Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleProceedings of the 4th International Symposium on Vitamin D and Analogs in Cancer Prevention and Therapy, May 20-21, 2011, Homburg/Saar, Germany

C15-functionalized 16-Ene-1α,25-dihydroxyvitamin D3 is a New Vitamin D Analog with Unique Biological Properties

GO KUMAGAI, MASASHI TAKANO, KANAKO SHINDO, DAISUKE SAWADA, NOZOMI SAITO, HIROSHI SAITO, SHINJI KAKUDA, KEN-ICHIRO TAKAGI, MIDORI TAKIMOTO-KAMIMURA, KAZUYA TAKENOUCHI, TAI C. CHEN and ATSUSHI KITTAKA
Anticancer Research January 2012, 32 (1) 311-317;
GO KUMAGAI
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MASASHI TAKANO
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KANAKO SHINDO
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DAISUKE SAWADA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
NOZOMI SAITO
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROSHI SAITO
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHINJI KAKUDA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KEN-ICHIRO TAKAGI
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MIDORI TAKIMOTO-KAMIMURA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KAZUYA TAKENOUCHI
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TAI C. CHEN
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ATSUSHI KITTAKA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: akittaka@pharm.teikyo-u.ac.jp
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The Δ16 structure as a vitamin D analog enhanced vitamin D receptor (VDR) binding affinity and induced significant cell differentiation, whereas its relative calcemic activity was reduced compared to 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3). Methodologies available to introduce a double bond at C16-C17 of the D-ring on the seco-steroidal skeleton were limited; therefore, a new synthetic strategy was developed to obtain not only the Δ16 structure, but also a new C15-functional group. Since C15-functionalization was unprecedented in vitamin D analog studies, the hybrid structure of Δ16 and the C15-OH group at the D-ring may provide important information on the structure-activity relationship with vitamin D analogs. The synthesized 16-ene-2α-methyl-1α,15α,25-trihydroxyvitamin D3 showed almost 3-times higher VDR binding affinity and an equipotent level of osteocalcin promoter transactivation activity in human osteosarcoma cells as compared to 1α,25(OH)2D3.

  • 16-ene-vitamin D3
  • SN2′ reaction
  • 15-substituted vitamin D3

The 16-ene structure of the CD-ring part of vitamin D3, first appeared in the total synthesis of 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3, 1 in Figure 1) reported by the Hoffmann-La Roche group in 1982 (1). To elongate the side chain at C20 (steroid numbering) from the (17Z)-ethylidene CD-ring of compound 5 creating the 20R-natural stereochemistry, they utilized the ene reaction with ethyl propionate in the presence of Lewis acid EtAlCl2 (Figure 2). After the successful ene reaction (88% yield and the desired 20R configuration) (1-3), the 16-ene structure 6 was obtained, and the double bond was then reduced stereo-selectively to the saturated CD-ring system 7 that was included in the natural active vitamin D3 skeleton. A great variety of unique 16-ene vitamin D analogs have been synthesized using the ene reaction, and their biological activities were evaluated (4-11). It was shown that 16-ene-1α,25-dihydroxyvitamin D3 had higher binding affinity for the vitamin D receptor (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 (11-16).

Another strategy for 16-ene construction was developed by Mouriño's group in 1999. They utilized SN2′-syn-facial displacement toward (17Z)-olefin carbamate (8) by high-order cuprate(s) (Li2Cu3Me5 etc.) to generate the 20R-natural configuration and the 16-ene double bond as in 9 with high yield(s) (Figure 3) (17). 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, stereoselectively, through this route.

C15 substituted vitamin D analogs have not been synthesized yet, and a new synthetic route for 16-ene analogs also via an SN2′ reaction was developed. In order to create a C15-substituent, a new substrate for the SN2′ reaction was used, since a C15-C16 epoxide ring system on the D-ring was desirable, in which the leaving group was still at the C16 position, and importantly, a required functional group (a hydroxy group) could remain at the C15 position after the SN2′ reaction.

Materials and Methods

Chemistry. A new epoxide 11 was synthesized from known ketone 10 in four steps (Figure 4). The stereochemistry of 15α,16α-epoxide 11 was determined by X-ray crystallographic analysis of its acetate 12 (Figure 5). Oxidation of the C17-OH group of 11 to the C17-oxo group followed by Wittig reaction with ethylidenetriphenylphosphorane gave the key intermediate of (17Z)-ethylidene epoxide 14 with a good yield (18).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Structures of active vitamin D3 (1α,25(OH)2D3), 1; 16-ene-1α,25(OH)2D3, 2; 16-ene-1α,15α,25(OH)3D3, 3 and 2α-methyl-16-ene-1α,15α,25(OH)3D3, 4.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Part of the first total and chiral synthesis of active vitamin D3 (1α,25(OH)2D3), in which 16-ene structure 6 was available by the ene reaction (1).

The next step was the SN2′ reaction of magnesium cyanocuprate prepared from 5-bromo-2-methyl-2-pentanol methoxymethyl ether (19) toward compound 14. This key SN2′ reaction proceeded smoothly in anti-displacement to afford the 16-ene product with the 15α-hydroxy group and the 20R-natural configuration side chain as the major isomer 15a (20R:20S=11:1).

The CD-ring 15a was converted to the 8-oxo compound 16 and subsequent Wittig reaction with bromomethylenetriphenylphosphorane gave bromoolefin 17. Trost coupling reaction (20) of the C15-substituted 16-ene-bromoolefin 17 and enynes 18 and 19 (21-23) gave the protected target molecules 20 and 21, respectively. The deprotection step took place under mild acidic conditions using (+)-camphorsulfonic acid in MeOH to yield 3 and 4 (Figure 6).

Human VDR binding assay. Binding affinity to the VDR was evaluated using a 1α,25(OH)2D3 assay kit (Polarscreen Vitamin D Receptor Competitor Assay, Red, Cat. No. PV4569) purchased from Invitrogen (Grand Island, NY, USA). The test compound solution (1 mM in EtOH) was initially diluted 10-fold with DMSO and then further 50-fold with the assay buffer provided in the kit. The assay was performed according to the procedures described in the kit insert. All the compounds were evaluated with N=2 within the range from 10−6 M to 10−10 M. IC50 values were calculated using the average of the measured values. The activities of each compound are presented as the relative value in which the activity of the natural hormone 1 was normalized to 100%.

Transactivation assay of human osteocalcin promoter. VDR transactivation of the osteocalcin promoter in HOS (human osteosalcoma) cells (American Type Culture Collection (ATCC) Manassas, VA, USA) by the test compounds was compared to the natural hormone. The human osteocalcin gene promoter fragment −838/+10 was cloned into the reporter plasmid pGL3 (Promega; Madison, WI, USA) as reported previously (24). Human VDR and RXR (Retinoid X receptor) genes were cloned into expression vector pcDNA3 (Invitrogen). The HOS cells were maintained in phenol red free DMEM (Invitrogen) containing 10% fetal calf serum (FCS) (Invitrogen). Prior to the transfections, the cells were plated in a 96 well plate at a density of 400,000 cells per well in Opti-MEM (Invitrogen). The cells were transfected with human osteocalcin reporter vector (pGL3-hOc: 100 ng/well), human VDR and RXR expression vector (pcDNA-hVDR, pcDNA-hRXR: 10 ng/well) and internal control plasmid phRL-TK (Promega: 25 ng/well) using 0.45 μL of Lipofectamine 2000 reagent (Invitrogen). After incubation at 37°C for 3 h, the culture media were replaced with phenol red free DMEM containing 10% FCS. The cells were treated with ethanol vehicle or various concentrations of the compounds (from 0.1 pM to 100 nM). After incubation at 37°C for 24 h, the luciferase activity of the cells was quantified by luminometer (Berthold Japan; Tokyo, Japan) using a Dual-Glo luciferase assay system (Promega).

Results and Discussion

Chemistry. In the first total synthesis of active vitamin D3 (1), trans-(8-hydroxy)hydrindan-17-one (22) was alkenylated through the Wittig reaction with ethylidenetriphenylphosphorane to afford (17Z)-ethylidene 23 as the major product. The steroid skeletons also gave (17Z)-ethylidene 25 after the Wittig reaction (Figure 7) (2,3,25). Furthermore, 15β,16β-epoxy-17-oxosteroid (26) was converted to (17E)-olefin 27, in which the direction of the ethylidene group was the same as in compounds 23 and 25 (Figure 7) (26-28). (17E)-Ethylidene-15β,16β-epoxysterols (27) were previously studied on the synthesis of 15β-hydroxysteroids such as oogoniol (28) (steroidal sex hormone of the water mold Achlya) (29). On the other hand, pavoninin-5 (29) (shark repellent of the sole Pardachirus pavoninus) has the 15α-hydroxysteroid structure (30, 31).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Part of 16-ene-1α,25(OH)2D3 synthesis through SN2′-syn displacement at C20 with the allylic carbamate system (17). MOM: methoxymethyl; TBS: tert-butyldimethylsilyl.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

(17Z)-Ethylidene-15α,16α-epoxyhydrindan (14) synthesis from ketone 10 (18).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

ORTEP (Oak Ridge Thermal Ellipsoid Plot Program) stereo drawing of acetate 12.

Interestingly, however, in the present process the 15α,16α-epoxyhydrindan-17-one (13) was converted to (17Z)-olefin (14) by the same Wittig reaction (the nomenclature of Z stereochemistry is the same in both compounds 23 and 25 coincidentally, but the direction of the ethylidene section is different, Figure 4). In the present case, the epoxide ring at the α-side would select orientation of the phosphorane reagent by steric repulsion as shown in Figure 8, i.e., first, the phosphorane reagent would attack the 17-keto carbonyl group from the less hindered α-side due to the C18-methyl group, and then, repulsion would occur between the methyl group and the α-epoxide ring, B, between the PPh3 group (which was the biggest group among H, CH3, and Ph3P) and the α-epoxide ring, C, and between both methyl and PPh3 groups and the α-epoxide ring, D; therefore, A would be the most favored conformation to give (17Z)-ethylidene stereochemistry.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Introduction of the side chain to C20 with R-configuration and conversion to bromoolefin 17, and subsequent Trost coupling reaction with A-ring precursor enynes (18 and 19). The coupling product was deprotected under mild acidic conditions. CSA: (+)-camphorsulfonic acid.

The side chain elongation reaction at the C20 position of (17Z)-ethylidene epoxide 14 in the anti-stereochemistry of SN2′ manner, proceeded smoothly with the magnesium cyanocuprate reagent (Figure 6) (18). Fortunately, the natural configuration of 20R was available as the major isomer, which could be explained by Corey's theory of a stereoelectronic effect arising from bidentate binding involving a d-orbital of nucleophilic copper and σ* and π* orbitals of the ethylidene epoxide 14 (Figure 9) (32). The 20R stereochemistry of 15a was determined without ambiguity by chemical conversion of 15a to the known 25-OMOM Grundmann's ketone, which has the 20R configuration (18).

Human VDR binding affinity. The hVDR binding affinity of the new 16-ene-vitamin D3 analogs 3 and 4 having the C15-OH group was determined using the Invitrogen receptor assay kit. Compounds 3 and 4 showed 65% and 278% binding affinity relative to that of 1α,25(OH)2D3. It was reported that the original 16-ene-1α,25(OH)2D3 (2) exhibited 161% binding affinity for calf-thymus VDR (33) and 240% for rat intestine VDR (12). The introduction of a double bond to the ring D at the C16-17 position of 1α,25(OH)2D3 enhanced the binding affinity for the VDR, but further modification with the C15α-OH group decreased the VDR binding affinity of compound 3, however, additional modification at the C2α position with the methyl group recovered the affinity of compound 4.

Osteocalcin promoter transactivation activity. The EC50 values of transactivation activity of the osteocalcin promoter in the HOS cells were 0.08 nM for compound 3 and 0.12 nM for compound 4, and 0.09 nM for 1α,25(OH)2D3. Thus the 16-ene analogs with the C15α-OH group 3 and 4 showed comparable or even greater transactivation activity than 1α,25(OH)2D3.

Our previous X-ray co-crystallographic study on 15α-methoxy-1α,25-dihydroxyvitamin D3 revealed that the 15α-methoxy group made a new contact with Ser275Oγ at a distance of 3.1 Å (18); therefore, the 15α-OH group of the analogs 3 and 4 could form a hydrogen bond with Ser275Oγ to stabilize the VDR-ligand complex. Importantly, the C15-OH group could be converted to a wide variety of other functional groups, and would be useful for further structure-activity relationship studies on the D-ring moiety of the active vitamin D to develop potential therapeutic agents for cancer (34-36).

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

Ethylidene introduction to the 17-position by Wittig reaction (1-3,25-28) and structures of oogoniol and pavoninin-5 as 15-hydroxysteroid natural products.

Figure 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 8.

(17Z)-Ethylidene formation via the most favored conformation A at the Wittig reaction step.

Figure 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 9.

Plausible reaction mechanism of anti-stereochemistry based on SN2′ reaction entering the full length side chain with 20R configuration.

Conclusion

The novel 16-ene-1α,15α,25-trihydroxyvitamin D3 analog 3 and its 2α-methylated analog 4 are synthesized efficiently utilizing the SN2′ reaction of organocuprate reagent toward (17Z)-ethylidene-15α,16α-epoxyhydrindan 14 with the anti-stereoselective displacement followed by Trost coupling with the A-ring precursor. These synthesized analogs show great VDR binding affinity and osteocalcin transactivation activity in HOS cells.

Acknowledgements

We thank Ms. Junko Shimode and Ms. Miki Takahashi (Teikyo University, Japan) for the spectroscopic measurements. This study was supported in part by Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (No. 21590022 to AK and No. 23590015 to DS) and Grant-in-Aid for Young Scientists from the Ministry of Education, Culture, Sports, Science and Technology, Japan, (No. 23790021 to MT).

Footnotes

  • ↵† Current Address: Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.

  • Received September 9, 2011.
  • Revision received November 17, 2011.
  • Accepted November 17, 2011.
  • Copyright© 2012 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved

References

  1. ↵
    1. Baggiolini EG,
    2. Iacobelli JA,
    3. Hennessy BM,
    4. Uskoković MR
    : Stereoselective total synthesis of 1α,25-dihydroxycholecalciferol. J Am Chem Soc 104: 2945-2948, 1982.
    OpenUrl
  2. ↵
    1. Dauben WG,
    2. Brookhart T
    : Stereocontrolled synthesis of steroidal side chains. J Am Chem Soc 103: 237-238, 1981.
    OpenUrl
  3. ↵
    1. Batcho AD,
    2. Berger DE,
    3. Uskoković MR,
    4. Snider BB
    : C-20 Stereospecific introduction of a steroid side chain. J Am Chem Soc 103: 1293-1295, 1981.
    OpenUrlCrossRef
  4. ↵
    1. Laverny G,
    2. Penna G,
    3. Uskokovic M,
    4. Marczak S,
    5. Maehr H,
    6. Jankowski P,
    7. Ceailles C,
    8. Vouros P,
    9. Smith B,
    10. Robinson M,
    11. Reddy GS,
    12. Adorini L
    : 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-cyclopropylvitamin D3. J Med Chem 52: 2204-2213, 2009.
    OpenUrlCrossRefPubMed
    1. Reichrath J,
    2. Friedrich M,
    3. Tilgen W
    1. Swami S,
    2. Zhao XY,
    3. Sarabia S,
    4. Siu-Caldera ML,
    5. Uskokovic M,
    6. Reddy SG,
    7. Feldman D
    : A low-calcemic vitamin D analog (Ro 25-4020) inhibits the growth of LNCaP human prostate cancer cells with increased potency by producing an active 24-oxo metabolite (Ro 29-9970). In: Vitamin D analogs in cancer prevention and therapy. Recent results in cancer research 164. Reichrath J, Friedrich M, Tilgen W (eds.). Springer-Verlag, Berlin, pp. 349-352, 2003.
    1. Norman AW,
    2. Bouillon R,
    3. Thomasset M
    1. Uskoković MR,
    2. Baggiolini E,
    3. Shiuey SJ,
    4. Iacobelli J,
    5. Hennessy B,
    6. Kiegiel J,
    7. Daniewski AR,
    8. Pizzolato G,
    9. Courtney LF,
    10. Horst RL
    : The 16-ene analogs of 1,25-dihydroxycholecalciferol. Synthesis and biological activity. In: Vitamin D: Gene Regulation, Structure-Function Analysis and Clinical Application. Norman AW, Bouillon R, Thomasset M (eds.). Walter de Gruyter, Berlin, pp. 139-145, 1991.
    1. Norman AW,
    2. Bouillon R,
    3. Thomasset M
    1. von Daehne W,
    2. Hansen CM,
    3. Hansen D,
    4. Mathiasen IS
    : Synthesis and biological activity of new side chain analogues of 16-dehydrocalcitriol and its 20-epimer. In: Vitamin D: Chemistry, Biology and Clinical Applications of the Steroid Hormone. Norman AW, Bouillon R, Thomasset M (eds.). Vitamin D Workshop, Inc, Riverside, CA, pp. 81-82, 1997.
    1. Posner GH,
    2. Wang Q,
    3. Han G,
    4. Lee JK,
    5. Crawford K,
    6. Zand S,
    7. Brem H,
    8. Peleg S,
    9. Dolan P,
    10. Kensler TW
    : Conceptually new sulfone analogues of the hormone 1α,25-dihydroxyvitamin D3: synthesis and preliminary biological evaluation. J Med Chem 42: 3425-3435, 1999.
    OpenUrlCrossRefPubMed
    1. Posner GH,
    2. Lee JK,
    3. Wang Q,
    4. Peleg S,
    5. Burke M,
    6. Brem H,
    7. Dolan P,
    8. Kensler TW
    : Noncalcemic, antiproliferative, transcriptionally active, 24-fluorinated hybrid analogues of the hormone 1α,25-dihydroxyvitamin D3. Synthesis and preliminary biological evaluation. J Med Chem 41: 3008-3014, 1998.
    OpenUrlCrossRefPubMed
    1. Posner GH,
    2. Lee JK,
    3. White MC,
    4. Hutchings RH,
    5. Dai H,
    6. Kachinski JL,
    7. Dolan P,
    8. Kensler TW
    : Antiproliferative hybrid analogs of the hormone 1α,25-dihydroxyvitamin D3: design, synthesis, and preliminary biological evaluation. J Org Chem 62: 3299-3314, 1997.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Feldman D,
    2. Glorieux FH,
    3. Pike JW
    1. Uskoković MR,
    2. Studzinski GP,
    3. Reddy GS
    : The 16-ene vitamin D analogs. In: Vitamin D. Feldman D, Glorieux FH, Pike JW (eds.). Academic Press, New York, Chapter 62, pp. 1045-1070, 1997.
  6. ↵
    1. Chen TC,
    2. Persons K,
    3. Uskokovic MR,
    4. Horst RL,
    5. Holick MF
    : 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 4: 49-57, 1993.
    OpenUrlCrossRef
    1. Bishop JE,
    2. Collins ED,
    3. Okamura WH,
    4. Norman AW
    : Profile of ligand specificity of the vitamin D binding protein for 1α,25-dihydroxyvitamin D3 and its analogs. J Bone Miner Res 9: 1277-1288, 1994.
    OpenUrlPubMed
    1. Siu-Caldera ML,
    2. Clark JW,
    3. Santos-Moore A,
    4. Peleg S,
    5. Liu YY,
    6. Uskoković MR,
    7. Sharma S,
    8. Reddy GS
    : 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 59: 405-412, 1996.
    OpenUrlCrossRefPubMed
    1. Jung SJ,
    2. Lee YY,
    3. Pakkala S,
    4. de Vos S,
    5. Elstner E,
    6. Norman AW,
    7. Green J,
    8. Uskokovic M,
    9. Koeffler HP
    : 1,25(OH)2-16Ene-vitamin D3 is a potent antileukemic agent with low potential to cause hypercalcemia. Leuk Res 18: 453-463, 1994.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Rhieu SY,
    2. Annalora AJ,
    3. Gathungu RM,
    4. Vouros P,
    5. Uskokovic MR,
    6. Schuster I,
    7. Palmore GTR,
    8. Reddy GS
    : A new insight into the role of rat cytochrome P450 24A1 in metabolism of selective analogs of 1α,25-dihydroxyvitamin D3. Arch Biochem Biophys 509: 33-43, 2011.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Rey MA,
    2. Martínez-Pérez JA,
    3. Fernández-Gacio A,
    4. Halkes K,
    5. Fall Y,
    6. Granja J,
    7. 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 64: 3196-3206, 1999 and references therein.
    OpenUrlPubMed
  9. ↵
    1. Shindo K,
    2. Kumagai G,
    3. Takano M,
    4. Sawada D,
    5. Saito N,
    6. Saito H,
    7. Kakuda S,
    8. Takagi K,
    9. Ochiai E,
    10. Horie K,
    11. Takimoto-Kamimura M,
    12. Ishizuka S,
    13. Takenouchi K,
    14. Kittaka A
    : New C15-substituted active vitamin D3. Org Lett 13: 2852-2855, 2011.
    OpenUrlPubMed
  10. ↵
    1. Larsen SD,
    2. Spilman CH,
    3. Yagi Y,
    4. Dinh DM,
    5. Hart KL,
    6. Hess GF
    : Design and synthesis of seco-oxysterol analogs as potential inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase gene transcription. J Med Chem 37: 2343-2351, 1994.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Trost BM,
    2. Dumas J,
    3. Villa M
    : New strategies for the synthesis of vitamin D metabolites via Pd-catalyzed reactions. J Am Chem Soc 114: 9836-9845, 1992.
    OpenUrl
  12. ↵
    1. Konno K,
    2. Fujishima T,
    3. Maki S,
    4. Liu Z,
    5. Miura D,
    6. Chokki M,
    7. Ishizuka S,
    8. Yamaguchi K,
    9. Kan Y,
    10. Kurihara M,
    11. Miyata N,
    12. Smith C,
    13. DeLuca HF,
    14. Takayama H
    : 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 43: 4247-4265, 2000.
    OpenUrlCrossRefPubMed
    1. Suhara Y,
    2. Kittaka A,
    3. Kishimoto S,
    4. Calverley MJ,
    5. Fujishima T,
    6. Saito N,
    7. Sugiura T,
    8. Waku K,
    9. Takayama H
    For the direct preparation of enyne 19, see: Suhara Y, Kittaka A, Kishimoto S, Calverley MJ, Fujishima T, Saito N, Sugiura T, Waku K, Takayama H: Synthesis and testing of 2α-modified 1α,25-dihydroxyvitamin D3 analogues with a double side chain: marked cell differentiation activity. Bioorg Med Chem Lett 12: 3255-3258, 2002.
    OpenUrlPubMed
  13. ↵
    1. Honzawa S,
    2. Yamamoto Y,
    3. Hirasaka K,
    4. Takayama H,
    5. 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-mannopyranoside. Heterocycles 61: 327-228, 2003.
    OpenUrl
  14. ↵
    1. Ozono K,
    2. Liao J,
    3. Kerner SA,
    4. Scott RA,
    5. Pike JW
    : The vitamin D-responsive element in the human osteocalcin promoter. J Biol Chem 265: 21881-21888, 1990.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Drefahl G,
    2. Ponsolt K,
    3. Schick H
    : Synthesen von Alkyliden-Steroiden durch Wittig-Reaktion. Chem Ber 98: 604-612, 1965.
    OpenUrl
  16. ↵
    1. Marino JP,
    2. Abe H
    : A new stereospecific approach to steroid side chains: conversion of dehydroepiandrosterone to cholesterol, isocholesterol, and their 15β-hydroxy derivatives. J Am Chem Soc 103: 2907-2909, 1981.
    OpenUrl
    1. Takahashi T,
    2. Ootake A,
    3. Tsuji J,
    4. Tachibana K
    : Regio- and stereoselective 15β-hydroxy group and side chains to steroids by the palladium-catalysed reaction of 1,3-diene monoepoxides with carbonucleophiles. Tetrahedron 41: 5747-5754, 1985.
    OpenUrl
  17. ↵
    1. Moon S,
    2. Stuhmiller LM,
    3. Chadha RK,
    4. McMorris TC
    : Synthesis of dehydro-oogoniol and oogoniol: the adrenosterone route. Tetrahedron 46: 2287-2306, 1990.
    OpenUrl
  18. ↵
    1. Wiersig JR,
    2. Waespe-Sarcevic N,
    3. Djerassi C
    : Stereospecific synthesis of the side chain of the steroidal plant sex hormone oogoniol. J Org Chem 44: 3374-3382, 1979.
    OpenUrl
  19. ↵
    1. Tachibana K,
    2. Sakaitanai M,
    3. Nakanishi K
    : Pavoninins: shark-repelling ichthyotoxins from the defense secretion of the pacific sole. Science 226: 703-705, 1984.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    1. Gong H,
    2. Williams JR
    : Synthesis of the aglycone of the shark repellent pavoninin-4 using remote functionalization. Org Lett 8: 2253-2255, 2006.
    OpenUrlPubMed
  21. ↵
    1. Corey EJ,
    2. Boaz NW
    : d-Orbital stereoelectronic control of the stereochemistry of SN2′ displacements by organocuprate reagents. Tetrahedron Lett 25: 3063-3066, 1984.
    OpenUrl
  22. ↵
    1. Nakagawa K,
    2. Sowa Y,
    3. Kurobe M,
    4. Ozono K,
    5. Siu-Caldera ML,
    6. Reddy GS,
    7. Uskokovic MR,
    8. Okano T
    : Differential activities of 1α,25-dihydroxy-16-ene-vitamin D3 analogs and their 3-epimers on human promyelocytic leukemia (HL-60) cell differentiation and apoptosis. Steroids 66: 327-337, 2001.
    OpenUrlPubMed
  23. ↵
    1. Sawada D,
    2. Tsukuda Y,
    3. Saito H,
    4. Kakuda S,
    5. Takimoto-Kamimura M,
    6. Ochiai E,
    7. Takenouchi K,
    8. Kittaka A
    : Development of 14-epi-19-nortachysterol and its unprecedented binding configuration for the human vitamin D receptor. J Am Chem Soc 133: 7215-7221, 2011.
    OpenUrlPubMed
    1. Saitoh H,
    2. Chida T,
    3. Takagi K,
    4. Horie K,
    5. Sawai Y,
    6. Nakamura Y,
    7. Harada Y,
    8. Takenouchi K,
    9. Kittaka A
    : Synthesis of C-2 substituted vitamin D derivatives having ringed side chains and biological evaluation, especially biological effect on bone by modification at C-2 position. Org Biomol Chem 9: 3954-3964, 2011.
    OpenUrlPubMed
  24. ↵
    1. Chen TC,
    2. Kittaka A
    : Novel Vitamin D Analogs for Prostate Cancer Therapy. International Scholarly Research Network (ISRN) Urology 2011: Article ID 301490, 9 pages, 2011.
PreviousNext
Back to top

In this issue

Anticancer Research: 32 (1)
Anticancer Research
Vol. 32, Issue 1
January 2012
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
C15-functionalized 16-Ene-1α,25-dihydroxyvitamin D3 is a New Vitamin D Analog with Unique Biological Properties
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
10 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
C15-functionalized 16-Ene-1α,25-dihydroxyvitamin D3 is a New Vitamin D Analog with Unique Biological Properties
GO KUMAGAI, MASASHI TAKANO, KANAKO SHINDO, DAISUKE SAWADA, NOZOMI SAITO, HIROSHI SAITO, SHINJI KAKUDA, KEN-ICHIRO TAKAGI, MIDORI TAKIMOTO-KAMIMURA, KAZUYA TAKENOUCHI, TAI C. CHEN, ATSUSHI KITTAKA
Anticancer Research Jan 2012, 32 (1) 311-317;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
C15-functionalized 16-Ene-1α,25-dihydroxyvitamin D3 is a New Vitamin D Analog with Unique Biological Properties
GO KUMAGAI, MASASHI TAKANO, KANAKO SHINDO, DAISUKE SAWADA, NOZOMI SAITO, HIROSHI SAITO, SHINJI KAKUDA, KEN-ICHIRO TAKAGI, MIDORI TAKIMOTO-KAMIMURA, KAZUYA TAKENOUCHI, TAI C. CHEN, ATSUSHI KITTAKA
Anticancer Research Jan 2012, 32 (1) 311-317;
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results and Discussion
    • Conclusion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • The Difference between 14-Epi-previtamin D3 and 14-Epi-19-norprevitamin D3: Their Synthesis and Binding Affinity for Human VDR
  • The Scots' Paradox: Can Sun Exposure, or Lack of it, Explain Major Paradoxes in Epidemiology?
  • Optimal Serum Calcidiol Concentration for Cancer Prevention
Show more Proceedings of the 4th International Symposium on Vitamin D and Analogs in Cancer Prevention and Therapy, May 20-21, 2011, Homburg/Saar, Germany

Similar Articles

Anticancer Research

© 2023 Anticancer Research

Powered by HighWire