Next Article in Journal
Advances in Parkinson’s Disease Drugs
Next Article in Special Issue
Impact of Vitamin D3 Deficiency on Phosphatidylcholine-/Ethanolamine, Plasmalogen-, Lyso-Phosphatidylcholine-/Ethanolamine, Carnitine- and Triacyl Glyceride-Homeostasis in Neuroblastoma Cells and Murine Brain
Previous Article in Journal
Anticancer Platinum Drugs Update
Previous Article in Special Issue
The Effects of Vitamin D on Immune System and Inflammatory Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Synthesis and Biological Evaluation of D-Ring-Modified Vitamin D Analogues

Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
*
Author to whom correspondence should be addressed.
Biomolecules 2021, 11(11), 1639; https://doi.org/10.3390/biom11111639
Submission received: 11 October 2021 / Revised: 28 October 2021 / Accepted: 28 October 2021 / Published: 4 November 2021
(This article belongs to the Special Issue Biochemistry and Molecular Biology of Vitamin D and Its Analog)

Abstract

:
The vitamin D3 structure consists of the A-ring, a linker originating from the B-ring, C-ring, D-ring, and side-chain moieties. Each unit has its unique role in expressing the biological activities of vitamin D3. Many efforts have been made to date to assess the possible clinical use of vitamin D. Some organic chemists focused on the D-ring structure of vitamin D and synthesized D-ring-modified vitamin D analogues, and their biological activities were studied. This review summarizes the synthetic methodologies of D-ring-modified vitamin D analogues, except for seco-D, and their preliminary biological profiles.

1. Introduction

Vitamin D3 is a fat-soluble vitamin produced from 7-dehydrocholesterol through a two-step pericyclic reaction on the B-ring: a photochemical electrocyclic reaction and a subsequent thermal [1,7]-sigmatropic rearrangement. 25-Hydroxyvitamin D3 [25(OH)D3 (1)] and 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3 (2)] are important compounds responsible for various physiological functions to maintain homeostasis in the human body. Vitamin D3 is divided into five constructive units: An A-ring, a linker originating from the B-ring, a C-ring, a D-ring, and a side-chain, each of which plays an essential role in expressing vitamin D-related activities [1,2].
Among these structures, both the A-ring and side-chain, in particular, are vitamin D3 activation and inactivation sites controlled by vitamin D-metabolizing CYPs [3,4,5], and the introduction of a substituent to and the structural transformation of these sites have been vigorously pursued. On the other hand, the CD-ring does not undergo direct hydroxylation in the activation and inactivation steps of the vitamin D3 molecule (Scheme 1).
For the synthesis of vitamin D3 analogues, the CD-ring part is often used directly from the natural vitamin D3 CD-ring part due to the difficulty of constructing the chiral CD-ring structure. Therefore, it is challenging to introduce functional groups to this part or transform the CD-ring to another skeleton. As a result, the synthesis of vitamin D3 analogues with a modified CD-ring and evaluation of their biological activities are still less explored than synthesis of vitamin D analogues with a modified A-ring or side-chain. However, organic chemists have attempted the introduction of substituents and/or structural transformation of both C- and D-ring parts and have succeeded in synthesizing a group of analogues and evaluating their biological activities to date [6,7,8,9,10]. The D-ring unit is characterized by the direct attachment of the side chain, and there have been many reports that the introduction of substituents to or structural transformation of the D-ring has significant effects on vitamin D activities.
In this review, we focus on the D-ring, which is one of the essential sites forming the basic framework of the vitamin D3 molecule. We describe the synthetic approach to vitamin D3 analogues with substituents or structural transformation on the D-ring, except for seco-D analogues, and their preliminary biological activities.

2. 16-Ene-Vitamin D3 Analogues

Among D-ring-modified vitamin D3 analogues, a wide variety of 16-ene-vitamin D3 analogues have been synthesized and evaluated regarding their biological properties as potential anticancer agents with low calcemic effects.
The first synthetic report on the 16-ene-vitamin D3 analogue (3) was described by Hoffmann-La Roche’s group in 1995 [11], and its biological activities were studied [12,13,14,15]. The synthetic route started from a commercially available steroid, dehydroepiandrosterone (4). The 16-ene unit was constructed utilizing an ene reaction between Z-olefin (5) and aldehyde (6) in the presence of Me2AlCl to give a mixture of C22-diastereomeric alcohols (7), and subsequent Barton deoxygenation afforded 16-en-23-yne (8). To construct the triene structure, photochemical conversion and subsequent thermal isomerization were applied on the B-ring diene system (Scheme 2). The final product, 3, was identified as a potential antipsoriatic agent at that time.
Posner and coworkers reported the synthesis of 16-ene-vitamin D3 analogues with the 1-hydroxymethyl group (9–14) and 24-oxo-16-ene vitamin D3 analogue (15) in 1997 (Scheme 3 and Scheme 4) [16,17].
In Scheme 3, the synthesis of 1-hydroxymethyl analogues (9–12) is described. Introduction of the key 16-ene unit was accomplished by a Me2AlCl-mediated ene reaction between olefin (16) and formaldehyde to afford alcohol (17). Next, 17 was converted to iodide (18), followed by the Zn/Ni-promoted Michael addition of iodide (18) to ethyl acrylate to produce ethyl ester (19). The ethyl ester (19) was treated with methyl magnesium bromide or ethyl magnesium bromide to give alcohol (20,21). After oxidation of 20 and 21, protection of the C25-hydroxy group afforded 8-keto-CD-ring (22) and (23). Each ketone (22,23) and the racemic A-ring moiety (24) were coupled using PhLi, and then protecting groups of the coupling products were removed in the presence of TBAF to afford 16-ene analogues (9–12) (Scheme 3).
For the synthesis of 24-oxo-16-ene analogues (1315), alcohol (17) was used as a starting material (Scheme 4). The synthesis began with the oxidation of primary alcohol to give aldehyde (25), followed by the addition of 26 to aldehyde (25) under basic conditions, to afford a mixture of C22-diastereomeric alcohols. Removal of the C22-hydroxy group using the Barton reaction yielded 24-oxo compound 27, which was converted to 8-keto-CD-ring (28) in four steps. Coupling reactions using 28 and A-rings (24,29) were performed under the same conditions as in Scheme 3. Under the coupling conditions, the 24-oxo unit was inert, i.e., no chemical conversion was observed at the C24 position.
The authors evaluated the antiproliferative activity of the newly synthesized analogues (10,12,14,15) in murine keratinocytes in vitro and found that the 26,27-diethyl-1β-hydroxymethyl analogue 12 and the 24-oxo with a natural A-ring analogue 15 showed activity comparable with that of the natural hormone 1α,25(OH)2D3 (2), even at physiologically relevant nanomolar concentrations. The 22-oxo-analogue 15 was an intermediary metabolite of 16-ene-1α,25(OH)2D3 (75 in Figure 1) formed through the (24R)-oxidation pathway (Scheme 1) with a longer half-life than the naturally occurring 24-oxo-1α,25(OH)2D3 and equipotent to parent 75 itself in regulating cell-growth and differentiation [18].
The same authors subsequently reported the 1-hydroxymethyl-24,24-difluoro-16-ene analogues (3033) in 1998 [19]. The 16-ene-homoallylic alcohol (35) provided by an ene reaction of olefin (34) was first converted to tosylate via tosylation of the primary hydroxy group and triethylsilylation of the secondary hydroxy group. Tosylate was converted to nitrile upon treatment with KCN, followed by reduction with DIBAL-H to give aldehyde (36). Introduction of the 24,24-difluoro unit was accomplished with the Reformatsky reaction using ethyl bromodifluoroacetate and activated zinc powder. Treatment of ester (37) with methyl lithium or ethyl lithium, followed by fluoride-induced deprotection at the C-8 position generated the natural side-chain-CD-ring (38) and 26,27-dihomo-CD-ring (39), which were converted to 8-keto-CD-rings (40,41) in two steps. A Wittig–Horner coupling reaction between a racemic phosphine oxide (24) and the 8-keto-CD-rings (40,41), followed by deprotection of the silyl protecting groups, produced the target analogues (3033) (Scheme 5).
The 1β-analogues (31,33) showed significant antiproliferative activity in murine keratinocytes and malignant melanoma cells, and this was equally potent to or even more potent than 1α,25(OH)2D3 (2). On the other hand, 1α-analogues (30,32) were much less potent than their 1β-counterparts (31,33). In addition, 31 and 33 showed no calcemic activity in vivo.
In 1999, 16-ene analogues with a sulfone unit (42–44,47,48) and their C24 fluorinated versions (45,46,49,50) were also reported (Scheme 6, Scheme 7, Scheme 8 and Scheme 9) [20].
For the synthesis of analogue 42, the known aldehyde (36) was used as a starting material (Scheme 6). Reduction of aldehyde (36) and iodination of the primary alcohol yielded iodide (51). After conversion of the iodide to sulfide (52), oxidation of the sulfide using oxone provided sulfone (53), and subsequent PDC oxidation of the C8-OH group gave ketone (54). Finally, the convergent coupling reaction to form the desired analogue (42) was performed with the ketone (54) and A-ring (29) carbanion (Scheme 6).
Next, one-carbon-elongated analogues (43,44) and their C24-fluorinated versions (45,46) were prepared in a similar manner. Treatment of iodide (51) with t-butyl methyl sulfone under basic conditions gave a 25-sulfone CD-ring (52). Introduction of the difluoro unit to the C24 position of sulfone (52) was achieved by utilizing a cationic fluorination reagent in the presence of nBuLi to give 53. Desilylation of 52 and 53, followed by oxidation at the C8-OH group, afforded ketones (54,55). Finally, these ketones (54,55) were converted into the desired vitamin D3 analogues (45–48) using the convergent coupling reaction with a lithium anion of the racemic A-ring moiety (56) (Scheme 7).
Synthesis of 23-ene analogues (47,48) started from aldehyde (36) (Scheme 8). Treatment of 36 with t-butyl methyl sulfone under basic conditions afforded C23-OH adducts (57). Mesylation of C23-OH, followed by elimination gave olefin (58). Although deprotection of the TES protective group was problematic using TBAF because of the Michael addition of fluoride to the C23 position, HF-MeCN instead of TBAF gave 59 in a nearly quantitative yield. The resulting alcohol (59) was subjected to PDC oxidation to afford ketone (60). Finally, 60 was converted to the desired vitamin D3 analogues (47,48) using the same method as above in Scheme 7.
Synthesis of 23-ene-24-fluoro analogues (49,50) is illustrated in Scheme 9. According to a similar methodology to that described in Scheme 8, an intermediate 23-hydroxy-24-fluoro-CD-ring (61) was prepared by the nucleophilic addition of t-butyl fluoromethyl sulfone to aldehyde (36) under basic conditions. Mesylation of the C23-hydroxy group and subsequent E2 elimination gave 23-ene-24-fluoro-olefin (62), which was converted to 23-ene-24-fluoro analogues (49,50) in four steps.
Antiproliferative activities of the newly synthesized 16-ene-vitamin D3 analogues (42–45,47,49) were tested in both murine keratinocytes and malignant melanoma cells in vitro. The data revealed that almost all analogues showed activities equivalent to or even stronger than those of 1α,25(OH)2D3 (2).
Next, the vitamin D receptor-mediated transcriptional activity of three analogues (42,45,49) in ROS 17/2.8 cells was evaluated, and 42 and 45 showed strong transcriptional activity only approximately twofold less active than that of 1α,25(OH)2D3 (2). For practical chemotherapeutic uses, the calcemic activity of five analogues (42,43,45,47,49) was tested in vivo, revealing that 42, 45, and 49 did not show calcemic activity at a 10 μg/kg dose for 1 week. In contrast, 47 was moderately calcemic, and 43 showed strong calcemic activity [20].
The 26,27-fluorinated-16-ene (63,64,66,69,71,73,80–83,85,87,90,9295) and non-fluorinated (3,65,67,68,70,72,74–79,84,86,88,89,91) analogues have been synthesized to date (Figure 1). Uskoković et al. published a comprehensive review article in this field, including synthetic routes and biological activities [21].
Briefly, the preparation of 16-ene-CD-ring moieties (104–118) is illustrated in Scheme 10. First, key intermediates (96,97) were synthesized from olefins (98,99), and then these were treated with hexafluoroacetone (HFA) (100) or 1,1,1-trifluoroacetone (101) under basic conditions to afford HFA-adducts and 26,26,26-trifluoroacetone-adducts, respectively. Hydrogenation of the 23,24-triple bond of the HFA-adducts with Lindlar catalyst gave 23,24-Z-olefins (108,109). On the other hand, reduction of the HFA-adducts and 26,26,26-trifluoroacetone-adducts with LiAlH4 in the presence of NaOCH3 afforded 23,24-E-olefins (106,107,110,111). Seven non-fluorinated CD-rings (112–118) were prepared using the same methodology. These synthetic CD-ring precursors (104–118) were coupled with the A-ring moieties to give 16-ene analogues (3,63–95) in Figure 1.
These 16-ene analogues possessed potential activity to induce HL-60 cell differentiation. The 1α-hydroxy analogues (69–75,80–89) showed stronger activity (IC50, 0.1–1.6 nM) than 1-deoxy analogues (3,63–68) (25– >1000 nM), 3-deoxy analogues (76–79) (6.5–50.0 nM), and 1α-fluoro analogues (90–95) (1.8–12.0 nM) [21,22].
Mouriño and coworkers described convergent synthetic routes for 16-ene-vitamin D3 analogues (119–122) in 1999 (Scheme 11 and Scheme 12) [23].
The synthesis of a C20-epi form of the 16-ene analogue (119) is shown in Scheme 11. The known C17-ketone (123) was used as a starting material, and stereoselective introduction of the 17Z-double bond was accomplished by a Wittig reaction. Treatment of carboxylic acid (124) with methyl lithium twice, followed by protection of the resulting C25-hydroxy group, afforded the desired CD-ring (125). The stereoselective oxidation of 125 with selenium dioxide afforded a C16-hydroxy-CD-ring (126). The C16-hydroxy group was subsequently converted to a leaving group, and the syn-SN2’ type reaction using a higher-order lithium cuprate (Li2Cu3R5, R = Me, nBu, cPr, tBu, Ph) gave varieties of C20-epi-16-ene-CD-rings (128–132). Removal of the TBS protective group of 128 and subsequent PDC oxidation afforded ketone (133). Synthesis of a C20-epi-16-ene analogue (119) with the triene structure was achieved by the convergent method.
It is possible to construct C20 natural form 16-ene-CD-rings using 17Z-carbamate (135) for a SN2’ syn-displacement reaction (Scheme 12). The key step for the C17-20 double-bond conversion from 17E (126) to 17Z (135) was accomplished by the Vedejs method [24]. Stereoselective epoxidation of 126, followed by the addition of lithium diphenylphosphine and subsequent elimination, gave 17Z-CD-ring (135). From 135, C20 natural type 16-ene analogues (120–122) were prepared, as shown in Scheme 11 (Scheme 12).
The synthesis and biological activities of C20-cyclopropyl-16-ene-1α,25-dihydroxyvitamin D3 analogues (136,137) were reported by Uskoković et al. in 2006 [25]. As shown in Scheme 13, 136 and a 19-nor type of 137 were prepared from ketone 138. After introduction of the cyclopropyl unit by the Wittig reaction, reduction and TBS protection gave 140. The 16-ene unit formation was accomplished by an ene reaction with formaldehyde to afford 141. The alcohol 141 was subsequently oxidized with pyridinium chlorochromate, and the formed aldehyde was converted to alkyne 143 in three steps. Acetylide prepared from alkyne 143 was reacted with acetone, followed by deprotection of the O-TBS group and reduction of the alkynyl moiety to give C25-OH-CD-ring 144. Oxidation of the C8-OH group, followed by trimethylsilylation of the C25-OH group, afforded 8-ketone 145. Finally, synthesis of 16-ene analogues (136,137) was achieved in a convergent manner by connecting with A-ring (29 or 146).
Immunomodulatory activity of C20-cyclopropyl-16-ene-1α,25-dihydroxyvitamin D3 analogues (136,137), the parent analogue (C20-cyclopropyl-1α,25(OH)2D3) (147), and 1α,25(OH)2D3 (2) was tested by suppression of interferon-γ (IFN-γ) release. Analogue (136) was found to be 45-times more potent than its parent analogue (147) and also 2440-times more active than 1α,25(OH)2D3 (2).
A metabolism study of 136 using the rat osteosarcoma cell line (UMR106) revealed that three main metabolites were detected from the HPLC profiles. Comparing the metabolic studies of 136 and 147, the authors concluded that these three metabolites were 24-hydroxy-136 (148), 24-oxo-136 (149), and 3-epi-136 (150) (Figure 2).
In 2009, the 16-ene-24-oxo-1α,25-dihydroxyvitamin D3 (149) was synthesized from the key intermediate 143 by a sequence of reactions illustrated in Scheme 14 [26]. Acetylide from alkyne 143 was reacted with acetone, followed by hydration of the resulting alkyne moiety using HgO in the presence of H2SO4 to obtain 24-oxo-CD-ring (151). The 24-oxo-CD-ring (151) was subsequently coupled with the A-ring (29) to give 16-ene-24-oxo-1α,25-dihydroxyvitamin D3 (149).
The authors compared the VDR-dependent CYP24A1 and cathelicidin antimicrobial peptide (CAMP) transcriptional activities of the 24-oxo analogue (149), its metabolic precursor 136, and the natural hormone, 1α,25(OH)2D3 (2), using peripheral blood mononuclear cells (PBMCs) as well as human acute monocytic leukemia cells (THP-1 cells), and revealed that both 16-ene analogues (136,149) exhibited similar transcriptional activities (EC50 ~0.7 nM) and greater activities than the natural hormone, 1α,25(OH)2D3 (2) (EC50 ~10 nM).
To compare the anti-inflammatory properties of 136, 149, and 2, the authors tested their potency to inhibit the production of cytokines (IFN-γ, TNF-α, IL-12/23p40, and IL-6) in vitro, and concluded that 136 and its 24-oxo metabolite 149 had a similar activity and significantly higher potency compared to the natural hormone (2).
Evaluation of the calcemic activity of 136, 149, and 2 revealed that 136 and 149 were less calcemic than 2, and 149 was the least calcemic among the three analogues.

3. 16-Modified Vitamin D3 Analogues

In 2003, the C16-substituted analogue 152 and its C20-epi-version 153 were reported by the LEO Pharma group in order to produce specific antibodies to 1α,25(OH)2D3 (2) and its C20-epi form (Scheme 15) [27]. The CD-ring synthons 154 and 155 were constructed from the known ketone 156. Reduction of 156 with NaBH4 yielded a mixture of (20R)-OH-CD-ring (157) and its 20S-isomer (158) in a ratio of 85:15. After separation of each isomer, 157 and 158 were converted to C20-natural form CD-ring (161) and C20-epi form CD-ring (162), respectively, in two steps. Tosylation of 161 and 162, followed by nucleophilic substitution to give 163 and C20-epi 164. The key step for introducing the C16-OH group was achieved by hydroboration/oxidation of 163 and C20-epi 164 to afford C16α-OH 165 and C20-epi-16α-OH 166 along with C16β-OH isomers as the minor products. Both C16α-OH-CD-rings (165,166) were converted to 8-keto-CD-ring (154) and C20-epi-8-keto-CD-ring (155), respectively, in four steps. The Wittig–Horner coupling reaction with the lithium salt of the A-ring precursor 29 gave the coupling products. The acetate group of the coupling products was replaced with glutaric acid esters, followed by deprotection with TBAF to afford 152 and 153.
Both analogues (152,153) were coupled with bovine serum albumin (BSA), and polyclonal antisera from the rabbits were obtained [28]. The antibodies to 152 had selective binding affinity for 1α,25(OH)2D3 (2) that was 12- to 13-times greater than for C20-epi-1α,25(OH)2D3. In contrast, antibodies to 153 exhibited poor selectivity between 1α,25(OH)2D3 (2) and C20-epi-1α,25(OH)2D3.
In 2019, Okamoto and coworkers reported the synthesis of 16-oxa-vitamin D3 analogues (169,170) [29]. They synthesized the 16-oxa-CD-ring moiety (171) starting from 1-chloro-3-methylbut-2-ene (172) in 20 steps (Scheme 16). For the construction of CD-ring parts, a Ti(II)-mediated enyne cyclization/Cu-catalyzed allylation and a subsequent ring-closing metathesis reaction (RCM) were applied. The synthetic 171 was coupled with the A-ring moieties (173,174) in the presence of PdCl2(dppf) and KOH to produce 16-oxa-vitamin D3 analogues (169,170). The VDR binding affinity of the newly synthesized 16-oxa analogues 169 and 170 was evaluated using the fluorescence polarization vitamin D-receptor competitor assay and time-resolved fluorescence resonance energy transfer VDR co-activator assay in comparison with those of the natural hormone (2) and its 19-nor form. It was demonstrated that both 169 and 170 were potent analogues. However, they were less potent than both the natural hormone (2) and its 19-nor-form.

4. Decalin-Vitamin D analogues

The natural hormone, 1α,25(OH)2D3 (2), contains a trans-hydrindane CD-ring fragment because of the biological synthetic pathway starting from lanosterol as the original steroidal skeleton. In 1996, Vandewalle et al. expanded the five-membered D-ring to a six-membered ring, and the CD-ring of the new vitamin D consisted of a trans-decalin ring system as shown in Scheme 17 [30]. (S)-Wieland–Miescher ketone 175 [31] was converted to trans-5-oxo-decalin 177a with the natural 20R-configuration at the vitamin D3 side chain and also 179a with the unnatural 20S-configuration side chain. During the synthesis of 177a and 179a, cis-fused decalins (177b, 179b) were also produced; however, treatment of the mixture of 5-oxo-decalins under basic conditions afforded trans-decalins as the major products in their equilibriums, respectively. These decalin-type CD-rings were connected to the A-ring precursors (29 or 146), respectively, to yield the new class of 1α,25(OH)2D3 analogues 180 and 182 as well as the 19-nor-versions 181 and 183. It is noteworthy that the natural 20R-configuration analogues of 180 and 181 showed higher VDR binding affinity, which was almost at the same level as that of 1α,25(OH)2D3 (2), than their 20-epi-counterparts 182 and 183, and the cell differentiation and proliferation activity of 180 and 181 was one order of magnitude higher than that of 2; however, 20-epi-counterparts 182 and 183 possessed weaker activity than 2. The trans-decalin pair represented one of the rare examples that the 20-epimer exhibited reduced biological activities when compared with the analogues with the natural 20R-configuration [32].
De Clercq’s group developed the trans-decalin core structure for pseudo-S2-symmetrical vitamin D analogues (188,190) [33]. For the synthesis, bis-triflate 185 was prepared from crystalline diketone 184 [34], and enyne 186 was connected in two-ways using Suzuki–Miyaura coupling and Sonogashira coupling reactions followed by appropriate chemical treatments to afford 188 and 190 in crystalline form, respectively (Scheme 18). However, these analogues did not show VDR-binding and antiproliferative activities.
As shown in Figure 3, the De Clercq group also developed various CD-ring modified analogues 191–195 [35,36,37]. These spiro-analogues showed low calcemic activity. From a natural product diosgenin, a spiro-vitamin D analogue 196 was synthesized by a Japanese group in 2005, and this compound, 196, induced apoptosis in Hep G2 cells through activation of p53 and Bax mRNAs [38].
Verlinden et al. published excellent review articles on CD-ring modified analogues of vitamin D, which included a D-ring- or C-ring lacking vitamin D molecules, and interestingly, these analogues showed moderate vitamin-D-related biological activities [39,40].

5. 15-Substituted Vitamin D3 Analogues

15-Hydroxyvitamin D3 and its derivatives were synthesized and reported by our group in 2011; i.e., 1α,15α,25-trihydroxyvitamin D3 (212) and 15α-methoxy-1α,25(OH)2D3 (213) as well as the 16-ene versions (220,221) were designed, synthesized, and biologically tested [41].
As shown in Scheme 19, vinyl acetate 197 from the known ketone 123 through transacetylation using isopropenyl acetate reacted with allyl methyl carbonate in the presence of Pd(OAc)2 and Bu3SnOMe to afford α,β-unsaturated ketone 198. The enone 198 was reduced with DIBAL-H to give allyl alcohol 199, stereoselectively. Next, mCPBA epoxidation afforded α-epoxide 200, whose stereochemistry was determined by X-ray crystallographic analysis of its acetate 201. Oxidation of 200 afforded 17-oxo-α-epoxide 202, and a Wittig reaction gave (17Z)-ethylidene 203 as a single isomer, although it was reported that 17-oxo-β-epoxysterols afforded only (17E)-ethylidene by the corresponding Wittig reaction [42,43,44]. 5-Bromo-2-methyl-2-pentanol MOM ether was converted to magnesium cyanocuprate, and 1,4-addition to ethylidene epoxide 203 yielded the 15α-hydroxy-16-ene-CD ring 204 with natural 20R- and unnatural 20S-configurations in a ratio of 11:1. The major isomer 204a results from attack of the alkyl cuprate on the β-face of (17Z)-alkene 181 in an SN2’ manner. The D-ring double bond of 204a was hydrogenated to afford 205 with a natural 17R- and unnatural 17S-configuration in a ratio of 14:1.
The secondary hydroxy group at C15 of 205a was methoxymethylated (206) or methylated (207), and C8-OH oxidation after deprotection afforded C8-ketone (Scheme 20). Ketones 208 and 209 were connected with A-ring phosphine oxide 29 by the Wittig–Horner reaction to yield the coupling products 210 and 211, respectively. The subsequent deprotection gave the C15-modified analogues 212 and 213. The C15-substituted 16-ene-vitamin D3 analogues were also available from 204a, which was converted to bromoolefin, 215, in four steps. A Trost coupling reaction with enyne 216 yielded 16-ene analog 218, which was deprotected to afford the desired 15-hydroxy-16-ene-analogue 220. To study the synergetic effects of the 2α-methyl group and the 16-ene-structure on biological activity, compound 221 was next synthesized using our original enyne 217 (Scheme 18) [45,46,47,48,49].
The basic biological activity of the novel compounds 212, 213, 220, and 221 was tested: (1) VDR binding affinity was 13% (212), 1.5% (213), 65% (220), and 278% (221) of that of 1α,25(OH)2D3 (2). (2) EC50 values of transactivation activity of the osteocalcin promoter in HOS cells were 0.12, 0.12, 0.08, and 0.12 nM, respectively, with 0.09 nM of 2. Interestingly, 16-ene analogs 220 and 221 exhibited comparable and even greater affinity for VDR and transactivation activity than the natural hormone (2) [41,50].

6. Conclusions

Actually, the 16-ene structure of the CD-ring part of vitamin D3 first appeared in the total synthesis of 1α,25(OH)2D3 (2) reported by the Hoffmann-La Roche group in 1982 [51]. To elongate the side chain at C20 from the (17Z)-ethylidene CD-ring creating the 20R-natural stereochemistry, they utilized the ene reaction with ethyl propionate in the presence of Lewis acid EtAlCl2. After the successful ene reaction, the first 16-ene structure was obtained, but the double bond was then reduced stereo-selectively to the saturated CD-ring system that was included in the natural active vitamin D3 skeleton. A great variety of unique 16-ene vitamin D analogs were synthesized using the ene reaction, and their biological activities were evaluated by Hoffmann-La Roche group and Posner’s group as shown in Section 2. It was proved that 16-ene-1α,25(OH)2D3 had higher binding affinity for the VDR, lower affinity for the vitamin D binding protein in circulation, and greater resistance to 24-oxo-mediated catabolism as compared with 1α,25(OH)2D3. Another strategy for 16-ene construction was developed by Mouriño’s group in 1999. They utilized SN2’-syn-facial displacement toward (17Z)-olefin carbamate from 135 by high-order cuprates (Li2Cu3Me5, etc.) to generate the 20R-natural configuration and the 16-ene double bond with high yield. The leaving group, a carbamate group, for the SN2’ reaction was located at the C16 position, and various types of substituents could be introduced at the C20 position, stereo-selectively, by this route. On the other hand, examples of 16-modified vitamin D analogues were few as described in Section 3, and only hydroxylated analogues were synthesized in which Mouriño’s synthetic intermediate was included as above. The unique example was Okamoto’s 16-oxa analogues of 1α,25(OH)2D3, even though these analogues exhibited weaker VDR binding affinity. Vandewalle and De Clercq developed a trans-decalin CD-ring system in the vitamin D structure, and these analogues showed potent biological activities, which is summarized in Section 4. Finally, our synthetic 15-substituted vitamin D3 analogues that possessed moderate biological activity were described in Section 5, but combination with 16-ene modification brought higher potency in VDR binding and osteocalcin transactivation activity than the natural hormone 2. Although it is known that seco-D-ring system as vitamin D analogues [39], this review covers real ring systems as the vitamin D derivatives with the chemically modified D-ring. Modification on the vitamin D skeleton is worthy of challenging, and only real synthetic molecules are able to tell us benefits for the specific disease treatment.

Author Contributions

Conceptualization, F.K. and A.K.; writing—original draft preparation, F.K. and S.M.; writing—review and editing, A.K.; supervision, A.K.; funding acquisition, A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the grant-in-aid from the Japan Society for the Promotion of Science (no. 18K06556 to AK).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Posner, G.H.; Kahraman, M. Overview: Rational design of 1α,25-dihydroxyvitamin D3 analogs (Deltanoids). In Vitamin D, 2nd ed.; Feldman, D., Pike, J.W., Glorieux, F.H., Eds.; Elsevier Academic Press: Burlington, MA, USA, 2005; Volume 2, pp. 1405–1422. [Google Scholar]
  2. Kulesza, U.; Sigüeiro, R.; Mouriño, A.; Sicinski, R.R. Synthesis of 9-alkylated calcitriol and two 1α,25-dihydroxy-9-methylene-10,19-dihydrovitamin D3 analogues with a non-natural triene system by thermal sigmatropic rearrangements. J. Org. Chem. 2013, 78, 1444–1450. [Google Scholar] [CrossRef] [PubMed]
  3. Rochel, N.; Wurtz, J.M.; 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. [Google Scholar] [CrossRef]
  4. Sakaki, T.; Sawada, N.; Komai, K.; Shiozawa, S.; Yamada, S.; Yamamoto, K.; Ohyama, Y.; Inouye, K. Dual metabolic pathway of 25-hydroxyvitamin D3 catalyzed by human CYP24. Eur. J. Biochem. 2000, 267, 6158–6165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sakaki, T.; Kagawa, N.; Yamamoto, K.; Inouye, K. Metabolism of vitamin D3 by cytochromes P450. Front. Biosci. 2005, 10, 119–134. [Google Scholar] [PubMed] [Green Version]
  6. Yasuda, K.; Nishikawa, M.; Okamoto, K.; Horibe, K.; Mano, H.; Yamaguchi, M.; Okon, R.; Nakagawa, K.; Tsugawa, N.; Okano, T.; et al. Elucidation of metabolic pathways of 25-hydroxyvitamin D3 mediated by CYP24A1 and CYP3A using Cyp24a1 knockout rats generated by CRISPR/Cas9 system. J. Biol. Chem. 2021, 296, 100668. [Google Scholar] [CrossRef] [PubMed]
  7. Ordentlich, P.; Heyman, R.A. Nonsteroidal Analogs. In Vitamin D, 2nd ed.; Feldman, D., Pike, J.W., Glorieux, F.H., Eds.; Elsevier Academic Press: Burlington, MA, USA, 2005; Volume 2, pp. 1557–1567. [Google Scholar]
  8. Kulesza, U.; Plum, L.A.; DeLuca, H.F.; Mouriño, A.; Sicinski, R.R. Novel 9-alkyl- and 9-alkylidene-substituted 1α,25-dihydroxyvitamin D3 analogues: Synthesis and biological examinations. J. Med. Chem. 2015, 58, 6237–6247. [Google Scholar] [CrossRef]
  9. Gogoi, P.; Seoane, S.; Sigüeiro, R.; Guiberteau, T.; Maestro, M.A.; Pérez-Fernández, R.; Rochel, N.; Mouriño, A. Aromatic-based design of highly active and noncalcemic vitamin D receptor agonists. J. Med. Chem. 2018, 61, 4928–4937. [Google Scholar] [CrossRef] [PubMed]
  10. Zhou, X.; Zhu, G.-D.; Van Haver, D.; Vandewalle, M.; De Clercq, P.J.; Verstuyf, A.; Bouillon, R. Synthesis, biological activity, and conformational analysis of four seco-D-15,19-bisnor-1α,25-dihydroxyvitamin D analogues, diastereomeric at C17 and C20. J. Med. Chem. 1999, 42, 3539–3556. [Google Scholar] [CrossRef]
  11. Okabe, M.; Sun, R.-C.; Scalone, M.; Jibilian, C.H.; Hutchings, S.D. Synthesis of 25-hydroxycholecalcifer-16-en-23-ynol: A potential antipsoriatic agent. J. Org. Chem. 1995, 60, 767–771. [Google Scholar] [CrossRef]
  12. Zhou, J.-Y.; Norman, A.W.; Chen, D.-L.; Sun, G.-W.; Uskokovic, M.; Koeffler, H.P. 1,25-Dihydroxy-16-ene-23-yne-vitamin D3 prolongs survival time of leukemic mice. Proc. Natl. Acad. Sci. USA 1990, 87, 3929–3932. [Google Scholar] [CrossRef] [Green Version]
  13. Jung, S.A.; Lee, Y.Y.; Pakkala, S.; de Vos, S.; Elstner, E.; Norman, A.W.; Green, J.; Uskokovic, M.; Koeffler, H.P. 1,25(OH)2-16-ene-vitamin D3 is a potent antileukemic agent with low potential to cause hypercalcemia. Leuk. Res. 1994, 18, 453–463. [Google Scholar] [CrossRef]
  14. Rao, L.G.; Sutherland, M.K.; Reddy, G.S.; Siu-Caldera, M.-L.; Uskokovic, M.R.; Murray, T.M. Effects of 1α,25-dihydroxy-16ene, 23yne-vitamin D3 on osteoblastic function in human osteosarcoma SaOS-2 cells: Differentiation-stage dependence and modulation by 17-β estradiol. Bone 1996, 19, 621–627. [Google Scholar] [CrossRef]
  15. Chen, T.C.; Persons, K.; Uskokovic, M.R.; Horst, R.L.; Holick, M.F. An evaluation of 1,25-dihydroxyvitamin D3 analogues on the proliferation and differentiation of cultured human keratinocytes, calcium metabolism and the differentiation of human HL-60 cells. J. Nutr. Biochem. 1993, 4, 49–57. [Google Scholar] [CrossRef]
  16. Posner, G.H.; Lee, J.K.; White, M.C.; Hutchings, R.H.; Dai, H.; Kachinski, J.L.; Dolan, P.; Kensler, T.W. Antiproliferative hybrid analogs of the hormone 1α,25-dihydroxyvitamin D3: Design, synthesis, and preliminary biological evaluation. J. Org. Chem. 1997, 62, 3299–3314. [Google Scholar] [CrossRef] [PubMed]
  17. Honzawa, S.; Yamamoto, Y.; Yamashita, A.; Sugiura, T.; Kurihara, M.; Arai, M.A.; Kato, S.; Kittaka, A. The 2α-(3-hydroxypropyl) group as an active motif in vitamin D3 analogues as agonists of the mutant vitamin D receptor (Arg274Leu). Bioorg. Med. Chem. 2008, 16, 3002–3024. [Google Scholar] [CrossRef] [PubMed]
  18. Siu-Caldera, M.-L.; Clark, J.W.; Santos-Moore, A.; Peleg, S.; Liu, Y.Y.; Uskoković, M.R.; Sharma, S.; Reddy, G.S. 1α,25-dihydroxy-24-oxo-16-ene vitamin D3, a metabolite of a synthetic vitamin D3 analog, 1α,25-dihydroxy-16-ene vitamin D3, is equipotent to its parent in modulating growth and differentiation of human leukemic cells. J. Steroid Biochem. Mol. Biol. 1996, 59, 405–412. [Google Scholar] [CrossRef]
  19. Posner, G.H.; Lee, J.K.; Wang, Q.; Peleg, S.; Burke, M.; Brem, H.; Dolan, P.; Kensler, T.W. Noncalcemic, antiproliferative, transcriptionally active, 24-fluorinated hybrid analogues of the hormone 1α,25-dihydroxyvitamin D3. Synthesis and preliminary biological evaluation. J. Med. Chem. 1998, 41, 3008–3014. [Google Scholar] [CrossRef] [PubMed]
  20. Posner, G.H.; Wang, Q.; Han, G.; Lee, J.K.; Crawford, K.; Zand, S.; Brem, H.; Peleg, S.; Dolan, P.; Kensler, T.W. Conceptually new sulfone analogues of the hormone 1α,25-dihydroxyvitamin D3: Synthesis and preliminary biological evaluation. J. Med. Chem. 1999, 42, 3425–3435. [Google Scholar] [CrossRef] [PubMed]
  21. Uskoković, M.R.; Studzinski, G.P.; Gardner, J.P.; Reddy, S.G.; Campbell, M.J.; Koeffler, H.P. The 16-ene vitamin D analogs. Curr. Pharm. Des. 1997, 3, 99–123. [Google Scholar]
  22. Uskoković, M.R.; Studzinski, G.P.; Reddy, G.S. The 16-ene vitamin D analogs. In Vitamin D; Feldman, D., Glorieux, F.H., Pike, J.W., Eds.; Academic Press: New York, NY, USA, 1997; pp. 1045–1070. [Google Scholar]
  23. Rey, M.A.; Martínez-Pérez, J.A.; Fernández-Gacio, A.; Halkes, K.; Fall, Y.; Granja, J.; Mouriño, A. New synthetic strategies to vitamin D analogues modified at the side chain and D ring. Synthesis of 1α,25-dihydroxy-16-ene-vitamin D3 and C-20 analogues. J. Org. Chem. 1999, 64, 3196–3206. [Google Scholar]
  24. Fuchs, P.L.; Vedejs, E. Olefin inversion by the phosphorus betaine method. J. Am. Chem. Soc. 1971, 93, 4070–4072. [Google Scholar]
  25. Uskoković, M.R.; Manchand, P.; Marczak, S.; Maehr, H.; Jankowski, P.; Adorini, L.; Reddy, G.S. C-20 Cyclopropyl vitamin D3 analogs. Curr. Top. Med. Chem. 2006, 6, 1289–1296. [Google Scholar] [CrossRef] [PubMed]
  26. Laverny, G.; Penna, G.; Uskoković, M.; Marczak, S.; Maehr, H.; Jankowski, P.; Ceailles, C.; Vouros, P.; Smith, B.; Robinson, M.; et al. Synthesis and anti-inflammatory properties of 1α,25-dihydroxy-16-ene-20-cyclopropyl-24-oxo-vitamin D3, a hypocalcemic, stable metabolite of 1α,25-dihydroxy-16-ene-20-cyclopropyl-vitamin D3. J. Med. Chem. 2009, 52, 2204–2213. [Google Scholar] [CrossRef] [PubMed]
  27. Blæhr, L.K.A.; Björkling, F.; Calverley, M.J.; Binderup, E.; Begtrup, M. Synthesis of novel hapten derivatives of 1α,25-dihydroxyvitamin D3 and its 20-epi analogue. J. Org. Chem. 2003, 68, 1367–1375. [Google Scholar] [PubMed]
  28. Hosoda, H.; Sakai, Y.; Yoshida, H.; Miyairi, S.; Ishii, K.; Nambara, T. The preparation of steroid N-hydroxysuccinimide esters and their reactivities with bovine serum albumin. Chem. Pharm. Bull. 1979, 27, 742–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ibe, K.; Yamada, T.; Okamoto, S. Synthesis and vitamin D receptor affinity of 16-oxa vitamin D3 analogues. Org. Biomol. Chem. 2019, 17, 10188–10200. [Google Scholar] [CrossRef]
  30. Chen, Y.-J.; De Clercq, P.; Vandewalle, M. Synthesis of new vitamin D3 analogues with a decalin-type CD-ring. Tetrahedron Lett. 1996, 37, 9361–9364. [Google Scholar] [CrossRef]
  31. Lutz, F.T.; Utecht, J. Improved preparation of enantiomerically pure (+)-(4aS,5S)-5-tert-butoxy-4a-methyl-4,4a,5,6,7,8-hexahydro-2(3H)-naphthalenone. Synthesis 1993, 957–958. [Google Scholar]
  32. Chen, Y.-J.; Gao, L.-J.; Murad, I.; Verstuyf, A.; Verlinden, L.; Verboven, C.; Bouillon, R.; Viterbo, D.; Milanesio, M.; Van Haver, D.; et al. Synthesis, biological activity, and conformational analysis of CD-ring modified trans-decalin 1α,25-dihydroxyvitamin D analogs. Org. Biomol. Chem. 2003, 1, 257–267. [Google Scholar] [CrossRef]
  33. Minne, G.; Verlinden, L.; Verstuyf, A.; De Clercq, P.J. Synthesis of 1α,25-dihydroxyvitamin D analogues featuring a S2-symmetric CD-ring core. Molecules 2009, 14, 894–903. [Google Scholar] [CrossRef] [Green Version]
  34. Minne, G.B.; De Clercq, P.J. Synthesis of 3,4,7,8-tetrahydronaphthalene-1,5(2H,6H)-dione. Molecules 2007, 12, 183–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Schepens, W.; Van Haver, D.; Vandewalle, M.; De Clercq, P.J.; Bouillon, R.; Verstuyf, A. Synthesis and biological activity of 22-oxa CD-ring modified analogues of 1α,25-dihydroxyvitamin D3: Spiro[5.5]undecane CF-ring analogues. Bioorg. Med. Chem. Lett. 2004, 14, 3889–3892. [Google Scholar] [CrossRef] [PubMed]
  36. De Buysser, F.; Verlinden, L.; Verstuyf, A.; De Clercq, P.J. Synthesis of 22-oxaspiro[4.5]decane CD-ring modified analogs of 1α,25-dihydroxyvitamin D3. Tetrahedron Lett. 2009, 50, 4174–4177. [Google Scholar] [CrossRef]
  37. Demin, S.; Van Haver, D.; Vandewalle, M.; De Clercq, P.J.; Bouillon, R.; Verstuyf, A. Synthesis and biological activity of 22-oxa CD-ring modified analogues of 1α,25-dihydroxyvitamin D3: Cis-perhydrindane CE-ring analogues. Bioorg. Med. Chem. Lett. 2004, 14, 3885–3888. [Google Scholar] [CrossRef] [PubMed]
  38. Quan, H.-J.; Koyanagi, J.; Komada, F.; Saito, S. Preparations of vitamin D analogs, spirostanols and furostanols from diosgenin and their cytotoxic activities. Eur. J. Med. Chem. 2005, 40, 662–673. [Google Scholar] [CrossRef]
  39. Verlinden, L.; Bouillon, R.; De Clercq, P.; Verstuyf, A. Analogs of Calcitriol. In Vitamin D, 4th ed.; Feldman, D., Pike, J.W., Bouillon, R., Giovannucci, E., Goltzman, D., Hewison, M., Eds.; Academic Press: London, UK, 2018; Volume 2, pp. 583–614. [Google Scholar]
  40. De Clercq, P.J.; De Buysser, F.; Minne, G.; Schepens, W.; Vrielynck, F.; van Haver, D.; Vandewalle, M.; Verstuyf, A.; Bouillon, R. The development of CD-ring modified analogs of 1α,25-dihydroxyvitamin D. J. Steroid Biochem. Mol. Biol. 2007, 103, 206–212. [Google Scholar] [CrossRef]
  41. Shindo, K.; Kumagai, G.; Takano, M.; Sawada, D.; Saito, N.; Saito, H.; Kakuda, S.; Takagi, K.; Ochiai, E.; Horie, K.; et al. New C15-Substituted Active Vitamin D3. Org. Lett. 2011, 13, 2852–2855. [Google Scholar] [CrossRef]
  42. Takahashi, T.; Ootake, A.; Tsuji, J.; Tachibana, K. Regio- and stereoselective introduction of 15β-hydroxy group and side chains to steroids by the palladium-catalyzed reaction of 1,3-diene monoepoxide. Tetrahedron 1985, 41, 5747–5754. [Google Scholar] [CrossRef]
  43. Marino, J.P.; Abe, H. New stereospecific approach to steroid side chains: Conversion of dehydroepiandrosterone to cholesterol, isocholesterol, and their 15β-hydroxy derivatives. J. Am. Chem. Soc. 1981, 103, 2907–2909. [Google Scholar] [CrossRef]
  44. Moon, S.; Stuhmiller, L.M.; Chadha, R.K.; McMorris, T.C. Synthesis of dehydro-oogoniol and oogoniol: The adrenosterone route. Tetrahedron 1990, 46, 2287–2306. [Google Scholar] [CrossRef]
  45. Konno, K.; Fujishima, T.; Maki, S.; Liu, Z.; Miura, D.; Chokki, M.; Ishizuka, S.; Yamaguchi, K.; Kan, Y.; Kurihara, M.; et al. Synthesis, biological evaluation, and conformational analysis of A-ring diastereomers of 2-methyl-1,25-dihydroxyvitamin D3 and their 20-epimers:  Unique activity profiles depending on the stereochemistry of the A-ring and at C-20. J. Med. Chem. 2000, 43, 4247–4265. [Google Scholar] [CrossRef] [PubMed]
  46. Saito, N.; Honzawa, S.; Kittaka, A. Recent results on A-ring modification of 1α,25-dihydroxyvitamin D3: Design and synthesis of VDR-agonists and antagonists with high biological activity. Curr. Top. Med. Chem. 2006, 6, 1273–1288. [Google Scholar] [CrossRef] [PubMed]
  47. Hourai, S.; Fujishima, T.; Kittaka, A.; Suhara, Y.; Takayama, H.; Rochel, N.; Moras, D. Probing a water channel near the A-ring of receptor-bound 1α,25-dihydroxyvitamin D3 with selected 2α-substituted analogues. J. Med. Chem. 2006, 49, 5199–5205. [Google Scholar] [CrossRef] [PubMed]
  48. Honzawa, S.; Yamamoto, Y.; Hirasaka, K.; Takayama, H.; Kittaka, A. Synthesis of A-ring synthon of 2α-substituted vitamin D3 analogues utilizing Grignard reaction towards methyl 2,3-anhydro-4,6-O-benzylidene-α-D-manno-pyranoside. Heterocycles 2003, 61, 327–338. [Google Scholar] [CrossRef]
  49. Saito, N.; Matsunaga, T.; Saito, H.; Anzai, M.; Takenouchi, K.; Miura, D.; Namekawa, J.; Ishizuka, S.; Kittaka, A. Further synthetic and biological studies on vitamin D hormone antagonists based on C24-alkylation and C2α-functionalization of 25-dehydro-1α-hydroxyvitamin D3-26,23-lactones. J. Med. Chem. 2006, 49, 7063–7075. [Google Scholar] [CrossRef] [PubMed]
  50. Kumagai, G.; Takano, M.; Shindo, K.; Sawada, D.; Saito, N.; Saito, H.; Kakuda, S.; Takagi, K.; Takimoto-Kamimura, M.; Takenouchi, K.; et al. C15-Functionalized 16-ene-1α,25-dihydroxyvitamin D3 is a new vitamin D analog with unique biological properties. Anticancer Res. 2012, 32, 311–318. [Google Scholar]
  51. Baggiolini, E.G.; Iacobelli, J.A.; Hennessy, B.M.; Uskoković, M.R. Stereoselective total synthesis of 1α,25-dihydroxycholecalciferol. J. Am. Chem. Soc. 1982, 104, 2945–2948. [Google Scholar] [CrossRef]
Scheme 1. (A) 7-Dehydrocholesterol isomerism to vitamin D3 via a two-step sequential pericyclic reaction and its activation pathway to 25(OH)D3 (1) and 1α,25(OH)2D3 (2). (B) Deactivation pathways of 1 and 2 to 26,23-lactone and calcitroic acid through (23S)-oxidation and (24R)-oxidation, respectively.
Scheme 1. (A) 7-Dehydrocholesterol isomerism to vitamin D3 via a two-step sequential pericyclic reaction and its activation pathway to 25(OH)D3 (1) and 1α,25(OH)2D3 (2). (B) Deactivation pathways of 1 and 2 to 26,23-lactone and calcitroic acid through (23S)-oxidation and (24R)-oxidation, respectively.
Biomolecules 11 01639 sch001
Scheme 2. Synthesis of the first 16-ene-25-hydroxyvitamin D (3) via an ene reaction from sterol (4).
Scheme 2. Synthesis of the first 16-ene-25-hydroxyvitamin D (3) via an ene reaction from sterol (4).
Biomolecules 11 01639 sch002
Scheme 3. Posner’s approach to 1-hydroxymethyl-25-hydroxy-16-ene-vitamin D3 analogues (912) using the Wittig–Horner reaction.
Scheme 3. Posner’s approach to 1-hydroxymethyl-25-hydroxy-16-ene-vitamin D3 analogues (912) using the Wittig–Horner reaction.
Biomolecules 11 01639 sch003
Scheme 4. Synthesis of 24-oxo-16-ene-vitamin D3 analogues.
Scheme 4. Synthesis of 24-oxo-16-ene-vitamin D3 analogues.
Biomolecules 11 01639 sch004
Figure 1. 16-ene analogues from Uskoković’s group.
Figure 1. 16-ene analogues from Uskoković’s group.
Biomolecules 11 01639 g001
Scheme 5. Synthesis of 24,24-difluoro-16-ene-1-hydroxymethyl-25-hydroxyvitamin D3 (3033).
Scheme 5. Synthesis of 24,24-difluoro-16-ene-1-hydroxymethyl-25-hydroxyvitamin D3 (3033).
Biomolecules 11 01639 sch005
Scheme 6. Synthesis of 23-tert-butylsulfonyl-16-ene-1α-hydroxyvitamin D analogue (42).
Scheme 6. Synthesis of 23-tert-butylsulfonyl-16-ene-1α-hydroxyvitamin D analogue (42).
Biomolecules 11 01639 sch006
Scheme 7. Synthesis of 24-tert-butylsulfonyl-16-ene-1-hydroxyvitamin D analogues (43–46).
Scheme 7. Synthesis of 24-tert-butylsulfonyl-16-ene-1-hydroxyvitamin D analogues (43–46).
Biomolecules 11 01639 sch007
Scheme 8. Synthesis of 24-tert-butylsulfonyl-16,23-diene-1-hydroxyvitamin D analogues (47,48).
Scheme 8. Synthesis of 24-tert-butylsulfonyl-16,23-diene-1-hydroxyvitamin D analogues (47,48).
Biomolecules 11 01639 sch008
Scheme 9. Synthesis of 24-tert-butylsulfonyl-16,23-diene-24-fluoro-1-hydroxyvitamin D analogues (49,50).
Scheme 9. Synthesis of 24-tert-butylsulfonyl-16,23-diene-24-fluoro-1-hydroxyvitamin D analogues (49,50).
Biomolecules 11 01639 sch009
Scheme 10. Synthesis of the 16-ene-CD-ring moieties (104118). *: an asymmetric carbon atom.
Scheme 10. Synthesis of the 16-ene-CD-ring moieties (104118). *: an asymmetric carbon atom.
Biomolecules 11 01639 sch010
Scheme 11. Mouriño’s convergent approach to C20-epi-16-ene-1α,25-dihydroxyvitamin D3 (119) using syn-SN2’ reaction as a key step.
Scheme 11. Mouriño’s convergent approach to C20-epi-16-ene-1α,25-dihydroxyvitamin D3 (119) using syn-SN2’ reaction as a key step.
Biomolecules 11 01639 sch011
Scheme 12. Construction of 17Z-CD-ring (135) using the Vedejs method and synthesis of 16-ene analogues (120122).
Scheme 12. Construction of 17Z-CD-ring (135) using the Vedejs method and synthesis of 16-ene analogues (120122).
Biomolecules 11 01639 sch012
Scheme 13. Synthesis of C20-cyclopropyl-16-ene analogues (136,137).
Scheme 13. Synthesis of C20-cyclopropyl-16-ene analogues (136,137).
Biomolecules 11 01639 sch013
Figure 2. Structures of the 16-ene analogues (147–150) with the cyclopropyl unit at the C20 position.
Figure 2. Structures of the 16-ene analogues (147–150) with the cyclopropyl unit at the C20 position.
Biomolecules 11 01639 g002
Scheme 14. Synthesis of 16-ene-24-oxo analogue (149).
Scheme 14. Synthesis of 16-ene-24-oxo analogue (149).
Biomolecules 11 01639 sch014
Scheme 15. Synthesis of the C16-substituted analogue 152 and its C20-epi-version 153 for the hapten formula. *: an asymmetric carbon atom.
Scheme 15. Synthesis of the C16-substituted analogue 152 and its C20-epi-version 153 for the hapten formula. *: an asymmetric carbon atom.
Biomolecules 11 01639 sch015
Scheme 16. Synthesis of 16-oxa-1α,25-dihydroxyvitamin D3 and its 19-nor counterpart (169,170).
Scheme 16. Synthesis of 16-oxa-1α,25-dihydroxyvitamin D3 and its 19-nor counterpart (169,170).
Biomolecules 11 01639 sch016
Scheme 17. Synthesis of decalin-type 1α,25-dihydroxyvitamin D analogues (180,182) including 19-nor-analogues (181,183).
Scheme 17. Synthesis of decalin-type 1α,25-dihydroxyvitamin D analogues (180,182) including 19-nor-analogues (181,183).
Biomolecules 11 01639 sch017
Scheme 18. Synthesis of pseudo-S2-symmetrical vitamin D analogues with decalin core (188,190).
Scheme 18. Synthesis of pseudo-S2-symmetrical vitamin D analogues with decalin core (188,190).
Biomolecules 11 01639 sch018
Figure 3. Unique examples of the modified D-ring moiety (191–196).
Figure 3. Unique examples of the modified D-ring moiety (191–196).
Biomolecules 11 01639 g003
Scheme 19. Synthesis of 15-substituted CD-rings (204,205).
Scheme 19. Synthesis of 15-substituted CD-rings (204,205).
Biomolecules 11 01639 sch019
Scheme 20. Synthesis of 15-substituted-1α,25-dihydroxyvitamin D3 and its 16-ene analogues (212,213,220,221).
Scheme 20. Synthesis of 15-substituted-1α,25-dihydroxyvitamin D3 and its 16-ene analogues (212,213,220,221).
Biomolecules 11 01639 sch020
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kawagoe, F.; Mototani, S.; Kittaka, A. The Synthesis and Biological Evaluation of D-Ring-Modified Vitamin D Analogues. Biomolecules 2021, 11, 1639. https://doi.org/10.3390/biom11111639

AMA Style

Kawagoe F, Mototani S, Kittaka A. The Synthesis and Biological Evaluation of D-Ring-Modified Vitamin D Analogues. Biomolecules. 2021; 11(11):1639. https://doi.org/10.3390/biom11111639

Chicago/Turabian Style

Kawagoe, Fumihiro, Sayuri Mototani, and Atsushi Kittaka. 2021. "The Synthesis and Biological Evaluation of D-Ring-Modified Vitamin D Analogues" Biomolecules 11, no. 11: 1639. https://doi.org/10.3390/biom11111639

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop