Vitamin D signalling pathways in cancer: potential for anticancer therapeutics

Page created by Harvey Wolf
 
CONTINUE READING
REVIEWS

                                  Vitamin D signalling pathways
                                  in cancer: potential for anticancer
                                  therapeutics
                                  Kristin K. Deeb*, Donald L. Trump‡ and Candace S. Johnson*
                                  Abstract | Epidemiological studies indicate that vitamin D insufficiency could have an
                                  aetiological role in various human cancers. Preclinical research indicates that the active
                                  metabolite of vitamin D, 1α,25(OH)2D3, also known as calcitriol, or vitamin D analogues might
                                  have potential as anticancer agents because their administration has antiproliferative
                                  effects, can activate apoptotic pathways and inhibit angiogenesis. In addition, 1α,25(OH)2D3
                                  potentiates the anticancer effects of many cytotoxic and antiproliferative anticancer agents.
                                  Here, we outline the epidemiological, preclinical and clinical studies that support the
                                  development of 1α,25(OH)2D3 and vitamin D analogues as preventative and therapeutic
                                  anticancer agents.

                                 The most widely accepted physiological role of vitamin            The seminal finding by Garland and Garland12 of
                                 D, which is mediated primarily by 1α,25(OH)2D3 (also          higher mortality rates from colon cancer in the northeast
                                 known as calcitriol), the most active product of vitamin      and lower rates in the south, southwest and west in the
                                 D synthesis, is in the physiological regulation of Ca2+ and   United States led to the important concept that exposure
                                 Pi transport and bone mineralization1. The importance of      to ultraviolet B or sunlight, which leads to vitamin D
                                 this role is demonstrated by studies with knockout mice       synthesis, can reduce the risk of colorectal cancer. Several
                                 deficient in key members of the vitamin D metabolic           epidemiological observations have shown an associa-
                                 pathway, such as 25-hydroxyvitamin D3‑1α-hydroxylase          tion between low serum 25(OH)D3 levels (the accepted
                                 (1α-OHase; encoded by Cyp27b1), an enzyme that gener-         measure of vitamin D body stores) and increased risk for
                                 ates 1α,25(OH)2D3, and 25-hydroxyvitamin D 24-hydrox-         colorectal13, breast14 and prostate15 cancers. There are
                                 ylase (24-OHase; encoded by Cyp24a1), the enzyme that         many epidemiological studies that have sought to
                                 degrades 1α,25(OH)2D3 (catabolism), and the vitamin D         determine associations between vitamin D status and
                                 receptor (Vdr), which binds 1α,25(OH)2D3 to affect target     the risk and mortality rates of a number of cancers16–19.
                                 gene transcription2–6 (TABLEs 1,2). Loss of these genes in    Giovannucci and colleagues16 recently performed an
                                 mice led to phenotypes with abnormal bone morphol-            extensive analysis that combined major determinants
                                 ogy. However, recent observations indicate a much             of vitamin D status on cancer risk and mortality with
                                 broader range of action for 1α,25(OH)2D3, including the       51,529 men that were enrolled in the Health Professionals
                                 regulation of differentiation, proliferation and apoptosis.   Follow-up Study (HPFS). Individuals were prospectively
                                 Furthermore, altered expression and function of proteins      followed for almost 20 years; diet, exercise and lifestyle
                                 crucial in vitamin D synthesis and catabolism have been       characteristics were analysed and health outcomes,
                                 observed in many tumour types (TABLE 3). Interestingly,       including cancer and cancer-related deaths, were
                                 Vdr–/– mice show hyperproliferation and increased mitotic     assessed. Giovannucci et al. developed a model to pre-
                                 activity in the descending colon, suggesting a role for       dict 25(OH)D3 levels based on the relationship between
Departments of
Pharmacology and                 1α,25(OH)2D3-mediated signalling in tumour suppres-           dietary and supplementary vitamin D, physical activity,
Therapeutics* and Medicine‡,     sion7. Zinser and colleagues8–10 showed that Vdr ablation     body mass index and sunlight exposure (a source of vita-
Roswell Park Cancer Institute,   in the mouse increased chemical carcinogenesis in mam-        min D). The model was applied to 47,800 individuals in
Buffalo, New York, USA.          mary, epidermis and lymphoid tissues but not in ovary,        the HPFS, and the analysis indicated a strong association
Correspondence to C.S.J.
e-mail: candace.johnson@
                                 uterus, lung or liver (TABLE 2). However, mice deficient in   between low levels of predicted 25(OH)D3 and increased
roswellpark.org                  key members of the vitamin D synthesis and catabolic          cancer incidence and cancer-related mortality, particu-
doi:10.1038/nrc2196              pathways do not develop spontaneous cancer2,3,11.             larly for cancers of the digestive system16. Furthermore,

684 | SEPTEMBER 2007 | volume 7                                                                                          www.nature.com/reviews/cancer
                                                         © 2007 Nature Publishing Group
REVIEWS

 At a glance                                                                                       resultant 25-hydroxycholecalciferol (25(OH)D3) is
                                                                                                   1α-hydroxylated in the kidney by mitochondrial
 • Epidemiological studies point to a relationship between vitamin D deficiency and                1α-hydroxylase (1α-OHase; encoded by the gene
   cancer risk.                                                                                    CYP27B1), this yields the hormonally active secosteroid
 • Alterations in vitamin D receptor expression, and in the synthesis (25-hydroxylase              1α,25(OH) 2D 3 (calcitriol) 23. 24-hydroxylation of
   and 1α-hydroxylase) and catabolism (24-hydroxylase) of vitamin D metabolites are                25(OH)D 3 and 1α,25(OH) 2D 3 by the cytochrome
   involved in the growth regulation of tumours; thus, compromising 1α,25(OH)2D3                   P450 enzyme 25-hydroxyvitamin D 24-hydroxylase
   (also known as calcitriol; the active metabolite of vitamin D signalling) sensitivity
                                                                                                   (24-OHase; encoded by the gene CYP24A1), to the
   and 1α,25(OH)2D3 signalling.
                                                                                                   metabolites 24,25(OH) 2D 3 and 1α,24,25(OH) 2D 3,
 • The antiproliferative effects of 1α,25(OH)2D3 have been demonstrated in various                 respectively, is the rate-limiting step for 25(OH)D 3
   tumour types, as determined by preclinical trials.
                                                                                                   and 1α,25(OH) 2 D 3 catabolism 23 . Additionally,
 • The anti-tumour effects of 1α,25(OH)2D3 involve mechanisms that are associated                  1α,25(OH)2D3 concentrations are feedback regulated:
   with G0/G1 arrest, differentiation, induction of apoptosis and modulating
                                                                                                   an increase in 24,25(OH)2D3 induces the synthesis of
   different signalling pathways in tumour cells, as well as inhibiting tumour
   angiogenesis.
                                                                                                   1α,25(OH)2D3; whereas Ca2+, Pi and 1α,25(OH)2D3
                                                                                                   itself suppress 1α,25(OH)2D3 synthesis23–26. CYP27B1
 • Glucocorticoids potentiate the anti-tumour effects of 1α,25(OH)2D3 and decrease
                                                                                                   (which encodes 1α-OHase) expression is induced
   1α,25(OH)2D3-induced hypercalcemia. 1α,25(OH)2D3 also potentiates the anti-
   tumour effects of many chemotherapeutic agents such as platinum analogues,
                                                                                                   by parathyroid hormone (PTH) 25 and repressed
   taxanes and DNA-intercalating agents.                                                           by 1α,25(OH) 2 D 3 24,27 . Furthermore, CYP24A1 is
                                                                                                   strongly induced by 1α,25(OH)2D3 to produce the less
 • Given that the major vitamin D catabolizing enzyme, CYP24A1 (24-hydroxylase), is
   often amplified and overexpressed in tumour cells, agents that inhibit this enzyme              active vitamin D metabolites 1α,24,25(OH)2D3 and
   can potentiate 1α,25(OH)2D3 anti-tumour effects.                                                24,25(OH)2D323.
 • Preclinical data indicate that maximal anti-tumour effects are seen with
                                                                                                       There are instances of tissue-specific regulation
   pharmacological doses of 1α,25(OH)2D3, and can be safely achieved in animals                    of the vitamin D synthetic enzymes. 1α,25(OH)2D3
   using a high-dose, intermittent schedule of administration. Some clinical trial data            functions in an autocrine and paracrine manner to
   indicates that 1α,25(OH)2D3 is well-tolerated in cancer patients within a proper                modulate vitamin D function and signalling. 1α-
   dosing schedule.                                                                                OHase is expressed at extra-renal sites such as nor-
 • Data support the hypothesis that vitamin D compounds may have an important                      mal colon, brain, placenta, pancreas, lymph nodes
   role in cancer therapy and prevention, and merit further investigation.                         and skin 28, allowing local conversion of 25(OH)D 3
                                                                                                   to 1α,25(OH)2D3. Importantly, increased CYP27B1
                                                                                                   expression is observed in breast29 and prostate30 can-
                                    Giovannucci et al. reported that an increase of 25 nmol        cers and during early colon tumour progression in
                                    per L in predicted 25(OH)D3 level is associated with a         well-to-moderately differentiated states, but decreased
                                    29% reduction in cancer-related mortality and a 17%            in poorly differentiated colon carcinomas 31–33 .
                                    reduction in cancer incidence, suggesting that high            Increased expression of CYP27B1 in cancer tis-
                                    25(OH)D3 levels might be associated with a decreased risk      sues could provide local conversion of 25(OH)D 3
                                    of some cancers16. A meta-analysis of case–control and         to 1α,25(OH)2D3, and may support the notion that
                                    cohort studies found that individuals with ≥ 33 ng per ml      25(OH)D3 and 1α,25(OH)2D3 might have a role in the
                                    (82 nmol per L) 25(OH)D3 had a 50% lower incidence of          chemoprevention of these cancers. However, CYP24A1
                                    colorectal cancer20. Additionally, patients with early stage   (encoding 24-OHase) mRNA expression is upregu-
                                    non-small-cell lung cancer with high 25(OH)D3 levels           lated in tumours, and may counteract 1α,25(OH)2D3
                                    and high vitamin D intake at the time of diagnosis and         antiproliferative activity, presumably by decreasing
                                    initiation of treatment had improved overall and recur-        1α,25(OH)2D3 levels34,35 (TABLE 3). Cross et al.35 have
                                    rence-free survival21. Therefore, these data suggest that      demonstrated that the upregulation of CYP24A1 and
                                    low levels of 25(OH)D3 are an important risk factor for        downregulation of CYP27B1 can occur in high-grade
                                    cancer incidence.                                              colon carcinomas. The chromosomal region 20q13.2,
                                                                                                   containing the CYP24A1 gene, is amplified in human
                                    Synthesis and catabolism of vitamin D                          breast tumours36, and CYP24A1 mRNA expression
                                    1α,25(OH) 2D 3 is synthesized from vitamin D in a              is upregulated in samples from human lung, colon
Secosteroid hormones                highly regulated multistep process (FIG. 1). The first         and ovarian tumours, suggesting that 1α,25(OH)2D3
Molecules that are very similar
in structure to steroids but with
                                    step in vitamin D synthesis is the formation of vita-          levels would be reduced in these cases35,37,38 (TABLE 3).
a ‘broken’ ring; two of the         min D3 in the skin through the action of ultraviolet           This suggests that inhibition of CYP24A1 expression
B‑ring carbon atoms (C-9 and        irradiation; vitamin D3 can also be taken in the diet          and activity is essential for prevention to be effective.
10) of the four steroid rings are   but in North America and Europe dietary vitamin D3             Small-molecule inhibitors with varying specificity for
not joined.
                                    intake is a minor component of vitamin D3 acquisi-             24-OHase39–42 render tumour cells more sensitive to the
Autocrine                           tion because dairy products, eggs, fish and fortified          action of 1α,25(OH)2D3 and its analogues. Consistent
A substance secreted by a cell      foods contain only small quantities of vitamin D22.            with the epidemiological studies discussed above, these
that acts on the surface            Decreased sun exposure further limits vitamin D                findings indicate that 1α,25(OH)2D3 catabolism could
receptors of the same cell.         synthesis.                                                     modulate tumour growth in some tissues, indicat-
Paracrine
                                        Vitamin D 3 (cholecalciferol) is hydroxylated by           ing the potential for the development of 24-OHase
A substance secreted by a cell      liver mitochondrial and microsomal 25-hydroxylases             inhibitors as cancer preventative and/or anticancer
that acts on adjacent cells.        (25-OHase) 23, encoded by the gene CYP27A1. The                therapeutic agents.

nature reviews | cancer                                                                                                 volume 7 | SEPTEMBER 2007 | 685
                                                             © 2007 Nature Publishing Group
REVIEWS

 Table 1 | Mouse models of vitamin D metabolic enzymes and receptor signalling
 Genetic modification          Mouse phenotype                                                                  Cancer phenotype                      Refs
 Cyp27b1 (which
          –/–
                               Pseudo-vitamin D deficiency rickets (PDDR); decreased serum Ca and  2+
                                                                                                                None                                    2,3
 encodes 1α-OHase)             Pi; secondary hyperthyroidism; undetectable 1α,25(OH)2D3 (calcitriol)
                               levels; disorganized growth plate structure and osteomalacia. In addition:
                               infertile females; uterine hypoplasia; decreased ovarian size; compromised
                               folliculogenesis; reduction in CD4+ and CD8+ peripheral T lymphocyte
 Cyp24a1–/– (24-OHase)         Lethal hypercalcemia; impaired intramembranous bone mineralization               None                                     11
 Vdr (VDR signalling)
    –/–
                               Vitamin D deficiency rickets type II (VDDR II) and osteomalacia; alopecia,       Hyperproliferation of descending     4,5,6,7
                               hypocalcemia, hyperparathyroidism; impaired bone formation, growth               colon (increased PCNA positivity
                               retardation; female infertility, uterine hypoplasia, impaired folliculogenesis   and cyclin D1 expression)
 PCNA, proliferating cell nuclear antigen.

                                1α,25(OH)2D3-mediated transcription of target genes.            in a transcriptionally repressed state48. Conformational
                                1α,25(OH)2D3 exerts transcriptional activation and              change also repositions the VDR activation function
                                repression of target genes by binding to the VDR (BOX 1).       2 (AF2) domains to bind to stimulatory coactivators,
                                The VDR is a member of the steroid hormone receptor             consisting of the steroid receptor coactivators (SRCs),
                                superfamily and regulates gene expression in a ligand-          nuclear coactivator 62 kDa–SKI-interacting protein
                                dependent manner43 (FIG. 2). Interestingly, N‑terminal VDR      (NCoA62–SKIP) and the chromatin modifiers, CREB
                                variants show tissue-specific expression44,45 that might also   binding protein (CBP)–p300 and PBAF (polybromo- and
                                contribute to the differential specificity of 1α,25(OH)2D3-     SWI‑2-related gene 1 associated factor), which acetylates
                                mediated regulation. 1α,25(OH)2D3–VDR-dependent                 histones in the nucleosomes to unravel DNA for transcrip-
                                transcriptional activity is modulated through synergistic       tion49. Once the chromatin is de-repressed, the vitamin
                                ligand-binding and dimerization with retinoic X receptor        D receptor-interacting proteins (DRIPs) form a complex
                                (RXR). The activated 1α,25(OH)2D3–VDR–RXR com-                  that binds to the AF2 domain of VDR and interacts with
                                plex specifically binds to vitamin D response elements          the transcription machinery, such as TF2B (transcription
                                (VDREs), composed of two hexanucleotide repeats inter-          factor 2B) and RNA polymerase II, and initiates transcrip-
                                spaced by varying numbers of nucleotides (for example,          tion (for a review see REF. 23). Recently, it has been shown
                                GGTCCA-NNN-GGTCCA, where N is any nucleotide;                   that epigenetic regulation of VDR through increased
                                this is denoted DR3), in the promoter regions of target         expression of NCoR1 and SMRT repress VDR-mediated
                                genes46. For transcriptional activation, VDR occupies the 3′    signalling in prostate50 and breast cancer51 cell lines, and
                                half-site whereas RXR binds the 5′ half-site of VDRE47.         may have a role in mediating the antiproliferative effects
                                    Co-factor proteins also have the ability to modulate        of 1α,25(OH)2D3 in these tissues.
                                VDR-mediated gene expression; these proteins possess                The mechanism by which 1α,25(OH)2D3 represses
                                intrinsic chromatin-modifying enzymatic activities, act         gene expression through the binding of VDR to negative
                                as a platform for the recruitment of chromatin-modifying        VDREs (DR3-type), placing VDR on the 5′ half-site of
                                proteins and recruit basal transcription factors to the pro-    the VDRE52,53, such as is the case with human PTH, may
                                moters23. 1α,25(OH)2D3 binding induces phosphoryla-             involve interference with transcriptional machinery but
                                tion and conformational changes in VDR, which causes            is less understood. Recently, transcriptional repression
                                the release of co-repressors (such as nuclear receptor          by 1α,25(OH)2D3 has been further elucidated for the
                                co-repressors (NCoRs) and the silencing mediator for            human CYP27B1 (REFS 27,54,55) and PTH56 genes. The
                                retinoid and thyroid hormone receptors (SMRT)–histone           VDR–RXR heterodimer represses gene transcription in
                                deacetylase (HDAC) complex) that maintain chromatin             a 1α,25(OH)2D3-dependent manner through E‑box-type

                                 Table 2 | Vdr knockout mice and carcinogenesis
                                 Oncogene/                Tissue                 Cancer phenotype                                                      Ref
                                 carcinogen
                                 MPA plus DMBA            Skin                   40% sebaceous, 25% squamous and 15% follicular papillomas                9
                                 carcinogens                                     compared with WT littermates; other infrequent lesions include
                                                                                 basal cell carcinoma and haemangioma
                                                          Mammary                Higher incidence of alveolar and ductal hyperplasias in                  8
                                                                                 Vdr–/– mice compared with WT mice; development of palpable
                                                                                 mammary tumours was not altered by Vdr ablation
                                                          Lymph nodes and/       Lymphoblastic and thymic lymphoma higher in Vdr–/– (27%)                 8
                                                          or thymus              compared with WT mice (11%)
                                 Vdr–/– ;neu oncogene     Mammary                Decreased survival of Vdr–/–; neu mice compared with their              10
                                                                                 Vdr+/+; neu and Vdr+/–; neu littermates; increased development of
                                                                                 mammary tumours driven by the neu oncogene
                                 DMBA, 7,12-dimethylbenzanthracene; MPA, medroxyprogesterone acetate; WT, wild-type.

686 | SEPTEMBER 2007 | volume 7                                                                                            www.nature.com/reviews/cancer
                                                         © 2007 Nature Publishing Group
REVIEWS

 Table 3 | Expression of molecules that function in vitamin D metabolism and signalling in human cancers.
Protein                  Altered expression observed          Prognostic or histological              Types of cancer
(gene)                   in human cancer tissues              observations
Vitamin D metabolic enzymes
25-OHase                 Increased mRNA                       NA                                      Breast34, cervical34 and ovarian cancer34, HCC172
(CYP27A1)
1α-OHase                 Increased mRNA                       NA                                      Basal cell carcinoma173, breast34,174, cervical34
(CYP27B1)                                                                                             and ovarian cancer34
                         Increased mRNA                       Moderately differentiated               Colon cancer31,35,175
                         Decreased mRNA                       Poorly differentiated                   Colon cancer35
                         Splice variants (Hyd‑V5, ‑V6, ‑V7    NA                                      Glioblastoma multiforme176, melanoma177,
                         and ‑V8)                                                                     cervical cancer177
                         Immunoreactivity                     NA                                      Pancreatic132, breast132 and colon cancer 33,
                                                                                                      renal cell carcinoma132
                         Increased immunoreactivity           Moderately differentiated               Colon cancer33,35
                         Decreased immunoreactivity           Poorly differentiated                   Colon cancer33,35
24-OHase                 Amplified at 20q13.2 locus           NA                                      Gastric adenocarcinoma37, breast cancer36
(CYP24A1)
                         Increased mRNA                       NA                                      Basal cell carcinoma173, SCC (cutaneous)178,
                                                                                                      lung38,42, breast34,36, colon35,38, cervical34 and
                                                                                                      ovarian cancer34,38
                         Increased mRNA                       Poor prognosis                          Oesophageal cancer179
                         Decreased mRNA                       NA                                      Breast cancer38
                         Increased mRNA and activity          Poorly differentiated                   Colon cancer32
                         Increased protein                    NA                                      Lung cancer (NSCLC)42
Vitamin D receptor
VDR                      Increased mRNA                       NA                                      Basal cell carcinoma173, SCC (cutaneous)178,
(VDR)                                                                                                 colon cancer31
                         Decreased mRNA                       Poorly differentiated                   Colon cancer31
                         Increased immunoreactivity           NA                                      Breast34, cervical34 and ovarian cancer34
                         Increased, predominantly             Well differentiated                     Colon cancer166
                         cytoplasmic
                         Decreased immunoreactivity           Moderately and poorly                   Colon cancer166
                                                              differentiated
                                                              Poorly differentiated                   Colon cancer35
 HCC, hepatocellular carcinoma; NA, non applicable; NSCLC, non-small cell lung carcinoma; SCC, squamous cell carcinoma.

                              negative VDREs, comprised of a CANNTG-like motif                  control of 1α,25(OH)2D3 biosynthesis55, as well as negative
                              in the promoter regions of the CYP27B1 (REFS 27,54,55)            regulation of other genes with nVDREs in their promot-
                              and PTH56 genes, which are distinct from the DR3-                 ers. Furthermore, Kim et al.57 demonstrated that not only
                              type response elements. VDR-interacting repressor                 is histone deacetylation crucial for chromatin structure
                              (VDIR), when bound to E‑box-type elements, induces                remodelling in suppression of the CYP27B1 gene, but
                              the transcriptional activation of CYP27B154. However,             that transrepression by VDR requires DNA methylation
                              the binding of 1α,25(OH)2D3 to VDR causes VDR to                  of the CYP27B1 gene promoter, suggesting complicated
                              interact with VDIR. 1α,25(OH)2D3-induced association              epigenetic modifications for transcriptional regulation
                              between VDR and VDIR induces dissociation of the                  of the CYP27B1 gene. Epigenetic regulation of CYP27B1
                              histone acetyltransferase (HAT) co-activator and recruit-         and CYP24A1 has been previously reported for the PNT‑2
                              ment of HDAC co-repressor for 1α,25(OH)2D3-induced                human normal prostate cells and DU‑145 prostate cancer
                              transrepression of CYP27B1 gene expression54. In addi-            cell line58. Histone methylation and demethylation are cru-
                              tion, Williams syndrome transcription factor (WSTF)               cial events that impose ligand- and signal-dependent gene
                              potentiates 1α,25(OH)2D3-induced transrepression by               activation by nuclear receptors and prevent the recruit-
                              VDR of the CYP27B1 gene promoter by facilitating the              ment of unliganded nuclear receptors and transcription
                              association between WINAC, a multifunctional, ATP-                factors from binding to their target promoters and causing
                              dependent chromatin-remodelling complex, and chro-                constitutive gene activation59.
                              matin55. This transrepression mechanism is an important               Examples of genes with DR3-type response elements
                              biological function of VDR to allow negative-feedback             that are transcriptionally activated by 1α,25(OH)2D3

nature reviews | cancer                                                                                                   volume 7 | SEPTEMBER 2007 | 687
                                                        © 2007 Nature Publishing Group
REVIEWS

                                                        UV-B

                                                                                   D3
                                                                                        DBP

                                   7-dehydrocholesterol          D3                       D3
                                                                                                                  Circulation
                                           Pre-D3

                                             D3
                                                     Skin
                                                                                                                                                         Intestine
                                                                                               Liver
                                              Pi, Ca2+ and            25-OHase
                                              other factors                                                                                                          D3
                                                                                               24-OHase
                                                                               25(OH)D3
                                                                   +/–
                                                                                                        24,25(OH)2D3

                          Parathyroid             PTH                 Kidney                     +        Excretion
                          glands
                                                                                                       1α,24,25(OH)2D3

                                                                                   1α-OHase
                                                                                                                                                      Dietary sources
                                                                                                24-OHase                                              of vitamin D
                                                                          1α,25(OH)2D3
                                        Intestine                                                             Tumour
                                        Increases                                                             microenvironment
                                        absorption                                                            • Inhibits proliferation
                                        of Ca2+                Bone                 Immune cells              • Induces differentiation
                                        and Pi                 Increases bone       Induces                   • Inhibits angiogenesis
                                                               mineralization       differentiation

                         Figure 1 | Vitamin D metabolism. Photochemical synthesis of vitamin D3 (cholecalciferol, D3) occurs cutaneously
                         where pro-vitamin D3 (7-dehydrocholesterol) is converted to pre-vitamin D3 (pre‑D3) in response to ultraviolet B
                                                                                                                           Nature
                         (sunlight) exposure. Vitamin D3, obtained from the isomerization of pre-vitamin D3 in the epidermal       Reviews
                                                                                                                                basal layers| Cancer
                                                                                                                                              or
                         intestinal absorption of natural and fortified foods and supplements, binds to vitamin D‑binding protein (DBP) in the
                         bloodstream, and is transported to the liver. D3 is hydroxylated by liver 25-hydroxylases (25-OHase). The resultant
                         25‑hydroxycholecalciferol (25(OH)D3) is 1α-hydroxylated in the kidney by 25-hydroxyvitamin D3‑1α-hydroxylase
                         (1α‑OHase). This yields the active secosteroid 1α,25(OH)2D3 (calcitriol), which has different effects on various target
                         tissues23. The synthesis of 1α,25(OH)2D3 from 25(OH)D3 is stimulated by parathyroid hormone (PTH) and suppressed by
                         Ca2+, Pi and 1α,25(OH)2D3 itself. The rate-limiting step in catabolism is the degradation of 25(OH)D3 and 1α,25(OH)2D3
                         to 24,25(OH)D3 and 1α,24,25(OH)2D3, respectively, which occurs through 24-hydroxylation by 25-hydroxyvitamin D 24-
                         hydroxylase (24-OHase), encoded by the CYP24A1 gene. 24,25(OH)D3 and 1α,24,25(OH)2D3 are consequently excreted.
                         The main effects of 1α,25(OH)2D3 on various target tissues are highlighted above.

                         consist of CYP24A1 (REF. 60) (encoding 24-OHase),                              Nongenomic action of 1α,25(OH)2D3. Nongenomic
                         BGLAP61 (osteocalcin; expressed in bone osteoblasts),                          actions mediated by 1α,25(OH)2D3 are rapid and not
                         and CDKN1A62 (which encodes the cyclin depend-                                 dependent on transcription. However, nongenomic
                         ent kinase (CDK) inhibitor p21). Those repressed                               signalling may indirectly affect transcription through
                         by 1α,25(OH)2D3 include PTH53. Although VDREs                                  cross-talk with other signalling pathways68,69. Although
                         are traditionally thought to occur in the promoter                             there is no agreement on how the nongenomic actions
                         regions of the target genes, a DR3-type VDRE was                               are initiated, data suggest that these effects begin at the
                         recently identified in exon 4 of the growth arrest                             plasma membrane and involve a non-classical membrane
                         and DNA-damage-inducible (GADD45) gene 63 .                                    receptor (memVDR; FIG. 2) described in intestinal caveo-
                         1α,25(OH)2D3-mediated repression or activation of                              lae70, and a 1α,25(OH)2D3-membrane-associated rapid-
                         many proto-oncogenes or tumour-suppressor genes                                response steroid binding protein (1α,25D3-MARRS)
                         is described in normal and tumour tissues 62,64–67;                            isolated from chick intestinal basal-lateral membrane71.
                         however, only a few such genes contain VDREs in the                                The most well-described nongenomic effect of
                         promoter regions and are under the direct transcrip-                           1α,25(OH)2D3 is the rapid intestinal absorption of Ca2+
                         tional control of 1α,25(OH)2D3, such as CDKN1A62                               (REF. 72). Binding of 1α,25(OH)2D3 to the proposed mem-
                         and CCNC (which encodes cyclin C, containing a                                 brane receptor can result in the activation of numerous
                         DR4-type VDRE)65. This suggests that 1α,25(OH)2D3                              signalling cascades68,69 (FIG. 2). Activation of these signalling
                         exerts many of its effects indirectly by modulating                            cascades, such as protein kinase C (PKC), can result in the
                         signalling cascades or by unknown nongenomic                                   rapid opening of voltage-gated Ca2+ channels and an increase
                         mechanisms (FIGS 2,3).                                                         in intracellular Ca2+ (REF. 73), which may subsequently

688 | SEPTEMBER 2007 | volume 7                                                                                                      www.nature.com/reviews/cancer
                                                     © 2007 Nature Publishing Group
REVIEWS

                           Box 1 | The vitamin D receptor
                           The human VDR gene (which encodes the vitamin D receptor), located on chromosome 12q, is composed of promoter and
                           regulatory regions (1a–1f) and exons 2–9, which encode 6 domains (A – F) of the full length VDR protein (see figure)23. VDR
                           nuclear localization signals (blue) direct the receptor into the nucleus155,156 along microtubule tracts to the nuclear pores157.
                           Upon 1α,25(OH)2D3 binding to the hormone ligand-binding domain (red), VDR is stabilized by the phosphorylation of
                           serine 51 in the DNA-binding domain (green) by protein kinase C76, and serine 208 in the hinge region by casein kinase II158.
                           VDR associates with the retinoic acid receptor (RXR) through the dimerization domains (yellow). The 1α,25(OH)2D3–VDR–
                           RXR complex binds to the vitamin D response elements (VDREs) through the DNA-binding domain in the promoters of
                           target genes. Conformational change in the VDR results in the dissociation of the co-repressor, silencing mediator for
                           retinoid and thyroid hormone receptors (SMRT), and allows interaction of the VDR activation function 2 (AF2)
                           transactivation domain (light grey) with stimulatory coactivators, such as steroid receptor coactivators (SRCs), vitamin D
                           receptor-interacting proteins complex and nuclear coactivator‑62 kDa–Ski-interacting protein (NCoA62–SKIP)23 that
                           mediate transcriptional activation.
                             Non-synonymous (FokI) and synonymous (BsmI, ApaI, TaqI and Tru9I) single-nucleotide polymorphisms (SNPs) have
                           been identified in VDR (defined by restriction enzymes, polymorphisms are indicated in parentheses). FokI
                           polymorphism at translation initiation codon results in a smaller VDR that interacts with transcription factor 2B (TF2B)
                           more efficiently and has greater transcriptional activity than the full length VDR159. Although the functional effects of
                           these SNPs remain unknown, they have been reported to be associated with increased susceptibility to primary and
                           metastatic breast cancer17, squamous cell carcinoma160, colorectal cancer161,162, and prostate cancer163,164, but may be
                           protective against head and neck cancer165.
                             The expression of VDR is an important determinant of the tumour cell response to 1α,25(OH)2D3. The VDR is
                           overexpressed or repressed in several histological types of cancer (TABLE 3), demonstrating tissue-type variations in
                           1α,25(OH)2D3 signalling (supplemental information S1 (table)). VDR expression increases in hyperplastic polyps and in
                           the early stages of tumorigenesis, but declines in late-stage poorly differentiated tumours and is absent in associated
                           metastases. Tumours of the colon with the highest expression of VDR were most responsive to 1α,25(OH)2D3
                           treatment85,166. However, downregulation of the VDR in colon cancer cells through the transcription factor SNA1L167
                           reduces the anticancer effect of the vitamin D analogue EB1089.
                                     VDR gene
                                       Chromosome 12
                                                       1f   1e 1a 1d 1b                     1c     2              3              45 6            7 8       9
                                             q13–14                                                                                                                   ~75 kb
                                                                                                                               Bsml (A60890G)
                                                                                                 Fokl (C27823T)                Tru9l (G61050A)
                                                                                                                               Apal (G61888T)          Taql (T61938C)
                                DNA binding
                                (aa 24–90, 91–115)                            VDR protein
                                Nuclear localization                                 S51                              S208
                                (aa 49–55, 79–105)                                    P                                 P
                                                                                                                                                               AF-2
                                Hormone ligand binding              N                                  Hinge region                                                   48 kDa
                                (aa 227–244, 268–316, 396–422)
                                                                          1     24   49 91 115                               227244268 317             396 422 427 aa
                                Dimerization
                                (aa 37, 91–92, 244–263, 317–395)              A/B    C                     D                             E/F
                                Transactivation
                                (aa 246, 416–422)

                                                                                                                                                 Nature Reviews | Cancer
                          activate the Raf–mitogen-activated protein kinase extracel-                  Anti-tumour effects of 1α,25(OH)2D3 signalling
                          lular signal-regulated kinase kinase (MEK)–mitogen-acti-                     1α,25(OH)2D3 has been examined preclinically for its
                          vated protein kinase (MAPK)–extracellular signal-regulated                   therapeutic efficacy in chemopreventive and anticancer
                          kinase (ERK) cascade in skeletal muscle cells74. Activation                  activity. A chemoprevention study used Nkx3‑1;Pten
                          of the Raf–MEK–MAPK–ERK cascade, which mediates                              mutant mice to recapitulate prostate carcinogenesis,
                          proliferative cellular effects, may be a response to increased               and showed that 1α,25(OH)2D3 administration delayed
                          Ca2+ in normal colon73 and skeletal muscle cells74, and may                  the onset of prostate intraepithelial neoplasias (PIN) and
                          not have a direct role in the antiproliferative activities of                had better anti-tumour activity when administered to
                          1α,25(OH)2D3 in tumour cells (discussed below). In addi-                     mice with early-stage (PIN) rather than advanced-stage
                          tion, ERK can also increase the transcriptional activity of                  prostate disease77. Furthermore, studies using model
                          the VDR75, and nongenomic activation of PKC may stabi-                       systems of squamous cell carcinoma (SCC)78, prostate
                          lize VDR (through phosphorylation)23,76, thereby affecting                   adenocarcinoma79, cancers of the ovary80, breast81 and
                          the transcriptional activity of the receptor. Therefore, the                 lung82 showed that the administration of 1α,25(OH)2D3
                          nongenomic activation of these pathways may cooperate                        or vitamin D analogues had significant anticancer effects.
                          with the classical genomic pathway to transactivate VDR                      The effects of 1α,25(OH)2D3 and its derivatives have
                          and elicit the antiproliferative effects of 1α,25(OH)2D3, but                been shown to function through the VDR to regulate
                          this remains to be elucidated.                                               proliferation, apoptosis and angiogenesis62,83–87.

nature reviews | cancer                                                                                                          volume 7 | SEPTEMBER 2007 | 689
                                                      © 2007 Nature Publishing Group
REVIEWS

                                                                                                       SOC                      1α,25(OH)2D3                 GPCR
                                                         1α,25(OH)2D3                                  channels

                                                                                                            P
                                                                                                                VDR
                                                                                             1α,25(OH)2D3                      PLCγ
                                                             d                                                                                                        AC
                                                                                  Caveolae                            Ca2+                                   PI3K

                                                              mem                            ?                                                                      [cAMP]
                                                                                                                               PKC                     Ras
                                                              VDR
                          9cRA                                                                                                                                       PKA
                                 RXR                P
                                             VDR                                                                                                Raf isoforms

                                                                                                                                                   MEK1/2
                                                                                                 PBAF
                                                                      a              CBP/        SWI/SNF
                                                                                                                                               ERK–MAPK1/2
                                                                                     P300              Chromatin
                                                                          NCoA62–                      remodeling
                                                    1α,25(OH)2D3          SKIP
                                 9cRA                                                SRC-1             (histone
                                        RXR VDR P                                                      acetylation)                                                 Nucleus
                                                    Transcriptional        9cRA
                                                                                            P                                             Cross-talk
                                                    activation                    RXR VDR
                          Transcriptional
                          repression
                                                                                  5′  3′
                                                                                  VDREs                 NCoA62–                       b      Gene
                                                                                                        SKIP                 05              transcription
                                                                                                                         IP2
                                                                                                            9cRA       DR  DRIPs      RNA         CDKN1A
                              c                                                                                                  TF2B Pol II
                                                     WINAC                                                               P                        CYP24A1
                                   HDAC                                     Gene                                 RXR VDR                          SPP1
                                complexes  NCoR–
                                           SMRT      WSTF                   repression
                                          9cRA                                       CYP27B1                           3′
                          Chromatin                                                  PTH                          5′
                          remodelling          RXR VDR P                                                           VDREs
                          (histone              VDIR
                          deacetylation)
                                               nVDREs

                                                                                                                        Nature Reviews | Cancer
                         Figure 2 | 1α,25(OH)2D3-mediated transcriptional regulation. Classical action of 1α,25(OH)       D is mediated by
                                                                                                                         2 3
                         binding of the vitamin D receptor (VDR)−9-cis­-retinoic acid receptor (RXR) complex at the vitamin‑D response
                         elements (VDREs). a | Transcriptional activation involves the co-activators, steroid receptor coactivators (SRCs),
                         nuclear coactivator‑62 kDa–Ski-interacting protein (NCoA62–SKIP) and histone acetyltransferases (HATs), CREB
                         binding protein (CBP)–p300 and polybromo- and SWI‑2-related gene 1 associated factor (PBAF–SNF) to acetylate
                         histones to derepress chromatin. b | Binding of the vitamin D receptor-interacting protein 205 (DRIP205) to the
                         activation function 2 (AF2) of VDR (and RXR) attracts a mediator complex containing other vitamin D receptor-
                         interacting proteins (DRIPs) that bridge the VDR–RXR–NCoA62–SKIP–DRIP205 complex with transcription factor
                         2B (TF2B) and RNA polymerase II (RNA Pol II) for transcription initiation. The presence of the multiprotein complex
                         facilitates increased transcription of genes, such as CDKN1A (which encodes the cyclin-dependent kinase
                         inhibitor p21), CYP24A1 (which encodes 24-OHase) and SPP1 (which encodes osteopontin)23.
                         c | 1α,25(OH)2D3-mediated transcriptional repression involves VDR–RXR heterodimer association with VDR-
                         interacting repressor (VDIR) bound to E‑box-type negative VDREs (nVDREs), dissociation of the HAT co-activator
                         and recruitment of histone deacetylase (HDAC) co-repressor54. Williams syndrome transcription factor (WSTF)
                         potentiates transrepression by interacting with a multifunctional, ATP-dependent chromatin-remodelling
                         complex (WINAC) and chromatin55. This leads to the repression of genes, such as CYP27B1 (which encodes 1α-
                         OHase) and PTH (which encodes parathyroid hormone). d | Non-genomic, rapid actions of 1α,25(OH)2D3 are
                         hypothesized to involve 1α,25(OH)2D3 binding to cytosolic (VDR) and membrane VDR (memVDR), also found in
                         caveolae, and speculated to activate the mitogen-activated protein kinase (MAPK)–extracellular signal-regulated
                         kinase (ERK) 1 and 2 cascade68 through the phosphorylation (P) and activation of Raf by protein kinase C (PKC) by
                         Ca2+ influx through store-operated Ca2+ (SOC) channels. 1α,25(OH)2D3 stimulates SOC Ca2+ influx (in muscle cells)
                         by trafficking of the classic VDR to the plasma membrane, where the VDR interacts with the SOC channel. Ca2+
                         influx activates Ca2+ messenger systems, such as PKC. Activated PKC can phosphorylate VDR. 1α,25(OH)2D3
                         binding to G‑protein coupled receptors (GPCRs) activates phospholipase Cγ (PLCγ), Ras, phosphatidylinositol
                         3‑kinase (PI3K) and protein kinase A (PKA) pathways, and induces MAPK–ERK1 and 2 signalling. Activated Raf–
                         MAPK–ERK may engage in cross-talk with the classical VDR pathway to modulate gene expression. AC, adenylate
                         cyclase; cAMP, cyclic adenosine monophosphate.

690 | SEPTEMBER 2007 | volume 7                                                                                                             www.nature.com/reviews/cancer
                                                        © 2007 Nature Publishing Group
REVIEWS

                                       a            b      c              d                   e                 f             g
                                                                              1α,25(OH)2D3
                                               IGF1
                                                                                                            TGFβ         ↑E-cadherin      Wnt

                                                                                                            TGFβR2
                                                                                                            TGFβR1
                                                   TGF1R
                                      EGFR
                                                                                                                          β-catenin                   Frizzled

                                Ras                P13K                                                                                                          Cytosol
                                                                  BCL2         BAX                         SMADs                         β-catenin
                            Raf isoforms                         BCL-XL       BCL-XS                                                            APC
                                             Akt                                                                    β-catenin VDR
                              MEK1/2                                                                                                      Degradation

                                                    Telomerase      Effector
                                                                    caspases
                           ERK–MAPK1/2

                                                                              p15    p21    p27    SKP2                   β-catenin
                                                     Apoptosis    MYC                                                                                MYC
                                                                                                                                       TCF4
                                                                                                                                                     TCF1
                             Differentiation                                                                                                         CD44
                             Apoptosis                                    CDK4/6        CDK2      Degradation                                        PARG
                             Growth inhibition
                                                                     Cyclin D1,2,3 Cyclin E

                                                                                                          p107/p130                                              Nucleus

                                                                          PPP                              E2F4/5
                                                                          pRB           pRB        +
                                                                                                             DP1          Cell cycle
                                                                                       E2F1,2,3

                                                                                                             Growth arrest
                          Figure 3 | Key cancer-related signalling pathways targeted by 1α,25(OH)2D3. 1α,25(OH)2D3 inhibits mitogen-activated
                                                                                                                             Nature Reviews | Cancer
                          protein kinase (MAPK)–extracellular signal-regulated kinase (ERK) 1 and 2 signalling through suppression of epidermal
                          growth factor (EGFR; a) and insulin-like growth factor 1 (IGF1; b), which both target Ras. 1α,25(OH)2D3 induces apoptosis
                          through the IGFR1−phosphatidylinositol 3‑kinase (PI3K)−Akt-dependent signalling pathway (b), inhibiting telomerase (c),
                          downregulating BCL2, inducing BAX and activating caspase cleavage (d). Cell-cycle progression is perturbed by
                          1α,25(OH)2D3 through S‑phase kinase-associated protein ubiquitin ligase (SKP2; targeting p27 for degradation; e), and
                          MYC, which results in pRB dephosphorylation; and transforming growth factor-β (TGFβ; f) cross-talk. Cell-cycle
                          perturbation by 1α,25(OH)2D3 ultimately affects the association of retinoblastoma pocket proteins (pRB and p107/p130)
                          and the E2F family of transcription factors and DP polypepitide (DP1) heterodimers that mediate the transcription of cell-
                          cycle genes. Association of E2F1, 2 and 3 with pRB in its hypophosphorylated state and interaction of the E2F4 and 5
                          transcriptional repressors and DP1 with p107/p130 prevent transcription of cell-cycle genes and restrain cell-cycle
                          progression. Activation of VDR by 1α,25(OH)2D3 induces the expression of E‑cadherin (g), thereby promoting the
                          translocation of β‑catenin from the nucleus to the plasma membrane and competing with T-cell transcription factor 4
                          (TCF4) for β‑catenin binding; thus inhibiting the Wnt–β-catenin–TCF4 signalling pathway, which leads to the induction of
                          MYC, TCF1 (transcription factor 1), CD44 and PPARG (peroxisome proliferator-activated receptor-γ). APC, adenomatosis
                          polyposis coli; CDK, cyclin-dependent kinase; pRB, phosphorylated retinoblastoma; Wnt, wingless-related MMTV
                          integration site.

                          Antiproliferative effects of 1α,25(OH)2D3. Cell-cycle                        contain VDREs, and their transcriptional activation
                          perturbation is central to 1α,25(OH) 2D3-mediated                            or repression may not be directly mediated by VDR.
                          antiproliferative activity in tumour cells (supplemen-                       1α,25(OH) 2D 3–VDR transcriptional activation of
                          tal information S1 (table)). Progression through the                         CDKN1A induces cell-cycle exit (differentiation) and
                          cell cycle is regulated by cyclins, and their association                    cell-cycle arrest in human U937 myelomonocytic cells62.
                          with CDKs and CDK inhibitors (CKIs). Expression                              Treatment of human breast cancer MCF7 cells with
                          of the CKIs p21 and p27 inhibits proliferation, in part                      1α,25(OH)2D3 also increases the expression of CDKN1A
                          by inducing G1 cell-cycle arrest and withdrawal from                         and CDKN1B, (which encodes p27) and represses
                          the cell cycle (G0). CDKN1A and GADD45A contain                              CCND1 (encoding cyclin D1), CCND3 (encoding cyc-
                          a functional VDRE and are direct transcriptional tar-                        lin D3), CCNA1 (which encodes cyclin A1) and CCNE1
                          gets of 1α,25(OH)2D3–VDR. However, many genes are                            (which encodes cyclin E1), and hence leads to the inhibi-
                          transcriptionally affected by 1α,25(OH)2D3 but do not                        tion of CDK activity and pRb hypophosphorylation88,89.

nature reviews | cancer                                                                                                             volume 7 | SEPTEMBER 2007 | 691
                                                           © 2007 Nature Publishing Group
REVIEWS

                         Similarly, the treatment of SCC cells with 1α,25(OH)2D3           of 1α,25(OH)2D3102. Recent findings reported by Palmer
                         induces G0/G1 cell-cycle arrest owing to the tran-                et al.103 indicate that 1α,25(OH)2D3 promotes differentia-
                         scriptional activation of CDKN1B and consequent                   tion through the induction of CDH1 (which encodes E
                         pRb hypophosphorylation90. However, in this context               cadherin) in adenomatosis polyposis coli (APC)-mutated
                         CDKN1A expression was repressed, indicating that the              human colorectal cancer SW480 cells. CDH1 activation
                         cell-cycle arrest is an indirect effect of 1α,25(OH)2D3           consequently restrained cell growth by facilitating the
                         treatment or that cell-type specificity might determine           translocation of β‑catenin from the nucleus to the plasma
                         the ability of activated 1α,25(OH)2D3–VDR to induce               membrane, thus inhibiting β‑catenin-mediated tran-
                         CDKN1A expression90. Other genes have been shown                  scription and allowing activated VDR to compete with
                         to be transcriptionally affected by 1α,25(OH)2D3 in               β‑catenin for transcription factor binding. Again, there
                         colon cancer, ovarian carcinoma and leukaemia cells,              appears to be no specific mechanism regarding the ability
                         such as activation of GADD45 (REF 63), which is involved          of 1α,25(OH)2D3 to induce differentiation in tumour cells
                         in DNA damage responses, repression of TYMS                       (supplemental information S1 (table)).
                         (which encodes thymidylate synthetase)91 and TK1
                         (which encodes thymidine kinase)91, which are involved            Apoptosis. In addition to the antiproliferative effects
                         in DNA replication, and activation of the INK4 fam-               of 1α,25(OH) 2D 3, there is increasing evidence that
                         ily92 of cyclin D‑dependent kinase inhibitors, which              1α,25(OH)2D3 exerts anti-tumour effects by regulat-
                         mediate G1 cell-cycle arrest; whereas cyclin E–CDK2               ing key mediators of apoptosis, such as repressing the
                         and the SKP2 (S-phase kinase-associated protein 2)                expression of the anti-apoptotic, pro-survival proteins
                         ubiquitin ligase, which targets CKIs to the proteasome,           BCL2 and BCL-XL, or inducing the expression of pro-
                         are downregulated93 by 1α,25(OH)2D3. 1α,25(OH)2D3                 apoptotic proteins (such as BAX, BAK and BAD). It
                         treatment also results in the repression of the proto-            has been reported that 1α,25(OH)2D3 downregulates
                         oncogene MYC89,94, which significantly contributes to             BCL2 expression in MCF‑7 breast tumour and HL‑60
                         the antiproliferative effects of 1α,25(OH)2D3.                    leukaemia cells and upregulates BAX and BAK expres-
                             1α,25(OH)2D3 can have many indirect effects on                sion in prostate cancer, colorectal adenoma and carci-
                         cell-cycle regulation owing to cross-talk with other              noma cells84. In addition to regulating the expression
                         pathways; for example, 1α,25(OH)2D3 treatment can                 of the BCL2 family, 1α,25(OH)2D3 might also directly
                         result in the upregulation of IGFBP3 (which encodes               activate caspase effector molecules, although it is
                         insulin growth factor binding protein 3) and trans-               unclear whether 1α,25(OH)2D3-induced apoptosis is
                         forming growth factor‑β (TGFβ)–SMAD3 signalling                   caspase-dependent84. In support of this idea, the treat-
                         cascades and by downregulating the epidermal growth               ment of mouse SCC tumour cells with 1α,25(OH)2D3
                         factor receptor (EGFR) signalling pathway67,95,96 (FIG. 3).       increased VDR expression and concomitantly inhib-
                         Although there appears to be an overall inhibition of             ited the phosphorylation of ERK104. Upstream of ERK,
                         cell-cycle progression in tumour cells treated with               the growth-promoting and pro-survival signalling
                         1α,25(OH)2D3, the precise molecular basis for such                molecule MEK is cleaved and inactivated in a caspase-
                         an effect differs from one tumour cell type to another            dependent manner in cells that undergo apoptosis
                         such that a unifying hypothesis with regard to the                after treatment with 1α,25(OH)2D3. Recently, a novel
                         exact mechanism of 1α,25(OH) 2D 3-mediated cell-                  mechanism of 1α,25(OH)2D3-mediated apoptosis in
                         cycle perturbation has not been possible (supplemental            epithelial ovarian cancer cells was proposed by Jiang
                         information S1 (table)).                                          et al. 105, wherein they showed that 1α,25(OH) 2D 3
                             Activation of the VDR by 1α,25(OH)2D3 can also                destabilizes telomerase reverse transcriptase (TERT)
                         inhibit tumour cell proliferation by inducing differentia-        mRNA, therefore inducing apoptosis through tel-
                         tion in various myeloid leukaemia cell lines and freshly          omere attrition resulting from the down-regulation
                         isolated leukaemia cells62,83, which is dependent on the          of telomerase activity. The diverse effects observed
                         formation of activated VDR and phosphatidylinositol 3-            for 1α,25(OH) 2D 3-mediated apoptosis suggest that
                         kinase (PI3K) complexes97. However, in haematopoeitic             although anti-proliferative effects directed against
                         progenitor cells, 1α,25(OH)2D3 inhibits differentiation           the tumour are clear in vitro and in vivo (supple-
                         through VDR-independent suppression of interleukin                mental information S1 (table)), dissecting the exact
                         12 (IL12) protein secretion and down-regulation of other          mechanism(s) central to these activities remains a
                         co-stimulatory molecules (CD40, CD80 and CD86)98. In              challenge.
                         cell lines of head and neck, colon and prostate tumours,
                         administration of 1α,25(OH)2D3 or vitamin D analogues             Angiogenesis. 1α,25(OH)2D3 inhibits the proliferation
                         induces the expression of genes that are associated with the      of endothelial cells in vitro and reduces angiogenesis
                         differentiated cell of origin91,99,100. In various colon cancer   in vivo106–108. Vascular endothelial growth factor (VEGF)-
                         cells, treatment with 1α,25(OH)2D3 induces differentia-           induced endothelial cell tube formation and tumour
                         tion either by increasing PKC- and JNK-dependent JUN              growth are inhibited in vivo by 1α,25(OH)2D3 admin-
                         activation101 or by differentially regulating the expression      istration to mice with VEGF-overexpressing MCF‑7
                         of inhibitor of DNA binding 1 and 2 (ID1 and ID2), which          xenografts86. 1α,25(OH)2D3 can increase VEGF mRNA
                         encode proteins that are transcriptional regulators of epi-       levels in vascular smooth muscle cells109 and upregu-
                         thelial cell proliferation (ID2) and differentiation (ID1);       late mRNA levels of the potent anti-angiogenic factor
                         the repression of ID2 mediated the antiproliferative effects      thrombospondin 1 (THBS1) in SW480-ADH human

692 | SEPTEMBER 2007 | volume 7                                                                                     www.nature.com/reviews/cancer
                                                   © 2007 Nature Publishing Group
REVIEWS

                                   colon tumour cells 102. In SCC cells, 1α,25(OH) 2D 3           prostaglandin activity, the induction of their degradation
                                   induces the angiogenic factor interleukin 8 (IL8)110, but      through the upregulation of 15-hydroxyprostaglandin
                                   in prostate cancer cells 1α,25(OH)2D3 interrupts IL8           dehydrogenase, and reduction of prostaglandin recep-
                                   signalling leading to the inhibition of endothelial cell       tors120. These findings support the rationale for clinical
                                   migration and tube formation111. A significant inhibition      evaluation of a combination of 1α,25(OH)2D3 and non-
                                   of metastasis is observed in prostate and lung murine          steroidal anti-inflammatory drugs (NSAIDs) for prostate
                                   models treated with 1α,25(OH)2D3, and these effects            cancer therapy120. Increased anti-tumour effects with
                                   may be based, at least in part, on the anti-angiogenic         1α,25(OH)2D3 combination therapy offers the opportu-
                                   effects described79,82. Interestingly, in tumour-derived       nity for the clinical use of 1α,25(OH)2D3 across several
Platinum analogues                 endothelial cells (TDECs), 1α,25(OH)2D3 induces apop-          tumour types where modest effects are observed with
Platinum-based                     tosis and cell-cycle arrest; however, these effects are not    chemotherapy alone.
chemotherapeutics that             seen in endothelial cells isolated from normal tissues
crosslink DNA and therefore        or from Matrigel plugs (Matrigel-derived endothelial           Clinical trials of 1α,25(OH)2D3
impair the progression of DNA
replication machinery.
                                   cells)106. Recently, Chung et al.112 demonstrated that         With the recognition of the preclinical antiproliferative
                                   TDECs may be more sensitive to 1α,25(OH)2D3 owing              and pro-differentiating effects of vitamin D in the 1970s
Taxanes                            to the epigenetic silencing of CYP24A1. Therefore,             and 1980s, a number of attempts were made to trans-
Drugs that inhibit microtubule     direct effects of 1α,25(OH)2D3 on endothelial cells may        late these findings into the clinic. Several investigators
dynamics by stabilizing GDP-
                                   have a primary role in the 1α,25(OH)2D3-mediated               attempted to administer 1α,25(OH)2D3 as a differenti-
bound tubulin. Microtubules
form the mitotic spindle and so    anti-tumour activity that is observed in animal models         ating agent in myelodysplasia and acute leukaemia121–123.
taxanes prevent the                of cancer.                                                     Although some patients seemed to respond to the ther-
completional of mitosis.                                                                          apy, these improvements were not enough to encour-
                                   Preclinical combination studies                                age further trials, as 20–30% of patients who received
Myelodysplasia
Any of a group of bone marrow
                                   In vitro and in vivo analyses indicate that 1α,25(OH)2D3       a daily dose of 1α,25(OH)2D3 developed hypercalcemia.
disorders that have markedly       acts synergistically with chemotherapeutic agents.             Such findings have reinforced the conviction that less
abnormal reduction in one or       1α,25(OH)2D3 potentiates the anticancer activity of            hypercalcemic analogues of vitamin D, with modified
more types of circulating blood    agents such as platinum analogues113–115, taxanes116,117 and   chemical structures to make them less prone to degra-
cells owing to defective growth
                                   DNA-intercalating agents117,118. Optimal potentiation          dation by 24-OHase (FIG. 4a), must be developed if the
and maturation of blood-
forming cells in the bone          is seen when 1α,25(OH)2D3 is administered before or            therapeutic advantages of vitamin D biological effects are
marrow.                            simultaneously with chemotherapy treatment; admin-             to be realized124,125. It is important to note that the early
                                   istration of 1α,25(OH)2D3 after the cytotoxic agent            anticancer studies of 1α,25(OH)2D3 were conducted
Hypercalcemia                      does not provide potentiation114,116. The combination of       using dosing schedules optimized for the treatment
Excess of Ca2+ in the blood.
Chronic elevated serum levels
                                   1α,25(OH)2D3 and cisplatin in SCC cells in vitro induced       of renal osteodystrophy and osteoporosis, and the doses
of Ca2+ (12.0 mg dL) can result    tumour cell apoptosis characteristic of 1α,25(OH)2D3           important for anticancer effects were not investigated
in urinary calculi (renal or       alone. The pro-apoptotic signalling molecule MEKK1             separately. Had the administration of 1α,25(OH)2D3
bladder stones) and abnormal       (mitogen-activated protein kinase kinase kinase 1), is         been developed from an anticancer standpoint, the
heart rhythms. Severe
                                   up-regulated in both apoptotic and pre-apoptotic SCC           following considerations would have been determined:
hypercalcemia (above 15–16
mg dL) can result in coma and      cells treated with 1α,25(OH)2D3 (REF 104). This up-regu-       first, optimal biologically-effective dose and maximum
cardiac arrest.                    lation of MEKK1 was potentiated in combination with            tolerated dose (MTD) across several cancers; second,
                                   cisplatin treatment, suggesting that 1α,25(OH)2D3 pre-         the most effective dosing schedules to achieve antican-
Osteodystrophy                     treatment commits cells to undergo apoptosis through           cer activity; third, 1α,25(OH)2D3-dependent signalling
Defective bone ossification that
occurs when the kidney fails to
                                   specific molecular pathways (probably the MEK signal-          targets and molecular end-points; fourth, 1α,25(OH)2D3
maintain proper levels of Pi and   ling pathway), and that this effect is increased when cells    interactions with other cytotoxic or other anticancer
Ca2+. This results in slowed       are treated with an additional genotoxic stimulus 113.         drugs that may be therapeutically advantageous; and
bone growth and causes bone        Similar effects are seen in MCF‑7 cells treated with the       finally, design of clinical trials that mirror, as much as
deformities in children. In
                                   vitamin D analogue ILX 23‑7553 in combination with             possible, the exposures active in preclinical models to
adults, renal osteodystrophy
results in thin and weak bones,    doxorubicin or ionizing radiation118. In these studies, ILX    determine whether biological effects can be achieved in
bone and joint pain and            23‑7553 increased doxorubicin cytotoxicity and blocked         human tumours in clinical therapy (FIG. 4b).
vulnerability to osteoporosis.     the induction of p53 expression. Increased anti-tumour             Several studies have attempted to define a safe
                                   activity with 1α,25(OH)2D3 and the taxane paclitaxel is        and effective clinical treatment regimen126–129. These
Osteoporosis
A condition that is
                                   associated with a significant decrease in p21 expression,      investigations were based on the recognition that most
characterized by a decrease in     which sensitizes cells to both DNA-damaging agents             positive preclinical studies used high-dose, intermit-
bone mass with decreased           (such as cisplatin and doxorubicin) and microtubule-           tent 1α,25(OH)2D3. Although it is clear that 20–30%
density and enlargement of         disrupting agents (such as paclitaxel and docetaxel)116.       of patients receiving 1α,25(OH)2D3 at a dose of 1.5–2.0
bone spaces producing
                                   In SCC and PC‑3 (prostate cancer) xenografts, pre-             µg a day develop hypercalcemia130, there have been few
porosity and brittleness of the
bone.                              treatment with 1α,25(OH)2D3 resulted in an increased           studies that have compared continuous and intermittent
                                   anti-tumour effect in combination with paclitaxel116.          dosing regimens in cancer patients. Muindi and col-
Pharmacokinetics                   Similar results have also been observed in vivo with           leagues131 have determined the pharmacokinetic profile of
The characteristic interactions    MCF‑7 xenografts in which mice were treated with               a 1α,25(OH)2D3 regimen that is active in a preclinical
of a drug and the body in
terms of its absorption,
                                   vitamin D analogues and paclitaxel119. 1α,25(OH)2D3-           animal model. High-dose 1α,25(OH)2D3 (daily for 3
distribution, metabolism and       mediated downregulation of cyclooxygenase 2 (COX2)             days 0.125 µg per mouse ~6.25 µg per kg (body weight))
excretion.                         expression in prostate cancer cells leads to decreased         resulted in growth inhibition of the syngeneic mouse SCC

nature reviews | cancer                                                                                                  volume 7 | SEPTEMBER 2007 | 693
                                                            © 2007 Nature Publishing Group
REVIEWS

a                                21                    Side chain
                                18         20 22
                                                      24
                       12
                                       17        23
              11            13                        25                                                          OH
                       C         14D
                                            16                                                                                                                              OH
              9
                        8             15
                        7
              6
                                             Seco-B-ring
              5                  19
          4                10          Vitamin D3                                            25(OH)D3                                                        1α,25(OH)2D3
               A                                                               HO
                       1               Cholecalciferol                                       25-Hydroxycholecalciferol                                       Calcitriol
    HO 3                                                                                                                            HO              OH
                   2

                                                           Vitamin D analogues                                                               Vitamin D receptor modulators

                                                                                                             OH
                                                           OH                                                                                            S
                                                                                                                                      HO                                             O
                                                                                                                                                                                     S
                                                                                                                                                                                 O
                                                                                                                                                             LY2108491               O

                                       Paricalcitol                                          EB1089
    HO                          OH                                                                                                                       S
                                                                      HO             OH                                               HO
                                                                                                                                                                                             OH
                                                                                                O                                                                            O
                                                                                                                                                             LY2109866
                                                                                                                                                                                         O
                                                                                                          OH
                                                           OH

                                                                                                                                                O                            O
    HO                      OH ILX23-7553                                                                                                O                   LG190119                    O
                                                                         HO            OH OCT

b    Epidemiology/risk factors                                                                                              1 ,25(OH)2D3 or new analogues
                                                                                          Prediction of                     and drug combinations
     • Nutritional composition
                                                                                          1α,25(OH)2D3 response
     • UV exposure
     • VDR polymorphisms                                                                                                 In vitro systems
     • Genetic background                                                                                                • Tumour cells                             Mechanisms of action
                                                                                                                         • Stromal cells                            • Genomic (VDR)
                                           Vitamin D levels                                                              • Progenitor cancer stem cells             • Non-genomic (memVDR)
                                           (serum and cancer tissue)          1α,25(OH)2D3    1α,25(OH)2D3                                                          • Apoptosis
                                                                              or new          associated toxicities:                                                • Cell cycle
                                           • Effectors of 1α,25(OH)2D3                                                                                              • Angiogenesis
                                           metabolism: expression             analogues       Improve drug dosing
                                                                              and drug        Pharmacokinetics           In vivo systems                            • Cell signaling cross-talk
                                           and activity of 25-OHase,                                                     • Preclinical animal models:               • Cell–cell interaction
                                           1α-OHase, 24-OHase, VDR            combinations    Pharmacodynamics
                                                                                                                         syngeneic, xenografts and
                                                                                                                         genetically-modified (VDR–/–)
                                       Clinical assessment
Cancer                                 • Therapeutic impact,                              Clinical trials
patient                                response and biomarkers                            • Prevention                         Clinical dosing
                                       evaluation                                         • Anti-tumour therapy                schedules

                                                      Figure 4 | Development of 1α,25(OH)2D3 and vitamin D analogues as anticancer drugs. a | Cholecalciferol               (vitamin
                                                                                                                                                              Nature Reviews | Cancer
                                                      D3) is 25-hydroxylated at C‑25 (denoted by carbon atom number on the structure of cholecalciferol) to form 25-hydroxyc-
                                                      holecalciferol (25(OH)D3). This is 1α-hydroxylated at C‑1 by 1α-OHase to yield 1α,25(OH)2D3 (calcitriol). 1α,25(OH)2D3 is a
                                                      secosteroid that is similar in structure to steroids but with a ‘broken’ B‑ring (denoted seco‑B-ring) where two of the carbon
                                                      atoms (C‑9 and C‑10) of the four steroid rings are not joined. Many vitamin D analogues (left) retain the secosteroid
                                                      structure with modified side chain structures around the C‑24 position, which makes them less hypercalcemic and less
                                                      prone to degradation by 24-OHase170,171. Several structures of vitamin D analogues referred to in the text are shown:
                                                      paricalcitol (19-nor‑1α(OH)2D2), ILX23‑7553 (16-ene‑23-yne‑1α,25(OH)2D3), OCT (Maxacalcitol, 22-oxa‑1α,25(OH)2D3)
                                                      and EB1089 (Seocalcitol, 1α-dihydroxy‑22,24-diene‑24,26,27-trihomo-vitamin D3). Vitamin D receptor modulators
                                                      (VDRMs, right) are non-secosteroidal in structure. Some of the representative compounds described are LY2108491,
                                                      LY2109866 and LG190119 (REFs 146,147). b | Paradigm for development and clinical translation of 1α,25(OH)2D3 as an
                                                      anticancer agent. Establishment of in vitro and in vivo experimental systems is crucial to developing 1α,25(OH)2D3 or
                                                      vitamin D analogues that target vitamin D metabolism and signalling. These systems allow the mechanisms of action of
                                                      1α,25(OH)2D3 to be studied along with novel analogues (also in combination with cytotoxic drugs) in multiple transformed
                                                      cell types and their biological effects (tumour and normal tissues) in animals. Importantly, studies on the pharmacokinetics
                                                      and pharmacodynamics of drug action will enable the development of better designed clinical dosing schedules for
                                                      clinical trials that will mirror the exposures active in preclinical models where optimal biological effects of 1α,25(OH)2D3
                                                      are demonstrated and are achievable in human tumours in clinical therapy.

694 | SEPTEMBER 2007 | volume 7                                                                                                                          www.nature.com/reviews/cancer
                                                                                 © 2007 Nature Publishing Group
You can also read