Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SIDE-CHAIN MODIFIED ERGOSTEROL AND STIGMASTEROL DERIVATIVES AS LIVER X RECEPTOR MODULATORS
Document Type and Number:
WIPO Patent Application WO/2019/021122
Kind Code:
A2
Abstract:
The present invention relates to novel Liver X Receptor (LXR) modulators, their use in diagnostic and therapy, more particularly in the treatment of diseases associated with LXR, such as cancer, inflammation, metabolic and autoimmune diseases.

Inventors:
RUSSO VINCENZO (IT)
MARINOZZI MAURA (IT)
CASTRO NAVAS FRANCISCO FERMIN (ES)
Application Number:
PCT/IB2018/055372
Publication Date:
January 31, 2019
Filing Date:
July 19, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
OSPEDALE SAN RAFFAELE SRL (IT)
International Classes:
A61K31/575; C07J9/00; A61K31/58; A61P35/00; C07J17/00; C07J53/00; C07J71/00
Other References:
POLI, G.; BIASI, F.; LEONARDUZZI, G.: "Redox biology oxysterols in the pathogenesis of major chronic diseases", REDOX BIOL., vol. 1, 2013, pages 125 - 130
BJORKHEM, I.; DICZFALUSY, U.: "Oxysterols: friends, foes, or just fellow passengers?", ARTERIOSCLER. THROMB. VASE. BIOL., 2002, pages 734 - 743
BOVENGA, F.; SABBA, C.; MOSCHETTA, A.: "Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer", CELL METAB., vol. 21, 2015, pages 517 - 526
TRAVERSARI, C.; SOZZANI, S.; STEFFENSEN, K. R.; RUSSO, V.: "LXR-dependent and - independent effects of oxysterols on immunity and tumor growth", EUR. J. IMMUNOL., vol. 44, 2014, pages 1896 - 1903
VENTECLEF, N.; FERRE, P.: "Liver X receptor: from metabolism to cancer", BIOCHEM. J., vol. 459, 2014, pages e1 - e3
DE BOUSSAC, H.; ALIOUI, A.; VIENNOIS, E.; DUFOUR, J.; TROUSSON, A.; VEGA, A.; GUY, L.; VOILE, D. H.; LOBACCARO, J.-M. A.; BARON, S: "Oxysterol receptors and their therapeutic applications in cancer conditions", EXPERT OPIN. THER. TARGETS, vol. 17, 2013, pages 1029 - 1038
JANOWSKY, BETHANY A.; WILLY, P. J.; RAMA DEVI, T.; FALCK, J. R.; MANGELSDORF, D. J.: "An oxysterol signalling pathway mediated by the nuclear receptor LXR", NATURE, vol. 383, 1996, pages 728 - 731, XP002104644, DOI: doi:10.1038/383728a0
JANOWSKI, B. A.; GROGAN, M. J.; JONES, S. A.; WISELY, G. B.; KLIEWER, S. A.; COREY, E. J.; MANGELSDORF, D. J.: "Structural requirements of ligands for the oxysterol liver X receptors LXR α and LXR β", PROC. NATL. ACAD. SCI. U. S. A., vol. 96, 1999, pages 266 - 271, XP002104645, DOI: doi:10.1073/pnas.96.1.266
SONG, C.; KOKONTIS, J. M.; HIIPAKKA, R. A.; LIAO, S.: "Ubiquitous receptor: a receptor that modulates gene activation by retinoic acid and thyroid hormone receptors", PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 10809 - 10813, XP002211667, DOI: doi:10.1073/pnas.91.23.10809
SHINAR, D. M.; ENDO, N.; RUTLEDGE, S. J.; VOGEL, R.; RODAN, G. A.; SCHMIDT, A.: "NER, a new member of the gene family encoding the human steroid hormone nuclear receptor", GENE, vol. 147, 1994, pages 273 - 276, XP023540956, DOI: doi:10.1016/0378-1119(94)90080-9
APFEL, R.; BENBROOK, D.; LERNHARDT, E.; ORTIZ, M. A.; SALBERT, G.; PFAHL, A. M.: "A novel orphan receptor specific for a subset of thyroid hormone-responsive elements and its interaction with the retinoid / thyroid hormone receptor subfamily", MOL. CELL. BIOL., vol. 14, 1994, pages 7025 - 7035
TEBOUL, M.; ENMARK, E.; LI, Q.; WIRKSTROEM, A. C.; PELTO-HUIKKO, M.; GUSTAFSSON, J.-A.: "OR-1, a member of the nuclear receptor superfamily that interacts with the 9-cis-retinoic acid receptor", PROC. NATL. ACAD. SCI. U. S. A., vol. 92, 1995, pages 2096 - 2100, XP002211668, DOI: doi:10.1073/pnas.92.6.2096
WILLY, P. J.; UMESONO, K.; ONG, E. S.; EVANS, R. M.; HEYMAN, R. A; MANGELSDORF, D. J.: "LXR, a nuclear receptor that defines a distinct retinoid response pathway", GENES DEV., vol. 9, 1995, pages 1033 - 1045, XP000972205
SPANN, N. J.; GLASS, C. K.: "Sterols and oxysterols in immune cell function", NAT. IMMUNOL., vol. 14, 2013, pages 893 - 900
GABBI, C.; WARNER, M.; GUSTAFSSON, J.-A.: "Action mechanisms of liver X receptors", BIOCHEM. BIOPHYS. RES. COMMUN., vol. 446, 2014, pages 647 - 650
DONG, X.-Y.; TANG, S.-Q.; CHEN, J.-D.: "Dual functions of Insig proteins in cholesterol homeostasis", LIPIDS HEALTH DIS., vol. 11, 2012, pages 173, XP021139564, DOI: doi:10.1186/1476-511X-11-173
OLKKONEN, V. M.; ZHOU, Y.; YAN, D.; VIHERVAARA, T.: "Oxysterol-binding proteins-emerging roles in cell regulation", EUR. J. LIPID SCI. TECHNOL., vol. 114, 2012, pages 634 - 643
CORMAN, A.; DEBERARDINIS, A. M.; HADDEN, M. K.: "Structure - activity relationships for side chain oxysterol agonists of the hedgehog signaling pathway", ACS MED. CHEM. LETT., vol. 3, 2012, pages 828 - 833, XP055486086, DOI: doi:10.1021/ml300192k
TICE, C. M.; NOTO, P. B.; FAN, K. Y.; ZHUANG, L.; LALA, D. S.; SINGH, S. B.: "The medicinal chemistry of liver X receptor (LXR) modulators", J. MED. CHEM., vol. 57, 2014, pages 7182 - 7205, XP055494920, DOI: doi:10.1021/jm500442z
LOREN, J.; HUANG, Z.; LAFFITTE, B. A; MOLTENI, V.: "Liver X Receptor Modulators: A review of recently patented compounds (2009 - 2012", EXPERT OPIN. THER. PAT., vol. 23, 2013, pages 1317 - 1335, XP055294658, DOI: doi:10.1517/13543776.2013.814640
SPENCER, T. A.; LI, D.; RUSSEL, J. S.; COLLINS, J. L.; BLEDSOE, R. K.; CONSLER, T. G.; MOORE, L. B.; GALARDI, C. M.; MCKEE, D. D.;: "Pharmacophore analysis of the nuclear oxysterol receptor LXRa", J. MED. CHEM., vol. 44, 2001, pages 886 - 897, XP002338735, DOI: doi:10.1021/jm0004749
QUINET, E. M.; SAVIO, D. A; HALPERN, A. R.; CHEN, L.; MILLER, C. P.; NAMBI, P.: "Gene-selective modulation by a synthetic oxysterol ligand of the liver X receptor", J. LIPID RES., vol. 45, 2004, pages 1929 - 1942
BERRODIN, T. J.; SHEN, Q.; QUINET, E. M.; YUDT, M. R.; FREEDMAN, L. P.: "Identification of 5a, 6a-epoxycholesterol as a novel modulator of liver X receptor activity", MOL. PHARMACOL., vol. 78, 2010, pages 1046 - 1058
PENG, D.; HIIPAKKA, R.; DAI, Q.; GUO, J.; REARDON, C.; GETZ, G. S.; LIAO, S.: "Antiatherosclerotic effects of a novel synthetic tissue-selective steroidal liver X receptor agonist in low-density lipoprotein receptor-deficient mice", J. PHARMACOL. EXP. THER., vol. 327, 2008, pages 332 - 342
PENG, D.; HIIPAKKA, R.; XIE, J. T.; DAI, Q.; KOKONTIS, J. M.; REARDON, C.; GETZ, G. S.; LIAO, S.: "A novel potent synthetic steroidal liver X receptor agonist lowers plasma cholesterol and triglycerides and reduces atherosclerosis in LDLR -/- mice", BR. J. PHARMACOL., vol. 162, 2011, pages 1792 - 1804, XP055044585, DOI: doi:10.1111/j.1476-5381.2011.01202.x
LI, L.; LIU, J.; ZHU, L.; CUTLER, S.; HASEGAWA, H.; SHAN, B.; MEDINA, J. C.: "Discovery and optimization of a novel series of liver X receptor-a Agonists", BIOORG. MED. CHEM. LETT., vol. 16, 2006, pages 1638 - 1642, XP025107064, DOI: doi:10.1016/j.bmcl.2005.12.015
JONES, P. J. H.: "Cholesterol-lowering effect of plant sterols", CURR. ATHEROSCLER. REP., vol. 1, 1999, pages 230 - 235
PLAT, J.; MENSINK, R. P.: "Increased intestinal ABCA1 expression contributes to the decrease in cholesterol absorption after plant stanol consumption", FASEB J., vol. 16, 2002, pages 1248 - 1253
KANEKO, E.; MATSUDA, M.; YAMADA, Y.; TACHIBANA, Y.; SHIMOMURA, I.; MAKISHIMA, M.: "Induction of intestinal ATP-binding cassette transporters by a phytosterol-derived liver X receptor agonist", J. BIOL. CHEM., vol. 278, 2003, pages 36091 - 36098, XP001199518, DOI: doi:10.1074/jbc.M304153200
CHISHOLM, J. W.; HONG, J.; MILLS, S. A.; LAWN, R. M.: "The LXR Ligand T0901317 induces severe lipogenesis in the db/db diabetic mouse", J. LIPID RES., vol. 44, 2003, pages 2039 - 2048, XP002469324, DOI: doi:10.1194/jlr.M300135-JLR200
PREMALATHA, R.; SRIKUMAR, K.; VIJAYALAKSMI, D.; KUMAR, G. N.; MATHUR, P. P.: "28-Homobrassinolide: a novel oxysterol transactivating LXR gene expression", MOL. BIOL. REP., vol. 41, 2014, pages 7447 - 7461
CHEN, Z.; LIU, J.; FU, Z.; YE, C.; ZHANG, R.; SONG, Y.; ZHANG, Y.; LI, H.; YING, H.; LIU, H.: "24(S)-Saringosterol from edible marine seaweed Sargassum Fusiforme is a novel selective LXRP agonist", J. AGRIC. FOOD CHEM., vol. 62, 2014, pages 6130 - 6137
YANG, C.; YU, L.; LI, W.; XU, F.; COHEN, J. C.; HOBBS, H. H.: "Disruption of cholesterol homeostasis by plant sterols", J. CLIN. INVEST., vol. 114, 2004, pages 813 - 822
FOLEY, D.; O'CALLAGHAN, Y.; O'BRIEN, N. M.; MCCARTHY, F. O.; MAGUIRE, A. R.: "Synthesis and characterization of stigmasterol oxidation products", J. AGRIC. FOOD CHEM., vol. 58, 2010, pages 1165 - 1173
MISHARIN, A. Y.; MEHTIEV, A. R.; MOROZEVICH, G. E.; TKACHEV, Y. V; TIMOFEEV, V. P.: "Synthesis and cytotoxicity evaluation of 22,23-oxygenated stigmastane derivatives", BIOORG. MED. CHEM., vol. 16, 2008, pages 1460 - 1473, XP022453115, DOI: doi:10.1016/j.bmc.2007.10.056
CRUMP, D. R.; WILLIAMS, D. H.; PELC, B.: "22S)-Hydroxyvitamin D4", J. C. S. PERKIN I, 1973, pages 2731 - 2733
TADA, M.; OIKAWA, A.: "Synthesis of 22,23-epoxyvitamin D2 (22,23-Epoxyergocalciferol", J. C. S. PERKIN I, 1979, pages 1858 - 1861
MIYASHITA, N.; YOSHIKOSHI, A.; GRIECO, P. A.: "Pyridinium p-toluenesulfonate. A mild and efficient catalyst for the tetrahydropyranylation of alcohols", J. ORG. CHEM., vol. 42, 1977, pages 3772 - 3774
ZHANG, R.; HE, H. P.; DI, Y. T.; LI, S. L.; ZUO, G. Y.; ZHANG, Y.; HAO, X. J.: "Chemical constituents from Aphanamixis Grandifolia", CHEM. NAT. COMPD., vol. 49, 2013, pages 100 - 104
WEI, X.; SHU, P.; LIU, T.; XIANG, M.; ZHANG, J.; XUE, Y.; LUO, Z.; YAO, G.; ZHANG, Y.: "Steroids and phenylpropanoids with immunomodulatory activities from the stem barks of Cinnamomum Wilsonii", CHINESE J. ORG. CHEM., vol. 33, 2013, pages 1273
BRYNJOLFFSSEN, J.; HANDS, D.; MIDGLEY, J. M.; WHALLEY, W. B.: "Unsaturated steroids. Part I. Synthesis of 22,23-dihydroergosterol", J. C. S. PERKIN, vol. 11976, pages 826 - 828
BARTON, D. H. R.; POYSTER, J. P.; SAMMES, P. G.; HURSTHOUSE, M. B.; NEIDLE, S.: "Stereospecific and regiospecific addition to an isolated, acyclic (steroidal) olefinic bond", CHEM. COMMUN., 1971, pages 715 - 716
HONG, C.; TONTONOZ, P.: "Liver X receptors in lipid metabolism: opportunities for drug discovery", NAT. REV. DRUG DISCOVERY, vol. 13, 2014, pages 433 - 444
REPA, J. J.; TURLEY, S. D.; LOBACCARO, J. A; MEDINA, J.; LI, L.; LUSTIG, K.; SHAN, B.; HEYMAN, R. A; DIETSCHY, J. M.; MANGELSDORF,: "Regulation of absorption and ABC 1-mediated efflux of cholesterol by RXR heterodimers", SCIENCE, vol. 289, 2000, pages 1524 - 1529, XP000952750, DOI: doi:10.1126/science.289.5484.1524
VENKATESWARAN, A.; LAFFITTE, B. A.; JOSEPH, S. B.; MAK, P. A.; WILPITZ, D. C.; EDWARDS, P. A.; TONTONOZ, P.: "Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXRa", PROC. NATL. ACAD. SCI. U. S. A., vol. 97, 2000, pages 12097 - 12102, XP000960875, DOI: doi:10.1073/pnas.200367697
GLASS, C. K.; SAIJO, K.: "Nuclear receptor transrepression pathways that regulate inflammation in macrophages and T cells", NAT. REV. IMMUNOL., vol. 10, 2010, pages 365 - 376
ITO, A.; HONG, C.; RONG, X.; ZHU, X.; TARLING, E. J.; HEDDE, P. N.; GRATTON, E.; PARKS, J.; TONTONOZ, P.: "LXRs link metabolism to inflammation through Abcal-dependent regulation of membrane composition and TLR signaling", ELIFE, vol. 4, 2015, pages 1 - 23
GONZALEZ SIERRA, M.; BUSTOS, D. A.; ZUDENIGO, M. E.; RUVEDA, E. A.: "Configurational assignment of epimeric 22,23-epoxides of steroids by Carbon-13 NMR Spectroscopy", TETRAHEDRON, vol. 42, 1986, pages 755 - 758, XP026648724, DOI: doi:10.1016/S0040-4020(01)87482-2
VILLABLANCA, R. J.; RACCOSTA, L.; ZHOU, D.; FONTANA, R.; MAGGIONI, D.; NEGRO, A.; SANVITO, F.; PONZONI, M.; VALENTINIS, B.; BREGNI: "Tumor-mediated liver X receptor-a activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor rsponses", NAT. MED., vol. 16, 2010, pages 98 - 105
SHELDRICK, G. M.: "A short history of SHELX", ACTA CRYST., vol. A64, 2008, pages 112 - 122
Attorney, Agent or Firm:
SANTORO, Tiziana et al. (IT)
Download PDF:
Claims:
CLAIMS

1. A compound of formula (I)

wherein

the bond indicated by the arrow is a single or a double bond; and

R' is a group selected from:

wherein

- the dotted line indicates the bond by which said group is linked to the remaining part of the molecule;

- R is a linear or branched, saturated Ci-Ce-alkyl; and

n is an integer from 1 to 6;

as well as all their possible stereoisomers.

2. The compound of formula (I) according to claim 1 , characterized in that it is selected from the following compounds:

PF 015 PFM016

51

PFM028 PFM027

29 30

3. The compound of formula (I) according to claim 2, characterized in that it is selected from the following compounds

4, The compound of formula (I) according to claim 2 characterized in that it is selected from the following compounds

and

5. A compound of formula (J) according to anyone of claims 1 to 4, for its use as LXR modulator.

6. Use of a compound of formula (1) according to anyone of claims 1 to 4, as a laboratory tool.

7. A compound of formula (I) according to anyone of claims 1 to 4, for its use in therapy and diagnostics.

8. A compound according to anyone of claims 1 to 4, for its use in the prevention and/or treatment of diseases linked to LXR as well as in the prevention and/or treatment of cancer, inflammatory and/or immunological diseases, melanoma, prostate cancer and skin carcinoma, chronic inflammatory diseases, metabolic diseases and autoimmune diseases.

9. A pharmaceutical composition comprising a compound according to anyone of claims 1 to 4 with at least a pharmaceutically acceptable carrier.

10. The composition of claim 9 for its use in the prevention and/or treatment of diseases linked to LXR as well as in the prevention and/or treatment of cancer, inflammatory and/or immunological diseases, melanoma, prostate cancer and skin carcinoma, chronic inflammatory diseases, metabolic diseases and autoimmune diseases.

1 1. The composition of claim 9 which comprises one or more additional active ingredients.

12. A compound of formula (Π)

wherein R" is a protecting group of the hydroxy, R5 is as defined in claim 1 and the bond indicated by the arrow is a single or a double bond.

A compound of formula (II) according to claim 12, wherein R" is an acetyl or a tetrahydropyranyJ group.

Use of a compound according to anyone of claims 12 or 13 as a synthesis intermediate.

Description:
"Side-Chain Modified Ergosterol and Stigmasterol Derivatives as Liver X

Receptor modulators"

***

Summary of the invention

The present invention relates to novel Liver X Receptor (LXR) modulators, their use in diagnostic and therapy, more particularly in the treatment of diseases associated with LXR, such as cancer, inflammation, metabolic and autoimmune diseases.

Technical Background

Oxysterols are 27-carbon intermediates or end-products of cholesterol metabolism, structurally characterized by the presence of oxygenated functions such as hydroxy, keto, hydroperoxy, epoxy and carboxy moieties. They are produced in vivo through both enzymatic- and non-enzymatic (auto-oxidation) processes. 1 ' 2 Specific enzymes of the cytochrome P450 (CYP) family preferentially oxidize the cholesterol side chain (7a-hydroxycholesterol (la), 24(,S)-hydroxycholesterol (2), 22(R)-hydroxycholesterol (22R-HC, 3), and 24( ( S),25-epoxycholesterol (4) are examples of oxysterols generated by CYPs, see below), whereas the double bond of the cholesterol B-ring represents a privileged target for free-radical-involving reactions. Thus, 7-ketocholesterol (5), 7β- hydroxycholesterol (lb), 5α,6α- and 5p,6P-epoxycholesterols (6a,b) constitute the

4 5 6a. b

a: 5 ,6 -epoxy

b: 5p,6p-epoxy

A broader definition for the class of oxysterols is not limited to cholesterol oxidation products, but includes also steroidal oxygenated derivatives that humans can assimilate by diet, either as primary constituents (plants and shellfish sterols) or as storage and cooking-derived components. 1

The past two decades have evidenced an exponential increase in the number of studies on the physiological roles of mammalian oxysterols, as well as on their contribution to the pathogenesis of different diseases. 3 ' 4 ' 5 6 The major breakthrough was the identification of a specific subset of oxysterols (2-4) 7 8 as endogenous ligands of Liver X Receptor a and β (LXRs). 9 ' 10 ' 11 ' 12 ' 13 Thus, given the action of LXRs (a and β isoforms) as whole-body cholesterol sensors and key regulators of lipogenesis, oxysterols have the potential to assume a key role in the modulation of lipid metabolism and glucose homeostasis.

LXRs and their ligands can also suppress inflammatory responses, either by activating the genes that encode anti-inflammatory proteins or by suppressing the genes that are under the control of proinflammatory transcription factors. 4 14

However, the functions of oxysterols are not limited to their LXR binding, 15 but they significantly interact with other cellular proteins, giving rise to different effects. Examples of proteins affected by oxysterols are: a) insulin-induced gene (INSIG) proteins, regulating the function of sterol response element binding protein (SREBP); 16 b) Niemann-Pick CI (NPC1) and oxysterol-binding protein family (OSBP/ORP), involved in cholesterol metabolism; 17 and c) Smoothened oncoprotein, interfering with the Hedgehog signalling. 18

So far the oxysterol medicinal chemistry has been mainly focused on the identification of LXR modulators, although the number of the studied natural and synthetic oxysterol derivatives is only marginal when compared to that of the non-steroidal ligands. 19 ' 20 The first series of synthetic steroidal ligands allowed the identification of the minimal pharmacophore for LXRa, i.e. a sterol with a hydrogen bond acceptor at C-24. 21 The most potent derivative of this series, namely cholenic acid dimethylamide 7, was an efficacious LXRa agonist, 21 able to promote a gene-selective modulation (see below). 22 5a,6a-Epoxycholesterol (6a), identified in processed food, was shown to be a LXR modulator with cell and gene-context-dependent activities, 23 whereas the two 5P-cholane derivatives 3a,6a,24-trihydroxy-24,24-di(trifluoromethyl)-5P-cholane (ATI-829, 8) 24 and 3a,6a,24-trihydroxy-22-en-24,24-di(trifluoromethyl)-5P-chola ne (ATI-111, 9), 25 whose design was inspired by the structure of the potent non-steroidal agonist N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro- 1-hydroxy- 1 -

(trifluoromethyl)ethyl]phenyl]benzene sulfonamide (T0901317), 26 demonstrated antiatherosclerotic effects. 24 ' 25 In view of the well-known effect of phytosterols in reducing blood cholesterol, 27 and considering the fact that the treatment of intestinal cells with these compounds was found to increase the expression of LXR target genes, 28 Kaneko et al 29 studied the LXR activity of a series of phytosterols, including natural and semi-synthetic derivatives. They identified (22£)-ergost-22-ene-la,3P- diol (YT-32, 10) 29 as a potent and non-isoform selective LXR agonist, able to selectively induce the expression of ABC transporter genes in the intestine. Interestingly, the oral administration of 10 resulted in the inhibition of the intestinal cholesterol adsorption without increasing plasma triglyceride levels, in contrast to what observed with non-steroidal ligands. 19 ' 30

The study of phytosterols as LXR agonists is limited to the mentioned compound 10, to the plant hormone 28-homobrassinolide (ll), 31 and to 24(,S)-saringosterol (12), a minor component isolated from marine seaweeds which showed to act as a selective LXRp agonist. 32

Indeed, the medicinal chemistry of Liver X Receptor (LXR) modulators has been so far mainly focused on non-steroidal compounds. Despite some compounds endowed with high potency have been discovered, these are characterized by a low or null gene- selectivity, thus actually preventing their clinical applications. Indeed, the nonsteroidal compounds T0901317 and GW3965 having the following formulas

T0901317 GW3965

induce lipogenic effects by increasing liver and circulating triglyceride levels. The lipogenic effects have precluded further clinical development of these compounds. To date, few steroidal modulator so far reported have shown cell and gene-context- dependent activities. There is therefore a need for steroid-based LXR modulators which are endowed with a better pharmacokinetic properties and lower toxicity with respect of LXR agonists of the prior art.

Objects of the invention

It is an object of the invention to provide novel steroid-based compounds which are LXR modulators, particularly agonists.

It is a further scope of the invention the use of said steroid-based LXR modulators, especially agonists, in diagnostic and therapy, especially in the treatment of diseases associated with LXR, such as cancer, inflammation, metabolic and autoimmune diseases.

It is further scope of the invention to provide pharmaceutical compositions comprising said steroid-based LXR modulators.

Definitions

In the present application the following abbreviation will be used:

ABCA1, ATP -binding cassette transporter Al;

CYP, cytochrome; DMF, dimethyl formamide;

DMSO, dimethyl sulfoxide;

FASN, fatty acid synthase;

FXR, farnesoid X receptor;

hLXR, human liver X receptor;

INSIG, insulin-induced gene; LPS, lipopolysaccharides;

LXR, liver X receptor;

MCP-1, monocyte chemoattractant protein- 1;

mCPBA, m-chloroperoxybenzoic acid;

mpc, medium pressure chromatography;

NPC1, Niemann-Pick CI;

ORP, OSBP-related protein;

OSBP, oxysterol-binding protein;

PCR, polymerase chain reaction;

PMA, phorbol 12-myristate 13-acetate;

PPARy, peroxisome proliferator-activated receptor γ;

PPTS, pyridinum p-toluenesulfonate;

PXR, pregnane X receptor;

qPCR, quantitative polymerase chain reaction;

RNA, ribonucleic acid; RXR, retinoid X receptor;

SCDl, stearoyl-CoA desaturase 1;

SD, standard deviation;

SREBP, sterol response element binding protein;

TLR, toll-like receptor;

TNF, tumor necrosis factor.

Brief description of the Figures

Figure 1 shows the flowchart for the structural assignment of the stigmastane derivatives.

Figure 2 shows the flowchart for the structural assignment of the ergostane derivatives. Figure 3 shows the regulation of ABCAl (A), SREBPlc (B), FASN (C), and SCDl (D) genes by the title compounds assessed by qPCR. U937 cells differentiated with PMA for 72 hours were treated with T0901317 (10 μΜ), 22R-HC (3) or with the tested compound (10 μΜ). The results show mean ± SD of three biological samples, (n = 3/group). *p < 0.05, **p < 0.01, ***p < 0.001.

Figure 4 shows the regulation of SREBPlc (A), FASN (B), and SCD1 (C) genes by the title compounds assessed by qPCR. HepG2 cells were treated with T0901317 (10 μΜ), 22R-HC (3) (10 μΜ) or with the tested compound (10 μΜ). The results show mean ± SD of three biological samples, (n = 3/group). *p < 0.05, **p < 0.01, ***p < 0.001.

Figure 5 shows the regulation of CCL2 (A) and T Fa (B) genes by the title compounds assessed by qPCR. U937 cells differentiated with PMA for 72 hours were treated with LPS (100 ng/ml) in combination with T0901317 (10 μΜ) or with the title compounds (10 μΜ). The results show mean ± SD of three biological samples, (n = 3/group). *p < 0.05, **p < 0.01, ***p < 0.001.

Figure 6 shows ergosterol derivatives activate LXRa. Human embryonic kidney 293 cells were co-transfected with the GAL4-hLXRa expression plasmid together with GAL4 responsive element pMHC100X4-TK-luc and Renilla plasmids. After 24 hours cells were incubated with different concentrations of synthetic agonists derived from the ergosterol and assayed for luciferase activity. Luciferase activity is expressed as fold induction of the compound over the vehicle. The values represent mean ±S.D. RLA, relative luciferase activity

Figure 7 shows stigmasterol derivatives activate LXRa. Human embryonic kidney 293 cells were co-transfected with the GAL4-hLXRa expression plasmid together with GAL4 responsive element pMHC100X4-TK-luc and Renilla plasmids. After 24 hours cells were incubated with different concentrations of synthetic agonists derived from the stigmasterol and assayed for luciferase activity. Luciferase activity is expressed as fold induction of the compound over the vehicle. The values represent mean ±S.D. RLA, relative luciferase activity.

Figure 8 shows ergosterol derivatives activate LXR . Human embryonic kidney 293 cells were co-transfected with the GAL4-hLXR expression plasmid together with GAL4 responsive element pMHC100X4-TK-luc and Renilla plasmids. After 24 hours cells were incubated with different concentrations of synthetic agonists derived from the ergosterol and assayed for luciferase activity. Luciferase activity is expressed as fold induction of the compound over the vehicle. The values represent mean ±S.D. RLA, relative luciferase activity.

Figure 9 Shows stigmasterol derivatives activate LXR . Human embryonic kidney 293 cells were co-transfected with the GAL4-hLXRJ3 expression plasmid together with GAL4 responsive element pMHC100X4-TK-luc and Renilla plasmids. After 24 hours cells were incubated with different concentrations of synthetic agonists derived from the stigmasterol and assayed for luciferase activity. Luciferase activity is expressed as fold induction of the compound over the vehicle. The values represent mean ±S.D. RLA, relative luciferase activity.

Figure 10 shows analysis of RXR, PPARy, PXR and FXR nuclear receptors' activation by the ergosterol and stigmasterol derivatives. Human embryonic kidney 293 cells were co-transfected with GAL4-hRXR (A), GAL4-hPPARy (B), GAL4-hPXR (C) and GAL4-hFXR (D) expression plasmids together with GAL4 responsive element pMHC100X4-TK-luc and Renilla plasmids. Cells were incubated with vehicle (DMSO), PFMs at the concentration of 10 μΜ, and the nuclear receptor specific ligands 9cis retinoic acid (100 nM) (A), Rosiglitazone (RSG 100 nM) (B), T0901317 (100 nM) (C) and GW4064 (100 nM) (D) for six hours. The values represent mean ±S.D. RLA, relative luciferase activity.

Figure 11 shows the in vitro effects of PFM009 (13) and PFM018 (25) on LLC tumor cells.

Figure 12 shows the expression of the LXR target gene Abcal in LLC tumor cells induced by PFM009 (13) and PFM018 (25).

Figure 13 shows the controlling effects of LLC tumor growth in mice were treated with PFM009 (13) and PFM018 (25).

Description of the invention

According to one of its aspects, the invention relates to novel compounds of formula (I)

wherein

the bond indicated by the arrow is a single or a double bond; and R' is a group selected from:

wherein

the dotted line indicates the bond by which said group is linked to the remaining part of the molecule;

R is a linear or branched, saturated Ci-C6-alkyl, preferably Ci-C4-alkyl; and

n is an integer from 1 to 6;

as well as all their possible stereoisomers.

According to the present invention, the linear or branched, saturated Ci-C 6 - alkyl, preferably Ci-C4-alkyl, is more preferably a methyl or an ethyl group.

According to the present invention, compound (I) is selected from the followin formulas

Particularl preferred compounds are those having the following formulas 13 to 30:

PFM015 PFM016

15 16 10

PFtMOB PFIVI0O5

27 28

PFM028 PFM027

29 30

Particularly preferred compounds are compound 13 and compound 25.

Other particularly preferred compounds are compound 21, compound 27 and compound 28.

Other preferred compounds are compounds 29 and 30.

The compounds of the invention are useful as LXR modulators, especially as LXRa and/or LXR agonists.

As it will be disclosed in the experimental part, some of the compounds of the invention are selective LXRa agonists, such as compounds 21, 27 and 28; compound 25 is a selective LXRp agonist, while compound 13 and 29 are non-selective LXR agonists.

The compounds of the present invention may be prepared according to methods known in the art.

For instance, for the above compounds, epoxides 13-16, may be obtained by oxidizing the double bond between positions C-22 and C-23 of stigmasterol and ergosterol; alcohols 17-24 derives from the reductive opening of each epoxide, and ketones 25-28 results from the oxidation of the corresponding alcohols.

(22E)-3a,5a-cyclo-6P-methoxystigmast-22-ene (29), obtained in two steps from stigmasterol, as already reported, 34 represented the starting material for the preparation of the stigmastane derivatives (Scheme 1). The epoxidation reaction of 29 resulted in the formation of the two diastereoisomeric epoxides 30 and 31, which were separated by chromatography in 30 and 18% yield, respectively.35 The recovery of the 3P-hydroxy-5,6-ene moiety was performed by the known two-step procedure,35 consisting first in the treatment with glacial acetic acid, followed by the alkaline hydrolysis in the presence of hydroalcoholic potassium carbonate solution. Thus, starting from 30 and 31, the desired (22R,23R)-22,23-epoxystigmast-5-ene-3p-ol (13) and its (22S,23S)-isomer 14, respectively were obtained.

Scheme 1. Synthesis of (22R,23R)-22,23-Epoxystigmast-5-ene-3p-ol (13) and

-22,23-Epoxystigmast-5-ene-3p-ol (14) a

"Reagents and conditions: (a) / ' . ra-CPBA, NaHCC , CH2CI2, reflux, 2 h; / ' / ' . mpc; (b) / ' . glacial AcOH, reflux, 5h; ii. K 2 C0 3 , MeOH/H 2 0, reflux, 3 h.

The LiAlH4-promoted reductive opening of the oxirane ring of 30 (Scheme 2) gave the inseparable mixture of the corresponding 23 S- and 22S-hydroxy derivatives 32 + 33, which was first treated with glacial acetic acid and then in basic conditions to afford, after medium pressure chromatography (mpc), pure samples of (23S)-3P- stigmast-5-ene-3,23-diol (17) and (22S)-3p-stigmast-5-ene-3,22-diol (18). Scheme 2. Synthesis of (23S)-3p-Stigmast-5-ene-3,23-diol (17) and (22S)-3p- Stigmast-5-ene-3,22-diol (18) a

"Reagents and conditions: (a) L1AIH4, THF, reflux, 36 h; (b) / ' . glacial AcOH, reflux, 6 h; / ' / ' . 2M KOH, MeOH, reflux, 3h; Hi. mpc.

Similarly, the reductive opening of the epoxide 31 gave the inseparable mixture of the corresponding 23R- and 22R-hydroxy derivatives 34 + 35 (Scheme 3). In this case, the chromatographic separation of the two components of the mixture was only possible as 3P-acetate form. Thus, the mixture 34 + 35 was heated in glacial acetic acid and the crude submitted to mpc to achieve the two pure isomers 36 and 37. Their final alkaline hydrolysis gave the desired (23R)-3p-stigmast-5-ene-3,23-diol (19) and (22R)-3p- stigmast-5-ene-3,22-diol (20), respectively, thus completing the series of stigmastanediols.

Scheme 3. Synthesis of (23R)-3p-Stigmast-5-ene-3,23-diol (19) and (22R)-3p- Stigmast-5-ene-3,22-diol (20) a

"Reagents and conditions: (a) L1AIH4, THF, reflux, 36 h; (b) / ' . glacial AcOH, reflux, 6h; / ' / ' . mpc; (c) 2M KOH, MeOH, reflux, 3h.

Swern oxidation of (23R)-3P-acetoxystigmast-5-ene-23-ol (36) afforded the corresponding 23-keto derivative 38 (Scheme 4), which under basic hydrolysis gave the desired 3P-hydroxystigmast-5-ene-23-one (25). Scheme 4. Synthesis of 3P-Hydroxystigmast-5-ene-23-one (25) a

R = Ac, 38

R = H, 25

aReagents and conditions: (a) (COCl) 2 , DMSO, CH 2 C1 2 , -78°C, 2h, then Et 3 N, r.t; (b) 2M KOH, acetone, reflux, 3h.

Analogously, (22R)-3P-acetoxystigmast-5-ene-22-ol (37) was converted into the desired 3P-hydroxystigmast-5-ene-22-one (26) (Scheme 5).

Scheme 5. Synthesis of 3P-Hydroxystigmast-5-ene-22-one (26) a

a Reagents and conditions: (a) (COCl) 2 , DMSO, CH 2 C1 2 , -78°C, 2h, then Et 3 N, r.t; (b) 2M KOH, acetone, reflux, 3h.

3P-Acetoxy cycloadduct 40, obtained by Diels-Alder cycloaddition between ergosterol-3P-acetate and 4-phenyl-l,2,4-triazoline-3,5-dione,36 constituted the starting material for the synthesis of the ergostane derivatives: Its epoxidation reaction with mCPBA gave access to the inseparable mixture of the two diastereoisomeric epoxides 4 la.36 In an analogous manner the corresponding mixture of 3β- tetrahydropyranyl-protected epoxides 41b was also prepared starting from 3β- tetrahydropyranyloxy cycloadduct.37 The treatment of the mixture 41a with anhydrous potassium carbonate resulted in the retro 1,4-cycloaddition reaction, affording, after mpc, the two single isomers 42 and 43 (Scheme 6). The minor, less polar component 42, whose absolute configuration was assigned as 22R,23R (vedi infra), was submitted to alkaline hydrolysis to furnish (22R,23R)-22,23-epoxyergosta- 5,7-diene-3P-ol (15). The same procedure starting from the major, more polar epoxide 43 gave the corresponding (22S,23S)-22,23-epoxyergosta-5,7-diene-3P-ol (16).

Scheme 6. Synthesis of (22R,23R)-22,23-Epoxy-3p-ergosta-5,7-diene-3-ol (15) and (22S,23S)-22,23-Epoxy-3p-ergosta-5,7-diene-3-ol (16) a

R = Ac, 42 R = Ac, 43

R = H, 15 R = H, 16

"Reagents and conditions: (a) raCPBA, CH 2 C1 2 , r. , 5h; (b) K 2 C0 3 , DMF, reflux, 6h; (c) 2M KOH, EtOH, reflux, 15 min.

The reductive opening of the epoxide mixture 41b gave, after separation by mpc, three different fractions, constituted by (23R)-3P-tetrahydropyranyloxyergost-5,7-diene-23- ol (44), the inseparable mixture of (23 S)- and (22S)-3P-tetrahydropyranyl-protected diols (45 + 46), and (22R)-3p-tetrahydropyranyloxyergost-5,7-diene-22-ol (47) (Scheme 7). The deprotection of the 3P-hydroxy group of 44 by pyridinium p- toluenesulfonate (PPTS)38 provided the desired (23R)-3P-ergost-5,7-diene-3,23-diol (23).

Scheme 7. Synthesis of (23R)-3p-Ergost-5,7-diene-3,23-diol (23), (22R)-3p-stigmast- 5-ene-3,22-diol (24), 3p-Hydroxyergosta-5,7-diene-23-one (27), and 3β- Hydroxyergosta-5,7-diene-22-one (28) a

a Reagents and conditions: (a) LiAlH 4 , THF-Et 2 0, reflux, 36 h; (b) (COCl) 2 , DMSO, CH 2 C1 2 , -78°C, 2h, then Et 3 N, r.t.; (c) PPTS, EtOH, reflux, lh; (d) PPTS, acetone, reflux, 5h.

Subsequent Swern oxidation of the single alcohol 44 afforded the 3β- tetrahydropyranyl-23-keto derivative 48, which was deprotected under analogous mild acidic conditions to finally afford 3P-hydroxyergosta-5,7-diene-23-one (27). An analogous sequence, starting from the more polar, pure 22R-hydroxy derivative 47 gave access to the desired (22R)-3P-ergost-5,7-diene-3,22-diol (24) and 3β- hydroxyergosta-5,7-diene-22-one (28).

The ergostanediol series was completed by reducing the 3P-tetrahydropyranyl-23-keto derivative 48 with sodium borohydride, achieving almost quantitatively the mixture of the two 23-hydroxy epimers, which, after deprotection at C-3 position, gave the already obtained (23R)-3P-ergost-5,7-diene-3,23-diol (23), and the missing 23 S- epimer 21 (Scheme 8). Analogously, starting from the 22-keto derivative 49, (22S)- 3P-ergost-5,7-diene-3,22-diol (22) was achieved along with the already obtained 24 (Scheme 9). Scheme 8. Synthesis of (23R)-3p-Ergost-5,7-diene-3,23-diol (23) and (23S)-3p- Ergost-5,7-diene-3,23-diols (21) a

23 21 a Reagents and conditions: (a) . NaBHt, THF, 2-propanol, H 2 0, r.t.; ii. PPTS, EtOH, reflux, lh; / ' / ' / ' . mpc.

Scheme 9. Synthesis of (22S)-3p-Ergost-5,7-diene-3,22-diol (22) and (22R)-3p- Ergost-5,7-diene-3,22-diol (24) a

22 24

"Reagents and conditions: (a) / ' . NaBH*, THF, 2-propanol, H 2 0, r.t.; / ' / ' . PPTS, EtOH, reflux, lh; / ' / ' / ' . mpc.

Scheme 10: Synthesis of 24(S)-stigmast-5-ene-3p,24-dio (29) a

a Reagent and conditions: (a) . Ph3P=CHC(0)CH(CH3)2, toluene, 92 h, reflux; ii. H2, 40 psi, 10% Pd/C, NaHC03, EtOAc/THF, 30 h, r.t.; 60% yield, (b) 1M CH2=CHMgBr in THF, Et20, 2 h, reflux; ii. mpc, 79% yield, (c) pTosOH H20, 75% aqueous dioxane, 75 °C, 1 h; (d) H2, 40 psi, 10% Pd/C, NaHC03, EtOAc/THF, 12 h, r.t.

Absolute Configuration Assignment The workflows for the stereochemical elucidation of the newly created asymmetric centers are depicted in Figures 1 and 2.

In the case of the members of the stigmastane series it was used the X-ray single crystal diffraction analysis reported for (22R)-3P-stigmast-5-ene-3,22-diol, the only known derivative among the stigmastanediols here reported.39,40 By comparison of its reported spectroscopic data with those of our compounds, it was established that the more polar diol 20 corresponded to (22R)-3P-stigmast-5-ene-3,22-diol. Since 20 had been obtained from the reductive opening of the more polar oxirane isomer 14, as a consequence, the latter had to be endowed with the 22S,23S-absolute configuration. Thus, the other diol deriving from its reductive opening, namely 19, was assigned instead with the 23R-configuration (Figure 1). By exclusion, the diols 17 and 18 were characterized by the S-configuration at the newly formed side-chain chiral center, and the less polar epoxide 13 by 22R,23R-configuration. The respective position of the hydroxyl group in the two diols 17 and 18 was definitively established by their comparison with the compounds resulting from the reduction of 3P-hydroxystigmast- 5-ene-22-one (26).

Although the synthesis of some of our ergostane derivatives had been already reported, their structural assignment had been only presumed.36,41,42 To unambiguously proceed with the structural elucidation, the diol 23, derived by the hydrolysis of 44, the less polar, major fraction obtained by the reductive opening of the epoxide mixture 41b (Scheme 7), was submitted to single crystal X-ray analysis (Figure 1) and thus characterized as the (23R)-isomer. Consequently, the diol 24, since obtained by the hydrolysis of the other more abundant isomer 47 resulting from the same opening reaction (Scheme 7), was assigned as (22R)-3P-ergost-5,7-diene-3,22-diol (Figure 2). Since these two major isomers surely derived from the opening of a unique epoxide, the absolute configuration 22S,23S was assigned to the more abundant epoxide 16, and consequently, the 22R,23R-configuration to the less abundant 15. The diol obtained by the reduction of the 23-keto derivative 27, different from 23, had to be the (23S)-isomer 21, as well as the other diol deriving from the 22-keto derivative 28 and distinct from 24, was the 22 S -derivative 22.

Other preferred compounds, according to the present invention, are those listed in Table (I) Table (I) 21

22







According to another of its aspect, the invention relates to the use of the compounds of formula (I) and especially of the preferred compounds, as LXR modulators, more particularly as LXR agonists.

According to another of its aspect, the invention relates to the use of the compounds of formula (I) and especially of the preferred compounds, as laboratory tools in researches involving LXR.

According to another of its aspect, the invention relates to the use of the compounds of formula (I) and especially of the preferred compounds, in therapy and diagnostics. More particularly, the above compounds are useful in the prevention and/or treatment of diseases linked to LXR, such as, but not limited to, cancer, inflammatory and/or immunological diseases. Particularly preferred are the use in the prevention and/or treatment of melanoma, prostate cancer and skin carcinoma. Also preferred are chronic inflammatory diseases, metabolic diseases and autoimmune diseases.

For their use in therapy, the compounds of the invention are formulated in pharmaceutical compositions along with at least one pharmaceutically acceptable carrier according to the methods known in the art; said compositions constitute another aspect of the present invention.

The skilled in the art is aware of the type of carrier to choose, based upon the intended route of administration. The compositions of the invention are suitable for intravenous, intraperitoneal, subcutaneous, intramuscular, topical, transdermal or oral administration. Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.

The compositions of the invention may also comprise additional active ingredients. According to another of its aspects, the invention relates to a method for the prevention and/or the treatment of a disease linked to LXR, which comprises administering to a mammal in need thereof, an effective amount of at least one compound of formula (I). A preferred method, according to the invention, is a method for the prevention and/or treatment of cancer, inflammatory and/or immunological diseases. Particularly preferred are the methods in the prevention and/or treatment of melanoma, prostate cancer and skin carcinoma. Also preferred are chronic inflammatory diseases, metabolic diseases and autoimmune diseases.

According to another of its aspects, the invention relates to synthesis intermediate compounds having the followin formula (II):

wherein the bond indicated by the arrow is a single or a double bond, R' is as above defined and R" is a protecting group of the hydroxy, such as for instance an acetyl or a tetrahydropyranyl group.

The invention is illustrated in the Experimental Section which follows, in a non- limiting way.

Experimental Section

LXRs Activity

All the synthesized compounds were first tested for their ability to activate LXRs by using luciferase assays with GAL-4 chimeric receptors. These were performed by co- transfecting plasmids encoding hLXRa- and β-binding domains fused to GAL-4, with the respective responsive element conjugated with the luciferase reporter gene into the human embryonic kidney 293 cells. Results of the assays are listed in Table 2: most of the compounds exhibited low micromolar LXRs activity retaining or, in some cases, improving the magnitude of activity of the endogenous ligand 22R-HC (3).

Table 2. LXR Agonist Profile of Compounds 13-28

LXRa ECso (μΜ) 3 LXRp ECso (μΜ) 3 Efficacy

Compd ± SD Efficacy (%) b ± SD (%) b

(95% C.I.) b ± SD (95% C.I.) b ± SD

22R- 6.71 ± 0.71 4.75 ± 0.12

100 100 HC (3) (5.4 - 8.2) (3.4 - 6.4)

2.09 ± 0.57 3.79 ± 0.82

13 488.8 ± 89.1 142.1 ±16.8

(1.1 - 3.4) (2.5 - 5.6)

aFifty% maximal activation (EC50) ± standard deviations (SD) was determined by

dose-response curve of titrating concentrations of compounds 13-28 (32, 16, 8, 4,

2 and 1 μΜ) tested by luciferase assays. The results were mean of three-five independent experiments; b Efficacy: % of compound effect ± SD versus 8 μΜ of

22R-HC; °NA: not active.

Concerning the isoform selectivity profile, besides non-selective and poorly preferential LXRa agonists (13, 16 and 19) (Figures 6-9), other compounds, such as 21, 27 and 28, deserve to be highlighted as selective LXRa agonists. Among them, the derivative 27 showed to be the most promising a-selective agonist thanks to its lower EC50 value and higher efficacy respect to the reference compound 3.

Furthermore, 20, 22 and 25 and 29 showed a good selectivity for the LXRP isoform in terms of EC50.

The reported compounds 25 and 29 showed to be endowed with improved isoform selectivity in comparison to the known derivative 12. The beta/alfa efficacy ratios for compounds 25 and 29, indeed, were 3.7 and 4.4, respectively, respect to the value of 1.4 for 12.

In particular, 25 can be considered a LXRP-selective agonist, being virtually inactive (EC50 = 14.51 μΜ) at LXRa (Figures 7 and 9). Of note, 25 while showing approximately 50% of efficacy in terms of LXRJ3 activation, was endowed with the lowest efficacy at LXRa, as compared to the 22R-HC (3) (Table 2); Thus, confirming its selectivity for the LXRP isoform. From a structural point of view, all the selective LXRa agonists are ergostane derivatives, whereas the preferential LXRp ligands belong to the two classes; however the most interesting compound in this sense, namely 25, is a stigmastane derivative.

Moreover, the skeleton system more than the nature and, where applicable, the stereochemistry of the side-chain modification, appeared to strictly influence both potency and isoform selectivity. Indeed, with R,R-epoxy derivatives 13 and 15, as the only exceptions, any equally side-chain modified ergostane and stigmastane derivatives did not show similar biological profile.

The selectivity of our compounds within the nuclear receptor superfamily was also evaluated by luciferase assays using GAL4-RXR, -PPARy, -PXR and -FXR plasmids. No compound was able to activate RXR or PPARy, whereas a slight activation of PXR by 21, 22 and 15, and a strong FXR activation by 20 was observed (Figure 10).

Gene expression profile

LXR agonists induce the expression of target genes, which are involved in cholesterol homeostasis, particularly in the reverse cholesterol transport pathway.43 Indeed, LXR agonists induce the expression of ABCAl both in macrophages and in many tissues of the periphery such as the intestine.44 Moreover, ABCAl regulates cholesterol efflux to APOAI acceptors.45 In the liver, LXR activation promotes the biosynthesis of fatty acids, a process also termed as de novo lipogenesis by inducing the expression of the master regulator of hepatic lipogenesis sterol-regulatory element-binding protein 1C (SREBP-lc), as well as several downstream genes in the SREBP-lc pathway, including steroyl CoA desaturase 1 (SCD1) and fatty acid synthase (FASN).43 Therefore, it was investigated, by quantitative PCR (qPCR), the expression of ABCAl , SREBPlc, FASN, and SCD1, by using RNA from monocytic U937 cells (Figure 3) and from hepatic HepG2 cells (Figure 4) stimulated with our compounds, the nonsteroidal agonist T0901317 or the endogenous ligand 22R-HC (3) as positive controls. As shown in Figure 3 A, all the compounds, except 18 and 15, were able to induce ABCAl expression, although to a different extent. With most derivatives, a mild up- regulation of the gene expression (2 fold) was observed, whereas, interestingly, with 13, 19, 20 and 25 a strong induction of ABCAl expression was detected, comparable to that caused by T0901317. Noteworthy, for all our compounds the level of up- regulation of SREBP-lc was much lower than that observed for T0901317 and comparable to the level obtained with the natural ligand 22R-HC (3) (Figure 3B). The effects observed on FASN and SCD1 genes were even more interesting: no compound up-regulated the mRNA levels of FASN (Figure 3C); a slight activation (below 2-fold) of SCD1 was detected only for 16 and 25, with the latter being statistically significant (Figure 3D). Also the natural ligand 22R-HC (3) did not induce up-regulation of FASN and SCD1 transcripts (Figures 3C and 3D). These data were confirmed at later time points (i.e. 16 hours). Then, the induction of genes involved in the lipogenesis using the hepatic cell line HepG2. 43,22 was evaluated. By qPCR analysis it was observed only a significant up-regulation of SREBP-lc induced by 13 and 16 compounds, while all the other compounds turned out to be negative (Figure 4A). No compound up- regulated the mRNA levels of FASN (Figure 4B) and SCD1 (Figure 4C). According to all these evidences, the derivatives 13, 19, 20 and 25, being strong inducers of ABCAl, poor activators of SREBPlc and SCD1 in the U937 cell line, showed to be very promising derivatives. Over the time, indeed, substantial efforts have been dedicated to the identification of LXR ligands able to turning on ABC transporter genes, without affecting lipogenic genes levels. This task is still one of the major challenge to the discovery of a clinically useful LXR modulator for atherosclerosis. According to the isoform selectivity profile, 13 and 19 were non-selective ligands, 20 and 25 were a preferential and a selective LXRp agonist, respectively, thus evidencing that in our model the ability to not up-regulate the genes involved in lipogenesis was not a phenomenon exclusive of LXRP-selective modulators.

Being LXRs not only transcriptional regulators of the cholesterol and lipid homeostasis, but also able to exert potent anti-inflammatory effects through the interference of TLRs 2, 4 and 9 signaling,46 it was decided to verify whether our compounds were also capable of modulating genes involved in the inflammatory pathways, such as the MCP-1/CCL2 and T Fa genes, which have been shown to be inhibited when LXRs are engaged in the presence of LPS.47 To this purpose, differentiated U937 cells was treated for 6 hours with our compounds in combination with LPS (100 ng/ml) and then the treated cells for the expression of CCL and TNFa by qPCR was evaluated. Most of the compounds were able to inhibit CCL2 expression with 21, 22, 19 and 16 showing the same grade of potency of the positive control T0901317 (Figure 5 A). Most of the compounds were also able to inhibit TNFa with 21, 13, 19 and 25 being the most active (Figure 5B). Similar results were obtained by using the endogenous ligand 22R-HC (3).

Chemistry.

Melting points were determined by the capillary method on a Biichi 535 electrothermal apparatus and are uncorrected. 1H- and 13C NMR spectra were taken on a Bruker AC 400 spectrometer as solutions in CDC13 unless otherwise indicated. The spin multiplicities are indicated by the symbols s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), and bs (broad). Flash chromatography was performed on Merck silica gel (0.040-0.063 mm). Medium pressure chromatography (mpc) was performed on Merck LiChroprep Si 60 Lobar columns. Microanalyses were carried out on a Carlo Erba 1106 elemental analyzer and the results were within ± 0.4% of the theoretical values. All solvents were distilled and dried according to standard procedures. Purity was determined by microanalysis to be >95% for all final compounds.

Example 1

(22R,23R)-22,23-Epoxystigmast-5-ene-3p-ol (13). The epoxide 30 (0.067 g, 0.15 mmol) was refluxed in glacial acetic acid (5 mL) for 5 h. The residue obtained by the removal of the solvent in vacuo was directly dissolved in methanol/water (2: 1, 12 mL) and the resulting solution treated with K2C03 (0.26 g, 1.86 mmol) and refluxed for 3 h. After cooling the reaction mixture was extracted with CH2C12 (3x10 mL) and the combined organic layers dried over Na2S04. The solvent was removed in vacuo and the residue submitted to mpc. Elution by light petroleum-ethyl acetate (80:20) afforded pure sample of 13 : 36% yield; mp 173.2-175.4 °C; 1H NMR (400 MHz) δ 0.69 (s, 3H), 2.28-2.29 (m, 2H), 2.49-2.50 (m, 1H), 2.75 (dd, 1H, J = 9.32 and 2.21 Hz), 3.52 (m, 1H), 5.35-5.36 (m, 1H); 13C NMR (100 MHz) δ 11.82, 12.45, 16.17, 19.37, 19.54, 20.17, 20.85, 21.01, 24.53, 27.93, 29.13, 31.61, 31.88 (2C), 36.48, 37.22, 38.66, 39.55, 42.24, 42.62, 48.28, 50.07, 53.42, 56.35, 62.14 (2C), 71.74, 121.54, 140.79; Anal. Calcd for C29H4802: C, 81.25%; H, 11.29%. Anal. Found: C, 81.17%; H, 11.24%.

Example 2

(22S,23S)-22,23-Epoxystigmast-5-ene-3p-ol (14). The epoxide 31 was treated as reported for compound 30 to furnish 14 in 29% yield; mp 127.8-130.2 °C; 1H NMR (400 MHz) δ 0.68 (s, 3H), 2.24-2.30 (m, 2H), 2.49-2.54 (m, 2H), 3.53 (m, 1H), 5.35- 5.37 (m, 1H); 13C NMR (100 MHz) 11.97, 12.36, 16.28, 19.36 (2C), 20.92, 21.06, 24.51, 27.07, 29.31, 29.68, 31.62, 31.87 (2C), 36.48, 37.25, 38.87, 39.67, 42.27, 42.67, 48.77, 50.17, 56.02, 56.32, 58.55, 63.13, 71.77, 121.67, 140.67; Anal. Calcd for C29H4802: C, 81.25%; H, 11.29%. Anal. Found: C, 81.33%; H, 11.27%.

Example 3

(22R,23R)-22,23-Epoxyergosta-5,7-diene-3p-ol (15). 2M KOH solution (0.2 mL) was added to a solution of 42 (0.037 g, 0.08 mmol) in EtOH (3.8 mL) and the resulting mixture was refluxed for 15 min. After cooling the reaction mixture was extracted with EtOAc (4x5 mL) and the combined organic layers were washed with brine (8 mL), dried over Na2S04, filtered and the solvent removed in vacuo to give a residue which was submitted to flash chromatography. Elution with light petroleum-ethyl acetate (80:20) afforded 15 in 64% yield; mp: 163.8-165.2 °C; 1H NMR (400 MHz) δ 0.61 (s, 3H), 3.61-3.65 (m, 1H), 5.39-5.41 (m, 1H), 5.57-5.58 (m, 1H); 13C NMR (100 MHz) 11.9, 13.7, 16.2, 16.3, 19.5, 20.4, 21.0, 23.2, 26.8, 31.1, 31.9, 37.0, 38.3, 39.0 (2C), 40.7, 42.3, 43.2, 46.2, 54.0, 55.6, 60.4, 64.3, 70.3, 116.5, 119.5, 139.8, 140.8; Anal. Calcd for C28H4402: C, 81.50%; H, 10.76%. Anal. Found: C, 81.17%; H, 10.74%. Example 4

(22S,23S)-22,23-Epoxyergosta-5,7-diene-3p-ol (16). The derivative 43 was treated as reported for 42 to furnish 16 in 89% yield: mp: 138.3-139.6 °C; 1H NMR (400 MHz) δ 0.60 (s, 3H), 3.60-3.66 (m, 1H), 5.39-5.41 (m, 1H), 5.56-5.58 (m, 1H); 13C NMR (100 MHz) 11.8, 12.5, 16.2, 17.1, 18.5, 20.2, 21.0, 23.3, 27.8, 31.0, 31.9, 37.0, 38.3, 39.0, 39.8, 40.7, 42.5, 43.2, 46.1, 53.3, 54.0, 63.1, 63.8, 70.3, 116.5, 119.4, 140.0, 140.7; Anal. Calcd for C28H4402: C, 81.50%; H, 10.76%. Anal. Found: C, 81.32%; H, 10.77%.

Example 5

(23S)-3p-Stigmast-5-ene-3,23-diol (17) and (22S)-3p-Stigmast-5-ene-3,22-diol (18). LiAlH4 (0.25 g, 6.71 mmol) was portion wise added to the solution of the epoxide 30 (0.27 g, 0.61 mmol) in anhydrous THF (15 mL). The resulting mixture was refluxed for 36 h under an argon atmosphere. After cooling, first EtOAc and then water were carefully added. The organic phase was separated and the water phase extracted with EtOAc (3x15 mL). The combined organic phases were washed with brine (20 mL) and then dried over Na2S04. After filtration, the solvent was evaporated in vacuo to give a residue, which was dissolved in glacial acetic acid (5 mL) and the resulting solution refluxed for 6 h. After cooling, the mixture of 32 + 33, obtained by the removal of the solvent in vacuo, was directly dissolved in methanol (16 mL) and treated with 2M KOH solution (8 mL). After refluxing for 3 h, the reaction mixture was extracted with EtOAc (3 x 15 ml). The combined organic layers were dried over Na2S04, filtered and the solvent removed in vacuo, to give a residue which was submitted to mpc. Elution by light petroleum-ethyl acetate (70:30) afforded pure samples of the desired compounds in 69% total yield; 17: mp 178.2-181.4 °C; 1H MR (400 MHz) δ 0.69 (s, 3H), 2.23-2.31 (m, 2H), 3.53 (m, 1H), 3.91 (m, 1H), 5.36 (m, 1H); 13C MR (100 MHz) 11.79, 13.82, 18.28, 19.38 (2C), 19.85, 21.03, 21.12, 24.24, 28.45, 28.54, 31.58, 31.82 (2C), 34.16, 36.44, 37.19, 39.73, 42.22, 42.35, 42.46, 49.13, 50.01, 56.66, 56.88, 70.55, 71.73, 121.62, 140.72; Anal. Calcd for C29H50O2: C, 80.87%; H, 11.70%. Anal. Found: C, 80.63%; H, 11.72%; 18: mp 168.9-172.4 °C; 1H NMR (400 MHz) δ 0.70 (s, 3H), 2.24-2.31 (m, 2H), 3.53 (m, 1H), 3.75 (t, 1H, J = 6.77 Hz), 5.35 (d, 1H, J = 5.21 Hz); 13C NMR (100 MHz) 11.39, 11.73, 11.85, 18.89, 19.10, 19.38, 21.05, 23.27, 24.18, 27.8, 28.84, 31.58, 31.80, 31.88, 35.77, 36.43, 37.20, 39.72, 39.92, 42.04, 42.20 (2C), 50.02, 52.58, 56.61, 71.71, 71.86, 121.60, 140.73; Anal. Calcd for (C29H50O2): C, 80.87%; H, 11.70%. Anal. Found: C, 80.70%; H, 11.65%.

Example 6 (23R)-3p-Stigmast-5-ene-3,23-diol (19). A solution of 36 (0.03 g, 0.06 mmol) in MeOH (3 mL) was treated with 2M KOH solution (1 mL) and the resulting mixture was refluxed for 30 min. After cooling the reaction mixture was extracted with EtOAc (3x10 mL) and the combined organic layers were dried over Na2S04, filtered and the solvent removed in vacuo. The residue, thus obtained, was purified by flash chromatography: elution with light petroleum-ethyl acetate (80:20) afforded 19 in 55% yield: mp 158.1-158.6 °C; 1H MR (400 MHz) δ 0.72 (s, 3H), 2.27-2.28 (m, 2H), 3.51 (m, 1H), 3.69-3.74 (m, 1H), 5.35 (d, 1H, J = 5.25 Hz); 13C MR (100 MHz) 11.94, 14.48, 18.63, 18.96, 19.17, 19.36, 21.09, 21.44, 24.25, 27.80, 28.50, 31.67, 31.90 (2C), 32.78, 36.51, 37.28, 39.87, 41.11, 42.32, 42.51, 50.17, 52.49, 56.92 (2C), 70.25, 71.76, 121.60, 140.81; Anal. Calcd for C29H50O2: C, 80.87%; H, 11.70%. Anal. Found: C, 80.67%; H, 11.66%.

Example 7

(22R)-3P-stigmast-5-ene-3,22-diol (20). The derivative 37 was treated as reported for compound 36 to furnish 20 in 72% yield: mp: 149.2-149.9 °C; 1H NMR (400 MHz) δ 0.72 (s, 3H), 3.51 (m, 1H), 3.69-3.74 (m, 1H), 5.35 (d, 1H, J = 5.25 Hz); 13C NMR (100 MHz) 11.75 (2C), 12.32, 17.74, 19.38, 20.45, 21.11, 23.60, 24.45, 27.50, 28.92, 29.65, 30.11, 31.70, 31.92, 36.54, 37.30, 39.81, 41.53, 42.33, 42.59, 42.70, 50.22, 53.07, 56.39, 71.39, 71.78, 121.59, 140.85; Anal. Calcd for C29H50O2: C, 80.87%; H, 11.70%. Anal. Found: C, 80.91%; H, 11.69%.

Example 8

(23R)-3p-Ergost-5,7-diene-3,23-diol (23) and (23S)-3p-Ergost-5,7-diene-3,23-diol (21). NaBH4 (0.13 g, 3.44 mmol) was added to a solution of the ketone 48 (0.10 g, 0.2 mmol) in THF-2-propanol (2: 1, 6 mL) and the resulting mixture was stirred at room temeprature overnight. The reaction mixture was then diluted with H20 (5 mL) and extracted with Et20 (3x5 mL). The combined organic layers were washed with brine (10 mL), dried over Na2S04, filtered and the solvent removed in vacuo. The residue, thus obtained, was dissolved in EtOH (10 mL) and treated with PPTS (0.012 g, 0.047 mmol). After refluxing for 1 h, the reaction mixture was allowed to cool to room temperature, and the solvent was removed in vacuo to give a residue, which was submitted to mpc. Elution with light petroleum-ethyl acetate (90: 10) afforded pure samples ofthe desired compounds in 78% total yield; 21 : mp 129.8-130.7 °C; 1H MR (400 MHz) δ 0.65 (s, 3H), 3.62-3.69 (m, 2H), 5.40-5.42 (m, 1H), 5.59 (dd, 1H, J = 7.89, 2.35 Hz). 13C MR (100 MHz) 10.54, 11.71, 16.27, 17.92, 20.67, 21.04, 21.75, 23.05, 27.74, 28.40, 31.92, 35.75, 36.97, 38.32, 39.09, 40.70 (2xC), 43.00, 45.32, 46.16, 54.37, 56.76, 70.44, 73.30, 116.33, 119.54, 139.80, 141.24; Anal. Calcd for C28H4602: C, 81.10%; H, 11.18%. Anal. Found: C, 80.97%; H, 11.19%.

Example 9

(22R)-3p-Ergost-5,7-diene-3,22-diol (24) and (22S)-3p-Ergost-5,7-diene-3,22-diol (22). The derivative 49 was treated as reported for 48 to furnish pure samples of the desired compounds 24 and 22 in 83% total yield. 22: mp 117.3-121.0 °C; 1H NMR (400 MHz) δ 0.65 (s, 3H), 0.79 (d, 3H, J = 6.84 Hz), 3.63-3.66 (m, 1H), 3.78-3.81 (m, 1H), 5.40-5.42 (m, 1H), 5.59-5.61 (m, 1H); 13C NMR (100 MHz) 11.79, 12.49, 15.55, 16.02, 16.22, 21.04, 21.12, 23.15, 23.80, 27.39, 29.53, 31.92, 34.60, 35.27, 36.98, 38.30, 39.09, 40.72, 43.00, 46.14, 52.73, 54.01, 70.40, 71.67, 116.43, 119.54, 139.90, 140.96; Anal. Calcd for C28H4602: C, 81.10%; H, 11.18%. Anal. Found: C, 81.09%; H, 11.17%.

Example 10

(23R)-3p-Ergost-5,7-diene-3,23-diol (23). PPTS (0.010 g, 0.039 mmol) was added to a solution of 44 (0.050 g, 0.1 mmol) in EtOH (5 mL) and the resulting mixture was refluxed for 5 h. After cooling the solvent was removed in vacuo and the residue was purified by flash chromatography. Elution with light petroleum-ethyl acetate (80:20) furnished 23 in 70% yield: mp 167.8-169.4 °C; 1H NMR (400 MHz) δ 0.68 (s, 3H), 3.68 (m, 1H), 3.82 (m, 1H), 5.44 (s, 1H), 5.61 (s, 1H). 13C NMR (100 MHz) 9.83, 11.88, 16.25, 18.45, 18.79, 21.05, 21.50, 22.98, 28.33, 29.55, 31.92, 33.11, 36.97, 38.33, 39.18, 40.73, 42.11, 43.01, 45.37, 46.17, 54.52, 56.47, 70.38, 70.64, 116.34, 119.53, 139.80, 141.19; Anal. Calcd for C28H4602: C, 81.10%; H, 11.18%. Anal. Found: C, 81.26%; H, 11.16%.

Example 11

(22R)-3P-Ergost-5,7-diene-3,22-diol (24). The derivative 47 was treated as reported for 44 to furnish 24 in 73% yield: mp 197.7-201.2 °C; 1H NMR (400 MHz) δ 0.65 (s, 3H), 0.79 (d, 3H, J = 6.84 Hz), 3.63-3.66 (m, 1H), 3.78-3.81 (m, 1H), 5.40-5.42 (m, 1H), 5.59-5.61 (m, 1H); 13C NMR (100 MHz) 11.79, 12.49, 15.55, 16.02, 16.22, 21.04, 21.12, 23.15, 23.80, 27.39, 29.53, 31.92, 34.60, 35.27, 36.98, 38.30, 39.09, 40.72, 43.00, 46.14, 52.73, 54.01, 70.40, 71.67, 116.43, 119.54, 139.90, 140.96; Anal. Calcd for C28H4602: C, 81.10%; H, 11.18%. Anal. Found: C, 80.86%; H, 11.20%. Example 12

3P-Hydroxystigmast-5-ene-23-one (25). A solution of DMSO (0.03 g, 0.38 mmol) in anhydrous CH2C12 (0.5 mL) was added to a solution of oxalyl chloride (0.025 g, 0.2 mmol) in anhydrous CH2C12 (1 mL), kept at -60 °C under an argon atmosphere. After the resulting mixture was stirred for 15 min at -60 °C, the solution of alcohol 36 (0.048 g, 0.1 mmol) in anhydrous CH2C12 (1 mL) was added. The mixture was stirred for 2 h at -55/60 °C before the addition of Et3N (0.08 g, 0.76 mmol). After the reaction mixture was allowed to reach room temperature, stirring was continued for 15 min, and then water (10 mL) was added. The reaction mixture was extracted with CH2C12 (3x5 mL), the combined organic layers were washed with brine (10 mL), dried over Na2S04, filtered and the solvent removed in vacuo, to give the crude ketone 38, which was dissolved in acetone (2 mL) and treated with 2M KOH solution (0.5 mL). The resulting solution was refluxed for 40 min, cooled and extracted with EtOAc (3x5 mL). The combined organic layers were dried over Na2S04, filtered and the solvent removed in vacuo to give a residue then submitted to flash chromatography. Elution with light petroleum-ethyl acetate (80:20) furnished 25 in 51% yield: mp: 174.2-174.8 °C; 1H NMR (400 MHz) δ 0.62 (s, 3H), 2.40-2.42 (m, 2H), 3.40-3.47 (m, 1H), 5.26 (d, 1H, J = 4.9 Hz); 13C NMR (100 MHz) 11.77, 12.02, 16.57, 18.46, 19.37, 19.61, 21.04, 23.90, 24.52, 27.64, 28.93, 31.64, 31.83, 31.90, 36.50, 37.27, 39.66, 39.82, 42.29, 42.47, 43.27, 49.66, 50.12, 51.95, 56.11, 71.72, 121.51, 140.78, 214.50; Anal. Calcd for C29H4802: C, 81.25%; H, 11.29%. Anal. Found: C, 81.32%; H, 11.31%. Example 13

3P-Hydroxystigmast-5-ene-22-one (26). The derivative 37 was treated as reported for the compound 36 to furnish 26 in 59% yield: mp: 151.7-152.8 °C; lHNMR (400 MHz) δ 0.72 (s, 3H), 3.48-3.53 (m, 1H), 5.32-5.33 (m, 1H); 13C NMR (100 MHz) 11.85, 19.35, 19.70, 20.06, 21.18, 21.57, 24.22, 28.37, 29.17, 31.62, 31.71, 31.85, 36.47, 37.24, 39.66, 42.26, 42.42, 50.06, 51.04, 55.70, 56.82, 60.82, 71.73, 121.52, 140.81, 214.58; Anal. Calcd for C29H4802: C, 81.25%; H, 11.29%. Anal. Found: C, 81.49%; H, 11.28%.

Example 14

3p-Hydroxyergost-5,7-diene-23-one (27). PPTS (0.010 g, 0.039 mmol) was added to a solution of 48 (0.050 g, 0.1 mmol) in acetone (5 mL) and the resulting mixture refluxed for 5 h. After cooling the solvent was removed in vacuo and the residue was purified by flash chromatography. Elution with light petroleum-ethyl acetate (80:20) furnished 27 in 78% yield: mp 104.4-105.6 °C; 1H MR (400 MHz) δ 5.57 (m, 1H), 5.39 (m, 1H), 3.6 (m, 1H), 5.38-5.40 (m, 1H), 5.56-5.58 (m, 1H); 13C MR (100 MHz) 11.8, 12.6, 16.2, 18.6, 20.0, 21.0, 21.4, 22.9, 28.2, 30.0, 31.8, 32.2, 36.9, 38.3, 39.0, 40.7, 42.9, 46.1, 49.0, 52.7, 54.4, 55.5, 70.3, 116.4, 119.4, 139.91, 140.9, 215.1; Anal. Calcd for C28H4402: C, 81.50%; H, 10.75%. Anal. Found: C, 81.73%; H, 10.71%.

Example 15

3P-Hydroxyergost-5,7-diene-22-one (28). The derivative 49 was treated as reported for 48 to furnish 28 in 58% yield: mp 118.2-122.6 °C. 1H NMR (400 MHz) δ 0.65 (s, 3H), 3.63-3.68 (m, 1H), 5.39-5.40 (m, 1H), 5.57-5.59 (m, 1H); 13C NMR (100 MHz) 11.94, 15.88, 16.23, 16.70, 18.17, 20.95, 23.19, 27.34, 31.82, 31.92, 33.61, 36.93, 38.26, 38.95, 40.65, 43.00, 46.07, 46.66, 49.98, 51.81, 53.66, 70.30, 116.56, 119.42, 139.97, 140.47, 214.79; Anal. Calcd for C28H4402: C, 81.50%; H, 10.75%. Anal. Found: C, 81.70%; H, 10.78%.

Example 16

(22R,23R)-22,23-Epoxy-3a,5a-cyclo-6p-methoxystigmastane (30) and (22S,23SR)- 22,23-Epoxy-3a,5a-cyclo-6P-methoxystigmastane (31). NaHC03 (7.34 g, 87 mmol,) and 77% m-CPBA (3.54 g, 16.8 mmol) were added to the solution of (22Ε)-3α,5α- cyclo-6P-methoxystigmast-22-ene34 (29) (3.0 g, 7.0 mmol) in CHC13 (60 mL) and the resulting mixture was refluxed for 2 h. After cooling the reaction mixture was washed with 10% Na2S203 solution (3x50 mL), water (50 mL), and then dried over Na2S04. The solvent was removed in vacuo and the residue submitted to mpc. Elution by light petroleum-ethyl acetate (95:5) afforded pure samples of 30 and 31 in 30% and 18% yields, respectively. Their spectral data were in agreement with those previously reported.48

Example 17

23R)-3p-Acetoxystigmast-5-ene-23-ol (36) and (22R)-3p-Acetoxystigmast-5-ene-22- ol (37). LiAlH4 (0.22 g, 5.94 mmol) was portion wise added to the solution of the epoxide 31 (0.24 g, 0.54 mmol) in anhydrous THF (15 mL). The resulting mixture was refluxed for 36 h under an argon atmosphere. After cooling, first EtO Ac and then water were carefully added. The organic phase was separated and the water phase extracted with EtO Ac (3x15 mL). The combined organic phases were washed with brine (20 mL) and then dried over Na2S04. After filtration, the solvent was evaporated in vacuo to give the mixture of 34 + 35, which was dissolved in glacial acetic acid (10 mL) and the resulting solution refluxed for 3 h. After cooling, the solvent was removed in vacuo to give a residue, which was submitted to mpc. Elution by light petroleum-ethyl acetate (80:20) afforded pure sample of (23R)-3P-acetoxystigmast-5-ene-23-ol (36): 36% yield; mp 132.1-132.6 °C; 1H NMR (400 MHz) δ 0.70 (s, 3H), 2.01 (s, 3H), 2.30 (d, 2H, J = 7.56 Hz), 3.67-3.71 (m, 1H), 4.58 (m, 1H), 5.36 (d, 1H, J = 4.25 Hz); 13C NMR (100 MHz) 11.86, 14.56, 18.53, 18.82, 19.03, 19.22, 20.93, 21.37 (2C), 24.17, 27.69 (2C), 28.45, 31.72, 31.79, 32.70, 36.47, 36.89, 38.02, 39.68, 40.93, 42.38, 49.88, 52.33, 56.70, 56.76, 70.04, 73.88, 122.53, 139.51, 170.48. Further elution with the same eluent afforded (22R)-3P-acetoxystigmast-5-ene-22-ol (37): 21% yield; mp 123.9-125.2 °C; 1H NMR (400 MHz) δ 0.70 (s, 3H), 2.03 (s, 3H), 2.31 (d, 2H, J = 7.11 Hz), 3.71 (d, 1H, J = 10.12 Hz), 4.60 (m, 1H), 5.37 (s, 1H); 13C NMR (100 MHz) 11.77, 11.81, 12.26, 17.53, 19.26, 20.52, 20.96, 21.41, 23.50, 24.36, 27.39, 27.69, 28.62, 29.77, 31.80 (2C), 36.50, 36.93, 38.04, 39.61, 41.30, 42.45, 42.57, 49.97, 52.90, 56.18, 71.19, 73.88, 122.51, 139.59, 170.54.

Example 18

3P-Acetoxy-5a,8a-(3,5-dioxo-4-phenyl-l,2,4-triazolidino)-22, 23-epoxyergost-6-ene (41a). 77% m-CPBA (0.42 g, 1.87 mmol) was added to the solution of ergosterol acetate adduct 4036 (1.0 g, 1.63 mmol) in CH2C12 (10 mL) and the resulting mixture was stirred at room temperature for 5 h. Then, the reaction mixture was filtered and the solution washed with 5% NaHC03 solution (2x10 mL) and brine (10 mL). The organic phase was dried over Na2S04, filtered and the solvent was removed in vacuo to give a residue which was submitted to flash chromatography. Elution by light petroleum-ethyl acetate (90: 10) afforded the desired compound 41a in 80% yield: mp: 138.1-144.1 °C; 1H NMR (400 MHz) δ 2.25-2.75 (m, 4H), 3.15-3.25 (m, 1H), 5.40 (m, 1H), 6.25 (m, 1H), 6.40 (m, 1H), 7.25-7.50 (m, 5H). 13C NMR (100 MHz) δ: 12.3, 12.8, 13.0, 13.4, 17.1, 17.3, 18.5, 19.2, 20.1, 20.3, 21.1, 22.2, 25.7, 30.7, 30.9, 33.5, 37.8, 39.3, 40.9, 42.3, 44.0, 48.8, 52.6, 54.9, 60.1, 62.8, 63.9, 64.6, 64.7, 65.1, 70.2, 126.0, 127.6, 128.6, 128.9, 131.5, 135.0, 135.3, 146.4, 148.8, 148.9, 169.8.

Example 19

(22R,23R)-3p-Acetoxy-22,23-epoxyergosta-5,7-diene (42) and (22S,23S)-3p- Acetoxy-22,23-Epoxyergosta-5,7-diene (43). Anhydrous K2C03 (0.13 g, 0.93 mmol) was added to a solution of epoxide 41a (0.59 g, 0.93 mmol) in anhydrous DMF (50 mL). The resulting mixture was refluxed for 6 h, then cooled to room temperature and neutral alumina was added. The resulting mixture was filtered, and treated with water to yield a precipitate, which was then filtered in vacuo washing with water. The solid was submitted to mpc. Elution with light petroleum-ethyl acetate (90: 10) afforded pure samples of the desired compounds in 65% total yield; 42: mp 158.8-160.2 °C; 1H NMR (400 MHz) δ 0.62 (s, 3H), 2.05 (s, 3H), 2.46-2.48 (m, 2H), 2.60-2.62 (m, 1H), 4.71 (m, 1H), 5.40-5.41 (m, 1H), 5.57-5.58 (m, 1H). 13C NMR (100 MHz) 11.92, 13.70, 16.09, 16.29, 19.51, 20.43, 20.94, 21.43, 23.21, 26.83, 28.06, 31.10, 36.60, 37.04, 37.87, 39.00, 42.29, 43.20, 46.00, 53.94, 55.63, 60.37, 64.22, 72.74, 116.52, 120.19, 138.59, 141.05, 170.56; 43 : mp 133.5-135.2 °C; 1H NMR (400 MHz) δ 0.61 (s, 3H), 2.05 (s, 3H), 2.37-2.52 (m, 3H), 2.69 (d, 1H, J = 7.70 Hz), 4.71 (m, 1H), 5.40 (bs, 1H), 5.57 (bs, 1H). 13C NMR (100 MHz) 11.88, 12.56, 16.13, 17.14, 18.57, 20.24, 20.92, 21.43, 23.32, 27.80, 28.05, 31.00, 36.61, 37.05, 37.86, 38.92, 39.89, 42.50, 43.22, 45.95, 53.28, 53.97, 63.07, 63.83, 72.72, 116.53, 120.08, 138.78, 140.94, 170.57.

Example 20

(23R)-3p-(Tetrahydro-2H-pyran-2-yloxy)ergost-5,7-diene-23-ol (44) and (22R)-3p- (Tetrahydro-2H-pyran-2-yloxy)ergost-5,7-diene-22-ol (47). ΠΑ1Η4 (1.87 g, 49 mmol) was portion wise added to the solution of the epoxide 41b (2.83 g, 4.2 mmol) in anhydrous THF (110 mL). The resulting mixture was refluxed for 36 h under an argon atmosphere. After cooling, first EtOAc and then water were carefully added. The organic phase was separated and the water phase extracted with EtOAc (3x25 mL). The combined organic phases were washed with brine (30 mL), dried over Na2S04, filtered and the solvent removed in vacuo to give a residue which was submitted to mpc. Elution with light petroleum-ethyl acetate (95:5) gave a pure sample of 44 in 20.5% yield: mp 95.1-96.9 °C; 1H MR (400 MHz) δ 0.66 (s, 3H), 3.49-3.51 (m, 1H), 3.62-3.74 (m, 2H), 3.93-3.95 (m, 1H), 4.74-4.77 (m, 1H), 5.39 (s, 1H), 5.57 (s, 1H); 13C MR (100 MHz) 9.79, 11.82, 16.14, 18.40, 18.76, 19.78, 19.95, 20.97, 21.46, 22.94, 25.42, 28.20, 28.28, 29.50, 29.92, 31.13, 31.22, 33.05, 37.19, 37.34, 38.17, 38.46, 38.68, 39.17, 42.09, 42.95, 45.35, 46.12, 54.46, 56.45, 62.54, 62.77, 70.53, 74.55, 74.69, 96.60, 97.00, 116.31, 116.40, 119.34, 119.47, 139.89, 140.14, 140.78, 141.01. Further elution gave the inseparable mixture of (23S)-3P- tetrahydropyranyloxyergost-5,7-diene-23-ol (45) and (22S)-3P- tetrahydropyranyloxyergost-5,7-diene-22-ol (46) in 30% yield. Following elution afforded a pure sample of 47 in 21% yield: mp 180.2-181.5 °C; 1H NMR (400 MHz) δ 0.63 (s, 3H), 1.07 (d, 3H, J = 6.50 Hz), 3.47-3.50 (m, 1H), 3.61-3.65 (m, 2H), 3.76 (d, 1H, J = 10.66 Hz), 3.91-3.93 (m, 1H), 4.73-4.75 (m, 1H), 5.37 (s, 1H), 5.55 (s, 1H); 13C NMR (100 MHz) 10.54, 11.68, 11.76, 12.48, 15.57, 16.06, 16.18, 17.91, 19.84, 20.01, 20.67, 21.01, 21.74, 23.05, 23.15, 25.45, 27.40, 27.73, 28.23, 28.38, 29.57, 29.96, 31.17, 31.26, 34.59, 35.31, 35.73, 37.23, 37.37, 38.22, 38.50, 38.73, 39.12, 40.72, 42.97, 43.05, 43.20, 45.35, 46.11, 46.16, 52.75, 53.98, 54.34, 56.78, 62.61, 62.84, 71.61, 74.20, 74.60, 74.68, 96.67, 97.05, 116.33, 116.43, 116.52, 119.36, 119.48, 139.98, 140.07, 140.22, 140.31, 140.57, 140.81, 141.07.

Example 21

3p-(Tetrahydro-2H-pyran-2-yloxy)ergost-5,7-diene-23-one (48). A solution of DMSO (0.20 g, 2.51 mmol) in anhydrous CH2C12 (0.5 mL) was added to a solution of oxalyl chloride (0.17 g, 1.32 mmol) in anhydrous CH2C12 (1 mL), kept at -60 °C under an argon atmosphere. After the resulting mixture was stirred for 15 min at -60 °C, the solution of alcohol 44 (0.33 g, 0.66 mmol) in anhydrous CH2C12 (2 mL) was added. The mixture was stirred for 2 h at -55/60 °C before the addition of Et3N (0.51 g, 5.0 mmol). After the reaction mixture was allowed to reach room temperature, stirring was continued for 15 min, and then water (10 mL) was added. The reaction mixture was extracted with CH2C12 (3x5 mL), the combined organic layers were washed with brine (10 mL), dried over Na2S04, filtered and the solvent removed in vacuo, to give a residue which was submitted to flash chromatography. Elution with light petroleum- ethyl acetate (90: 10) furnished 48 in 65% yield: mp 135.9-136.3 °C; 1H NMR (400 MHz) δ 0.66 (s, 3H), 3.49 (m, 1H), 3.63 (m, 1H), 3.93 (m, 2H), 5.38 (s, 1H), 5.56 (s, 1H); 13C NMR (100 MHz) 11.82, 12.58, 16.23, 18.67, 20.07, 21.38, 22.98, 25.48, 28.20, 30.08, 31.30, 32.28, 37.28, 38.00, 39.09, 43.01, 46.16, 49.09, 52.80, 54.47, 55.66, 62.91, 74.70, 74.76, 97.13, 116.53, 119.45, 139.89, 140.91, 214.81.

Biology.

T0901317, GW4064 and 9-cis-retinoic acid were purchased from Sigma. Rosiglitazone was purchased from Cayman Chemical (Ann Arbor, MI).

Cell Culture and Co-transfection Assays. Human embryonic Kidney 293 cells (American Type Culture Collection) were cultured in Dulbecco's Modified Eagle's medium containing 10% of fetal bovine serum at 37°C in humidified atmosphere of 5% C02. HEK293 cells (4x104 cells per well) were transiently transfected in 48 well plate with the reporter plasmids pMH100X4-TK-luc (100 ng/well), Renilla (22 ng/well) together with 100 ng/well of pCMX-Gal4-RXR, pCMX-GaW-PPAR-γ, pCMX-Gal4-PXR, pFA-CMV-FXR pCMX-Gal4-LXR-a or pCMX-GaW-LXR-β plasmids using X-tremeGENE 9 DNA Transfection Reagent (Roche). Six hours after transfection, the cells were treated with the appropriate compound for 24 hours. Luciferase activities were analyzed by luciferase Dual Reporter Assay Systems (Promega) according to the manufacturer's protocol. GAL4-LXRs, GAL4-PPAR-y, GAL4-RXR and TK-MHClOO-luc plasmids were described in Villablanca et al.49 GAL4-PXR was a kind gift of Dr. Enrique Sainz (The Scripps Research Institute, La Jolla, USA). GAL4-FXR was a kind gift of Dr. Daniel Merk (Goethe-University Frankfurt am Main). The results obtained by luciferase assays and reported in Table 1 are from three to five independent experiments.

Quantitative Real-Time-PCR. U937 cell line was differentiated in foam macrophages with phorbol 12-myristate 13-acetate (PMA) 10 ng/ml (Sigma) for 72 hours at 37°C in 10 mm dish at the concentration of 3x106 cells in 10 ml RPMI 10% FBS. At day 3 nuclear receptor ligands were added for 6 hours. HepG2 cells were treated with the ligands as described by Quinet et al.22 Total RNA was purified by TRIZOL (Invitrogen, Carlsbad, CA, USA). Reverse transcription was performed incubating 2 μg of total RNA 1 hour at 42 °C with MLV-reverse transcriptase (Promega). Quantitative PCR was performed using Sybr Green Master Mix (Applied Biosystems) and real-time PCR (Viia 7 Real Time PCR System, Applied Biosystems). All PCR reactions were done in triplicate. The comparative Ct method was used to quantify transcripts that were normalized for human GAPDH. the following primer pairs were used:

GAPDH-F ACA TCA TCC CTG CCT CTA CTG

GAPDH-R ACC ACC TGG TGC TCA GTG TA

ABCAl-F CCA GGC CAG TAC GGA ATT C

ABCAl-R CCT CGC CAA ACC AGT AGG A

SREBP-lc-F GGC GGG CGC AGA TC

SREBP-lc-R TTG TTG ATA AGC TGA AGC ATG TCT

MCP-l-F AGA AGC TGT GAT CTT CAA GAC CAT T

MCP-l-R TGC TTG TCC AGG TGG TCC AT

FAS-F ACA GCG GGG AAT GGG TAC T

FAS-R GAC TGG TAC AAC GAG CGG AT

SCD1-F TTC AGA AAC ACA TGC TGA TCC TCA TAA TTC

SCD1-R ATT AAG CAC CAC AGC ATA TCG CAA GAA AGT

TNFa-F TCT TCT CGA ACC CCG AGT GA

TNFa-R CCT CTG ATG GCA CCA CCA G

Statistical analysis. Data are expressed as mean ± SEM and were analyzed for significance by ANOVA with Dunnet's multiple comparison tests. The analysis was performed with Prism software. Data in Table 1 are expressed as EC50 ± SD. In particular, the standard deviations were obtained by calculating the mean of the EC50 of each experiment (three to five independent experiments). The efficacy (%) of the compounds was calculated as the percentage of the compound effect, in terms of LXRa or β activation, versus 8 μΜ of 22R-HC ± SD. The analyses were performed with Prism software. X-ray Analysis. A single crystal of compound 23 was submitted to X-ray data collection on an Oxford-Diffraction Xcalibur Sapphire 3 diffractometer with a graphite monochromated Μο-Κα radiation (λ = 0.71073 A) at 293 K. The structure was solved by direct methods implemented in SHELXS program (version 2013/1).50 The refinement was carried out by full-matrix anisotropic least-squares on F2 for all reflections for non-H atoms by means of the SHELXL program (version 2013/4).50 Crystallographic data (excluding structure factors) of 23 have been deposited with the Cambridge Crystallographic Data Centre as supplementary publication no. CCDC 1526884. Copies of the data can be obtained, free of charge, on application to CCDC, 12 Union Road, Cambridge CB2 1EZ, UK

Pharmacological assays on murine Lewis lung carcinoma cells (LLC) in vitro and in vivo

We have performed a set of experiments to evaluate whether some LXR modulators, particularly PFM009 (13) and PFM018 (25) influence the growth of the murine Lewis lung carcinoma cells (LLC) in vitro and in vivo. The treatment of LLC tumor cells for 96 hours with 10 μΜ of the above-reported compounds was not able to alter LLC tumor growth in vitro (Figure 11). We tested by qPCR whether these compounds were able to induce the expression of the LXR target genes in LLC cells, and we observed that both PFM009 and PFM018 induced the expression of the LXR target gene Abcal in LLC tumor cells (Figure 12).

Then, we tested whether these compounds could influence LLC tumor growth in vivo. To do that, C57BL/6 mice were challenged with lxlO 6 LLC tumor cells subcutaneously. Six days later, tumor-bearing mice were treated with PFM009 or PFM018 (30 mg/Kg/day) for five consecutive days and then evaluated for tumor growth. In accordance with data obtained by qPCR experiments, we observed a significant control of LLC tumor growth when mice were treated with both PFM009 and PFM018 (Figure 13 A and 13B). While not willing to be bound to any particular theory, these experiments seem to indicate that PFM009 and PFM018, which are LXR and β selective agonists, respectively, control tumor growth possibly interfering with non-tumor microenvironmental cells, as these compounds do not block tumor cell proliferation in vitro (Figure 11). REFERENCES

(1) Poli, G.; Biasi, F.; Leonarduzzi, G. Redox biology oxysterols in the pathogenesis of major chronic diseases. Redox Biol. 2013, 1, 125-130.

(2) Bjorkhem, I.; Diczfalusy, U. Oxysterols: friends, foes, or just fellow passengers? Arterioscler. Thromb. Vase. Biol. 2002, 734-743.

(3) Bovenga, F.; Sabba, C; Moschetta, A. Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer. Cell Metab. 2015, 21, 517-526.

(4) Traversari, C; Sozzani, S.; Steffensen, K. R.; Russo, V. LXR-dependent and - independent effects of oxysterols on immunity and tumor growth. Eur. J. Immunol. 2014, 44, 1896-1903.

(5) Venteclef, N; Ferre, P. Liver X receptor: from metabolism to cancer. Biochem. J. 2014, 459, el-e3.

(6) De Boussac, H.; Alioui, A; Viennois, E.; Dufour, J.; Trousson, A.; Vega, A.; Guy, L.; Voile, D. H.; Lobaccaro, J.-M. A.; Baron, S. Oxysterol receptors and their therapeutic applications in cancer conditions. Expert Opin. Ther. Targets 2013, 17, 1029-1038.

(7) Janowsky, Bethany A.; Willy, P. J.; Rama Devi, T.; Falck, J. R.; Mangel sdorf, D. J. An oxysterol signalling pathway mediated by the nuclear receptor LXR. Nature 1996, 383, 728-731.

(8) Janowski, B. A.; Grogan, M. J.; Jones, S. A.; Wisely, G. B.; Kliewer, S. A.; Corey, E. J.; Mangelsdorf, D. J. Structural requirements of ligands for the oxysterol liver X receptors LXR a and LXR β. Proc. Natl. Acad. Sci. U. S. A. 1999, 96, 266- 271.

(9) Song, C; Kokontis, J. M.; Hiipakka, R. A.; Liao, S. Ubiquitous receptor : a receptor that modulates gene activation by retinoic acid and thyroid hormone receptors. Proc. Natl. Acad. Sci. USA 1994, 91, 10809-10813.

(10) Shinar, D. M.; Endo, N.; Rutledge, S. J.; Vogel, R.; Rodan, G. A.; Schmidt, A. NER, a new member of the gene family encoding the human steroid hormone nuclear receptor. Gene 1994, 147, 273-276.

(11) Apfel, R.; Benbrook, D.; Lernhardt, E.; Ortiz, M. A.; Salbert, G; Pfahl, A. M. A novel orphan receptor specific for a subset of thyroid hormone-responsive elements and its interaction with the retinoid / thyroid hormone receptor subfamily. Mol. Cell. Biol. 1994, 14, 7025-7035.

(12) Teboul, M; Enmark, E.; Li, Q.; Wirkstroem, A. C; Pelto-Huikko, M; Gustafsson, J. -A. OR-1, a member of the nuclear receptor superfamily that interacts with the 9-cis-retinoic acid receptor. Proc. Natl. Acad. Sci. U. S. A. 1995, 92, 2096- 2100.

(13) Willy, P. J.; Umesono, K.; Ong, E. S.; Evans, R. M.; Heyman, R. a; Mangel sdorf, D. J. LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev. 1995, 9, 1033-1045.

(14) Spann, N. J.; Glass, C. K. Sterols and oxysterols in immune cell function. Nat. Immunol. 2013, 14, 893-900.

(15) Gabbi, C; Warner, M.; Gustafsson, J.-A. Action mechanisms of liver X receptors. Biochem. Biophys. Res. Commun. 2014, 446, 647-650.

(16) Dong, X.-Y.; Tang, S.-Q.; Chen, J.-D. Dual functions of Insig proteins in cholesterol homeostasis. Lipids Health Dis. 2012, 11, 173.

(17) Olkkonen, V. M.; Zhou, Y.; Yan, D.; Vihervaara, T. Oxysterol-binding proteins-emerging roles in cell regulation. Eur. J. Lipid Sci. Technol. 2012, 114, 634- 643.

(18) Corman, A.; Deberardinis, A. M.; Hadden, M. K. Structure - activity relationships for side chain oxysterol agonists of the hedgehog signaling pathway.

ACS Med. Chem. Lett. 2012, 3, 828-833.

(19) Tice, C. M.; Noto, P. B.; Fan, K. Y.; Zhuang, L.; Lala, D. S.; Singh, S. B. The medicinal chemistry of liver X receptor (LXR) modulators. J. Med. Chem. 2014, 57, 7182-7205.

(20) Loren, J.; Huang, Z.; Laffitte, B. a; Molteni, V. Liver X Receptor Modulators: A review of recently patented compounds (2009 - 2012). Expert Opin. Ther. Pat. 2013, 23, 1317-1335.

(21) Spencer, T. a.; Li, D.; Russel, J. S.; Collins, J. L.; Bledsoe, R. K.; Consler, T. G; Moore, L. B.; Galardi, C M.; McKee, D. D.; Moore, J. T.; Watson, M. a.; Parks, D. J.; Lambert, M. H; Willson, T. M. Pharmacophore analysis of the nuclear oxysterol receptor LXRa. J. Med. Chem. 2001, 44, 886-897. (22) Quinet, E. M; Savio, D. a; Halpern, A. R.; Chen, L.; Miller, C. P.; Nambi, P. Gene- selective modulation by a synthetic oxysterol ligand of the liver X receptor. J. Lipid Res. 2004, 45, 1929-1942.

(23) Berrodin, T. J.; Shen, Q.; Quinet, E. M.; Yudt, M. R.; Freedman, L. P. Identification of 5a, 6a-epoxycholesterol as a novel modulator of liver X receptor activity. Mol. Pharmacol. 2010, 78, 1046-1058.

(24) Peng, D.; Hiipakka, R.; Dai, Q.; Guo, J.; Reardon, C; Getz, G. S.; Liao, S. Antiatherosclerotic effects of a novel synthetic tissue-selective steroidal liver X receptor agonist in low-density lipoprotein receptor-deficient mice. J. Pharmacol. Exp. Ther. 2008, 327, 332-342.

(25) Peng, D.; Hiipakka, R.; Xie, J. T.; Dai, Q.; Kokontis, J. M; Reardon, C; Getz, G. S.; Liao, S. A novel potent synthetic steroidal liver X receptor agonist lowers plasma cholesterol and triglycerides and reduces atherosclerosis in LDLR -/- mice. Br. J. Pharmacol. 201 1, 162, 1792-1804.

(26) Li, L.; Liu, J.; Zhu, L.; Cutler, S.; Hasegawa, H.; Shan, B.; Medina, J. C. Discovery and optimization of a novel series of liver X receptor-a Agonists. Bioorg. Med. Chem. Lett. 2006, 16, 1638-1642.

(27) Jones, P. J. H. Cholesterol-lowering effect of plant sterols. Curr. Atheroscler. Rep. 1999, 1, 230-235.

(28) Plat, J.; Mensink, R. P. Increased intestinal ABCAl expression contributes to the decrease in cholesterol absorption after plant stanol consumption. FASEB J. 2002, 16, 1248-1253.

(29) Kaneko, E.; Matsuda, M.; Yamada, Y.; Tachibana, Y.; Shimomura, I; Makishima, M. Induction of intestinal ATP -binding cassette transporters by a phytosterol-derived liver X receptor agonist. J. Biol. Chem. 2003, 278, 36091-36098.

(30) Chisholm, J. W.; Hong, J.; Mills, S. A.; Lawn, R. M. The LXR Ligand T0901317 induces severe lipogenesis in the db/db diabetic mouse. J. Lipid Res. 2003, 44, 2039-2048.

(31) Premalatha, R.; Srikumar, K.; Vijayalaksmi, D.; Kumar, G. N.; Mathur, P. P. 28-Homobrassinolide: a novel oxysterol transactivating LXR gene expression. Mol.

Biol. Rep. 2014, 41, 7447-7461. (32) Chen, Z.; Liu, J.; Fu, Z.; Ye, C; Zhang, R.; Song, Y.; Zhang, Y.; Li, H.; Ying, H.; Liu, H. 24(S)-Saringosterol from edible marine seaweed Sargassum Fusiforme is a novel selective LXRp agonist. J. Agric. Food Chem. 2014, 62, 6130-6137.

(33) Yang, C; Yu, L.; Li, W.; Xu, F.; Cohen, J. C; Hobbs, H. H. Disruption of cholesterol homeostasis by plant sterols. J. Clin. Invest. 2004, 114, 813-822.

(34) Foley, D.; O'Callaghan, Y.; O'Brien, N. M.; McCarthy, F. O.; Maguire, A. R. Synthesis and characterization of stigmasterol oxidation products. J. Agric. Food Chem. 2010, 58, 1165-1173.

(35) Misharin, A. Y.; Mehtiev, A. R.; Morozevich, G. E.; Tkachev, Y. V; Timofeev, V. P. Synthesis and cytotoxicity evaluation of 22,23 -oxygenated stigmastane derivatives. Bioorg. Med. Chem. 2008, 16, 1460-1473.

(36) Crump, D. R; Williams, D. H.; Pelc, B. (22 S)-Hydroxy vitamin D4. J. C. S. Perkin l 1973, 2731-2733.

(37) Tada, M.; Oikawa, A. Synthesis of 22,23 -epoxy vitamin D2 (22,23- Epoxyergocalciferol). J. C. S. Perkin I 1979, 1858-1861.

(38) Miyashita, N.; Yoshikoshi, A.; Grieco, P. A. Pyridinium p-toluenesulfonate. A mild and efficient catalyst for the tetrahydropyranylation of alcohols. J. Org. Chem. 1977, 42, 3772-3774.

(39) Zhang, R; He, H. P.; Di, Y. T.; Li, S. L.; Zuo, G. Y.; Zhang, Y.; Hao, X. J. Chemical constituents from Aphanamixis Grandifolia. Chem. Nat. Compd. 2013, 49,

100-104.

(40) Wei, X.; Shu, P.; Liu, T.; Xiang, M.; Zhang, J.; Xue, Y.; Luo, Z.; Yao, G; Zhang, Y. Steroids and phenylpropanoids with immunomodulatory activities from the stem barks of Cinnamomum Wilsonii. Chinese J. Org. Chem. 2013, 33, 1273.

(41) Brynjolffssen, J.; Hands, D.; Midgley, J. M.; Whalley, W. B. Unsaturated steroids. Part i. Synthesis of 22,23 -dihydroergosterol. J. C. S. Perkin l 1976, 826-828.

(42) Barton, D. H. R.; Poyster, J. P.; Sammes, P. G.; Hursthouse, M. B.; Neidle, S.

Stereospecific and regiospecific addition to an isolated, acyclic (steroidal) olefinic bond. Chem. Commun. 1971, 715-716.

(43) Hong, C; Tontonoz, P. Liver X receptors in lipid metabolism: opportunities for drug discovery. Nat. Rev. Drug Discovery 2014, 13, 433-444. (44) Repa, J. J.; Turley, S. D.; Lobaccaro, J. a; Medina, J.; Li, L.; Lustig, K.; Shan, B.; Heyman, R. a; Dietschy, J. M.; Mangelsdorf, D. J. Regulation of absorption and ABC 1 -mediated efflux of cholesterol by RXR heterodimers. Science 2000, 289, 1524- 1529.

(45) Venkateswaran, A.; Laffitte, B. A.; Joseph, S. B.; Mak, P. A.; Wilpitz, D. C; Edwards, P. A.; Tontonoz, P. Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXRa. Proc. Natl. Acad. Sci. U. S. A. 2000, 97, 12097-12102. (46) Glass, C. K.; Saijo, K. Nuclear receptor transrepression pathways that regulate inflammation in macrophages and T cells. Nat. Rev. Immunol. 2010, 10, 365-376. (47) Ito, A.; Hong, C; Rong, X.; Zhu, X.; Tarling, E. J.; Hedde, P. N.; Gratton, E.; Parks, J.; Tontonoz, P. LXRs link metabolism to inflammation through Abcal- dependent regulation of membrane composition and TLR signaling. Elife 2015, 4, 1- 23.

(48) Gonzalez Sierra, M.; Bustos, D. A.; Zudenigo, M. E.; Riiveda, E. A. Configurational assignment of epimeric 22,23 -epoxides of steroids by Carbon- 13

NMR Spectroscopy. Tetrahedron 1986, 42, 755-758.

(49) Villablanca, R. J.; Raccosta, L.; Zhou, D.; Fontana, R.; Maggioni, D.; Negro, A.; Sanvito, F.; Ponzoni, M.; Valentinis, B.; Bregni, M.; Prinetti, A.; Steffensen, K. R.; Sonnino, S.; Gustafsson, J. -A.; Doglioni, C; Bordignon, C; Traversari, C; Russo, V. Tumor-mediated liver X receptor-a activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor rsponses. Nat. Med. 2010, 16, 98-105.

(50) Sheldrick, G. M. A short history of SHELX. Acta Cryst. 2008, A64, 112-122.