Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-18T17:20:42.372Z Has data issue: false hasContentIssue false

Vitamin D and colorectal cancer: molecular, epidemiological and clinical evidence

Published online by Cambridge University Press:  09 March 2016

Ruoxu Dou
Affiliation:
Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangdong 510655, People’s Republic of China
Kimmie Ng
Affiliation:
Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
Edward L. Giovannucci
Affiliation:
Department of Nutrition, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
JoAnn E. Manson
Affiliation:
Department of Nutrition, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02115, USA
Zhi Rong Qian
Affiliation:
Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
Shuji Ogino*
Affiliation:
Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
*
*Corresponding author: S. Ogino, fax +1 617 582 8558, email shuji_ogino@dfci.harvard.edu
Rights & Permissions [Opens in a new window]

Abstract

In many cells throughout the body, vitamin D is converted into its active form calcitriol and binds to the vitamin D receptor (VDR), which functions as a transcription factor to regulate various biological processes including cellular differentiation and immune response. Vitamin D-metabolising enzymes (including CYP24A1 and CYP27B1) and VDR play major roles in exerting and regulating the effects of vitamin D. Preclinical and epidemiological studies have provided evidence for anti-cancer effects of vitamin D (particularly against colorectal cancer), although clinical trials have yet to prove its benefit. In addition, molecular pathological epidemiology research can provide insights into the interaction of vitamin D with tumour molecular and immunity status. Other future research directions include genome-wide research on VDR transcriptional targets, gene–environment interaction analyses and clinical trials on vitamin D efficacy in colorectal cancer patients. In this study, we review the literature on vitamin D and colorectal cancer from both mechanistic and population studies and discuss the links and controversies within and between the two parts of evidence.

Type
Full Papers
Copyright
Copyright © The Authors 2016 

Although a well-recognised physiological role of vitamin D is the regulation of Ca and phosphate metabolism( Reference Holick 1 ), recent studies suggest a much broader range of biological functions of vitamin D, including potential anti-neoplastic effects. Garland & Garland( Reference Garland and Garland 2 ) discovered in 1980 that colon cancer mortality rates in the USA were highest in places where populations were exposed to the least amount of sunlight, and proposed that vitamin D might be a protective factor against colon cancer. Since then, extensive studies have reported anti-neoplastic actions of vitamin D, particularly in colorectal cancer( Reference Deeb, Trump and Johnson 3 , Reference Feldman, Krishnan and Swami 4 ). If adequate vitamin D does have a protective effect, ensuring that people have sufficient vitamin D can be an effective way to reduce cancer incidence and mortality( Reference Feldman, Krishnan and Swami 4 ). In this review, we discuss relevant basic science and preclinical studies, which examined the mechanisms including the regulation of proliferation, differentiation, apoptosis, angiogenesis and immunity. We also discuss epidemiological and human intervention studies and address possible reasons why evidence for an effect of vitamin D supplementation remains inconclusive. In addition, we remark on molecular pathological epidemiology (MPE)( Reference Ogino, Chan and Fuchs 5 , Reference Ogino and Stampfer 6 ), which can bridge the gap between basic science and human population studies of vitamin D and colorectal cancer.

We conducted a literature research in the Web of Science database under the topics ‘Vitamin D’ AND ‘Colorectal Neoplasms’, and in the PubMed database using the MeSH terms ‘Vitamin D’ AND ‘Colorectal Neoplasms’, for papers published in English from January 1995 to November 2015. We manually searched for references cited in the chosen articles and in published reviews.

Source and metabolism of vitamin D

Vitamin D belongs to a group of steroids known as secosteroids. In humans, the most common forms of vitamin D are vitamin D3 (cholecalciferol) and vitamin D2 (ergocalciferol); both can be obtained from the diet and from diet supplements. Vitamin D3 can also be synthesised in adequate amounts in the skin, under exposure to sunlight( Reference Holick 7 ). As vitamin D can be produced in the human body, strictly speaking it is not a vitamin per se, but rather is the precursor to the potent steroid hormone calcitriol (also known as 1,25-dihydroxyvitamin D3).

Vitamin D from the skin and diet is activated to calcitriol by two cytochrome P450-mediated hydroxylation steps. The first step takes place mostly in the liver, where the enzyme vitamin D-25-hydroxylase (predominantly CYP2R1, cytochrome P450 family 2 subfamily R member 1) catalyses the first hydroxylation of vitamin D at C25. This reaction yields 25-hydroxyvitamin D (25(OH)D) – the circulating form with a half-life of 2 weeks – which is used to determine an individual’s vitamin D status( Reference Holick 7 , Reference Ponchon and DeLuca 8 ). In the second step, 25(OH)D is metabolised by the enzyme 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1, cytochrome P450 family 27 subfamily B member 1) in the kidneys and certain extrarenal sites to yield the active form calcitriol( Reference Fraser and Kodicek 9 ). Calcitriol then performs its biological functions, inhibits CYP27B1 activity( Reference Young, Schwartz and Wang 10 ) and induces expression of the enzyme 25-hydroxyvitamin D-24-hydroxylase (CYP24A1, cytochrome P450 family 24 subfamily A member 1), which catabolises 25(OH)D and calcitriol into biologically inactive forms (Fig. 1)( Reference Pike and Meyer 11 ).

Fig. 1 The metabolism of vitamin D in human body. Vitamin D that is taken up via the diet, or synthesised from 7-dehydrocholesterol by the skin following UV exposure, binds to vitamin D-binding protein (DBP) in the circulation and is transported to the liver. Vitamin D is hydroxylated at C25 by CYP2R1 in the liver to 25-hydroxyvitamin D (25(OH)D), the major circulating form of vitamin D in the human body. In the kidney and some extrarenal sites, 25(OH)D is further hydroxylated at C1 by CYP27B1 into 1,25-dihydroxyvitamin D3 (1,25(OH)2D) (calcitriol), the bioactive form. Both 25(OH)D and 1,25(OH)2D are deactivated by CYP24A1 through additional hydroxylation at C24. Both CYP27B1 and CYP24A1 are regulated by calcitriol.

Mechanism of calcitriol action

Calcitriol exerts its biological effects by binding and activating the nuclear vitamin D receptor (VDR) and regulating gene expression( Reference Deeb, Trump and Johnson 3 , Reference Christakos, Dhawan and Verstuyf 12 ). The binding of calcitriol induces a conformational change in VDR that allows the receptor to dimerise with the retinoid X receptor (RXR); this heterodimer specifically docks on vitamin D response elements (VDRE) in the promoter regions of target genes( Reference Carlberg, Bendik and Wyss 13 ). The conformational change of VDR also recruits the co-activator and detaches the co-repressor to acetylate nucleosome histones and unravel DNA, thus enabling transcription (Fig. 2(a))( Reference Tagami, Lutz and Kumar 14 ).

Fig. 2 The mechanism of calcitriol (1,25(OH)2D) action through vitamin D receptor (VDR). Calcitriol binds and activates nuclear VDR, which then dimerises with retinoid X receptor (RXR). (a) Transcriptional activation involves the VDR–RXR heterodimer binding with vitamin D response element (VDRE) and recruitment of histone acetyltransferase co-activator. (b) Transcriptional depression involves VDR–RXR binding with negative VDRE (nVDRE) and recruitment of histone deacetylase co-repressor. RNA POL II, RNA polymerase II.

Calcitriol-dependent repression of gene transcription is documented for the CYP27B1 ( Reference Murayama, Kim and Yanagisawa 15 ) and PTH (parathyroid hormone)( Reference Kim, Fujiki and Murayama 16 ) genes. Haussler et al. ( Reference Haussler, Whitfield and Kaneko 17 ) postulated that VDR-mediated repression initiates with the docking of liganded VDR–RXR on a negative VDRE in the promoter regions of target genes, which then conforms liganded VDR such that it binds the co-repressor rather than the co-activator (Fig. 2(b)).

In addition to its genomic actions that occur over a period of hours or days, calcitriol also rapidly initiates many biological responses( Reference Haussler, Jurutka and Mizwicki 18 ). For instance, calcitriol can bind with a plasma membrane VDR of the intestinal epithelial cells and cause the coupled opening of Ca2+ channels, resulting in the rapid hormonal stimulation of intestinal Ca transport (transcaltachia) within minutes( Reference Norman, Mizwicki and Norman 19 , Reference Ordonez-Moran, Larriba and Palmer 20 ). Furthermore, the binding of calcitriol with membrane VDR may engage in cross-talk with the classical VDR pathway to modulate gene expression, possibly through Ca2+ influx activation of the Ca2+ messenger system such as protein kinase C( Reference Deeb, Trump and Johnson 3 ).

Vitamin D metabolism in colorectal cancer

The response of cancer cells to calcitriol depends not only on VDR expression but also on the intracellular concentrations of calcitriol( Reference Hsu, Feldman and McNeal 21 , Reference Swami, Krishnan and Wang 22 ). Intracellular calcitriol concentrations are determined by the circulating concentrations of 25(OH)D and calcitriol, and by the activity of CYP27B1 and CYP24A1 within the cell. CYP27B1 and CYP24A1 were previously known as enzymes within the kidney, but are now also found in extrarenal sites including the colon( Reference Zehnder, Bland and Williams 23 , Reference Cross, Bises and Lechner 24 ). As described below, the levels of CYP27B1, CYP24A1 and VDR in colorectal cancer cells are studied in relation to differentiation and response to treatment.

CYP27B1

CYP27B1, as the synthesising enzyme of calcitriol, is normally expressed at low levels in the colon( Reference Bises, Kallay and Weiland 25 , Reference Tangpricha, Flanagan and Whitlatch 26 ). In well-differentiated and moderately differentiated colorectal cancer samples, expression of CYP27B1 is elevated, whereas in poorly differentiated colorectal cancer samples the expression is repressed( Reference Bises, Kallay and Weiland 25 Reference Bareis, Bises and Bischof 28 ). Ogunkolade et al. ( Reference Ogunkolade, Boucher and Fairclough 29 ) reported that CYP27B1 mRNA expression levels are similar in colorectal cancer samples and in healthy colon samples, but are decreased in adjacent normal colon mucosa, 10 cm from the tumour border; this finding suggests that CYP27B1 expression in adjacent colon cells is regulated by the tumour, or that low expression of CYP27B1 in the colon is a risk for carcinogenesis. Bareis et al. ( Reference Bareis, Kallay and Bischof 30 ) showed that the slowly dividing, highly differentiated colorectal cancer cell line Caco-2/15 responds in a dose-dependent manner to epidermal growth factor (EGF) or calcitriol by up-regulating the expressions of VDR and CYP27B1, whereas highly proliferative, less-differentiated cell lines (Caco-2/AQ, COGA-1A and COGA-1E) show a down-regulation of VDR and CYP27B1 after EGF or calcitriol treatment. Although definite in vivo evidence is lacking, local production of calcitriol in the colon has been indirectly suggested by human studies. The serum concentration of 25(OH)D, rather than that of calcitriol, was inversely associated with colonic epithelial cell proliferation in a chemoprevention study( Reference Holt, Arber and Halmos 31 ). Wagner et al. ( Reference Wagner, Dias and Schnabl 32 ) showed a positive correlation between serum and colon calcitriol concentrations (r 0·58, P=0·0008), with a positive colon calcitriol intercept (21·5 pmol/kg, P<0·001) at zero serum calcitriol, supporting the notion of synthesis of calcitriol within the colon. To summarise, elevated CYP27B1 expression suggests possible benefit from treatment with vitamin D, especially in well-differentiated and moderately differentiated tumours, whereas the relatively low expression of CYP27B1 in poorly differentiated colorectal cancer indicates a mechanism of resistance of the cancer cells to calcitriol actions.

CYP24A1

As the main enzyme determining the biological half-life of calcitriol, CYP24A1 is found in low levels in normal human colon mucosa and in colorectal adenomas, but in elevated levels in the majority of adenocarcinomas( Reference Horvath, Lakatos and Kosa 33 ). CYP24A1 mRNA expression is also increased in poorly differentiated and late-stage colorectal cancers, compared with well-differentiated, early-stage tumours( Reference Bareis, Bises and Bischof 28 ). Anderson et al. ( Reference Anderson, Nakane and Ruan 34 ) showed that CYP24A1 mRNA expression is not only significantly up-regulated in human HT29 cells but also profoundly stimulated by calcitriol treatment, abrogating the anti-proliferative effect of calcitriol. Kosa et al. ( Reference Kosa, Horvath and Wolfling 35 ) also observed that CYP24A1 mRNA is induced by calcitriol treatment in Caco-2 – a human colon adenocarcinoma cell line. Cell viability and proliferation are not influenced by calcitriol alone, but are markedly reduced when calcitriol is co-administered with KD-35 – a CYP24A1 inhibitor. Together, these findings suggest that CYP24A1 exhibits a potent negative-feedback effect, and that inhibition of CYP24A1 may be a good strategy for enhancing the anti-tumour effect of calcitriol.

Vitamin D receptor

As the major receptor to mediate the biological effects of calcitriol, VDR is present in most cells of the human body, and is especially abundant in intestinal epithelial cells( Reference Wang, Zhu and DeLuca 36 ). VDR expression is increased in adenoma and in well-differentiated or moderately differentiated colorectal cancer tissues, but is decreased in poorly differentiated tumours, and is negligible in metastatic lymph nodes( Reference Cross, Bareis and Hofer 27 , Reference Matusiak, Murillo and Carroll 37 ). Palmer et al. ( Reference Palmer, Larriba and Garcia 38 ) discovered that the transcription factors SNAI1 (snail family zinc finger 1) and SNAI2 (snail family zinc finger 2) repress VDR expression in SW480-ADH cells and block the anti-tumour action of the calcitriol analogue EB1089. RNA expressions of SNAI1 and SNAI2 are up-regulated in human colorectal cancers, and are inversely correlated with VDR mRNA expression( Reference Palmer, Larriba and Garcia 38 , Reference Larriba, Bonilla and Munoz 39 ). These findings suggest that high levels of SNAI1 and SNAI2 are a probable cause for VDR down-regulation and for vitamin D unresponsiveness in advanced colorectal cancer, and that vitamin D therapy may not be a good treatment choice for patients who overexpress SNAI1 and SNAI2.

Anti-cancer actions of vitamin D on colorectal cancer

The anti-cancer effects of calcitriol are mostly studied in vitro by binding to the VDR and causing transcriptional activation and repression of the target genes. Given the pivotal role of nuclear VDR as a transcriptional regulator, researchers have investigated the genome-wide targets of calcitriol-stimulated VDR in human cells by chromatin immunoprecipitation-sequencing (ChIP-Seq). In one such study profiling human lymphoblastoid cells, VDR-binding sites were significantly enriched near colorectal cancer-associated genes identified from genome-wide association studies( Reference Ramagopalan, Heger and Berlanga 40 ). Meyer et al. ( Reference Meyer, Goetsch and Pike 41 ) performed ChIP-Seq for VDR/RXR on the human colorectal cancer cell LS180, and identified FOS (FBJ murine osteosarcoma viral oncogene homolog) and MYC (v-myc avian myelocytomatosis viral oncogene homolog) among the target genes. In addition, several transcription factors regulated by calcitriol subsequently amplify and diversify the transcriptional output( Reference Goeman, De Nicola and D’Onorio De Meo 42 ). The most studied anti-cancer effects of calcitriol are listed below.

Proliferation

Previous studies have established VDR as a biomarker for vitamin D-mediated inhibition of human colon cancer cell growth( Reference Shabahang, Buras and Davoodi 43 ). The anti-proliferative effect of vitamin D on colorectal cancer involves multiple pathways. In Caco-2 cells, calcitriol and its analogues (F6-D3, ZK 156718 and BGP-13) increase the expressions of the cyclin-dependent kinase (CDK) inhibitors CDKN1A (cyclin-dependent kinase inhibitor 1A (p21, Cip1)) and CDKN1B (cyclin-dependent kinase inhibitor 1B (p27, Kip1)), which inhibit CDK2 and CDK6, leading to G1 phase arrest( Reference Scaglione-Sewell, Bissonnette and Skarosi 44 Reference Berkovich, Sintov and Ben-Shabat 46 ). Calcitriol also results in the activation of latent transforming growth factor-β1 (TGFB1) in Caco-2 cells, and sensitises these cells to the growth inhibitory effects of TGFB1( Reference Chen, Davis and Sitrin 47 ). Synthetic low-calcemic vitamin D analogues (EB1089 and CB1093) decrease proliferation of HT29 human cancer cells by inhibiting the secretion of insulin-like growth factor 2 (IGF2) and by inducing the insulin-like growth factor-binding protein-6, which sequesters IGF2 with high affinity( Reference Oh, Kim and Schaffer 48 ). Calcitriol also counteracts EGF-stimulated Caco-2 cell growth by markedly decreasing epidermal growth factor receptor expression( Reference Tong, Hofer and Ellinger 49 ).

Differentiation

Calcitriol has multiple pro-differentiation effects in colorectal cancer cells. The classic marker for differentiation is the expression of alkaline phosphatase, which is found along the brush border of the colon mucosa but is poorly expressed in proliferating colorectal cancer cells. Calcitriol and its analogues (ZK 156718 and EB1089) increase the activity of alkaline phosphatase in colorectal adenoma cell lines (RG/C2 and AA/C1) and colorectal cancer cells (Caco-2, PC/JW, HT29 and SW620)( Reference Gaschott, Steinmeyer and Steinhilber 45 , Reference Diaz, Paraskeva and Thomas 50 ). Chen et al. ( Reference Chen, Davis and Bissonnette 51 ) reported that calcitriol increases alkaline phosphatase activity in Caco-2 cells by stimulating activator protein-1 (FOS/JUN (jun proto-oncogene)) activation, which is accomplished via a protein kinase C α- and mitogen-activated protein kinase (MAPK)-dependent mechanism.

Apart from affecting the expression of alkaline phosphatase, calcitriol also induces the expression of E-cadherin (CDH1, cadherin 1) and other adhesion proteins, causing β-catenin (CTNNB1, catenin beta 1) to translocate from the nucleus to E-cadherin complexes at the plasma membrane in the human colon cancer SW480-ADH cell line( Reference Palmer, Gonzalez-Sancho and Espada 52 , Reference Palmer, Sanchez-Carbayo and Ordonez-Moran 53 ); similar effect on Cdh1 has been observed in an Apc min/+ (adenomatous polyposis coli) mouse model( Reference Xu, Posner and Stevenson 54 ). Meanwhile, ligand-activated VDR competes with the T cell-specific transcription factor 7-like 2 (TCF7L2) for CTNNB1 binding and represses downstream gene expression in SW480-ADH cells( Reference Palmer, Gonzalez-Sancho and Espada 52 ). Calcitriol–VDR also inhibits CTNNB1 activity in Caco-2 cells, and the inhibition is enhanced by wild-type APC( Reference Egan, Thompson and Vitanov 55 ). Finally, the WNT (wingless-type MMTV integration site family) antagonist DKK1 (dickkopf WNT signaling pathway inhibitor 1) is induced by calcitriol in association with E-cadherin in SW480-ADH cells( Reference Aguilera, Pena and Garcia 56 ). As a result, calcitriol and its analogues inhibit the WNT/CTNNB1 pathway and the activation of its target genes in colorectal cancer cells; this in turn contributes to the inhibition of cell proliferation and to the maintenance of the differentiated phenotype.

Apoptosis

Calcitriol induces apoptosis in colorectal adenoma and colorectal cancer by up-regulating the pro-apoptotic proteins BAK1 (BCL2-antagonist/killer 1) and BAX (BCL2-associated X protein) and by down-regulating the expressions of anti-apoptotic proteins BAG1, BIRC5 (baculoviral IAP repeat containing 5) and BCL2 (B-cell CLL/lymphoma 2). In two colorectal adenoma and three colorectal cancer cell lines, calcitriol and the vitamin D analogue EB1089 induced p53-independent apoptosis in a dose-dependent manner, and the levels of the pro-apoptotic protein BAK1 were consistently increased in all the cell lines examined( Reference Diaz, Paraskeva and Thomas 50 ). Barnes et al. ( Reference Barnes, Arhel and Lee 57 ) showed that EB1089 induces apoptosis in a colorectal adenoma S/RG/C2 cell line by re-distributing the anti-apoptotic protein BAG1 from the nucleus to the cytoplasm. Liu et al. ( Reference Liu, Hu and Chakrabarty 58 ) discovered that calcitriol suppresses the expression of BIRC5 (survivin) and promotes a cytotoxic response to 5-fluorouracil in human colon cancer cells (CBS, Moser, Caco-2 and HCT116) in a Ca-sensing receptor (CASR)-dependent manner, possibly by binding the VDRE in CASR promoters( Reference Canaff and Hendy 59 , Reference Chakrabarty, Wang and Canaff 60 ). In an Apc1638N/+ mouse model of intestinal cancer, a western-pattern diet decreased the expression of the pro-apoptotic protein BAX and increased the expression of the anti-apoptotic protein BCL2; treatment with vitamin D and Ca reverses these effects of the western-style diet and markedly inhibits tumour growth( Reference Yang, Lamprecht and Shinozaki 61 ). In a human colorectal cancer xenograft model in nude mice, treatment with the vitamin D analogues BGP-13 and BGP-15 activated cell apoptosis( Reference Berkovich, Sintov and Ben-Shabat 46 ). However, the pro-apoptotic effect of calcitriol appears to be not always true: Stambolsky et al. ( Reference Stambolsky, Tabach and Fontemaggi 62 ) reported that mutant TP53 (tumor protein p53) is recruited to VDR-regulated genes and converts calcitriol into an anti-apoptotic agent in SW480 cells. Thus, TP53 mutation status might be a predictive marker for vitamin D treatment response.

Angiogenesis

Calcitriol also inhibits angiogenesis. Mantell et al. ( Reference Mantell, Owens and Bundred 63 ) showed that calcitriol significantly inhibits the sprouting and elongation of vascular endothelial growth factor A (VEGFA)-induced endothelial cells in a dose-dependent manner. In human colorectal cancer SW480 cells, calcitriol treatment for 24 h at 0·1 and 1 µm decreases the expression of hypoxia-inducible factor-1α and at 1 µm inhibits the secretion of VEGFA under conditions of hypoxia( Reference Ben-Shoshan, Amir and Dang 64 ). However, Fernandez-Garcia et al. ( Reference Fernandez-Garcia, Palmer and Garcia 65 ) reported that calcitriol increases the levels of VEGFA and the anti-angiogenic factor thrombospondin 1, leading to a minimal balanced change in the angiogenic potential of SW480-ADH cells. Calcitriol also represses the expression of DKK4 (dickkopf WNT signaling pathway inhibitor 4) in SW480-ADH cells; DKK4 is induced by the TCF7L2/CTNNB1 pathway and enhances the migratory, invasive and pro-angiogenic potential of colorectal cancer( Reference Pendas-Franco, Garcia and Pena 66 ). In a rat model of colon tumourigenesis induced by azoxymethan, intraperitoneal administration of calcitriol significantly reduced the incidence of colon tumours and also decreased the level of VEGFA and microvessel counts in tumours, suggesting that anti-angiogenesis is a mechanism for the anti-tumourigenic effect of vitamin D( Reference Iseki, Tatsuta and Uehara 67 ).

Immune modulation

Calcitriol modulates innate and adaptive immunity in the colon( Reference van Harten-Gerritsen, Balvers and Witkamp 68 ). Calcitriol induces the expression of the cathelicidin anti-microbial peptide, a major component of the innate immune system, in HT29 cells( Reference Gombart, Borregaard and Koeffler 69 ). Lithocholic acid, a secondary bile acid and a vitamin D analogue, decreases NF-κB activity via the VDR in colonic cancer cells (Caco-2 and HT29C19A)( Reference Sun, Mustafi and Cerda 70 ). CYP27B1-knockout mice show increased IL1 and IL17 expressions in the colon and are more susceptible to colitis compared with heterozygote controls( Reference Liu, Nguyen and Chun 71 ). In a Smad3 –/– mouse model of bacteria-induced colitis, increased dietary vitamin D suppressed MAPK and NF-κB activation, severity of colitis and incidence of intestinal cancer( Reference Meeker, Seamons and Paik 72 ). In addition, calcitriol has effects on several immune cell types including dendritic cells, B cells and T cells throughout the human body( Reference Veldhoen and Brucklacher-Waldert 73 ). Specifically, the Vdr-knockout mouse model showed that VDR is required for the maturation and proliferation of intestinal CD8αα + intra-epithelial lymphocytes( Reference Bruce and Cantorna 74 ), which might have a regulatory role within the gut( Reference van Wijk and Cheroutre 75 ). On the other hand, the effect of calcitriol and the level of expression of VDR may both be affected by the immune environment of the colon: in human colon ductal epithelium, VDR expression is considerably decreased in patients with ulcerative colitis and is even lower in patients with colitis-associated colorectal cancer( Reference Wada, Tanaka and Maeda 76 ). In line with this, treatment with TNF and IL-6 leads to decreased expression of CYP27B1 in colonic epithelial COGA-1A cells( Reference Hummel, Fetahu and Groschel 77 ).

Recent studies have shown interactions between gut microbiota and immunity in colon carcinogenesis( Reference Johnson, Dejea and Edler 78 Reference O’Keefe, Li and Lahti 80 ), and vitamin D has been reported to regulate the gut microbiome. In a dextran sodium sulphate-induced colitis model, mice on vitamin D-deficient diet showed more prominent symptoms of colitis and elevated concentrations of bacteria compared with mice on vitamin D-sufficient diet( Reference Lagishetty, Misharin and Liu 81 ). Similarly, in the same colitis model, Ooi et al. ( Reference Ooi, Li and Rogers 82 ) showed that Cyp27b1-knockout mice had higher concentrations of the Helicobacter species in the faeces and more severe symptoms of colitis compared with wild-type littermates. In addition, calcitriol supplementation (1·25 µg/100 g diet) to Cyp27b1-knockout mice reduced Helicobacter numbers and colitis severity( Reference Ooi, Li and Rogers 82 ). Given the data from mouse models, it would be interesting to investigate changes in the human gut microbiome after vitamin D supplementation.

MicroRNA

MicroRNAs (miR) are implicated in the anti-neoplastic influence of vitamin D( Reference Christakos, Dhawan and Verstuyf 12 ). Alvarez-Diaz et al. ( Reference Alvarez-Diaz, Valle and Ferrer-Mayorga 83 ) reported that miR-22 is induced by calcitriol in a time-, dose- and VDR-dependent manner in multiple human colorectal cancer cell lines. Specifically, in SW480-ADH and HCT116 cells that express VDR, miR-22 is required for the anti-proliferative and anti-migratory effects of calcitriol, and regulates the expression of several target genes of calcitriol. Consistently, miR-22 expression is associated with VDR expression in human colorectal cancer samples, suggesting that miR-22 has a role in the VDR-mediated anti-tumour effect of vitamin D.

Padi et al. ( Reference Padi, Zhang and Rustum 84 ) found that calcitriol up-regulates miR-627, which in turn mediates the anti-growth effect of calcitriol in HT29 cells; they reported that miR-627 down-regulates the expression of KDM3A (lysine demethylase 3A, a histone demethylase), increases methylation of histone H3K9, and thereby suppresses the expression of proliferative factors such as GDF15 (growth differentiation factor 15). This same effect of miR-627 is also found in the HCT116 xenograft model of nude mice( Reference Padi, Zhang and Rustum 84 ). Collectively, these findings suggest that enhancing the effect of miR-627, or suppressing its target KDM3A, has the same anti-tumour effect as does vitamin D, and may bypass the side-effects of hypercalcaemia.

Vitamin D in animal models of colorectal cancer

Studies in various animal models of colorectal cancer support a protective role of vitamin D. A western-style diet (high in fat and low in vitamin D and Ca) induces benign and malignant tumours in various mouse models of intestinal tumourigenesis, and supplementation with vitamin D plus Ca produces a significant decrease in the incidence and multiplicity of colon tumours( Reference Newmark, Yang and Kurihara 85 ). In murine models of colorectal carcinogenesis induced by exogenous carcinogens, administration of calcitriol or vitamin D also impedes the neoplastic process( Reference Iseki, Tatsuta and Uehara 67 , Reference Mokady, Schwartz and Shany 86 , Reference Hummel, Thiem and Hobaus 87 ).

Tumour cells implanted into mice are commonly used to evaluate anti-cancer treatments. In a human colorectal cancer (MC26) xenograft model, mice fed a vitamin D-sufficient diet had smaller tumours than those fed a vitamin D-deficient diet( Reference Tangpricha, Spina and Yao 88 ); in nude mice, treatment with vitamin D analogues (BGP-13 and BGP-15) inhibited the growth of human HT29 xenograft( Reference Berkovich, Sintov and Ben-Shabat 46 ). Add-on of the vitamin D analogues PRI-2191 and PRI-2205 showed improved anti-tumour effects compared with chemotherapy alone, which included 5-fluorouracil, capecitabine, irinotecan or oxaliplatin( Reference Milczarek, Psurski and Kutner 89 , Reference Milczarek, Rosinska and Psurski 90 ).

Mouse models of intestinal cancer are also generated by introducing specific germ-line mutations. The Apc +/min mice develop more than 100 intestinal tumours per animal, and calcitriol significantly decreases the surface area with polyps in the gastrointestinal tract( Reference Xu, Posner and Stevenson 54 , Reference Huerta, Irwin and Heber 91 ). In the Apc +/1638N mouse model of intestinal cancer, when the animals were fed a western-style diet, adding dietary vitamin D and Ca induced apoptosis of epithelial cells and inhibited tumourigenesis in the intestine( Reference Yang, Lamprecht and Shinozaki 61 ). A protective effect by vitamin D was also observed in Smad3–/– mice, a model of bacteria-driven colitis and colon cancer when infected with Helicobacter bilis ( Reference Meeker, Seamons and Paik 72 ). Finally, a Vdr-knockout mouse model, compared with wild-type and heterozygote mice, showed increased markers of cell proliferation and oxidative stress in the colon descendens( Reference Kallay, Pietschmann and Toyokuni 92 ). Compared with Apc +/min Vdr +/+ mice, Apc +/min Vdr –/– mice have increased nuclear Ctnnb1, higher expressions of Ctnnb1/Tcf7l2 target genes and larger tumours in the intestine( Reference Jesus Larriba, Ordonez-Moran and Chicote 93 ), supporting the anti-neoplastic effect of VDR in the colon.

Vitamin D action in human colon and rectum

Beyond cell lines and animal models, researchers have studied the effects of supplemental vitamin D in the colon and rectum of humans. In a randomised, double-blinded, controlled trial of 2×2 factorial design, Bostick( Reference Bostick 94 ) and colleagues tested the efficacy of 20 µg of vitamin D and/or 2 g of Ca daily for 6 months on subjects with recently diagnosed colorectal adenoma. Normal-appearing rectal mucosa was biopsied, and immunohistochemistry was performed for markers of differentiation and proliferation. A statistically significant increase of expressions in the vitamin D group relative to the placebo group was found for BAX (56 %)( Reference Fedirko, Bostick and Flanders 95 ), CDKN1A (142 %)( Reference Fedirko, Bostick and Flanders 96 ), APC (48 %), CDH1 (78 %)( Reference Ahearn, Shaukat and Flanders 97 ), MSH2 (mutS homolog 2; 169 %)( Reference Sidelnikov, Bostick and Flanders 98 ), CASR (39 %) and CYP27B1 (159 %)( Reference Ahearn, McCullough and Flanders 99 ). These findings, in line with preclinical studies, indicate that supplemental vitamin D can favourably modulate multiple biomarkers of colorectal cancer risk in normal colon tissues.

Epidemiological studies of vitamin D and colorectal cancer

Epidemiological studies have extensively investigated the relationship between vitamin D status and colorectal cancer, not only on the incidence of the disease but also on the survival of its patients. Regarding the surrogates for vitamin D status, the evidence of association is strong for plasma 25(OH)D concentration but less so for vitamin D intake. For a better interpretation of the data, the strengths and weaknesses of the surrogates are discussed in the context of study design.

Measurement of vitamin D in human populations

Determination of the vitamin D status of individuals in population-based studies needs a consideration of both biology and logistics. The plasma concentration of total 25(OH)D, the major circulating metabolite of vitamin D, is commonly used to determine vitamin D status( Reference Holick 100 ). For instance, a 25(OH)D concentration of <20 ng/ml (50 nmol/l) is considered vitamin D insufficiency( Reference Ross, Manson and Abrams 101 ), and 25(OH)D concentration of >150 ng/ml (375 nmol/l) may cause vitamin D intoxication( Reference Holick 100 ). However, the association of 25(OH)D with colorectal cancer may be confounded by other risk factors. For example, both obesity and low physical activity have been associated with lower plasma 25(OH)D concentrations, as well as with increased colorectal cancer risk( Reference Davis and Dwyer 102 ). Inflammation has been postulated as another confounder based on the assumption that inflammation reduces 25(OH)D concentrations( Reference Autier, Boniol and Pizot 103 ), although there is some evidence against this theory( Reference Song, Wu and Chan 104 ). Moreover, especially for cohorts, the time of blood sampling will likely precede the diagnosis of colorectal cancer for a variety of years for different patients, and it might be helpful to have an additional 25(OH)D measurement that is within a comparable time from diagnosis among all patients( Reference Hofmann, Yu and Horst 105 , Reference Grant 106 ). However, serial blood sampling may not be feasible in many large-scale cohort studies.

Alternatively, dietary or supplementary intakes of vitamin D can be assessed repeatedly with questionnaires. Nevertheless, recall of diet and supplement use is imprecise. Moreover, as skin exposed to sunlight also produces vitamin D, vitamin D intake does not necessarily represent overall vitamin D status or the plasma concentration of 25(OH)D. In 3345 subjects of the Women’s Health Initiative (WHI) observational study, total vitamin D intake calculated based on information from questionnaires explained 9 % variance in serum 25(OH)D concentrations( Reference Cheng, Millen and Wactawski-Wende 107 ).

Recently, a predicted 25(OH)D score using dietary and lifestyle information collected from questionnaires has been used as a surrogate of vitamin D status( Reference Bertrand, Giovannucci and Liu 108 , Reference Jung, Qian and Yamauchi 109 ). Using multivariate linear regression, Bertrand et al. ( Reference Bertrand, Giovannucci and Liu 108 ) derived this score based on known determinants of circulating 25(OH)D, including age, race, UV radiation exposure, vitamin D intake, BMI, physical activity, alcohol intake, postmenopausal hormone use and season of blood collection from more than 4500 participants with available blood samples in three US nationwide cohorts. The predicted score explained 25–33 % variance in plasma 25(OH)D concentrations in different cohorts. This approach of using information from questionnaires estimates vitamin D status data in cohorts where plasma concentrations are not available, and incorporates not only dietary vitamin D intake but also non-dietary exposures, which are associated with increased plasma 25(OH)D concentrations. Of note, the predicted score was derived from the original cohorts, and its application to other cohorts will require further validation.

Plasma concentrations of 25-hydroxyvitamin D and incidence of colorectal cancer

Table 1 summarises previous studies investigating plasma 25(OH)D concentrations and incidence of colorectal cancer with at least 300 cases( Reference Jung, Qian and Yamauchi 109 Reference Song, Nishihara and Wang 122 ). Evidence for the association of plasma 25(OH)D concentration or 25(OH)D score with lower colorectal cancer incidence is quite strong. To further support this, two meta-analyses reported inverse associations between plasma 25(OH)D concentration and risk of colorectal adenoma, a well-established precancerous lesion for colorectal cancer( Reference Lee 123 , Reference Yin, Grandi and Raum 124 ).

Table 1 Major studies (no. of cases ≥300) investigating plasma 25-hydroxyvitamin D (25(OH)D) concentrations and incidence of colorectal cancer (Odds ratios, hazard ratios (HR) and 95 % confidence intervals)

WHI, Women’s Health Initiative; NHS, Nurses’ Health Study; HPFS, Health Professionals Follow-up Study; JPHC Study, Japan Public Health Center-based Prospective Study; EPIC, European Prospective Investigation into Cancer and Nutrition; Ref., referent values; MCCS, Melbourne Collaborative Cohort Study; ATBC, Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study; VDR, vitamin D receptor; DBP, vitamin D-binding protein; PLCO, Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial; MPE, molecular pathological epidemiology.

* P heterogeneity is for colon cancer v. rectal cancer.

P heterogeneity is for VDR (–) v. VDR (+).

P heterogeneity is for low v. high DBP.

§ P heterogeneity is for high v. mild v. absent reaction.

By integrating exposure data such as vitamin D status and tumour molecular/immune features of colorectal cancer tissue, MPE( Reference Ogino, Chan and Fuchs 5 , Reference Ogino and Stampfer 6 , Reference Ogino, Lochhead and Chan 125 , Reference Ogino, Campbell and Nishihara 126 ) research provides new insights into the relationship between vitamin D and colorectal cancer. Jung et al. ( Reference Jung, Qian and Yamauchi 109 ) studied the risk of colorectal cancer in relation to the predicted score for 25(OH)D concentration (with 1059 incident cases during the follow-up of 140 418 participants). A higher predicted 25(OH)D score was inversely associated with colorectal cancer risk (P<0·001), regardless of VDR expression levels in tumour cells (P heterogeneity=0·75). Considering the role of vitamin D in the immune system, another MPE study showed that high plasma 25(OH)D concentration was associated with lower risk of colorectal cancer with high-level immune reaction (P trend<0·001), but not with risk of tumour with lower-level reaction (P trend>0·50, P heterogeneity=0·001)( Reference Song, Nishihara and Wang 122 ). This statistical analysis of heterogeneity is critical, as the hypotheses address differential effects of vitamin D on subtypes( Reference Wang, Kuchiba and Ogino 127 , Reference Wang, Spiegelman and Kuchiba 128 ). These data support the hypothesis that effect of vitamin D might be strong in tumours enriched with immune cells( Reference Song, Nishihara and Wang 122 ), because immune cells in tumour can activate vitamin D, and thereby increase local levels of active vitamin D( Reference Edfeldt, Liu and Chun 129 Reference Sigmundsdottir, Pan and Debes 131 ). Although a replication by additional studies is needed, these findings suggest an interplay of vitamin D status and the immune system in inhibiting the tumourigenesis of colorectal cancer. In addition, a possible interaction may exist between vitamin D status and tumour immunity status in colorectal cancer patient survival analyses, requiring further investigation. With complex immune and inflammatory processes suggested to be involved in colorectal cancer progression and regulated by vitamin D, it has been recommended that future epidemiological studies should measure both vitamin D and inflammatory markers, preferably multiple times, and perform mediation analysis( Reference Nishihara, VanderWeele and Shibuya 132 ) to study the role of inflammation as a mediator between vitamin D and colorectal cancer( Reference van Harten-Gerritsen, Balvers and Witkamp 68 ).

Plasma 25-hydroxyvitamin D concentration and survival of colorectal cancer

Table 2 shows previous studies with at least 300 cases on plasma 25(OH)D concentration and survival of patients with diagnosed colorectal cancer( Reference Ng, Meyerhardt and Wu 133 Reference Ng, Venook and Sato 139 ). Of note, to reduce potential reverse causation associated with undiagnosed tumours at the time of blood sampling that might lower plasma 25(OH)D concentrations, the Nurses’ Health Study( Reference Ng, Meyerhardt and Wu 133 , Reference Ng, Wolpin and Meyerhardt 134 ), the Health Professionals Follow-up Study( Reference Ng, Meyerhardt and Wu 133 , Reference Ng, Wolpin and Meyerhardt 134 ) and the European Prospective Investigation into Cancer and Nutrition study( Reference Fedirko, Riboli and Tjonneland 136 ) measured plasma 25(OH)D concentration before diagnosis of colorectal cancer, and excluded cases diagnosed within 2 years after blood collection. In contrast, the Study of Colorectal Cancer in Scotland( Reference Zgaga, Theodoratou and Farrington 137 ) and the Cancer and Leukemia Group B/Southwest Oncology Group 80405( Reference Ng, Venook and Sato 139 ) studies measured 25(OH)D shortly after diagnosis, a timing more subject to reverse causation. Despite the different timing of blood collection, there is a consistent prognostic association of plasma 25(OH)D concentration with colorectal cancer patient survival.

Table 2 Major studies (no. of cases ≥300) investigating plasma 25-hydroxyvitamin D (25(OH)D) concentrations and survival of patients with diagnosed colorectal cancer (CRC) (Hazard ratios (HR) and 95 % confidence intervals)

NHS, Nurses’ Health Study; HPFS, Health Professionals Follow-up Study; NCCTG, North Central Cancer Treatment Group; mCRC, metastatic colorectal cancer; PFS, progression-free survival; EPIC, European Prospective Investigation into Cancer and Nutrition; SOCCS, Study of Colorectal Cancer in Scotland; CALGB, Cancer and Leukemia Group B; SWOG, Southwest Oncology Group.

Vitamin D intake and incidence of colorectal cancer

Table 3 lists previous studies exploring the relationship between vitamin D intake and risk of colorectal cancer with at least 500 cases( Reference Jenab, Bueno-de-Mesquita and Ferrari 113 , Reference Ma, Zhang and Wang 115 , Reference Martinez, Giovannucci and Colditz 140 Reference McCullough, Robertson and Rodriguez 150 ). In contrast to the consistent and strong evidence from the studies measuring plasma 25(OH)D, the association between vitamin D intake and incidence of colorectal cancer is conflicting. Nevertheless, a 2011 meta-analysis( Reference Ma, Zhang and Wang 115 ) of prospective studies reported an inverse association between vitamin D intake and colorectal cancer incidence.

Table 3 Major studies (no. of cases ≥500) investigating vitamin D intake and incidence of colorectal cancer (CRC) (Odds ratios, relative risks (RR) and 95 % confidence intervals)

NHS, Nurses’ Health Study; SMC, Swedish Mammography Cohort; CPS II, Cancer Prevention study II; JPHC Study, Japan Public Health Center-based Prospective Study; EPIC, European Prospective Investigation into Cancer and Nutrition.

* Indoor, subjects engaged in sedentary or standing work (including no job) and no outdoor physical activity at leisure; outdoor, subjects engaged in work with labour or walking or outdoor physical activity at leisure at least of 120 min/week.

Vitamin D intake and survival of colorectal cancer

Observational studies on the impact of vitamin D intake in patients with diagnosed colorectal cancer are limited. In a paper published in 2014, Yang et al. ( Reference Yang, McCullough and Gapstur 151 ) included 1111 participants in the Cancer Prevention Study II Nutrition Cohort who were diagnosed with invasive, non-metastatic colorectal cancer. The researchers evaluated associations of Ca, vitamin D and diary product intakes after colorectal cancer diagnosis with all-cause and colorectal cancer-specific mortality. After a mean follow-up of 7·6 years, both Ca and milk intakes were inversely associated with all-cause mortality and colorectal cancer-specific mortality, but vitamin D intake was not associated with either mortality outcomes( Reference Yang, McCullough and Gapstur 151 ).

Randomised controlled trials

Randomised placebo-controlled trials are the ‘gold standard’ in establishing causal association; however, such evidence to date has been inconclusive on the effect of vitamin D on colorectal cancer. The findings and limitations of completed clinical trials are discussed, with a preview of ongoing trials, which might hopefully conclude the controversy.

Completed clinical trials of vitamin D intake and incidence of colorectal cancer

To date, four completed randomised controlled trials of vitamin D have a reasonable number of cancer cases (Table 4)( Reference Wactawski-Wende, Kotchen and Anderson 110 , Reference Trivedi, Doll and Khaw 152 Reference Avenell, MacLennan and Jenkinson 154 ). In a substudy of the WHI, 36 282 postmenopausal women were given 5µg of vitamin D and 500 mg of Ca twice daily (10µg of vitamin D and 1000 mg of Ca daily), or a matching placebo, for an average of 7 years( Reference Wactawski-Wende, Kotchen and Anderson 110 ). The incidence of invasive colorectal cancer in this study did not differ significantly between women assigned to Ca plus vitamin D and those assigned to placebo (168 v. 154 cases, hazard ratio 1·08; 95 % CI 0·86, 1·34; P=0·51), and tumour characteristics were similar in the two groups. This study has several limitations. First, the modest dose of vitamin D used in the trial led to only a small rise in plasma 25(OH)D concentrations( Reference Ng, Scott and Drake 155 ), which was measured only in a small sample of the study population. Second, the limited compliance in the treatment group and the allowance for the placebo group to take supplements could have further reduced the actual contrast of 25(OH)D between groups. In fact, as shown in a post hoc analysis of WHI, in 15 646 women (43 %) who were not taking personal Ca or vitamin D supplements at randomisation, Ca and vitamin D treatment non-significantly reduced the risk of colorectal cancer by 17 %( Reference Bolland, Grey and Gamble 156 ). Third, the 7-year follow-up may not be sufficient to show a benefit for the prevention of colorectal cancer, which has a long natural history and a relatively low incidence.

Table 4 Major completed randomised trials (n ≥1000) investigating vitamin D supplementation and cancer (Hazard ratios (HR) and 95 % confidence intervals)

CRC, colorectal cancer; WHI, Women’s Health Initiative; RECORD, Randomised Evaluation of Calcium Or vitamin D.

A second completed randomised trial was carried out in the UK, with 2686 participants (2037 men and 649 women)( Reference Trivedi, Doll and Khaw 152 ). An oral supplement of 2500µg vitamin D or a matching placebo was given every 4 months for 5 years. Over the 5-year period, twenty-eight and twenty-seven cases of colon cancer were documented in the treatment and control group, respectively, with no association with vitamin D treatment (relative risk 1·02; 95 % CI 0·60, 1·74; P=0·94). This study applied a dosage of vitamin D that had a moderate effect upon the measured plasma 25(OH)D concentration (74·3 nmol/l in the treatment group v. 53·4 nmol/l in the control group, P<0·001); nevertheless, it was limited by the small sample size and the short follow-up period.

Two other studies have investigated the association of vitamin D and Ca supplement intake with cancer incidence. The Nebraska trial( Reference Lappe, Travers-Gustafson and Davies 153 ) detected lower incidence of cancer in patients treated with vitamin D plus Ca than with placebo (P<0·03), whereas the Randomised Evaluation of Calcium Or vitamin D trial( Reference Avenell, MacLennan and Jenkinson 154 ) found no association. However, neither study was designed to detect the association of supplement use with colorectal cancer incidence as the primary end point.

In the recently published Vitamin D/Calcium Polyp Prevention trial (Table 4)( Reference Baron, Barry and Mott 157 ), patients with recently diagnosed adenomas were randomly assigned 25µg of vitamin D daily or no vitamin D in a factorial design. After 3 or 5 years of treatment, participants given vitamin D had a mean net increase in serum 25(OH)D concentrations of 7·83 ng/ml, relative to participants given placebo. Overall, 43 % of the participants had one or more adenomas diagnosed during follow-up, and the adjusted risk ratio for recurrent adenoma was 0·99 (95 % CI 0·89, 1·09) with vitamin D v. no vitamin D.

Two points are worth noting for comparison of this null finding with preexisting epidemiological evidence. First, as the authors admitted, the vitamin D dose in the Polyp Prevention trial (25µg daily) was lower than the dose many experts now recommend( Reference Bischoff-Ferrari, Giovannucci and Willett 158 , Reference Bischoff-Ferrari, Shao and Dawson-Hughes 159 ), and it was used for a limited time( Reference Baron, Barry and Mott 157 ). This resulted in a net increase of 7·83 ng/ml of serum 25(OH)D, in contrast with a generally >20 ng/ml difference between the high and low quartiles or quintiles of 25(OH)D in observational studies( Reference Giovannucci 160 ). Thus, the moderate dose of vitamin D might not cause a change in adenoma incidence that was detectable by the power of this trial. Second, the risk of incidence for recurrent adenoma is not a direct translation of the risk for incident adenoma or colorectal cancer. For instance, in a colorectal cancer screening trial, elevated dietary fibre intake was associated with reduced risk of incident colorectal adenoma and colorectal cancer (OR 0·76 and 0·85, respectively), but not with the risk of recurrent adenoma (OR 1·08)( Reference Kunzmann, Coleman and Huang 161 ). Similarly, a meta-analysis has also shown different associations of higher serum 25(OH)D with incident or recurrent colorectal adenoma (OR 0·82 or 0·87 for a 20 ng/ml increase, respectively)( Reference Yin, Grandi and Raum 124 ). Therefore, the null finding should not be generalised to persons without a recent history of colorectal adenoma. On the basis of the clinical literature included in this review, high vitamin D status might have the greatest anti-neoplastic effects early in colorectal carcinogenesis and later in disease progression, but less so in the metastatic stage or adenoma recurrence.

Ongoing clinical trials of vitamin D intake and incidence of colorectal cancer

Several randomised controlled trials are under way to study whether vitamin D supplementation reduces the risk of cancer (Table 5)( Reference Manson and Bassuk 162 ). These trials are applying higher dosages of vitamin D than previous trials, and are measuring baseline and/or follow-up plasma 25(OH)D concentrations. For example, the VITamin D and OmegA-3 TriaL study collects baseline blood samples on 17 000 participants and follow-up samples on 6000( Reference Pradhan and Manson 163 ). In aggregate, these trials have already recruited over 53 000 participants, and the first results are expected to be available in 2015.

Table 5 Major ongoing randomised trials (n ≥1000) investigating vitamin D supplementation and cancer

VITAL, VITamin D and OmegA-3 TriaL; FIND, Finnish Vitamin D Trial; CAPS, Clinical Trial of Vitamin D3 to Reduce Cancer Risk in Postmenopausal Women; VIDAL, Vitamin D and Longevity Trial.

Clinical trial of vitamin D intake and survival of colorectal cancer

Accumulating evidence of the involvement of vitamin D in cancer progression demands clinical trials for patients diagnosed with colorectal cancer. The study of mortality, rather than incidence, of colorectal cancer will likely require fewer subjects and shorter follow-up. To date, only one clinical trial is registered on ClinicalTrials.gov addressing this question (NCT01516216); it is recruiting 120 participants with previously untreated metastatic colorectal cancer and randomising them into two arms. Together with the standard chemotherapy with FOLFOX and bevacizumab, arm 1 gets 10 µg/d of vitamin D, whereas arm 2 gets a loading dose of 200 µg/d for 2 weeks followed by a maintenance dose of 100 µg/d. Although the sample size is small, the study does collect plasma 25(OH)D concentrations, and thus analyses of the relationships between high-dose vitamin D treatment, 25(OH)D status and prognosis are possible.

Genetic variation, vitamin D status and colorectal cancer

Heritable factors explain approximately 35 % of the risk of colorectal cancer( Reference Lichtenstein, Holm and Verkasalo 164 ), and contribute substantially to the variability of vitamin D status( Reference Shea, Benjamin and Dupuis 165 ). Thus, genetic variation related to vitamin D status might have an impact on the risk of colorectal cancer. A genome-wide association study of circulating 25(OH)D concentrations in 33 996 individuals has identified SNP loci near four genes including GC (group-specific component, which encodes vitamin D-binding protein), DHCR7 (7-dehydrocholesterol reductase, which can remove the substrate from vitamin D synthesis in skin), CYP2R1 and CYP24A1 ( Reference Wang, Zhang and Richards 166 ). To gain insight into the genetic link between vitamin D status and colorectal cancer, Hiraki et al. ( Reference Hiraki, Qu and Hutter 167 ) investigated these four SNP loci in 10 061 colorectal cancer cases and 12 768 controls, but found no significant association between the loci and risk of colorectal cancer. A similar null finding was reported in another cohort containing 438 colorectal cancer cases( Reference Jorde, Schirmer and Wilsgaard 168 ). Moreover, the four loci do not overlap with the risk variants identified from previous genome-wide association studies for colorectal cancer( Reference Peters, Bien and Zubair 169 ). As the SNP identified by Wang et al. ( Reference Wang, Zhang and Richards 166 ) can explain only a small variation (1–4 %) in 25(OH)D concentrations, the reduction in overall colorectal cancer risk by increased vitamin D levels due to the SNP might be too small to be detectable. In addition to genes related to vitamin D metabolism, VDR polymorphism has also been studied with regard to risk of colorectal cancer, although most results are inconclusive( Reference Koestner, Denzer and Mueller 170 ). Nevertheless, two meta-analyses have shown significant associations of risk for colorectal cancer with two VDR polymorphisms, BsmI (rs1544410) (relative risk (RR) 0·57; 95 % CI 0·36, 0·89 for BB v. bb)( Reference Touvier, Chan and Lau 171 ) and TaqI (rs731236) (OR 1·43; 95 % CI 1·30, 1·58 for tt v. TT)( Reference Serrano, Gnagnarella and Raimondi 172 ), respectively.

As one future direction, the MPE approach may link vitamin-D-related SNP to specific subtypes of colorectal cancer. Another future direction is to investigate interactions between SNP of the vitamin D pathway genes and vitamin D status variables in analyses of colorectal cancer incidence and mortality( Reference Hiraki, Joshi and Ng 173 ). In addition to such a candidate gene approach, analyses of genome-wide gene–environment interactions with vitamin D status variables may enable us to discover potentially important SNP and pathways for colorectal cancer( Reference Peters, Bien and Zubair 169 ). Next-generation sequencing technologies, with greater depth and finer resolution, will draw a broader picture for the targets and interacting factors of vitamin D and VDR, and relate them with specific diseases including colorectal cancer( Reference Ferguson, Laing and Marlow 174 ).

Conclusion

Since Garland & Garland( Reference Garland and Garland 2 ) proposed vitamin D for colon cancer prevention 25 years ago, functional studies on vitamin D or its analogues have provided supportive evidence for its anti-tumour effect in colorectal cancer. Evidence from both in vitro and in vivo experiments suggests that anti-proliferation, pro-differentiation, pro-apoptosis, anti-angiogenesis, immune modulation and miR regulation are involved in the anti-tumour effect of vitamin D. Recent studies also have explored the local expressions and impacts of vitamin D-metabolising enzymes and VDR, which may lead to the discovery of predictive biomarkers for vitamin D treatment response.

Epidemiological studies have consistently demonstrated a strong inverse association of plasma 25(OH)D concentration with colorectal cancer incidence and mortality. The MPE approach is valuable in generating hypotheses on potential mechanisms of the observed protective effect of vitamin D, and in identifying molecular pathological signatures as predictive markers for benefit from vitamin D. On the other hand, the effect of vitamin D intake on colorectal cancer prevention is controversial, largely due to the following reasons: the slow development of colorectal cancer, the confounding effects caused by sunlight exposure, outdoor physical activity, BMI, dairy and Ca intakes, etc. in observational studies, and the suboptimal dosage of vitamin D applied in previous clinical trials. Ongoing large randomised controlled trials with high-dose vitamin D treatment are promising to tackle these problems and decide the value of vitamin D supplementation. Meanwhile, clinical trials of vitamin D on colorectal cancer survival are scarce and logistically more feasible, suggesting a new direction for future studies. Finally, next-generation sequencing and studies of genome-wide gene–environment interactions will likely shed more light on the mechanisms of association between vitamin D and colorectal cancer.

Acknowledgements

The authors thank Sonal Jhaveri, who is supported by the Dana-Farber Cancer Institute, for editing the manuscript.

This work was supported by US National Institutes of Health grants (R01 CA151993 and R35 CA197735 to S. O.; U01 CA138962 to J. E. M.; K07 CA148894 to K. N.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The funders had no role in the study design, data collection and analysis, decision to publish or preparation of the manuscript.

R. D. and Z. R. Q. designed the research; R. D. and S. O. wrote the paper; Z. R. Q., K. N., E. L. G. and J. E. M. reviewed the paper; and R. D. and S. O. had primary responsibility for the final content. All authors read and approved the final manuscript.

J. E. M. and colleagues at Brigham and Women’s Hospital, Harvard Medical School, are recipients of funding from the National Institutes of Health to conduct the VITamin D and OmegA-3 TriaL, a large-scale randomised trial of vitamin D and n-3 in the prevention of cancer and CVD (U01 CA138962). The other authors declare that they have no conflicts of interest.

References

1. Holick, MF (2006) The role of vitamin D for bone health and fracture prevention. Curr Osteoporos Rep 4, 96102.Google Scholar
2. Garland, CF & Garland, FC (1980) Do sunlight and vitamin D reduce the likelihood of colon cancer? Int J Epidemiol 9, 227231.Google Scholar
3. Deeb, KK, Trump, DL & Johnson, CS (2007) Vitamin D signalling pathways in cancer: potential for anticancer therapeutics. Nat Rev Cancer 7, 684700.Google Scholar
4. Feldman, D, Krishnan, AV, Swami, S, et al. (2014) The role of vitamin D in reducing cancer risk and progression. Nat Rev Cancer 14, 342357.CrossRefGoogle ScholarPubMed
5. Ogino, S, Chan, AT, Fuchs, CS, et al. (2011) Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut 60, 397411.Google Scholar
6. Ogino, S & Stampfer, M (2010) Lifestyle factors and microsatellite instability in colorectal cancer: the evolving field of molecular pathological epidemiology. J Natl Cancer Inst 102, 365367.Google Scholar
7. Holick, MF (2006) Resurrection of vitamin D deficiency and rickets. J Clin Invest 116, 20622072.Google Scholar
8. Ponchon, G & DeLuca, HF (1969) The role of the liver in the metabolism of vitamin D. J Clin Invest 48, 12731279.Google Scholar
9. Fraser, DR & Kodicek, E (1970) Unique biosynthesis by kidney of a biological active vitamin D metabolite. Nature 228, 764766.Google Scholar
10. Young, MV, Schwartz, GG, Wang, L, et al. (2004) The prostate 25-hydroxyvitamin D-1 alpha-hydroxylase is not influenced by parathyroid hormone and calcium: implications for prostate cancer chemoprevention by vitamin D. Carcinogenesis 25, 967971.Google Scholar
11. Pike, JW & Meyer, MB (2012) Regulation of mouse Cyp24a1 expression via promoter-proximal and downstream-distal enhancers highlights new concepts of 1,25-dihydroxyvitamin D(3) action. Arch Biochem Biophys 523, 28.Google Scholar
12. Christakos, S, Dhawan, P, Verstuyf, A, et al. (2016) Vitamin D: metabolism, molecular mechanism of action, and pleiotropic effects. Physiol Rev 96, 365408.Google Scholar
13. Carlberg, C, Bendik, I, Wyss, A, et al. (1993) Two nuclear signalling pathways for vitamin D. Nature 361, 657660.CrossRefGoogle ScholarPubMed
14. Tagami, T, Lutz, WH, Kumar, R, et al. (1998) The interaction of the vitamin D receptor with nuclear receptor corepressors and coactivators. Biochem Biophys Res Commun 253, 358363.CrossRefGoogle ScholarPubMed
15. Murayama, A, Kim, MS, Yanagisawa, J, et al. (2004) Transrepression by a liganded nuclear receptor via a bHLH activator through co-regulator switching. EMBO J 23, 15981608.CrossRefGoogle Scholar
16. Kim, MS, Fujiki, R, Murayama, A, et al. (2007) 1Alpha,25(OH)2D3-induced transrepression by vitamin D receptor through E-box-type elements in the human parathyroid hormone gene promoter. Mol Endocrinol 21, 334342.Google Scholar
17. Haussler, MR, Whitfield, GK, Kaneko, I, et al. (2013) Molecular mechanisms of vitamin D action. Calcif Tissue Int 92, 7798.Google Scholar
18. Haussler, MR, Jurutka, PW, Mizwicki, M, et al. (2011) Vitamin D receptor (VDR)-mediated actions of 1alpha,25(OH)(2)vitamin D(3): genomic and non-genomic mechanisms. Best Pract Res Clin Endocrinol Metab 25, 543559.CrossRefGoogle Scholar
19. Norman, AW, Mizwicki, MT & Norman, DP (2004) Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nat Rev Drug Discov 3, 2741.Google Scholar
20. Ordonez-Moran, P, Larriba, MJ, Palmer, HG, et al. (2008) RhoA-ROCK and p38MAPK-MSK1 mediate vitamin D effects on gene expression, phenotype, and Wnt pathway in colon cancer cells. J Cell Biol 183, 697710.Google Scholar
21. Hsu, JY, Feldman, D, McNeal, JE, et al. (2001) Reduced 1alpha-hydroxylase activity in human prostate cancer cells correlates with decreased susceptibility to 25-hydroxyvitamin D3-induced growth inhibition. Cancer Res 61, 28522856.Google Scholar
22. Swami, S, Krishnan, AV, Wang, JY, et al. (2012) Dietary vitamin D(3) and 1,25-dihydroxyvitamin D(3) (calcitriol) exhibit equivalent anticancer activity in mouse xenograft models of breast and prostate cancer. Endocrinology 153, 25762587.CrossRefGoogle Scholar
23. Zehnder, D, Bland, R, Williams, MC, et al. (2001) Extrarenal expression of 25-hydroxyvitamin d(3)-1 alpha-hydroxylase. J Clin Endocrinol Metab 86, 888894.Google Scholar
24. Cross, HS, Bises, G, Lechner, D, et al. (2005) The vitamin D endocrine system of the gut – its possible role in colorectal cancer prevention. J Steroid Biochem Mol Biol 97, 121128.Google Scholar
25. Bises, G, Kallay, E, Weiland, T, et al. (2004) 25-Hydroxyvitamin D3-1alpha-hydroxylase expression in normal and malignant human colon. J Histochem Cytochem 52, 985989.Google Scholar
26. Tangpricha, V, Flanagan, JN, Whitlatch, LW, et al. (2001) 25-Hydroxyvitamin D-1 alpha-hydroxylase in normal and malignant colon tissue. Lancet 357, 16731674.Google Scholar
27. Cross, HS, Bareis, P, Hofer, H, et al. (2001) 25-Hydroxyvitamin D(3)-1alpha-hydroxylase and vitamin D receptor gene expression in human colonic mucosa is elevated during early cancerogenesis. Steroids 66, 287292.CrossRefGoogle Scholar
28. Bareis, P, Bises, G, Bischof, MG, et al. (2001) 25-Hydroxy-vitamin D metabolism in human colon cancer cells during tumor progression. Biochem Biophys Res Commun 285, 10121017.Google Scholar
29. Ogunkolade, BW, Boucher, BJ, Fairclough, PD, et al. (2002) Expression of 25-hydroxyvitamin D-1-α-hydroxylase mRNA in individuals with colorectal cancer. Lancet 359, 18311832.Google Scholar
30. Bareis, P, Kallay, E, Bischof, MG, et al. (2002) Clonal differences in expression of 25-hydroxyvitamin D(3)-1alpha-hydroxylase, of 25-hydroxyvitamin D(3)-24-hydroxylase, and of the vitamin D receptor in human colon carcinoma cells: effects of epidermal growth factor and 1alpha, 25-dihydroxyvitamin D(3). Exp Cell Res 276, 320327.CrossRefGoogle Scholar
31. Holt, PR, Arber, N, Halmos, B, et al. (2002) Colonic epithelial cell proliferation decreases with increasing levels of serum 25-hydroxy vitamin D. Cancer Epidemiol Biomarkers Prev 11, 113119.Google ScholarPubMed
32. Wagner, D, Dias, AG, Schnabl, K, et al. (2012) Determination of 1,25-dihydroxyvitamin D concentrations in human colon tissues and matched serum samples. Anticancer Res 32, 259263.Google Scholar
33. Horvath, HC, Lakatos, P, Kosa, JP, et al. (2010) The candidate oncogene CYP24A1: a potential biomarker for colorectal tumorigenesis. J Histochem Cytochem 58, 277285.Google Scholar
34. Anderson, MG, Nakane, M, Ruan, X, et al. (2006) Expression of VDR and CYP24A1 mRNA in human tumors. Cancer Chemother Pharmacol 57, 234240.Google Scholar
35. Kosa, JP, Horvath, P, Wolfling, J, et al. (2013) CYP24A1 inhibition facilitates the anti-tumor effect of vitamin D3 on colorectal cancer cells. World J Gastroenterol 19, 26212628.CrossRefGoogle ScholarPubMed
36. Wang, Y, Zhu, J & DeLuca, HF (2012) Where is the vitamin D receptor? Arch Biochem Biophys 523, 123133.Google Scholar
37. Matusiak, D, Murillo, G, Carroll, RE, et al. (2005) Expression of vitamin D receptor and 25-hydroxyvitamin D3-1{alpha}-hydroxylase in normal and malignant human colon. Cancer Epidemiol Biomarkers Prev 14, 23702376.Google Scholar
38. Palmer, HG, Larriba, MJ, Garcia, JM, et al. (2004) The transcription factor SNAIL represses vitamin D receptor expression and responsiveness in human colon cancer. Nat Med 10, 917919.Google Scholar
39. Larriba, MJ, Bonilla, F & Munoz, A (2010) The transcription factors Snail1 and Snail2 repress vitamin D receptor during colon cancer progression. J Steroid Biochem Mol Biol 121, 106109.Google Scholar
40. Ramagopalan, SV, Heger, A, Berlanga, AJ, et al. (2010) A ChIP-seq defined genome-wide map of vitamin D receptor binding: associations with disease and evolution. Genome Res 20, 13521360.Google Scholar
41. Meyer, MB, Goetsch, PD & Pike, JW (2012) VDR-RXR and TCF4/beta-catenin cistromes in colonic cells of colorectal tumor origin: impact on c-FOS and c-MYC gene expression. Mol Endocrinol 26, 3751.Google Scholar
42. Goeman, F, De Nicola, F, D’Onorio De Meo, P, et al. (2014) VDR primary targets by genome-wide transcriptional profiling. J Steroid Biochem Mol Biol 143, 348356.CrossRefGoogle ScholarPubMed
43. Shabahang, M, Buras, RR, Davoodi, F, et al. (1993) 1,25-Dihydroxyvitamin D3 receptor as a marker of human colon carcinoma cell line differentiation and growth inhibition. Cancer Res 53, 37123718.Google Scholar
44. Scaglione-Sewell, BA, Bissonnette, M, Skarosi, S, et al. (2000) A vitamin D3 analog induces a G1-phase arrest in CaCo-2 cells by inhibiting cdk2 and cdk6: roles of cyclin E, p21Waf1, and p27Kip1. Endocrinology 141, 39313939.CrossRefGoogle ScholarPubMed
45. Gaschott, T, Steinmeyer, A, Steinhilber, D, et al. (2002) ZK 156718, a low calcemic, antiproliferative, and prodifferentiating vitamin D analog. Biochem Biophys Res Commun 290, 504509.Google Scholar
46. Berkovich, L, Sintov, AC & Ben-Shabat, S (2013) Inhibition of cancer growth and induction of apoptosis by BGP-13 and BGP-15, new calcipotriene-derived vitamin D3 analogs, in-vitro and in-vivo studies. Invest New Drugs 31, 247255.Google Scholar
47. Chen, AP, Davis, BH, Sitrin, MD, et al. (2002) Transforming growth factor-beta 1 signaling contributes to Caco-2 cell growth inhibition induced by 1,25(OH)(2)D-3. Am J Physiol Gastrointest Liver Physiol 283, G864G874.Google Scholar
48. Oh, YS, Kim, EJ, Schaffer, BS, et al. (2001) Synthetic low-calcaemic vitamin D(3) analogues inhibit secretion of insulin-like growth factor II and stimulate production of insulin-like growth factor-binding protein-6 in conjunction with growth suppression of HT-29 colon cancer cells. Mol Cell Endocrinol 183, 141149.Google Scholar
49. Tong, WM, Hofer, H, Ellinger, A, et al. (1999) Mechanism of antimitogenic action of vitamin D in human colon carcinoma cells: relevance for suppression of epidermal growth factor-stimulated cell growth. Oncol Res 11, 7784.Google ScholarPubMed
50. Diaz, GD, Paraskeva, C, Thomas, MG, et al. (2000) Apoptosis is induced by the active metabolite of vitamin D3 and its analogue EB1089 in colorectal adenoma and carcinoma cells: possible implications for prevention and therapy. Cancer Res 60, 23042312.Google Scholar
51. Chen, A, Davis, BH, Bissonnette, M, et al. (1999) 1,25-Dihydroxyvitamin D(3) stimulates activator protein-1-dependent Caco-2 cell differentiation. J Biol Chem 274, 3550535513.Google Scholar
52. Palmer, HG, Gonzalez-Sancho, JM, Espada, J, et al. (2001) Vitamin D-3 promotes the differentiation of colon carcinoma cells by the induction of E-cadherin and the inhibition of beta-catenin signaling. J Cell Biol 154, 369387.CrossRefGoogle ScholarPubMed
53. Palmer, HG, Sanchez-Carbayo, M, Ordonez-Moran, P, et al. (2003) Genetic signatures of differentiation induced by 1alpha,25-dihydroxyvitamin D3 in human colon cancer cells. Cancer Res 63, 77997806.Google Scholar
54. Xu, H, Posner, GH, Stevenson, M, et al. (2010) Apc(MIN) modulation of vitamin D secosteroid growth control. Carcinogenesis 31, 14341441.CrossRefGoogle ScholarPubMed
55. Egan, JB, Thompson, PA, Vitanov, MV, et al. (2010) Vitamin D receptor ligands, adenomatous polyposis coli, and the vitamin d receptor FokI polymorphism collectively modulate beta-catenin activity in colon cancer cells. Mol Carcinog 49, 337352.Google Scholar
56. Aguilera, O, Pena, C, Garcia, JM, et al. (2007) The Wnt antagonist DICKKOPF-1 gene is induced by 1alpha, 25-dihydroxyvitamin D3 associated to the differentiation of human colon cancer cells. Carcinogenesis 28, 18771884.Google Scholar
57. Barnes, JD, Arhel, NJ, Lee, SS, et al. (2005) Nuclear BAG-1 expression inhibits apoptosis in colorectal adenoma-derived epithelial cells. Apoptosis 10, 301311.Google Scholar
58. Liu, G, Hu, X & Chakrabarty, S (2010) Vitamin D mediates its action in human colon carcinoma cells in a calcium-sensing receptor-dependent manner: downregulates malignant cell behavior and the expression of thymidylate synthase and survivin and promotes cellular sensitivity to 5-FU. Int J Cancer 126, 631639.Google Scholar
59. Canaff, L & Hendy, GN (2002) Human calcium-sensing receptor gene. Vitamin D response elements in promoters P1 and P2 confer transcriptional responsiveness to 1,25-dihydroxyvitamin D. J Biol Chem 277, 3033730350.CrossRefGoogle Scholar
60. Chakrabarty, S, Wang, HM, Canaff, L, et al. (2005) Calcium sensing receptor in human colon carcinoma: interaction with Ca2+ and 1,25-dihydroxyvitamin D-3. Cancer Res 65, 493498.Google Scholar
61. Yang, K, Lamprecht, SA, Shinozaki, H, et al. (2008) Dietary calcium and cholecalciferol modulate cyclin D1 expression, apoptosis, and tumorigenesis in intestine of adenomatous polyposis coli1638N/+ mice. J Nutr 138, 16581663.CrossRefGoogle ScholarPubMed
62. Stambolsky, P, Tabach, Y, Fontemaggi, G, et al. (2010) Modulation of the vitamin D3 response by cancer-associated mutant p53. Cancer Cell 17, 273285.CrossRefGoogle ScholarPubMed
63. Mantell, DJ, Owens, PE, Bundred, NJ, et al. (2000) 1 alpha, 25-Dihydroxyvitamin D(3) inhibits angiogenesis in vitro and in vivo . Circ Res 87, 214220.Google Scholar
64. Ben-Shoshan, M, Amir, S, Dang, DT, et al. (2007) 1 alpha, 25-Dihydroxyvitamin D-3 (calcitriol) inhibits hypoxia-inducible factor-1/vascular endothelial growth factor pathway in human cancer cells. Mol Cancer Ther 6, 14331439.Google Scholar
65. Fernandez-Garcia, NI, Palmer, HG, Garcia, M, et al. (2005) 1 alpha, 25-Dihydroxyvitamin D-3 regulates the expression of Id1 and Id2 genes and the angiogenic phenotype of human colon carcinoma cells. Oncogene 24, 65336544.Google Scholar
66. Pendas-Franco, N, Garcia, JM, Pena, C, et al. (2008) DICKKOPF-4 is induced by TCF/beta-catenin and upregulated in human colon cancer, promotes tumour cell invasion and angiogenesis and is repressed by 1alpha,25-dihydroxyvitamin D3 . Oncogene 27, 44674477.CrossRefGoogle Scholar
67. Iseki, K, Tatsuta, M, Uehara, H, et al. (1999) Inhibition of angiogenesis as a mechanism for inhibition by 1alpha-hydroxyvitamin D3 and 1,25-dihydroxyvitamin D3 of colon carcinogenesis induced by azoxymethane in Wistar rats. Int J Cancer 81, 730733.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
68. van Harten-Gerritsen, AS, Balvers, MG, Witkamp, RF, et al. (2015) Vitamin D, inflammation and colorectal cancer progression: a review of mechanistic studies and future directions for epidemiological studies. Cancer Epidemiol Biomarkers Prev 24, 18201828. Google Scholar
69. Gombart, AF, Borregaard, N & Koeffler, HP (2005) Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D-3. FASEB J 19, 10671077.Google Scholar
70. Sun, J, Mustafi, R, Cerda, S, et al. (2008) Lithocholic acid down-regulation of NF-kappa B activity through vitamin D receptor in colonic cancer cells. J Steroid Biochem Mol Biol 111, 3740.CrossRefGoogle ScholarPubMed
71. Liu, N, Nguyen, L, Chun, RF, et al. (2008) Altered endocrine and autocrine metabolism of vitamin D in a mouse model of gastrointestinal inflammation. Endocrinology 149, 47994808.Google Scholar
72. Meeker, S, Seamons, A, Paik, J, et al. (2014) Increased dietary vitamin D suppresses MAPK signaling, colitis, and colon cancer. Cancer Res 74, 43984408.Google Scholar
73. Veldhoen, M & Brucklacher-Waldert, V (2012) Dietary influences on intestinal immunity. Nat Rev Immunol 12, 696708.Google Scholar
74. Bruce, D & Cantorna, MT (2011) Intrinsic requirement for the vitamin D receptor in the development of CD8alphaalpha-expressing T cells. J Immunol 186, 28192825.CrossRefGoogle ScholarPubMed
75. van Wijk, F & Cheroutre, H (2009) Intestinal T cells: facing the mucosal immune dilemma with synergy and diversity. Semin Immunol 21, 130138.Google Scholar
76. Wada, K, Tanaka, H, Maeda, K, et al. (2009) Vitamin D receptor expression is associated with colon cancer in ulcerative colitis. Oncol Rep 22, 10211025.Google Scholar
77. Hummel, DM, Fetahu, IS, Groschel, C, et al. (2014) Role of proinflammatory cytokines on expression of vitamin D metabolism and target genes in colon cancer cells. J Steroid Biochem Mol Biol 144, 9195.Google Scholar
78. Johnson, CH, Dejea, CM, Edler, D, et al. (2015) Metabolism links bacterial biofilms and colon carcinogenesis. Cell Metab 21, 891897.Google Scholar
79. Mima, K, Sukawa, Y, Nishihara, R, et al. (2015) Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol 1, 653661.Google Scholar
80. O’Keefe, SJ, Li, JV, Lahti, L, et al. (2015) Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun 6, 6342.Google Scholar
81. Lagishetty, V, Misharin, AV, Liu, NQ, et al. (2010) Vitamin D deficiency in mice impairs colonic antibacterial activity and predisposes to colitis. Endocrinology 151, 24232432.CrossRefGoogle ScholarPubMed
82. Ooi, JH, Li, Y, Rogers, CJ, et al. (2013) Vitamin D regulates the gut microbiome and protects mice from dextran sodium sulfate-induced colitis. J Nutr 143, 16791686.Google Scholar
83. Alvarez-Diaz, S, Valle, N, Ferrer-Mayorga, G, et al. (2012) MicroRNA-22 is induced by vitamin D and contributes to its antiproliferative, antimigratory and gene regulatory effects in colon cancer cells. Hum Mol Genet 21, 21572165.Google Scholar
84. Padi, SK, Zhang, Q, Rustum, YM, et al. (2013) MicroRNA-627 mediates the epigenetic mechanisms of vitamin D to suppress proliferation of human colorectal cancer cells and growth of xenograft tumors in mice. Gastroenterology 145, 437446.Google Scholar
85. Newmark, HL, Yang, K, Kurihara, N, et al. (2009) Western-style diet-induced colonic tumors and their modulation by calcium and vitamin D in C57Bl/6 mice: a preclinical model for human sporadic colon cancer. Carcinogenesis 30, 8892.Google Scholar
86. Mokady, E, Schwartz, B, Shany, S, et al. (2000) A protective role of dietary vitamin D3 in rat colon carcinogenesis. Nutr Cancer 38, 6573.Google Scholar
87. Hummel, DM, Thiem, U, Hobaus, J, et al. (2013) Prevention of preneoplastic lesions by dietary vitamin D in a mouse model of colorectal carcinogenesis. J Steroid Biochem Mol Biol 136, 284288.Google Scholar
88. Tangpricha, V, Spina, C, Yao, M, et al. (2005) Vitamin D deficiency enhances the growth of MC-26 colon cancer xenografts in Balb/c mice. J Nutr 135, 23502354.Google Scholar
89. Milczarek, M, Psurski, M, Kutner, A, et al. (2013) Vitamin D analogs enhance the anticancer activity of 5-fluorouracil in an in vivo mouse colon cancer model. BMC Cancer 13, 294.Google Scholar
90. Milczarek, M, Rosinska, S, Psurski, M, et al. (2013) Combined colonic cancer treatment with vitamin D analogs and irinotecan or oxaliplatin. Anticancer Res 33, 433444.Google Scholar
91. Huerta, S, Irwin, RW, Heber, D, et al. (2002) 1 alpha,25-(OH)(2)-D-3 and its synthetic analogue decrease tumor load in the Apc(min) mouse. Cancer Res 62, 741746.Google Scholar
92. Kallay, E, Pietschmann, P, Toyokuni, S, et al. (2001) Characterization of a vitamin D receptor knockout mouse as a model of colorectal hyperproliferation and DNA damage. Carcinogenesis 22, 14291435.Google Scholar
93. Jesus Larriba, M, Ordonez-Moran, P, Chicote, I, et al. (2011). Vitamin D receptor deficiency enhances Wnt/beta-catenin signaling and tumor burden in colon cancer. PLoS ONE 6, e23524.CrossRefGoogle Scholar
94. Bostick, RM (2015) Effects of supplemental vitamin D and calcium on normal colon tissue and circulating biomarkers of risk for colorectal neoplasms. J Steroid Biochem Mol Biol 148, 8695.CrossRefGoogle ScholarPubMed
95. Fedirko, V, Bostick, RM, Flanders, WD, et al. (2009) Effects of vitamin D and calcium supplementation on markers of apoptosis in normal colon mucosa: a randomized, double-blind, placebo-controlled clinical trial. Cancer Prev Res 2, 213223.Google Scholar
96. Fedirko, V, Bostick, RM, Flanders, WD, et al. (2009) Effects of vitamin d and calcium on proliferation and differentiation in normal colon mucosa: a randomized clinical trial. Cancer Epidemiol Biomarkers Prev 18, 29332941.Google Scholar
97. Ahearn, TU, Shaukat, A, Flanders, WD, et al. (2012) A randomized clinical trial of the effects of supplemental calcium and vitamin D3 on the APC/beta-catenin pathway in the normal mucosa of colorectal adenoma patients. Cancer Prev Res 5, 12471256.Google Scholar
98. Sidelnikov, E, Bostick, RM, Flanders, WD, et al. (2010) Effects of calcium and vitamin D on MLH1 and MSH2 expression in rectal mucosa of sporadic colorectal adenoma patients. Cancer Epidemiol Biomarkers Prev 19, 10221032.CrossRefGoogle ScholarPubMed
99. Ahearn, TU, McCullough, ML, Flanders, WD, et al. (2011) A randomized clinical trial of the effects of supplemental calcium and vitamin D3 on markers of their metabolism in normal mucosa of colorectal adenoma patients. Cancer Res 71, 413423.Google Scholar
100. Holick, MF (2007) Vitamin D deficiency. N Engl J Med 357, 266281.Google Scholar
101. Ross, AC, Manson, JE, Abrams, SA, et al. (2011) The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab 96, 5358.Google Scholar
102. Davis, CD & Dwyer, JT (2007) The ‘sunshine vitamin’: benefits beyond bone? J Natl Cancer Inst 99, 15631565.Google Scholar
103. Autier, P, Boniol, M, Pizot, C, et al. (2014) Vitamin D status and ill health: a systematic review. Lancet Diabetes Endocrinol 2, 7689.Google Scholar
104. Song, M, Wu, K, Chan, AT, et al. (2014) Plasma 25-hydroxyvitamin D and risk of colorectal cancer after adjusting for inflammatory markers. Cancer Epidemiol Biomarkers Prev 23, 21752180.Google Scholar
105. Hofmann, JN, Yu, K, Horst, RL, et al. (2010) Long-term variation in serum 25-hydroxyvitamin D concentration among participants in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Cancer Epidemiol Biomarkers Prev 19, 927931.Google Scholar
106. Grant, WB (2011) Effect of interval between serum draw and follow-up period on relative risk of cancer incidence with respect to 25-hydroxyvitamin D level: implications for meta-analyses and setting vitamin D guidelines. Dermatoendocrinol 3, 199204.CrossRefGoogle Scholar
107. Cheng, TY, Millen, AE, Wactawski-Wende, J, et al. (2014) Vitamin D intake determines vitamin d status of postmenopausal women, particularly those with limited sun exposure. J Nutr 144, 681689.Google Scholar
108. Bertrand, KA, Giovannucci, E, Liu, Y, et al. (2012) Determinants of plasma 25-hydroxyvitamin D and development of prediction models in three US cohorts. Br J Nutr 108, 18891896.Google Scholar
109. Jung, S, Qian, ZR, Yamauchi, M, et al. (2014) Predicted 25(OH)D score and colorectal cancer risk according to vitamin D receptor expression. Cancer Epidemiol Biomarkers Prev 23, 16281637.Google Scholar
110. Wactawski-Wende, J, Kotchen, JM, Anderson, GL, et al. (2006) Calcium plus vitamin D supplementation and the risk of colorectal cancer. N Engl J Med 354, 684696.Google Scholar
111. Wu, K, Feskanich, D, Fuchs, CS, et al. (2007) A nested case-control study of plasma 25-hydroxyvitamin D concentrations and risk of colorectal cancer. J Natl Cancer Inst 99, 11201129.Google Scholar
112. Otani, T, Iwasaki, M, Sasazuki, S, et al. (2007) Plasma vitamin D and risk of colorectal cancer: the Japan Public Health Center-Based Prospective Study. Br J Cancer 97, 446451.Google Scholar
113. Jenab, M, Bueno-de-Mesquita, HB, Ferrari, P, et al. (2010) Association between pre-diagnostic circulating vitamin D concentration and risk of colorectal cancer in European populations:a nested case-control study. BMJ 340, b5500.Google Scholar
114. Lee, JE, Li, H, Chan, AT, et al. (2011) Circulating levels of vitamin D and colon and rectal cancer: the Physicians’ Health Study and a Meta-analysis of Prospective Studies. Cancer Prev Res 4, 735743.Google Scholar
115. Ma, Y, Zhang, P, Wang, F, et al. (2011) Association between vitamin D and risk of colorectal cancer: a systematic review of prospective studies. J Clin Oncol 29, 37753782.Google Scholar
116. Chung, M, Lee, J, Terasawa, T, et al. (2011) Vitamin D with or without calcium supplementation for prevention of cancer and fractures: an updated meta-analysis for the U.S. Preventive Services Task Force. Ann Inter Med 155, 827838.Google Scholar
117. Neuhouser, ML, Manson, JE, Millen, A, et al. (2012) The influence of health and lifestyle characteristics on the relation of serum 25-hydroxyvitamin D with risk of colorectal and breast cancer in postmenopausal women. Am J Epidemiol 175, 673684.Google Scholar
118. English, DR, Williamson, EJ, Heath, AK, et al. (2013) Abstract A54: vitamin D and risk of colorectal cancer: the Melbourne Collaborative Cohort Study. Cancer Prev Res 6, A54.Google Scholar
119. Anic, GM, Weinstein, SJ, Mondul, AM, et al. (2014) Serum vitamin D, vitamin D binding protein, and risk of colorectal cancer. PLOS ONE 9, e102966.Google Scholar
120. Theodoratou, E, Tzoulaki, I, Zgaga, L, et al. (2014) Vitamin D and multiple health outcomes: umbrella review of systematic reviews and meta-analyses of observational studies and randomised trials. BMJ 348, g2035.Google Scholar
121. Weinstein, SJ, Purdue, MP, Smith-Warner, SA, et al. (2015) Serum 25-hydroxyvitamin D, vitamin D binding protein and risk of colorectal cancer in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. Int J Cancer 136, E654E664.Google Scholar
122. Song, M, Nishihara, R, Wang, M, et al. (2015) Plasma 25-hydroxyvitamin D and colorectal cancer risk according to tumour immunity status. Gut 65, 269304.Google Scholar
123. Lee, JE (2011) Circulating levels of vitamin D, vitamin D receptor polymorphisms, and colorectal adenoma: a meta-analysis. Nutr Res Pract 5, 464470.Google Scholar
124. Yin, L, Grandi, N, Raum, E, et al. (2011) Meta-analysis: serum vitamin D and colorectal adenoma risk. Preven Med 53, 1016.CrossRefGoogle ScholarPubMed
125. Ogino, S, Lochhead, P, Chan, AT, et al. (2013) Molecular pathological epidemiology of epigenetics: emerging integrative science to analyze environment, host, and disease. Mod Pathol 26, 465484.Google Scholar
126. Ogino, S, Campbell, PT, Nishihara, R, et al. (2015) Proceedings of the second international molecular pathological epidemiology (MPE) meeting. Cancer Causes Control 26, 959972.CrossRefGoogle ScholarPubMed
127. Wang, M, Kuchiba, A & Ogino, S (2015) A meta-regression method for studying etiological heterogeneity across disease subtypes classified by multiple biomarkers. Am J Epidemiol 182, 263270.Google Scholar
128. Wang, M, Spiegelman, D, Kuchiba, A, et al. (2015) Statistical methods for studying disease subtype heterogeneity. Stat Med 35, 782800.Google Scholar
129. Edfeldt, K, Liu, PT, Chun, R, et al. (2010) T-cell cytokines differentially control human monocyte antimicrobial responses by regulating vitamin D metabolism. Proc Natl Acad Sci U S A 107, 2259322598.Google Scholar
130. Liu, PT, Stenger, S, Li, H, et al. (2006) Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 311, 17701773.CrossRefGoogle ScholarPubMed
131. Sigmundsdottir, H, Pan, J, Debes, GF, et al. (2007) DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol 8, 285293.Google Scholar
132. Nishihara, R, VanderWeele, TJ, Shibuya, K, et al. (2015) Molecular pathological epidemiology gives clues to paradoxical findings. Eur J Epidemiol 30, 11291135.Google Scholar
133. Ng, K, Meyerhardt, JA, Wu, K, et al. (2008) Circulating 25-hydroxyvitamin D levels and survival in patients with colorectal cancer. J Clin Oncol 26, 29842991.CrossRefGoogle ScholarPubMed
134. Ng, K, Wolpin, BM, Meyerhardt, JA, et al. (2009) Prospective study of predictors of vitamin D status and survival in patients with colorectal cancer. Br J Cancer 101, 916923.Google Scholar
135. Ng, K, Sargent, DJ, Goldberg, RM, et al. (2011) Vitamin D status in patients with stage IV colorectal cancer: findings from Intergroup trial N9741. J Clin Oncol 29, 15991606.Google Scholar
136. Fedirko, V, Riboli, E, Tjonneland, A, et al. (2012) Prediagnostic 25-hydroxyvitamin D, VDR and CASR polymorphisms, and survival in patients with colorectal cancer in western European populations. Cancer Epidemiol Biomarkers Prev 21, 582593.Google Scholar
137. Zgaga, L, Theodoratou, E, Farrington, SM, et al. (2014) Plasma vitamin D concentration influences survival outcome after a diagnosis of colorectal cancer. J Clin Oncol 32, 24302439.Google Scholar
138. Maalmi, H, Ordonez-Mena, JM, Schottker, B, et al. (2014) Serum 25-hydroxyvitamin D levels and survival in colorectal and breast cancer patients: systematic review and meta-analysis of prospective cohort studies. Eur J Cancer 50, 15101521.Google Scholar
139. Ng, K, Venook, AP, Sato, K, et al. (2015) Vitamin D status and survival of metastatic colorectal cancer patients: results from CALGB/SWOG 80405 (alliance). J Clin Oncol 33, Suppl. 3, 507.Google Scholar
140. Martinez, ME, Giovannucci, EL, Colditz, GA, et al. (1996) Calcium, vitamin D, and the occurrence of colorectal cancer among women. J Natl Cancer Inst 88, 13751382.Google Scholar
141. Pritchard, RS, Baron, JA & deVerdier, MG (1996) Dietary calcium, vitamin D, and the risk of colorectal cancer in Stockholm, Sweden. Cancer Epidemiol Biomarkers Prev 5, 897900.Google Scholar
142. Terry, P, Baron, JA, Bergkvist, L, et al. (2002) Dietary calcium and vitamin D intake and risk of colorectal cancer: a prospective cohort study in women. Nutr Cancer 43, 3946.CrossRefGoogle ScholarPubMed
143. Slattery, ML, Neuhausen, SL, Hoffman, M, et al. (2004) Dietary calcium, vitamin D, VDR genotypes and colorectal cancer. Int J Cancer 111, 750756.Google Scholar
144. Park, SY, Murphy, SP, Wilkens, LR, et al. (2007) Calcium and vitamin D intake and risk of colorectal cancer: the Multiethnic Cohort Study. Am J Epidemiol 165, 784793.Google Scholar
145. Mizoue, T, Kimura, Y, Toyomura, K, et al. (2008) Calcium, dairy foods, vitamin D, and colorectal cancer risk: the Fukuoka Colorectal Cancer Study. Cancer Epidemiol Biomarkers Prev 17, 28002807.Google Scholar
146. Ishihara, J, Inoue, M, Iwasaki, M, et al. (2008) Dietary calcium, vitamin D, and the risk of colorectal cancer. Am J Clin Nutr 88, 15761583.Google Scholar
147. Lipworth, L, Bender, TJ, Rossi, M, et al. (2009) Dietary vitamin D intake and cancers of the colon and rectum: a case-control study in Italy. Nutr Cancer 61, 7075.Google Scholar
148. Huncharek, M, Muscat, J & Kupelnick, B (2009) Colorectal cancer risk and dietary intake of calcium, vitamin D, and dairy products: a meta-analysis of 26335 cases from 60 observational studies. Nutr Cancer 61, 4769.Google Scholar
149. Marcus, PM & Newcomb, PA (1998) The association of calcium and vitamin D, and colon and rectal cancer in Wisconsin women. Int J Epidemiol 27, 788793.Google Scholar
150. McCullough, ML, Robertson, AS, Rodriguez, C, et al. (2003) Calcium, vitamin D, dairy products, and risk of colorectal cancer in the Cancer Prevention Study II Nutrition Cohort (United States). Cancer Causes Control 14, 112.Google Scholar
151. Yang, B, McCullough, ML, Gapstur, SM, et al. (2014) Calcium, vitamin D, dairy products, and mortality among colorectal cancer survivors: the Cancer Prevention Study-II Nutrition Cohort. J Clin Oncol 32, 23352343.Google Scholar
152. Trivedi, DP, Doll, R & Khaw, KT (2003) Effect of four monthly oral vitamin D-3 (cholecalciferol) supplementation on fractures and mortality in men and women living in the community: randomised double blind controlled trial. BMJ 326, 469472.Google Scholar
153. Lappe, JM, Travers-Gustafson, D, Davies, KM, et al. (2007) Vitamin D and calcium supplementation reduces cancer risk: results of a randomized trial. Am J Clin Nutr 85, 15861591.Google Scholar
154. Avenell, A, MacLennan, GS, Jenkinson, DJ, et al. (2012) Long-term follow-up for mortality and cancer in a randomized placebo-controlled trial of vitamin D(3) and/or calcium (RECORD trial). J Clin Endocrinol Metab 97, 614622.Google Scholar
155. Ng, K, Scott, JB, Drake, BF, et al. (2014) Dose response to vitamin D supplementation in African Americans: results of a 4-arm, randomized, placebo-controlled trial. Am J Clin Nutr 99, 587598.Google Scholar
156. Bolland, MJ, Grey, A, Gamble, GD, et al. (2011) Calcium and vitamin D supplements and health outcomes: a reanalysis of the Women’s Health Initiative (WHI) limited-access data set. Am J Clin Nutr 94, 11441149.Google Scholar
157. Baron, JA, Barry, EL, Mott, LA, et al. (2015) A trial of calcium and vitamin D for the prevention of colorectal adenomas. N Engl J Med 373, 15191530.Google Scholar
158. Bischoff-Ferrari, HA, Giovannucci, E, Willett, WC, et al. (2006) Estimation of optimal serum concentrations of 25-hydroxyvitamin D for multiple health outcomes. Am J Clin Nutr 84, 1828.Google Scholar
159. Bischoff-Ferrari, HA, Shao, A, Dawson-Hughes, B, et al. (2010) Benefit-risk assessment of vitamin D supplementation. Osteoporos Int 21, 11211132.Google Scholar
160. Giovannucci, E (2013) Epidemiology of vitamin D and colorectal cancer. Anticancer Agents Med Chem 13, 1119.Google Scholar
161. Kunzmann, AT, Coleman, HG, Huang, WY, et al. (2015) Dietary fiber intake and risk of colorectal cancer and incident and recurrent adenoma in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Am J Clin Nutr 102, 881890.Google Scholar
162. Manson, JE & Bassuk, SS (2015) Vitamin D research and clinical practice: at a crossroads. JAMA 313, 13111312.Google Scholar
163. Pradhan, AD & Manson, JE (2015) Update on the Vitamin D and OmegA-3 trial (VITAL). J Steroid Biochem Mol Biol 155, 252256.Google Scholar
164. Lichtenstein, P, Holm, NV, Verkasalo, PK, et al. (2000) Environmental and heritable factors in the causation of cancer – analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med 343, 7885.Google Scholar
165. Shea, M, Benjamin, E, Dupuis, J, et al. (2009) Genetic and non-genetic correlates of vitamins K and D. Eur J Clin Nutr 63, 458464.Google Scholar
166. Wang, TJ, Zhang, F, Richards, JB, et al. (2010) Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet 376, 180188.Google Scholar
167. Hiraki, LT, Qu, C, Hutter, CM, et al. (2013) Genetic predictors of circulating 25-hydroxyvitamin d and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev 22, 20372046.Google Scholar
168. Jorde, R, Schirmer, H, Wilsgaard, T, et al. (2012) Polymorphisms related to the serum 25-hydroxyvitamin D level and risk of myocardial infarction, diabetes, cancer and mortality. The Tromso Study. PLOS ONE 7, e37295.Google Scholar
169. Peters, U, Bien, S & Zubair, N (2015) Genetic architecture of colorectal cancer. Gut 64, 16231636.Google Scholar
170. Koestner, K, Denzer, N, Mueller, CSL, et al. (2009) The relevance of vitamin D receptor (VDR) gene polymorphisms for cancer: a review of the literature. Anticancer Res 29, 35113536.Google Scholar
171. Touvier, M, Chan, DSM, Lau, R, et al. (2011) Meta-analyses of vitamin D intake, 25-hydroxyvitamin D status, vitamin D receptor polymorphisms, and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 20, 10031016.Google Scholar
172. Serrano, D, Gnagnarella, P, Raimondi, S, et al. (2015) Meta-analysis on vitamin D receptor and cancer risk: focus on the role of TaqI, ApaI, and Cdx2 polymorphisms. Eur J Cancer Prev 25, 8596.Google Scholar
173. Hiraki, LT, Joshi, AD, Ng, K, et al. (2014) Joint effects of colorectal cancer susceptibility loci, circulating 25-hydroxyvitamin D and risk of colorectal cancer. PLOS ONE 9, e92212.Google Scholar
174. Ferguson, LR, Laing, B, Marlow, G, et al. (2015) The role of vitamin D in reducing gastrointestinal disease risk and assessment of individual dietary intake needs: focus on genetic and genomic technologies. Mol Nutr Food Res 60, 119133.Google Scholar
Figure 0

Fig. 1 The metabolism of vitamin D in human body. Vitamin D that is taken up via the diet, or synthesised from 7-dehydrocholesterol by the skin following UV exposure, binds to vitamin D-binding protein (DBP) in the circulation and is transported to the liver. Vitamin D is hydroxylated at C25 by CYP2R1 in the liver to 25-hydroxyvitamin D (25(OH)D), the major circulating form of vitamin D in the human body. In the kidney and some extrarenal sites, 25(OH)D is further hydroxylated at C1 by CYP27B1 into 1,25-dihydroxyvitamin D3 (1,25(OH)2D) (calcitriol), the bioactive form. Both 25(OH)D and 1,25(OH)2D are deactivated by CYP24A1 through additional hydroxylation at C24. Both CYP27B1 and CYP24A1 are regulated by calcitriol.

Figure 1

Fig. 2 The mechanism of calcitriol (1,25(OH)2D) action through vitamin D receptor (VDR). Calcitriol binds and activates nuclear VDR, which then dimerises with retinoid X receptor (RXR). (a) Transcriptional activation involves the VDR–RXR heterodimer binding with vitamin D response element (VDRE) and recruitment of histone acetyltransferase co-activator. (b) Transcriptional depression involves VDR–RXR binding with negative VDRE (nVDRE) and recruitment of histone deacetylase co-repressor. RNA POL II, RNA polymerase II.

Figure 2

Table 1 Major studies (no. of cases ≥300) investigating plasma 25-hydroxyvitamin D (25(OH)D) concentrations and incidence of colorectal cancer (Odds ratios, hazard ratios (HR) and 95 % confidence intervals)

Figure 3

Table 2 Major studies (no. of cases ≥300) investigating plasma 25-hydroxyvitamin D (25(OH)D) concentrations and survival of patients with diagnosed colorectal cancer (CRC) (Hazard ratios (HR) and 95 % confidence intervals)

Figure 4

Table 3 Major studies (no. of cases ≥500) investigating vitamin D intake and incidence of colorectal cancer (CRC) (Odds ratios, relative risks (RR) and 95 % confidence intervals)

Figure 5

Table 4 Major completed randomised trials (n ≥1000) investigating vitamin D supplementation and cancer (Hazard ratios (HR) and 95 % confidence intervals)

Figure 6

Table 5 Major ongoing randomised trials (n ≥1000) investigating vitamin D supplementation and cancer