Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2018 Nov 22;85(6):1095–1102. doi: 10.1111/bcp.13781

Update on pharmacologically‐relevant vitamin D analogues

Glenville Jones 1,, Martin Kaufmann 1
PMCID: PMC6533488  PMID: 30308088

Abstract

Pharmacologists have been interested in vitamin D since its metabolism was elucidated in the early 1970s. Despite the synthesis of thousands of vitamin D analogues in the hope of separating its calcemic and anti‐proliferative properties, few molecules have reached the market for use in the treatment of clinical conditions from psoriasis to chronic kidney disease. This review discusses vitamin D drugs, recently developed or still under development, for use in various diseases, but in particular bone disease. In the process we explore the mechanisms postulated to explain the action of these vitamin D analogues including action through the vitamin D receptor, action through other receptors e.g. FAM57B2 and dual action on transcriptional processes.

Keywords: bioequivalence, cytochrome P450, drug development, genetics and pharmacogenetics, pharmacokinetics, therapeutic drug monitoring

Introduction

Since the metabolism of vitamin D was elucidated in the early 1970s, organic chemists and pharmacologists have sought to design and synthesize vitamin D analogues that would mimic some, if not all, of the biological actions of the parent vitamin. Thousands of molecules have been prepared and tested but only a few of these have been successfully translated into marketed drug forms that can be used to treat bone disease or in other areas of clinical medicine. These synthetic vitamin D analogues have been reviewed extensively in recent years 1, 2, 3, 4, 5 and this chapter will not attempt to duplicate those reviews. Furthermore, this review will not spend much time exploring the precise biochemical mechanism by which the active form of vitamin D3, 1,25‐dihydroxyvitamin D3 [1,25‐(OH)2D3] is able to modulate gene expression of target genes involved in the physiological effects of vitamin D on calcium/phosphate homeostasis or its effects on cell proliferation and differentiation covered in recent summaries of this subject 6, 7, 8. Rather, this short treatise will focus on the pharmacologically important vitamin D agents recently introduced or are under active development that might be employed ultimately in the treatment of bone disease or related conditions. We start by reviewing new work on two early metabolites of vitamin D3: 25‐hydroxyvitamin D3 [25‐OH‐D3] and 24,25‐dihydroxyvitamin D3 [24,25‐(OH)2D3], then move on to review newer analogues of 1,25‐(OH)2D3 that are modified chemically to be more selective in their actions, especially in bone.

25‐OH‐D3

Prior to any discussion of vitamin D analogues, we must mention the parent vitamin D molecules, vitamin D3 (cholecalciferol) and vitamin D2 (ergocalciferol) that are still used in the treatment of vitamin D deficiency and its accompanying bone diseases, rickets and osteomalacia. Doses of 600–800 IU day–1 have been recommended to maintain serum 25‐OH‐D (defined as the sum of 25‐OH‐D3 + 25‐OH‐D2) levels above 20 ng ml–1 (50 nmol l–1) in the normal healthy population by the Institute of Medicine 9 but the Endocrine Society suggests higher doses of 1000–2000 IU day–1 are needed to cure vitamin D deficiency 10. Oral supplements of vitamin D (D2 or D3) are usually given at daily, weekly or monthly intervals in order to raise serum 25‐OH‐D levels, though a more frequent dosing regimen seems to be the more effective alternative. Much recent testing suggests that monthly dosing of vitamin D2 is less effective than vitamin D3 11, 12, although the differences are not so evident when the two are compared in daily dosing protocols 13. A recent clinical trial 14 suggests that large doses of vitamin D3 given every few months is associated with an increase in the frequency of falls and fractures, alhough the exact mechanism for how such side effects manifest is not known. Nevertheless, vitamin D supplements with their low cost and over‐the‐counter availability remain the agent of choice to treat vitamin D deficiency and associated bone diseases.

25‐OH‐D3 (Table 1), the first metabolite of vitamin D3 is produced in the liver and constitutes the main circulating form. Although, like vitamin D3, 25‐OH‐D3 is biologically inactive until it is 1α‐hydroxylated in the kidney, the provision of this prodrug form overcomes certain barriers sometimes observed with the parent vitamin D3, namely: malabsorption of orally administered drug; sequestration of the lipophilic molecule by the adipose tissue; and slow or inefficient 25‐hydroxylation by the liver enzyme, CYP2R1 21. A synthetic drug form of 25‐OH‐D3 (also known as calcifediol) named Calderol was first introduced in the mid‐1970s for the treatment of renal osteodystrophy and other bone diseases and, although it is still available in Europe 22, it was discontinued in North America in the late 1990s because of poor demand in the face of the initial success of 1,25‐(OH)2D3 and its analogues. Recently, 25‐OH‐D3 was reintroduced in North America as an extended‐release form named Rayaldee (OPKO‐Renal, Miami, FL, USA) and approved for the treatment of secondary hyperparathyroidism experienced by stage 3 and 4 chronic kidney disease patients, the forerunner of renal osteodystrophy 23. Data from stage 3 and 4 chronic kidney disease patients show that 25‐OH‐D3 effectively raises serum 25‐OH‐D3 levels from low pretreatment values (<20 ng ml–1) to normal levels (>30 ng ml–1) and effectively lowers parathyroid hormone without causing hypercalcaemic side effects 24. Since these stage 3 and 4 patients have been reported to still possess residual CYP27B1 (1α‐hydroxylase) enzyme activity, it is believed that they can adequately convert 25‐OH‐D3 to 1,25‐(OH)2D3 in a regulated manner. Thus, the expectation is that the prodrug, Rayaldee might provide a safer alternative to 1,25‐(OH)2D3 and its commonly prescribed 1α‐hydroxylated analogues, paricalcitol and doxacalciferol, which are now suspected of causing excessive calcification of soft tissues during long‐term use 25. Early use of Calderol in the 1980s and 1990s suggested that even dialysis patients who have lost renal function and the ability to 1α‐hydroxylate 25‐OH‐D3 in the kidney could still make 1,25‐(OH)2D3 from large doses of 25‐OH‐D3 in extra‐renal tissues due the presence of the extra‐renal CYP27B1 enzyme 26. Accordingly, it remains to be seen if Rayaldee will generate a sufficient rise in serum 1,25‐(OH)2D3 to reduce hyperparathyroidism in dialysis patients or can be used more widely to benefit other patients with bone diseases caused by low serum levels of 25‐OH‐D3.

Table 1.

Natural metabolites of vitamin D and analogues of 1α,25‐(OH)2D3

Vitamin D analogue [ringstructure]a Side chain structure Company Status Possible target diseases Mode of delivery Reference
25‐OH‐D 3 1

chemical structure image

Generic
 
OPKO‐Renal
In use Europe
 
In use N. America
Vitamin D Deficiency
and/or
Chronic Kidney Disease
Stage 3/4
Hyperparathyroidism
Rapid release,
oral
Extended release
oral
 
 
US Patent No. 3 565 924
24 R ,25‐(OH) 2 D 3 1

chemical structure image

Bone fracture repair Martineau et al. 15
Calcitriol, 1α,25‐(OH) 2 D 3 2

chemical structure image

Roche, Duphar In use worldwide Hypocalcemia, Psoriasis Systemic Topical Baggiolini et al. 16
2‐methylene‐19‐nor‐ 20‐epi‐1α,25‐(OH) 2 D 3 (2MD) 3

chemical structure image

Deltanoids Preclinical Osteoporosis Systemic Shevde et al. 17
2‐methylene‐22‐dehydro‐19‐nor‐1,24 R ,25‐(OH) 3 D 3 (WT‐51) 3

chemical structure image

Deltanoids Preclinical Osteoporosis Systemic US Patent Application No. 2014/0206655
ED71 (Eldecalcitol) 4

chemical structure image

Chugai In use Japan Osteoporosis Systemic Nishii et al. 18
19‐nor‐14‐epi‐23‐yne‐ 1,25‐(OH) 2 D 3 (Inecalcitol, TX527) 5

chemical structure image

Hybrigenics Clinical trials Prostate Cancer Leukemia Systemic Ma et al. 19
C8‐alkyl derivatives 6

chemical structure image

Hyperproliferative disorders Gogoi et al. 20
a

Structure of the vitamin D nucleus (secosterol ring structure).

As described above, current dogma suggests that ALL vitamin D analogues have their therapeutic effects by mimicking the action of 1,25‐(OH)2D3 through a nuclear vitamin D receptor (VDR)‐mediated transcriptional mechanism, up‐and down‐regulating expression of vitamin D‐dependent genes 6, 7, 8. Although an additional theory involving a 1,25‐(OH)2D3 membrane‐receptor has been proposed 27, it has largely been dismissed because of the lack of convincing supporting evidence. Epidemiological studies have previously demonstrated an inverse association between serum 25‐OH‐D levels and lipid levels, as well as severity of metabolic diseases and body‐mass index 28, 29, 30, 31. However, Uesugi and colleagues 32 have recently shown in vitro that 25‐OH‐D3 can inhibit lipogenesis by mediating the degradation of sterol regulatory‐binding element proteins (SREBPs) involved in transactivation of lipogenic genes, via a novel VDR‐independent mechanism 33. SREBP precursors reside in the membrane of the endoplasmic reticulum, bound to SREBP cleavage‐activating protein (SCAP). Under low cellular concentrations of sterols, the SREBP–SCAP complex translocates to the Golgi, where the N‐terminal transcription factor domain is cleaved by sequential action of site‐1 and ‐2 proteases. The mature form of SREBP translocates to the nucleus to mediate the transcription of lipogenic genes. The presence of cholesterol and 25‐hydroxycholesterol promote binding and sequestration of SREBP–SCAP complex to insulin‐induced genes, preventing translocation to the Golgi, transcription factor maturation and consequently lipogenesis during a sterol‐replete state, as a part of a negative feedback loop 33, 34, 35. 25‐OH‐D3 inhibits lipogenesis by binding to SCAP, mediating its ubiquitination and degradation. The unbound SREBP is unstable and is ultimately degraded as well 34. While inhibition of lipogenesis via the 25‐OH‐D3–SCAP–SREBP axis has not yet been tested in vivo, it presents a novel pathway that could potentially be targeted for vitamin D analogue design for use in treatment of metabolic diseases. It is possible that 25‐OH‐D3 is simply mimicking 25‐hydroxycholesterol, which is a known oxysterol regulator of cholesterol metabolism. However, these preliminary data point to a possible beneficial effect of high levels of 25‐OH‐D3 to lower lipid levels in bone disease patients treated with 25‐OH‐D3. In addition to 25‐OH‐D3, there is ample justification for the Japanese consortium to screen a library of vitamin D analogues to potentially find agents capable of more potent binding to SREBP/SCAP. This work is reportedly in progress 36.

24,25‐(OH)2D3

Since its discovery in the early 1970s, 24,25‐(OH)2D3 (Table 1) has been considered a degradatory product of 25‐OH‐D3, devoid of biological activity and like its 1α‐hydroxylated counterpart, 1,24,25‐(OH)3D3, the first step on a catabolic pathway to truncated water‐soluble metabolites destined for excretion 37. The enzyme involved is CYP24A1, the deletion or mutation of which causes hypercalceamia in both mice and humans, evidence that is consistent with 24‐hydroxylation being a catabolic step 38. However, for 3 decades there has been the minority opinion that 24‐hydroxylated vitamin D metabolites might play an anabolic role in addition to the catabolic role evident when CYP24A1 exhibits loss of function. This role would be in mineralization or calcification of unmineralized osteoid 39 or tibial fracture repair in chicks 40. Until now, there has been a lack of any plausible mechanism for how 24,25‐(OH)2D3 could achieve this effect given the fact that it shows poor VDR binding and other evidence to support this theory has been meagre although a putative 24,25‐(OH)2D3 receptor has been claimed 41.

However, a very recent publication 15 has provided, for the first time, evidence for a possible novel mechanism to explain acceleration of bone‐fracture repair. Matrineau et al. have identified and cloned an effector molecule named FAM57B2 that binds 24,25‐(OH)2D3 and produces a lipid‐signalling molecule lactosyl ceramide (LacCer) thatmediates endochondral ossification during bone callus formation 15. Ablation of CYP24A1 gene or inactivation of Fam57b2 gene in chondrocytes caused loss of the bone‐healing effect while treatment with exogenous synthetic 24R,25‐(OH)2D3 or lactosyl ceramide, but not 1,25‐(OH)2D3, restores the effect 15. It is not yet clear if other 24‐hydroxylated vitamin D analogues or even other vitamin D analogues in general can reproduce the same bone‐healing effect as 24R,25‐(OH)2D3 at equivalent doses to those used here. 24,25‐(OH)2D3 is an allosteric modulator of FAM57B2 that catalyses formation of lactosylceramide from glucosylceramide. Although the specific role of LacCer in the chondrocyte remains unclear, it has been shown to participate in pathways such as cell proliferation, adhesion, apoptosis and angiogenesis. FAM57B2 ablation was found to phenocopy the callus defect observed in cyp24a1 –/– mice. Although the callus defect in cyp24a1 –/– mice could be rescued by either 24,25‐(OH)2D3 or LacCer administration; only LacCer treatment could restore the callus defect in Fam57b2 –/– animals indicating the requirement of both 24,25‐(OH)2D3 and its effector molecule FAM57B2 in this novel pathway of bone fracture repair. These data offer exciting evidence that 24‐hydroxylated vitamin D metabolites might be used to accelerate bone repair mechanisms.

Analogues of 1,25‐(OH)2D3

The physiological actions of 1,25‐(OH)2D3 include stimulation of intestinal calcium and phosphate absorption; conservation of calcium in the kidney; and stimulation of bone resorption through a VDR‐mediated mechanism of action 6, 7, 8. All of these functions are potentially useful for an agent used in the treatment of bone disease. Furthermore, although the VDR is present in both osteoblasts and immature cells of the osteoclast lineage and it was thought that the effects of 1,25‐(OH)2D3 on mature osteoclasts may be through effects on the rates of osteoclastogenesis and be regulated indirectly via osteoblasts involving osteoblast/osteoclast communication factors 42. At the molecular level, 1,25‐(OH)2D3 achieves its multiple effects by modulating the genes for key intestinal, kidney and bone proteins such as calbindins, Ca‐ATPases, osteocalcin, osteopontin and RANKL 6, 7, 8. It is not then surprising that various 1α‐hydroxylated vitamin D analogues (Table 1), such as 1,25‐(OH)2D3, 1α‐OH‐D3 and 1α‐OH‐D2 have undergone clinical trials for the treatment of osteoporosis 42. Four small trials of 1α‐OH‐D3, three of which were performed in Japan, suggested that it was effective in reducing hip fractures while in other trials the results were inconclusive 43, 44, 45, 46, 47. In early clinical trials, 1,25‐(OH)2D3‐treated patients showed modest gains in bone mineral density and reductions in fracture rates, but a more recent trial demonstrated no statistically significant effect of 1,25‐(OH)2D3 on the incidence of nonvertebral fractures or vertebral deformities. Moreover, in these trials, there was an increased risk of hypercalcaemia. This is not a big surprise; 3 decades ago, Hock and colleagues treated rats with pharmacological doses of 1,25(OH)2D3 and found that animals developed hypercalcaemia and nephrocalcinosis along with a hyperosteoid or under‐mineralized bone histology 48. Thus, based upon these clinical trials, 1,25‐(OH)2D3 has not been recommended as a first‐line drug to treat osteoporosis.

More recently, several newer 1,25‐(OH)2D3 analogues (Table 1) have been developed that are claimed to be more selective towards bone, particularly promoting bone formation rather than resorption. As such, they are less calcaemic than 1,25‐(OH)2D3, but not noncalcaemic. Of particular note are the C2‐substituted vitamin D analogues such as ED‐71 (now called Eldecalcitol), currently in use in Japan for treatment of osteoporosis 49; and 2‐MD [2‐methylene‐19‐nor, 20‐epi‐1,25‐(OH)2D3] and its 22‐dehydro, 24‐hydroxy derivative WT‐51 17, 50, 51. Despite the increased size of the A‐ring, these C2‐substituted derivatives have been shown to still fit neatly into the ligand‐binding pocket of the VDR in X‐ray crystallographic studies and also modulate gene expression. Eldecalcitol appears to have a tighter affinity for the vitamin D‐binding protein and as a result is reported to have a longer half‐life in the plasma 18. Eldecalcitol restored bone mass without causing hypercalcaemia in long‐term studies involving ovariectomized rats 49. Both 2MD and WT‐51 represent newer 2‐substituted vitamin D analogues which promote bone nodule formation in vitro and cause increases in bone mineral density and bone strength in ovariectomized rats in vivo without causing hypercalcaemia 17, 50, 51. However, in a randomized, double‐blind, placebo‐controlled study of osteopaenic women, daily oral dosing with 2‐MD caused a marked increase in bone formation markers but also resulted in a marked increase in bone resorption 51. Consequently, the net effect was increased bone remodelling and to date 2‐MD has not been developed further. WT‐51 has yet to reach the clinic.

Another family of 1,25‐(OH)2D3 analogues, the C/D‐ring modified derivatives (Table 1), that are commanding attention are under development for use in other fields include the Hybrigenics drug, Inecalcitol [14‐epi, 23‐yne derivative of 1,25‐(OH)2D3], which induces apoptosis in squamous cell carcinoma models and is being tested in a clinical trial for treatment of leukaemia 19, 52; and the C‐8 alkyl, C‐ring‐less, m‐phenylene aromatic D‐ring derivatives of 1,25‐(OH)2D3 20, which are purported to be noncalcaemic. Although there is optimism with every new family of vitamin D analogues, caution prevails as many have claimed that vitamin D analogues are non‐calcaemic or low‐calcaemic during in vitro testing only to find that the agents are rejected during clinical trials due to their calcaemic side‐effects in vivo.

Antagonists, nonsteroidal scaffolds and hybrid vitamin D molecules

The elucidation of the ligand‐binding domain of the VDR by X‐ray crystallography 53 has allowed for rational drug design of potential vitamin D analogues. The 1.8 A resolution crystal structure of the ligand‐binding domain reveals a folded helical arrangement around a pocket harbouring the 1,25(OH)2D3 molecule enclosed by a trapdoor composed of a helix‐12 53. During the binding of the ligand, the helix‐12 folds relative to the rest of the VDR structure to take up a different orientation and block exit of the ligand. The natural 1,25‐(OH)2D3 hormone occupies only 56% of the available space within the ligand‐binding pocket 53. Certain bile acids e.g. lithocholic acid will also fit into this pocket, suggesting that other molecules might trigger modulation of vitamin D‐dependent genes in vivo 54. Work has not only shown that synthetic vitamin D ligands can be modified in several new directions and still fit comfortably into the ligand‐binding domain (e.g. the 2‐substituted family of useful analogues, as noted earlier) but still allow for the helix‐12 rearrangement of the VDR necessary to trigger the recruitment of co‐activators and transactivation of vitamin D‐dependent genes 6, 7, 8. Agents that bind to the VDR but block the helix‐12 rearrangement such as the extended side‐chain vitamin D analogues (e.g. TEI‐9647 which is a 26,23‐lactone 55, 56 or the Schering ZK‐159222 compound, which is a side‐chain extended ester 57) are VDR antagonists (Table 2). TEI‐9647 was in clinical trial for the treatment of Paget's Disease 58.

Table 2.

Miscellaneous vitamin D compounds

Name Structure Name Structure
TEI‐9647
Teijin
 
VDR antagonist
Dehydration product of 1α,25( R )‐(OH) 2 D 3 –26,23( S )‐lactone
 
Saito and Kittaka 56
Ochiai et al . 55
Toell et al . 57
Ishizuka et al . 58

chemical structure image

 
 
 
ZK159222
Schering
 
VDR antagonist
 
Toell et al. 57

chemical structure image

LG190090
Ligand Pharmaceuticals
 
Non‐steroidal VDR agonist
 
Boehm et al . 59

chemical structure image

LY2108491
Eli Lilly
 
Non‐steroidal VDR agonist
 
Ma et al. 60

chemical structure image

 
DK 406
 
VDR‐Histone deacetylase inhibitor hybrid
Bijian et al . 62

chemical structure image

Modelling of the ligand‐binding pocket of VDR has also allowed for the design of vitamin D analogues based upon nonsteroidal scaffolds (Table 2). Biphenyl derivatives with a variable inter‐ring linker and side chains (e.g. Eli Lilly LY2108491 59 or the ligand LG190090 60) allow for the creation of a rigid molecule with the length, shape and functional groups to mimic the hormone, 1,25‐(OH)2D3. The main advantage of such molecules is that, unlike 1,25‐(OH)2D3, they lack chiral centres and are cheap to manufacture. Nonetheless, no nonsteroidal scaffold has entered clinical trials for the treatment of vitamin D‐related disease.

Another interesting concept is to make a hybrid vitamin D analogue that combines vitamin D functions with some complementary action. Retiferols combining vitamin D and retinoid functions have been successfully created by the Kutner group 61 could have some utility of the treatment of bone disease, while the VDR agonist has also been combined with a histone deacetylase inhibitor in the form of DK‐406 (Table 2) by Gleason and colleagues 62, its value arising from the fact that both agents are regulators of gene transcription and might be expected to act synergistically on certain vitamin D‐dependent genes.

Conclusions

As this review indicates, the same vitamin D metabolites that formed the original focus of pharmacologists' interest in the vitamin D field are currently undergoing a renaissance. Furthermore, the interest caused by 25‐OH‐D3 and 24,25‐(OH)2D3 is not all through the same 1,25‐(OH)2D3‐VDR‐mediated gene transcriptional mechanism that has driven the synthesis of thousands of calcitriol analogues for the past 3 or more decades. Novel ideas about how these agents might be proven to work therapeutically have emerged that involve the extra‐renal CYP27B1 enzyme, binding to the SREBP‐SCAP complex or an effector molecule FAM57B2, which binds 24R,25‐(OH)2D3. Although none of these operate in the clinical arena, there is promise that the work may result in novel drugs for treatment of vitamin D deficiency and bone disease. Time will tell.

Competing Interests

There are no competing interests to declare.

Jones G., and Kaufmann M. (2019) Update on pharmacologically‐relevant vitamin D analogues, Br J Clin Pharmacol;85:1095–1102. 10.1111/bcp.13781.

References

  • 1. Calverley MJ, Jones G. Vitamin D In: Antitumor Steroids, ed Blickenstaff RT. San Diego: Academic Press, 1992; 193–270. [Google Scholar]
  • 2. Bouillon R, Okamura WH, Norman AW. Structure‐function relationships in the vitamin D endocrine system. Endocr Rev 1995; 16: 200–257. [DOI] [PubMed] [Google Scholar]
  • 3. Jones G. Chapter 83: Vitamin D and analogues In: Principles of Bone Biology, Third edn, Section: Pharmacological Mechanisms of Therapeutics, eds Bilezikian J, Raisz L, Rodan G. San Diego: Academic Press Inc, 2008; 1777–1799. [Google Scholar]
  • 4. Jones G. Vitamin D analogs. Endocrinol Metab Clin North Am 2010; 39: 447–472. [DOI] [PubMed] [Google Scholar]
  • 5. Verlinden L, Eelen G, Bouillon R, Vanderwalle M, De Clercq P, Verstuyf A. Analogs of calcitriol In: Vitamin D, Third edn, eds Feldman D, Pike W, Adams J. San Diego: Elsevier, 2011; 1461–1487. [Google Scholar]
  • 6. Jones G, Strugnell S, DeLuca HF. Current understanding of the molecular actions of vitamin D. Physiol Rev 1998; 78: 1193–1231. [DOI] [PubMed] [Google Scholar]
  • 7. Pike JW, Zella LA, Meyer MB, Fretz JA, Kim S. Molecular actions of 1,25‐dihydroxyvitamin D3 on genes involved in calcium homeostasis. J Bone Miner Res 2007; 22 (Suppl. 2): V16–V19. [DOI] [PubMed] [Google Scholar]
  • 8. Pike JW, Meyer MB, Lee SM, Onal M, Benkusky NA. The vitamin D receptor: contemporary genomic approaches reveal new basic and translational insights. J Clin Invest 2017; 127: 1146–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Institute of Medicine . Dietary reference intakes for calcium and vitamin D. Washington DC: The National Academies Press, 2011. [PubMed] [Google Scholar]
  • 10. Holick MF, Binkley NC, Bischoff‐Ferrari HA, Gordon CM, Hanley DA, Heaney RP, et al Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011; 96: 1911–1930. [DOI] [PubMed] [Google Scholar]
  • 11. Armas LA, Hollis BW, Heaney RP. Vitamin D2 is much less effective than vitamin D3 in humans. J Clin Endocrinol Metab 2004; 89: 5387–5391. [DOI] [PubMed] [Google Scholar]
  • 12. Rapuri PB, Gallagher JC, Haynatzki G. Effect of vitamins D2 and D3 supplement use on serum 25OHD concentration in elderly women in summer and winter. Calcif Tissue Int 2004; 74: 150–156. [DOI] [PubMed] [Google Scholar]
  • 13. Holick MF, Biancuzzo RM, Chen TC, Klein EK, Young A, Bibuld D, et al Vitamin D2 is as effective as vitamin D3 in maintaining circulating concentrations of 25‐hydroxyvitamin D. J Clin Endocrinol Metab 2008; 93: 677–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Sanders KM, Stuart AL, Williamson EJ, Simpson JA, Kotowicz MA, Young D, et al Annual high‐dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA 2010; 303: 1815–1822. [DOI] [PubMed] [Google Scholar]
  • 15. Martineau C, Naja RP, Husseini A, Hamade B, Kaufmann M, Akhouayri O, et al Optimal bone fracture repair requires 24R,25‐dihydroxyvitamin D3 and its effector molecule FAM57B2. J Clin Invest 2018; 128: 3546–3557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Baggiolini EG, Wovkulich PM, Iacobelli JA, Hennessy BM, Uskokovic MR. Preparation of 1α‐hydroxylated vitamin D metabolites by total synthesis In: Vitamin D: Chemical, Biochemical and Clinical Endocrinology of Calcium Metabolism, eds Norman AW, Schaefer K, von Herrath D, Grigoleit HG. Berlin: De Gruyter, 1982; 1089–1100. [Google Scholar]
  • 17. Shevde NK, Plum LA, Clagett‐Dame M, Yamamoto H, Pike JW, DeLuca HF. A potent analog of 1α,25‐dihydroxyvitamin D3 selectively induces bone formation. Proc Natl Acad Sci U S A 2002; 99: 13487–13491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Nishii Y, Sato K, Kobayashi T. The development of vitamin D analogues for the treatment of osteoporosis. Osteoporosis Int 1993; 1 (Suppl): S190–S193. [DOI] [PubMed] [Google Scholar]
  • 19. Ma Y, Yu WD, Hidalgo AA, Luo W, Delansorne R, Johnson CS, et al Inecalcitol, an analog of 1,25D3, displays enhanced antitumor activity through the induction of apoptosis in a squamous cell carcinoma model system. Cell Cycle 2013; 12: 743–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Gogoi P, Seoane S, Sigüeiro R, Guiberteau T, Maestro MA, Pérez‐Fernández R, et al Aromatic‐based design of highly active and noncalcemic vitamin D receptor agonists. J Med Chem 2018; 61: 4928–4937. [DOI] [PubMed] [Google Scholar]
  • 21. Jones G, Prosser DE, Kaufmann M. Chapter 5: The activating enzymes of vitamin D metabolism (25‐ and 1α‐hydroxylases) In: Vitamin D, 4th edn, eds Feldman D, Pike W, Bouillon R, Giovannucci E, Goltzman D, Hewison M. San Diego, CA: Elsevier, 2017. [Google Scholar]
  • 22. Jetter A, Egli A, Dawson‐Hughes B, Staehelin HB, Stoecklin E, Goessl R, et al Pharmacokinetics of oral vitamin D(3) and calcifediol. Bone 2014; 59: 14–19. [PubMed] [Google Scholar]
  • 23. Sprague SM, Strugnell SA, Bishop CW. Extended‐release calcifediol for secondary hyperparathyroidism in stage 3‐4 chronic kidney disease. Expert Rev Endocrinol Metab 2017; 12: 289–301. [DOI] [PubMed] [Google Scholar]
  • 24. Petkovich M, Bishop CW. Chapter 91: extended‐release calcifediol in renal disease In: Vitamin D, 4th edn, eds Feldman D, Pike W, Bouillon R, Giovannucci E, Goltzman D, Hewison M. San Diego, CA: Elsevier, 2017; 667–678. [Google Scholar]
  • 25. Razzaque MS. The dualistic role of vitamin D in vascular calcifications. Kidney Int 2011; 79: 708–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Dusso A, Lopez‐Hilker S, Rapp N, Slatopolsky E. Extra‐renal production of calcitriol in chronic renal failure. Kidney Int 1988; 34: 368–375. [DOI] [PubMed] [Google Scholar]
  • 27. Nemere I, Dormanen MC, Hammond MW, Okamura WH, Norman AW. Identification of a specific binding proteins for 1 alpha,25‐dihydroxyvitamin D3 in basal‐lateral membranes of chick intestinal epithelium and relationship to transcaltachia. J Biol Chem 1994; 269: 23750–23756. [PubMed] [Google Scholar]
  • 28. Auwerx J, Bouillon R, Kesteloot H. Relation between 25‐hydroxyvitamin D3, apolipoprotein AI, and high density lipoprotein cholesterol. Arterioscler Thromb J Vasc Biol 1992; 12: 671–674. [DOI] [PubMed] [Google Scholar]
  • 29. Botella‐Carretero JI, Alvarez‐Blasco F, Villafruela JJ, Balsa JA, Vázquez C, Escobar‐Morreale HF. Vitamin D deficiency is associated with the metabolic syndrome in morbid obesity. Clin Nutr Edinb Scotl 2007; 26: 573–580. [DOI] [PubMed] [Google Scholar]
  • 30. Bouillon R, Carmeliet G, Lieben L, Watanabe M, Perino A, Auwerx J, et al Vitamin D and energy homeostasis: of mice and men. Nat Rev Endocrinol 2014; 10: 79–87. [DOI] [PubMed] [Google Scholar]
  • 31. Ford ES, Ajani UA, McGuire LC, Liu S. Concentrations of serum vitamin D and the metabolic syndrome among US adults. Diabetes Care 2005; 28: 1228–1230. [DOI] [PubMed] [Google Scholar]
  • 32. Asano L, Watanabe M, Ryoden Y, Usuda K, Yamaguchi T, Khambu B, et al Vitamin D metabolite, 25‐hydroxyvitamin D, regulates lipid metabolism by inducing degradation of SREBP/SCAP. Cell Chem Biol 2017; 24: 207–217. [DOI] [PubMed] [Google Scholar]
  • 33. Brown MS, Goldstein JL. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane‐bound transcription factor. Cell 1997; 89: 331–340. [DOI] [PubMed] [Google Scholar]
  • 34. Goldstein JL, DeBose‐Boyd RA, Brown MS. Protein sensors for membrane sterols. Cell 2006; 124: 35–46. [DOI] [PubMed] [Google Scholar]
  • 35. Radhakrishnan A, Sun LP, Kwon HJ, Brown MS, Goldstein JL. Direct binding of cholesterol to the purified membrane region of SCAP: mechanism for a sterol‐sensing domain. Mol Cell 2004; 15: 259–268. [DOI] [PubMed] [Google Scholar]
  • 36. Uesugi M. 25‐hydroxyvitamin D and lipid metabolism, oral presentation. In: 21st Workshop on Vitamin D. Abstract Book, Barcelona, Spain, 16–19 May, 2018; 72.
  • 37. Jones G, Kaufmann M, Prosser D. 25‐hydroxyvitamin D3‐24‐hydroxylase (CYP24A1): its important role in the degradation of vitamin D. Arch Biochem Biophys 2012; 523: 9–18. [DOI] [PubMed] [Google Scholar]
  • 38. Schlingmann KP, Kaufmann M, Weber S, Irwin A, Goos C, John U, et al Mutations in CYP24A1 and idiopathic infantile hypercalcemia. N Engl J Med 2011; 365: 410–421. [DOI] [PubMed] [Google Scholar]
  • 39. Seo EG, Einhorn TA, Norman AW. 24R,25‐dihydroxyvitamin D3: an essential vitamin D3 metabolite for both normal bone integrity and healing of tibial fracture in chicks. Endocrinology 1997; 138: 3864–3872. [DOI] [PubMed] [Google Scholar]
  • 40. Seo EG, Norman AW. Three‐fold induction of renal 25‐hydroxyvitamin D3–24‐hydroxylase activity and increased serum 24,25‐dihydroxyvitamin D3 levels are correlated with the healing process after chick tibial fracture. J Bone Miner Res Off J Am Soc Bone Miner Res 1997; 12: 598–606. [DOI] [PubMed] [Google Scholar]
  • 41. Kato A, Seo EG, Einhorn TA, Bishop JE, Norman AW. Studies on 24R,25dihydroxyvitamin D3: evidence for a nonnuclear membrane receptor in the chick tibial fracturehealing callus. Bone 1998; 23: 141–146. [DOI] [PubMed] [Google Scholar]
  • 42. Ebeling PR, Eisman JA. Vitamin D and osteoporosis In: Vitamin D, Third edn, eds Feldman D, Pike W, Adams J. San Diego, CA: Elsevier, 2011; 1129–1144. [Google Scholar]
  • 43. Orimo H, Shiraki M, Hayashi T, Nakamura T. Reduced occurrence of vertebral crush fractures in senile osteoporosis treated with 1α(OH)‐vitamin D3 . Bone Miner 1987; 3: 47–52. [PubMed] [Google Scholar]
  • 44. Gallagher JC, Bishop CW, Knutson JC, Mazess RB, DeLuca HF. Effects of increasing doses of 1α‐hydroxyvitamin D2 on calcium homeostasis in postmenopausal osteopenic women. J Bone Miner Res 1994; 9: 607–614. [DOI] [PubMed] [Google Scholar]
  • 45. Gallagher JC, Riggs BL, Recker RR, Goldgar D. The effect of calcitriol on patients with postmenopausal osteoporosis with special reference to fracture frequency. Proc Soc Exp Biol Med 1989; 191: 287–292. [DOI] [PubMed] [Google Scholar]
  • 46. Ott S, Chesnut CH. Calcitriol treatment is not effective in 2post‐menopausal osteoporosis. Ann Intern Med 1989; 110: 267–264. [DOI] [PubMed] [Google Scholar]
  • 47. Tilyard MW, Spears GFS, Thomson J, Dovey S. Treatment of post‐menopausal osteoporosis with calcium. N Engl J Med 1992; 326: 357–362. [DOI] [PubMed] [Google Scholar]
  • 48. Hock JM, Gunness‐Hey M, Poser J, Olson H, Bell NH, Raisz LG. Stimulation of undermineralized matrix formation by 1,25dihydroxyvitamin D3 in long bones of rats. Calcif Tissue Int 1986; 38: 79–86. [DOI] [PubMed] [Google Scholar]
  • 49. Matsumoto T, Kubodera N. ED‐71, a new active vitamin D3, increases bone mineral density regardless of serum 25(OH) D levels in osteoporotic subjects. J Steroid Biochem Mol Biol 2007; 103: 584–586. [DOI] [PubMed] [Google Scholar]
  • 50. Ke HZ, Qi H, Crawford DT, Simmons HA, Xu G, Li M, et al A new vitamin D analog, 2MD, restores trabecular and cortical bone mass and strength in ovariectomized rats with established osteopenia. J Bone Miner Res 2005; 20: 1742–1755. [DOI] [PubMed] [Google Scholar]
  • 51. DeLuca HF, Bedale W, Binkley N, Gallagher JC, Bolognese M, Peacock M, et al The vitamin D analogue 2MD increases bone turnover but not BMD in postmenopausal women with osteopenia: results of a 1‐year phase 2 double‐blind, placebo‐controlled, randomized clinical trial. J Bone Miner Res 2011; 26: 538–545. [DOI] [PubMed] [Google Scholar]
  • 52. Medioni J, Deplanque G, Ferrero JM, Maurina T, Rodier JM, Raymond E, et al Phase I safety and pharmacodynamic of inecalcitol, a novel VDR agonist with docetaxel in metastatic castration‐resistant prostate cancer patients. Clin Cancer Res 2014; 20: 4471–4477. [DOI] [PubMed] [Google Scholar]
  • 53. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D. The crystal structure of the nuclear receptor for vitamin D bound to its natural ligand. Mol Cell 2000; 5: 173–179. [DOI] [PubMed] [Google Scholar]
  • 54. Jurutka PW, Thompson PD, Whitfield GK, Eichhorst KR, Hall N, Dominguez CE, et al. Molecular and functional comparison of 1,25‐dihydroxyvitamin D(3) and the novel vitamin D receptor ligand, lithocholic acid, in activating transcription of cytochrome P450 3A4. J Cell Biochem 2005; 94: 917–943. [DOI] [PubMed] [Google Scholar]
  • 55. Ochiai E, Miura D, Eguchi H, Ohara S, Takenouchi K, Azuma Y, et al Molecular mechanism of the vitamin D antagonistic actions of (23S)‐25‐dehydro‐1alpha‐hydroxyvitamin D3–26,23‐lactone depends on the primary structure of the carboxyl‐terminal region of the vitamin D receptor. Mol Endocrinol 2005; 19: 1147–1157. [DOI] [PubMed] [Google Scholar]
  • 56. Saito N, Kittaka A. Highly potent vitamin D receptor antagonists: design, synthesis, and biological evaluation. Chembiochem 2006; 7: 1479–1490. [DOI] [PubMed] [Google Scholar]
  • 57. Toell A, Gonzalez MM, Ruf D, Steinmeyer A, Ishizuka S, Carlberg C. Different molecular mechanisms of vitamin D3 receptor antagonists. Mol Pharmacol 2001; 59: 1478–1485. [DOI] [PubMed] [Google Scholar]
  • 58. Ishizuka S, Kurihara N, Reddy SV, Cornish J, Cundy T, Roodman GD. (23S)‐25‐Dehydro‐1α‐hydroxyvitamin D3‐26,23‐lactone, a vitamin D receptor antagonist that inhibits osteoclast formation and bone resorption in bone marrow cultures from patients with Paget's disease. Endocrinology 2005; 146: 2023–2030. [DOI] [PubMed] [Google Scholar]
  • 59. Boehm MF, Fitzgerald P, Zou A, Elgort MG, Bischoff ED, Mere L, et al Novel nonsecosteroidal vitamin D mimics exert VDR modulating activities with less calcium mobilization than 1,25‐dihydroxyvitamin D3 . Chem Biol 1999; 6: 265–275. [DOI] [PubMed] [Google Scholar]
  • 60. Ma Y, Khalifa B, Yee YK, Lu J, Memezawa A, Savkur RS, et al Identification and characterization of noncalcemic, tissue‐selective, nonsecosteroidal vitamin D receptor modulators. J Clin Invest 2006; 116: 892–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Bolla NR, Marcinkowska E, Brown G, Kutner A. Retiferols: synthesis and biological activity of a conceptually novel class of vitamin D analogs. Expert Opin Ther Pat 2014; 24: 633–646. [DOI] [PubMed] [Google Scholar]
  • 62. Bijian K, Kaldre D, Wang TT, Su J, Bouttier M, Boucher A, et al Efficacy of hybrid vitamin D receptor agonist/histone deacetylase inhbitors in vitamin Dresistant triple negative 4T1 breast cancer. J Steroid Biochem Mol Biol 2018; 177: 135–139. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES