3 that have greater potency may become adjuvant therapy for breast and prostate cancers, myelodysplastic syndrome, acute myelogenous leukemia in remission and other cell types, especially in the setting of low disease burden. A new class of analogs of 1,25(OH)2D3 has been synthesized that has a novel 19-nor motif, as well as incorporating many structural elements previously shown to increase potency. These analogs were examined for their effects on prostate cancer cell lines (PC-3, LNCaP, and DU 145), a human breast cell line (MCF-7), and an acute myeloid leukemia cell line (HL-60). Dose-response clonogenic studies showed that each of these analogs had more potent antiproliferative activities against the cancer cells than 1,25(OH)2D3, and 1,25-(OH)2-16,23Z-diene-26,27-bishomo-19-nor-D3 (Ro 27-2014) was the most potent analog [10-fold increased activity compared to 1,25(OH)2D3]. Further studies were performed using Ro 27-2014. Pulse-exposure studies showed that a 5-day pulse-exposure to Ro 27-2014 (10(-7) M) in liquid culture was adequate to achieve a 50% inhibition of MCF-7 clonal growth in soft agar in the absence of the analog, suggesting that the growth inhibition mediated by the analog was irreversible. Cell cycle analyses using MCF-7 cells showed that Ro 27-2014 (10(-7) M for 4 days) induced a significant increase in the number of cells in G0-G1 (72.8+/-8.9% versus 49.9+/-3.5% in control cells), with a concomitant decrease in the percent of cells in S phase (13.1+/-6.2% versus 35.8+/-3.5% in control cells). The chief toxicity of vitamin D3 compounds is hypercalcemia, and therefore, we examined calcemic activity of Ro 27-2014 in mice and found it not to induce hypercalcemia at doses of 0.05 microg i.p. three times per week. In contrast, the same dose of a 19-nor vitamin D3 compound with 6 fluorines on the side chain (1,25-(OH)2-16-ene-23-yne-26,27-F6-19-nor-D3), although also having potent anticancer activity, caused severe hypercalcemia (18 mg/dl). In summary, 19-nor vitamin D3 compounds with desaturation and lengthening of their side chains result in a series of compounds with a good therapeutic index, having potent anticancer activity and low toxicity.

PMID: 9699668 [PubMed - indexed for MEDLINE]

Vitamin D, sunlight and prostate cancer risk

References

1. Lipscomb J. Estimating the cost of cancer care in the United States: a work very much in progress. Journal of the National Cancer Institute. 2008;100(9):607–610. [PubMed]
2. Bostwick DG, Burke HB, Djakiew D, et al. Human prostate cancer risk factors. Cancer. 2004;101(supplement 10):2371–2490. [PubMed]
3. Klein EA. Chemoprevention of prostate cancer. Annual Review of Medicine. 2006;57:49–63.
4. Lowe JF, Frazee LA. Update on prostate cancer chemoprevention. Pharmacotherapy. 2006;26(3):353–359. [PubMed]
5. Lippman SM, Lee JJ. Reducing the “risk”of chemoprevention: defining and targeting high risk—2005 AACR cancer research and prevention foundation award lecture. Cancer Research. 2006;66(6):2893–2903. [PubMed]
6. Shukla S, Gupta S. Dietary agents in the chemoprevention of prostate cancer. Nutrition and Cancer. 2005;53(1):18–32. [PubMed]
7. Thompson IM, Tangen CM, Goodman PJ, Lucia MS, Klein EA. Chemoprevention of prostate cancer. Journal of Urology. 2009;182(2):499–508. [PubMed]
8. Kennel KA, Drake MT, Hurley DL. Vitamin D deficiency in adults: when to test and how to treat. Mayo Clinic Proceedings. 2010;85(8):752–757. [PMC free article] [PubMed]
9. Ross AC, Manson JE, Abrams SA, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the institute of medicine: what clinicians need to know. The Journal of Clinical Endocrinology & Metabolism. 2011;96(1):53–58. [PMC free article] [PubMed]
10. Holick MF. Vitamin D and sunlight: strategies for cancer prevention and other health benefits. Clinical Journal of the American Society of Nephrology. 2008;3(5):1548–1554. [PubMed]
11. Toner CD, Davis CD, Milner JA. The vitamin D and cancer conundrum: aiming at a moving target. Journal of the American Dietetic Association. 2010;110(10):1492–1500. [PubMed]
12. Holick MF, Smith E, Pincus S. Skin as the site of vitamin D synthesis and target tissue for 1,25-dihydroxyvitamin D3: use of calcitriol (1,25-dihydroxyvitamin D3) for treatment of psoriasis. Archives of Dermatology. 1987;123(12):1677–1683. [PubMed]
13. Norman AW. Sunlight, season, skin pigmentation, vitamin D, and 25-hydroxyvitamin D: integral components of the vitamin D endocrine system. American Journal of Clinical Nutrition. 1998;67(6):1108–1110. [PubMed]
14. Glossmann HH. Origin of 7-dehydrocholesterol (provitamin D) in the skin. Journal of Investigative Dermatology. 2010;130(8):2139–2141. [PubMed]
15. Ingraham BA, Bragdon B, Nohe A. Molecular basis of the potential of vitamin D to prevent cancer. Current Medical Research and Opinion. 2008;24(1):139–149. [PubMed]
16. Gupta D, Lammersfeld CA, Trukova K, Lis CG. Vitamin D and prostate cancer risk: a review of the epidemiological literature. Prostate Cancer and Prostatic Diseases. 2009;12(3):215–226. [PubMed]
17. McCullough ML, Bostick RM, Mayo TL. Vitamin D gene pathway polymorphisms and risk of colorectal, breast, and prostate cancer. Annual Review of Nutrition. 2009;29:111–132.
18. Mucci LA, Spiegelman D. Vitamin D and prostate cancer risk—a less sunny outlook? Journal of the National Cancer Institute. 2008;100(11):759–761. [PubMed]
19. Jones G. Pharmacokinetics of vitamin D toxicity. American Journal of Clinical Nutrition. 2008;88(2):582S–586S. [PubMed]
20. Talwar SA, Aloia JF, Pollack S, Yeh JK. Dose response to vitamin D supplementation among postmenopausal African American women. American Journal of Clinical Nutrition. 2007;86(6):1657–1662. [PMC free article] [PubMed]
21. Heaney RP, Davies KM, Chen TC, Holick MF, Barger-Lux MJ. Human serum 25-hydroxycholecalciferol response to extended oral dosing with cholecalciferol. American Journal of Clinical Nutrition. 2003;77(1):204–210. [PubMed]
22. Aloia JF, Patel M, DiMaano R, et al. Vitamin D intake to attain a desired serum 25-hydroxyvitamin D concentration. American Journal of Clinical Nutrition. 2008;87(6):1952–1958. [PubMed]
23. Cashman KD, Hill TR, Lucey AJ, et al. Estimation of the dietary requirement for vitamin D in healthy adults. American Journal of Clinical Nutrition. 2008;88(6):1535–1542. [PubMed]
24. Trang HM, Cole DE, Rubin LA, Pierratos A, Siu S, Vieth R. Evidence that vitamin D3 increases serum 25-hydroxyvitamin D more efficiently than does vitamin D2. American Journal of Clinical Nutrition. 1998;68(4):854–858. [PubMed]
25. Armas LAG, Hollis BW, Heaney RP. Vitamin D2 is much less effective than vitamin D3 in humans. Journal of Clinical Endocrinology & Metabolism. 2004;89(11):5387–5391. [PubMed]
26. Holick MF, Biancuzzo RM, Chen TC, et al. Vitamin D2 is as effective as vitamin D3 in maintaining circulating concentrations of 25-hydroxyvitamin D. Journal of Clinical Endocrinology and Metabolism. 2008;93(3):677–681. [PMC free article] [PubMed]
27. Looker AC. Do body fat and exercise modulate vitamin D status? Nutrition Reviews. 2007;65(8):S124–S126. [PubMed]
28. Yanoff LB, Parikh SJ, Spitalnik A, et al. The prevalence of hypovitaminosis D and secondary hyperparathyroidism in obese black Americans. Clinical Endocrinology. 2006;64(5):523–529. [PMC free article] [PubMed]
29. Chennaiah S, Vijayalakshmi V, Suresh C. Effect of the supplementation of dietary rich phytoestrogens in altering the vitamin D levels in diet induced osteoporotic rat model. The Journal of Steroid Biochemistry and Molecular Biology. 2010;21(1-2):268–272. [PubMed]
30. Cross HS, Kallay E. Regulation of the colonic vitamin D system for prevention of tumor progression: an update. Future Oncology. 2009;5(4):493–507. [PubMed]
31. Hossein-nezhad A, Mirzaei K, Maghbooli Z, Najmafshar A, Larijani B. The influence of folic acid supplementation on maternal and fetal bone turnover. Journal of Bone and Mineral Metabolism. 2010;29(2):186–192. [PubMed]
32. Cross HS, Nittke T, Peterlik M. Modulation of vitamin D synthesis and catabolism in colorectal mucosa: a new target for cancer preventio. Anticancer Research. 2009;29(9):3705–3712. [PubMed]
33. Chen TC. 25-hydroxyvitamin D-1 alpha-hydroxylase (CYP27B1) is a new class of tumor suppressor in the prostate. Anticancer Research. 2008;28(4A):2015–2017. [PubMed]
34. Whitlatch LW, Young MV, Schwartz GG, et al. 25-hydroxyvitamin D-1alpha-hydroxylase activity is diminished in human prostate cancer cells and is enhanced by gene transfer. The Journal of Steroid Biochemistry and Molecular Biology. 2002;81(2):135–140. [PubMed]
35. Rungby J, Mortensen L, Jakobsen K, Brock A, Mosekilde L. Distribution of hydroxylated vitamin D metabolites [25OHD3 and 1,25(OH)2D3] in domestic pigs: evidence that 1,25(OH)2D3] is stored outside the blood circulation? Comparative Biochemistry and Physiology. 1993;104(3):483–484. [PubMed]
36. Konety BR, Somogyi G, Atan A, Muindi J, Chancellor MB, Getzenberg RH. Evaluation of intraprostatic metabolism of 1,25-dihydroxyvitamin D3 (calcitriol) using a microdialysis technique. Urology. 2002;59(6):947–952. [PubMed]
37. Lou YR, Qiao S, Talonpoika R, Syvälä H, Tuohimaa P. The role of vitamin D3 metabolism in prostate cancer. The Journal of Steroid Biochemistry and Molecular Biology. 2004;92(4):317–325. [PubMed]
38. Chen TC, Wang L, Whitlatch LW, Flanagan JN, Holick MF. Prostatic 25-hydroxyvitamin D-1alpha-hydroxylase and its implication in prostate cancer. Journal of Cellular Biochemistry. 2003;88(2):315–322. [PubMed]
39. Ma JF, Nonn L, Campbell MJ, Hewison M, Feldman D, Peehl DM. Mechanisms of decreased Vitamin D 1alpha-hydroxylase activity in prostate cancer cells. Molecular and Cellular Endocrinology. 2004;221(1-2):67–74. [PubMed]
40. Khorchide M, Lechner D, Cross HS. Epigenetic regulation of vitamin D hydroxylase expression and activity in normal and malignant human prostate cells. The Journal of Steroid Biochemistry and Molecular Biology. 2005;93(2-5):167–172. [PubMed]
41. Wang L, Persons KS, Jamieson D, et al. Prostate 25-hydroxyvitamin D-1alpha-hydroxylase is up-regulated by suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor. Anticancer Research. 2008;28(4A):2009–2013. [PubMed]
42. Lou YR, Laaksi I, Syvälä H, et al. 25-hydroxyvitamin D3 is an active hormone in human primary prostatic stromal cells. The FASEB journal. 2004;18(2):332–334. [PubMed]
43. Skowronski RJ, Peehl DM, Feldman D. Vitamin D and prostate cancer: 1,25 dihydroxyvitamin D3 receptors and actions in human prostate cancer cell lines. Endocrinology. 1993;132(5):1952–1960. [PubMed]
44. Miller GJ, Stapleton GE, Hedlund TE, Moffatt KA. Vitamin D receptor expression, 24-hydroxylase activity, and inhibition of growth by 1alpha,25-dihydroxyvitamin D3 in seven human prostatic carcinoma cell lines. Clinical Cancer Research. 1995;1(9):997–1003. [PubMed]
45. Luo W, Karpf AR, Deeb KK, et al. Epigenetic mechanisms of promigratory chemokine CXCL14 regulation in human prostate cancer cells. Cancer Research. 2010;70(14):5953–5962. [PMC free article] [PubMed]
46. Roff A, Wilson RT. A novel SNP in a vitamin D response element of the CYP24A1 promoter reduces protein binding, transactivation, and gene expression. The Journal of Steroid Biochemistry and Molecular Biology. 2008;112(1–3):47–54. [PMC free article] [PubMed]
47. Muindi JR, Nganga A, Engler KL, Coignet LJ, Johnson CS, Trump DL. CYP24 splicing variants are associated with different patterns of constitutive and calcitriol-inducible CYP24 activity in human prostate cancer cell lines. The Journal of Steroid Biochemistry and Molecular Biology. 2007;103(3–5):334–337. [PubMed]
48. Lou YR, Tuohimaa P. Androgen enhances the antiproliferative activity of vitamin D3 by suppressing 24-hydroxylase expression in LNCaP cells. The Journal of Steroid Biochemistry and Molecular Biology. 2006;99(1):44–49. [PubMed]
49. Lou YR, Nazarova N, Talonpoika R, et al. 5alpha-dihydrotestosterone inhibits 1alpha,25-dihydroxyvitamin D3-induced expression of CYP24 in human prostate cancer cells. Prostate. 2005;63(3):222–230. [PubMed]
50. Lou YR, Miettinen S, Kagechika H, Gronemeyer H, Tuohimaa P. Retinoic acid via RARalpha inhibits the expression of 24-hydroxylase in human prostate stromal cells. Biochemical and Biophysical Research Communications. 2005;338(4):1973–1981. [PubMed]
51. Farhan H, Wähälä K, Cross HS. Genistein inhibits vitamin D hydroxylases CYP24 and CYP27B1 expression in prostate cells. The Journal of Steroid Biochemistry and Molecular Biology. 2003;84(4):423–429. [PubMed]
52. Swami S, Krishnan AV, Peehl DM, Feldman D. Genistein potentiates the growth inhibitory effects of 1,25-dihydroxyvitamin D3 in DU145 human prostate cancer cells: role of the direct inhibition of CYP24 enzyme activity. Molecular and Cellular Endocrinology. 2005;241(1-2):49–61. [PubMed]
53. Muindi JR, WD Yu, Ma Y, et al. CYP24A1 inhibition enhances the antitumor activity of calcitriol. Endocrinology. 2010;151(9):4301–4312. [PMC free article] [PubMed]
54. Krishnan AV, Trump DL, Johnson CS, Feldman D. The role of vitamin D in cancer prevention and treatment. Endocrinology and Metabolism Clinics of North America. 2010;39(2):401–418. [PubMed]
55. Pike JW, Meyer MB. The vitamin D receptor: new paradigms for the regulation of gene expression by 1,25-dihydroxyvitamin D3. Endocrinology and Metabolism Clinics of North America. 2010;39(2):255–269. [PMC free article] [PubMed]
56. Mizwicki MT, Norman AW. The vitamin D sterol-vitamin D receptor ensemble model offers unique insights into both genomic and rapid-response signaling. Science Signaling. 2009;2(75):p. re4.
57. Zhang Z, Kovalenko P, Cui M, Desmet M, Clinton SK, Fleet JC. Constitutive activation of the mitogen-activated protein kinase pathway impairs vitamin D signaling in human prostate epithelial cells. Journal of Cellular Physiology. 2010;224(2):433–442. [PMC free article] [PubMed]
58. Koszewski NJ, Herberth J, Malluche HH. Retinoic acid receptor gamma 2 interactions with vitamin D response elements. The Journal of Steroid Biochemistry and Molecular Biology. 2010;120(4-5):200–207. [PubMed]
59. Murthy S, Agoulnik IU, Weigel NL. Androgen receptor signaling and vitamin D receptor action in prostate cancer cells. Prostate. 2005;64(4):362–372. [PubMed]
60. Mordan-McCombs S, Brown T, Wang WL, Gaupel AC, Welsh J, Tenniswood M. Tumor progression in the LPB-Tag transgenic model of prostate cancer is altered by vitamin D receptor and serum testosterone status. The Journal of Steroid Biochemistry and Molecular Biology. 2010;121(1-2):368–371. [PubMed]
61. Zanello LP, Norman AW. Rapid modulation of osteoblast ion channel responses by 1alpha,25(OH)2-vitamin D3 requires the presence of a functional vitamin D nuclear receptor. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(6):1589–1594. [PMC free article] [PubMed]
62. Crescioli C, Maggi M, Vannelli GB, et al. Effect of a vitamin D3 analogue on keratinocyte growth factor-induced cell proliferation in benign prostate hyperplasia. Journal of Clinical Endocrinology and Metabolism. 2000;85(7):2576–2583. [PubMed]
63. Hedlund TE, Moffatt KA, Miller GJ. Stable expression of the nuclear vitamin D receptor in the human prostatic carcinoma cell line JCA-1: evidence that the antiproliferative effects of 1 alpha, 25-dihydroxyvitamin D3 are mediated exclusively through the genomic signaling pathway. Endocrinology. 1996;137(5):1554–1561. [PubMed]
64. Pedrozo HA, Schwartz Z, Rimes S, et al. Physiological importance of the 1,25(OH)2D3membrane receptor and evidence for a membrane receptor specific for 24,25(OH)2D3. Journal of Bone and Mineral Research. 1999;14(6):856–867. [PubMed]
65. Nemere I, Farach-Carson MC, Rohe B, et al. Ribozyme knockdown functionally links a 1,25(OH)2D3 membrane binding protein (1,25D3-MARRS) and phosphate uptake in intestinal cells. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(19):7392–7397. [PMC free article] [PubMed]
66. Karlsson S, Olausson J, Lundh D, et al. Vitamin D and prostate cancer: the role of membrane initiated signaling pathways in prostate cancer progression. The Journal of Steroid Biochemistry and Molecular Biology. 2010;121(1-2):413–416. [PubMed]
67. Krishnan AV, Feldman D. Molecular pathways mediating the anti-inflammatory effects of calcitriol: implications for prostate cancer chemoprevention and treatment. Endocrine-Related Cancer. 2010;17(1):R19–R38. [PubMed]
68. Gocek E, Studzinski GP. Vitamin D and differentiation in cancer signaling differentiation. Critical Reviews in Clinical Laboratory Sciences. 2009;46(4):190–209. [PMC free article] [PubMed]
69. Rohan JN, Weigel NL. 1α,25-dihydroxyvitamin D3 reduces c-Myc expression, inhibiting proliferation and causing G1 accumulation in C4-2 prostate cancer cells. Endocrinology. 2009;150(5):2046–2054. [PMC free article] [PubMed]
70. Toropainen S, Väisänen S, Heikkinen S, Carlberg C. The down-regulation of the human MYC gene by the nuclear hormone 1alpha,25-dihydroxyvitamin D3 is associated with cycling of corepressors and histone deacetylases. Journal of Molecular Biology. 2010;400(3):284–294. [PubMed]
71. Ikeda N, Uemura H, Ishiguro H, et al. Combination treatment with 1alpha,25-dihydroxyvitamin D3 and 9-cis-retinoic acid directly inhibits human telomerase reverse transcriptase transcription in prostate cancer cells. Molecular Cancer Therapeutics. 2003;2(8):739–746. [PubMed]
72. Blutt SE, Mcdonnell TJ, Polek TC, Weigel NL. Calcitriol-induced apoptosis in LNCaP cells is blocked by overexpression of Bcl-2. Endocrinology. 2000;141(1):10–17. [PubMed]
73. Sung V, Feldman D. 1,25-Dihydroxyvitamin D3 decreases human prostate cancer cell adhesion and migration. Molecular and Cellular Endocrinology. 2000;164(1-2):133–143. [PubMed]
74. Yang ES, Burnstein KL. Vitamin D inhibits G1 to S progression in LNCaP prostate cancer cells through p27Kip1 stabilization and Cdk2 mislocalization to the cytoplasm. Journal of Biological Chemistry. 2003;278(47):46862–46868. [PubMed]
75. Jensen SS, Madsen MW, Lukas J, Binderup L, Bartek J. Inhibitory effects of 1α,25-dihydroxyvitamin D3 on the G1-S phase-controlling machinery. Molecular Endocrinology. 2001;15(8):1370–1380. [PubMed]
76. Liu M, Lee MH, Cohen M, Bommakanti M, Freedman LP. Transcriptional activation of the Cdk inhibitor p21 by vitamin D3 leads to the induced differentiation of the myelomonocytic cell line U937. Genes and Development. 1996;10(2):142–153. [PubMed]
77. Blutt SE, Allegretto EA, Pike JW, Weigel NL. 1,25-dihydroxyvitamin D3and 9-cis-acid act synergistically to inhibit the growth of LNCaP prostate cells and cause accumulation of cells in G1. Endocrinology. 1997;138(4):1491–1497. [PubMed]
78. Campbell MJ, Elstner E, Holden S, Uskokovic M, Koeffler HP. Inhibition of proliferation of prostate cancer cells by a 19-nor-hexafluoride vitamin D3 analogue involves the induction of p21waf1, p27kip1 and E-cadherin. Journal of Molecular Endocrinology. 1997;19(1):15–27. [PubMed]
79. Flores O, Burnstein KL. GADD45gamma: a new vitamin D-regulated gene that is antiproliferative in prostate cancer cells. Endocrinology. 2010;151(10):4654–4664. [PMC free article] [PubMed]
80. Bao BY, Yeh SD, Lee YF. 1alpha,25-dihydroxyvitamin D3 inhibits prostate cancer cell invasion via modulation of selective proteases. Carcinogenesis. 2006;27(1):32–42. [PubMed]
81. Culig Z, Steiner H, Bartsch G, Hobisch A. Interleukin-6 regulation of prostate cancer cell growth. Journal of Cellular Biochemistry. 2005;95(3):497–505. [PubMed]
82. Wegiel B, Bjartell A, Culig Z, Persson JL. Interleukin-6 activates PI3K/Akt pathway and regulates cyclin A1 to promote prostate cancer cell survival. International Journal of Cancer. 2008;122(7):1521–1529.
83. Araki S, Omori Y, Lyn D, et al. Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer. Cancer Research. 2007;67(14):6854–6862. [PubMed]
84. Vasto S, Carruba G, Candore G, Italiano E, Di Bona D, Caruso C. Inflammation and prostate cancer. Future Oncology. 2008;4(5):637–645. [PubMed]
85. Mantell DJ, Owens PE, Bundred NJ, Mawer EB, Canfield AE. 1α,25-dihydroxyvitamin D3 inhibits angiogenesis in vitro and in vivo. Circulation Research. 2000;87(3):214–220. [PubMed]
86. Nonn L, Peng L, Feldman D, Peehl DM. Inhibition of p38 by vitamin D reduces interleukin-6 production in normal prostate cells via mitogen-activated protein kinase phosphatase 5: implications for prostate cancer prevention by vitamin D. Cancer Research. 2006;66(8):4516–4524. [PubMed]
87. Bao BY, Yao J, Lee YF. 1alpha, 25-dihydroxyvitamin D3 suppresses interleukin-8-mediated prostate cancer cell angiogenesis. Carcinogenesis. 2006;27(9):1883–1893. [PubMed]
88. Xu Y, Fang F, Clair DK, et al. Suppression of RelB-mediated manganese superoxide dismutase expression reveals a primary mechanism for radiosensitization effect of 1alpha,25-dihydroxyvitamin D3 in prostate cancer cells. Molecular Cancer Therapeutics. 2007;6(7):2048–2056. [PMC free article] [PubMed]
89. Ben-Shoshan M, Amir S, Dang DT, Dang LH, Weisman Y, Mabjeesh NJ. 1α,25-dihydroxyvitamin D3 (Calcitriol) inhibits hypoxia-inducible factor-1/vascular endothelial growth factor pathway in human cancer cells. Molecular Cancer Therapeutics. 2007;6(4):1433–1439. [PubMed]
90. Chung I, Han G, Seshadri M, et al. Role of vitamin D receptor in the antiproliferative effects of calcitriol in tumor-derived endothelial cells and tumor angiogenesis in vivo. Cancer Research. 2009;69(3):967–975. [PMC free article] [PubMed]
91. Moreno J, Krishnan AV, Swami S, Nonn L, Peehl DM, Feldman D. Regulation of prostaglandin metabolism by calcitriol attenuates growth stimulation in prostate cancer cell. Cancer Research. 2005;65(17):7917–7925. [PubMed]
92. Krishnan AV, Moreno J, Nonn L, et al. Novel pathways that contribute to the anti-proliferative and chemopreventive activities of calcitriol in prostate cancer. The Journal of Steroid Biochemistry and Molecular Biology. 2007;103(3–5):694–702. [PubMed]
93. Krishnan AV, Srinivas S, Feldman D, et al. Inhibition of prostaglandin synthesis and actions contributes to the beneficial effects of calcitriol in prostate cancer. Dermato-Endocrinology. 2009;1(1):7–11. [PMC free article] [PubMed]
94. Rhee Hvander, Coebergh JW, Vries ED. Sunlight, vitamin D and the prevention of cancer: a systematic review of epidemiological studies. European Journal of Cancer Prevention. 2009;18:458–475.
95. John EM, Koo J, Schwartz GG. Sun exposure and prostate cancer risk: evidence for a protective effect of early-life exposure. Cancer Epidemiology Biomarkers and Prevention. 2007;16(6):1283–1286.
96. Hanchette CL, Schwartz GG. Geographic patterns of prostate cancer mortality. evidence for a protective effect of ultraviolet radiation. Cancer. 1992;70(12):2861–2869. [PubMed]
97. Schwartz GG, Hanchette CL. UV, latitude, and spatial trends in prostate cancer mortality: all sunlight is not the same (United States) Cancer Causes and Control. 2006;17(8):1091–1101. [PubMed]
98. Grant WB. An estimate of premature cancer mortality in the U.S. due to inadequate doses of solar ultraviolet-B radiation. Cancer. 2002;94(6):1867–1875. [PubMed]
99. Luscombe CJ, Fryer AA, French ME, et al. Exposure to ultraviolet radiation: association with susceptibility and age at presentation with prostate cancer. The Lancet. 2001;358(9282):641–642.
100. John EM, Dreon DM, Koo J, et al. Residential sunlight exposure is associated with a decreased risk of prostate cancer. Journal of Steroid Biochemistry & Molecular Biology. 2004;89-90(1–5):549–552. [PubMed]
101. Bodiwala D, Luscombe CJ, French ME, et al. Susceptibility to prostate cancer: studies on interactions between UVR exposure and skin type. Carcinogenesis. 2003;24(4):711–717. [PubMed]
102. Lagunova Z, Porojnicu AC, Dahlback A, Berg JP, Beer TM, Moan J. Prostate cancer survival is dependent on season of diagnosis. Prostate. 2007;67(12):1362–1370. [PubMed]
103. Robsahm TE, Tretli S, Dahlback A, Moan J. Vitamin D3 from sunlight may improve the prognosis of breast-, colon- and prostate cancer (Norway) Cancer Causes and Control. 2004;15(2):149–158. [PubMed]
104. Colli JL, Colli A. International comparisons of prostate cancer mortality rates with dietary practices and sunlight levels. Urologic Oncology. 2006;24(3):184–194. [PubMed]
105. Ben-Shlomo Y, Evans S, Ibrahim F, et al. The risk of prostate cancer amongst black men in the United Kingdom: the process cohort study. European Urology. 2008;53(1):99–105. [PubMed]
106. Bodiwala D, Luscombe CJ, French ME, et al. Polymorphisms in the vitamin D receptor gene, ultraviolet radiation, and susceptibility to prostate cancer. Environmental and Molecular Mutagenesis. 2004;43(2):121–127. [PubMed]
107. Moon S, Holley S, Bodiwala D, et al. Associations between G/A1229, A/G3944, T/C30875, C/T48200 and C/T65013 genotypes and haplotypes in the vitamin D receptor gene, ultraviolet radiation and susceptibility to prostate cancer. Annals of Human Genetics. 2006;70:226–236. [PubMed]
108. Luscombe CJ, French ME, Liu S, et al. Outcome in prostate cancer associations with skin type and polymorphism in pigmentation-related genes. Carcinogenesis. 2001;22(9):1343–1347. [PubMed]
109. Rukin N, Blagojevic M, Luscombe CJ, et al. Associations between timing of exposure to ultraviolet radiation, T-stage and survival in prostate cancer. Cancer Detection and Prevention Journal. 2007;31(6):443–449.
110. Colli JL, Grant WB. Solar ultraviolet B radiation compared with prostate cancer incidence and mortality rates in United States. Urology. 2008;71(3):531–535. [PubMed]
111. de Vries E, Soerjomataram I, Houterman S, et al. Decreased risk of prostate cancer after skin cancer diagnosis: a protective role of ultraviolet radiation? American Journal of Epidemiology. 2007;165(8):966–972. [PubMed]
112. Rukin NJ, Zeegers MP, Ramachandran S, et al. A comparison of sunlight exposure in men with prostate cancer and basal cell carcinoma. British Journal of Cancer. 2007;96(3):523–528. [PMC free article] [PubMed]
113. Tuohimaa P, Pukkala E, Scélo G, et al. Does solar exposure, as indicated by the non-melanoma skin cancers, protect from solid cancers: vitamin D as a possible explanation. European Journal of Cancer. 2007;43(11):1701–1712. [PubMed]
114. Levi F, Randimbison L, Te VC, Conconi MM, La Vecchia C. Risk of prostate, breast and colorectal cancer after skin cancer diagnosis. International Journal of Cancer. 2008;123(12):2899–2901.
115. Gilbert R, Metcalfe C, Oliver SE, et al. Life course sun exposure and risk of prostate cancer: population-based nested case-control study and meta-analysis. International Journal of Cancer. 2009;125(6):1414–1423.
116. Grant WB. Geographic variation of prostate cancer mortality rates in the United States: implications for prostate cancer risk related to vitamin D. International Journal of Cancer. 2004;111(3):470–471.
117. Waltz P, Chodick G. International comparisons of prostate cancer mortality rates with dietary practices and sunlight levels. Urologic Oncology. 2007;25(1):p. 85.
118. Waltz P, Chodick G. Assessment of ecological regression in the study of colon, breast, ovary, non-Hodgkin’s lymphoma, or prostate cancer and residential UV. European Journal of Cancer Prevention. 2008;17(3):279–286. [PubMed]
119. Corder EH, Guess HA, Hulka BS, et al. Vitamin D and prostate cancer: a prediagnostic study with stored sera. Cancer Epidemiology Biomarkers and Prevention. 1993;2(5):467–472.
120. Ahonen MH, Tenkanen L, Teppo L, Hakama M, Tuohimaa P. Prostate cancer risk and prediagnostic serum 25-hydroxyvitamin D levels (Finland) Cancer Causes and Control. 2000;11(9):847–852. [PubMed]
121. Li H, Stampfer MJ, Hollis JB, et al. A prospective study of plasma vitamin D metabolites, vitamin D receptor polymorphisms, and prostate cancer. PLoS Medicine. 2007;4(3, article e103)
122. Tretli S, Hernes E, Berg JP, Hestvik UE, Robsahm TE. Association between serum 25(OH)D and death from prostate cancer. British Journal of Cancer. 2009;100(3):450–454. [PMC free article] [PubMed]
123. Braun MM, Helzlsouer KJ, Hollis BW, Comstock GW. Prostate cancer and prediagnostic levels of serum vitamin D metabolites (Maryland, United States) Cancer Causes and Control. 1995;6(3):235–239. [PubMed]
124. Gann PH, Ma J, Hennekens CH, et al. Circulating vitamin D metabolites in relation to subsequent development of prostate cancer. Cancer Epidemiology, Biomarkers & Prevention. 1996;5(2):121–126.
125. Nomura AM, Stemmermann GN, Lee J, et al. Serum vitamin D metabolite levels and the subsequent development of prostate cancer (Hawaii, United States) Cancer Causes and Control. 1998;9(4):425–432. [PubMed]
126. Platz EA, Leitzmann MF, Hollis BW, Willett WC, Giovannucci E. Plasma 1,25-dihydroxy- and 25-hydroxyvitamin D and subsequent risk of prostate cancer. Cancer Causes and Control. 2004;15(3):255–265. [PubMed]
127. Gandini S, Boniol M, Haukka J, et al. Meta-analysis of observational studies of serum 25-hydroxyvitamin D levels and colorectal, breast and prostate cancer and colorectal adenoma. International Journal of Cancer. 2011;128(6):1414–1424.
128. Barnett CM, Nielson CM, Shannon J, et al. Serum 25-OH vitamin D levels and risk of developing prostate cancer in older men. Cancer Causes and Control. 2010;21(8):1297–1303. [PMC free article] [PubMed]
129. Travis RC, Crowe FL, Allen NE, et al. Serum vitamin D and risk of prostate cancer in a case-control analysis nested within the European prospective investigation into cancer and nutrition (EPIC) American Journal of Epidemiology. 2009;169(10):1223–1232. [PMC free article] [PubMed]
130. Ahn J, Peters U, Albanes D, et al. Serum vitamin D concentration and prostate cancer risk: a nested case-control study. Journal of the National Cancer Institute . 2008;100(11):796–804. [PubMed]
131. Tuohimaa P, Tenkanen L, Ahonen M, et al. Both high and low levels of blood vitamin D are associated with a higher prostate cancer risk: a longitudinal, nested case-control study in the nordic countries. International Journal of Cancer. 2004;108(1):104–108.
132. Mikhak B, Hunter DJ, Spiegelman D, Platz EA, Hollis BW, Giovannucci E. Vitamin D receptor (VDR) gene polymorphisms and haplotypes, interactions with plasma 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D, and prostate cancer risk. Prostate. 2007;67(9):911–923. [PubMed]
133. Ma J, Stampfer MJ, Gann PH, et al. Vitamin D receptor polymorphisms, circulating vitamin D metabolites, and risk of prostate cancer in United States physicians. Cancer Epidemiology Biomarkers and Prevention. 1998;7(5):385–390.
134. Park SY, Murphy SP, Wilkens LR, Stram DO, Henderson BE, Kolonel LN. Calcium, vitamin D, and dairy product intake and prostate cancer risk: the multiethnic cohort study. American Journal of Epidemiology. 2007;166(11):1259–1269. [PubMed]
135. Kristal AR, Arnold KB, Neuhouser ML, et al. Diet, supplement use, and prostate cancer risk: results from the prostate cancer prevention trial. American Journal of Epidemiology. 2010;172(5):566–577. [PMC free article] [PubMed]
136. Huncharek M, Muscat J, Kupelnick B. Dairy products, dietary calcium and vitamin D intake as risk factors for prostate cancer: a meta-analysis of 26,769 cases from 45 observational studies. Nutrition and Cancer. 2008;60(4):421–441. [PubMed]
137. Uitterlinden AG, Fang Y, Van Meurs JB, Pols HAP, Van Leeuwen JPTM. Genetics and biology of vitamin D receptor polymorphisms. Gene. 2004;338(2):143–156. [PubMed]
138. Gennari L, De Paola V, Merlotti D, Martini G, Nuti R. Steroid hormone receptor gene polymorphisms and osteoporosis: a pharmacogenomic review. Expert Opinion on Pharmacotherapy. 2007;8(5):537–553. [PubMed]
139. Whitfield GK, Remus LS, Jurutka PW, et al. Functionally relevant polymorphisms in the human nuclear vitamin D receptor gene. Molecular and Cellular Endocrinology. 2001;177(1-2):145–159. [PubMed]
140. Orton SM, Morris AP, Herrera BM, et al. Evidence for genetic regulation of vitamin D status in twins with multiple sclerosis. American Journal of Clinical Nutrition. 2008;88(2):441–447. [PMC free article] [PubMed]
141. Arai H, Miyamoto KI, Yoshida M, et al. The polymorphism in the caudal-related homeodomain protein Cdx-2 binding element in the human vitamin D receptor gene. Journal of Bone and Mineral Research. 2001;16(7):1256–1264. [PubMed]
142. Durrin LK, Haile RW, Ingles SA, Coetzee GA. Vitamin D receptor 3’-untranslated region polymorphisms: lack of effect on mRNA stability. Biochimica et Biophysica Acta. 1999;1453(3):311–320. [PubMed]
143. Carling T, Rastad J, Akerström G, Westin G. Vitamin D receptor (VDR) and parathyroid hormone messenger ribonucleic acid levels correspond to polymorphic VDR alleles in human parathyroid tumors. Journal of Clinical Endocrinology and Metabolism. 1998;83(7):2255–2259. [PubMed]
144. Verbeek W, Gombart AF, Shiohara M, et al. Vitamin D receptor: no evidence for allele-specific mRNA stability in cells which are heterozygous for the Taq I restriction enzyme polymorphism. Biochemical and Biophysical Research Communications. 1997;238(1):77–80. [PubMed]
145. Morrison NA, Qi JC, Tokita A, et al. Prediction of bone density from vitamin D receptor alleles. Nature. 1994;387(6628):p. 106.
146. Taylor JA, Hirvonen A, Watson M, Pittman G, Mohler JL, Bell DA. Association of prostate cancer with vitamin D receptor gene polymorphism. Cancer Research. 1996;56(18):4108–4110. [PubMed]
147. Ingles SA, Ross RK, Yu MC, et al. Association of prostate cancer risk with genetic polymorphisms in vitamin D receptor and androgen receptor. Journal of the National Cancer Institute. 1997;89(2):166–170. [PubMed]
148. Habuchi T, Suzuki T, Sasaki R, et al. Association of vitamin D receptor gene polymorphism with prostate cancer and benign prostatic hyperplasia in a Japanese population. Cancer Research. 2000;60(2):305–308. [PubMed]
149. Suzuki K, Matsui H, Ohtake N, et al. Vitamin D receptor gene polymorphism in familial prostate cancer in a Japanese population. International Journal of Urology. 2003;10(5):261–266. [PubMed]
150. Ntais C, Polycarpou A, Ioannidis JP. Vitamin D receptor gene polymorphisms and risk of prostate cancer: a meta-analysis. Cancer Epidemiology Biomarkers and Prevention. 2003;12(12):1395–1402.
151. Oakley-Girvan I, Feldman D, Eccleshall TR, et al. Risk of early-onset prostate cancer in relation to germ line polymorphisms of the vitamin D receptor. Cancer Epidemiology Biomarkers and Prevention. 2004;13(8):1325–1330.
152. Maistro S, Snitcovsky I, Sarkis AS, da Silva IA, Brentani MM. Vitamin D receptor polymorphisms and prostate cancer risk in Brazilian men. International Journal of Biological Markers. 2004;19(3):245–249. [PubMed]
153. Cheteri MB, Stanford JL, Friedrichsen DM, et al. Vitamin D receptor gene polymorphisms and prostate cancer risk. Prostate. 2004;59(4):409–418. [PubMed]
154. Huang SP, Chou YH, Wayne Chang WS, et al. Association between vitamin D receptor polymorphisms and prostate cancer risk in a Taiwanese population. Cancer Letters. 2004;207(1):69–77. [PubMed]
155. Williams H, Powell IJ, Land SJ, et al. Vitamin D receptor gene polymorphisms and disease free survival after radical prostatectomy. Prostate. 2004;61(3):267–275. [PubMed]
156. John EM, Schwartz GG, Koo J, et al. Sun exposure, vitamin D receptor gene polymorphisms, and risk of advanced prostate cancer. Cancer Research. 2005;65(12):5470–5479. [PubMed]
157. Mishra DK, Bid HK, Srivastava DS, Mandhani A, Mittal RD. Association of vitamin D receptor gene polymorphism and risk of prostate cancer in India. Urologia Internationalis. 2005;74(4):315–318. [PubMed]
158. Hayes VM, Severi G, Padilla EJ, et al. Genetic variants in the vitamin D receptor gene and prostate cancer risk. Cancer Epidemiology Biomarkers and Prevention. 2005;14(4):997–999.
159. Kidd LC, Paltoo DN, Wang S, et al. Sequence variation within the 5’ regulatory regions of the vitamin D binding protein and receptor genes and prostate cancer risk. Prostate. 2005;64(3):272–282. [PubMed]
160. Berndt SI, Dodson JL, Huang WY, Nicodemus KK. Systematic review of vitamin D receptor gene polymorphisms and prostate cancer risk. Journal of Urology. 2006;175(5):1613–1623. [PubMed]
161. Holick CN, Stanford JL, Kwon EM, et al. Comprehensive association analysis of the vitamin D pathway genes, VDR, CYP27B1, and CYP24A1, in prostate cancer. Cancer Epidemiology, Biomarkers & Prevention. 2007;16(10):1990–1999.
162. Li H, Stampfer MJ, Hollis JB, et al. A prospective study of plasma vitamin D metabolites, vitamin D receptor polymorphisms, and prostate cancer. PLoS Medicine. 2007;4(3, article e103)
163. Rukin NJ, Luscombe C, Moon S, et al. Prostate cancer susceptibility is mediated by interactions between exposure to ultraviolet radiation and polymorphisms in the 5’ haplotype block of the vitamin D receptor gene. Cancer Letters. 2007;247(2):328–335. [PubMed]
164. Onen IH, Ekmekci A, Eroglu M, et al. Association of genetic polymorphisms in vitamin D receptor gene and susceptibility to sporadic prostate cance. Experimental Biology and Medicine. 2008;233(12):1608–1614. [PubMed]
165. Torkko KC, van Bokhoven A, Mai P, et al. VDR and SRD5A2 polymorphisms combine to increase risk for prostate cancer in both non-hispanic white and hispanic white men. Clinical Cancer Research. 2008;14(10):3223–3229. [PubMed]
166. Ahn J, Albanes D, Berndt SI, et al. Vitamin D-related genes, serum vitamin D concentrations and prostate cancer risk. Carcinogenesis. 2009;30(5):769–776. [PMC free article] [PubMed]
167. Chen L, Smith GD, Evans DM, et al. Genetic variants in the vitamin d receptor are associated with advanced prostate cancer at diagnosis: findings from the prostate testing for cancer and treatment study and a systematic review. Cancer Epidemiology, Biomarkers & Prevention. 2009;18(11):2874–2881.
168. Raimondi S, Johansson H, Maisonneuve P, Gandini S. Review and meta-analysis on vitamin D receptor polymorphisms and cancer risk. Carcinogenesis. 2009;30(7):1170–1180. [PubMed]
169. Yin M, Wei S, Wei Q. Vitamin D receptor genetic polymorphisms and prostate cancer risk: a meta-analysis of 36 published studies. International Journal of Clinical and Experimental Medicine. 2009;2(2):159–175. [PMC free article] [PubMed]
170. Holt SK, Kwon EM, Peters U, Ostrander EA, Stanford JL. Vitamin D pathway gene variants and prostate cancer risk. Cancer Epidemiology Biomarkers and Prevention. 2009;18(6):1929–1933.
171. Bai Y, Yu Y, Yu B, et al. Association of vitamin D receptor polymorphisms with the risk of prostate cancer in the Han population of Southern China. BMC Medical Genetics. 2009;10, article 125
172. Risio M, Venesio T, Kolomoets E, et al. Genetic polymorphisms of CYP17A1, vitamin D receptor and androgen receptor in Italian heredo-familial and sporadic prostate cancers. Cancer Epidemiology. In press.

Plaats een reactie ...

Reageer op "Vitamine D maar ook het seizoen van diagnose hebben grote invloed op bepaalde vormen van kanker, prostaatkanker - borstkanker . en ziekteverloop"


Gerelateerde artikelen
 

Gerelateerde artikelen

Vitamine D - calcifediol is >> Vitamine D-suppletie met of >> Vitamine D: Kostenpost of >> Vitamine D-suppletie reguleert >> Hoge dosis vitamine D toediening >> Hoge vitamine D waarden - >> Vitamine D tekort bij ouderen >> Zonlicht - (vitamine D) beschermt >> Vitamine D tekort bij kinderen >> Vitamine D3 plus calcium aanvullend >>