WO2006117684A1 - Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds - Google Patents

Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds Download PDF

Info

Publication number
WO2006117684A1
WO2006117684A1 PCT/IB2006/001541 IB2006001541W WO2006117684A1 WO 2006117684 A1 WO2006117684 A1 WO 2006117684A1 IB 2006001541 W IB2006001541 W IB 2006001541W WO 2006117684 A1 WO2006117684 A1 WO 2006117684A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydroxy
vitamin
pent
trifluoro
compound
Prior art date
Application number
PCT/IB2006/001541
Other languages
French (fr)
Inventor
Luciano Adorini
Milan R. Uskokovic
Michèle RESCHE-RIGNON
Original Assignee
Bioxell S.P.A.
Galapagos S.A.S.U.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bioxell S.P.A., Galapagos S.A.S.U. filed Critical Bioxell S.P.A.
Priority to CA002602464A priority Critical patent/CA2602464A1/en
Priority to EP06765498A priority patent/EP1898924A1/en
Priority to US11/886,922 priority patent/US20090298799A1/en
Priority to TW095110651A priority patent/TW200736207A/en
Publication of WO2006117684A1 publication Critical patent/WO2006117684A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/59Compounds containing 9, 10- seco- cyclopenta[a]hydrophenanthrene ring systems
    • A61K31/5939,10-Secocholestane derivatives, e.g. cholecalciferol, i.e. vitamin D3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones
    • A61P5/20Drugs for disorders of the endocrine system of the parathyroid hormones for decreasing, blocking or antagonising the activity of PTH

Definitions

  • vitamin D cholesterol calcium and phosphorous homeostasis
  • the operation of the vitamin D endocrine system depends on the following: first, on the presence of cytochrome P450 enzymes in the liver (Bergman, T. and Postlind, H. (1991) Biochem. J. 276:427-432; Ohyama, Y and Okuda, K. (1991) J. Biol. Chem. 266:8690-8695) and kidney (Henry, HX. and Norman, A.W. (1974) J. Biol. Chem. 249:7529-7535; Gray, R.W. and Ghazarian, IG. (1989) Biochem. J.
  • Vitamin D 3 and its hormonally active forms are well-known regulators of calcium and phosphorous homeostasis. These compounds are known to stimulate, at least one of, intestinal absorption of calcium and phosphate, mobilization of bone mineral, and retention of calcium in the kidneys. Furthermore, the discovery of the presence of specific vitamin D receptors in more than 30 tissues has led to the identification of vitamin D 3 as a pluripotent regulator outside its classical role in calcium/bone homeostasis.
  • vitamin D 3 hormone and active metabolites have been found to be capable of regulating cell proliferation and differentiation of both normal and malignant cells (Reichcl, H. et al. (1989) Ann. Rev. Med. 40: 71-78). Given the activities of vitamin D 3 and its metabolites, much attention has focused on the development of synthetic analogs of these compounds.
  • vitamin D .- ⁇ - indications/ applications of vitamin D compounds. Therefore, structural analogs of vitamin D having improved therapeutic activity, particularly for the treatment of osteoporosis and secondary hyperparathyroidism and/or reduced undesirable side effects are needed.
  • the invention provides novel vitamin D 3 compounds having improved therapeutic activity for the treatment of osteoporosis and secondary hyperparathyroidism and/or reduced undesirable side effects useful for the treatment of osteoporosis and secondary.
  • the invention provides a method for treating osteoporosis in a subject comprising administering to a subject in need thereof a therapeutically effective amount of a vitamin D 3 compound selected from the group consisting of (20S)- 1 ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluororriethyl-pent-2-ynyl)- cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-l ⁇ -Fluoro-25-hydroxy-20- (5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (6), thereby treating the subject for osteoporosis.
  • a vitamin D 3 compound selected from the group consisting of (20S)- 1 ,25-Di
  • the invention provides a method for treating a subject for secondary hyperparathyroidism comprising administering to a subject in need thereof a therapeurically effective amount of a vitamin D 3 compound selected from the group consisting of (20S)-l,25-Dihydroxy-20-((2Z)-5 I 5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl)cholecalciferol (2); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5- trifluoro-4-hyd ⁇ oxy-4-trifluoromcthyl-pent-2-enyrj-cholecalciferol (3); (20R)-1, 25-
  • the invention provides a pharmaceutical composition for use in the treatment of osteroporosis, comprising a therapeutically effective amount of a vitamin D 3 compound selected from the group consisting of (20S)-1,25-Dihydroxy-20- (S ⁇ .S-trifluoro ⁇ -hydroxy ⁇ -trifluoromethyl-pent ⁇ -ynylJ-cholecalcifero ⁇ l); (20S)- l 3 25-Dihydroxy-20-[(2E)-5 1 5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (3); and (20S)-l ⁇ -Fluoro-25-hydroxy-20-(5,5,5-trifiuoro-4-hydroxy-4- trifl ⁇ oromcthyl-pcn1.-2-ynyl)-cholecalciferol (6), and a pharmaceutically acceptable diluent or carrier.
  • a vitamin D 3 compound selected from the group consisting of (20S)-1,25-
  • the invention provides a pharmaceutical composition for use in the treatment of secondary hyperparathyroidism comprising a therapeutically effective amount of a vitamin D3 compound selected from the group consisting of (20S)- I,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- enyl)cholecalcifcrol (2); (20S)-l,25 ⁇ Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl]-cholecalciferol (3); (20R)-l,25-Dihydroxy-20-(5,5,5- trifluoro-4-hydroxy-4-trifiuoromethyl-pent-2-ynyl)-cholecalciferol (4); and (20R)- 1,25- Dihydroxy-20-[(2E)-5,5 5 5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl)cholecalc
  • Another aspect of the invention provides a packaged formulation for use in the treatment of osteoporosis, comprising a pharmaceutical composition comprising a vitamin D 3 compound selected from the the group consisting of (20S)-1,25-Dihydroxy- 20-(5 J 5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (1); (20S)- l,25-Dihydroxy-20-[(2E)-5 ) 5,5-trifluoro-4-hydroxy-4-trifluoromethyI-pent-2-enyl]- cholecalciferol (3); and (20S)-l ⁇ -Fluoro-25-hydroxy-20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-ynyl)-cholecalciferol (6); and instructions for use in the treatment of osteoporosis.
  • a vitamin D 3 compound selected from the group consisting of (20S)-1,25-Dihydroxy- 20
  • Yet another aspect of the invention provides a packaged formulation for use in the treatment of secondary hyperparathyroidism, comprising a pharmaceutical composition comprising a vitamin D 3 compound selected from the group consisting of (20S)-l,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl- ⁇ ent-2- enyl)cholecalciferol (2); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- mfluoromethyl-pent-2-enyl]-cholecalciferol (3); (20R)-l,25-Dihydroxy-20-(5,5,5- trifluoro-4-hydroxy-4-t ⁇ ifluoromethyl-pent-2-ynyl)-choleca]ciferol (4); and (2QR)-1, 25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2
  • Figure 1 shows tibia proximal metaphysic bone volume (uCT) measurements in 3 month old OVX rats.
  • Figure 2 shows lumbar spine BMD (DEXA) measurements in 3 month old OVX rats.
  • Figure 3 shows urinary calcium levels in 3 month old OVX rats.
  • Figure 4 shows bone volume in 3 month old OVX rats using (1).
  • Figure 5 shows a reevaluation of 3 month old OVX rats for tibia proximal metaphysic bone volume ( ⁇ CT).
  • Figure 6A shows serum Ca levels in 3 month old rats.
  • Figure 6B shows urinary
  • Figure 7 shows travecular bone volume ( ⁇ CT) measurements in 6 month old OVX rats.
  • Figure 8 shows urinary calcium levels in 6 month old OVX rats.
  • Figure 9 shows BMD (DEXA) measurements in 6 month old OVX rats.
  • Figure 1OA shows serum calcium levels.
  • Figure 1OB shows urinary calcium levels.
  • Figure 11 shows BMD (DEXA) measurements in 6 month old OVX rats.
  • Figure 12A shows parathyroid hormone (PTH) levels in rats with moderate renal failure.
  • Figure 12B shows serum Ca levels in rats with moderate renal failure.
  • Figures 13A and 13B show a model of safety parameters, measuring serum and serum Ca levels.
  • Figure 14A shows a decrease in PTH levels in rats with severe chronic renal failure.
  • Figure 14B shows the serum calcium levels in rats with severe chronic renal failure.
  • Figures 15A and 15B show measurements of serum and serum Ca to determine safety profiles in rats with severe renal failure.
  • Figure 16A shows the trabecular bone volume measurements in uremic rats.
  • Figure 16B shows bine m ⁇ narl density (pQCT) measurements in uremic rats.
  • Figures 17A, 17B, and 17C show tibia histomorphometry analysis, measuring bone formation rate, osteoblast surface, and osteoclast number in uremic rats with moderate renal failure.
  • Figure 18A is a picture of a rat tibia using optical microscopy (x50) of a normal trabeculae.
  • Figure 18B is a picture of a rat tibia using optical microscopy (x50) of osteoid thickening.
  • Figure 18C is a picture of a rat tibia using optical microscopy (x50) of peritrabecular fibrosis.
  • Figure 19 shows bone mineral density (DEXA) in uremic rats.
  • Figure 20 is a picture of a rat femur cortical porosity using fluorescence microscopy (x8), showing normal porosity, mild porosity, medium porosity, and marked porosity.
  • Figure 21 is a picture of a cross section of a rat aorta using Von Kossa staining (xlOO), showing a control, moderate aorta calcification, and severe aortic calcification.
  • xlOO Von Kossa staining
  • administration includes routes of introducing the vitamin D 3 compound(s) to a subject to perform their intended function.
  • routes of administration include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecal), oral, inhalation, rectal and transdermal.
  • the pharmaceutical preparations are, of course, given by forms suitable for each administration route. For example, these preparations are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc.
  • the vitamin D3 compound can be coated with or disposed in a selected material to protect it from natural conditions which may detrimentally effect its ability to perform its intended function.
  • the vitamin D 3 compound can be administered alone, or in conjunction with either another agent as described above or with a pharmaceutically-acceptable carrier, or both.
  • the vitamin D 3 compound can be administered prior to the administration of the other agent, simultaneously with the agent, or after the administration of the agent.
  • the vitamin D 3 compound can also be administered in a proform which is converted into its active metabolite, or more active metabolite in vivo.
  • alkyl refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • alkyl further includes alkyl groups, which can further include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone, e.g., oxygen, nitrogen, sulfur or phosphorous atoms.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chain, C 3 -C 30 for branched chain), preferably 26 or fewer, and more preferably 20 or fewer.
  • preferred cycloalkyis have from 3-10 carbon atoms in their ring structure, and more preferably have 3, 4, 5, 6 or 7 carbons in the ring structure.
  • alkyl as used throughout the specification and claims is intended to include both “unsubstituted alkyls” and “substituted alkyls,” the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alky lth iocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonate, sulfamoyl, sulfonamido, nitro, triflu
  • lower alky as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six, and most preferably from one to four carbon atoms in its backbone structure, which may be straight or branched-chain.
  • lower alkyl groups include methyl, ethyl, n-propyl, i-propyl, tert-butyl, hexyl, heptyl, octyl and so forth.
  • the term "lower alkyl” includes a straight chain alkyl having 4 or fewer carbon atoms in its backbone, e.g., C 1 -C 4 alkyl.
  • alkoxyalkyl refers to alkyl groups, as described above, which further include oxygen, nitrogen or sulfur atoms replacing one or more carbons of the hydrocarbon backbone, e.g., oxygen, nitrogen or sulfur atoms.
  • alkenyl and alkynyl refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond, respectively.
  • the invention contemplates cyano and propargyl groups.
  • aryl refers to the radical of aryl groups, including 5- and 6-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, benzoxazole, benzothiazolc, triazole, tetrazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • Aryl groups also include polycyclic fused aromatic groups such as naphthyl, quinolyl, indolyl, and the like.
  • aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles," “heteroaryls” or “heteroaromatics.”
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, s
  • vitamin D 3 includes all activities elicited by vitamin D 3 compounds in a responsive cell. It includes genomic and n ⁇ n-genomic activities elicited by these compounds (Gniadecki R. and Calverley MJ. (1998) Pharmacology & Toxicology 82: 173-176; Bouillon, R. et al. (1995) Endocrinology Reviews 16(2):206-207; Norman A.W. et al. (1992) J. Steroid Biochem MoI Biol 41 :231-240; Baran D.T. et al. (1991) /. Bone Miner Res. 6:1269-1275; Caffrey J.M. and Farach-Carson M.C. (1989) J. Biol. Chem. 264:20265-20274; Nemere 1 et al. (1984) Endocrinology 1 15:1476-1483).
  • bone metabolism includes direct or indirect effects in the formation or degeneration of bone structures, e.g., bone formation, bone resorption, etc., which may ultimately affect the concentrations in serum of calcium and phosphate. This term is also intended to include effects of compounds of the invention in bone cells, e.g., osteoclasts and osteoblasts, that may in turn result in bone formation and degeneration.
  • calcium and phosphate homeostasis refers to the careful balance of calcium and phosphate concentrations, intraccllularly and extracellularly, triggered by fluctuations in the calcium and phosphate concentration in a cell, a tissue, an organ or a system.
  • deuteroalkyl refers to alky] groups in which one or more of the of the hydrogens has been replaced with deuterium.
  • an effective amount includes an amount effective, at dosages and for periods of time necessary, to achieve the desired result, e.g., sufficient treat a vitamin Dj associated state or to modulate ILT3 expression in a cell.
  • An effective amount of vitamin D 3 compound may vary according to factors such as the disease state, age, and weight of the subject, and the ability of the vitamin Dj compound to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response.
  • An effective amount is also one in which any toxic or detrimental effects (e.g., side effects) of compound are outweighed by the therapeutically beneficial effects.
  • a therapeutically effective amount of vitamin D 3 compound may range from about 0.001 to 30 ⁇ g/kg body weight, preferably about 0.01 to 25 ⁇ g/kg body weight, more preferably about 0.1 to 20 ⁇ g/kg body weight, and even more preferably about 1 to 10 ⁇ g/kg, 2 to 9 ⁇ g/kg, 3 to 8 ⁇ g/kg, 4 to 7 ⁇ g/kg, or 5 to 6 ⁇ g/kg body weight.
  • the skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present.
  • treatment of a subject with a therapeutically effective amount of a vitamin D 3 compound can include a single treatment or, preferably, can include a series of treatments.
  • a subject is treated with a vitamin D 3 compound in the range of between about 0.1 to 20 ⁇ g/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.
  • the effective dosage of a vitamin D3 compound used for treatment may increase or decrease over the course of a particular treatment.
  • the term “enan Homers” refers to two stereoisomers of a compound which are non-superimposable mirror images of one another. An equimolar mixture of two enantiomers is called a “racemic mixture” or a “racemate.”
  • the language “Gemini vitamin D 3 compounds” is intended to include vitamin D 3 compounds and analogs thereof having bis C20 side chains. Vitamin D 3 compounds are characterized by an "A" ring (monocycle) which is connected to a "B" ring (bicycle) which is connected to a side chain at carbon C20 of the side chain.
  • the Gemini compounds of the invention have two side chains and are, therefore, conspicuously distinguishable from vitamin D3 compounds having a single side chain.
  • Candidate A and B rings for the Gemini compounds of the invention arc disclosed in U.S. Patent Nos. 6,559,138, 6,329,538 , 6,331,642 , 6,452,028 , 6,492,353, 6,040,461 , 6,030,963, 5,939,408, 5,872,113, 5,840,718, 5,612,328, 5,512,554, 5,451,574, 5,428,029, 5,145,846, and 4,225,525.
  • Examples of Gemini compounds in accordance with the invention are disclosed in U.S. Patent No. 6,030,962.
  • halogen designates -F, -Cl, -Br or -I.
  • haioalkyl is intended to include alkyl groups as defined above that are mono-, di- or polysubstituted by halogen, e.g., fluoromethyl and trifluoromethyl.
  • halogen e.g., fluoromethyl and trifluoromethyl.
  • hydroxy means -OH.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus.
  • hormone secretion is art-recognized to mean maintenance of static, or constant, conditions in an internal environment.
  • hormone secretion is art-recognized and includes activities of vitamin D 3 compounds that control the transcription and processing responsible for secretion of a given hormone e.g,, a parathyroid hormone (PTH) of a vitamin D3 responsive cell (Bouillon, R. et al. (1995) Endocrine Reviews 16(2):235-237).
  • PTH parathyroid hormone
  • hypercalcemia or “hypercalcemic activity” is intended to have its accepted clinical meaning, namely, increases in calcium serum levels that are manifested in a subject by the following side effects, depression of central and peripheral nervous system, muscular weakness, constipation, abdominal pain > lack of appetite and, depressed relaxation of the heart during diastole.
  • Symptomatic manifestations of hypercalcemia are triggered by a stimulation of at least one of the following activities, intestinal calcium transport, bone calcium metabolism and osteocalcin synthesis (reviewed in Bouliion, R. et al. (1995) Endocrinology Reviews 16(2): 200-257).
  • improved biological properties refers to any activity inherent in a compound of the invention that enhances its effectiveness in vivo. In a preferred embodiment, this term refers to any qualitative or quantitative improved therapeutic property of a vitamin D 3 compound, such as reduced toxicity, e.g. , reduced hypercalcemic activity.
  • isomers or “stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • the term “modulate” refers to increases or decreases in the activity of a cell in response to exposure to a compound of the invention, e.g., the inhibition of proliferation and/or induction of differentiation of at least a sub-population of cells in an animal such that a desired end result is achieved, e.g., a therapeutic result.
  • this phrase is intended to include hyperactive conditions that result in pathological disorders.
  • non-genomic vitamin D 3 activities include cellular (e.g., calcium transport across a tissue) and subcellular activities (e.g., membrane calcium transport opening of voltage-gated calcium channels, changes in intracellular second messengers) elicited by vitamin D 3 compounds in a responsive cell.
  • Electrophysiological and biochemical techniques for detecting these activities are known in the art.
  • An example of a particular well-studied non-genomic activity is the rapid hormonal stimulation of intestinal calcium mobilization, termed "transcaltachia" (Nemere I. et al. (1984) Endocrinology 115: 1476-1483; Lieberherr M. et al (1989) J. Biol Chem. 264:20403- 20406; WaIi R.K. et al. (1992) Endocrinology 131 : 1125-1133; WaIi R.K. et al. (1992) Am. J. Physiol. 262:G945-G953; WaIi R.K. et al (199O) J.
  • obtaining as in “obtaining a vitamin D 3 compound” is intended to include purchasing, synthesizing or otherwise acquiring the compound.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • polycyclyl or “polycycHc radical” refer to the radical of two or more cyclic rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Rings that are joined through non-adjacent atoms are termed "bridged" rings.
  • Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, hydroxy!, alkylcarbonyloxy, aryJcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbo ⁇ yl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, aryiamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfam
  • prodrug includes compounds with moieties which can be metabolized in vivo. Generally, the prodrugs are metabolized in vivo by esterases or by other mechanisms to active drugs. Examples of prodrugs and their uses are well known in the art (See, e.g., Be ⁇ ge et al. (1977) "Pharmaceutical Salts", J. Pharm. Set. 66:3-19).
  • the prodrugs can be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form or hydroxyl with a suitable esterifying agent. Hydroxyl groups can be converted into esters via treatment with a carboxylic acid.
  • prodrug moieties include substituted and unsubstiruted, branch or unbranched lower alkyl ester moieties, (e.g. , propionoic acid esters), lower alkenyl esters, di-iower alkyl-amino lower-alkyl esters (e.g., dimethylaminoethyi ester), acylamino lower alkyl esters (e.g., acetyloxymethy] ester), acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters (phenyl ester), aryl-lower alkyl esters (e.g., benzyl ester), substituted (e.g., with methyl, haJo, or methoxy substituents) aryl and aryl-lower alkyl esters, amides, lower-alkyl amides, di- lower alkyl amides, and hydroxy amides
  • reduced toxicity is intended to include a reduction in any undesired side effect elicited by a vitamin D 3 compound when administered in vivo, e.g., a reduction in the hypercalcemic activity.
  • the term "secosteroid" is art-recognized and includes compounds in which one of the cyclopentanoperhydro- phenanthrene rings of the steroid ring structure is broken. l ⁇ ,25(OH) 2 D 3 and analogs thereof are hormonally active secosteroids. In the case of vitamin Oy, the 9-10 carbon-carbon bond of the B-ring is broken, generating a seco-B- steroid.
  • the official IUPAC name for vitamin D 3 is 9,10-secocholesta-5,7,iO(19)-trien- 3B-ol.
  • a 6-s-trans confo ⁇ ner of 1 ⁇ ,25(OH)2D 3 is illustrated herein having all carbon atoms numbered using standard steroid notation.
  • a dotted line ( — ) indicating a substituent which is in the ⁇ -orientation ⁇ i.e. , above the plane of the ring
  • a wedged solid line ( ⁇ ) indicating a substituent which is in the ⁇ -orientation ⁇ i.e. , below the plane of the molecule
  • a wavy line ( ' ⁇ n ⁇ ' v ⁇ - ) indicating that a substituent may be either above or below the plane of the ring.
  • ring A it should be understood that the stereochemical convention in the vitamin D field is opposite from the general chemical field, whexein a dotted line indicates a substituent on Ring A which is in an a- orientation (i.e. , below the plane of the molecule), and a wedged solid line indicates a substituent on ring A which is in the ⁇ -ori ⁇ ntation (i.e. , above the plane of the ring).
  • the A ring of the hormone l ⁇ ,25(OH) 2 D 3 contains two asymmetric centers at carbons 1 and 3, each one containing a hydroxyl group in well-characterized configurations, namely the l ⁇ - and 3 ⁇ - hydroxyl groups.
  • carbons 1 and 3 of the A ring are said to be "chiral carbons" or "carbon centers”.
  • the indication of stereochemistry across a carbon-carbon double bond is also opposite from the general chemical field in that "Z” refers to what is often referred to as a “cis” (same side) conformation whereas "E” refers to what is often referred to as a "trans” (opposite side) conformation.
  • the A ring of the hormone l-alpha,25(O HThDs contains two asymmetric centers at carbons 1 and 3, each one containing a hydroxyl group in well-characterized configurations, namely the 1- alpha- and 3-beta- hydroxyl groups.
  • carbons 1 and 3 of the A ring are said to be “chiral carbons” or “chiral carbon centers.” Regardless, both configurations, cis/trans and/or ZfB are encompassed by the compounds of the present invention.
  • the tenns "d" and "1" configuration are as defined by the IUPAC Recommendations. As to the use of the terms, diastereomer, racemate, epimer and enantiomer, these will be used in their normal context to describe the stereochemistry of preparations.
  • subject includes organisms which are capable of suffering from a vitamin D 3 associated state or who could otherwise benefit from the administration of a vitamin D 3 compound of the invention, such as human and non-human animals.
  • Preferred human animals include human patients suffering from or prone to suffering from a vitamin D 3 associated state, as described herein.
  • non-human animals of the invention includes all vertebrates, e.g., , mammals, e.g., rodents, e.g., mice, and non-mammals, such as non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc.
  • siilfhydryl or "thiol” means -SH.
  • systemic administration means the administration of a vitamin D 3 com ⁇ ound(s), drug or other material, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • VDR vitamin D response element
  • VDRE refers to DNA sequences composed of half-sites arranged as direct repeats. It is known in the art that type II receptors do not bind to their respective binding site as homodimers but require an auxiliary factor, RXR (e.g. RXR ⁇ , RXR ⁇ , RXR ⁇ ) for high affinity binding Yu et al. (1991) Cell 67:1251-1266; Bugge et al
  • vitamin D 3 associated state is a state which can be prevented, treated or otherwise ameliorated by administration of one or more compounds of the invention.
  • Vitamin D 3 associated states include ILT3 -associated disorders, disorders characterized by an aberrant activity of a vitamin D 3 -responsive cell, disorders characterized by a deregulation of calcium and phosphate metabolism, and other disorders or states described herein.
  • vitamin D 3 -responsive cell includes any cell which is is capable of responding to a vitamin D 3 compound described herein, or is associated with disorders involving an aberrant activity of hyperproliferative skin cells, parathyroid cells, neoplastic cells, immune cells, and bone cells. These cells can respond to vitamin D% activation by triggering genomic and/or non-genomic responses that ultimately result in the modulation of cell proliferation, differentiation survival, and/or other cellular activities such as hormone secretion. In a preferred embodiment, the ultimate responses of a cell are inhibition of cell proliferation and/or induction of differentiation-specific genes.
  • Exemplary vitamin D 3 responsive cells include immune cells, bone cells, neuronal cells, endocrine cells, neoplastic cells, epidermal cells, endodermal cells, smooth muscle cells, among others.
  • terms "d” and "1" configuration are as defined by the IUPAC Recommendations.
  • diastereomer, racemate, epimer and enantiomer will be used in their normal context to describe the stereochemistry of preparations.
  • the invention provides for the use of vitamin D compounds to treat osteoporosis and secondary hyperparathyroidism.
  • Preferred compounds for use in the methods of the invention include the following compounds:
  • especially preferred compounds include (20S)- 1,25- Dihydroxy ⁇ O ⁇ S.S.S-trifluoro ⁇ -hydroxy ⁇ -trifluoroinethyl-pent ⁇ -yny ⁇ -choIecalciferol (1) and (20S)-1 ,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl)choleca]ciferol (2).
  • the structures of some of the compounds of the invention include asymmetric carbon atoms. Accordingly, the isomers arising from such asymmetry (e.g., all enantiomers and diastereomers) are included within the scope of this invention, unless indicated otherwise. Such isomers can be obtained in substantially pure form by classical separation techniques and/or by stereochemical ⁇ controlled synthesis.
  • Naturally occurring or synthetic isomers can be separated in several ways known in the art. Methods for separating a racemic mixture of two enantiomers include chromatography using a chiral stationary phase (see, e.g., "Chiral Liquid Chromatography,” WJ. Lough, Ed. Chapman and Hall, New York (1989)). W 2
  • Enantiomers can also be separated by classical resolution techniques. For example, formation of diastereomeric salts and fractional crystallization can be used to separate enantiomers.
  • the diastereomeric salts can be formed by addition of enantiomerically pure chiral bases such as brucine, 5 quinine, ephedrine, strychnine, and the like.
  • diastereomeric esters can be formed with enantiomerically pure chiral alcohols such as menthol, followed by separation of the diastereomeric esters and hydrolysis to yield the free, enantiomerically enriched carboxylic acid.
  • the 20-mehtyl Gemini vitamin D 3 compounds of the invention can provide a less toxic alternative to current methods of treatment for osteoporosis and secondary hyperparathyroidism.
  • the invention provides a method for treating osteoporosis in a subject comprising administering to a subject in need thereof a therapeurically effective
  • Preferred compounds for this aspect of the invention include (20S)- 1 ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (t); (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifiuoromethyl-pent-2 ⁇ enyl]-cholecalcifcrol (3); and (20S)-l ⁇ -Fluoro-25-hydroxy-20-
  • An especially preferred compound of the invention is (20S)-l,25-Dihydroxy-20-(5,5,5- trifluoro ⁇ -hydroxy ⁇ -trifluoromethyl-pent ⁇ -ynyO-cholecalciferol (1).
  • the method further comprises identifying a subject as being in need of treatment for osteoporosis.
  • the method further comprises obtaining the vitamin D3 compound.
  • Another aspect of the invention provides a method for treating a subject for secondary hyperparathyroidism comprising administering to a subject in need thereof a therapeurically effective amount of a 20-methyl Gemini vitamin D 3 compound of the invention, thereby treating the subject for secondary hyperparathyroidism.
  • Preferred compounds for this aspect of the invention include (20S)-1,25-Dihydroxy-20- ((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl)cholecalciferol (2); (20S)- l,25-Dihydroxy-20-[(2E)-5 5 5 J 5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (3); (20R)-1 ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-perit-2-ynyl)-cholecalciferol (4); and (20R)-1,25-Dihydroxy-20-[(2E)- 5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (5).
  • An especially preferred compound of the invention is is (20S)-1 ,25-Dihydroxy-20-((2Z)- 5,5,5-trifJuoro-4-hydroxy-4-triiluorornethyI-pent-2-enyl)cholecalciferol (2).
  • the method further comprises identifying a subject as being in need of treatment for secondary hyperparathyroidism. In another embodiment the method further comprises obtaining the vitamin D 3 compound.
  • the subject is a mammal. In preferred embodiments, the subject is human.
  • the vitamin D 3 compound is administered to the subject using a pharmaceutically-acceptable formulation.
  • the vitamin D 3 compound is advantageously administered in combination with a pharmaceutically acceptable diluent or carrier.
  • the pharmaceutically-acceptable formulation provides sustained delivery of the vitamin Dj compound to a subject for at least four weeks after the pharmaceutically-acceptable formulation is administered to the subject.
  • the vitamin D 5 compound is administered orally, intravenously, topically, or parentcrally.
  • dosages may vary depending on the particular indication, route of administration and subject, the 20- methyl Gemini vitamin D 3 compounds of the invention are administered at a concentration of about 0.001 ⁇ g to about 100 ⁇ g/kg of body weight. In certain embodiments, the 20-methyl Gemini vitamin D 3 compounds of the invention are administered at a concentration of about 5 ⁇ g/kg of body weight.
  • Another aspect of the invention provides a pharmaceutical composition for use in the treatment of osteroporosis, comprising a therapeutically effective amount of a 20- methyl Gemini vitamin D 3 compound of the invention and a pharmaceutically acceptable diluent or carrier.
  • Preferred compounds of this aspect of the invention include (20S)-l,25-Dihydroxy-20-(5,5,S-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trif!uoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-l ⁇ -Fluoro-25-hydroxy ⁇ 20- (5,5,5-trifluoro-4-riydiOxy-4-trifiuoromethyl-pent-2-ynyl)-cholecalciferol (6).
  • an especially preferred compound of the invention is (20S)-l,25-Dihydroxy-20-(5,5,5- trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (I).
  • the invention provides a pharmaceutical composition for use in the treatment of secondary hyperparathyroidism comprising a therapeutically effective amount of a 20-methyl Gemini vitamin D 3 compound of the invention and a pharmaceutically acceptable diluent or carrier.
  • Preferred compounds of this aspect of the invention include (20S) ⁇ l,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl)choIecalciferol (2); (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5- trifluoro-4-hydroxy-4-trifl ⁇ oromethyl-pent-2-enyl]-cholecalciferoJ (3); (20R)- 1 ,25- Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (4); and (20R)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-tri fluoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl]-cholecalciferol (5).
  • a particularly preferred compound is (20S)-1 ,25- Dihydroxy-20-(
  • the invention provides a packaged formulation for use in the treatment of osteoporosis, comprising a pharmaceutical composition comprising a 20- methyl Gemini vitamin D 3 compound and instructions for use in the treatment of osteoporosis in accordance with the methods of the invention.
  • Preferred compounds of this aspect of the invention include (20S)-l J 25-Dihydroxy-20-(5,5,5-trifluoro-4- hydroxy ⁇ -trifluoromethyl-pent ⁇ -ynyO-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20- [(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-l ⁇ -Fluoro-25-hydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trif]uoromethyl-pcnt-2- ynyl)-cholecalciferol (6).
  • An especially preferred compound of the invention is (20S)- l ) 25-Dihydroxy-20-(5,5,5-triflxioro-4-hydroxy-4-trif]uoromethyI-pent-2-ynyl)- cholecalcifcrol (1).
  • Yet another aspect of the invention provides a packaged formulation for use in the treatment of secondary hyperparathyroidism, comprising a pharmaceutical composition comprising a 20-methyl Gemini vitamin D 3 compound and instructions for use in the treatment of secondary hyperparathyroidism in accordance with the methods of the invention.
  • Preferred compounds of this aspect of the invention include (20S)- l ,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluororaethyl-pent-2- enyOcholecalciferol (2); (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl]-cholecalciferol (3); (20R)-l,25-Dihydroxy-20-(5,5 J 5- trifluoro-4-hydroxy-4-trifl ⁇ oromethyl-pent-2-ynyl)-choIecalciferol (4); and (20R)-1, 25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-cnyl]- cholecalciferol (5), A particularly preferred compound is (20S)-1,25-Dihydroxy-20-
  • the invention also provides a pharmaceutical composition, comprising an effective amount a vitamin D3 compound described herein and a pharmaceutically acceptable carrier.
  • the effective amount is effective to treat a vitamin D3 associated state, as described previously.
  • the vitamin D 3 compound is administered to the subject using a pharmaceuticaily-aceeptable formulation, e.g., a pharmaceutically-acceptable formulation that provides sustained delivery of the vitamin D 3 compound to a subject for at least 12 hours, 24 hours, 36 hours, 48 hours, one week, two weeks, three weeks, or four weeks after the pharmaceutically-acceptable formulation is administered to the subject.
  • a pharmaceuticaily-aceeptable formulation e.g., a pharmaceutically-acceptable formulation that provides sustained delivery of the vitamin D 3 compound to a subject for at least 12 hours, 24 hours, 36 hours, 48 hours, one week, two weeks, three weeks, or four weeks after the pharmaceutically-acceptable formulation is administered to the subject.
  • these pharmaceutical compositions are suitable for topical or oral administration to a subject.
  • the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aq ⁇ eous solutions or suspensions), tablets, boluses, powders, granules, pastes; (2) parenteral administration * for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; or (5) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the compound.
  • the subject is a mammal, e.g., a primate, e.g., a human.
  • the methods of the invention further include administering to a subject a therapeutically effective amount of a vitamin D 3 compound in combination with another pharmaceutically active compound.
  • pharmacuctically active compounds include compounds known to treat autoimmune disorders, e.g., immunosuppressant agents such as cyclosporin A, rapamycin, desoxyspergualine, FK 506, steroids, azathioprine, anti-T cell antibodies and monoclonal antibodies to T cell subpopulatio ⁇ s.
  • Other pharmaceutically active compounds that may be used can be found in Harrison's Principles of Internal Medicine, Thirteenth Edition, Eds. T.R.
  • angiogenesis inhibitor compound and the pharmaceutically active compound may be administered to the subject in the same pharmaceutical composition or in different pharmaceutical compositions (at the same time or at different times).
  • pharmaceutically acceptable is refers to those vitamin D 3 compounds of the present invention, compositions containing such compounds, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically-acceptable carrier includes pharmaceutically- acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipicnt, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (S) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflowcr oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (I3) agar; (14) buffering agents, such as magnesium hydroxide and
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine telraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), le
  • compositions containing a vitamin D 3 compound(s) include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.
  • compositions include the step of bringing into association a vitamin D 3 compound(s) with the carrier and, optionally, one or more accessory ingredicnis.
  • the formulations are prepared by uniformly and intimately bringing into association a vitamin D 3 compound with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • compositions of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a vitamin D 3 compound(s) as an active ingredient.
  • a compound may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylccllulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the vitamin D 3 compound(s) include pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethy] carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3- butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can include adjuvants such as we
  • Suspensions in addition to the active vitamin D 3 compound(s) may contain suspending agents as, for example, cthoxylatcd isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, cthoxylatcd isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • compositions of the invention for rectal or vaginal administration may be presented as a suppository, which maybe prepared by mixing one or more vitamin D 3 compound(s) with one or more suitable no ⁇ irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • suitable no ⁇ irritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • compositions of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a vitamin D3 compound(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active vitamin D 3 compound(s) may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to vitamin D3 compound(s) of the present invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to a vitamin D 3 compound(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellanls, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • the vitamin D 3 compound(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound, A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically-acceptable carriers and stabilizers.
  • the carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid., lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols.
  • Aerosols generally are prepared from isotonic solutions.
  • Transdermal patches have the added advantage of providing controlled delivery of a vitamin D 3 compound(s) to the body.
  • dosage forms can be made by dissolving or dispersing the agent in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the active ingredient across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the active ingredient in a polymer matrix or gel.
  • Ophthalmic formulations are also contemplated as being within the scope of this invention.
  • compositions of this invention suitable for parenteral administration comprise one or more vitamin D 3 cornpound(s) in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials;, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutan
  • Injectable depot forms arc made by forming microencapsule matrices of vitamin D3 compound(s) in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthocsters) and poly(a ⁇ hydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • vitamin D 3 compound(s) When administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically-acceptablc carrier.
  • the vitamin D 3 compound(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels and time course of administration of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • An exemplary dose range is from 0.1 to 10 mg per day.
  • a preferred dose of the vitamin D 3 compound for the present invention is the maximum that a patient can tolerate and not develop serious hypercalcemia.
  • the vitamin D3 compound of the present invention is administered at a concentration of about 0.001 ⁇ g to about 100 ⁇ g per kilogram of body weight, about 0.001 - about 10 ⁇ g/kg or about 0.001 ⁇ g - about 100 ⁇ g/kg of body weight. Ranges intermediate to the above-recited values are also intended to be part of the invention.
  • Schemes 1-9 below depict the reaction steps for the synthesis of the highly fluori ⁇ ated-20-metb.yl gemini vitamin D 3 compounds of the invention.
  • Scheme 1 shows the synthetic route for the production of the diol 15 and its epimer, 16.
  • Alcohol 7 was protected with a silyl group to compound 8, then cyclopropanated to provide cyclopropane 9.
  • Conversion of the ester to the aldehyde was accomplished over two steps to provide 11. Chain elongation using a modified Wittig- Horner reaction provided 12. Reduction of the double bond and cyclopropane opening liberated ester 13, which was reduced to alcohol 14.
  • Deprotection and chromatographic separation yielded intermediate 15 and its epimer 16.
  • Scheme 2 shows the chain elongation of 15 to triol 21.
  • Oxidation of the primary alcohol of 15 provided the corresponding aldehyde 17 and chain elongation provided alkyne 18. Protection of the tertiary alcohol to compound 19 was followed by lithiation of the alkyne and reaction with hexafluoroacetone to produce 20. Silyl group deprotection provided triol 21.
  • Scheme 3 shows that 21 is oxidized to ketone 22 which is amenable to Wittig- Homer coupling with an appropriate phosphine oxide. Further reduction of the alkyne of 21 was carried out to form either the cis or trans olefins, 23 and 25, respectively. Oxidation provided ketones 24 and 26.
  • Scheme 4 shows the Wittig-Horner coupling of ketone 22 with phosphine oxide 27 in the presence of base to provide the corresponding coupled product
  • Scheme 4 also shows the coupling of hydroxyl protected ketone 28 with phosphine oxide 29 to provide the corresponding coupled product.
  • Scheme 5 shows the Wittig-Horner coupling of ketone 24 with phosphine oxide 27 in the presence of base to provide the corresponding coupled product. Deprotection of the silyl group(s) with tetrabutyl ammonium fluoride afforded compound 2.
  • Scheme 6 shows the Wittig-Horner coupling of ketone 26 with phosphine oxide 27 in the presence of base to provide the corresponding coupled product. Deprotection of the silyl group(s) with tetrabutyl ammonium fluoride afforded compound 3.
  • Vitamin D3 compounds 4 and 5 were synthesized in accordance with the reactions described in schemes 1-6 above.
  • the starting material for the production of 4 and 5 was 16.
  • Scheme 7 shows the chain elongation of 16 to triol 34. Oxidation of the primary alcohoS of 16 provided the corresponding aldehyde and chain elongation provided alkyne 31. Protection of the tertiary alcohol was followed by lithiation of the alkyne and reaction with hexafluoroacetone to produce 33. Silyl group deprotection provided triol 34.
  • Scheme 9 shows the coupling of ketones 35 and 38 with phosphine oxide 27 to provide compounds 4 and 5.
  • Chiral syntheses can result in products of high stereoisomer purity. However, in some cases, the stereoisomer purity of the product is not sufficiently high.
  • the skilled artisan will appreciate that the separation methods described herein can be used to further enhance the stereoisomer purity of the vitamin D 3 -epimer obtained by chiral synthesis. Any novel syntheses, described herein, of the compounds of the invention, and of intermediates thereof, are also intended to be included within the scope of the present invention.
  • reaction mixture was filtrated through column with silica gel (200 cm 3 ) and celite (2 cm) and using dichloromethane as a mobile phase, The fractions containing product were pooled and evaporated to give oil (ca. 5.71 g). Product was used to the next reaction without purification.
  • reaction mixture was quenched with 10 ml of aqueous saturated solution of ammonium chloride, diluted with 100 ml of saturated solution of ammonium chloride and extracted four times with 50 ml of toluene and then 50 ml of ethyl acetate.
  • the organic layer was washed with 50 ml of brine, dried and evaporated.
  • the residue was purified over silica gel (200 cm 3 ) using hexanerethyl acetate
  • the mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wate ⁇ brine (1: 1) and 50 ml of brine, dried over Na 2 SO,) and evaporated.
  • reaction mixture was filtrated through column with silica gel (50 cm 3 ) and celite (1 cm) using dichloromethane, dichloromcthane:ethyl acetate (4:1). The fractions containing product were pooled and evaporated to give 1.58 g of product as yellow oil. The product was used to the next reaction without farther purification.
  • reaction mixture was cooled to -7O 0 C and 5.00 ml (8.00 mmol) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. After 30 min h ⁇ xafl ⁇ oroacetone was added (the contener's valve was opened three times). The reaction was stirred at -7O 0 C for 2h then 5.0 ml of saturated solution of ammonium chloride was added.
  • the mixture was dissolved by the addition of 100 ml of saturated solution of ammonium chloride and extracted three times with 80 ml of ethyl acetate, dried over Na 2 SO 4 and evaporated.
  • the oil residue was chromatographed twice to remove a large amount of polymer compounds.
  • the first column (100 cm 3 ) using hexane:ethyl acetate (10:1) as mobile phase.
  • the second column (100 cm 3 ) using hexanexthyl acetate (25:1, 15:1) as mobile phase. Fractions containing product were pooled and evaporated to give 1.959 g of colorless oil. Product was used to the next reaction without farther purification.
  • the mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wate ⁇ brine (1 :1) and 50 ml of brine, dried over Na 2 SC ⁇ and evaporated.
  • reaction mixture was stirred, for 4h and then the dry ice was removed from bath and the solution was allowed to warm up to -4O 0 C in Ih.
  • the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine.
  • the water fraction was extracted three times with 50 ml of ethyl acetate, dried .over Na 2 SO- J and evaporated.
  • the mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wate ⁇ brine (1 :1) and 50 ml of brine, dried over NazSO 4 and evaporated.
  • the substrate was hydrogenated at ambient temperature and atmospheric pressure of hydrogen.
  • the reaction was monitoring by TI-C (hexane:eihyl acetate - 2:1).
  • reaction mixture was filtrated through column with silica gel (75 cm 3 ) and celite (2 cm) and using dichloromethane : ethyl acetate (4:1) as a mobile phase.
  • dichloromethane : ethyl acetate (4:1) as a mobile phase.
  • the fractions containing product were pooled and evaporated to give yellow oil.
  • the product was used to the next reaction without farther purification.
  • reaction mixture was stirred for 5h (last 0.5h at -2O 0 C) and then the bath was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine. The water fraction was extracted three times with 50 ml of ethyl acetate, dried over Na 2 SO 4 and evaporated.
  • the mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of water:brine (1:1) and 50 ml of brine, dried over Na 2 SO 4 and evaporated.
  • reaction mixture was stirred at SO 0 C for 5h and then was cooled to O 0 C.
  • the mixture was stirred at room temp for 30 min, 2.2 g of MgSO 4 was added and mixture was stirred for next 15 min.
  • the suspension was filtrated and solvent evaporated.
  • reaction mixture was filtrated through column with silica gel (100 cm 3 ) using dichloromethane : ethyl acetate (4:1) as a mobile phase.
  • the fractions containing product were pooled and evaporated to give 253 mg of yellow oil.
  • the product was used to the next reaction without farther purification
  • reaction mixture was stirred for 5h (last 0.5h at -2O 0 C) and then the bath, was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine.
  • the water fraction was extracted three times with 50 ml of ethyi acetate, dried over NajSCU and evaporated.
  • the oil residue was chromatographed on column (60 cm 3 , protected from light) using hexane:ethyl acetate (4:1) as mobile phase. Fractions containing product were pooled and evaporated to give colorless oil (304 mg) which was treated with 5 ml of 1 M tetrabutylammoni ⁇ jm fluoride in tetrahydrofurane.
  • the reaction mixture was stirred at room temperature for 2 Ih.
  • the mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times'with 50 ml of wate ⁇ brine (1 :1) and 50 ml of brine, dried over NajS ⁇ 4 and evaporated.
  • the reaction mixture was filtrated through column with silica gel (75 cm 3 ) and celite (2 cm) and ysing dichloromethane, dichloromethane:ethyl acetate (4:1) as a mobile phase.
  • the fractions containing product were pooled and evaporated to give 1.298 g of yellow oil.
  • the product was used to the next reaction without farther purification.
  • the mixture was dissolved by the addition of 100 ml of saturated solution of ammonium chloride and extracted three times with 80 ml of ethyl acetate, dried over Na 2 SO 4 and evaporated.
  • the mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wate ⁇ brine (1 :1) and 50 ml of brine, dried over
  • reaction mixture was filtrated through column with silica gel (75 cm 3 ) using dichloromethane, dichloromethane:ethyl acetate (4:1, 3:1). The fractions containing product were pooled and evaporated to give 271 mg (94%) of product as yellow oil.
  • reaction mixture was stirred at -78 0 C for 5h and then the bath was removed and the mixture was poured into 100 ml of saturated solution of ammonium chloride and extracted three times with 50 ml of ethyl acetate, dried over Na 2 SC ⁇ and evaporated.
  • reaction mixture was starred for 4h (last 0.5h at -2O 0 C) and then the bath was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine.
  • the water fraction was extracted three times with 50 ml of ethyl acetate, dried OVeFNa 2 SO 4 and evaporated. ;
  • the maximum tolerated dose of the vitamin D 3 compounds of the invention were determined in eight Week-old female C57BL/6 mice (3 mice/group) dosed orally (0.1 ml/mouse) with various concentrations of Vitamin D 3 analogs daily for four days.
  • Analogs were formulated in miglyol for a final concentration of 10, 30, 100 and 300 ⁇ g/kg when given atj ⁇ .1 ml/mouse p.o. daily. Blood for serum calcium assay was drawn by tail bleed on day five, the final day of the study. Serum calcium levels were determined using a colorimetric assay (Sigma Diagnostics, procedure no. 597). The highest dose of ajnalog tolerated without inducing hypercalcemia (serum calcium >10.7 mg/dl) was taken as the maximum tolerated does (MTD). Table 1 shows the relative MTD for vitamj ⁇ D 3 compounds.
  • Immature dendritic cells were prepared as described in Romani, N. et al., J. Immunol. Meth. 196:137.
  • IFN- ⁇ production by allogeneic T cell activation in the mixed leukocyte response (MLR) was determined as described in Penna, G., et al, J. Immunol., 164: 2405-2411 (2000). Briefly, peripheral blood mononuclear cells (PBMC) were separated from buffy coats by Ficoll gradient and the same number (3x105) of allogeneic PBMC from 2 different donors were co-cultured in 96-well flat-bottom plates. After 5 days, IFN- ⁇ production in the ⁇ t ⁇ LR. assay was measured by EL ⁇ SA and the results expressed as amount (nM) of test compound required to induce 50% inhibition of IFN- ⁇ production (IC 50 ) (TaWe I). :
  • Rats Three-month old Sprague Dawley female rats were purchased from CERJ or Charles River, France. Rats underwent bilateral ovariectomy (OVX) or sham-operation (Sham) under anejsthesia with intraperitoneal ketamine hydrochloride (50 mg/kg BW). The success of ovariectomy was evidenced at necropsy by weighing the uterus and visualizing the absence of ovarian tissue.
  • OVX bilateral ovariectomy
  • Sham sham-operation
  • intraperitoneal ketamine hydrochloride 50 mg/kg BW
  • rats were housed at 22 ⁇ 2°C with a 12h:12h light- dark cycle.
  • the animals were pair-fed a standard diet (Safe, 0.6% P, 0,8% Ca) and received Eau de Volvic ad libitum.
  • Experimental procedures were approved by the Animal Ethics Committee of
  • ⁇ CT tibia and femur
  • histomorphometry tibia
  • the whole left legs were collected for pQCT analysis.
  • the fourth and fifth lumbar vertebrae were dissected for DXA analysis.
  • .Excised tibias were scanned by a ⁇ CT machine (Scanco Medical) with software version 3j.l for a 2D-evaluation. The scans started at a distance of lmm of the reference line in the proximal tibia metaphysis. Five 0.5mm-spaced slices were analyzed. The slice thickness was 20-30 ⁇ m.
  • Bone volume (BV/TV) and trabeculae number (TbN) an ⁇ l thickness (TbTh) were assessed.
  • Excised tibias were scanned by a pQCT machine (Stratec XCT Research SA+) with software version 5.4 to assess trabecular and coijtical Bone Mineral Density (BMD) of proximal tibia metaphysis.
  • BMD Bone Mineral Density
  • the cortical BMD was analyzed at 20 mm from the reference line.
  • the voxel size was 0.10 mm.
  • the peel made used was 20/50.
  • the BMD of excised fourth (L4) and fifth (L5) lumbar vertebrae were scanned by a DXA machine (Hologic QDR 4500) with software optimized for small animal studies.
  • the regional high-resolution sofware selected a thin X-ray aperture.
  • the large region was 68-71 and the narrow one was 35-21 to assess the body of the vertebrae of L4 and L5. ;
  • trabecular boneivolume BV/TV (%), trabecular thickness ( ⁇ m), connections and number, as well! as cellular parameters (osteoblast, osteoid surfaces and osteoclast number), and dynamic parameters (mineralizing surface, mineral apposition rate, bone formation rate) were evaluated, i Serum and Urinary Biochemistry
  • PTH Immutopics
  • Figure 2 shows lumbar spine BMD (DEXA) measurements in 3 month old OVX rats.
  • Figure 3 shows urinary calcium levels in 3 month old OVX rats.
  • Figure 4 shows bone volume in 3 month old OVX rats using (1). The efficacy of
  • Figure 5 sh'ows a reevaluation of 3 month old OVX rats for tibia proximal metaphysic bone volume ( ⁇ CT).
  • Figure 6A shows serum Ca levels in 3 month old rats.
  • Figure 6B shows urinary Ca levels in 3 month old rats. Three month old female rats were orally dosed for three weeks, five days per week, with eight rats per group.
  • Tibia ( ⁇ CT) was found'to be 90% increased over OVX controls with 0.3 ⁇ g/kg in rats treated with (1).
  • Tibia (]uCT) was found to be 114% increased over OVX controls with I ⁇ g/kg in rats treated with (X).
  • the vertebrae (L5) was found to have an increase of 8% over OVX control rats when treated with 0.3 ⁇ g/kg of (1).
  • the vertebrae (L5) was found to have an increases' of 12% over OVX control rats when treated with 1 ⁇ g/kg of (1).
  • compound (1) provided rats with S Ca>10,7 mg/L in two of sixteen rats at 1 i ⁇ g/kg,
  • sham, OVX control rats Compounds or vehicle (sham, OVX control rats) were given by daily oral gavage (5ml/kg) , 5/7 days for 8 weeks. Just before operation a group of intact rats were sacrificed for bas ⁇ line parameters. Before the beginning of treatment, groups of sham and OVX rats we're sacrificed to serve as basal controls. Ten and three days before sacrifice, rats were given subcutaneous injections of calcein (lOml/kg, Sigma) to determine dynamic changes in bone tissue. The day before sacrifice, the rats were fasted and housed in metabolic cages to collect overnight urines.
  • calcein calcein
  • ⁇ CT tibia and f ⁇ mur
  • histomorphometry tibia
  • the whole left legs were collected for pQCT analysis.
  • the fourth and fifth lumbar vertebrae were dissected for DXA analysis.
  • $xcised tibias were scanned by a ⁇ CT machine (Scanco Medical) with software version; 3.1 for a 2D-evaluation. The scans started at a distance of lmm of the reference line in'the proximal tibia metaphysis. Five 0.5mm-spaced slices were analyzed. The slice thickness was 20-30 ⁇ m.
  • Bone volume (BV/TV) and trabecule number (TbN) aind thickness (TbTh) were assessed.
  • Excised tibias were scanned by a pQCT machine ⁇ Stratec XCT Research SA+) with software version 5.4 to assess trabecular and cortical Bone Mineral Density (BMD) of proximal tibia metaphysis.
  • BMD Bone Mineral Density
  • the cortical BMD was analyzed at 20 mm from the reference line.
  • the voxel size was 0.10 mm.
  • the peel njiode used was 20/50.
  • the BMD of excised fourth (L4) and fifth (L5) lumbar vertebrae were scanned by a DXA machine (Hologic QDR 4500) with software optimized for small animal studies.
  • the regional high-resolution sofware selected a thin X-ray aperture. ' The large region was 68-71 and the narrow one was 35-21 to assess the body of the vertebrae of L4 and L5.
  • Structural and dynamic parameters were measured in the secondary spongiosa of the proximal tibial metaphysis situated about 1 mm distal from the growth plate-epiphyseal junction. Structural parameters, i.e.
  • trabecular bone volume BV/TV (%), trabecular thickness ( ⁇ m), connections and number, as well as cellular parameters (osteoblast, osteoid surfaces and osteoclast number), and dynamic parameters (mineralizing surface, mineral apposition rate, bone formation rate) were evaluated.
  • Osteocalcin (IRMA kit, Immutopics), DPyr (Merra DPD EIA kit), CTx (Ratlaps ELISA, Nordic Hioscience Daignostics) Ca, P, creatinin (Cobas Mira analyser) and rat PTH (Immutopics) were assessed in serum or urine material according to the manufacturer's instructions.
  • Figure 7 shows travecular bone volume ( ⁇ CT) measurements in 6 month old OVX rats. ;
  • Figure 8 Shows urinary calcium levels in 6 month old OVX rats.
  • Figure 9l shows BMD (DEXA) measurements in 6 month old OVX rats.
  • Figure ! OA shows serum calcium levels.
  • Figure 1OB shows urinary calcium levels.
  • Figure IiI shows BMD (DEXA) measurements in 6 month old OVX rats.
  • Sprague Dawley male rats (220-25Og BW) were purchased from CERJ, Fiance, Rats were 5/6 nephrectomized (right kidney and 2/3 left kidney removed by surgery) or sham-operated (controls) by the supplier according to Prostrakan' procedures. The success of nephrectomy was evidenced at necropsy by visualizing the kidney tissue and measuring serum creatinin and urea. There were 10-12 rats in each group. The rats were housed at 22 ⁇ 2°C with a 12h:12h light dark cycle. At the beginning ofthe study, the animals were fed a standard diet (Safe, 0.6% P, 0.8% Ca) and received Eau de Volvic ad libitum.
  • phosphate Na2HPO4
  • Eau de Volvic 6g/l
  • Experimental procedures were approved by the Animal Ethics Committee of Prostrakan and by DDSV of Seine St Denis, France.
  • the treatment started 82 days post-nephrcctomy.
  • Compounds were firstly dissolved in ethano ⁇ (lmg/ml). The other dilutions were done in Miglyol 812N.
  • Compounds or vehicle were given by daily oral gavage (5 ml/kg), 5/7 days for 49 days. Just b ⁇ fore operation a group of intact rats were sacrificed for baseline parameters.
  • rats were sacrificed to served as basal controls. Ten and three days before sacrifice, rats were given subcutaneous injections of calcein (10ml/kg, Sigma) to determine dynamic changes in bone tissue. The day before sacrifice, the rats were fasted and housed in metabolic cages to' collect overnight urines.
  • ⁇ CT tibia and fe ⁇ ur
  • histomorphometry tibia
  • the whole left legs were collected for pQCJT analysis.
  • the fourth and fifth lumbar vertebrae were dissected for DXA analysis.
  • Excised t ⁇ bias were scanned by a ⁇ CT machine (Scanco Medical) with software version 3.1 for a 2D-evaluation. The scans started at a distance of lmm of the reference line in the proximal tibia metaphysis. Five 0.5mm-spaced slices were analyzed, The slide thickness was 20-30 ⁇ rn.
  • Bone volume (BV/TV) and trabeculae number (TbN) and thickness (TbTh) were assessed.
  • Excised tibias were scanned by a pQCT machine (Stratec XCT Research SA+) with software version 5.4 to assess trabecular and coftical Bone Mineral Density (BMD) of proximal tibia metaphysis.
  • BMD Bone Mineral Density
  • the BMD of excised fourth (L4) and fifth (L5) lumbar vertebrae' were scanned by a DXA machine (Ho logic QDR 4500) with software optimized for SmJaIl animal studies.
  • the regional high-resolution sofware selected a thin X-ray aperture.
  • the large region was 68-71 and the narrow one was 35-21 to assess the body of the vertejbrae of L4 and L5. 006/001541
  • trabecular bone volume BV/TV (%), trabecular thickness ( ⁇ m), connections and number, as well as cellular parameters (osteoblast, osteoid surfaces and osteoclast number), and dynamic parameters (mineralizing surface, mineral i apposition rate, bone formation rate) were evaluated.
  • PTH (Immutopies) were assessed in serum or urine material according to the manufacturer's instructions.
  • Figure 12Ai shows parathyroid hormone (PTH) levels in rats with moderate renal failure.
  • Figure 12B shows serum Ca levels in rats with moderate renal failure.
  • Figures 13A and 13B show a model of safety parameters, measuring serum and serum Ca levels.
  • Figure 14 A shows a decrease in PTH levels in rats with severe chronic renal failure.
  • Figure 14B shows the serum calcium levels in rats with severe chronic renal failure. ;
  • Figures 15 A and 15B show measurements of serum and serum Ca to determine safety profiles in rats with severe renal failure.
  • Figure 16 A shows the trabecular bone volume measurements in uremic rats.
  • Figure 16B shows' bone mineral density (pQCT) measurements in uremic rats.
  • Figures 17,A 5 17B, and 17C show tibia histomorphornetry analysis, measuring bone formation rate, osteoblast surface, and osteoclast number in uremic rats with moderate renal failure.
  • Figure 18A is a picture of a rat tibia using optical microscopy (x50) of a normal trabecular Figur
  • e 18B is a picture of a rat tibia using optical microscopy (x50) of osteoid thickening
  • Figure 18C is a picture of a rat tibia using optical microscopy (x50) of perit ⁇ abecular fibrosis.
  • Compound (2) provided normal levels of peritrabecular fibrosis at a dose of 3 ⁇ g/kg.
  • Compound (4) provided normal levels of peritrabecular fibrosis at a dose of 0.1 ⁇ lg/kg. At 0.03 ⁇ g/kg, only one of nine rats demonstrated peritrabecular Fibrosis.
  • Compound (2) provided normal levels of osteoid thickening at a dose of 3 ⁇ g/kg and 1 ⁇ g/kg.
  • Compound (4) provided normal levels of osteoid thickening at a dose of 0.03 ⁇ g/kg- At 0.1 ⁇ g/kg, only one of nine rats demonstrated osteoid thickening.
  • Figure 19 shows bone mineral density (DEXA) in uremic rats.
  • Figure 20 is a picture of a rat femur cortical porosity using fluorescence microscopy (xS), showing normal porosity, mild porosity, medium porosity, and marked porosity.
  • Compjound (2) provided mild porous levels of femur cortical porosity in four of ten rats at 1 ⁇ g/kg. At 3 ⁇ g/kg, nine rats demonstrated normal levels of femur cortical porosity.
  • Compound (4) provided mild porous levels of femur cortical porosity in four of nine rats at 0103 ⁇ g/kg.
  • Compound (4) provided mild porous levels of femur cortical porosity in one of nine rats at 0.1 ⁇ g/kg.
  • FIG. 21 ils a picture of a cross section of a rat aorta using Von Kossa staining (xlOO), showing a; control, moderate aorta calcification, and severe aortic calcification.
  • Compound (4) wais administered to seven rats at 0.03 ⁇ g/kg which provided uremia of 20.52 mM and S. breatinin value of 283.23 ⁇ M. No calcification was found in CRF control rats, but those rats presenting calcifications had severe renal failure.
  • Compouiwjls (2) and (4) demonstrated stronger inhibitition of renin in vitro over calcitriol. Both cojmpounds demonstrated efficacy in PTH suppression and bone porosity. Both compounds also demonstrated equal or more beneficial results in terms of safety profiles,; when compared to Zemplar. Compounds (2) and (4) demonstrated greater or equal positive results compared to Zemplar in the renin inhibition in vitro experiments, PTH suppression, CaxP, bone porosity, and aortic calcification.

Landscapes

  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • Diabetes (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Endocrinology (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The invention provides for methods of using 20-methyl Gemini vitamin D3 compounds to treat osteoporosis and secondary hyperparathyroidism.

Description

METHODS OF TREATING OSTEOPOROSIS AND SECONDARY HYPERPARATHYROIDISM USING 20-METHYL, GEMINI VITAMIN D3
COMPOUNDS
Related Applications
This application claims the benefit of U.S. provisional patent application No. 60/664,397, filed March 23, 2005. This application is related to international patent application No. PCT/US2006/XXXXXX, filed on March 23, 2006 (Attorney Docket No. 49949-63097PCT(A), Express Mail Label No. EV 756031949 US). The disclosures of both applications are incorporated herein in their entireties by this reference.
Background of the Invention
The importance of vitamin D (cholecalciferol) in the biological systems of higher animals has been recognized since its discovery by Mellanby in 1920 (Mellanby, E. (1921) Spec. Rep. Ser. Med. Res. Council (GB) SRS 61 :4). It was in the interval of 1920-1930 that vitamin D officially became classified as a "vitamin" essential for the normal development of the skeleton and maintenance of calcium and phosphorous homeostasis.
Studies involving the metabolism of vitamin D3 were initiated with the discovery and chemical characterization of the plasma metabolite, 25-hydroxyvitamin Dj
[25(OH)D3] (Blunt J.W. et al. (1968) Biochemistry 6:3317-3322) and the hormonally active form, lα,25(OH)2D3 (Myrtle, J.F. et al. (1970) J. Biol. Chem. 245:1190-1 196; Norman, A.W. et al (1971) Science 173:51-54; Lawson, D.E.M. et al (1971) Nature 230:228-230; Holick, M.F. (197I) PrOc. Natl. Acad. ScL USA 68:803-804). The formulation of the concept of a vitamin D endocrine system was dependent upon the appreciation of the key role of the kidney in producing lα, 25(OH)iD3 in a carefully regulated fashion (Fraser, O.K. and Kodicek, E (1970) Nature 288:764-766; Wong, R.G. et al. (1972) /. CHn. Invest. 51:1287-1291), and the discovery of a nuclear receptor for 1 α,25(OH)2D3 (VD3R) in the intestine (Haussler, M.R. et al. (1969) Exp. Cell Res. 58:234-242; Tsai, H.C. and Norman, A.W. (1972) J. Biol Chem. 248:5967-5975).
The operation of the vitamin D endocrine system depends on the following: first, on the presence of cytochrome P450 enzymes in the liver (Bergman, T. and Postlind, H. (1991) Biochem. J. 276:427-432; Ohyama, Y and Okuda, K. (1991) J. Biol. Chem. 266:8690-8695) and kidney (Henry, HX. and Norman, A.W. (1974) J. Biol. Chem. 249:7529-7535; Gray, R.W. and Ghazarian, IG. (1989) Biochem. J. 259:561-568), and in a variety of other tissues to effect the conversion of vitamin D3 into biologically active metabolites such as lα, 25(OH)2D3 and 24R,25(OH)2D3; second, on the existence of the plasma vitamin D binding protein (DBP) to effect the selective transport and delivery of these hydrophobic molecules to the various tissue components of the vitamin D endocrine system (Van Baelen, H. et al. (1988) Ann NY Acad. ScL 538:60-68; Cooke, NJE. and Haddad, J.G. (1989) Endocr. Rev. 10:294-307; Bikle, D.D. et al. (1986) J. Clin. Endocrinol. Metab. 63:954-959); and third, upon the existence of stereoselective receptors in a wide variety of target tissues that interact with the agonist lα,25(OH)zD3 to generate the requisite specific biological responses for this secosteroid hormone (Pike> J.W, (1991) Annu. Rev. Nutr. 11:189-216). To date, there is evidence that nuclear receptors for lα,25(OH)2D3 (VD3R) exist in more than 30 tissues and cancer cell lines (Reichel, H. and Norman, A.W. (1989) Annu. Rev. Med. 40:71-78).
Vitamin D3 and its hormonally active forms are well-known regulators of calcium and phosphorous homeostasis. These compounds are known to stimulate, at least one of, intestinal absorption of calcium and phosphate, mobilization of bone mineral, and retention of calcium in the kidneys. Furthermore, the discovery of the presence of specific vitamin D receptors in more than 30 tissues has led to the identification of vitamin D3 as a pluripotent regulator outside its classical role in calcium/bone homeostasis. A paracrine role for lα,25(OH)2 D3 has been suggested by the combined presence of enzymes capable of oxidizing vitamin D3 into its active forms, e.g., 25-OHD-lα.-hydroxylase, and specific receptors in several tissues such as bone, keratinocytes, placenta, and immune cells. Moreover, vitamin D3 hormone and active metabolites have been found to be capable of regulating cell proliferation and differentiation of both normal and malignant cells (Reichcl, H. et al. (1989) Ann. Rev. Med. 40: 71-78). Given the activities of vitamin D3 and its metabolites, much attention has focused on the development of synthetic analogs of these compounds. A large number of these analogs involve structural modifications in the A ring, B ring, C/D rings, and, primarily, the side chain (Bouillon, R. et al. , Endocrine Reviews 16(2):201-204). Although a vast majority of the vitamin D3 analogs developed to date involve structural modifications in the side chain, a few studies have reported the biological profile of A-ring diastereomers (Norman, A.W. et al. J. Biol. Chem. 268 (27): 20022-20030). Furthermore, biological esterification of steroids has been studied (Hochberg, R.B., (1998) Endocr Rev. 19(3): 331-348), and esters of vitamin D3 are known (WO 97/11053).
Moreover, despite much effort in developing synthetic analogs, clinical applications of vitamin D and its structural analogs have been limited by the undesired side effects elicited by these compounds after administration to a subject for known
.-}- indications/ applications of vitamin D compounds. Therefore, structural analogs of vitamin D having improved therapeutic activity, particularly for the treatment of osteoporosis and secondary hyperparathyroidism and/or reduced undesirable side effects are needed.
Summary of the Invention
The invention provides novel vitamin D3 compounds having improved therapeutic activity for the treatment of osteoporosis and secondary hyperparathyroidism and/or reduced undesirable side effects useful for the treatment of osteoporosis and secondary.
Thus, in one aspect, the invention provides a method for treating osteoporosis in a subject comprising administering to a subject in need thereof a therapeutically effective amount of a vitamin D3 compound selected from the group consisting of (20S)- 1 ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluororriethyl-pent-2-ynyl)- cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-lα-Fluoro-25-hydroxy-20- (5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (6), thereby treating the subject for osteoporosis. In another aspect, the invention provides a method for treating a subject for secondary hyperparathyroidism comprising administering to a subject in need thereof a therapeurically effective amount of a vitamin D3 compound selected from the group consisting of (20S)-l,25-Dihydroxy-20-((2Z)-5I5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl)cholecalciferol (2); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5- trifluoro-4-hydτoxy-4-trifluoromcthyl-pent-2-enyrj-cholecalciferol (3); (20R)-1, 25-
Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalcifero] (4); and (20R)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl]-cholecalciferol (5), thereby treating the subject for secondary hyperparathyroidism. In yet another aspect, the invention provides a pharmaceutical composition for use in the treatment of osteroporosis, comprising a therapeutically effective amount of a vitamin D3 compound selected from the group consisting of (20S)-1,25-Dihydroxy-20- (S^.S-trifluoro^-hydroxy^-trifluoromethyl-pent^-ynylJ-cholecalcifero^l); (20S)- l325-Dihydroxy-20-[(2E)-515,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (3); and (20S)-lα-Fluoro-25-hydroxy-20-(5,5,5-trifiuoro-4-hydroxy-4- triflυoromcthyl-pcn1.-2-ynyl)-cholecalciferol (6), and a pharmaceutically acceptable diluent or carrier.
In still another aspect, the invention provides a pharmaceutical composition for use in the treatment of secondary hyperparathyroidism comprising a therapeutically effective amount of a vitamin D3 compound selected from the group consisting of (20S)- I,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- enyl)cholecalcifcrol (2); (20S)-l,25~Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl]-cholecalciferol (3); (20R)-l,25-Dihydroxy-20-(5,5,5- trifluoro-4-hydroxy-4-trifiuoromethyl-pent-2-ynyl)-cholecalciferol (4); and (20R)- 1,25- Dihydroxy-20-[(2E)-5,555-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (5), and a pharmaceutically acceptable diluent or carrier.
Another aspect of the invention provides a packaged formulation for use in the treatment of osteoporosis, comprising a pharmaceutical composition comprising a vitamin D3 compound selected from the the group consisting of (20S)-1,25-Dihydroxy- 20-(5J5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (1); (20S)- l,25-Dihydroxy-20-[(2E)-5)5,5-trifluoro-4-hydroxy-4-trifluoromethyI-pent-2-enyl]- cholecalciferol (3); and (20S)-lα-Fluoro-25-hydroxy-20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-ynyl)-cholecalciferol (6); and instructions for use in the treatment of osteoporosis. Yet another aspect of the invention provides a packaged formulation for use in the treatment of secondary hyperparathyroidism, comprising a pharmaceutical composition comprising a vitamin D3 compound selected from the group consisting of (20S)-l,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-ρent-2- enyl)cholecalciferol (2); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- mfluoromethyl-pent-2-enyl]-cholecalciferol (3); (20R)-l,25-Dihydroxy-20-(5,5,5- trifluoro-4-hydroxy-4-tτifluoromethyl-pent-2-ynyl)-choleca]ciferol (4); and (2QR)-1, 25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (5), and instructions for use in the treatment of secondary hyperparathyroidism.
Brief Description of the Drawings
Figure 1 shows tibia proximal metaphysic bone volume (uCT) measurements in 3 month old OVX rats. Figure 2 shows lumbar spine BMD (DEXA) measurements in 3 month old OVX rats. Figure 3 shows urinary calcium levels in 3 month old OVX rats.
Figure 4 shows bone volume in 3 month old OVX rats using (1).
Figure 5 shows a reevaluation of 3 month old OVX rats for tibia proximal metaphysic bone volume (μCT). Figure 6A shows serum Ca levels in 3 month old rats. Figure 6B shows urinary
Ca levels in 3 month old rats.
Figure 7 shows travecular bone volume (μCT) measurements in 6 month old OVX rats.
Figure 8 shows urinary calcium levels in 6 month old OVX rats. Figure 9 shows BMD (DEXA) measurements in 6 month old OVX rats.
Figure 1OA shows serum calcium levels. Figure 1OB shows urinary calcium levels.
Figure 11 shows BMD (DEXA) measurements in 6 month old OVX rats.
Figure 12A shows parathyroid hormone (PTH) levels in rats with moderate renal failure. Figure 12B shows serum Ca levels in rats with moderate renal failure.
Figures 13A and 13B show a model of safety parameters, measuring serum and serum Ca levels.
Figure 14A shows a decrease in PTH levels in rats with severe chronic renal failure. Figure 14B shows the serum calcium levels in rats with severe chronic renal failure.
Figures 15A and 15B show measurements of serum and serum Ca to determine safety profiles in rats with severe renal failure.
Figure 16A shows the trabecular bone volume measurements in uremic rats. Figure 16B shows bine mϊnarl density (pQCT) measurements in uremic rats. Figures 17A, 17B, and 17C show tibia histomorphometry analysis, measuring bone formation rate, osteoblast surface, and osteoclast number in uremic rats with moderate renal failure.
Figure 18A is a picture of a rat tibia using optical microscopy (x50) of a normal trabeculae. Figure 18B is a picture of a rat tibia using optical microscopy (x50) of osteoid thickening. Figure 18C is a picture of a rat tibia using optical microscopy (x50) of peritrabecular fibrosis.
Figure 19 shows bone mineral density (DEXA) in uremic rats.
Figure 20 is a picture of a rat femur cortical porosity using fluorescence microscopy (x8), showing normal porosity, mild porosity, medium porosity, and marked porosity. Figure 21 is a picture of a cross section of a rat aorta using Von Kossa staining (xlOO), showing a control, moderate aorta calcification, and severe aortic calcification.
Detailed Description of the Invention
1. DEFINITIONS
Before further description of the present invention, and in order that the invention may be more readily understood, certain terms are first defined and collected here for convenience. The term "administration" or "administering" includes routes of introducing the vitamin D3 compound(s) to a subject to perform their intended function. Examples of routes of administration which can be used include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecal), oral, inhalation, rectal and transdermal. The pharmaceutical preparations are, of course, given by forms suitable for each administration route. For example, these preparations are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred. The injection can be bolus or can be continuous infusion. Depending on the route of administration, the vitamin D3 compound can be coated with or disposed in a selected material to protect it from natural conditions which may detrimentally effect its ability to perform its intended function. The vitamin D3 compound can be administered alone, or in conjunction with either another agent as described above or with a pharmaceutically-acceptable carrier, or both. The vitamin D3 compound can be administered prior to the administration of the other agent, simultaneously with the agent, or after the administration of the agent.
Furthermore, the vitamin D3 compound can also be administered in a proform which is converted into its active metabolite, or more active metabolite in vivo.
The term "alkyl" refers to the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. The term alkyl further includes alkyl groups, which can further include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone, e.g., oxygen, nitrogen, sulfur or phosphorous atoms. In preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chain, C3-C30 for branched chain), preferably 26 or fewer, and more preferably 20 or fewer. Likewise, preferred cycloalkyis have from 3-10 carbon atoms in their ring structure, and more preferably have 3, 4, 5, 6 or 7 carbons in the ring structure.
Moreover, the term alkyl as used throughout the specification and claims is intended to include both "unsubstituted alkyls" and "substituted alkyls," the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alky lth iocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonate, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. Cycloalkyis can be further substituted, e.g., with the substituents described above. An "alkyiaryl" moiety is an alkyl substituted with an aryl (e.g., phenylmcthyl (benzyl)). The term "alky!" also includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
Unless the number of carbons is otherwise specified, "lower alky]" as used herein means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six, and most preferably from one to four carbon atoms in its backbone structure, which may be straight or branched-chain. Examples of lower alkyl groups include methyl, ethyl, n-propyl, i-propyl, tert-butyl, hexyl, heptyl, octyl and so forth. In preferred embodiment, the term "lower alkyl" includes a straight chain alkyl having 4 or fewer carbon atoms in its backbone, e.g., C1-C4 alkyl. The terms "alkoxyalkyl," "polyaminoalkyl" and "thioalkoxyalkyl" refer to alkyl groups, as described above, which further include oxygen, nitrogen or sulfur atoms replacing one or more carbons of the hydrocarbon backbone, e.g., oxygen, nitrogen or sulfur atoms.
The terms "alkenyl" and "alkynyl" refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond, respectively. For example, the invention contemplates cyano and propargyl groups. The term "aryl" as used herein, refers to the radical of aryl groups, including 5- and 6-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, pyrrole, furan, thiophene, imidazole, benzoxazole, benzothiazolc, triazole, tetrazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Aryl groups also include polycyclic fused aromatic groups such as naphthyl, quinolyl, indolyl, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles," "heteroaryls" or "heteroaromatics." The aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonate, sulfamoyl, sulfonamide, nitro, trifiuoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. Aryl groups can also be fused or bridged with alicyclic or heterocyclic rings which are not aromatic so as to form a polycycle {e.g., tetraiin).
The language "biological activities" of vitamin D3 includes all activities elicited by vitamin D3 compounds in a responsive cell. It includes genomic and nσn-genomic activities elicited by these compounds (Gniadecki R. and Calverley MJ. (1998) Pharmacology & Toxicology 82: 173-176; Bouillon, R. et al. (1995) Endocrinology Reviews 16(2):206-207; Norman A.W. et al. (1992) J. Steroid Biochem MoI Biol 41 :231-240; Baran D.T. et al. (1991) /. Bone Miner Res. 6:1269-1275; Caffrey J.M. and Farach-Carson M.C. (1989) J. Biol. Chem. 264:20265-20274; Nemere 1 et al. (1984) Endocrinology 1 15:1476-1483).
The language "bone metabolism" includes direct or indirect effects in the formation or degeneration of bone structures, e.g., bone formation, bone resorption, etc., which may ultimately affect the concentrations in serum of calcium and phosphate. This term is also intended to include effects of compounds of the invention in bone cells, e.g., osteoclasts and osteoblasts, that may in turn result in bone formation and degeneration. The language "calcium and phosphate homeostasis" refers to the careful balance of calcium and phosphate concentrations, intraccllularly and extracellularly, triggered by fluctuations in the calcium and phosphate concentration in a cell, a tissue, an organ or a system. Fluctuations in calcium levels that result from direct or indirect responses to compounds of the invention are intended to be included by these terms. The term "chiral" refers to molecules which have the property of non- superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner. The term "diastereomers" refers to stereoisomers with two or more centers of dissymmetry and whose molecules are not mirror images of one another.
The term "deuteroalkyl" refers to alky] groups in which one or more of the of the hydrogens has been replaced with deuterium.
The term "effective amount" includes an amount effective, at dosages and for periods of time necessary, to achieve the desired result, e.g., sufficient treat a vitamin Dj associated state or to modulate ILT3 expression in a cell. An effective amount of vitamin D3 compound may vary according to factors such as the disease state, age, and weight of the subject, and the ability of the vitamin Dj compound to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. An effective amount is also one in which any toxic or detrimental effects (e.g., side effects) of compound are outweighed by the therapeutically beneficial effects.
A therapeutically effective amount of vitamin D3 compound (i.e., an effective dosage) may range from about 0.001 to 30 μg/kg body weight, preferably about 0.01 to 25 μg/kg body weight, more preferably about 0.1 to 20 μg/kg body weight, and even more preferably about 1 to 10 μg/kg, 2 to 9 μg/kg, 3 to 8 μg/kg, 4 to 7 μg/kg, or 5 to 6 μg/kg body weight. The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a vitamin D3 compound can include a single treatment or, preferably, can include a series of treatments. In one example, a subject is treated with a vitamin D3 compound in the range of between about 0.1 to 20 μg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. It will also be appreciated that the effective dosage of a vitamin D3 compound used for treatment may increase or decrease over the course of a particular treatment.
The term "enan Homers" refers to two stereoisomers of a compound which are non-superimposable mirror images of one another. An equimolar mixture of two enantiomers is called a "racemic mixture" or a "racemate." The language "Gemini vitamin D3 compounds" is intended to include vitamin D3 compounds and analogs thereof having bis C20 side chains. Vitamin D3 compounds are characterized by an "A" ring (monocycle) which is connected to a "B" ring (bicycle) which is connected to a side chain at carbon C20 of the side chain. The Gemini compounds of the invention have two side chains and are, therefore, conspicuously distinguishable from vitamin D3 compounds having a single side chain. Candidate A and B rings for the Gemini compounds of the invention arc disclosed in U.S. Patent Nos. 6,559,138, 6,329,538 , 6,331,642 , 6,452,028 , 6,492,353, 6,040,461 , 6,030,963, 5,939,408, 5,872,113, 5,840,718, 5,612,328, 5,512,554, 5,451,574, 5,428,029, 5,145,846, and 4,225,525. Examples of Gemini compounds in accordance with the invention are disclosed in U.S. Patent No. 6,030,962.
The term "halogen" designates -F, -Cl, -Br or -I.
The term "haioalkyl" is intended to include alkyl groups as defined above that are mono-, di- or polysubstituted by halogen, e.g., fluoromethyl and trifluoromethyl. The term "hydroxy!" means -OH.
The term "heteroatom" as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus.
The term "homeostasis" is art-recognized to mean maintenance of static, or constant, conditions in an internal environment. The language "hormone secretion" is art-recognized and includes activities of vitamin D3 compounds that control the transcription and processing responsible for secretion of a given hormone e.g,, a parathyroid hormone (PTH) of a vitamin D3 responsive cell (Bouillon, R. et al. (1995) Endocrine Reviews 16(2):235-237).
The language "hypercalcemia" or "hypercalcemic activity" is intended to have its accepted clinical meaning, namely, increases in calcium serum levels that are manifested in a subject by the following side effects, depression of central and peripheral nervous system, muscular weakness, constipation, abdominal pain> lack of appetite and, depressed relaxation of the heart during diastole. Symptomatic manifestations of hypercalcemia are triggered by a stimulation of at least one of the following activities, intestinal calcium transport, bone calcium metabolism and osteocalcin synthesis (reviewed in Bouliion, R. et al. (1995) Endocrinology Reviews 16(2): 200-257).
The language "improved biological properties" refers to any activity inherent in a compound of the invention that enhances its effectiveness in vivo. In a preferred embodiment, this term refers to any qualitative or quantitative improved therapeutic property of a vitamin D3 compound, such as reduced toxicity, e.g. , reduced hypercalcemic activity. The term "isomers" or "stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
The term "modulate" refers to increases or decreases in the activity of a cell in response to exposure to a compound of the invention, e.g., the inhibition of proliferation and/or induction of differentiation of at least a sub-population of cells in an animal such that a desired end result is achieved, e.g., a therapeutic result. In preferred embodiments, this phrase is intended to include hyperactive conditions that result in pathological disorders. The language "non-genomic" vitamin D3 activities include cellular (e.g., calcium transport across a tissue) and subcellular activities (e.g., membrane calcium transport opening of voltage-gated calcium channels, changes in intracellular second messengers) elicited by vitamin D3 compounds in a responsive cell. Electrophysiological and biochemical techniques for detecting these activities are known in the art. An example of a particular well-studied non-genomic activity is the rapid hormonal stimulation of intestinal calcium mobilization, termed "transcaltachia" (Nemere I. et al. (1984) Endocrinology 115: 1476-1483; Lieberherr M. et al (1989) J. Biol Chem. 264:20403- 20406; WaIi R.K. et al. (1992) Endocrinology 131 : 1125-1133; WaIi R.K. et al. (1992) Am. J. Physiol. 262:G945-G953; WaIi R.K. et al (199O) J. CUn. Invest. 85: 1296-1303; Bolt M.J.G. et al (1993) Biochem. J. 292:271-276). Detailed descriptions of experimental transcaltachia are provided in Norman, A.W. (1993) Endocrinology 268(27):20022-20030; Yoshimoto, Y. and Norman, A.W. (1986) Eridocrinologyl 18:2300-2304. Changes in calcium activity and second messenger systems are well known in the art and are extensively reviewed in Bouillion, R. et al. (1995) Endocrinology Review 16(2): 200-257; the description of which is incorporated herein by reference.
The term "obtaining" as in "obtaining a vitamin D3 compound" is intended to include purchasing, synthesizing or otherwise acquiring the compound.
The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. The terms "polycyclyl" or "polycycHc radical" refer to the radical of two or more cyclic rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Rings that are joined through non-adjacent atoms are termed "bridged" rings. Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, hydroxy!, alkylcarbonyloxy, aryJcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarboπyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, aryiamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfamoyl, sulfonamide, nitro, trifluoromcthyl, cyano, azido, heterocyclyl, alkyl, alkylaryl, or an aromatic or heterαaromatic moiety.
The term "prodrug" includes compounds with moieties which can be metabolized in vivo. Generally, the prodrugs are metabolized in vivo by esterases or by other mechanisms to active drugs. Examples of prodrugs and their uses are well known in the art (See, e.g., Beτge et al. (1977) "Pharmaceutical Salts", J. Pharm. Set. 66:3-19). The prodrugs can be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form or hydroxyl with a suitable esterifying agent. Hydroxyl groups can be converted into esters via treatment with a carboxylic acid. Examples of prodrug moieties include substituted and unsubstiruted, branch or unbranched lower alkyl ester moieties, (e.g. , propionoic acid esters), lower alkenyl esters, di-iower alkyl-amino lower-alkyl esters (e.g., dimethylaminoethyi ester), acylamino lower alkyl esters (e.g., acetyloxymethy] ester), acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters (phenyl ester), aryl-lower alkyl esters (e.g., benzyl ester), substituted (e.g., with methyl, haJo, or methoxy substituents) aryl and aryl-lower alkyl esters, amides, lower-alkyl amides, di- lower alkyl amides, and hydroxy amides. Preferred prodrug moieties are propionoic acid esters and acyl esters. Prodrugs which are converted to active forms through other mechanisms in vivo are also included.
The language "reduced toxicity" is intended to include a reduction in any undesired side effect elicited by a vitamin D3 compound when administered in vivo, e.g., a reduction in the hypercalcemic activity.
The term "secosteroid" is art-recognized and includes compounds in which one of the cyclopentanoperhydro- phenanthrene rings of the steroid ring structure is broken. lα,25(OH)2D3 and analogs thereof are hormonally active secosteroids. In the case of vitamin Oy, the 9-10 carbon-carbon bond of the B-ring is broken, generating a seco-B- steroid. The official IUPAC name for vitamin D3 is 9,10-secocholesta-5,7,iO(19)-trien- 3B-ol. For convenience, a 6-s-trans confoπner of 1 α,25(OH)2D3 is illustrated herein having all carbon atoms numbered using standard steroid notation.
Figure imgf000014_0001
In the formulas presented herein, the various substituents on ring A are illustrated as joined to the steroid nucleus by one of these notations: a dotted line ( — ) indicating a substituent which is in the β-orientation {i.e. , above the plane of the ring), a wedged solid line (<) indicating a substituent which is in the α-orientation {i.e. , below the plane of the molecule), or a wavy line ( 'λn^'- ) indicating that a substituent may be either above or below the plane of the ring. In regard to ring A, it should be understood that the stereochemical convention in the vitamin D field is opposite from the general chemical field, whexein a dotted line indicates a substituent on Ring A which is in an a- orientation (i.e. , below the plane of the molecule), and a wedged solid line indicates a substituent on ring A which is in the β-oriεntation (i.e. , above the plane of the ring). As shown, the A ring of the hormone lα,25(OH)2D3 contains two asymmetric centers at carbons 1 and 3, each one containing a hydroxyl group in well-characterized configurations, namely the lα- and 3β- hydroxyl groups. In other words, carbons 1 and 3 of the A ring are said to be "chiral carbons" or "carbon centers".
Also, throughout the patent literature, the A ring of a vitamin D compound is often depicted in generic formulae as any one of the following structures:
Figure imgf000014_0002
wherein X, is defined as H (or H2 ) or =CH2; or
Figure imgf000015_0001
wherein Xi is defined as H2 or CH2. Although there does not appear to be any set convention, it is clear that one of ordinary skill in the art understands either formula I or =CH2, as follows:
Figure imgf000015_0002
For purposes of the instant invention, the representation of the A ring as shown immediately above in formula II will be used in all generic structures.
Furthermore the indication of stereochemistry across a carbon-carbon double bond is also opposite from the general chemical field in that "Z" refers to what is often referred to as a "cis" (same side) conformation whereas "E" refers to what is often referred to as a "trans" (opposite side) conformation. As shown, the A ring of the hormone l-alpha,25(O HThDs contains two asymmetric centers at carbons 1 and 3, each one containing a hydroxyl group in well-characterized configurations, namely the 1- alpha- and 3-beta- hydroxyl groups. In other words, carbons 1 and 3 of the A ring are said to be "chiral carbons" or "chiral carbon centers." Regardless, both configurations, cis/trans and/or ZfB are encompassed by the compounds of the present invention. With respect to the nomenclature of a chiral center, the tenns "d" and "1" configuration are as defined by the IUPAC Recommendations. As to the use of the terms, diastereomer, racemate, epimer and enantiomer, these will be used in their normal context to describe the stereochemistry of preparations.
The term "subject" includes organisms which are capable of suffering from a vitamin D3 associated state or who could otherwise benefit from the administration of a vitamin D3 compound of the invention, such as human and non-human animals. Preferred human animals include human patients suffering from or prone to suffering from a vitamin D3 associated state, as described herein. The term "non-human animals" of the invention includes all vertebrates, e.g., , mammals, e.g., rodents, e.g., mice, and non-mammals, such as non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc. The term "siilfhydryl" or "thiol" means -SH.
The phrases "systemic administration," "administered systemically", "peripheral administration" and "administered peripherally" as used herein mean the administration of a vitamin D3 comρound(s), drug or other material, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
The term "VDR" is intended to include members of the type II class of steroid/thyroid superfamily of receptors (Srunnenberg, H.G. (1993) Bio Essays 15(5):309-15), which are able to bind and transactivate through the vitamin D response element (VDRE) in the absence of a ligand (Damm et al. (1989) Nature 339:593-97; Sap et al. Nature 343: 177-180).
The term "VDRE" refers to DNA sequences composed of half-sites arranged as direct repeats. It is known in the art that type II receptors do not bind to their respective binding site as homodimers but require an auxiliary factor, RXR (e.g. RXRα, RXRβ, RXRγ) for high affinity binding Yu et al. (1991) Cell 67:1251-1266; Bugge et al
(1992) EMBOJ. 1 1:1409-1418; Kliewer et at. (1992) Nature 355:446-449; Leid et al. (1992) EMBO J. 1 1 :1419-1435; Zhang er a/. (1992) Nature 355:441 -446).
The language "vitamin D3 associated state" is a state which can be prevented, treated or otherwise ameliorated by administration of one or more compounds of the invention. Vitamin D3 associated states include ILT3 -associated disorders, disorders characterized by an aberrant activity of a vitamin D3-responsive cell, disorders characterized by a deregulation of calcium and phosphate metabolism, and other disorders or states described herein.
The term "vitamin D3-responsive cell" includes any cell which is is capable of responding to a vitamin D3 compound described herein, or is associated with disorders involving an aberrant activity of hyperproliferative skin cells, parathyroid cells, neoplastic cells, immune cells, and bone cells. These cells can respond to vitamin D% activation by triggering genomic and/or non-genomic responses that ultimately result in the modulation of cell proliferation, differentiation survival, and/or other cellular activities such as hormone secretion. In a preferred embodiment, the ultimate responses of a cell are inhibition of cell proliferation and/or induction of differentiation-specific genes. Exemplary vitamin D3 responsive cells include immune cells, bone cells, neuronal cells, endocrine cells, neoplastic cells, epidermal cells, endodermal cells, smooth muscle cells, among others. With respect to the nomenclature of a chiral center, terms "d" and "1" configuration are as defined by the IUPAC Recommendations. As to the use of the terms, diastereomer, racemate, epimer and enantiomer will be used in their normal context to describe the stereochemistry of preparations.
2. GEMINI VITAMIN O1 COMPOUNDS
In certain aspects, the invention provides for the use of vitamin D compounds to treat osteoporosis and secondary hyperparathyroidism. Preferred compounds for use in the methods of the invention include the following compounds:
(20S)-l ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (1):
Figure imgf000017_0001
(20S)-l ,25-Dihydroxy-20-((2Z)-5,5,5-trifiuoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl)cholecalciferol (2):
Figure imgf000017_0002
(20S)-l)25-Dihydroxy-20-[(2E)-5,5)5-trifluoro-4-hydroxy-4-trifluoromethyl- (3):
Figure imgf000017_0003
(2OR)-1 , 25-Dihydroxy-20-(5,555-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (4):
Figure imgf000018_0001
(20R)-l,25-Dihydroxy-20-[(2E)-5,5,5-trifiuoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl]-cholecaIciferol (5):
Figure imgf000018_0002
(20S)- 1 α-F!uoro-25-hydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyI-pent-2- ynyl)-cho]ecalciferol (6):
Figure imgf000018_0003
In certain embodiments, especially preferred compounds include (20S)- 1,25- Dihydroxy^O^S.S.S-trifluoro^-hydroxy^-trifluoroinethyl-pent^-yny^-choIecalciferol (1) and (20S)-1 ,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl)choleca]ciferol (2).
The structures of some of the compounds of the invention include asymmetric carbon atoms. Accordingly, the isomers arising from such asymmetry (e.g., all enantiomers and diastereomers) are included within the scope of this invention, unless indicated otherwise. Such isomers can be obtained in substantially pure form by classical separation techniques and/or by stereochemical^ controlled synthesis.
Naturally occurring or synthetic isomers can be separated in several ways known in the art. Methods for separating a racemic mixture of two enantiomers include chromatography using a chiral stationary phase (see, e.g., , "Chiral Liquid Chromatography," WJ. Lough, Ed. Chapman and Hall, New York (1989)). W 2
Enantiomers can also be separated by classical resolution techniques. For example, formation of diastereomeric salts and fractional crystallization can be used to separate enantiomers. For the separation of enantiomers of carboxylic acids, the diastereomeric salts can be formed by addition of enantiomerically pure chiral bases such as brucine, 5 quinine, ephedrine, strychnine, and the like. Alternatively, diastereomeric esters can be formed with enantiomerically pure chiral alcohols such as menthol, followed by separation of the diastereomeric esters and hydrolysis to yield the free, enantiomerically enriched carboxylic acid. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric 10 acid, mandclic acid, or lactic acid can result in formation of the diastereomeric salts.
3. USES OF THE VITAMIN P^ COMPOUNDS OF THE INVENTION
In current methods, the use of vitamin D3 compounds has been limited because
15 of their hypercalcemic effects. The 20-mehtyl Gemini vitamin D3 compounds of the invention can provide a less toxic alternative to current methods of treatment for osteoporosis and secondary hyperparathyroidism.
In one aspect, the invention provides a method for treating osteoporosis in a subject comprising administering to a subject in need thereof a therapeurically effective
20 amount of a 20-methyl Gemini vitamin Dj compound of the invention, thereby treating said subject for osteoporosis. Preferred compounds for this aspect of the invention include (20S)- 1 ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (t); (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifiuoromethyl-pent-2~enyl]-cholecalcifcrol (3); and (20S)-lα-Fluoro-25-hydroxy-20-
25 (S^jS-trifluoro^-hydroxy^-trifJuoromethyl-pent^-ynyO-cholecalciferol (6). An especially preferred compound of the invention is (20S)-l,25-Dihydroxy-20-(5,5,5- trifluoro^-hydroxy^-trifluoromethyl-pent^-ynyO-cholecalciferol (1).
Irt one embodiment of this aspect of the invention, the method further comprises identifying a subject as being in need of treatment for osteoporosis. In another
30 embodiment the method further comprises obtaining the vitamin D3 compound.
Another aspect of the invention provides a method for treating a subject for secondary hyperparathyroidism comprising administering to a subject in need thereof a therapeurically effective amount of a 20-methyl Gemini vitamin D3 compound of the invention, thereby treating the subject for secondary hyperparathyroidism.
35 Preferred compounds for this aspect of the invention include (20S)-1,25-Dihydroxy-20- ((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl)cholecalciferol (2); (20S)- l,25-Dihydroxy-20-[(2E)-555J5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (3); (20R)-1 ,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-perit-2-ynyl)-cholecalciferol (4); and (20R)-1,25-Dihydroxy-20-[(2E)- 5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (5). An especially preferred compound of the invention is is (20S)-1 ,25-Dihydroxy-20-((2Z)- 5,5,5-trifJuoro-4-hydroxy-4-triiluorornethyI-pent-2-enyl)cholecalciferol (2).
In one embodiment of this aspect of the invention, the method further comprises identifying a subject as being in need of treatment for secondary hyperparathyroidism. In another embodiment the method further comprises obtaining the vitamin D3 compound.
In certain embodiments of the methods of the invention, the subject is a mammal. In preferred embodiments, the subject is human.
In one embodiment, the vitamin D3 compound is administered to the subject using a pharmaceutically-acceptable formulation. In certain embodiments, the vitamin D3 compound is advantageously administered in combination with a pharmaceutically acceptable diluent or carrier.
In another embodiment, the pharmaceutically-acceptable formulation provides sustained delivery of the vitamin Dj compound to a subject for at least four weeks after the pharmaceutically-acceptable formulation is administered to the subject. In accordance with the methods of the invention, the vitamin D5 compound is administered orally, intravenously, topically, or parentcrally. Although dosages may vary depending on the particular indication, route of administration and subject, the 20- methyl Gemini vitamin D3 compounds of the invention are administered at a concentration of about 0.001 μg to about 100 μg/kg of body weight. In certain embodiments, the 20-methyl Gemini vitamin D3 compounds of the invention are administered at a concentration of about 5 μg/kg of body weight.
Another aspect of the invention provides a pharmaceutical composition for use in the treatment of osteroporosis, comprising a therapeutically effective amount of a 20- methyl Gemini vitamin D3 compound of the invention and a pharmaceutically acceptable diluent or carrier. Preferred compounds of this aspect of the invention include (20S)-l,25-Dihydroxy-20-(5,5,S-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trif!uoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-lα-Fluoro-25-hydroxy~20- (5,5,5-trifluoro-4-riydiOxy-4-trifiuoromethyl-pent-2-ynyl)-cholecalciferol (6). An especially preferred compound of the invention is (20S)-l,25-Dihydroxy-20-(5,5,5- trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (I). In yet another aspect, the invention provides a pharmaceutical composition for use in the treatment of secondary hyperparathyroidism comprising a therapeutically effective amount of a 20-methyl Gemini vitamin D3 compound of the invention and a pharmaceutically acceptable diluent or carrier. Preferred compounds of this aspect of the invention include (20S)~l,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl)choIecalciferol (2); (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5- trifluoro-4-hydroxy-4-triflυoromethyl-pent-2-enyl]-cholecalciferoJ (3); (20R)- 1 ,25- Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol (4); and (20R)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-tri fluoro-4-hydroxy-4-trifluoromethyl- pent-2-enyl]-cholecalciferol (5). A particularly preferred compound is (20S)-1 ,25- Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyi-pent-2- enyl)cholecalciferol (2).
In another aspect, the invention provides a packaged formulation for use in the treatment of osteoporosis, comprising a pharmaceutical composition comprising a 20- methyl Gemini vitamin D3 compound and instructions for use in the treatment of osteoporosis in accordance with the methods of the invention. Preferred compounds of this aspect of the invention include (20S)-lJ25-Dihydroxy-20-(5,5,5-trifluoro-4- hydroxy^-trifluoromethyl-pent^-ynyO-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20- [(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-lα-Fluoro-25-hydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trif]uoromethyl-pcnt-2- ynyl)-cholecalciferol (6). An especially preferred compound of the invention is (20S)- l)25-Dihydroxy-20-(5,5,5-triflxioro-4-hydroxy-4-trif]uoromethyI-pent-2-ynyl)- cholecalcifcrol (1).
Yet another aspect of the invention provides a packaged formulation for use in the treatment of secondary hyperparathyroidism, comprising a pharmaceutical composition comprising a 20-methyl Gemini vitamin D3 compound and instructions for use in the treatment of secondary hyperparathyroidism in accordance with the methods of the invention. Preferred compounds of this aspect of the invention include (20S)- l ,25-Dihydroxy-20-((2Z)-5,5,5-trifluoro-4-hydroxy-4-trifluororaethyl-pent-2- enyOcholecalciferol (2); (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-enyl]-cholecalciferol (3); (20R)-l,25-Dihydroxy-20-(5,5J5- trifluoro-4-hydroxy-4-triflυoromethyl-pent-2-ynyl)-choIecalciferol (4); and (20R)-1, 25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-cnyl]- cholecalciferol (5), A particularly preferred compound is (20S)-1,25-Dihydroxy-20- ((2Z)-5,555-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl)cho5ecalciferol (2). 4. PHARMACEUTICAL COMPOSITIONS
The invention also provides a pharmaceutical composition, comprising an effective amount a vitamin D3 compound described herein and a pharmaceutically acceptable carrier. In a further embodiment, the effective amount is effective to treat a vitamin D3 associated state, as described previously.
In an embodiment, the vitamin D3 compound is administered to the subject using a pharmaceuticaily-aceeptable formulation, e.g., a pharmaceutically-acceptable formulation that provides sustained delivery of the vitamin D3 compound to a subject for at least 12 hours, 24 hours, 36 hours, 48 hours, one week, two weeks, three weeks, or four weeks after the pharmaceutically-acceptable formulation is administered to the subject.
In certain embodiments, these pharmaceutical compositions are suitable for topical or oral administration to a subject. In other embodiments, as described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqυeous solutions or suspensions), tablets, boluses, powders, granules, pastes; (2) parenteral administration* for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; or (5) aerosol, for example, as an aqueous aerosol, liposomal preparation or solid particles containing the compound.
In certain embodiments, the subject is a mammal, e.g., a primate, e.g., a human. The methods of the invention further include administering to a subject a therapeutically effective amount of a vitamin D3 compound in combination with another pharmaceutically active compound. Examples of pharmacuctically active compounds include compounds known to treat autoimmune disorders, e.g., immunosuppressant agents such as cyclosporin A, rapamycin, desoxyspergualine, FK 506, steroids, azathioprine, anti-T cell antibodies and monoclonal antibodies to T cell subpopulatioπs. Other pharmaceutically active compounds that may be used can be found in Harrison's Principles of Internal Medicine, Thirteenth Edition, Eds. T.R. Harrison er a/. McGraw- Hill N.Y., NY; and the Physicians Desk Reference 50th Edition 1997, Oradell New Jersey, Medical Economics Co., the complete contents of which are expressly incorporated herein by reference. The angiogenesis inhibitor compound and the pharmaceutically active compound may be administered to the subject in the same pharmaceutical composition or in different pharmaceutical compositions (at the same time or at different times).
The phrase "pharmaceutically acceptable" is refers to those vitamin D3 compounds of the present invention, compositions containing such compounds, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically-acceptable carrier" includes pharmaceutically- acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipicnt, solvent or encapsulating material, involved in carrying or transporting the subject chemical from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (S) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflowcr oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (I3) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine telraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Compositions containing a vitamin D3 compound(s) include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent. Methods of preparing these compositions include the step of bringing into association a vitamin D3 compound(s) with the carrier and, optionally, one or more accessory ingredicnis. In general, the formulations are prepared by uniformly and intimately bringing into association a vitamin D3 compound with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. Compositions of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a vitamin D3 compound(s) as an active ingredient. A compound may also be administered as a bolus, electuary or paste.
In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylccllulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol rnonostcarate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent.
The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration of the vitamin D3 compound(s) include pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethy] carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3- butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
Suspensions, in addition to the active vitamin D3 compound(s) may contain suspending agents as, for example, cthoxylatcd isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which maybe prepared by mixing one or more vitamin D3 compound(s) with one or more suitable noπirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
Compositions of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a vitamin D3 compound(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active vitamin D3 compound(s) may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams and gels may contain, in addition to vitamin D3 compound(s) of the present invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to a vitamin D3 compound(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellanls, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. The vitamin D3 compound(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound, A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.
Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically-acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular compound, but typically include nonionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid., lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.
Transdermal patches have the added advantage of providing controlled delivery of a vitamin D3 compound(s) to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absorption enhancers can also be used to increase the flux of the active ingredient across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the active ingredient in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more vitamin D3 cornpound(s) in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials;, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in tum, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms arc made by forming microencapsule matrices of vitamin D3 compound(s) in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthocsters) and poly(aπhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
When the vitamin D3 compound(s) are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically-acceptablc carrier.
Regardless of the route of administration selected, the vitamin D3 compound(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.
Actual dosage levels and time course of administration of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. An exemplary dose range is from 0.1 to 10 mg per day.
A preferred dose of the vitamin D3 compound for the present invention is the maximum that a patient can tolerate and not develop serious hypercalcemia. Preferably, the vitamin D3 compound of the present invention is administered at a concentration of about 0.001 μg to about 100 μg per kilogram of body weight, about 0.001 - about 10 μg/kg or about 0.001 μg - about 100 μg/kg of body weight. Ranges intermediate to the above-recited values are also intended to be part of the invention.
5. SYNTHESIS OF COMPOUNDS OF THE INVENTION
Compounds of the invention can be synthesized by methods described in this section, the examples, and the chemical literature.
Schemes 1-9 below depict the reaction steps for the synthesis of the highly fluoriπated-20-metb.yl gemini vitamin D3 compounds of the invention.
Scheme 1 shows the synthetic route for the production of the diol 15 and its epimer, 16. Alcohol 7 was protected with a silyl group to compound 8, then cyclopropanated to provide cyclopropane 9. Conversion of the ester to the aldehyde was accomplished over two steps to provide 11. Chain elongation using a modified Wittig- Horner reaction provided 12. Reduction of the double bond and cyclopropane opening liberated ester 13, which was reduced to alcohol 14. Deprotection and chromatographic separation yielded intermediate 15 and its epimer 16.
Scheme 1
Figure imgf000029_0001
Figure imgf000030_0001
Scheme 2 shows the chain elongation of 15 to triol 21. Oxidation of the primary alcohol of 15 provided the corresponding aldehyde 17 and chain elongation provided alkyne 18. Protection of the tertiary alcohol to compound 19 was followed by lithiation of the alkyne and reaction with hexafluoroacetone to produce 20. Silyl group deprotection provided triol 21.
Scheme 2
Figure imgf000030_0002
Scheme 3 shows that 21 is oxidized to ketone 22 which is amenable to Wittig- Homer coupling with an appropriate phosphine oxide. Further reduction of the alkyne of 21 was carried out to form either the cis or trans olefins, 23 and 25, respectively. Oxidation provided ketones 24 and 26. Scheme 3
Figure imgf000031_0001
Scheme 4 shows the Wittig-Horner coupling of ketone 22 with phosphine oxide 27 in the presence of base to provide the corresponding coupled product Scheme 4 also shows the coupling of hydroxyl protected ketone 28 with phosphine oxide 29 to provide the corresponding coupled product. Dcprotection of the silyl group(s) with tetxabutyl ammonium fluoride afforded compounds 1 and 6.
Scheme 4
Figure imgf000031_0002
Figure imgf000032_0001
29
Scheme 5 shows the Wittig-Horner coupling of ketone 24 with phosphine oxide 27 in the presence of base to provide the corresponding coupled product. Deprotection of the silyl group(s) with tetrabutyl ammonium fluoride afforded compound 2.
Scheme 5
Figure imgf000032_0002
Scheme 6 shows the Wittig-Horner coupling of ketone 26 with phosphine oxide 27 in the presence of base to provide the corresponding coupled product. Deprotection of the silyl group(s) with tetrabutyl ammonium fluoride afforded compound 3.
Scheme 6
Figure imgf000032_0003
Vitamin D3 compounds 4 and 5 were synthesized in accordance with the reactions described in schemes 1-6 above. The starting material for the production of 4 and 5 was 16. Scheme 7 shows the chain elongation of 16 to triol 34. Oxidation of the primary alcohoS of 16 provided the corresponding aldehyde and chain elongation provided alkyne 31. Protection of the tertiary alcohol was followed by lithiation of the alkyne and reaction with hexafluoroacetone to produce 33. Silyl group deprotection provided triol 34.
Scheme 7
Figure imgf000033_0001
Scheme 8 shows that 34 is oxidized to form ketone 35 that is amenable to Wittig-
Horner coupling with an appropriate phosphine oxide. Further reduction of the alkyne of 34 was carried out to form the trans olefin 36. Oxidation and hydroxyl protection provided ketone 38.
Scheme 8
Figure imgf000033_0002
Figure imgf000034_0001
Scheme 9 shows the coupling of ketones 35 and 38 with phosphine oxide 27 to provide compounds 4 and 5.
Scheme 9
Figure imgf000034_0002
Chiral syntheses can result in products of high stereoisomer purity. However, in some cases, the stereoisomer purity of the product is not sufficiently high. The skilled artisan will appreciate that the separation methods described herein can be used to further enhance the stereoisomer purity of the vitamin D3-epimer obtained by chiral synthesis. Any novel syntheses, described herein, of the compounds of the invention, and of intermediates thereof, are also intended to be included within the scope of the present invention.
EXEMPLIFICATION OF THE INVENTION
The invention is further illustrated by the following examples which should in no way should be construed as being further limiting.
Synthesis of Compounds of the Invention Experimental
All operations involving vitamin D3 analogs were conducted in amber-colored glassware in a nitrogen atmosphere. Tetτahydrofuran was distilled from sodium- benzophenone ketyl just prior to its use and solutions of solutes were dried with sodium sulfate. Melting points were determined on a Thomas-Hoover capillary apparatus and arc uncorrected. Optical rotations were measured at 25 0C. 1H NMR spectra were recorded at 400 MHz in CDCI3 unless indicated otherwise. TLC was carried out on silica gel plates (Merck PF-254) with visualization under short-wavelength UV light or by spraying the plates with 10% phosphomolybdic acid in methanol followed by heating. Flash chromatography was carried out on 40-65 μm mesh silica gel. Preparative HPLC was performed on a 5x50 cm column and 15-30 μm mesh silica gel at a flow rate of 100 mL/min.
EXAMPLE 1
Synthesis of (20S)-1, 25~Dihydroxy~20-(5,5)5-tήflιιoro-4-hydroxy-4-trifluoromethyl- pent-2-ynyl)cholecalciferol (1)
(JR, 3aR, 4S, 7aR)-4-(tert-Bυty!-dimethyI-silanyloxy)-l-[3-(tert-butyl-dimethyl- si)anyloxy)-l-methylene-propyl]~7a-methyl-octahydro-indene (8)
Figure imgf000035_0001
7 «
A 250 ml round bottom flask equipped with stir bar, Claisen adapter with rubber septum and nitrogen sweep was charged with 17.53 g (51.77 mmol) of 3-[(1R, 3aR, 4S, ya^-^ζtert-butyl-diniethyl-silanyloxy^Ta-iTiethyl-octahydro-inden-l-yll-but-S-en-l-ol and 75 ml of dichloromethane. A 7.05 g (103.54 mmol) imidazole was added followed by 9.36 g (62.124 mrnol) of t-butyldimethylsilyl chloride. The mixture was stirred for 2.5h. The mixture was then diluted with 100 ml of water and extracted four times with
50 ml of dichloromethane. The combined organic layers were dried over Na24 and evaporated.
The oil residue was chromatographed on column (400 cm3) using hexane, hexane:ethyl acetate (50:1 , 25:1) as mobile phase and collecting ca. 40 ml fractions to give 22.32 g (95%) of product as colorless oil.
1H NMR (CDCl3): 4.87(1H, s), 4.8O(1H, s), 4.02(1H, br s), 3.67(2H, t, J=7.3 Hz), 2.34- 2.14(2H, m), 2.06-2.00(1H, m), 1.85-1.27(911, m), 1.20-1.08(2H, m), 0.89(18H, s), 0.79(3H5 s), O.O5(6H, s), O.02(3H, s), 0.01(311, s)
2-[2-(tert-ButyI-dimethyl-silany]oxy)-ethyl]-2-[(lS, 3aR, 4S, 7aR)-4-(tert-butyl- dimethyl-silanyloxy)-7a-methyl-octahydro-inden-l-yl}-cyclopropanecarboxylic acid
Figure imgf000036_0001
S 9 A 250 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 10.00 g (22.08 mrnol) of (IR, 3aR, 4S, 7aR)-4-(tert- butyl-dimethyl-silanyloxy)-l-[3-(tert-butyl-dimethyl-silanyloxy)-l-methylene-propyl]- 7a-methyl-octahydro-indene, 200 mg Of Rh2(OAc)4 and 40 ml of dichloromethane. A solution of 5.304 g (46.486 mmol) of ethyl diazoacetate in 30 ml of dichloromethane was added dropwise (12 ml/h) at room temperature.
The reaction mixture was concentrated in vacuo and the remaining residue was filtrated on column (200 cm3) using hexaneiethyl acetate (3 :1) as mobile phase. The solvent was evaporated and the oil residue was chromatographed on column (250 cm3) using hcxane:ethyl acetate (25: 1, 10: 1 and 5: 1) as mobile phase to give 8.44 g (71%) of products as a mixture of isomers. {2-I2-(tert-Butyl-dimethyl-silaπyIoxy)-ethyl]-2-[(lS, 3aR, 4S5 7aR)-4-(tert-butyl- dmiethyl-silanyloxy^Ta-methyl-octahydro-inden-l-ylj-cyclopropyO-methanol (10)
Figure imgf000037_0001
Λ 50 ml round bottom flask equipped with stir bar and CIaisen adapter with rubber septum was charged with 4.140 g (7.682 rnmol) of 2-[2-(tert~butyl-dimethyl- silanyIoxy)-ethyl]-2-[(lS, 3aR, 4S, 7aR)-4-(tert-butyl-dimethyl-si]anyIoxy)-7a-mcthyl- octahydro-inden-l-yl]-cyclopropanecarboxylic acid ethyl ester and 20 ml of dichloromethane, The reaction mixture was cooled to -7O0C and 10.0 ml (15.0mmol) of 1 -5M DIBAL-H in toluene was added dropwise during 45 min. The reaction was stirred at -70° C for 1 h and then 5 ml of saturated solution of ammonium chloride was added dropwise.
The mixture was dissolved by the addition of 100 ml of water and 50 ml of IN HCl, extracted three times with 50 ml of ethyl acetate, dried over Na2SO4 and evaporated. The oil residue was chromatographed on column (200 cm" ) using hexane:ethyl acetate (10:1, 3: 1) as mobile phase. The fractions containing product were pooled and evaporated to give 3.610 g, (94%) of products (mixture of isomers) as colorless oil.
2-[2-(tert-Butyl-dimethyl-sllanyIoxy)-ethyl]-2-((lS, 3aR54S, 7aR)-4-(tert-butyl- dimcthy]-silanyioxy)-7a-metbyl-octahydro-inden-l-yl]-cyclopropanecarbaldehyde
Figure imgf000037_0002
10 11
A 250 ml round bottom flask equipped with stir bar and CIaisen adapter with rubber septum was charged with 6.074 g (28.178 mmol) of pyridinium chlorochromate, 7.00 g of celite and 100 ml of dichloromethane. A 6.970 g (14.027 mmol) of {2-[2-(tert- butyl-dimethyl-silanyloxy)-ethyl]-2-[(lS, 3aR, 4S, 7aR)-4-(tert-butyl-dimethyl- silanyloxy)-7a-methyl-octahydro-inden-l-y]]-cyclopropyl} -methanol in 10 ml of dichloromethane was added dropwise and mixture was stirred in room temperature for Ih.
The reaction mixture was filtrated through column with silica gel (200 cm3) and celite (2 cm) and using dichloromethane as a mobile phase, The fractions containing product were pooled and evaporated to give oil (ca. 5.71 g). Product was used to the next reaction without purification.
3-{2-[2-(tert-Butyl-diraethyI-siIanyloxy)-etήyll-2-{(lS, 3aR, 4S, 7aR)-4-(tcrt-butyl- dimethyl-silanyloxy)-7a-methyl-octahydro-inden-l-yl]-cyc]opropyl}-acrylic acid
Figure imgf000038_0001
A 250 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 80 ml of toluene and 35.0 ml (35.0 mmol) of IM potassium tert-hutoxiάe in tetrahydrofurane was added. A 7.850 g (35.015 mmol) of triethyl phosphonoacetate in 5 ml of toluene was added dropwise at ca. 5 0C. The mixture was stirred at room temperature for Ih. Then the mixture was cooled to -15 0C and crude (ca. 11.54 mmol) 2-[2-(tert-butyl-dimethyl-silanyloxy)-ethyl]-2-[(lS, 3aR, 4S, 7aR)-4-(tert-butyl-dimethyl-silanyloxy)-7a-methyI-octahydro-inden-l-yl]- cyclopropanecarbaldehyde in 5 ml of toluene was added and stirring was continued at - 10° C for 3h.
The reaction mixture was quenched with 10 ml of aqueous saturated solution of ammonium chloride, diluted with 100 ml of saturated solution of ammonium chloride and extracted four times with 50 ml of toluene and then 50 ml of ethyl acetate. The organic layer was washed with 50 ml of brine, dried and evaporated. The residue was purified over silica gel (200 cm3) using hexanerethyl acetate
(20:1) as a mobile phase to give 5.750 g (88%) of products (mixture of isomers). 7-(tert-ButyI-dimethyI-silanyloxy)-5-[(lR, 3aR, 4S, 7aR)-4-(tert-butyl-dimetbyl- siIanyIoxy)-7a-methyl-octahydro-inden-l-yl]-5-methyl-heptanoic acid ethyl ester
Figure imgf000039_0001
12 » A 5.750 g (10.177 mmol) oF3-{2-[2-(tert-butyl-dimethyl-silanyloxy)-ethyl]-2-
[(1S, 3aR, 4S, 7aR)-4-(tert-butyl-diτnethyl-silanyloxy)-7a-τnethyl-octahydro-inden-l- ylj-cyclopropyl) -acrylic acid ethyl ester was hydrogenated over 1.60 g of 10% Pd/C in 40 ml of ethanol at room temperature and atmospheric pressure of hydrogen. The reaction was monitoring by TLC (hexane:ethyl acetate-50:l). After 18h the catalyst was filtered off and solvent evaporated. The residue was purified over silica gel (300cm3) using hexane:ethyl acetate (100: 1 , 50: 1 , 20: 1) as a mobile phase to give 5.150 g (89%) of products (mixture of isomers).
8-(tert-Butyl-dimethyl-silanyloxy)-6-[(lR, 3aR, 4S, 7aR)-4-(tert-butyI-dimethyl- si!anyloxy)-7a-methyl-octahydro-inden-t-yll-2,6-dimethyi-octan-2-oI (14)
Figure imgf000039_0002
13 >d
A 250 ml round bottom flask equipped with stir bar, Claisen adapter with rubber septum was charged with 5.110 g (8.980 mmol) of 7-(tert-butyl-dimethyl-silanyIoxy)-5- [(1R, 3aR, 4S, 7aR)-4-(tert-butyl-dimethyl-silanyloxy)-7a-methyl-octahydro-inden-l- yl]-5~methyl-lieptanoie acid ethyl ester ester and SO ml of diethyl ether. The solution was cooled in ace-water bath and 17.4 ml (54.3 mmol) of 3.12M solution of mcthylmagnesium bromide in diethyl ether was added dropwise. After completion of the addition the mixture was stirred at room temperature for 2.5h then cooled again in an ice bath. A 10 ml of saturated solution of ammonium chloride was added dropwise. The resulting precipitate was dissolved by the addition of 50 ml of saturated solution of ammonium chloride. The aqueous layer was extracted three times with 100 ml of ethyl acetate. The combined organic layers were dried (NaOSO4) and evaporated. The product was used to the next reaction without farther purification.
3-[(1R, 3aR, 4S, 7aR)-4-(terr-ButyI-dimethyl-siIanyloxy)-7a-methyl-octahydro-
Figure imgf000040_0001
14 15
A 50 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with crude (ca. 8.98mmol) 8-(tert-buryl-dimethyl- silanyloxy)-6-[4-(tert-butyl-dimethyl-silanyloxy)-7a-methyl-octahydro-inden-l-yl]-2,6-- dimethyl-octan-2-ol, 10 ml of tetrahydrofurane and 15.0 ml (15.0 mmol) of IM tetrabutylammoπium fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 2.5h.
The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1: 1) and 50 ml of brine, dried over Na2SO,) and evaporated.
The oil residue was chromatographed four times on columns (400cm3) using hexane;ethyl acetate (1 : 1) as a mobile phase to give: Is1 - 1.456g (low polar epimer); 2nd - 0,852g, (mixture of epimers); 3rd - 1.132g (more polar epimer); All products 3.44Og (88% two steps)
Low polar epimer: (3S)-3-[(lR, 3aR, 4S, 7aR)-4-(tert-Butyl-dimethyl-siIanyloxy)- 7a-methyl-octahydro-inden-1-yl]-3,7-dimethyl-octane-l,7-diol
Figure imgf000040_0002
[α] 3^ = +26.1° c=0.44, CHCl3
1H NMR (CDCl3): 3.9O(1H, br s), 3.67(2H, br t, J=S-I Hz),2.06-1.99(IH, m), 1.87- ) 50(4H, m), I .73(2H, t, J=7.9 Hz), 1.40-1.06(14H, m), 1.22(6H, s), 1.06(3H, s), 0.95(3H, s), 1.95-O.82(1H, m), 0.88(9H, s), 0.00(3H, s), -0.01(3H, s) 13C NMR (CDCl3): 71.03, 69.58, 59.79, 57.32, 52.99, 44.78, 43.81 , 41.64, 41.58, 40.26, 38,68, 34.37, 29.4S. 29.36, 25.86, 23.49, 22.78, 21.72, 18.18, 18.09, 37.78, 16.78, -4.70, -5.07
MS ΪIRES Calculated for: C26H52O3Si [M+Naf 463.3578
Observed: [M+Naf 463.3580
More polar epimer: (3R)-3-[(lR? 3aR, 4S, 7aR)-4-(tert-Butyl-d.methyI-silanyloxy)- 7a-methyl-octahydro-inden-l-yl)-3,7-dimethyl-octane-l,7-diol
Figure imgf000041_0001
[αj » = +22.7° c=θ'.44, CHCl3
1H NMR (CDCl3): 3.99-3.97(1H, m), 3.65-3.61 (2H, m), 1.97(1H, br d, J=I 2,3 Hz),
1.84-1 J2(lH, m) " 1.66-1.50(6H, m), 1.45-1.15(14H, m), l ,21(6H, s), 1.05(3H, s),
0.95(3H3 s), 0.87(9H, s), -0.01(3H, s), -0.02(3H, s) 13C NMR (CDCl3): 71.05, 69.57, 59.47, 57.46, 53.02, 44.87, 43.90, 41.83, 41.61 , 39 99,
38.93, 34.37, 29.43, 29.42, 25.87, 23 42, 22.84, 22.12, 18.57, 18.09, 17.81, 16.79, -4.69,
-5.06
MS HJRES Calculated for: C26H52O3Si [M+Naf 463.3578
Observed: [M+Naf 463.3575
(3S)-3-[(lR, 3aR, 4S, 7aR)-4-(tert~Butyi~dimethyl-silaπyloxy)-7a-methyl-octahydro- 1 indcn-l-yl]-7~hydroxy-3,7-dimethyl-oetanaI (17)
Figure imgf000041_0002
JS 17
A 25 ml round bottom flask equipped with stir bar and Claiscn adapter with rubber septum was charged with 1.572 g (7.292 mmol) of pyridinium chlorochromate, 1.60 g of celite and 25 ml of dichloromethane. A 1.607 g (3.646 mmol) of (3S)-3-[(lR, 3 aR, 4S, 7aR)-4-(tert-butyl-dimethyl-silanyloxy)-7a-rnethy]-octahydro-inden- 1 -yl]-3 ,7- dimethyl~octane-l,7-diol in 6 ml of dichloromethane was added dropwise and mixture was stirred at room temperature for Ih 45 min and additional portion 300 mg (1.392 mmol) of pyridinium chlorochromate was added. The reaction was stirred for next Ih 15 min.
The reaction mixture was filtrated through column with silica gel (50 cm3) and celite (1 cm) using dichloromethane, dichloromcthane:ethyl acetate (4:1). The fractions containing product were pooled and evaporated to give 1.58 g of product as yellow oil. The product was used to the next reaction without farther purification.
(6S)-6-[(lR, 3aR, 4S, 7aR)-4-(tert-Butyl-dimethyl-si]any}oxy)-7a-methyl-octahydro- inden~l-yl)-2,6-dimethyl~non-8-yn-2-ol (18)
with 4S, 7aR)-4-
Figure imgf000042_0001
(tert-butyl-dimethyi-silanyloxy)-7a-methyl-octahydro-inden-l-yl]-7-hydroxy-3,7- dimsthyl-octanal and 30 ml of methanol. A 1.416 g (7.37 mmol) of 1 -diazo-2-oxo- propyl)-phosphonic acid dimethyl ester in 3 ml of methanol was added and the resulting mixture was cooled in an ice bath. A 1.416 g (10.245 mmol) of potassium carbonate was added and the reaction mixture was stirred in the ice bath for 30 min and then at room temperature for 3h. A 100 ml of water was added and the mixture was extracted three times with 80 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The oil residue was chromatographed on column (250 cm3) using hexane:ethyl acetate (7:1) as mobile phase. Fractions containing product were pooled and evaporated to give 1.310 g (83%, 2 steps) of product as colorless oil.
[αj 3 D" = +15.7° c=0.61 , CHCl3
1H NMR (CDQ3): 3.98(111, br s), 2.28(2H, d, J=2.1 Hz), 1.95-1.91(2H, m), 1.7S(IH, dt, J= 13.4, 3.8 Hz), 1.68-1.62(1H, m), 1.58-1.48(6H7 m), 1.44-U7(15H, m), 1.22(6H, s), 1.04QH, s), 1.00(3H, s), 0.93-0.83(IH, m), 0.88(9H5 s), -0.00(3H, s), -0.01(3H, s) 13C NMR (CDCl3): 83.09, 71.03, 69.84, 69.64, 56.68, 52.95, 44.80, 43.71, 41.31, 40.21, 39.28, 34.33, 29.44, 29.29, 28.80, 25.85, 22.74, 22.69, 22.18, 18.14, 18.05, 17.73, 16.68, -4.77, -5.13 1541
S HRES Calculated for: C27H50O2Si [M+Na]+ 457.3472
Observed: [M+Na]+ 4573473
(IR, 3aR, 4S, 7aF)-4-(tert-Butyl-(iimethyI-silanyloxy)-l-[(lS)-l,5-djmethyl-l-prop- 2-ynyl-5-trι!methylsilanyloxy-hexyI]-7a-πiethyl-octahydro-indene (19)
Figure imgf000043_0001
J8 19
A 50 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 1.300 g (2.990 mmol) of (6S)-6-[(lR, 3aR, 4S, 7aR)-4-(tert- butyl-dimethyl-silahy)oxy)-7a-methyl-octahydro-inden-l -yϊ]-2,6-dimethyl-non-8-yn-2- ol and 25 ml of dichloromethanc. A 2.00 ml (13.63 mmol) of l-(trimethylsilyl)imidasole was added dropwise. The mixture was stirredoat room temperature for 1 h,
A 100 ml of water was added and the mixture was extracted three times with 80 ml of hexane, dried over Na2SO4 and evaporated. The oil residue was chromatographed on column (75 cm3) using hexane:ethyl acetate
(25:1) as mobile phase. Fractions containing product were pooled and evaporated to give
1.409 g (93%) of product as colorless oil.
1H NMR (CDCl3): 3.98(1H, br s), 2.27(2H, d, J=2.9 Hz), 1.97-1.91(2H, m), 1.82- 1.75(lH, m), 1.69- 1.62(1 H, m), 1.59-1.50(2H, m), 1.42-1.20(12H1 m), 1.20(6H, s), 1.05(3H, s), 1.00(3H, s), 0.93-0.85(1H5 m), O.88(9H, s), 0.10(9H3 s), 0.00(3H, s), - 0.01(3H, s)
(6S)-6-|(lR, 3aR^ 4S, 7aR)-4-(tert-Butyl-dimethyl-silanyloxy)-7a-methyl-octahydro- inden-l-yπ-ljUl-trifluoro-θjlO-dimethyl^-trifluoromethyl-lO-trimethylsilanyloxy- undec-3-yn-2-ol (20)
Figure imgf000044_0001
19 ZO
Λ two neck '50 ml round bottom flask equipped with stir bar, Claisen adapter with rubber septum and funnel (with cooling bath) was charged with 1.390 g (2.742 mmol) Of (IR, 3aR, 4S, 7aR)-4-(tert-butyl-dimethyl-silanyloxy)-l-[(15)-l,5-dimethyl-l- prop-2->τiyl-5-trimethyIsilanyloxy-hexyl]-7a-methy]-octahydro-indene and 30 ml of tetrahydrofurane. The funnel was connected to container with hexafiuoroacctone and cooled (acetone, dry ice). The reaction mixture was cooled to -7O0C and 5.00 ml (8.00 mmol) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. After 30 min hεxaflυoroacetone was added (the contener's valve was opened three times). The reaction was stirred at -7O0C for 2h then 5.0 ml of saturated solution of ammonium chloride was added.
The mixture was dissolved by the addition of 100 ml of saturated solution of ammonium chloride and extracted three times with 80 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The oil residue was chromatographed twice to remove a large amount of polymer compounds. The first column (100 cm3) using hexane:ethyl acetate (10:1) as mobile phase. The second column (100 cm3) using hexanexthyl acetate (25:1, 15:1) as mobile phase. Fractions containing product were pooled and evaporated to give 1.959 g of colorless oil. Product was used to the next reaction without farther purification.
(6S)-l,l,l-Trifiuoro-6-[(lR, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-inden- l-yl]-6,10-dimethyl-2-trifluoromethyl-undec-3-yne-2,lO-dio) (2l)
Figure imgf000044_0002
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with crude (ca. 2.74 mmol) (6S)-6-[(lR, 3aR, 4S, 7aR)-4- (tert-butyl-dimethyl-silanyloxy)-7a-methyl-octahydro-inden-l -yl]-l , 1 , 1 -trifiuoro-6,10- dimethyl-2-trifluoromethyl-10-trimethylsilanyloxy-undec-3-yn-2-ol and 12.0 ml (12.0 mmol) of IM tetrabutylammoniutn fluoride in tetrahydrofurane and reaction was stirred at 7O0C. After ISIl new portion 5.0 ml of IM terrabutylammonium fluoride in tetrahydrofiirane was added. The reaction mixture was stirred at 700C for next 80h. The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1:1) and 50 ml of brine and dried over NaϊSCU and evaporated.
The oil residue was chromatographed on column (200 cm3) using hexanetethyl acetate (3:1 , 2: 1) as mobile phase. The fractions containing product were pooled and evaporated. The residue was crystallized from hexane-ethyl acetate to give 917 mg
(69%, two steps) of product as a white crystal.
m.p. 146-1470C
[α] 3^ = -3.5° c=0.'43, CHCl3
1H NMR (CDCl3): 4.08(1H5 br s), 2.45(1H, AB, J=I 7 Hz), 2.36(1H, AB, J=17 Hz),
1.98-1.92(1H, m), 1.85-1.74(2H, m), 1.67-1.18(18H, m), 1.25(6H, s), 1.07(3H, s), 1.02(3H, s)
MS HRES Calculated for: C24H36F6O3 [M+Na]+ 509.2461
Observed: [M+Na]" 509.2459
(IR, 3aRs 4S, 7aR)-7a-MethyI-l-{(lS)-6^6-trifluoro-5-hydroxy-l-(4-hydroxy-4- methyl-pentyl)'-l-niethyl-5-triiluoromethyl-hex-3-ynyl]-octahydro-mdcn-4-oaε (22)
Figure imgf000045_0001
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 300 mg (0.617 mmol) of (6S)-I ,l5l-trifluoro-6-[(lR) 3aR, 4S, 7aR)~4-hydroxy-7a-methyl-octahydro-indcn-l -yl]-6,10-dimethyl-2- trifluoromethyi-undec-3-yne-2,10-diol and 10 ml of dϊchloromethanc. A 696 mg (1.851 mmol) of pyridinium dichromate and 710 mg of celite were added and mixture was stirred in room temperature for 3h.
The reaction mixture was filtrated through column with silica gel (50 cm3) and celite (2 cm) and using dichloromethane : ethyl acetate (4:1) as a mobile phase. The fractions containing product were pooled and evaporated to give yellow oil. The product was used to the next reaction without farther purification. (20S)-l,25-DiUydroxy-20-(5,5,5-trjfluoro-4-hydroxy-4-trifluoromethyl-pent-2- : yπyl)cholecalciferol (1)
Figure imgf000046_0001
A 25 ml round bottom flask equipped with stir bar and Cϊaisen adapter with rubber septum was' charged with 1.798 g (3.084 mmol) of (1 S,5R)-1 ,5-bis-((^^~ butyldimethyl)silaήyloxy)-3-[2-(diphenylfosphinoyl)-eth-(Z)-ylidenc]-2-methylene- cyclohexane and ]2 ml of tetrahydrofurane. The reaction mixture was cooled to -780C and 1.9 ml (3.04 mmol) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. The resulting deep red solution was stirred at -780C for 20 min and crude (ca 0.617mmol) (IR, 3aR, 4S, 7aR)-7a-methyl-l-[(lS)-6,6,6-trifluoro-5-hydroxy-l-(4- hydroxy-4-methyl-pcntyl)-l-methyl-5-trifluoromethyl-hex-3-ynyl]-octahydro-inden-4- one was added dropwise in 1.5 ml of tetrahydrofurane. The reaction mixture was stirred for 5h and then the, bath was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine. The water fraction was extracted three times with 50 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The oil residue was chromatographed on column (75 cm3, protected from light) using hexane:ethyl acetate (5: 1) as mobile phase. Fractions containing product were pooled and evaporated to give colorless oil (293 mg) which was treated with 5 ml of IM tetrabutylammonium fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 4Oh.
The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1 :1) and 50 ml of brine, dried over Na2SC^ and evaporated.
The oil residue was chromatographed on column (50 cm3, protected from light) using ethyl acetate as mobile phase. Fractions containing product were pooled and evaporated to give product as colorless oil. Oil was dissolved in methyl acetate and evaporated (4 times) to give 190 mg (50% three steps) of product as white foam.
Figure imgf000046_0002
UV λmax (EtOH): 205.50 nm (ε 16586), 266.00 nm (ε 14319) 1H NMR (CDCl3):! 6.36(1 H, d, J=I 1.3 Hz), 6.23QH, br s), 6.00(1H, d, J=I 1.1 Hz), 5.32(1H, s), 4.98(1H, s), 4.43(1H, dd, J=7.75 4.3 Hz), 4.25-4.20(1H, m), 2.82-2.79(1H, m), 2.59(1 H, dd, J=*13.1, 3.1 Hz), 2.44(1H, AB, J=I 7.2Hz), 2.37(1H, AB, J=I 7.2 Hz), 2.3O(1H, dd, J=I 3.2, 6.2 Hz, ), 2.06-1.87(4H, m), 1.72-1.36(1 IH, m), 1.26- 1.21(1H, m), 1.24(6H, s), 0.99(3.H, s), 0.64(3H, s)
13C NMR (CDCl3): 147.48, 142.29, 133.16, 124.72, 121.32(q, J=142.7 Hz), 1 17.59, 1 1.68, 90.08, 72.62, 71.39, 70.73, 66.89, 57.28, 56.52, 46.65, 45.18, 43.20, 42.81, 41.04, 40.89, 40.03, 29.79, 29.35, 28.95, 23.45, 22.86, 22.60, 21 ,84, 17.77, 14.93 MS HRES Calculated for: C33H46FeO4 [M-HNa]+ 643.3192
Observed: ■ y [M+Na]f 643.3192
EXAMPLE 2
Synthesis ofζlOSyia-Fluoro-IS-hydroxy^O^S^^-trifluoro-^hydroxy-^ trijluorotnethyl-pent-l-ynyiy-cholecalciferol (6)
(IR, 3aR, 4S, 7aR)-7a-Methyl-l-l(lS)-6,6,6-trifIuoro-l-methyl-l-(4-metfayl-4- trimethylsiJanyloxy-pentyl)-5-trifluoroniethyl-5-trimethylsiIanyloxy-hex-3-ynyl3-
Figure imgf000047_0001
A 25 rηl round bottom flask equipped with stir bar and Claisen adapter with rubber septum'was charged with 585 mg (1.207 mmol) of (IR, 3aR, 4S, 7aR)-7a- methyl-1 -[(1 S)-6,6,6-trifluoro-5-hydroxy- 1 -(4-hydroxy-4-methyl-peπtyl)- 1 -methyI-5- trifluoromethyl-hex-3-ynyl]-octahydro-inden-4-oπe and 10 ml of dichlorom ethane. A 1.5 ml (10.2 ramol) of l-(trimethylsilyl)imidazole was added dropwise. The mixture was stirred at room temperature for 3h. A 150 ml of ethyl acetate was added and the mixture was washed three times with 50 ml of water, dried over Na2SOn and evaporated.
The oil residue was chromatographed on column (50 cm3) using hexane:ethyl acetate (10: 1) as mobile phase. Fractions containing product were pooled and evaporated to give 660 mg (87%) of product as colorless oil.
1H NMR (CDCl3): 2.44-2.39(3H, m), 2.32-2.16(2H, m), 2.10-1.99(2H, m), 1.95- 1.84(211, m), 1.77-1.56(41-1, m), 1.38-1.19(7H, m), 1.20(6H, s), 1.03(3H1 s), 0.74(3H, s), 0.28(9H, s), 0.10(9H, s)
(20S)-lα-FIuoro-2S-hydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-
2-ynyl)-cholecalciferol (6)
Figure imgf000048_0001
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 495 mg (1.052 mmol) of (lS,5R)-l-((/er/- butyldimethyl)silanyloxy)-3-[2-(diphenylfosphmoyl)-eth-(Z)-ylidene]-5-fluoro-2- methylene-cyclohexane and 10 ml of telrahydrofurane. The reaction mixture was cooled to -70° C and 0.65 ml (1.04 mmol) of 1.6M n-butyllithium was added dropwise. The resulting deep red solution was stirred at -70° C for 20 min and 300 mg (0.477 mmol) of (IR, 3aR, 4S, 7aR)-7a-methyl-l-[(lS)-6,6,6-trJfluoro-l -methyl- 1 -(4-methyl-4- trimethylsilanyloxy|-pentyl)-5-trifluoromethyl-5-trimethy]silanyloxy-hex-3-ynyl]- octahydro-inden-4 -one was added dropwise in 1.5 ml of tetrahydrofurane. The reaction mixture was stirred, for 4h and then the dry ice was removed from bath and the solution was allowed to warm up to -4O0C in Ih. The mixture was poured into 50 ml of ethyl acetate and 100 ml of brine. The water fraction was extracted three times with 50 ml of ethyl acetate, dried .over Na2SO-J and evaporated.
The oil residue was chromatographed on column (50 cm3, protected from light) using hexane:ethy! acetate (10:1) as mobile phase. Fractions containing product were pooled and evaporated to give colorless oil (ca. 429 mg) which was treated with 10 ml of IM tetrabutylammbnium fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 18h.
The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1 :1) and 50 ml of brine, dried over NazSO4 and evaporated.
The oil residue was ehromatographed on column (50 cm3, protected from light) using ethyl acctate:hexane (1 : 1) as mobile phase. Fractions containing product were pooled and evaporated to give product as colorless oil. The product was dissolved in methyl acetate and evaporated (2 times) to give 274 mg 92%) of product as white foam, {a] o = +27.0 c=0.50, EtOH
UV λmax (EtOH): 212 nm (ε 34256), 243 nm (ε 15866), 271 nm (ε 16512) MS HRES Calculated for: C33H45F7O3 [M+Na}+ 645.3149
Observed: [MB-Na]+ 645.3348
EXAMPLE 3
Synthesis of (20S)-l,25Φihydroxy~20-(5,5,5-trifluoro-4~hydroxy~4-trifltιoromeilιyl- pent-(2Z)-enyl)chotecaIciferol (2)
(3Z,6S)-l,l5I-Trifluoro-6-[(lR, 3aR, 4S5 7aR)-4-hydroxy-7a-methyl-octahydro- inden-l-ylJ-δ^O-dimethyi-Z-trifluoromethyl-undec^-ene-ZjlO-dioHlS)
Figure imgf000049_0001
A 25 ml round bottom flask was charged with 250 mg (0.514 mmoi) of (6S)- l ,l,l-trifiuoro-6-[(lR, 3aR> 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-inden-l-yl]-6,10- dimethyl-2-trifluoromethyl-undec-3-yne-2,10-diol, 70 mg of 5% Pd/CaCθ3, 6.0 ml of hexane, 2.4 ml of ethyl acetate and 0.23 ml of solution of qumoline in ethanol (prepared from 3.1 ml ofethano! and 168μi of quinoline).
The substrate was hydrogenated at ambient temperature and atmospheric pressure of hydrogen. The reaction was monitoring by TI-C (hexane:eihyl acetate - 2:1).
After 7h the catalyst was filtered off and solvent evaporated. The residue was purified over silica gel (125 cm ) using hexane:ethyl acetate (2:1) as a mobile phase. Fractions containing product were pooled and evaporated to give 243 mg (97%) of product as colorless oil. 1H NMR (CDCl3):' 6.14-6.05(1 H, m), 5.49(1 H, d, J=12.5 Hz), 4.08(1H, br s), 2.83(1H5 dd, J=I 5.9, 9.7 Hz), 2.48-2.3S(IH, m), 1.85-1.75(2H, m), 1.65-1.20(17H5 m), 1.22(3H, s), 1.20(3H, s), l.O8(3H, s), 1.03-0.96(1H3 m), 1.00(3H, s) 13C NMR (CDCi3): 140.22, 117.44, 71.79, 69.66, 56.74, 52.58, 44.1 1, 43.45, 41.19, 40.24, 39.64, 36.88, 33.44, 30.09, 28.88, 22.55, 22.21, 21.70, 17.63, 17.58, 16.54
(IR, 3aR, 4S, 7aR)-7a-Methyi-l-[(lS,3Z)-6,6,6-trifluoro-5-faydroxy-l-(4-hyctroxy-4- methyl-pentyl)-l-raethyI-S-trifluoromethyl-hex-3-enyl]-octahydro-inden-4-one (24)
Figure imgf000050_0001
» w
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 290 mg (0.594 mmbl) of (3Z,6S)-1,1 ,1-triflυoro-ό- [(1R, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-inden-l-y]]-6;10-dimcthyl-2- trifluoromethyl-u,ndcc-3-ene-2,10-diol and 10 ml of dichloromcthanc. A 700 mg (1.861 mmol) pyridiimirn dichromate and 750 mg of cclite was added and mixture was stirred in room temperature for 3h.
The reaction mixture was filtrated through column with silica gel (75 cm3) and celite (2 cm) and using dichloromethane : ethyl acetate (4:1) as a mobile phase. The fractions containing product were pooled and evaporated to give yellow oil. The product was used to the next reaction without farther purification.
(20S)-],25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-tπfluoroniethyl-pent-(2Z)-
Figure imgf000050_0002
A 25 rail round bottom flask equipped with stir bar and Claisen adapter with rubber septum'was charged with 1.800 g (3.088 mmol) of (lS,5R)-l,5-bis-((tert- bιityldimethyl)sxlanyloxy)-3-[2-(diplienylfosphinoy])-eth-(Z)-yliάene3-2-inethylcne- cyclohexane and 1 CLO ml of tetrahydrofurane. The reaction mixture was cooled to -780C and 1.9 ml (3.04 mmol) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. The resulting deep red solution was stirred at -780C for 20 min and 278 mg (0.571 mrnol) of (IR, 3aR, 4S, 7aR)-7a-meQiyl-l-[(lS,3Z)-6,6,6-trifluoio-5-hydroxy-H4- hydroxy-4-methyl-'pentyl)-l-methyl-5-trifiuoromethyl-hex-3-eny]]-octahydro-inden-4- one was added drqpwisc in 1.5 ml of tetrahydrofurane. The reaction mixture was stirred for 5h (last 0.5h at -2O0C) and then the bath was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine. The water fraction was extracted three times with 50 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The oil residue was chromatographed on column (75 cm3, protected from light) using hexane:etbyl acetate (4:1) as mobile phase. Fractions containing product were pooled and evaporated to give colorless oil (309 mg) which was treated with 5 ml of IM tetrabutylammonium fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 22h.
The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of water:brine (1:1) and 50 ml of brine, dried over Na2SO4 and evaporated.
The oil residue was chromatographed on column (50 cmJ, protected from light) using ethyl acetate as mobile phase. Fractions containing product were pooled and evaporated to give product as colorless oil. Oil was dissolved in methyl acetate and evaporated (4 tijnes) to give 192 mg (54%, two steps) of product as white foam.
OV λmax (EtOH): 204.08 nm (ε 27522), 266.03 nm (ε 20144) 1H NMR (CDCb): 6.37(1 H, d, J-I l.1 Hz), 6.1O(1H, ddd, J=12.5, 9.0, 6.0 Hz)3 6.00(1H, d, J=I 1.3 Hz), 5.47(1 H, d, 3=12.2 Hz), 5.32(1H, s), 5.07(1H, br, s), 4.99(1 H. s), 4.43(1 H, dd, J*7.8, 4.2 Hz), 4.25-4.20(1H, m), 2.85-2.79(2H, m), 2.59(1H, dd, J=I 3.4, 3.0 Hz), 2.46(IiH, dd, J-16.4, 4.9 Hz), 2.31(1H, dd, J=I 3.4, 6.4 Hz), 2.04-1.97(3H, m), 1.90(lH, ddd, 1=12.0, 8.2, 3.2 Hz), 1.76-1.20(17H, m), 1.21(3H, s), 1.20(3H, s), 1.06- 1.00(1 H, m)3 0,96(3H, s), 0.64(3H, s)
13C NMR (CTJfCI3): 147.51, 142.74, 140.17, 132,92, 124.88, 122.95(q, J=142.6 Hz), 122.80(q, J=14I.9 Hz), 117.52, 117.39, 11 1.65, 71.94, 70.73, 66.88, 56.86, 56.65, 46.79, 45.20, 43.95, 42.83, 41.06, 40.09, 39.75, 37.22, 30.35, 29.05, 28.82, 23.58, 22.50, 22.19, 21.93, 17.53, 15.04 MS HRES : Calculated for: C33H48F6O4 [M+Naf 645.3349 i Observed: [M+Naf 645.3350 J ! EXAMPLE 4
Synthesis pf(20S)-l,25-Dihydroxy~20-[(2E)-5,5,5-trifluoro-4~hydroxy-4- trifluoromethyl-pent-2-enyl]~cholecalciferol (3)
(3E,6S)-l,l,l-Trifluoro-6-[(lR, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro- indeii-l-yij-όjlO-dimcthyl^-trifluoromethyl-undec-S-ene-ljIO-dioI (25)
Figure imgf000052_0001
A 25 ml round bottom flask equipped with stir bar and condenser with nitrogen sweep was diaτged with 4.0 ml (4.0 mmol) of IM Jithium aluminum hydride in tetrahydrofurane. The mixture was cooled to 00C and 216"mg (4.00 mmol) of sodium methoxide was added slowly followed by 300 nig (0.637 mmol) of (6S)-I3I , I -trifluoro- 6-([(1R, 3aR, 4S5 7aR)-4-hydroxy-7a-methyl-octahydro-inden-l-yl>6,10-dimethyl-2- trifluoromethyl-undec-3-yne-2,10-diol in 4.0 ml of tetrahydrofurane. The reaction mixture was stirred at SO0C for 5h and then was cooled to O0C. A 1.0 ml of water, 1.0 ml of 2N NaOH aηd 20.0 ml of diethyl ether were added. The mixture was stirred at room temp for 30 min, 2.2 g of MgSO4 was added and mixture was stirred for next 15 min. The suspension was filtrated and solvent evaporated.
The oil residue was chromatographed on columns (100 cm3 and 30 cm3) using dichloromethane:ethyl acetate (4: 1) as mobile phase. Fractions containing product were pooled and evaporated to give 279 mg (93%) of product as colorless oil.
1H NMR (CDCl3): 6.32(1H, dt, J=15.7, 7.8 Hz), 5.59(1H, 15.7 Hz), 4.09(1H, br s), 2.29(2H, d, J=7.6 Hz), 2.01 (IH, br d, J=3.3 Hz), 1.86-1-75(2H, m), 1.63-1.04(18H, m), 1.21(6H, s), 1.09(3H, s), 0.98(3H, s)
13C NMR (CDCl3): 137.07, 119.81, 71.52, 69.54, 69.57, 57.20, 52.53, 44.16, 43.50, 42.29, 41.43, 40.10, 40.04, 33.39, 29.33, 29.29, 23.01, 22.17, 21.69, 17.86, 17.51 , 16.58
(lR, 3aR, 4S,;7aR)-7a-Methyl-l-[(lS,3E)-6,656-triπuoro-5-hydroxy-l-(4-hydroxy-4- methyI-pentyl)-l-metbyI-5-trifluαromethyϊ-hex-3-enyl]-octahydro-inden-4-oπe (26)
Figure imgf000053_0001
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 274 mg (0.561 mmol) of (6Ss3E)-l,l.,l-trifluoro~6- [(1R, 3aR, 4S, 7aR}-4-hydroxy-7a-methyl-octahydro-indeπ-l-yl]-6,10-dimethyl-2- trifluoromethyl-undec-3-ene-2,10-diol and 10 mi of dichloromethane. A 704 mg (1.871 mmol) of pyridinium dichromate and 740 mg of celite was added and mixture was stirred in room temperature for 2h.
The reaction mixture was filtrated through column with silica gel (100 cm3) using dichloromethane : ethyl acetate (4:1) as a mobile phase. The fractions containing product were pooled and evaporated to give 253 mg of yellow oil. The product was used to the next reaction without farther purification
(20S)-l,25~Dihy(iroxy-20-[(2E)-5,5,5-trifi«oro-4-hydroxy-4-trifluoromethyl-pent-2- cnylj-cholecaiciferol (3)
Figure imgf000053_0002
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 1.765 g (3 028 mmol) of (lS,5R)-l,5-bis-((fert- butyldimethyl)silanyloxy)-3-[2-(diphenylfosphinoyl)-cth-(Z)-ylideneJ-2-methylene- cyclohexane and 10.0 ml of tetrahydrofurarte. I'he reaction mixture was cooled to -780C and 1.8 ml (2.88 mmol) of 1.6M n-butyUithium in tetrahydrofurane was added dropwise. The resulting deep red solution was stirred at -780C for 20 min and 253 mg (0.520 mmol) Of (IR, 3aR, 43, 7aR)-7a-methyl-l-[(lS,3E)-6,6,6-trifluoro-5-hydroxy-l-(4- hydroxy^-methyl-penty^-l-mcthyl-D-trifluoromethyl-hex-S-enylj-octahydro-inden^- one was added dropwise in 1.5 ml of tetrahydrofurane. The reaction mixture was stirred for 5h (last 0.5h at -2O0C) and then the bath, was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine. The water fraction was extracted three times with 50 ml of ethyi acetate, dried over NajSCU and evaporated. The oil residue was chromatographed on column (60 cm3, protected from light) using hexane:ethyl acetate (4:1) as mobile phase. Fractions containing product were pooled and evaporated to give colorless oil (304 mg) which was treated with 5 ml of 1 M tetrabutylammoniυjm fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 2 Ih.
The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times'with 50 ml of wateπbrine (1 :1) and 50 ml of brine, dried over NajSθ4 and evaporated.
The oil residue was chromatographed on column (50 cm3, protected from light) using ethyl acetate fts mobile phase. Fractions containing product were pooled and evaporated to give product as colorless oil. Oil was dissolved in methyl acetate and evaporated (4 times) to give 176 mg (54%, two steps) of product as white foam.
[α] p = -4.5° C-0.33J CHCl3 UV λmax (EtOH): 204.50 nra (ε 1784ό),266.17 nm (ε 16508)
1H NMR (CDCl3): 6.36(1 H, d, J=11.3 Hz), 6.32(1H, dt, J=15.1, 7.5 Hz), 6.00(1H, d, J=I Ll Hz), 5.59(1 Ii, d, J=15.8 Hz, 5.33(1H, s), 4.99(1 H, s), 4.53(1H, br s), 4.43(1H3 dd, J=7,7, 4.3 Hz), 4.25-4.00(1H, m), 2.Sl(IH3 dd, J=12.1, 3.8 Hz), 2.59(1H1 dd, J=13.3, 2.9 Hz), 2.34-2.29(3H, m), 2.05-1.96(3H, m), 1.93-1.87(1H, m), L71-1.21(17H, τn), 1.21(6H, s), 1.12-1.{J5(1R, m), 0.95(3H, s), 0,66(3H, s)
13C NMR (CDCl3): si 47.48,- 142.53, 136.92, 133.05, 124.83, 122.39(q, J= 141.5 Hz), 1 19.76, 117.58, 1 17.49, 1 1 1, 71 , 71.61, 70.73, 66.90, 57.39, 56.62, 46.79, 45.18, 43.99, 42.83, 42.48, 41.29, 40.13, 40.04, 29.62, 29.28, 28.98, 23.50, 23.06, 22.24, 21.90, 17.74, 15.11 MS HRES Calculated for: C33H48F6O4 [M+Naf 645.3349
Observed: [M+Na]1 645.3346
i EXAMPLE 5
[ Synthesis of (20M)-l,25-Dihydroxy-20~(5,5s5-triflιιoro-4-hydroxy-4-trifluorotnethyL-
; pent-2-ynyl)-cholecatciferol (4)
(3R)-3-[(lR, 3aR, 4S, 7aR)~4-(tert-Butyl-dimethyl-sHanyIoxy)-7a~raethyI- octahydiϊo-inden-l-yI]-7-hydroxy-3,7-dirnethyl-octanal (30)
Figure imgf000055_0001
16 30
A 50 ml round bottom flask equipped with stir bar and CIaiscn adapter with rubber septum was charged with 1.558 g (7.228 mrnol) of pyridinium chlorochromate, 1.60 g of celite and 20 ml of dichloromethane. A 1.440 g (3.267 mmol) of (3R)-3-[(lR, 3aR, 4S3 7aR)-4-(tert-butyl-dimethyl-sihnyloxy)-7a-methyl-octahydro-mden-l-yl]-3,7- dimethyl-octane-l,7-diol in 10 mi of dichloromethane was added dropwise and mixture was stirred in
Figure imgf000055_0002
temperature for 2h 50min.
The reaction mixture was filtrated through column with silica gel (75 cm3) and celite (2 cm) and ysing dichloromethane, dichloromethane:ethyl acetate (4:1) as a mobile phase. The fractions containing product were pooled and evaporated to give 1.298 g of yellow oil. The product was used to the next reaction without farther purification.
I
(6R)~6-[(1R, 3aR} 4S, 7aR)-4-(tert-Buty!-dimethyl-siIanyloxy)-7a-methyl- octahydro-mden-l-ylJ-2,6-dimethyI-πon-8-yn-2-ol (31)
Figure imgf000055_0003
A 50 ml round bottom flask equipped with stir bar and Ciaiscn adapter with rubber septum was charged with 1.298 g (2.958 mmol) of (3R)-3-[(lR, 3aR, 4S, 7aR)-4- (tert-butyl-dimcthyl-silanyloxy)-7a-methyl-octahydro-indcn-l -yI]-7-hydroxy-317- dimethyl-octanal and 30 ml of methanol. A 1.137 g (5.916 mmol) of l-diazo-2-oxo- propyl)-phosphonic acid dimethyl ester in 3 ml of methanol was added and the resulting mixture was cooled in an ice bath to O0C. A 1.140 g (8.248 mmol) of potassium carbonate was addled and the reaction mixture was stirred in the ice bath for 30 min and then at room temperature for 2h 50 min. A 100 ml of water was added and the mixture was extracted three times with 80 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The oil residue wφ chromatographed on column (200 cm3) using hexane:ethyl acetate (7:1) as mobile phase. Fractions containing product were pooled and evaporated to give 1.151 g (81%) of product as colorless oil. [α] :p = +l 8.3° c=0.54, CHCl3
1HL NMR (CDCl3)I 3.99(1H, br s), 2.16-2.07(2H5 m), 2.00-1.97(1H, m), 1.92(1H, t, J=2.6 Hz), 1.84- 1.74(1 H, m), 1.67-1.64(1H, m), 1.58-1.22(16H, m), 1.22(6H, s), 1.04(3H, s), 0.99(3H, s), 0.88(9H, s), 0.00(3H, s), -0.01(3H, s)
MS HRES Calculated for: C27H50O2Si [M+Naf 457.3472
Observed: [M+Naf 457.3473
(1R? 3aR, 4S, 7aR)-4-(tert-ButyJ-dimethyI-silanyloxy)-l-l(lR)-l,5-dimethyl-l-prop- 2-ynyl-δ-ti"ϊmethyIsilaτιyloxy-hexyl]-7a-methyl-octahy(iro-indene (32)
,
Figure imgf000056_0001
31 M
A 50 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum w<jis charged with 1.151 g (2.647 mmol) of (6R)-6-[(iR, 3aR, 4S, 7aR)-4-
(tert-butyl-dimetiiyl-silanyloxy)-7a-methyl-octahydro-inden-l-yl]-2,6-dimethyl-non-8- yn-2-ol and 20 ml of dichloromethane. A 2.0 ml (13.63 mmol) of 1-
(trimethylsilyt)irήidazolc was added dropwise. The mixture was stirred at room temperature for ^ h.
A 100 rπϊ of water was added and the mixture was extracted three times with 50 ml of ethyl acetate, dried over Na24 and evaporated. The oil residue was chromatographed on column (75 cm3) using hexanc:ethyl acetate (25: 1) as; mobile phase. Fractions containing product were pooled and evaporated to give 1.260 g (94%) of product as colorless oil.
[α] % = +18.5° Of 0.46, CHCl3 1H NMR (CI)ClI3): 3.98(lH, br s), 2.12-2.08(2H, m), 20.5-1.95(2H, πi)s 1.92-1.9O(1H, m), l.S3-1.21(16H, m), 1.21(6H3 s), 1.04(3H, s), 0.98(3H, s), 0.88(9H, s), 0.11(9H, s),
O.00(3H, s), -O.φl(3H, s)
13C NMR (CI)Cl3): 83.00, 74.07, 69.70, 69.50, 56.63, 53.03, 45.66, 43.74, 41.35, 39.59,
39.45, 34.38, 29.99, 29.60, 25.85, 22.81, 22.43, 22.06, 18.56, 18.05, 17.76, 16.49, 2.65, - 4.77, -5.13
MS HRES Calculated for: C30H58O2Si2 [M+Naf 529.3867
Observed: [M+Naf 529.3868 (6R)-6-I(lR, 3aR, 4S, 7aR)-4-(tert-Butyl-dimethyl-silanyloxy)-7a-methyl- octahy hyl-10-
Figure imgf000057_0001
32 3S
A two neck SO ml round bottom flask equipped with stir bar, Claisen adapter with rubber septum and runnel (with cooling bath) was charged with 1.252 g (2.470 mmol) of (!R, 3aR, 4S, 7aR)-4-(tert-butyl-dimethyl-silanyloxy)-l-[(lR)-l,5-dimethyl-l- prop-2-ynyl-5-trime^hylsilanyloxy-hexyl]-7a-methyl-octahydro-indene and 25 ml of tetrahydrofurane. The funnel was connected to container with hexafluoroacetone and cooled (acetone, dry ice). The reaction mixture was cooled to
-7O0C and 2.4 nil (3.84 mmo3) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. After 30 niiin hexafluoroacetone was added (the container's valve was opened three times). The reaction was stirred at -700C for 2h then 5.0 ml of saturated solution of ammonium chloride ;was added.
The mixture was dissolved by the addition of 100 ml of saturated solution of ammonium chloride and extracted three times with 80 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The residue was chromatographed twice on columns (75 cm3) using hexane:ethyl acetate (10:1) as mobile phase to give 1.711 g of mixture of product and polymer (from hexafluoroacetone).
(6R)~l,l,ϊ-Trifluoro-6-[(lR, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-iπden- l-yl]-6,10*dimethyl-2-trifluoromethyl-undec-3-yne-2,10-diol (34)
Figure imgf000057_0002
A 25 ml rourid bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with erode (ca 2.470 mmol) (6R)-6-[(lR, 3aR, 4S, 7aR)-4- (tert-butyl-dimethyl-silany1oxy)~7a-methyl-octahydro-mden-l -yl]-l , 1 ,1 -trifluoro-6,10- dimethyl-24rifluofomethyl-10-tτimethylsilanyloxy~undec-3-yn-2-ol and 15.0 ml (15.0 mmol) of IM tetraibutylammonium fluoride in tetrahydrofurane. The reaction mixture was stirred at 7O0Q for 96h.
The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1 :1) and 50 ml of brine, dried over
Na2SO4 and evaporated. The oil residue was chromatographed on columns, 200cm3 and 75 cm3 using hexarie:ethyl acetate (2: 1). The fractions containing product were pooled and evaporated to !give 979 mg (81%) of product as colorless oil.
Figure imgf000058_0001
CHCl3
1H NMR (CDCl3]I: 4.08(1H, br s), 2.24(1H, AB, J=17.2 Hz), 2.17(1H, AB, J=17.2 Hz),
2.05-2.02(1H, m),;1.85-1.76(2H, m), 1.664.20(18H, m), 1.26(3H, s), 1.25(3H1 s),
1.07(3H, s), 1.01(3H, s)
MS HRES Calculated for: C24H36F6O3 [M+Na]+ 509.2461 Observed: [M+Naf 509.2463
(IR, 3aR, 4S, 7aR)-7a-Methyl-l-[(lR)-6,6,6-trifluoro-5-hydroxy-l-(4-hydroxy-4- methyi-pentyl)-l-methyl-5-trifluoromethyl-hex-3-yπyJ]-octahydro-inden-4-one (35)
Figure imgf000058_0002
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 291 mg (0.598 mmol) of (6R)-l,l,l-trifluoro-6-[(lR, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-inden-l -ylj-6,10-dimethyl-2- trifluoromethyl-uqdec-3-yne-2,10-diol and 10 ml of dichloromethane. A 700 mg (1.861 mmol) of pyridiniiam dichromate and 720 mg of celite was added and mixture was stirred in room temperature for 3h.
The reaction mixture was filtrated through column with silica gel (75 cm3) using dichloromethane, dichloromethane:ethyl acetate (4:1, 3:1). The fractions containing product were pooled and evaporated to give 271 mg (94%) of product as yellow oil.
(20R)-l,25-DiIjydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyI-pent-2- ynyl)-cholecalciferol (4)
Figure imgf000059_0001
A 25 ml round bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 2.118 g (3.634 mmol) of (lS,5R)-l ,5-bis-((/er/- butyldimethyl)silanyloxy)-3-[2-(diphenylfosphinoyl)-eth-(Z)-ylidenc]-2-methylene- cyclohexane and 10 mi of tetrahydrofurane. The reaction mixture was cooled to -78°C and 2.2 ml (3.52 mmol) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. The resulting deep red solution was stirred at -780C for 20 min and 271 mg, (0.559 mmol) Of (IR5 3aR, 4S, 7aR)-7a-methyl-l-[(lR,3E)-6,6,6-trifluoro-5-hydroxy-l-(4- hydroxy-4-methyl-pcntyl)-l-methyl-5-trifiuoromethyl-hex-3-ynyl]-octahydro-inden-4- one was added dropwise in 1.5 ml of tetrahydrofurane. The reaction mixture was stirred at -780C for 5h and then the bath was removed and the mixture was poured into 100 ml of saturated solution of ammonium chloride and extracted three times with 50 ml of ethyl acetate, dried over Na2SC^ and evaporated.
The oil residue was chromatographed on column (50 cm1, protected from light) using hexane:ethyl acetate (4: 1) as mobile phase. The fractions contains impurities was chromatographed on column (50 cm3, protected from light) using hexanerethyl acetate (5:1) as mobile phase. Fractions containing product were pooled and evaporated to give colorless oil (250 mg) which was treated with 5 ml of IM tetrabutylammonium fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 18h. The mixture was dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1 :1) and 50 ml of brine, dried over Na2SCi and evapprated.
The oil residue was chromatographed on column (50 cmJ, protected from light) using ethyl acetate as mobile phase. Fractions containing product were pooled and evaporated to give product as colorless oil. Oil was dissolved in methyl acetate and evaporated (4 times) to give 194 mg (56%) of product as white foam.
[α] 3 D° = +7.9° c=0;38, EtOH
UV λmax (EtOI-I): 212.33 nm (ε 141 13), 265.00 nra (ε 15960) 1H NMR (D6-DMSO): 8.93(1H, s), 6.18(1H, d, J=11.3 Hz), 5.96(1H, d, J=I 1.3 Hz), 5.22(1H, s)5 4.86(1H7 d, J=4.83 Hz), 4.75(1H, s), 4.54(1H, d, J=3.63 Hz)1 4.20-4.15(1H5 m), 4.06(1H, s),i3.98(lH, br s), 2.77(1H, d, J=13.7 Hz), 2.4O-2.33(1H, in), 2.27- 2.14(3H, m), 2.00-1.90(2H, m), 1.82-1.78(2H, m), 1.64-1.54(5H, m), 1.47-1.18(1OH, m), 1.05(3H9 s); 1.05(3H, s), 0.95(3H, s), 0.59(3H, s)
13C NMR (D6-DMSO): 149.38, 139,51 , 135.94, 122.32, 121.47(q, J=142.9 Hz), 1 17.99, 109.77, 89.53, 7^0.58, 68.72, 68.35, 65.06, 56.02, 55.91, 46.06, 44.S5, 44.65, 43.11, 29.30, 29.03, 28.78, 28.32, 23.05, 22.40, 21.90, 21.52, 18.27, 14.29 MS HRES Calculated for: Cj3H46F6O4 [M+Na]1" 643.3192 Observed: [M+Na]+ 643.3190
EXAMPLE 6 Synthesiβ of (20R)-l>25-Dihydroxy~20-[(2E)-5,5,5-trifluoro-4-hydrøxy~4- trifluoromethyl-pent-2-enylJ-cholecalciferol (5)
(3E,6R)-l,l,l-Trifluoro-6-{(lR, 3aR, 4S, 7aR)-4-hγdroxy-7a-methyl-octahydro- inden-l-tyl]-6,10-dimethyl-2-trifluoromethyl-un'dec-3-ene-2,10-(liol (36)
Figure imgf000060_0001
A 25 ml round bottom flask equipped with stir bar and condenser with nitrogen sweep was charged with 4.5 ml (4.5 mmol) of IM lithium aluminum hydride in tetrahydrofurane and the mixture was cooled to O0C. A 243 mg (4.50 mmol) of sodium methoxide was added slowly followed by substrate 337 mg (0.693 mmol) of (3E,6R)- 1 ,1,1 -trifluoro-6-[( 1 R, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-mden-l -yl]-6, 10- dimethyl-2-trifluoromethyl-undec-3-yne-2,10-diol in 5 ml of tetrahydrofurane. The reaction mixture was stirred at SO0C for 6h 30 min and then was cooled to O0C. A 1 ml of water, 1 ml of 2N NaOH and 20 ml of diethyl ether were added. The mixture was stirred at room;temp for 30 min and 2.2 g OfMgSO4 was added and mixture was stirred for next 15 miij. The suspension was filtrated and solvent evaporated. The oil Residue was chromatographed on column (100 cm3) using dichloromethane;ethyl acetate (4:1) as mobile phase. Fractions containing product were pooled and evaipαrated to give 330 mg (97%) of product as colorless oil. 1H NMR (CDCl3): 6.28(1H, dt, J=I 5.7, 7.3 Hz), 5.59(1H, d, J=15.4 Hz), 6.12(1H, br s), 2.12(2H, d, J=7.7 Hz), 2.06-1.98(1H5 m), 1.85-1.74(2H, m), 1.68-1.16(18H, m), 1.22(6H, s), 1.08(3H, s), 0.98(3H, s)
(IR, 3aR, methyl-p (37)
Figure imgf000061_0001
A 25 ml round bottom flask equipped with stir bar and Claiseii adapter with rubber septum was charged with 330 mg (0.675 mmol) of (3E,6Z)- 1,1,1 -trifluoro-6- [(1R, 3aR, 4S, 7aR)-4-hydroxy-7a-methyl-octahydro-inden-l-yl]-6,10-dimethyl-2- trifluoτomethyl-undec-3-ene-2,10-diol and 10 ml of dichloromethane. A 920 mg (2.445 mmol) of pyridinium dichromate was added and mixture was stirred in room temperature for 7h. The read ion mixture was filtrated through column with silica gel (60 cm3) using dichloromethane !: ethyl acetate (4: 1) as mobile phase. The fractions containing product were pooled and evaporated to give 302 mg (92%) of product as colorless oil.
[αl 3° = -l 7.7° c=0.46, CHCl3 1H NMR (CDCIi,): 6.30(1H, dt, J=15.6, 7.7 Hz), 5.60(IH, d, J=I 5.6 Hz), 2.40(1H, dd,
J=I l .1 , 7.3 Hz), 2.30-2.14(6H, m), 2.06-1.98(lH, m), 1.96-1.81(lH, m), l .78-1.30(13H, m), 1.24(3H, s), 1.23(3H, s), 0.98(3H, s), 0.74(3H, s)
13C NMR (CDCl3): 212.12, 136.27, 120.28, 71.45, 62.27, 57.44, 50.69, 44.28, 42.02,
40.76, 40.17, 39.69, 39.65, 29.34, 29.23, 23.98, 22.66, 22.24, 18.67, 18.19, 15.47 MS HRES Calculated for: C24H36F6O3 [M+Naf 509.2461
Observed: [M+Naf 509.2463
(IR, 3aR, 4S, 7aR)-7a-MethyM-[(lR,3E)-6,6,6-trifluoro-l-methyl-l-(4-methyl-4- trimethylsnaniyloxy-pentyl)-5-trifluororaethyl-5-trimetfaylsilanyloxy-hex-3-euyl]- octahydro-inden-4-one (38)
Figure imgf000062_0001
A 25 ml rouhd bottom flask equipped with stir bar and Claisen adapter with rubber septum was charged with 292 mg (0.600 mmol) of (IR, 3aR, 4S, 7aR)-7a- methyl-l-((lR,3E)-$,6,6-trifluoro-5-hydroxy-l-(4-hydroxy-4-methyl-pentyl)-l-methyl- 5-triflυoromethyi-hex-3-enyl]-octahydro-inden-4-one and 8 ml of dichloromethane. A 0.7 ml (4.8 mmol) of 1 -(trimethylsilyl)imidazole was added dropwise. The mixture was stirred at room temperature for 2h.
A 100 ml of water was added and the mixture was extracted three times with 50 ml of ethyl acetate, dried over Na2SO4 and evaporated.
The oil residue was chromatographed on column (60 cm3) using hexane:ethyl acetate (10:1, 4:1) as mobile phase. Fractions containing product were pooled and evaporated to give 360 mg (95%) of product as colorless oil.
-
with
Figure imgf000062_0002
to -78°C and 0.8 ml (1.28 mmol) of 1.6M n-butyllithium in tetrahydrofurane was added dropwise. The resulting deep red solution was stirred at -780C for 20 min and 358 mg (0.567 mmol) Of (IR, 3aR, 4S, 7aR)-7a-methyl-l-[(lR,3E)-6,6,6-trifluoro-l-methyl-l-(4- methy]-4-trimethylsilaπyloxy-penty!)-5-triflυoromethyl-5-trimethylsilanyloxy-hex-3- enylJ-octahydro-inden-4-one was added dropwise in 1.5 ml of tetrahydrofurane. The reaction mixture was starred for 4h (last 0.5h at -2O0C) and then the bath was removed and the mixture was poured into 50 ml of ethyl acetate and 100 ml of brine. The water fraction was extracted three times with 50 ml of ethyl acetate, dried OVeFNa2SO4 and evaporated. ;
The oil residjue was chrσmatographed on column (50 cm3, protected from light) using hexane:ethyl acetate (10;l) as mobile phase. Fractions containing product and some mono deprotected compound were pooled and evaporated to give colorless oil (440 mg) which was treated with 10 ml of IM tetrabutylammonium fluoride in tetrahydrofurane. The reaction mixture was stirred at room temperature for 2 Ih.
The mixtureαvas dissolved by the addition of 150 ml of ethyl acetate and extracted six times with 50 ml of wateπbrine (1 :1) and 50 ml of brine, dried over Na2SO4 and evaporated.
The oil residue was chromatographed on column (50 cm3, protected from light) using ethyl acetate as mobile phase. Fractions containing product were pooled and evaporated to give 3;0 5mg (86%, two steps) of product as colorless solid.
[o] so = +13.4° v=0M, EtOH
IJV λmax (EtOH): 212.76 nm (ε 15453), 265.03(ε 17341)
1H NMR (D6-DMSJO): 8.04(1H, s), 6.28(1H5 dt, J=15.5, 7.6 Hz)5 6.18(1H, d, J-I l .1 Hz), 5.97(1H, d, J=IjLl Hz), 5.61(IH, d, J=15.5 Hz), 5.22(1H, s), 4.75(1H5 s), 4.19- 4.16(1H, m), 3.98(I H, br s), 2.77(1H, d, 13.9 Hz), 2.35(1H, d, J=I 1.7 Hz), 2.16(1H, dd, J=13.6, 5.3 Hz), 2.07(2H, d, J=7.3 Hz)1 1.99-1.90(21-1, m), 1.81-1.78(1H, m), 1.64- 1.55(6H, m), 1.48-1 il7(12H, m), l .O5(6H, s), 0.90(3H, s), O.84(1H. s), 0.61(3H5 s) 13C NMR (D6-DMSO): 149.34, 139.65, 136.40, 135.82, 122.60(q, J=143.0 Hz), 122.32, 119.80, 117.90, 109J76, 68.68, 68.36, 65.04, 56.35, 56.00, 46.18, 44.85, 44.64, 43.09, 41.05, 40.42, 29.34, 129.12, 28.31, 23.08, 22.47, 21.79, 21.58, 17.91, 14.57 MS HRES Calculated for: Cj3H48F6O4 [M+Naf 645.3349
Observed: [M+Naf 645.3355
; EXAMPLE 7
Determination of Maximum Tolerated Dose (MTD) of Vitamin D3 Analogs i
The maximum tolerated dose of the vitamin D3 compounds of the invention were determined in eight Week-old female C57BL/6 mice (3 mice/group) dosed orally (0.1 ml/mouse) with various concentrations of Vitamin D3 analogs daily for four days.
Analogs were formulated in miglyol for a final concentration of 10, 30, 100 and 300 μg/kg when given atjθ.1 ml/mouse p.o. daily. Blood for serum calcium assay was drawn by tail bleed on day five, the final day of the study. Serum calcium levels were determined using a colorimetric assay (Sigma Diagnostics, procedure no. 597). The highest dose of ajnalog tolerated without inducing hypercalcemia (serum calcium >10.7 mg/dl) was taken as the maximum tolerated does (MTD). Table 1 shows the relative MTD for vitamjψ D3 compounds.
; Table 1
Figure imgf000064_0001
! EXAMPLE 8
: Immunological Assay of Vitamin D 3 Compounds
Immature dendritic cells (DC) were prepared as described in Romani, N. et al., J. Immunol. Meth. 196:137. IFN-γ production by allogeneic T cell activation in the mixed leukocyte response (MLR) was determined as described in Penna, G., et al, J. Immunol., 164: 2405-2411 (2000). Briefly, peripheral blood mononuclear cells (PBMC) were separated from buffy coats by Ficoll gradient and the same number (3x105) of allogeneic PBMC from 2 different donors were co-cultured in 96-well flat-bottom plates. After 5 days, IFN-γ production in the ϊtøLR. assay was measured by ELΪSA and the results expressed as amount (nM) of test compound required to induce 50% inhibition of IFN-γ production (IC50) (TaWe I). :
EXAMPLE 9
; Osteoporosis-Early curative treatment
Animals
Three-month old Sprague Dawley female rats were purchased from CERJ or Charles River, France. Rats underwent bilateral ovariectomy (OVX) or sham-operation (Sham) under anejsthesia with intraperitoneal ketamine hydrochloride (50 mg/kg BW). The success of ovariectomy was evidenced at necropsy by weighing the uterus and visualizing the absence of ovarian tissue.
Throughout the whole experiment, rats were housed at 22 ± 2°C with a 12h:12h light- dark cycle. The animals were pair-fed a standard diet (Safe, 0.6% P, 0,8% Ca) and received Eau de Volvic ad libitum. Experimental procedures were approved by the Animal Ethics Committee of
Prostrakan and by DDSV of Seine St Denis, France.
The treatment started 3 weeks post-ovariectomy. Compounds were firstly dissolved in ethatiol (lmg/ml). The other dilutions were done in Miglyol 812N. Compounds or vehicle (sham, OVX control rats) were given by daily oral gavage (5ml/kg), 5/7 days for 3 weeks. Just before operation a group of intact rats were sacrificed for baseline parameters. Before the beginning of treatment, groups of sham and OVX rats were sacrificed to serve as basal controls. Ten and three days before sacrifice, rats were given subcutaneous injections of calcein (lOmVkg, Sigma) to determine dynamic changes in bone tissue. The day before sacrifice, the rats were fasted and housed in metabolic cages to collect overnight urines.
Evaluation of bone parameters: pQCT, μCT, DEXA analysis
At necropsy, right long bones were removed and fixed in 70% ethanol for further μCT (tibia and femur) and histomorphometry (tibia) analysis. The whole left legs were collected for pQCT analysis. The fourth and fifth lumbar vertebrae were dissected for DXA analysis. .Excised tibias were scanned by a μCT machine (Scanco Medical) with software version 3j.l for a 2D-evaluation. The scans started at a distance of lmm of the reference line in the proximal tibia metaphysis. Five 0.5mm-spaced slices were analyzed. The slice thickness was 20-30μm. Bone volume (BV/TV) and trabeculae number (TbN) an<l thickness (TbTh) were assessed. Excised tibias were scanned by a pQCT machine (Stratec XCT Research SA+) with software version 5.4 to assess trabecular and coijtical Bone Mineral Density (BMD) of proximal tibia metaphysis. For analysis of trabecular bone, the distance between the reference line and the first measurement line; was 3 mm. Three otherlines separated by 1 mm were analyzed. The cortical BMD was analyzed at 20 mm from the reference line. The voxel size was 0.10 mm. The peel made used was 20/50. The BMD of excised fourth (L4) and fifth (L5) lumbar vertebrae ;were scanned by a DXA machine (Hologic QDR 4500) with software optimized for small animal studies. The regional high-resolution sofware selected a thin X-ray aperture. The large region was 68-71 and the narrow one was 35-21 to assess the body of the vertebrae of L4 and L5. ;
Histomorphometi-y analysis
Left tibia; from each rat was removed and dissected free of adjacent tissues. The bones were fixed in 70% cthanol, dehydrated in graded concentrations of ethanol, defatted in xylene, then embedded without decalcification in methyl methacrylate. Five μm-thick sections were made and stained with toluidine blue and cyanin solochrome and used for structural and cellular parameters evaluation. Ten μm-thick sections remained unstained for fluorescence microscopy observations. Structural and dynamic parameters were measured in the secondary spongiosa of the proximal tibial metaphysis situated about 1 mm distjal from the growth plate-epiphyseal junction. Structural parameters, i.e. trabecular boneivolume BV/TV (%), trabecular thickness (μm), connections and number, as well! as cellular parameters (osteoblast, osteoid surfaces and osteoclast number), and dynamic parameters (mineralizing surface, mineral apposition rate, bone formation rate) were evaluated, i Serum and Urinary Biochemistry
Osteocalcin (IRMA kit, Immutopics), DPyr ( Metra DPD EIA kit), CTx (Ratlaps
ELISA, Nordic! Bioscience Daignostics) Ca, P, creatinin (Cobas Mira analyser) and rat
PTH (Immutopics) were assessed in serum or urine material according to the manufacturer's; instructions.
Results Data were expressed as mean ± sem. Statistics were calculated using StatVievv version 5.0 for Windows (SAS Institute Inc.). The ANOVA test was used for all groups. Significant difference between groups was determined by Student's t-test. p<0.05 or lower was considered a significant difference. Figure 1 shows tibia proximal metaphysic bone volume (μCT) measurements in
3 month old OVX rέts.
Figure 2 shows lumbar spine BMD (DEXA) measurements in 3 month old OVX rats.
Figure 3 shows urinary calcium levels in 3 month old OVX rats. Figure 4 shows bone volume in 3 month old OVX rats using (1). The efficacy of
(1) vs. caicitrol on travccular bone volume was higher at 0.3 μg/kg.
Figure 5 sh'ows a reevaluation of 3 month old OVX rats for tibia proximal metaphysic bone volume (μCT).
Figure 6A.shows serum Ca levels in 3 month old rats. Figure 6B shows urinary Ca levels in 3 month old rats. Three month old female rats were orally dosed for three weeks, five days per week, with eight rats per group.
Compounds (1) and (3) demonstrated greater efficacy than calcitriol. Tibia (μCT) was found'to be 90% increased over OVX controls with 0.3 μg/kg in rats treated with (1). Tibia (]uCT) was found to be 114% increased over OVX controls with I μg/kg in rats treated with (X). The vertebrae (L5) was found to have an increase of 8% over OVX control rats when treated with 0.3 μg/kg of (1). The vertebrae (L5) was found to have an increases' of 12% over OVX control rats when treated with 1 μg/kg of (1).
Regarding safety, compound (1) provided rats with S Ca>10,7 mg/L in two of sixteen rats at 1 iμg/kg,
EXAMPLE 10
Osteoporosis- Long term curative treatment
Animals
Six month-old Sprague Dawley female rats were purchased from CERJ or
Charles River,! France. Rats underwent bilateral ovariectomy (OVX) or sham-operation
(Sham) under anesthesia with intraperitoneal ketamine hydrochloride (50 mg/kg BW).
The success of ovariectomy was evidenced at necropsy by weighing the uterus and visualizing the absence of ovarian tissue. Throughout the whole experiment, rats were housed at 22i 20C with a 12: 12h light-dark cycle. The animals were pair-fed a standard diet (Safe, 0.6% P; 0.8% Ca) and received Eau de Volvic ad libitum. Experimental procedures were approved by the Animal Ethics Committee of Prostrakan and by DDSV of Seine St Denis,: France. The treatment started 8 weeks post-ovariectomy. Compounds were firstly dissolved in ethanol (lmg/ml). The other dilutions were done in Migiyol 812N. Compounds or vehicle (sham, OVX control rats) were given by daily oral gavage (5ml/kg) , 5/7 days for 8 weeks. Just before operation a group of intact rats were sacrificed for basφline parameters. Before the beginning of treatment, groups of sham and OVX rats we're sacrificed to serve as basal controls. Ten and three days before sacrifice, rats were given subcutaneous injections of calcein (lOml/kg, Sigma) to determine dynamic changes in bone tissue. The day before sacrifice, the rats were fasted and housed in metabolic cages to collect overnight urines.
Evaluation of bone parameters: pQCT, μCT, DEXA analysis
At necropsy, right long bones were removed and fixed in 70% ethanol for further μCT (tibia and fέmur) and histomorphometry (tibia) analysis. The whole left legs were collected for pQCT analysis. The fourth and fifth lumbar vertebrae were dissected for DXA analysis. $xcised tibias were scanned by a μCT machine (Scanco Medical) with software version; 3.1 for a 2D-evaluation. The scans started at a distance of lmm of the reference line in'the proximal tibia metaphysis. Five 0.5mm-spaced slices were analyzed. The slice thickness was 20-30μm. Bone volume (BV/TV) and trabecule number (TbN) aind thickness (TbTh) were assessed. Excised tibias were scanned by a pQCT machine {Stratec XCT Research SA+) with software version 5.4 to assess trabecular and cortical Bone Mineral Density (BMD) of proximal tibia metaphysis. For analysis of trabecular bone, the distance between the reference line and the first measurement line was 3 mm. Three other lines separated by 1 mm were analyzed. The cortical BMD was analyzed at 20 mm from the reference line. The voxel size was 0.10 mm. The peel njiode used was 20/50. The BMD of excised fourth (L4) and fifth (L5) lumbar vertebrae were scanned by a DXA machine (Hologic QDR 4500) with software optimized for small animal studies. The regional high-resolution sofware selected a thin X-ray aperture.' The large region was 68-71 and the narrow one was 35-21 to assess the body of the vertebrae of L4 and L5.
Histomorphometry analysis
Left tibia from each rat was removed and dissected free of adjacent tissues. The bones were fixed in 70% ethanol, dehydrated in graded concentrations of ethanol, defatted in xylene, then embedded without decalcification in methyl methacrylate. B2006/001541
Five μm-thick sections were made and stained with toluidine blue and cyanin solochrome and uged for structural and cellular parameters evaluation. Ten μm-thick sections remained ;unstained for fluorescence microscopy observations. Structural and dynamic parameters were measured in the secondary spongiosa of the proximal tibial metaphysis situated about 1 mm distal from the growth plate-epiphyseal junction. Structural parameters, i.e. trabecular bone volume BV/TV (%), trabecular thickness (μm), connections and number, as well as cellular parameters (osteoblast, osteoid surfaces and osteoclast number), and dynamic parameters (mineralizing surface, mineral apposition rate, bone formation rate) were evaluated.
Serum and Urinary Biochemistry
Osteocalcin (IRMA kit, Immutopics), DPyr (Merra DPD EIA kit), CTx (Ratlaps ELISA, Nordic Hioscience Daignostics) Ca, P, creatinin (Cobas Mira analyser) and rat PTH (Immutopics) were assessed in serum or urine material according to the manufacturer's instructions.
Results '
Data were expressed as mean ± sem. Statistics were calculated using StatView version 5.0 for Windows (SAS Institute Inc.). The ANOVA test was used for all groups. Significant difference between groups was determined by Student's t-test. p<0.05 or lower was considered a significant difference.
Figure 7 :shows travecular bone volume (μCT) measurements in 6 month old OVX rats. ;
Figure 8|Shows urinary calcium levels in 6 month old OVX rats. Figure 9l shows BMD (DEXA) measurements in 6 month old OVX rats.
Figure ! OA shows serum calcium levels. Figure 1OB shows urinary calcium levels.
Figure IiI shows BMD (DEXA) measurements in 6 month old OVX rats.
' EXAMPLE 11
Secondary hyperparathyroidism
Animals ;
Three month-old Sprague Dawley male rats (220-25Og BW) were purchased from CERJ, Fiance, Rats were 5/6 nephrectomized (right kidney and 2/3 left kidney removed by surgery) or sham-operated (controls) by the supplier according to Prostrakan' procedures. The success of nephrectomy was evidenced at necropsy by visualizing the kidney tissue and measuring serum creatinin and urea. There were 10-12 rats in each group. The rats were housed at 22 ± 2°C with a 12h:12h light dark cycle. At the beginning ofthe study, the animals were fed a standard diet (Safe, 0.6% P, 0.8% Ca) and received Eau de Volvic ad libitum. Twenty days after operation, phosphate (Na2HPO4) was added to Eau de Volvic (6g/l). Experimental procedures were approved by the Animal Ethics Committee of Prostrakan and by DDSV of Seine St Denis, France. The treatment started 82 days post-nephrcctomy. Compounds were firstly dissolved in ethanoϊ (lmg/ml). The other dilutions were done in Miglyol 812N. Compounds or vehicle (control rats) were given by daily oral gavage (5 ml/kg), 5/7 days for 49 days. Just bøfore operation a group of intact rats were sacrificed for baseline parameters. Before' the beginning of treatment, groups of control and nephrectomized rats were sacrificed to served as basal controls. Ten and three days before sacrifice, rats were given subcutaneous injections of calcein (10ml/kg, Sigma) to determine dynamic changes in bone tissue. The day before sacrifice, the rats were fasted and housed in metabolic cages to' collect overnight urines.
Evaluation of bonέ parameters: pOCT, uCT, DEXA analysis
At necropsy, right long bones were removed and fixed in 70% ethanol for further μCT (tibia and feφur) and histomorphometry (tibia) analysis. The whole left legs were collected for pQCJT analysis. The fourth and fifth lumbar vertebrae were dissected for DXA analysis. Excised tϊbias were scanned by a μCT machine (Scanco Medical) with software version 3.1 for a 2D-evaluation. The scans started at a distance of lmm of the reference line in the proximal tibia metaphysis. Five 0.5mm-spaced slices were analyzed, The slide thickness was 20-30μrn. Bone volume (BV/TV) and trabeculae number (TbN) and thickness (TbTh) were assessed. Excised tibias were scanned by a pQCT machine (Stratec XCT Research SA+) with software version 5.4 to assess trabecular and coftical Bone Mineral Density (BMD) of proximal tibia metaphysis. For analysis of trabecular bone, the distance between the reference line and the first measurement line was 3 mm. Three other lines separated by 1 mm were analyzed. The cortical BMD was analyzed at 20 mm from the reference line. The voxel size was 0.10 mm. The peel mode used was 20/50. The BMD of excised fourth (L4) and fifth (L5) lumbar vertebrae' were scanned by a DXA machine (Ho logic QDR 4500) with software optimized for SmJaIl animal studies. The regional high-resolution sofware selected a thin X-ray aperture. The large region was 68-71 and the narrow one was 35-21 to assess the body of the vertejbrae of L4 and L5. 006/001541
Histomorphometrv analysis
Left tibia from each rat was removed and dissected free of adjacent tissues. The bones were fixed in 70% ethanol, dehydrated in graded concentrations of ethanol, defatted in xylene, then embedded without decalcification in methyl methacrylate. Five μm-thick sections were made and stained with toluidinε blue and cyanin solochrome and ujsed for structural and cellular parameters evaluation. Ten μm-thick sections remained unstained for fluorescence microscopy observations. Structural and dynamic parameters were measured in the secondary spongiosa of the proximal tibial metaphysis situated about 1 mm distal from the growth plate-epiphyseal junction. Structural parameters, i.e. trabecular bone volume BV/TV (%), trabecular thickness (μm), connections and number, as well as cellular parameters (osteoblast, osteoid surfaces and osteoclast number), and dynamic parameters (mineralizing surface, mineral i apposition rate, bone formation rate) were evaluated. j
Serum and Urinairy Biochemistry
Osteocalcin (IRMA kit, ϊmmutopics), DPyr ( Metra DPD EIA kit), CTx (Ratlaps
ELISA, Nordic Bioscience Daignostics) Ca, P, creatinin (Cobas Mira analyser) and rat
PTH (Immutopies) were assessed in serum or urine material according to the manufacturer's instructions.
Aortic calcifications
To assesj* the calcification, aortas (6 cm-segment starting at arch), hearts and remaining kidnqy tissues were removed and fixed for histology analysis. Aortas segment? were fixed in 3.7% formaldehyde and embedded in paraffin. Five μm cross sections wjere made and stained with a Von Kossa method for the calcification evaluation. Thejfollowing grading of the calcification was used: moderate when less
50% of the aortjc perimeter was calcified; severe when 100% of the aortic perimeter was calcified.
Results
Data w^re expressed as mean ± sem. Statistics were calculated using StatView version 5.0 for jWindows (SAS Institute Inc.). The ANOVA test was used for all groups.
Significant difference between groups was determined by Student's t-test. p<0.05 or lower was considered a significant difference. Figure 12Ai shows parathyroid hormone (PTH) levels in rats with moderate renal failure. Figure 12B shows serum Ca levels in rats with moderate renal failure.
Figures 13A and 13B show a model of safety parameters, measuring serum and serum Ca levels. Figure 14 A; shows a decrease in PTH levels in rats with severe chronic renal failure. Figure 14B shows the serum calcium levels in rats with severe chronic renal failure. ;
Figures 15 A and 15B show measurements of serum and serum Ca to determine safety profiles in rats with severe renal failure. Figure 16 A shows the trabecular bone volume measurements in uremic rats.
Figure 16B shows' bone mineral density (pQCT) measurements in uremic rats.
Figures 17,A5 17B, and 17C show tibia histomorphornetry analysis, measuring bone formation rate, osteoblast surface, and osteoclast number in uremic rats with moderate renal failure. Figure 18A is a picture of a rat tibia using optical microscopy (x50) of a normal trabecular Figur,e 18B is a picture of a rat tibia using optical microscopy (x50) of osteoid thickening. Figure 18C is a picture of a rat tibia using optical microscopy (x50) of peritτabecular fibrosis. Compound (2) provided normal levels of peritrabecular fibrosis at a dose of 3 μg/kg. At 1 μg/kg, only one often rats demonstrated peritravecular fibrosis. Compound (4) provided normal levels of peritrabecular fibrosis at a dose of 0.1 μlg/kg. At 0.03 μg/kg, only one of nine rats demonstrated peritrabecular Fibrosis. Compound (2) provided normal levels of osteoid thickening at a dose of 3 μg/kg and 1 μg/kg. Compound (4) provided normal levels of osteoid thickening at a dose of 0.03 μg/kg- At 0.1 μg/kg, only one of nine rats demonstrated osteoid thickening. Figure 19 shows bone mineral density (DEXA) in uremic rats.
Figure 20 is a picture of a rat femur cortical porosity using fluorescence microscopy (xS), showing normal porosity, mild porosity, medium porosity, and marked porosity. Compjound (2) provided mild porous levels of femur cortical porosity in four of ten rats at 1 μg/kg. At 3 μg/kg, nine rats demonstrated normal levels of femur cortical porosity. Compound (4) provided mild porous levels of femur cortical porosity in four of nine rats at 0103 μg/kg. Compound (4) provided mild porous levels of femur cortical porosity in one of nine rats at 0.1 μg/kg.
In rats with moderate renal failure, bone loss in CRF rats increased via bone turn over. Compounds (2) and (4) provided bone protection on the tibia and vertebrae; and demonstrated good efficacy. Figure 21 ils a picture of a cross section of a rat aorta using Von Kossa staining (xlOO), showing a; control, moderate aorta calcification, and severe aortic calcification. Compound (4) wais administered to seven rats at 0.03 μg/kg which provided uremia of 20.52 mM and S. breatinin value of 283.23 μM. No calcification was found in CRF control rats, but those rats presenting calcifications had severe renal failure.
Compouiwjls (2) and (4) demonstrated stronger inhibitition of renin in vitro over calcitriol. Both cojmpounds demonstrated efficacy in PTH suppression and bone porosity. Both compounds also demonstrated equal or more beneficial results in terms of safety profiles,; when compared to Zemplar. Compounds (2) and (4) demonstrated greater or equal positive results compared to Zemplar in the renin inhibition in vitro experiments, PTH suppression, CaxP, bone porosity, and aortic calcification.
Incorporation by Reference
The contdnts of all references (including literature references, issued patents, published patent Applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated herein in their entireties by reference.
Equivalents
Those skulled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described hereini Such equivalents are intended to be encompassed by the following claims.

Claims

1541What is claimed is:
1. A method ;for treating osteoporosis in a subject comprising administering to a subject in need thereof a therapeutically effective amount of a vitamin D3 compound selected from the group consisting of (20S)-l,25-Dihydroxy-20-(5,5,5-trifluoro-4- hydroxy^-tTifluoromethyl-pent^-yny^-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20- [(2E)-5,5 ,5-trifluόro-4-hydroxy-4-trif!uoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-lα-Fluoro-25-hydroxy-20-(5,5,5-trifIuoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (6), thereby treating said subject for osteoporosis.
2. The methpd of ciaim 1, wherein the vitamin D3 compound is (20S)-1 ,25- Dihydroxy-20-(5?5,5-trifluoro-4-hydroxy-4-trif}uoromethyl-pent-2-ynyl)-cholecaIciferol
(l). i
3 , The method of claim 1 , wherein the vitamin D3 compound is (20S)-1 ,25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4~hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (3).
4. The method of claim 1, wherein the vitamin D3 compound is (20S)- lα-Fluoro- 25-hydroxy-20-(j5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pcnt-2-ynyl)- cholecalciferol (ό).
5. The method of any of claims 1-4, which further comprises identifying said subject as being; in need of treatment for osteoporosis.
6. A rnethdd for treating a subject for secondary hyperparathyroidism comprising administering to; a subject in need thereof a therapeurically effective amount of a vitamin D3 compound selected from the group consisting of (20S)-1,25-Dihydroxy-20-((2Z)- 5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl)cholecalciferol (2); (20S)-1, 25- Dihydroxy-20-[i(2E)-5,5,5-trifluoro-4-hydroxy-4-trifiuoromethyl-pent-2-enylj- cholecalciferol (3); (20R)-1 ,25-Dihydroxy-20-(5,5,5-trifiuoro-4-hydroxy-4- trifluoromethyl ■pent-2-ynyl)-chokcalciferol (4); and (20R)-1,25-Dihydroxy-20-[(2E)-
S.S.S-trifluoro-i-hydroxy^-trifluoromethyl-pent^-enylj-cholecalciferol (5), thereby treating said subject for secondary hyperparathyroidism.
7. The method of claim 6, wherein the vitamin D3 compound is (20S)- 1 ,25- Dihydroxy-20-((2Z)i5)5,5-trifluoro-4-hydroxy-4-triπuoromethyl-pent-2- cnyl)cholecalciferol !{2).
8. The method of claim 6, wherein the vitamin D3 compound is (20S)-1 ,25- Dihydroxy-20-[(2E)'-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-eny3]- chokcalciferol (3).
9. The method ;of claim 6, wherein the vitamin D3 compound is (20R)-1 ,25- Dihydroxy-20-(535,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)-cholecalciferol
(4). ;
10. The method, of claim 6, wherein the vitamin D3 compound is (20R)-1 ,25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (5). '
1 1. The method! of any of claims 6-10, which further comprises identifying said subject as being in need of treatment for secondary hyperparathyroidism.
12. The method! of any of claims 1-11, further comprising obtaining the vitamin D3 compound.
13. The method of any of claims 1-12, wherein the subject is a mammal.
14. The method of claim 13, wherein the subject is human.
15. The method of any one of claims 1-14, wherein said vitamin D3 compound is administered to the subject using a pharmaceutically-acceptable formulation.
16. The method of any one of claims 1-14, wherein said vitamin D3 compound is administered in cotnbination with a pharmaceutically acceptable diluent or carrier.
17. The method of claim 15, wherein said pharmaceutically-acceptable formulation provides sustained! delivery of said vitamin D3 compound to a subject for at least four weeks after the pharmaceutically-acceptable formulation is administered to the subject.
18. The method: of any one of claims 1-17, wherein said compound is administered orally, intravenously, topically, or parenterally.
19, The method of any one of claims 1-18, wherein said compound is administered 5 at a concentration of 0.001 μg - 100 μg/kg of body weight.
20. A pharmaceutical composition for use in the treatment of osteroporosis, comprising a therapeutically effective amount of a vitamin Dj compound selected from
10 the group consisting of (20S)-1 ,25-Dihydroxy~20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethy]-psnt-2-ynyl)-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5- trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-Ia- Fluoro-25-hydroxy-20-(5,5,5-trifiuoro-4-hydroxy-4-rrifluoromethyl-pent-2-ynyl)- cholecalciferol (6), and a pharmaceutically acceptable diluent or carrier. i s ;
21. The pharmaceutical composition of claim 20, wherein the vitamin D3 compound is (20S)- 1 ,25-Dihydroxy-20-(5J5J5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)- cholecalciferol ( I). i
20 22. The pharmaceutical composition of claim 20, wherein the vitamin D3 compound is (20S)-l,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- enylj-cholecalciferol (3).
23. The pharmaceutical composition of claim 20, wherein the vitamin D3 compound 25 is (20S)-lα-Fluoiro-25-hydroxy-20-(5J5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciierol (6).
24. A pharmaceutical composition for use in the treatment of secondary hyperparathyroidism comprising a therapeutically effective amount of a vitamin D3
30 compound selected from the group consisting of (20S)-1 ,25-Dihydroxy-20-((2Z)-5,5.5- trifluoro-4-hydτpxy-4-trifluoromethyl-pent-2-enyl)cholecalciferol (2); (20S)-1 ,25- Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoroτnethyl-pent-2-enyl]- cholecalciferol (3); (20R)-1 ,25-Dihydroxy~20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl4pent-2-ynyl)-cholecalciferol (4); and (20R)-1,25-Dihydroxy-20-[(2E)-
35 5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pcnt-2-enyl]-cholecalciferol (5), and a pharmaceutically acceptable diluent or carrier.
25. The pharmaceutical composition of claim 24, wherein the vitamin Dj compound is (20S)-l,25-Dihy^roxy-20-((2Z)-5,5,5-trifiuoro-4-hydroxy-4-tiifluoromethyl-pent-2- enyl)cholecalcifeτol (2). '■
26. The pharmaceutical composition of claim 24, wherein the vitamin D3 compound is (20S)- 1 ,25-Dihydroxy-20-[(2E)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- enyl]-cholecalciferjot (3).
27. The pharmaceutical composition of claim 24, wherein the vitamin D3 compound is (20R)-l,25-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-ρent-2-ynyl)- cholecalciferol (4).
28. The pharmaceutical composition of claim 24, wherein the vitamin D3 compound is (20R)-1 ,25-Dihjydroxy-20-[(2E)-5,5,5-tril]uoro-4-hydroxy-4-trifluoromethyl-pent-2- enylj-cholecalciferol (5).
29. A packaged formulation for use in the treatment of osteoporosis, comprising a pharmaceutical composition comprising a vitamin D3 compound selected from the the group consisting of (20S)-1325-Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4- trifluoromethyl-pent-2-ynyl)-cholecalciferol (1); (20S)-1 ,25-Dihydroxy-20-[(2E)-5,5,5- trifluoro-4-hydroχy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (3); and (20S)-Ia- Fluoro-25-hydroxy-20-(5,5>5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)- cholecalciferol (6); and instructions for use in the treatment of osteoporosis.
30. The packaged formulation of claim 29, wherein the vitamin D3 compound is (20S)-l,25-Dihyjdroxy-20-(5,5J5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-ynyl)- cholecalciferol (1).
31 , The packaged formulation of claim 29, wherein the vitamin D3 compound is (20S)-l,25-Dthydroxy-20-[(2E)-5,5,5-trifiuoro-4-hydroxy-4-trifluoromethyl-pent-2- enyl]-cholecalci!ferol (3).
32. The packaged formulation of claim 29, wherein the vitamin D3 compound is (20S)-lα-Fluorό-25-hydroxy-20-(5,5J5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- ynyl)-cholecalciferol (6).
33. A packaged;formulation for use in the treatment of secondary hyperparathyroidism, comprising a pharmaceutical composition comprising a vitamin D3 compound seiectcd'from the group consisting of (20S)-l,25-Dihydroxy-20-((2Z)-5,5,5- trifluoro-4-hydroxyr4-rrif!uoromethyl-pent-2-eny])cholecalciferol (2); (20S)- 1 ,25- Dihydroxy-20-[(2B)-5,5,5-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]- cholecalciferol (3);; (20R)-1 ,2S~Dihydroxy-20-(5,5,5-trifluoro-4-hydroxy-4- tafluorornethyl-pent-2-ynyl)-choiecalciferol (4); and (20R)-1,25-Dihydroxy-20-[(2E)- 5,5,5-trif!uoro-4-hydroxy-4-trifluoromethyl-pent-2-enyl]-cholecalciferol (5), and instructions for use in the treatment of secondary hyperparathyroidism.
34. The packaged formulation of claim 33, wherein the vitamin D3 compound is (20S)-l525-Dihydroxy-20-((2Z)-5,5,5-trifluoτo-4-hydroxy-4-trifluoromethyl-pent-2- enyl)cholecalcifeφl (2), and instructions for use in the treatment of a subject for secondary hyperparathyroidism.
35. The packaged formulation of claim 33, wherein the vitamin D3 compound is (20S)-l,25-Dihydroxy-20-[(2E)-5,555-trifluoro-4-hydroxy-4-trifluoromethyl-pent-2- enyl]-cholecalciferol (3), and instructions for use in the treatment of a subject for secondary hyperparathyroidism.
36. The packaged formulation of claim 33, wherein the vitamin D3 compound is (20R)-l,25-Dihyjdroxy-20-(5,5,5-trifiuoro-4-hydroxy-4-trifiuoromethyl-pent-2-yπyl)- cholecalci feral (4), and instructions for use in the treatment of a subject for secondary hyperparathyroidism.
37. The packaged formulation of claim 33, wherein the vitamin D3 compound is (20R)-l:,25-Dihydroxy-20-[(2E)-5,5>5-trifluoro-4-hydroxy-4-trifluoromcthyl-pent-2- eny]]-cholecalcϊferol (5), and instructions for use in the treatment of a subject for secondary hyperparathyroidism.
PCT/IB2006/001541 2005-03-23 2006-03-23 Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds WO2006117684A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002602464A CA2602464A1 (en) 2005-03-23 2006-03-23 Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds
EP06765498A EP1898924A1 (en) 2005-03-23 2006-03-23 Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds
US11/886,922 US20090298799A1 (en) 2005-03-23 2006-03-23 Methods of Treating Osteoporosis and Secondary Hyperparathyroidism Using 20-Methyl, Gemini Vitamin D3 Compounds
TW095110651A TW200736207A (en) 2005-03-23 2006-03-28 Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin D3 compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US66439705P 2005-03-23 2005-03-23
US60/664,397 2005-03-23

Publications (1)

Publication Number Publication Date
WO2006117684A1 true WO2006117684A1 (en) 2006-11-09

Family

ID=36793191

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2006/001541 WO2006117684A1 (en) 2005-03-23 2006-03-23 Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds

Country Status (6)

Country Link
US (1) US20090298799A1 (en)
EP (1) EP1898924A1 (en)
CN (1) CN101198336A (en)
CA (1) CA2602464A1 (en)
TW (1) TW200736207A (en)
WO (1) WO2006117684A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1879596A1 (en) * 2005-03-23 2008-01-23 Bioxell S.p.a. 20-alkyl, gemini vitamin d3 compounds and methods of use thereof
US10406202B2 (en) 2014-10-22 2019-09-10 Extend Biosciences, Inc. Therapeutic vitamin D conjugates
US10420819B2 (en) 2014-10-22 2019-09-24 Extend Biosciences, Inc. Insulin vitamin D conjugates

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6030962A (en) * 1997-04-28 2000-02-29 Synttex (U.S.A.) Inc. Vitamin D3 analogs with bis C-20 side chains
WO2004098522A2 (en) * 2003-04-30 2004-11-18 Bioxell S.P.A. Gemini vitamin d3 compounds and methods of use thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6521608B1 (en) * 1998-03-27 2003-02-18 Oregon Health & Science University Vitamin D and its analogs in the treatment of tumors and other hyperproliferative disorders
US6516294B1 (en) * 1999-07-01 2003-02-04 The Regents Of The University Of California Nuclear receptor for 1α,25-dihydroxyvitamin D3 useful for selection of vitamin D3 ligands and a method therefor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6030962A (en) * 1997-04-28 2000-02-29 Synttex (U.S.A.) Inc. Vitamin D3 analogs with bis C-20 side chains
WO2004098522A2 (en) * 2003-04-30 2004-11-18 Bioxell S.P.A. Gemini vitamin d3 compounds and methods of use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WEYTS FRANCI A ET AL: "Novel Gemini analogs of 1alpha,25-dihydroxyvitamin D3 with enhanced transcriptional activity.", BIOCHEMICAL PHARMACOLOGY, vol. 67, no. 7, 1 April 2004 (2004-04-01), pages 1327 - 1336, XP002395606, ISSN: 0006-2952 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1879596A1 (en) * 2005-03-23 2008-01-23 Bioxell S.p.a. 20-alkyl, gemini vitamin d3 compounds and methods of use thereof
JP2008538114A (en) * 2005-03-23 2008-10-09 ビオクセル エッセ ピ ア 20-alkyl, geminivitamin D3 compounds and methods of use thereof
EP1879596A4 (en) * 2005-03-23 2009-01-21 Bioxell Spa 20-alkyl, gemini vitamin d3 compounds and methods of use thereof
US10406202B2 (en) 2014-10-22 2019-09-10 Extend Biosciences, Inc. Therapeutic vitamin D conjugates
US10420819B2 (en) 2014-10-22 2019-09-24 Extend Biosciences, Inc. Insulin vitamin D conjugates
US10702574B2 (en) 2014-10-22 2020-07-07 Extend Biosciences, Inc. Therapeutic vitamin D conjugates
US11116816B2 (en) 2014-10-22 2021-09-14 Extend Biosciences, Inc. Therapeutic vitamin d conjugates
US12076366B2 (en) 2014-10-22 2024-09-03 Extend Biosciences, Inc. Therapeutic vitamin D conjugates

Also Published As

Publication number Publication date
CN101198336A (en) 2008-06-11
US20090298799A1 (en) 2009-12-03
TW200736207A (en) 2007-10-01
CA2602464A1 (en) 2006-11-09
EP1898924A1 (en) 2008-03-19

Similar Documents

Publication Publication Date Title
DE69832699T2 (en) CYCLIC ETHERIC VITAMIN D3 COMPOUNDS, 1ALFA (OH) 3-EPI-VITAMIN D3 COMPOUNDS AND ITS USES
US20060258630A1 (en) Method for the prevention and treatment of uveitis
JP2006096759A (en) 2-alkyl-19-nor-vitamin d compound
US6372731B1 (en) Vitamin D derivatives with C-25 substituents, process for their preparation, intermediate products and their use in preparing medicaments
PL170447B1 (en) Method of obtaining novel 1 alpha-hydroxyvitamin d4 and novel transition and analog compounds
AU2005289664A1 (en) 20-cycloalkyl,26,27-alkyl/haloalkyl vitamin D3 compounds and methods of use thereof
EP0578494A1 (en) 24-cyclopropane vitamin D derivatives
US20020045606A1 (en) Esters of vitamin D3 and uses thereof
JP5005340B2 (en) 2-propylidene-19-nor-vitamin D compound
JP2015212294A (en) 1-deoxy analog of vitamin d-related compound
JP5770261B2 (en) (20S) -2-methylene-19-nor-22-dimethyl-1α, 25-dihydroxyvitamin D3 and (20R) -2-methylene-19-nor-22-dimethyl-1α, 25-hydroxyvitamin D3
EP1898924A1 (en) Methods of treating osteoporosis and secondary hyperparathyroidism using 20-methyl, gemini vitamin d3 compounds
JP2007505883A (en) Pharmaceutical composition and method comprising a combination of 2-alkylidene-19-nor-vitamin D derivative and parathyroid hormone
CA2601914A1 (en) 20-alkyl, gemini vitamin d3 compounds and methods of use thereof
RU2175318C2 (en) Derivatives of vitamin d3, methods of their synthesis, pharmaceutical composition
EP1663250A1 (en) Methods for treating bladder dysfunction
JP2021167319A (en) Synthesis and biological activity of 2-methylene analogs of calcitriol and related compounds
WO2012166938A2 (en) (22e)-2-methylene-26,27-cyclo-22-dehydro-1alpha-hydroxy-19-norvitamin d3 derivatives
AU2010324596A1 (en) 2-methylene-19,26-nor-(20S)-1alpha-hydroxyvitamin D3
RU2261244C2 (en) New analogues of vitamin d
WO2007110109A1 (en) Novel method
WO2006035075A1 (en) Use of vitamin d compounds for the prevention or treatment of chronic prostatitis
ZA200602097B (en) Methods for treating bladder dysfunction

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2602464

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006765498

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: RU

WWW Wipo information: withdrawn in national office

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2006765498

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11886922

Country of ref document: US